Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Brain Multiphys ; 52023 Dec.
Article in English | MEDLINE | ID: mdl-38187909

ABSTRACT

Rhenium-186 (186Re) labeled nanoliposome (RNL) therapy for recurrent glioblastoma patients has shown promise to improve outcomes by locally delivering radiation to affected areas. To optimize the delivery of RNL, we have developed a framework to predict patient-specific response to RNL using image-guided mathematical models. Methods: We calibrated a family of reaction-diffusion type models with multi-modality imaging data from ten patients (NCR01906385) to predict the spatio-temporal dynamics of each patient's tumor. The data consisted of longitudinal magnetic resonance imaging (MRI) and single photon emission computed tomography (SPECT) to estimate tumor burden and local RNL activity, respectively. The optimal model from the family was selected and used to predict future growth. A simplified version of the model was used in a leave-one-out analysis to predict the development of an individual patient's tumor, based on cohort parameters. Results: Across the cohort, predictions using patient-specific parameters with the selected model were able to achieve Spearman correlation coefficients (SCC) of 0.98 and 0.93 for tumor volume and total cell number, respectively, when compared to the measured data. Predictions utilizing the leave-one-out method achieved SCCs of 0.89 and 0.88 for volume and total cell number across the population, respectively. Conclusion: We have shown that patient-specific calibrations of a biology-based mathematical model can be used to make early predictions of response to RNL therapy. Furthermore, the leave-one-out framework indicates that radiation doses determined by SPECT can be used to assign model parameters to make predictions directly following the conclusion of RNL treatment. Statement of Significance: This manuscript explores the application of computational models to predict response to radionuclide therapy in glioblastoma. There are few, to our knowledge, examples of mathematical models used in clinical radionuclide therapy. We have tested a family of models to determine the applicability of different radiation coupling terms for response to the localized radiation delivery. We show that with patient-specific parameter estimation, we can make accurate predictions of future glioblastoma response to the treatment. As a comparison, we have shown that population trends in response can be used to forecast growth from the moment the treatment has been delivered.In addition to the high simulation and prediction accuracy our modeling methods have achieved, the evaluation of a family of models has given insight into the response dynamics of radionuclide therapy. These dynamics, while different than we had initially hypothesized, should encourage future imaging studies involving high dosage radiation treatments, with specific emphasis on the local immune and vascular response.

2.
Biomed Phys Eng Express ; 7(4)2021 05 28.
Article in English | MEDLINE | ID: mdl-34050041

ABSTRACT

Convection-enhanced delivery of rhenium-186 (186Re)-nanoliposomes is a promising approach to provide precise delivery of large localized doses of radiation for patients with recurrent glioblastoma multiforme. Current approaches for treatment planning utilizing convection-enhanced delivery are designed for small molecule drugs and not for larger particles such as186Re-nanoliposomes. To enable the treatment planning for186Re-nanoliposomes delivery, we have developed a computational fluid dynamics approach to predict the distribution of nanoliposomes for individual patients. In this work, we construct, calibrate, and validate a family of computational fluid dynamics models to predict the spatio-temporal distribution of186Re-nanoliposomes within the brain, utilizing patient-specific pre-operative magnetic resonance imaging (MRI) to assign material properties for an advection-diffusion transport model. The model family is calibrated to single photon emission computed tomography (SPECT) images acquired during and after the infusion of186Re-nanoliposomes for five patients enrolled in a Phase I/II trial (NCT Number NCT01906385), and is validated using a leave-one-out bootstrapping methodology for predicting the final distribution of the particles. After calibration, our models are capable of predicting the mid-delivery and final spatial distribution of186Re-nanoliposomes with a Dice value of 0.69 ± 0.18 and a concordance correlation coefficient of 0.88 ± 0.12 (mean ± 95% confidence interval), using only the patient-specific, pre-operative MRI data, and calibrated model parameters from prior patients. These results demonstrate a proof-of-concept for a patient-specific modeling framework, which predicts the spatial distribution of nanoparticles. Further development of this approach could enable optimizing catheter placement for future studies employing convection-enhanced delivery.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/diagnostic imaging , Convection , Glioblastoma/diagnostic imaging , Humans , Neoplasm Recurrence, Local , Radioisotopes , Rhenium
3.
Sci Rep ; 7(1): 2574, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566692

ABSTRACT

At present, 64Cu(II) labeled tracers including 64CuCl2 have been widely applied in the research of molecular imaging and therapy. Human copper transporter 1 (hCTR1) is the major high affinity copper influx transporter in mammalian cells, and specially responsible for the transportation of Cu(I) not Cu(II). Thus, we investigated the feasible application of 64Cu(I) for PET imaging. 64Cu(II) was reduced to 64Cu(I) with the existence of sodium L-ascorbate, DL-Dithiothreitol or cysteine. Cell uptake and efflux assay was investigated using B16F10 and A375 cell lines, respectively. Small animal PET and biodistribution studies were performed in both B16F10 and A375 tumor-bearing mice. Compared with 64Cu(II), 64Cu(I) exhibited higher cellular uptake by melanoma, which testified CTR1 specially influx of Cu(I). However, due to oxidation reaction in vivo, no significant difference between 64Cu(I) and 64Cu(II) was observed through PET images and biodistribution. Additionally, radiation absorbed doses for major tissues of human were calculated based on the mouse biodistribution. Radiodosimetry calculations for 64/67Cu(I) and 64/67Cu(II) were similar, which suggested that although melanoma were with high radiation absorbed doses, high radioactivity accumulation by liver and kidney should be noticed for the further application. Thus, 64Cu(I) should be further studied to evaluate it as a PET imaging radiotracer.


Subject(s)
Copper Radioisotopes/pharmacology , Melanoma/diagnostic imaging , Molecular Imaging/methods , Positron-Emission Tomography/methods , Animals , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Line, Tumor , Copper Radioisotopes/chemistry , Copper Transporter 1 , Humans , Melanoma/pathology , Mice , Pilot Projects , Tissue Distribution
4.
Methods Mol Biol ; 1522: 155-178, 2017.
Article in English | MEDLINE | ID: mdl-27837538

ABSTRACT

Liposomes can serve as carriers of radionuclides for diagnostic imaging and therapeutic applications. Herein, procedures are outlined for radiolabeling liposomes with the gamma-emitting radionuclide, technetium-99m (99mTc), for noninvasive detection of disease and for monitoring the pharmacokinetics and biodistribution of liposomal drugs, and/or with therapeutic beta-emitting radionuclides, rhenium-186/188 (186/188Re), for radionuclide therapy. These efficient and practical liposome radiolabeling methods use a post-labeling mechanism to load 99mTc or 186/188Re into preformed liposomes prepared in advance of the labeling procedure. For all liposome radiolabeling methods described, a lipophilic chelator is used to transport 99mTc or 186/188Re across the lipid bilayer of the preformed liposomes. Once within the liposome interior, the pre-encapsulated glutathione or ammonium sulfate (pH) gradient provides for stable entrapment of the 99mTc and 186/188Re within the liposomes. In the first method, 99mTc is transported across the lipid bilayer by the lipophilic chelator, hexamethylpropyleneamine oxime (HMPAO) and 99mTc-HMPAO becomes trapped by interaction with the pre-encapsulated glutathione within the liposomes. In the second method, 99mTc or 186/188Re is transported across the lipid bilayer by the lipophilic chelator, N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA), and 99mTc-BMEDA or 186/188Re-BMEDA becomes trapped by interaction with pre-encapsulated glutathione within the liposomes. In the third method, an ammonium sulfate (pH) gradient loading technique is employed using liposomes with an extraliposomal pH of 7.4 and an interior pH of 5.1. BMEDA, which is lipophilic at pH 7.4, serves as a lipophilic chelator for 99mTc or 186/188Re to transport the radionuclides across the lipid bilayer. Once within the more acidic liposome interior, 99mTc/186/188Re-BMEDA complex becomes protonated and more hydrophilic, which results in stable entrapment of the 99mTc/186/188Re-BMEDA complex within the liposomes. Since many commercially available liposomal drugs use an ammonium sulfate (pH) gradient for drug loading, these liposomal drugs can be directly radiolabeled with 99mTc-BMEDA for noninvasive monitoring of tissue distribution during treatment or with 186/188Re-BMEDA for combination chemo-radionuclide therapy.


Subject(s)
Diagnostic Imaging/methods , Liposomes/chemistry , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Rhenium/chemistry , Technetium/chemistry , Ammonium Sulfate/chemistry , Ethylenediamines/chemistry , Glutathione/chemistry , Humans , Hydrogen-Ion Concentration , Organometallic Compounds/chemistry , Organotechnetium Compounds/chemistry , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Whole Body Imaging
5.
Expert Opin Drug Deliv ; 13(6): 873-89, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26981891

ABSTRACT

INTRODUCTION: A major limitation of current liposomal cancer therapies is the inability of liposome therapeutics to penetrate throughout the entire tumor mass. This inhomogeneous distribution of liposome therapeutics within the tumor has been linked to treatment failure and drug resistance. Both liposome particle transport properties and tumor microenvironment characteristics contribute to this challenge in cancer therapy. This limitation is relevant to both intravenously and intratumorally administered liposome therapeutics. AREAS COVERED: Strategies to improve the intratumoral distribution of liposome therapeutics are described. Combination therapies of intravenous liposome therapeutics with pharmacologic agents modulating abnormal tumor vasculature, interstitial fluid pressure, extracellular matrix components, and tumor associated macrophages are discussed. Combination therapies using external stimuli (hyperthermia, radiofrequency ablation, magnetic field, radiation, and ultrasound) with intravenous liposome therapeutics are discussed. Intratumoral convection-enhanced delivery (CED) of liposomal therapeutics is reviewed. EXPERT OPINION: Optimization of the combination therapies and drug delivery protocols are necessary. Further research should be conducted in appropriate cancer types with consideration of physiochemical features of liposomes and their timing sequence. More investigation of the role of tumor associated macrophages in intratumoral distribution is warranted. Intratumoral infusion of liposomes using CED is a promising approach to improve their distribution within the tumor mass.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Animals , Humans , Liposomes
6.
Adv Drug Deliv Rev ; 76: 39-59, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25016083

ABSTRACT

One of the major limitations of current cancer therapy is the inability to deliver tumoricidal agents throughout the entire tumor mass using traditional intravenous administration. Nanoparticles carrying beta-emitting therapeutic radionuclides that are delivered using advanced image-guidance have significant potential to improve solid tumor therapy. The use of image-guidance in combination with nanoparticle carriers can improve the delivery of localized radiation to tumors. Nanoparticles labeled with certain beta-emitting radionuclides are intrinsically theranostic agents that can provide information regarding distribution and regional dosimetry within the tumor and the body. Image-guided thermal therapy results in increased uptake of intravenous nanoparticles within tumors, improving therapy. In addition, nanoparticles are ideal carriers for direct intratumoral infusion of beta-emitting radionuclides by convection enhanced delivery, permitting the delivery of localized therapeutic radiation without the requirement of the radionuclide exiting from the nanoparticle. With this approach, very high doses of radiation can be delivered to solid tumors while sparing normal organs. Recent technological developments in image-guidance, convection enhanced delivery and newly developed nanoparticles carrying beta-emitting radionuclides will be reviewed. Examples will be shown describing how this new approach has promise for the treatment of brain, head and neck, and other types of solid tumors.


Subject(s)
Nanoparticles , Neoplasms/diagnostic imaging , Neoplasms/therapy , Radiopharmaceuticals , Radiotherapy, Image-Guided , Animals , Humans , Hyperthermia, Induced , Nanoparticles/therapeutic use , Radionuclide Imaging , Radiopharmaceuticals/therapeutic use
7.
Nucl Med Commun ; 34(9): 909-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23708872

ABSTRACT

INTRODUCTION: Radiolabeled Arg-Gly-Asp (RGD) and bombesin (BBN) heterodimers have been investigated for dual targeting of tumor integrin αvß3 receptors and gastrin-releasing peptide receptors. The goal of this study was to evaluate the potential use of a Lu-labeled RGD-BBN heterodimer for targeted prostate cancer therapy. MATERIALS AND METHODS: A 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-conjugated RGD-BBN peptide (DO3A-RGD-BBN) was radiolabeled with Lu and purified by high-performance liquid chromatography. The in-vivo biodistribution study of Lu-DO3A-RGD-BBN was carried out in mice bearing human prostate cancer PC3 xenografts. The receptor-targeting specificity of the radiolabeled peptide was assayed by injecting the tracer with the unlabeled RGD-BBN peptide. Radiation absorbed doses in adult male patients, based on biodistribution data from mice, were also calculated. RESULTS: DO3A-RGD-BBN peptides were successfully labeled with Lu, and high radiochemical purity (>95%) could be achieved after high-performance liquid chromatography purification. In human PC3 xenograft-bearing mice, the tumor accumulation of Lu-DO3A-RGD-BBN was 5.88±1.12, 2.77±0.30, 2.04±0.19, and 1.18±0.19%ID/g at 0.5, 2, 24, and 48 h, respectively. With rapid clearance from normal tissues, the radiolabeled probe displayed high tumor-to-blood and tumor-to-muscle ratios. On calculating the radiation absorbed doses for Lu-DO3A-RGD-BBN, we found that the prostate tumor and the pancreas were the organs receiving the highest radiation absorbed doses. CONCLUSION: Dual integrin αvß3 and GPRP-targeted agent Lu-DO3A-RGD-BBN shows excellent prostate cancer-targeting ability, and it is worthy of further evaluation for prostate cancer-targeted therapy.


Subject(s)
Bombesin/chemistry , Bombesin/therapeutic use , Dimerization , Lutetium/therapeutic use , Molecular Targeted Therapy/methods , Oligopeptides/chemistry , Prostatic Neoplasms/radiotherapy , Animals , Aza Compounds/chemistry , Bombesin/metabolism , Bombesin/pharmacokinetics , Cell Line, Tumor , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Male , Mice , Prostatic Neoplasms/pathology , Radiation Dosage , Radiochemistry , Radioisotopes/therapeutic use
8.
J Liposome Res ; 23(1): 70-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23210622

ABSTRACT

Camptothecin (CPT) represents a potent anticancer drug. However, its therapeutic use is impaired by both drug solubility, hydrolysis, and protein interactions in vivo. Use of liposomes as a drug-formulation approach could overcome some of these challenges. The aim of this study was to perform a mechanistic study of the incorporation and retention of the lipophilic parent CPT compound in different liposome formulations using radiolabeled CPT and thus to be able to identify promising CPT delivery systems. In this context, we also wanted to establish an appropriate mouse tumor model, in vivo scintigraphic imaging, and biodistribution methodology for testing the most promising formulation. CPT retention in various liposome formulations after incubation in buffer and serum was determined. The HT-29 mouse tumor model, (111)In-labeled liposomes, as well as (3)H-labeled CPT were used to investigate the biodistribution of liposomes and drug. The ability of different liposome formulations to retain CPT in buffer was influenced by lipid concentration and drug/lipid ratio, rather than lipid composition. The tested formulations were cleared from the blood in the following order: CPT solution > CPT liposomes > (111)In-labeled liposomes, and liposomes mainly accumulated in the liver. Lipid composition did not influence CPT retention to the same extent as earlier observed from incorporation studies. The set-up for the biodistribution study works well and is suited for future in vivo studies on CPT liposomes. The biodistribution study showed that liposomes circulated longer than free drug, but premature release of drug from liposomes occurred. Further studies to develop formulations with higher retention potential and prolonged circulation are desired.


Subject(s)
Camptothecin/administration & dosage , Drug Delivery Systems , Liposomes/administration & dosage , Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/chemistry , Humans , Lipids , Liposomes/chemistry , Mice , Neoplasms/pathology , Solubility , Tissue Distribution
9.
Mol Pharm ; 9(9): 2513-22, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22894603

ABSTRACT

Most diagnosed early stage breast cancer cases are treated by lumpectomy and adjuvant radiation therapy, which significantly decreases the locoregional recurrence but causes inevitable toxicity to normal tissue. By using a technique of preparing liposomes carrying technetium-99m ((99m)Tc), rhenium-186 ((186)Re), or rhenium-188 ((188)Re) radionuclides, as well as chemotherapeutic agents, or their combination, for cancer therapy with real time image-monitoring of pharmacokinetics and prediction of therapy effect, this study investigated the potential of a novel targeted focal radiotherapy with low systemic toxicity using radioactive immunoliposomes to treat both the surgical cavity and draining lymph nodes in a rat breast cancer xenograft positive surgical margin model. Immunoliposomes modified with either panitumumab (anti-EGFR) or bevacizumab (anti-VEGF) were remote loaded with (99m)Tc diagnostic radionuclide, and injected into the surgical cavity of female nude rats with positive margins postlumpectomy. Locoregional retention and systemic distribution of (99m)Tc-immunoliposomes were investigated by nuclear imaging, stereofluorescent microscopic imaging, and gamma counting. Histopathological examination of excised draining lymph nodes was performed. The locoregional retention of (99m)Tc-immunoliposomes in each animal was influenced by the physiological characteristics of the surgical site of individual animals. Panitumumab- and bevacizumab-liposome groups had higher intracavitary retention compared with the control liposome groups. Draining lymph node uptake was influenced by both the intracavitary radioactivity retention level and metastasis status. The panitumumab-liposome group had higher accumulation on the residual tumor surface and in the metastatic lymph nodes. Radioactive liposomes that were cleared from the cavity were metabolized quickly and accumulated at low levels in vital organs. Therapeutic radionuclide-carrying specifically targeted panitumumab- and bevacizumab-liposomes have increased potential compared to non-antibody targeted liposomes for postlumpectomy focal therapy to eradicate remaining breast cancer cells inside the cavity and draining lymph nodes with low systemic toxicity.


Subject(s)
Immunotoxins/administration & dosage , Liposomes/administration & dosage , Lymph Nodes/diagnostic imaging , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/radiotherapy , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Bevacizumab , Female , Immunotoxins/chemistry , Liposomes/chemistry , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/surgery , Mastectomy, Segmental/methods , Panitumumab , Particle Size , Radioisotopes/chemistry , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Rats , Rats, Nude , Rhenium/administration & dosage , Rhenium/chemistry , Technetium/administration & dosage , Technetium/chemistry
10.
Bioconjug Chem ; 23(6): 1322-32, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22577859

ABSTRACT

Liposomes are effective lipid nanoparticle drug delivery systems, which can also be functionalized with noninvasive multimodality imaging agents with each modality providing distinct information and having synergistic advantages in diagnosis, monitoring of disease treatment, and evaluation of liposomal drug pharmacokinetics. We designed and constructed a multifunctional theranostic liposomal drug delivery system, which integrated multimodality magnetic resonance (MR), near-infrared (NIR) fluorescent and nuclear imaging of liposomal drug delivery, and therapy monitoring and prediction. The premanufactured liposomes were composed of DSPC/cholesterol/Gd-DOTA-DSPE/DOTA-DSPE with the molar ratio of 39:35:25:1 and having ammonium sulfate/pH gradient. A lipidized NIR fluorescent tracer, IRDye-DSPE, was effectively postinserted into the premanufactured liposomes. Doxorubicin could be effectively postloaded into the multifunctional liposomes. The multifunctional doxorubicin-liposomes could also be stably radiolabeled with (99m)Tc or (64)Cu for single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging, respectively. MR images displayed the high-resolution micro-intratumoral distribution of the liposomes in squamous cell carcinoma of head and neck (SCCHN) tumor xenografts in nude rats after intratumoral injection. NIR fluorescent, SPECT, and PET images also clearly showed either the high intratumoral retention or distribution of the multifunctional liposomes. This multifunctional drug carrying liposome system is promising for disease theranostics allowing noninvasive multimodality NIR fluorescent, MR, SPECT, and PET imaging of their in vivo behavior and capitalizing on the inherent advantages of each modality.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Carcinoma, Squamous Cell/diagnosis , Doxorubicin/analogs & derivatives , Drug Delivery Systems , Head and Neck Neoplasms/diagnosis , Liposomes/chemistry , Animals , Carcinoma, Squamous Cell/drug therapy , Copper Radioisotopes/chemistry , Doxorubicin/administration & dosage , Fluorescent Dyes/chemistry , Head and Neck Neoplasms/drug therapy , Heterocyclic Compounds/chemistry , Male , Optical Imaging , Organometallic Compounds/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/administration & dosage , Positron-Emission Tomography , Rats , Rats, Nude , Squamous Cell Carcinoma of Head and Neck , Technetium/chemistry , Tomography, Emission-Computed, Single-Photon
11.
Int J Nanomedicine ; 7: 2227-38, 2012.
Article in English | MEDLINE | ID: mdl-22619558

ABSTRACT

BACKGROUND: Gold nanoshells are excellent agents for photothermal ablation cancer therapy and are currently under clinical trial for solid tumors. Previous studies showed that passive delivery of gold nanoshells through intravenous administration resulted in limited tumor accumulation, which represents a major challenge for this therapy. In this report, the impact of direct intratumoral administration on the pharmacokinetics and biodistribution of the nanoshells was systematically investigated. METHODS: The gold nanoshells were labeled with the radionuclide, copper-64 ((64)Cu). Intratumoral infusion of (64)Cu-nanoshells and two controls, ie, (64)Cu-DOTA (1,4,7,10-tetraazaciclododecane- 1,4,7,10-tetraacetic acid) and (64)Cu-DOTA-PEG (polyethylene glycol), as well as intravenous injection of (64)Cu-nanoshells were performed in nude rats, each with a head and neck squamous cell carcinoma xenograft. The pharmacokinetics was determined by radioactive counting of serial blood samples collected from the rats at different time points post-injection. Using positron emission tomography/computed tomography imaging, the in vivo distribution of (64)Cu-nanoshells and the controls was monitored at various time points after injection. Organ biodistribution in the rats at 46 hours was analyzed by radioactive counting and compared between the different groups. RESULTS: The resulting pharmacokinetic curves indicated a similar trend between the intratumorally injected agents, but a significant difference with the intravenously injected (64)Cu-nanoshells. Positron emission tomography images and organ biodistribution results on rats after intratumoral administration showed higher retention of (64)Cu-nanoshells in tumors and less concentration in other healthy organs, with a significant difference from the controls. It was also found that, compared with intravenous injection, tumor concentrations of (64)Cu-nanoshells improved substantially and were stable at 44 hours post-injection. CONCLUSION: There was a higher intratumoral retention of (64)Cu-nanoshells and a lower concentration in other healthy tissues, suggesting that intratumoral administration is a potentially better approach for nanoshell-based photothermal therapy.


Subject(s)
Copper Radioisotopes/administration & dosage , Nanoshells/administration & dosage , Animals , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Copper Radioisotopes/pharmacokinetics , Feasibility Studies , Gold , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/radiotherapy , Humans , Injections, Intralesional , Male , Multimodal Imaging , Nanomedicine , Positron-Emission Tomography , Rats , Rats, Nude , Tissue Distribution , Tomography, X-Ray Computed , Xenograft Model Antitumor Assays
12.
J Biol Inorg Chem ; 17(5): 709-18, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22418921

ABSTRACT

Epidermal growth factor receptor 1 (EGFR) is an attractive target for radionuclide therapy of head and neck carcinomas. Affibody molecules against EGFR (Z(EGFR)) show excellent tumor localizations in imaging studies. However, one major drawback is that radiometal-labeled Affibody molecules display extremely high uptakes in the radiosensitive kidneys which may impact their use as radiotherapeutic agents. The purpose of this study is to further explore whether radiometal-labeled human serum albumin (HSA)-Z(EFGR) bioconjugates display desirable profiles for the use in radionuclide therapy of EGFR-positive head and neck carcinomas. The Z(EFGR) analog, Ac-Cys-Z(EGFR:1907), was site-specifically conjugated with HSA. The resulting bioconjugate 1,4,7,10-tetraazacyclododecane-1,4,7-triacetic acid (DO3A)-HSA-Z(EGFR:1907) was then radiolabeled with either (64)Cu or (177)Lu and subjected to in vitro cell uptake and internalization studies using the human oral squamous carcinoma cell line SAS. Positron emission tomography (PET), single photon emission computed tomography (SPECT), and biodistribution studies were conducted using SAS-tumor-bearing mice. Cell studies revealed a high (8.43 ± 0.55 % at 4 h) and specific (0.95 ± 0.09 % at 4 h) uptake of (177)Lu-DO3A-HSA-Z(EGFR:1907) as determined by blocking with nonradioactive Z(EGFR:1907). The internalization of (177)Lu-DO3A-HSA-Z(EGFR:1907) was verified in vitro and found to be significantly higher than that of (177)Lu-labeled Z(EFGR) at 2-24 h of incubation. PET and SPECT studies showed good tumor imaging contrasts. The biodistribution of (177)Lu-DO3A-HSA-Z(EGFR:1907) in SAS-tumor-bearing mice displayed high tumor uptake (5.1 ± 0.44 % ID/g) and liver uptake (31.5 ± 7.66 % ID/g) and moderate kidney uptake (8.5 ± 1.08 % ID/g) at 72 h after injection. (177)Lu-DO3A-HSA-Z(EGFR:1907) shows promising in vivo profiles and may be a potential radiopharmaceutical for radionuclide therapy of EGFR-expressing head and neck carcinomas.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , ErbB Receptors/metabolism , Head and Neck Neoplasms/diagnostic imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Amino Acid Sequence , Animals , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Copper/chemistry , Copper/pharmacokinetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/radiotherapy , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Lutetium/chemistry , Lutetium/pharmacokinetics , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Positron-Emission Tomography , Serum Albumin/chemistry , Serum Albumin/pharmacokinetics , Squamous Cell Carcinoma of Head and Neck , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
13.
Neuro Oncol ; 14(4): 416-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22427110

ABSTRACT

Although external beam radiation is an essential component to the current standard treatment of primary brain tumors, its application is limited by toxicity at doses more than 80 Gy. Recent studies have suggested that brachytherapy with liposomally encapsulated radionuclides may be of benefit, and we have reported methods to markedly increase the specific activity of rhenium-186 ((186)Re)-liposomes. To better characterize the potential delivery, toxicity, and efficacy of the highly specific activity of (186)Re-liposomes, we evaluated their intracranial application by convection-enhanced delivery in an orthotopic U87 glioma rat model. After establishing an optimal volume of 25 µL, we observed focal activity confined to the site of injection over a 96-hour period. Doses of up to 1850 Gy were administered without overt clinical or microscopic evidence of toxicity. Animals treated with (186)Re-liposomes had a median survival of 126 days (95% confidence interval [CI], 78.4-173 days), compared with 49 days (95% CI, 44-53 days) for controls. Log-rank analysis between these 2 groups was highly significant (P = .0013) and was even higher when 100 Gy was used as a cutoff (P < .0001). Noninvasive luciferase imaging as a surrogate for tumor volume showed a statistically significant separation in bioluminescence by 11 days after 100 Gy or less treatment between the experimental group and the control animals (χ(2)[1, N= 19] = 4.8; P = .029). MRI also supported this difference in tumor size. Duplication of tumor volume differences and survival benefit was possible in a more invasive U251 orthotopic model, with clear separation in bioluminescence at 6 days after treatment (χ(2)[1, N= 9] = 4.7; P = .029); median survival in treated animals was not reached at 120 days because lack of mortality, and log-rank analysis of survival was highly significant (P = .0057). Analysis of tumors by histology revealed minimal areas of necrosis and gliosis. These results support the potential efficacy of the highly specific activity of brachytherapy by (186)Re-liposomes convection-enhanced delivery in glioma.


Subject(s)
Brachytherapy/methods , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Glioma/radiotherapy , Radioisotopes/therapeutic use , Rhenium/therapeutic use , Animals , Brain Neoplasms/pathology , Convection , Glioblastoma/pathology , Glioma/pathology , Liposomes , Nanoparticles/therapeutic use , Rats , Tumor Burden , Xenograft Model Antitumor Assays
14.
Radiology ; 261(3): 813-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22025735

ABSTRACT

PURPOSE: To determine the therapeutic efficacy of rhenium 186 ((186)Re)-labeled PEGylated liposomal doxorubicin ((186)Re-liposomal doxorubicin) in combination with radiofrequency (RF) ablation of human head and neck squamous cell carcinoma (HNSCC) xenograft in nude rats. MATERIALS AND METHODS: This investigation was approved by the animal care committee. Sixty nude rats with subcutaneously implanted HNSCC xenografts (six per group) were treated with (a) RF ablation (70 °C for 5 minutes), (b) PEGylated liposomes, (c) liposomal doxorubicin, (d) (186)Re-PEGylated liposomes (1295 MBq/kg), (e) (186)Re-liposomal doxorubicin (555 MBq/kg), (f) PEGylated liposomes plus RF ablation, (g) liposomal doxorubicin plus RF ablation, (h) (186)Re-PEGylated liposomes plus RF ablation, or (i) (186)Re-liposomal doxorubicin plus RF ablation. Six rats did not receive any treatment (control group). Tumor uptake in (186)Re therapy groups was monitored with small-animal single photon emission computed tomography for 5 days. Therapeutic efficacy was monitored for 6 weeks with measurement of tumor volume, calculation of the percentage injected dose of fluorine 18 fluorodeoxyglucose (FDG) in tumor from small-animal positron emission tomography (PET) images, and determination of viable tumor volume at histopathologic examination. Significant differences between groups were determined with analysis of variance. RESULTS: The average tumor volume (± standard deviation) on the day of therapy was 1.32 cm(3) ± 0.17. At 6 weeks after therapy, control of tumor growth was better with (186)Re-liposomal doxorubicin than with liposomal doxorubicin alone (tumor volume, 2.26 cm(3) ± 0.89 vs 5.43 cm(3) ± 0.93, respectively; P < .01). The use of RF ablation with liposomal doxorubicin and (186)Re-liposomal doxorubicin further improved tumor control (tumor volume, 2.05 cm(3) ± 1.36 and 1.49 cm(3) ± 1.47, respectively). The tumor growth trend correlated with change in percentage of injected dose of FDG in tumor for all groups (R(2) = 0.85, P < .001). Viable tumor volume was significantly decreased in the group treated with (186)Re-liposomal doxorubicin plus RF ablation (0.54 cm(3) ± 0.38; P < .001 vs all groups except (186)Re-liposomal doxorubicin alone). CONCLUSION: Triple and dual therapies had an observable trend ((186)Re-liposomal doxorubicin plus RF ablation > (186)Re-liposomal doxorubicin > liposomal doxorubicin plus RF ablation > liposomal doxorubicin) of improved tumor growth control and decreased viable tumor compared with other therapies. FDG PET could be used as a noninvasive surrogate marker for tumor growth and viability in this tumor model.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/therapy , Doxorubicin/pharmacokinetics , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/therapy , Radiopharmaceuticals/pharmacokinetics , Rhenium/pharmacokinetics , Analysis of Variance , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Catheter Ablation , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease Models, Animal , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Isotope Labeling , Liposomes , Nuclear Medicine/methods , Radionuclide Imaging , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacology , Random Allocation , Rats , Rats, Nude , Rhenium/administration & dosage , Rhenium/pharmacology , Transplantation, Heterologous
15.
Phys Med Biol ; 56(17): 5721-34, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21841210

ABSTRACT

Crucial to all cancer therapy modalities is a strong correlation between treatment and effect. Predictability of therapy success/failure allows for the optimization of treatment protocol and aids in the decision of whether additional treatment is necessary to prevent tumour progression. This work evaluated the relationship between cancer treatment and effect for intratumoural infusions of liposome-encapsulated ¹86Re to head and neck squamous cell carcinoma xenografts of nude rats. Absorbed dose calculations using a dose-point kernel convolution technique showed significant intratumoural dose heterogeneity due to the short range of the beta-particle emissions. The use of three separate tumour infusion locations improved dose homogeneity compared to a single infusion location as a result of a more uniform radioactivity distribution. An improved dose-response correlation was obtained when using effective uniform dose (EUD) calculations based on a generic set of radiobiological parameters (R² = 0.84) than when using average tumour absorbed dose (R² = 0.22). Varying radiobiological parameter values over ranges commonly used for all types of tumours showed little effect on EUD calculations, which suggests that individualized parameter use is of little significance as long as the intratumoural dose heterogeneity is taken into consideration in the dose-response relationship. The improved predictability achieved when using EUD calculations for this cancer therapy modality may be useful for treatment planning and evaluation.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/radiotherapy , Radiotherapy Planning, Computer-Assisted/methods , Rhenium/administration & dosage , Algorithms , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Head and Neck Neoplasms/metabolism , Humans , Infusions, Intralesional , Liposomes , Radioisotopes/administration & dosage , Radiotherapy Dosage , Rats , Rats, Nude , Transplantation, Heterologous , Treatment Outcome
16.
Cancer Biother Radiopharm ; 26(5): 603-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21834653

ABSTRACT

This study was performed to determine the maximum tolerated dose (MTD) and therapeutic effects of rhenium-186 ((186)Re)-labeled liposomal doxorubicin (Doxil), investigate associated toxicities, and calculate radiation absorbed dose in head and neck tumor xenografts and normal organs. Doxil and control polyethylene glycol (PEG)-liposomes were labeled using (186)Re-N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA) method. Tumor-bearing rats received either no therapy (n=6), intravenous Doxil (n=4), or escalating radioactivity of (186)Re-Doxil (185-925 MBq/kg) or (186)Re-PEG-liposomes (1110-1665 MBq/kg) and were monitored for 28 days. Based on body weight loss and systemic toxicity, MTD for (186)Re-Doxil and (186)Re-PEG-liposomes were established at injected radioactivity/body weight of 740 and 1480 MBq/kg, respectively. (186)Re-injected radioactivity/body weight for therapy studies was determined to be 555 MBq/kg for (186)Re-Doxil and 1295 MBq/kg for (186)Re-PEG-liposomes. All groups recovered from their body weight loss, leucopenia, and thrombocytopenia by 28 days postinjection. Normalized radiation absorbed dose to tumor was significantly higher for (186)Re-Doxil (0.299±0.109 Gy/MBq) compared with (186)Re-PEG-liposomes (0.096±0.120 Gy/MBq) (p<0.05). In a separate therapy study, tumor volumes were significantly smaller for (186)Re-Doxil (555 MBq/kg) compared with (186)Re-PEG-liposomes (1295 MBq/kg) (p<0.01) at 42 days postinjection. In conclusion, combination chemoradionuclide therapy with (186)Re-Doxil has promising potential, because good tumor control was achieved with limited associated toxicity.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Doxorubicin/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Radioisotopes/pharmacology , Radiopharmaceuticals/pharmacology , Rhenium/pharmacology , Animals , Combined Modality Therapy , Disease Models, Animal , Doxorubicin/toxicity , Humans , Male , Radioisotopes/toxicity , Radiometry , Radiopharmaceuticals/toxicity , Rats , Rats, Nude , Rhenium/toxicity , Xenograft Model Antitumor Assays
17.
Med Phys ; 38(3): 1339-47, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21520844

ABSTRACT

PURPOSE: Focused radiation therapy by direct intratumoral infusion of lipid nanoparticle (liposome)-carried beta-emitting radionuclides has shown promising results in animal model studies; however, little is known about the impact the intratumoral liposomal radionuclide distribution may have on tumor control. The primary objective of this work was to investigate the effects the intratumoral absorbed dose distributions from this cancer therapy modality have on tumor control and treatment planning by combining dosimetric and radiobiological modeling with in vivo imaging data. METHODS: 99mTc-encapsulated liposomes were intratumorally infused with a single injection location to human head and neck squamous cell carcinoma xenografts in nude rats. High resolution in vivo planar imaging was performed at various time points for quantifying intratumoral retention following infusion. The intratumoral liposomal radioactivity distribution was obtained from 1 mm resolution pinhole collimator SPECT imaging coregistered with CT imaging of excised tumors at 20 h postinfusion. Coregistered images were used for intratumoral dosimetric and radiobiological modeling at a voxel level following extrapolation to the therapeutic analogs, 186Re/ 18Re liposomes. Effective uniform dose (EUD) and tumor control probability (TCP) were used to assess therapy effectiveness and possible methods of improving upon tumor control with this radiation therapy modality. RESULTS: Dosimetric analysis showed that average tumor absorbed doses of 8.6 Gy/MBq (318.2 Gy/mCi) and 5.7 Gy/MBq (209.1 Gy/mCi) could be delivered with this protocol of radiation delivery for 186Re/188Re liposomes, respectively, and 37-92 MBq (1-2.5 mCi)/g tumor administered activity; however, large intratumoral absorbed dose heterogeneity, as seen in dose-volume histograms, resulted in insignificant values of EUD and TCP for achieving tumor control. It is indicated that the use of liposomes encapsulating radionuclides with higher energy beta emissions, dose escalation through increased specific activity, and increasing the number of direct tumor infusion sites improve tumor control. For larger tumors, the use of multiple infusion locations was modeled to be much more efficient, in terms of activity usage, at improving EUD and TCP to achieve a tumoricidal effect. CONCLUSIONS: Direct intratumoral infusion of beta-emitting radionuclide encapsulated liposomes shows promise for cancer therapy by achieving large focally delivered tumor doses. However, the results of this work also indicate that average tumor dose may underestimate tumoricidal effect due to substantial heterogeneity in intratumoral liposomal radionuclide distributions. The resulting intratumoral distribution of liposomes following infusion should be taken into account in treatment planning and evaluation in a clinical setting for an optimal cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/radiotherapy , Liposomes/administration & dosage , Radiation Dosage , Radioisotopes/therapeutic use , Rhenium/therapeutic use , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Male , Models, Biological , Radiometry , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Rats , Treatment Outcome
18.
Langmuir ; 27(8): 4447-55, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21413760

ABSTRACT

A novel asparagine-derived lipid analogue (ALA(11,17)) bearing a tetrahydropyrimidinone headgroup and two fatty chains (11 and 17 indicate the lengths of linear alkyl groups) was synthesized in high yield and purity. The thin film hydration of formulations containing 5 mol % or greater ALA(11,17) in distearoylphosphatidylcholine (DSPC) generated multilamellar vesicles (MLVs) that remained unaggregated according to optical microscopy, while those formed from DSPC only were highly clustered. The MLVs were processed into unilamellar liposomes via extrusion and were characterized by dynamic light scattering (DLS), zeta potential, turbidity, and scanning electron microscopy (SEM) analysis. Results show that the presence of ALA(11,17) in DSPC liposomes significantly alters the morphology, colloidal stability, and retention of encapsulated materials in both acidic and neutral conditions. The ability of ALA(11,17)-hybrid liposomes to encapsulate and retain inclusions under neutral and acidic conditions (pH < 2) was demonstrated by calcein dequenching experiments. DLS and SEM confirmed that ALA(11,17)/DSPC liposomes remained intact under these conditions. The bilayer integrity observed under neutral and acidic conditions and the likely biocompatibility of these fatty amino acid analogues suggest that ALA(11,17) is a promising additive for modulating phosphatidylcholine lipid bilayer properties.


Subject(s)
Acids/pharmacology , Asparagine/chemistry , Lipid Bilayers/chemistry , Lipids/chemistry , Phosphatidylcholines/chemistry , Biocompatible Materials , Capsules , Liposomes
19.
Int J Nanomedicine ; 6: 259-69, 2011.
Article in English | MEDLINE | ID: mdl-21423588

ABSTRACT

PURPOSE: Gold nanoshells (NSs) have already shown great promise as photothermal actuators for cancer therapy. Integrin αvß3 is a marker that is specifically and preferentially overexpressed on multiple tumor types and on angiogenic tumor neovasculature. Active targeting of NSs to integrin αvß3 offers the potential to increase accumulation preferentially in tumors and thereby enhance therapy efficacy. METHODS: Enzyme-linked immunosorbent assay (ELISA) and cell binding assay were used to study the in vitro binding affinities of the targeted nanoconjugate NS-RGDfK. In vivo biodistribution and tumor specificity were analyzed using 64Cu-radiolabeled untargeted and targeted NSs in live nude rats bearing head and neck squamous cell carcinoma (HNSCC) xenografts. The potential thermal therapy applications of NS-RGDfK were evaluated by subablative thermal therapy of tumor xenografts using untargeted and targeted NSs. RESULTS: ELISA and cell binding assay confirmed the binding affinity of NS-RGDfK to integrin αvß3. Positron emission tomography/computed tomography imaging suggested that tumor targeting is improved by conjugation of NSs to cyclo(RGDfK) and peaks at ~20 hours postinjection. In the subablative thermal therapy study, greater biological effectiveness of targeted NSs was implied by the greater degree of tumor necrosis. CONCLUSION: The results presented in this paper set the stage for the advancement of integrin αvß3-targeted NSs as therapeutic nanoconstructs for effective cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/radiotherapy , Integrin alphaVbeta3 , Nanoconjugates/chemistry , Peptides, Cyclic/pharmacology , Animals , Carcinoma, Squamous Cell/blood supply , Cell Line, Tumor , Copper Radioisotopes , Enzyme-Linked Immunosorbent Assay , Gold/chemistry , Hot Temperature , Humans , Hyperthermia, Induced/methods , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Laser Therapy , Mice , Mice, Nude , Models, Animal , Nanoshells/chemistry , Protein Binding , Rats , Rats, Nude , Tissue Distribution , Tomography, Emission-Computed/methods , Transplantation, Heterologous
20.
Phys Med Biol ; 56(3): 703-19, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21299006

ABSTRACT

Post-operative radiotherapy has commonly been used for early stage breast cancer to treat residual disease. The primary objective of this work was to characterize, through dosimetric and radiobiological modeling, a novel focal brachytherapy technique which uses direct intracavitary infusion of ß-emitting radionuclides (186Re/188Re) carried by lipid nanoparticles (liposomes). Absorbed dose calculations were performed for a spherical lumpectomy cavity with a uniformly injected activity distribution using a dose point kernel convolution technique. Radiobiological indices were used to relate predicted therapy outcome and normal tissue complication of this technique with equivalent external beam radiotherapy treatment regimens. Modeled stromal damage was used as a measure of the inhibition of the stimulatory effect on tumor growth driven by the wound healing response. A sample treatment plan delivering 50 Gy at a therapeutic range of 2.0 mm for 186Re-liposomes and 5.0 mm for 188Re-liposomes takes advantage of the dose delivery characteristics of the ß-emissions, providing significant EUD (58.2 Gy and 72.5 Gy for 186Re and 188Re, respectively) with a minimal NTCP (0.046%) of the healthy ipsilateral breast. Modeling of kidney BED and ipsilateral breast NTCP showed that large injected activity concentrations of both radionuclides could be safely administered without significant complications.


Subject(s)
Brachytherapy/methods , Liposomes/chemistry , Mastectomy, Segmental , Radiobiology/methods , Radioisotopes/therapeutic use , Rhenium/therapeutic use , Animals , Brachytherapy/adverse effects , Breast/radiation effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Transformation, Neoplastic , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans , Kidney/radiation effects , Organs at Risk/radiation effects , Probability , Radioisotopes/adverse effects , Radiometry , Rats , Rhenium/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL