Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
BMC Complement Med Ther ; 22(1): 58, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35255889

ABSTRACT

BACKGROUND: Gastric signet ring cell carcinoma (SRCC) is an aggressive gastric adenocarcinoma with a poor prognosis when diagnosed at an advanced stage. As alternative medicine, two natural supplements (ascorbate (AA) and sodium alpha lipoate (LA)) have been shown to inhibit various cancers with mild side effects. METHODS: These two natural supplements and a series of combinations (AA&LA, AA+LA and LA + AA) were incubated with non-SRCC cells (GPM-1), patient-derived gastric origin SRCC (GPM-2), gastric-origin SRCCs (HSC-39 and KATO-3), human pancreatic (MIA PaCa-2) and ovarian (SKOV-3) cells for evaluating their therapeutic effects. Moreover, these treatments were applied in 3D-cultured organoids to reveal the feasibility of these approaches for in vivo study. RESULTS: Analyzing their antioxidant capabilities and dose-response curves, we observed that all four gastric cell lines, including three patient-derived cell lines were sensitive to ascorbate (~ 10 mM). The influence of ascorbate incubation time was studied, with a 16-h incubation found to be optimal for in vitro studies. Moreover, a simultaneous combination of AA and LA (AA&LA) did not significantly inhibit cell proliferation, while prior LA treatment increased the growth inhibition of AA therapy (LA + AA). Anti-cancer efficacy of AA was further confirmed in 3D-cultured SRCC (KATO-3) organoids. CONCLUSIONS: This study highlights the potential of AA and LA + AA in treating gastric origin SRCC, and demonstrates the influence of order in which the drugs are administered.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Complementary Therapies , Stomach Neoplasms , Carcinoma, Signet Ring Cell/drug therapy , Carcinoma, Signet Ring Cell/pathology , Humans , Sodium , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
2.
J Control Release ; 330: 1080-1094, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33189786

ABSTRACT

Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 µg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.


Subject(s)
Hyperthermia, Induced , Neoplasms , Animals , Imidazoles , Immunotherapy , Liposomes , Mice , Neoplasms/drug therapy
3.
J Control Release ; 309: 277-288, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31301340

ABSTRACT

Gemcitabine delivery to pancreatic ductal adenocarcinoma is limited by poor pharmacokinetics, dense fibrosis and hypo-vascularization. Activatable liposomes, with drug release resulting from local heating, enhance serum stability and circulation, and the released drug retains the ability to diffuse within the tumor. A limitation of liposomal gemcitabine has been the low loading efficiency. To address this limitation, we used the superior solubilizing potential of copper (II) gluconate to form a complex with gemcitabine at copper:gemcitabine (1:4). Thermosensitive liposomes composed of DPPC:DSPC:DSPE-PEG2k (80:15:5, mole%) then reached 12 wt% loading, 4-fold greater than previously reported values. Cryo transmission electron microscopy confirmed the presence of a liquid crystalline gemcitabine­copper mixture. The optimized gemcitabine liposomes released 60% and 80% of the gemcitabine within 1 and 5 min, respectively, at 42 °C. Liposomal encapsulation resulted in a circulation half-life of ~2 h in vivo (compared to reported circulation of 16 min for free gemcitabine in mice), and free drug was not detected within the plasma. The resulting gemcitabine liposomes were efficacious against both murine breast cancer and pancreatic cancer in vitro. Three repeated treatments of activatable gemcitabine liposomes plus ultrasound hyperthermia regressed or eliminated tumors in the neu deletion model of murine breast cancer with limited toxicity, enhancing survival when compared to treatment with gemcitabine alone. With 5% of the free gemcitabine dose (5 rather than 100 mg/kg), tumor growth was suppressed to the same degree as gemcitabine. Additionally, in a more aggressive tumor model of murine pancreatic cancer, liposomal gemcitabine combined with local hyperthermia induced cell death and regions of apoptosis and necrosis.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Breast Neoplasms/therapy , Delayed-Action Preparations/chemistry , Deoxycytidine/analogs & derivatives , Liposomes/chemistry , Pancreatic Neoplasms/therapy , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Drug Delivery Systems , Drug Liberation , Female , Humans , Hyperthermia, Induced , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/pathology , Temperature , Gemcitabine
4.
J Control Release ; 303: 42-54, 2019 06 10.
Article in English | MEDLINE | ID: mdl-30978432

ABSTRACT

A successful chemotherapy-immunotherapy solid-tumor protocol should accomplish the following goals: debulk large tumors, release tumor antigen for cross-presentation and cross-priming, release cancer-suppressive cytokines and enhance anti-tumor immune cell populations. Thermally-activated drug delivery particles have the potential to synergize with immunotherapeutics to accomplish these goals; activation can release chemotherapy within bulky solid tumors and can enhance response when combined with immunotherapy. We set out to determine whether a single protocol, combining locally-activated chemotherapy and agonist immunotherapy, could accomplish these goals and yield a potentially translational therapy. For effective delivery of free doxorubicin to tumors with minimal toxicity, we stabilized doxorubicin with copper in temperature-sensitive liposomes that rapidly release free drug in the vasculature of cancer lesions upon exposure to ultrasound-mediated hyperthermia. We found that in vitro exposure of tumor cells to hyperthermia and doxorubicin resulted in immunogenic cell death and the local release of type I interferons across murine cancer cell lines. Following intravenous injection, local activation of the liposomes within a single tumor released doxorubicin and enhanced cross-presentation of a model antigen at distant tumor sites. While a variety of protocols achieved a complete response in >50% of treated mice, the complete response rate was greatest (90%) when 1 week of immunotherapy priming preceded a single activatable chemotherapeutic administration. While repeated chemotherapeutic delivery reduced local viable tumor, the complete response rate and a subset of tumor immune cells were also reduced. Taken together, the results suggest that activatable chemotherapy can enhance adjuvant immunotherapy; however, in a murine model the systemic adaptive immune response was greatest with a single administration of chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Hyperthermia, Induced , Immunotherapy , Mammary Neoplasms, Experimental/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Liposomes , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/administration & dosage
5.
Clin Cancer Res ; 24(20): 4976-4987, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29967252

ABSTRACT

Purpose: Noninvasive and quantitative tracking of CD8+ T cells by PET has emerged as a potential technique to gauge response to immunotherapy. We apply an anti-CD8 cys-diabody, labeled with 64Cu, to assess the sensitivity of PET imaging of normal and diseased tissue.Experimental Design: Radiolabeling of an anti-CD8 cys-diabody (169cDb) with 64Cu was developed. The accumulation of 64Cu-169cDb was evaluated with PET/CT imaging (0, 5, and 24 hours) and biodistribution (24 hours) in wild-type mouse strains (n = 8/group studied with imaging and IHC or flow cytometry) after intravenous administration. Tumor-infiltrating CD8+ T cells in tumor-bearing mice treated with CpG and αPD-1 were quantified and mapped (n = 6-8/group studied with imaging and IHC or flow cytometry).Results: We demonstrate the ability of immunoPET to detect small differences in CD8+ T-cell distribution between mouse strains and across lymphoid tissues, including the intestinal tract of normal mice. In FVB mice bearing a syngeneic HER2-driven model of mammary adenocarcinoma (NDL), 64Cu-169cDb PET imaging accurately visualized and quantified changes in tumor-infiltrating CD8+ T cells in response to immunotherapy. A reduction in the circulation time of the imaging probe followed the development of treatment-related liver and splenic hypertrophy and provided an indication of off-target effects associated with immunotherapy protocols.Conclusions: 64Cu-169cDb imaging can spatially map the distribution of CD8+ T cells in normal organs and tumors. ImmunoPET imaging of tumor-infiltrating cytotoxic CD8+ T cells detected changes in T-cell density resulting from adjuvant and checkpoint immunotherapy protocols in our preclinical evaluation. Clin Cancer Res; 24(20); 4976-87. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal , CD8-Positive T-Lymphocytes/metabolism , Copper Radioisotopes , Lymphocyte Count , Molecular Imaging , Positron-Emission Tomography , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Humans , Immunotherapy , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Molecular Imaging/methods , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/therapy , Positron Emission Tomography Computed Tomography , Xenograft Model Antitumor Assays
6.
J Control Release ; 243: 232-242, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27746275

ABSTRACT

We previously developed a pH-responsive copper-doxorubicin (CuDox) cargo in lysolipid-based temperature-sensitive liposomes (LTSLs). The CuDox complex is released from the particle by elevated temperature; however, full release of doxorubicin from CuDox requires a reduced pH, such as that expected in lysosomes. The primary goal of this study is to evaluate the cellular uptake and intracellular trafficking of the drug-metal complex in comparison with intact liposomes and free drug. We found that the CuDox complex was efficiently internalized by mammary carcinoma cells after release from LTSLs. Intracellular doxorubicin and copper were 6-fold and 5-fold greater, respectively, after a 0.5h incubation with the released CuDox complex, as compared to incubation with intact liposomes containing the complex. Total cellular doxorubicin fluorescence was similar following CuDox and free doxorubicin incubation. Imaging and mass spectrometry assays indicated that the CuDox complex was initially internalized intact but breaks down over time within cells, with intracellular copper decreasing more rapidly than intracellular doxorubicin. Doxorubicin fluorescence was reduced when complexed with copper, and nuclear fluorescence was reduced when cells were incubated with the CuDox complex as compared with free doxorubicin. Therapeutic efficacy, which typically results from intercalation of doxorubicin with DNA, was equivalent for the CuDox complex and free doxorubicin and was superior to that of liposomal doxorubicin formulations. Taken together, the results suggest that quenched CuDox reaches the nucleus and remains efficacious. In order to design protocols for the use of these temperature-sensitive particles in cancer treatment, the timing of hyperthermia relative to drug administration must be examined. When cells were heated to 42°C prior to the addition of free doxorubicin, nuclear drug accumulation increased by 1.8-fold in cancer cells after 5h, and cytotoxicity increased 1.4-fold in both cancer and endothelial cells. Endothelial cytotoxicity was similarly augmented with mild hyperthermia applied prior to treatment with released CuDox. In summary, we find that the drug-metal complex formed in temperature-sensitive particles can be internalized by cancer and endothelial cells resulting in therapeutic efficacy that is similar to free doxorubicin, and this efficacy can be enhanced by elevated temperature.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Copper/chemistry , Doxorubicin/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/metabolism , Doxorubicin/administration & dosage , Doxorubicin/metabolism , Doxorubicin/pharmacology , Endothelial Cells/metabolism , Female , Humans , Hydrogen-Ion Concentration , Hyperthermia, Induced/methods , Mass Spectrometry , Mice , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacology , Temperature , Time Factors
7.
Biomaterials ; 104: 339-51, 2016 10.
Article in English | MEDLINE | ID: mdl-27479049

ABSTRACT

The overall prognosis of bladder cancer has not been improved over the last 30 years and therefore, there is a great medical need to develop novel diagnosis and therapy approaches for bladder cancer. We developed a multifunctional nanoporphyrin platform that was coated with a bladder cancer-specific ligand named PLZ4. PLZ4-nanoporphyrin (PNP) integrates photodynamic diagnosis, image-guided photodynamic therapy, photothermal therapy and targeted chemotherapy in a single procedure. PNPs are spherical, relatively small (around 23 nm), and have the ability to preferably emit fluorescence/heat/reactive oxygen species upon illumination with near infrared light. Doxorubicin (DOX) loaded PNPs possess slower drug release and dramatically longer systemic circulation time compared to free DOX. The fluorescence signal of PNPs efficiently and selectively increased in bladder cancer cells but not normal urothelial cells in vitro and in an orthotopic patient derived bladder cancer xenograft (PDX) models, indicating their great potential for photodynamic diagnosis. Photodynamic therapy with PNPs was significantly more potent than 5-aminolevulinic acid, and eliminated orthotopic PDX bladder cancers after intravesical treatment. Image-guided photodynamic and photothermal therapies synergized with targeted chemotherapy of DOX and significantly prolonged overall survival of mice carrying PDXs. In conclusion, this uniquely engineered targeting PNP selectively targeted tumor cells for photodynamic diagnosis, and served as effective triple-modality (photodynamic/photothermal/chemo) therapeutic agents against bladder cancers. This platform can be easily adapted to individualized medicine in a clinical setting and has tremendous potential to improve the management of bladder cancer in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Microscopy, Fluorescence/methods , Nanoparticles/administration & dosage , Photochemotherapy/methods , Porphyrins/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Combined Modality Therapy/methods , Doxorubicin/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Nanoparticles/chemistry , Peptides, Cyclic/administration & dosage , Photosensitizing Agents/administration & dosage , Phototherapy/methods , Theranostic Nanomedicine/methods , Treatment Outcome
8.
Phys Med Biol ; 61(14): 5275-96, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27353347

ABSTRACT

A 1.5 MHz prolate spheroidal therapeutic array with 128 circular elements was designed to accommodate standard imaging arrays for ultrasonic image-guided hyperthermia. The implementation of this dual-array system integrates real-time therapeutic and imaging functions with a single ultrasound system (Vantage 256, Verasonics). To facilitate applications involving small animal imaging and therapy the array was designed to have a beam depth of field smaller than 3.5 mm and to electronically steer over distances greater than 1 cm in both the axial and lateral directions. In order to achieve the required f number of 0.69, 1-3 piezocomposite modules were mated within the transducer housing. The performance of the prototype array was experimentally evaluated with excellent agreement with numerical simulation. A focal volume (2.70 mm (axial) × 0.65 mm (transverse) × 0.35 mm (transverse)) defined by the -6 dB focal intensity was obtained to address the dimensions needed for small animal therapy. An electronic beam steering range defined by the -3 dB focal peak intensity (17 mm (axial) × 14 mm (transverse) × 12 mm (transverse)) and -8 dB lateral grating lobes (24 mm (axial) × 18 mm (transverse) × 16 mm (transverse)) was achieved. The combined testing of imaging and therapeutic functions confirmed well-controlled local heating generation and imaging in a tissue mimicking phantom. This dual-array implementation offers a practical means to achieve hyperthermia and ablation in small animal models and can be incorporated within protocols for ultrasound-mediated drug delivery.


Subject(s)
Hyperthermia, Induced/instrumentation , Phantoms, Imaging , Therapy, Computer-Assisted/methods , Transducers , Ultrasonography/instrumentation , Animals , Equipment Design , Humans , Hyperthermia, Induced/methods , Image Interpretation, Computer-Assisted , Models, Theoretical , Ultrasonography/methods
9.
J Control Release ; 220(Pt A): 253-264, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26471394

ABSTRACT

Ultrasonic activation of nanoparticles provides the opportunity to deliver a large fraction of the injected dose to insonified tumors and produce a complete local response. Here, we evaluate whether the local and systemic response to chemotherapy can be enhanced by combining such a therapy with locally-administered CpG as an immune adjuvant. In order to create stable, activatable particles, a complex between copper and doxorubicin (CuDox) was created within temperature-sensitive liposomes. Whereas insonation of the CuDox liposomes alone has been shown to produce a complete response in murine breast cancer after 8 treatments of 6 mg/kg delivered over 4 weeks, combining this treatment with CpG resolved local cancers within 3 treatments delivered over 7 days. Further, contralateral tumors regressed as a result of the combined treatment, and survival was extended in systemic disease. In both the treated and contralateral tumor site, the combined treatment increased leukocytes and CD4+ and CD8+ T-effector cells and reduced myeloid-derived suppressor cells (MDSCs). Taken together, the results suggest that this combinatorial treatment significantly enhances the systemic efficacy of locally-activated nanotherapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , DNA/administration & dosage , Doxorubicin/administration & dosage , Immunotherapy/methods , Nanoparticles , Organometallic Compounds/administration & dosage , Ultrasonic Therapy/methods , Adjuvants, Immunologic/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , DNA/chemistry , Doxorubicin/chemistry , Drug Compounding , Female , Liposomes , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Nanotechnology , Organometallic Compounds/chemistry , Technology, Pharmaceutical/methods , Temperature , Time Factors , Tumor Burden/drug effects , Tumor Microenvironment
10.
PLoS One ; 10(8): e0134938, 2015.
Article in English | MEDLINE | ID: mdl-26244783

ABSTRACT

Mild hyperthermia has been successfully employed to induce reversible physiological changes that can directly treat cancer and enhance local drug delivery. In this approach, temperature monitoring is essential to avoid undesirable biological effects that result from thermal damage. For thermal therapies, Magnetic Resonance Imaging (MRI) has been employed to control real-time Focused Ultrasound (FUS) therapies. However, combined ultrasound imaging and therapy systems offer the benefits of simple, low-cost devices that can be broadly applied. To facilitate such technology, ultrasound thermometry has potential to reliably monitor temperature. Control of mild hyperthermia was previously achieved using a proportional-integral-derivative (PID) controller based on thermocouple measurements. Despite accurate temporal control of heating, this method is limited by the single position at which the temperature is measured. Ultrasound thermometry techniques based on exploiting the thermal dependence of acoustic parameters (such as longitudinal velocity) can be extended to create thermal maps and allow an accurate monitoring of temperature with good spatial resolution. However, in vivo applications of this technique have not been fully developed due to the high sensitivity to tissue motion. Here, we propose a motion compensation method based on the acquisition of multiple reference frames prior to treatment. The technique was tested in the presence of 2-D and 3-D physiological-scale motion and was found to provide effective real-time temperature monitoring. PID control of mild hyperthermia in presence of motion was then tested with ultrasound thermometry as feedback and temperature was maintained within 0.3°C of the requested value.


Subject(s)
Hyperthermia, Induced/methods , Magnetic Resonance Imaging/methods , Thermometry/methods , Ultrasonic Therapy/methods , Animals , Humans , Hyperthermia, Induced/instrumentation , Magnetic Resonance Imaging/instrumentation , Motion , Phantoms, Imaging , Reproducibility of Results , Thermometry/instrumentation , Ultrasonic Therapy/instrumentation
11.
Prostaglandins Other Lipid Mediat ; 113-115: 13-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25019221

ABSTRACT

Epidemiological and pre-clinical studies support the anti-tumor effects of ω-3 PUFAs; however, the results from human trials are mixed, making it difficult to provide dietary guidelines or recommendations of ω-3 PUFAs for disease prevention or treatment. Understanding the molecular mechanisms by which ω-3 PUFAs inhibit cancer could lead to better nutritional paradigms and human trials to clarify their health effects. The ω-3 PUFAs exert their biological activities mainly through the formation of bioactive lipid metabolites. Here we discuss the biology of cyclooxygenase, lipoxygenase and cytochrome P450 enzymes-derived ω-3-series lipid metabolites on angiogenesis, inflammation and cancer.


Subject(s)
Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Animals , Cytochromes/metabolism , Humans , Inflammation/enzymology , Lipid Metabolism , Lipoxygenases/metabolism , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism
12.
J Control Release ; 172(1): 266-273, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-23994755

ABSTRACT

The development of treatment protocols that result in a complete response to chemotherapy has been hampered by free drug toxicity and the low bioavailability of nano-formulated drugs. Here, we explore the application of temperature-sensitive liposomes that have been formulated to enhance stability in circulation. We formed a pH-sensitive complex between doxorubicin (Dox) and copper (CuDox) in the core of lysolipid-containing temperature-sensitive liposomes (LTSLs). The complex remains associated at neutral pH but dissociates to free Dox in lower pH environments. The resulting CuDox-LTSLs were injected intravenously into a syngeneic murine breast cancer model (6 mg Dox/kg body weight) and intravascular release of the drug was triggered by ultrasound. The entire tumor was insonified for 5 min prior to drug administration and 20 min post drug injection. A single-dose administration of CuDox-LTSLs combined with insonation suppressed tumor growth. Moreover, after twice per week treatment over a period of 28 days, a complete response was achieved in which the NDL tumor cells and the tumor interstitium could no longer be detected. All mice treated with ultrasound combined with CuDox-LTSLs survived, and tumor was undetectable 8 months post treatment. Iron and copper-laden macrophages were observed at early time points following treatment with this temperature sensitive formulation. Systemic toxicity indicators, such as cardiac hypertrophy, leukopenia, and weight and hair loss were not detected with CuDox-LTSLs after the 28-day therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/therapy , Doxorubicin/administration & dosage , Liposomes/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Combined Modality Therapy , Copper/chemistry , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Female , Hydrogen-Ion Concentration , Hyperthermia, Induced/methods , Mice , Temperature , Ultrasonics
13.
Med Phys ; 40(8): 083301, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23927360

ABSTRACT

PURPOSE: Ultrasound-induced mild hyperthermia has advantages for noninvasive, localized and controlled drug delivery. In this study, a tissue-mimicking agarose-based phantom with a thermally sensitive indicator was developed for studying the spatial drug delivery profile using ultrasound-induced mild hyperthermia. METHODS: Agarose powder, regular evaporated milk, Dulbecco's phosphate-buffered saline (DPBS), n-propanol, and silicon carbide powder were homogeneously mixed with low temperature sensitive liposomes (LTSLs) loaded with a self-quenched near-infrared (NIR) fluorescent dye. A dual-mode linear array ultrasound transducer was used for insonation at 1.54 MHz with a total acoustic power and acoustic pressure of 2.0 W and 1.5 MPa, respectively. After insonation, the dye release pattern in the phantom was quantified based on optical images, and the three-dimensional release profile was reconstructed and analyzed. A finite-difference time-domain-based algorithm was developed to simulate both the temperature distribution and spatial dye diffusion as a function of time. Finally, the simulated dye diffusion patterns were compared to experimental measurements. RESULTS: Self-quenching of the fluorescent dye in DPBS was substantial at a concentration of 6.25×10(-2) mM or greater. The transition temperature of LTSLs in the phantom was 35 °C, and the release reached 90% at 37 °C. The simulated temperature for hyperthermia correlated with the thermocouple measurements with a mean error between 0.03±0.01 and 0.06±0.02 °C. The R2 value between the experimental and simulated spatial extent of the dye diffusion, defined by the half-peak level in the elevation, lateral and depth directions, was 0.99 (slope=1.08), 0.95 (slope=0.99), and 0.80 (slope=1.04), respectively, indicating the experimental and simulated dye release profiles were similar. CONCLUSIONS: The combination of LTSLs encapsulating a fluorescent dye and an optically transparent phantom is useful for visualizing and modeling drug release in vitro following ultrasound-induced mild hyperthermia. The coupled temperature simulation and dye-diffusion simulation tools were validated with the experimental system and can be used to optimize the thermal dose and spatial and temporal dye release pattern.


Subject(s)
Drug Delivery Systems/methods , Hyperthermia, Induced , Phantoms, Imaging , Ultrasonics , Buffers , Carbocyanines/chemistry , Diffusion , Fluorescent Dyes/chemistry , Liposomes , Optical Imaging , Phosphates/chemistry , Phosphatidylcholines/chemistry , Temperature
14.
Proc Natl Acad Sci U S A ; 110(16): 6530-5, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23553837

ABSTRACT

Epidemiological and preclinical evidence supports that omega-3 dietary fatty acids (fish oil) reduce the risks of macular degeneration and cancers, but the mechanisms by which these omega-3 lipids inhibit angiogenesis and tumorigenesis are poorly understood. Here we show that epoxydocosapentaenoic acids (EDPs), which are lipid mediators produced by cytochrome P450 epoxygenases from omega-3 fatty acid docosahexaenoic acid, inhibit VEGF- and fibroblast growth factor 2-induced angiogenesis in vivo, and suppress endothelial cell migration and protease production in vitro via a VEGF receptor 2-dependent mechanism. When EDPs (0.05 mg · kg(-1) · d(-1)) are coadministered with a low-dose soluble epoxide hydrolase inhibitor, EDPs are stabilized in circulation, causing ~70% inhibition of primary tumor growth and metastasis. Contrary to the effects of EDPs, the corresponding metabolites derived from omega-6 arachidonic acid, epoxyeicosatrienoic acids, increase angiogenesis and tumor progression. These results designate epoxyeicosatrienoic acids and EDPs as unique endogenous mediators of an angiogenic switch to regulate tumorigenesis and implicate a unique mechanistic linkage between omega-3 and omega-6 fatty acids and cancers.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Cell Transformation, Neoplastic/drug effects , Docosahexaenoic Acids/metabolism , Epoxy Compounds/pharmacology , Fatty Acids, Omega-3/chemistry , Fatty Acids, Unsaturated/pharmacology , Neoplasm Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Animals , Cell Movement/drug effects , Dose-Response Relationship, Drug , Epoxide Hydrolases/antagonists & inhibitors , Epoxy Compounds/metabolism , Fatty Acids, Unsaturated/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy
15.
PLoS One ; 7(4): e35509, 2012.
Article in English | MEDLINE | ID: mdl-22536396

ABSTRACT

While Magnetic Resonance Thermometry (MRT) has been extensively utilized for non-invasive temperature measurement, there is limited data on the use of high field (≥7T) scanners for this purpose. MR-guided Focused Ultrasound (MRgFUS) is a promising non-invasive method for localized hyperthermia and drug delivery. MRT based on the temperature sensitivity of the proton resonance frequency (PRF) has been implemented in both a tissue phantom and in vivo in a mouse Met-1 tumor model, using partial parallel imaging (PPI) to speed acquisition. An MRgFUS system capable of delivering a controlled 3D acoustic dose during real time MRT with proportional, integral, and derivative (PID) feedback control was developed and validated. Real-time MRT was validated in a tofu phantom with fluoroptic temperature measurements, and acoustic heating simulations were in good agreement with MR temperature maps. In an in vivo Met-1 mouse tumor, the real-time PID feedback control is capable of maintaining the desired temperature with high accuracy. We found that real time MR control of hyperthermia is feasible at high field, and k-space based PPI techniques may be implemented for increasing temporal resolution while maintaining temperature accuracy on the order of 1°C.


Subject(s)
Hyperthermia, Induced , Magnetic Resonance Imaging , Mammary Neoplasms, Experimental/therapy , Thermometers , Ultrasonic Therapy , Animals , Cell Line, Tumor , Female , Mice , Models, Biological , Neoplasm Transplantation , Soy Foods , Temperature , Water
16.
IEEE Trans Biomed Eng ; 58(7): 2002-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21402506

ABSTRACT

Gold nanoparticles (GNPs) are nontoxic, can be functionalized with ligands, and preferentially accumulate in tumors. We have developed a 13.56-MHz RF-electromagnetic field (RF-EM) delivery system capable of generating high E-field strengths required for noninvasive, noncontact heating of GNPs. The bulk heating and specific heating rates were measured as a function of NP size and concentration. It was found that heating is both size and concentration dependent, with 5 nm particles producing a 50.6 ± 0.2 °C temperature rise in 30 s for 25 µg/mL gold (125 W input). The specific heating rate was also size and concentration dependent, with 5 nm particles producing a specific heating rate of 356 ± 78 kW/g gold at 16 µg/mL (125 W input). Furthermore, we demonstrate that cancer cells incubated with GNPs are killed when exposed to 13.56 MHz RF-EM fields. Compared to cells that were not incubated with GNPs, three out of four RF-treated groups showed a significant enhancement of cell death with GNPs (p<0.05). GNP-enhanced cell killing appears to require temperatures above 50 °C for the experimental parameters used in this study. Transmission electron micrographs show extensive vacuolization with the combination of GNPs and RF treatment.


Subject(s)
Gold/chemistry , Hyperthermia, Induced/instrumentation , Metal Nanoparticles/chemistry , Neoplasms/therapy , Cell Death/radiation effects , Cell Line, Tumor , Citric Acid , Electromagnetic Fields , Equipment Design , Hot Temperature , Humans , Hyperthermia, Induced/methods , Microscopy, Electron, Transmission , Nanotechnology , Particle Size
17.
Cancer Res ; 71(4): 1362-73, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21212415

ABSTRACT

Genetic and molecular studies suggest that activin receptor-like kinase 1 (ALK1) plays an important role in vascular development, remodeling, and pathologic angiogenesis. Here we investigated the role of ALK1 in angiogenesis in the context of common proangiogenic factors [PAF; VEGF-A and basic fibroblast growth factor (bFGF)]. We observed that PAFs stimulated ALK1-mediated signaling, including Smad1/5/8 phosphorylation, nuclear translocation and Id-1 expression, cell spreading, and tubulogenesis of endothelial cells (EC). An antibody specifically targeting ALK1 (anti-ALK1) markedly inhibited these events. In mice, anti-ALK1 suppressed Matrigel angiogenesis stimulated by PAFs and inhibited xenograft tumor growth by attenuating both blood and lymphatic vessel angiogenesis. In a human melanoma model with acquired resistance to a VEGF receptor kinase inhibitor, anti-ALK1 also delayed tumor growth and disturbed vascular normalization associated with VEGF receptor inhibition. In a human/mouse chimera tumor model, targeting human ALK1 decreased human vessel density and improved antitumor efficacy when combined with bevacizumab (anti-VEGF). Antiangiogenesis and antitumor efficacy were associated with disrupted co-localization of ECs with desmin(+) perivascular cells, and reduction of blood flow primarily in large/mature vessels as assessed by contrast-enhanced ultrasonography. Thus, ALK1 may play a role in stabilizing angiogenic vessels and contribute to resistance to anti-VEGF therapies. Given our observation of its expression in the vasculature of many human tumor types and in circulating ECs from patients with advanced cancers, ALK1 blockade may represent an effective therapeutic opportunity complementary to the current antiangiogenic modalities in the clinic.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/therapeutic use , Animals , Cells, Cultured , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , Mice , Mice, SCID , Molecular Targeted Therapy/methods , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
Article in English | MEDLINE | ID: mdl-21156363

ABSTRACT

Mild hyperthermia is increasingly important for the activation of temperature-sensitive drug delivery vehicles. Noninvasive ultrasound thermometry based on a 2-D speckle tracking algorithm was examined in this study. Here, a commercial ultrasound scanner, a customized co-linear array transducer, and a controlling PC system were used to generate mild hyperthermia. Because the co-linear array transducer is capable of both therapy and imaging at widely separated frequencies, RF image frames were acquired during therapeutic insonation and then exported for off-line analysis. For in vivo studies in a mouse model, before temperature estimation, motion correction was applied between a reference RF frame and subsequent RF frames. Both in vitro and in vivo experiments were examined; in the in vitro and in vivo studies, the average temperature error had a standard deviation of 0.7°C and 0.8°C, respectively. The application of motion correction improved the accuracy of temperature estimation, where the error range was 1.9 to 4.5°C without correction compared with 1.1 to 1.0°C following correction. This study demonstrates the feasibility of combining therapy and monitoring using a commercial system. In the future, real-time temperature estimation will be incorporated into this system.


Subject(s)
Hyperthermia, Induced/methods , Signal Processing, Computer-Assisted , Thermography/methods , Ultrasonography/methods , Algorithms , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Female , Image Processing, Computer-Assisted/methods , Mice , Neoplasm Transplantation , Phantoms, Imaging , Sepharose , Temperature , Thermography/instrumentation , Transducers , Ultrasonography/instrumentation
19.
IEEE Trans Biomed Eng ; 57(1): 155-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20064754

ABSTRACT

A new system is presented for generating controlled tissue heating with a clinical ultrasound scanner, and initial in vitro and in vivo results are presented that demonstrate both transient and sustained heating in the mild-hyperthermia range of 37 ( degrees )C-42 ( degrees )C. The system consists of a Siemens Antares ultrasound scanner, a custom dual-frequency three-row transducer array and an external temperature feedback control system. The transducer has two outer rows that operate at 1.5 MHz for tissue heating and a center row that operates at 5 MHz for B-mode imaging to guide the therapy. We compare the field maps obtained using a hydrophone against calculations of the ultrasound beam based on monochromatic and linear assumptions. Using the finite-difference time-domain (FDTD) method, we compare predicted time-dependent thermal profiles to measured profiles for soy tofu as a tissue-mimicking phantom. In vitro results show differential heating of 6 ( degrees )C for chicken breast and tofu. In vivo tests of the system were performed on three mice bearing Met-1 tumors, which is a model of aggressive, metastatic, and highly vascular breast cancer. In superficially implanted tumors, we demonstrate controlled heating to 42 ( degrees )C. We show that the system is able to maintain the temperature to within 0.1 ( degrees )C of the desired temperature both in vitro and in vivo.


Subject(s)
Hyperthermia, Induced/methods , Neoplasms, Experimental/diagnostic imaging , Ultrasonic Therapy/methods , Animals , Chickens , Hyperthermia, Induced/instrumentation , Meat , Mice , Soy Foods , Ultrasonic Therapy/instrumentation , Ultrasonography
20.
J Control Release ; 141(2): 128-36, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-19748536

ABSTRACT

To provide a continuous and prolonged delivery of the substrate D-luciferin for bioluminescence imaging in vivo, luciferin was encapsulated into liposomes using either the pH gradient or acetate gradient method. Under optimum loading conditions, 0.17 mg luciferin was loaded per mg of lipid with 90-95% encapsulation efficiency, where active loading was 6 to 18-fold higher than that obtained with passive loading. Liposomal luciferin in a long-circulating formulation had good shelf stability, with 10% release over 3-month storage at 4 degrees C. Pharmacokinetic profiles of free and liposomal luciferin were then evaluated in transgenic mice expressing luciferase. In contrast to rapid in vivo clearance of free luciferin (t(1/2)=3.54 min), luciferin encapsulated into long-circulating liposomes showed a prolonged release over 24h. The first-order release rate constant of luciferin from long-circulating liposomes, as estimated from the best fit of the analytical model to the experimental data, was 0.01 h(-1). Insonation of luciferin-loaded temperature-sensitive liposomes directly injected into one tumor of Met1-luc tumor-bearing mice resulted in immediate emission of light. Systemic injection of luciferin-loaded long-circulating liposomes into Met1-luc tumor-bearing mice, followed by unilateral ultrasound-induced hyperthermia, produced a gradual increase in radiance over time, reaching a peak at 4-7 h post-ultrasound.


Subject(s)
Benzothiazoles/administration & dosage , Drug Delivery Systems , Luminescence , Luminescent Agents/administration & dosage , Mammary Neoplasms, Experimental/pathology , Animals , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Cell Line, Tumor , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Compounding , Drug Stability , Female , Hydrogen-Ion Concentration , Hyperthermia, Induced , Injections, Intralesional , Injections, Intravenous , Liposomes , Luciferases/genetics , Luciferases/metabolism , Luminescent Agents/chemistry , Luminescent Agents/pharmacokinetics , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Models, Biological , Permeability , Solubility , Temperature , Transfection , Ultrasonics
SELECTION OF CITATIONS
SEARCH DETAIL