Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Nanoscale Res Lett ; 16(1): 9, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33411055

ABSTRACT

The greatest contributors to cancer mortality are metastasis and the consequences of its treatment. Here, we present a novel treatment of metastatic breast cancer that combines photothermal therapy with targeted single-walled carbon nanotubes (SWCNTs) and immunostimulation with a checkpoint inhibitor. We find that the selective near-infrared photothermal ablation of primary orthotopic EMT6 breast tumors in syngeneic BALB/cJ mice using an annexin A5 (ANXA5) functionalized SWCNT bioconjugate synergistically enhances an anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4)-dependent abscopal response, resulting in an increased survival (55%) at 100 days after tumor inoculation. In comparison, there was no survival at 100 days for either photothermal therapy by itself or immunostimulation by itself. Prior to photothermal therapy, the SWCNT-ANXA5 bioconjugate was administered systemically at a relatively low dose of 1.2 mg/kg, where it then accumulated in tumor vasculature via ANXA5-dependent binding. During photothermal therapy, the average maximum temperature in the tumor reached 54 °C (duration 175 s). The mechanism of prolonged survival resulting from combinatorial photothermal ablation and immune stimulation was evaluated by flow cytometric quantification of splenic antitumoral immune effector cells and serum cytokine quantification.

2.
Magn Reson Imaging ; 76: 52-60, 2021 02.
Article in English | MEDLINE | ID: mdl-33220448

ABSTRACT

PURPOSE: Hypoxia measurements can provide crucial information regarding tumor aggressiveness, however current preclinical approaches are limited. Blood oxygen level dependent (BOLD) Magnetic Resonance Imaging (MRI) has the potential to continuously monitor tumor pathophysiology (including hypoxia). The aim of this preliminary work was to develop and evaluate BOLD MRI followed by post-image analysis to identify regions of hypoxia in a murine glioblastoma (GBM) model. METHODS: A murine orthotopic GBM model (GL261-luc2) was used and independent images were generated from multiple slices in four different mice. Image slices were randomized and split into training and validation cohorts. A 7 T MRI was used to acquire anatomical images using a fast-spin-echo (FSE) T2-weighted sequence. BOLD images were taken with a T2*-weighted gradient echo (GRE) and an oxygen challenge. Thirteen images were evaluated in a training cohort to develop the MRI sequence and optimize post-image analysis. An in-house MATLAB code was used to evaluate MR images and generate hypoxia maps for a range of thresholding and ΔT2* values, which were compared against respective pimonidazole sections to optimize image processing parameters. The remaining (n = 6) images were used as a validation group. Following imaging, mice were injected with pimonidazole and collected for immunohistochemistry (IHC). A test of correlation (Pearson's coefficient) and agreement (Bland-Altman plot) were conducted to evaluate the respective MRI slices and pimonidazole IHC sections. RESULTS: For the training cohort, the optimized parameters of "thresholding" (20 ≤ T2* ≤ 35 ms) and ΔT2* (±4 ms) yielded a Pearson's correlation of 0.697. These parameters were applied to the validation cohort confirming a strong Pearson's correlation (0.749) when comparing the respective analyzed MR and pimonidazole images. CONCLUSION: Our preliminary study supports the hypothesis that BOLD MRI is correlated with pimonidazole measurements of hypoxia in an orthotopic GBM mouse model. This technique has further potential to monitor hypoxia during tumor development and therapy.


Subject(s)
Glioblastoma/pathology , Magnetic Resonance Imaging , Oxygen/blood , Tumor Hypoxia , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Glioblastoma/blood , Humans , Image Processing, Computer-Assisted , Male , Mice
3.
PLoS One ; 15(7): e0236245, 2020.
Article in English | MEDLINE | ID: mdl-32706818

ABSTRACT

We have previously demonstrated that endothelial targeting of gold nanoparticles followed by external beam irradiation can cause specific tumor vascular disruption in mouse models of cancer. The induced vascular damage may lead to changes in tumor physiology, including tumor hypoxia, thereby compromising future therapeutic interventions. In this study, we investigate the dynamic changes in tumor hypoxia mediated by targeted gold nanoparticles and clinical radiation therapy (RT). By using noninvasive whole-body fluorescence imaging, tumor hypoxia was measured at baseline, on day 2 and day 13, post-tumor vascular disruption. A 2.5-fold increase (P<0.05) in tumor hypoxia was measured two days after combined therapy, resolving by day 13. In addition, the combination of vascular-targeted gold nanoparticles and radiation therapy resulted in a significant (P<0.05) suppression of tumor growth. This is the first study to demonstrate the tumor hypoxic physiological response and recovery after delivery of vascular-targeted gold nanoparticles followed by clinical radiation therapy in a human non-small cell lung cancer athymic Foxn1nu mouse model.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Metal Nanoparticles/therapeutic use , Tumor Hypoxia , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Gold/therapeutic use , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Optical Imaging/methods , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Xenograft Model Antitumor Assays
4.
Phys Med Biol ; 65(21): 21RM02, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32380492

ABSTRACT

This roadmap outlines the potential roles of metallic nanoparticles (MNPs) in the field of radiation therapy. MNPs made up of a wide range of materials (from Titanium, Z = 22, to Bismuth, Z = 83) and a similarly wide spectrum of potential clinical applications, including diagnostic, therapeutic (radiation dose enhancers, hyperthermia inducers, drug delivery vehicles, vaccine adjuvants, photosensitizers, enhancers of immunotherapy) and theranostic (combining both diagnostic and therapeutic), are being fabricated and evaluated. This roadmap covers contributions from experts in these topics summarizing their view of the current status and challenges, as well as expected advancements in technology to address these challenges.


Subject(s)
Metal Nanoparticles/therapeutic use , Theranostic Nanomedicine/methods , Humans , Hyperthermia, Induced
5.
Ultrasound Med Biol ; 46(2): 369-376, 2020 02.
Article in English | MEDLINE | ID: mdl-31694771

ABSTRACT

A cost-effective method for serial in vivo imaging of tumor microvasculature has been developed. We evaluated acoustic angiography (AA) for visualizing and assessing non-small cell lung tumor (A549) microvasculature in mice before and after tumor vascular disruption by vascular-targeted gold nanoparticles and radiotherapy. Standard B-mode and microbubble-enhanced AA images were acquired at pre- and post-treatment time points. Using these modes, a new metric, 50% vessel penetration depth, was developed to characterize the 3-D spatial heterogeneity of microvascular networks. We observed an increase in tumor perfusion after radiation-induced vascular disruption, relative to control animals. This was also visualized in vessel morphology mode, which revealed a loss in vessel integrity. We found that tumors with poorly perfused vasculature at day 0 exhibited a reduced growth rate over time. This suggested a new method to reduce in-group treatment response variability using pre-treatment microvessel maps to objectively identify animals for study removal.


Subject(s)
Angiography/methods , Contrast Media , Imaging, Three-Dimensional , Metal Nanoparticles , Microbubbles , Microvessels/diagnostic imaging , Neoplasms/blood supply , Neoplasms/diagnostic imaging , Animals , Female , Image Enhancement , Mice , Ultrasonography/methods
6.
Cancer Lett ; 425: 174-182, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29574275

ABSTRACT

Approximately 75% of ovarian cancer is diagnosed once metastasis to the peritoneal cavity has occurred. A large proportion of patients eventually develop platinum-resistive tumors, which are considered terminal. In order to provide an alternative a novel fusion protein, mCTH-ANXA5, has been developed for the treatment of recurrent, metastatic ovarian cancer. The fusion protein combines annexin V (ANXA5), an ovarian tumor and tumor vasculature targeting protein, with mutated cystathionine gamma-lyase (mCTH), an enzyme that converts selenomethionine (SeMet) into toxic methylselenol, which generates reactive oxygen species and eventual tumor cell death. In order to further enhance the therapeutic efficacy, anti-CD73 and anti-OX40 immunostimulants were combined with mCTH-ANXA5, resulting in an increase of survival by 100% from 12 to 24 days post-therapy and decrease tumor burden in mice with orthotopic metastatic ovarian cancer. Further evaluation of the combination therapy revealed a strong antibody-mediated immune response, and an increased infiltration of cytotoxic T-cells along with a decrease in tumor promoting immune cells. This study demonstrates the efficacy of a synergistic, multi-drug system by attacking the tumor as well as enlisting the body's own defense system to treat the patient.


Subject(s)
Antibodies/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Prodrugs/administration & dosage , Recombinant Fusion Proteins/administration & dosage , 5'-Nucleotidase/antagonists & inhibitors , Animals , Annexin A5/genetics , Annexin A5/metabolism , Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Drug Synergism , Female , Humans , Immunotherapy , Mice , Neoplasm Metastasis , Neoplasm Recurrence, Local/immunology , OX40 Ligand/antagonists & inhibitors , Ovarian Neoplasms/immunology , Prodrugs/pharmacology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
7.
Nanotechnology ; 29(3): 035101, 2018 Jan 19.
Article in English | MEDLINE | ID: mdl-29160225

ABSTRACT

Bladder cancer has a 60%-70% recurrence rate most likely due to any residual tumour left behind after a transurethral resection (TUR). Failure to completely resect the cancer can lead to recurrence and progression into higher grade tumours with metastatic potential. We present here a novel therapy to treat superficial tumours with the potential to decrease recurrence. The therapy is a heat-based approach in which bladder tumour specific single-walled carbon nanotubes (SWCNTs) are delivered intravesically at a very low dose (0.1 mg SWCNT per kg body weight) followed 24 h later by a short 30 s treatment with a 360° near-infrared light that heats only the bound nanotubes. The energy density of the treatment was 50 J cm-2, and the power density that this treatment corresponds to is 1.7 W cm-2, which is relatively low. Nanotubes are specifically targeted to the tumour via the interaction of annexin V (AV) and phosphatidylserine, which is normally internalised on healthy tissue but externalised on tumours and the tumour vasculature. SWCNTs are conjugated to AV, which binds specifically to bladder cancer cells as confirmed in vitro and in vivo. Due to this specific localisation, NIR light can be used to heat the tumour while conserving the healthy bladder wall. In a short-term efficacy study in mice with orthotopic MB49 murine bladder tumours treated with the SWCNT-AV conjugate and NIR light, no tumours were visible on the bladder wall 24 h after NIR light treatment, and there was no damage to the bladder. In a separate survival study in mice with the same type of orthotopic tumours, there was a 50% cure rate at 116 days when the study was ended. At 116 days, no treatment toxicity was observed, and no nanotubes were detected in the clearance organs or bladder.


Subject(s)
Hyperthermia, Induced , Nanotubes, Carbon/chemistry , Phosphatidylserines/chemistry , Phototherapy , Urinary Bladder Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Humans , Lasers , Mice, Inbred C57BL , Tissue Distribution , Treatment Outcome , Urinary Bladder Neoplasms/diagnostic imaging
8.
Mol Cancer Ther ; 16(9): 1855-1865, 2017 09.
Article in English | MEDLINE | ID: mdl-28522586

ABSTRACT

Mutant cystathionine gamma-lyase was targeted to phosphatidylserine exposed on tumor vasculature through fusion with Annexin A1 or Annexin A5. Cystathionine gamma-lyase E58N, R118L, and E338N mutations impart nonnative methionine gamma-lyase activity, resulting in tumor-localized generation of highly toxic methylselenol upon systemic administration of nontoxic selenomethionine. The described therapeutic system circumvents systemic toxicity issues using a novel drug delivery/generation approach and avoids the administration of nonnative proteins and/or DNA required with other enzyme prodrug systems. The enzyme fusion exhibits strong and stable in vitro binding with dissociation constants in the nanomolar range for both human and mouse breast cancer cells and in a cell model of tumor vascular endothelium. Daily administration of the therapy suppressed growth of highly aggressive triple-negative murine 4T1 mammary tumors in immunocompetent BALB/cJ mice and MDA-MB-231 tumors in SCID mice. Treatment did not result in the occurrence of negative side effects or the elicitation of neutralizing antibodies. On the basis of the vasculature-targeted nature of the therapy, combinations with rapamycin and cyclophosphamide were evaluated. Rapamycin, an mTOR inhibitor, reduces the prosurvival signaling of cells in a hypoxic environment potentially exacerbated by a vasculature-targeted therapy. IHC revealed, unsurprisingly, a significant hypoxic response (increase in hypoxia-inducible factor 1 α subunit, HIF1A) in the enzyme prodrug-treated tumors and a dramatic reduction of HIF1A upon rapamycin treatment. Cyclophosphamide, an immunomodulator at low doses, was combined with the enzyme prodrug therapy and rapamycin; this combination synergistically reduced tumor volumes, inhibited metastatic progression, and enhanced survival. Mol Cancer Ther; 16(9); 1855-65. ©2017 AACR.


Subject(s)
Cyclophosphamide/pharmacology , Neoplasms/enzymology , Neoplasms/pathology , Neovascularization, Pathologic/enzymology , Prodrugs/pharmacology , Sirolimus/pharmacology , Animals , Annexin A5/genetics , Carbon-Sulfur Lyases/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neovascularization, Pathologic/drug therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Protein Eng Des Sel ; 30(2): 85-94, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27986920

ABSTRACT

Enzyme prodrug therapy has the potential to remedy the lack of selectivity associated with the systemic administration of chemotherapy. However, most current systems are immunogenic and constrained to a monotherapeutic approach. We developed a new class of fusion proteins centered about the human enzyme ß-glucuronidase (ßG), capable of converting several innocuous prodrugs into chemotherapeutics. We targeted ßG to phosphatidylserine on tumor cells, tumor vasculature and metastases via annexin A1/A5. Phosphatidylserine shows promise as a universal marker for solid tumors and allows for tumor type-independent targeting. To create fusion proteins, human annexin A1/A5 was genetically fused to the activity-enhancing 16a3 mutant of human ßG, expressed in chemically defined, fed-batch suspension culture, and chromatographically purified. All fusion constructs achieved >95% purity with yields up to 740 µg/l. Fusion proteins displayed cancer selective cell-surface binding with cell line-dependent binding stability. One fusion protein in combination with the prodrug SN-38 glucuronide was as effective as the drug SN-38 on Panc-1 pancreatic cancer cells and HAAE-1 endothelial cells, and demonstrated efficacy against MCF-7 breast cancer cells. ßG fusion proteins effectively enable localized combination therapy that can be tailored to each patient via prodrug selection, with promising clinical potential based on their near fully human design.


Subject(s)
Annexin A1/genetics , Annexin A5/genetics , Glucuronidase/metabolism , Recombinant Fusion Proteins/metabolism , Cell Line, Tumor , Glucuronidase/chemistry , Glucuronidase/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Molecular Targeted Therapy , Mutation , Prodrugs/metabolism , Protein Conformation , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL