Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Ultrasound Med ; 41(9): 2365-2371, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34866197

ABSTRACT

The feasibility of activating phase change contrast agents (PCCA) made from Definity (Lantheus Medical Imaging) using X-rays was investigated. A 10 mL of Definity PCCA (0.1 mL PCCA/mL) were injected into gelatin phantoms and irradiated using doses of 0, 30, 50, and 100 Gy. Size distribution and PCCA activation were measured after irradiation. Definity PCCAs were activated at a threshold of 50 Gy. Changes were visible with microscopy, visual inspection of T-flasks, and ultrasound imaging of gelatin phantoms. Moreover, increasing the radiation dose above 50 Gy appeared to further activate PCCA. These results indicate Definity PCCAs may be useful for ultrasound-based radiation dosimetry.


Subject(s)
Contrast Media , Gelatin , Humans , Phantoms, Imaging , Radiation, Ionizing , Ultrasonography/methods
2.
Int J Hyperthermia ; 37(1): 1189-1201, 2020.
Article in English | MEDLINE | ID: mdl-33047639

ABSTRACT

AIM: Hyperthermia (HT) has been shown to improve clinical response to radiation therapy (RT) for cancer. Synergism is dramatically enhanced if HT and RT are combined simultaneously, but appropriate technology to apply treatments together does not exist. This study investigates the feasibility of delivering HT with RT to a 5-10mm annular rim of at-risk tissue around a tumor resection cavity using a temporary thermobrachytherapy (TBT) balloon implant. METHODS: A balloon catheter was designed to deliver radiation from High Dose Rate (HDR) brachytherapy concurrent with HT delivered by filling the balloon with magnetic nanoparticles (MNP) and immersing it in a radiofrequency magnetic field. Temperature distributions in brain around the TBT balloon were simulated with temperature dependent brain blood perfusion using numerical modeling. A magnetic induction system was constructed and used to produce rapid heating (>0.2°C/s) of MNP-filled balloons in brain tissue-equivalent phantoms by absorbing 0.5 W/ml from a 5.7 kA/m field at 133 kHz. RESULTS: Simulated treatment plans demonstrate the ability to heat at-risk tissue around a brain tumor resection cavity between 40-48°C for 2-5cm diameter balloons. Experimental thermal dosimetry verifies the expected rapid and spherically symmetric heating of brain phantom around the MNP-filled balloon at a magnetic field strength that has proven safe in previous clinical studies. CONCLUSIONS: These preclinical results demonstrate the feasibility of using a TBT balloon to deliver heat simultaneously with HDR brachytherapy to tumor bed around a brain tumor resection cavity, with significantly improved uniformity of heating over previous multi-catheter interstitial approaches. Considered along with results of previous clinical thermobrachytherapy trials, this new capability is expected to improve both survival and quality of life in patients with glioblastoma multiforme.


Subject(s)
Brachytherapy , Brain Neoplasms , Hyperthermia, Induced , Magnetite Nanoparticles , Brain Neoplasms/radiotherapy , Feasibility Studies , Heating , Humans , Quality of Life
3.
J Ultrasound Med ; 38(12): 3221-3228, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31124171

ABSTRACT

OBJECTIVES: Hypoxic cancer cells have been shown to be more resistant to radiation therapy than normoxic cells. Hence, this study investigated whether ultrasound (US)-induced rupture of oxygen-carrying microbubbles (MBs) would enhance the response of breast cancer metastases to radiation. METHODS: Nude mice (n = 15) received stereotactic injections of brain-seeking MDA-MB-231 breast cancer cells into the right hemisphere. Animals were randomly assigned into 1 of 5 treatment groups: no intervention, 10 Gy radiation using a small-animal radiation research platform, nitrogen-carrying MBs combined with US-mediated MB rupture immediately before 10 Gy radiation, oxygen-carrying MBs immediately before 10 Gy radiation, and oxygen-carrying MBs with US-mediated MB rupture immediately before 10 Gy radiation. Tumor progression was monitored with 3-dimensional US, and overall survival was noted. RESULTS: All groups except those treated with oxygen-carrying MB rupture and radiation had continued rapid tumor growth after treatment. Tumors treated with radiation alone showed a mean increase in volume ± SD of 337% ± 214% during the week after treatment. Tumors treated with oxygen-carrying MBs and radiation without MB rupture showed an increase in volume of 383% ± 226%. Tumors treated with radiation immediately after rupture of oxygen-carrying MBs showed an increase in volume of only 41% ± 1% (P = 0.045), and this group also showed a 1 week increase in survival time. CONCLUSIONS: Adding US-ruptured oxygen-carrying MBs to radiation therapy appears to delay tumor progression and improve survival in a murine model of metastatic breast cancer.


Subject(s)
Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Drug Carriers , Microbubbles , Oxygen/administration & dosage , Animals , Disease Models, Animal , Female , Mice , Mice, Nude , Random Allocation , Ultrasonography
4.
J Biol Chem ; 291(10): 5157-71, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26703469

ABSTRACT

A network model for the determination of tumor metabolic fluxes from (13)C NMR kinetic isotopomer data has been developed and validated with perfused human DB-1 melanoma cells carrying the BRAF V600E mutation, which promotes oxidative metabolism. The model generated in the bonded cumomer formalism describes key pathways of tumor intermediary metabolism and yields dynamic curves for positional isotopic enrichment and spin-spin multiplets. Cells attached to microcarrier beads were perfused with 26 mm [1,6-(13)C2]glucose under normoxic conditions at 37 °C and monitored by (13)C NMR spectroscopy. Excellent agreement between model-predicted and experimentally measured values of the rates of oxygen and glucose consumption, lactate production, and glutamate pool size validated the model. ATP production by glycolytic and oxidative metabolism were compared under hyperglycemic normoxic conditions; 51% of the energy came from oxidative phosphorylation and 49% came from glycolysis. Even though the rate of glutamine uptake was ∼ 50% of the tricarboxylic acid cycle flux, the rate of ATP production from glutamine was essentially zero (no glutaminolysis). De novo fatty acid production was ∼ 6% of the tricarboxylic acid cycle flux. The oxidative pentose phosphate pathway flux was 3.6% of glycolysis, and three non-oxidative pentose phosphate pathway exchange fluxes were calculated. Mass spectrometry was then used to compare fluxes through various pathways under hyperglycemic (26 mm) and euglycemic (5 mm) conditions. Under euglycemic conditions glutamine uptake doubled, but ATP production from glutamine did not significantly change. A new parameter measuring the Warburg effect (the ratio of lactate production flux to pyruvate influx through the mitochondrial pyruvate carrier) was calculated to be 21, close to upper limit of oxidative metabolism.


Subject(s)
Melanoma/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , Cell Line, Tumor , Citric Acid Cycle , Glucose/metabolism , Glutamine/metabolism , Humans , Models, Theoretical , Mutation, Missense , Oxygen/metabolism , Proto-Oncogene Proteins B-raf/genetics
5.
J Biol Chem ; 291(1): 42-57, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26521302

ABSTRACT

The antitumor agent lonidamine (LND; 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid) is known to interfere with energy-yielding processes in cancer cells. However, the effect of LND on central energy metabolism has never been fully characterized. In this study, we report that a significant amount of succinate is accumulated in LND-treated cells. LND inhibits the formation of fumarate and malate and suppresses succinate-induced respiration of isolated mitochondria. Utilizing biochemical assays, we determined that LND inhibits the succinate-ubiquinone reductase activity of respiratory complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through complex II, which reduced the viability of the DB-1 melanoma cell line. The ability of LND to promote cell death was potentiated by its suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. Using stable isotope tracers in combination with isotopologue analysis, we showed that LND increased glutaminolysis but decreased reductive carboxylation of glutamine-derived α-ketoglutarate. Our findings on the previously uncharacterized effects of LND may provide potential combinational therapeutic approaches for targeting cancer metabolism.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex II/antagonists & inhibitors , Indazoles/pharmacology , Mitochondria/metabolism , Cell Death/drug effects , Cell Line, Tumor , Citric Acid Cycle/drug effects , Diacetyl/analogs & derivatives , Diacetyl/pharmacology , Electron Transport Complex II/metabolism , Fumarates/metabolism , Glutamine/metabolism , Glutathione/metabolism , Humans , Malates/metabolism , Melanoma/metabolism , Melanoma/pathology , Metabolic Flux Analysis , Mitochondria/drug effects , Models, Biological , NADP/metabolism , Naphthalenes/pharmacology , Oxidation-Reduction/drug effects , Pentose Phosphate Pathway/drug effects , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism
6.
Biochim Biophys Acta ; 1866(2): 151-162, 2016 12.
Article in English | MEDLINE | ID: mdl-27497601

ABSTRACT

Lonidamine (LND) was initially introduced as an antispermatogenic agent. It was later found to have anticancer activity sensitizing tumors to chemo-, radio-, and photodynamic-therapy and hyperthermia. Although the mechanism of action remained unclear, LND treatment has been known to target metabolic pathways in cancer cells. It has been reported to alter the bioenergetics of tumor cells by inhibiting glycolysis and mitochondrial respiration, while indirect evidence suggested that it also inhibited l-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). Recent studies have demonstrated that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki 2.5µM) and cooperatively inhibits l-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevis oocytes with K0.5 and Hill coefficient values of 36-40µM and 1.65-1.85, respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~7µM) than other substrates including glutamate (IC50~20µM). LND inhibits the succinate-ubiquinone reductase activity of respiratory Complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through Complex II and has been reported to promote cell death by suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated l-lactic acid efflux, Complex II and glutamine/glutamate oxidation.


Subject(s)
Antineoplastic Agents/pharmacology , Indazoles/pharmacology , Animals , Hexokinase/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Indazoles/toxicity , Membrane Transport Proteins/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/metabolism
7.
Biochem J ; 473(7): 929-36, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26831515

ABSTRACT

Lonidamine (LND) is an anti-tumour drug particularly effective at selectively sensitizing tumours to chemotherapy, hyperthermia and radiotherapy, although its precise mode of action remains unclear. It has been reported to perturb the bioenergetics of cells by inhibiting glycolysis and mitochondrial respiration, whereas indirect evidence suggests it may also inhibit L-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). In the present study, we test these possibilities directly. We demonstrate that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki2.5 µM) and co-operatively inhibits L-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevisoocytes with K0.5 and Hill coefficient values of 36-40 µM and 1.65-1.85 respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~ 7 µM) than other substrates including glutamate (IC50~ 20 µM). In isolated DB-1 melanoma cells 1-10 µM LND increased L-lactate output, consistent with MPC inhibition, but higher concentrations (150 µM) decreased L-lactate output whereas increasing intracellular [L-lactate] > 5-fold, consistent with MCT inhibition. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated L-lactic acid efflux and glutamine/glutamate oxidation. Together these actions can account for published data on the selective tumour effects of LND onL-lactate, intracellular pH (pHi) and ATP levels that can be partially mimicked by the established MPC and MCT inhibitor α-cyano-4-hydroxycinnamate (CHC).


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/antagonists & inhibitors , Cell Membrane/metabolism , Indazoles/pharmacology , Membrane Transport Proteins , Mitochondrial Proteins/antagonists & inhibitors , Monocarboxylic Acid Transporters/antagonists & inhibitors , Symporters/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/genetics , Ion Transport/drug effects , Ion Transport/genetics , Lactic Acid/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Solute Carrier Proteins , Symporters/genetics , Symporters/metabolism
8.
NMR Biomed ; 28(3): 281-90, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25504852

ABSTRACT

We demonstrate that the effects of lonidamine (LND, 100 mg/kg, i.p.) are similar for a number of xenograft models of human cancer including DB-1 melanoma and HCC1806 breast, BT-474 breast, LNCaP prostate and A2870 ovarian carcinomas. Following treatment with LND, each of these tumors exhibits a rapid decrease in intracellular pH, a small decrease in extracellular pH, a concomitant monotonic decrease in nucleoside triphosphate and an increase in inorganic phosphate over a 2-3 h period. We have previously demonstrated that selective intracellular tumor acidification potentiates response of this melanoma model to melphalan (7.5 mg/kg, i.v.), producing an estimated 89% cell kill based on tumor growth delay analysis. We now show that, in both DB-1 melanoma and HCC1806 breast carcinoma, LND potentiates response to doxorubicin, producing 95% cell kill in DB-1 melanoma at 7.5 mg/kg, i.v. doxorubicin and 98% cell kill at 10.0 mg/kg doxorubicin, and producing a 95% cell kill in HCC1806 breast carcinoma at 12.0 mg/kg doxorubicin. Potentiation of doxorubicin may result from cation trapping of the weakly basic anthracycline. Recent experience with the clinical treatment of melanoma and other forms of human cancer suggests that these diseases will probably not be cured by a single therapeutic procedure other than surgery. A multimodality therapeutic approach will be required. As a potent modulator of tumor response to N-mustards and anthracyclines as well as tumor thermo- and radiosensitivity, LND promises to play an important clinical role in the management and possible complete local control of a number of prevalent forms of human cancer.


Subject(s)
Adenosine Triphosphate/deficiency , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Indazoles/pharmacology , Ovarian Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Xenograft Model Antitumor Assays , Acids/metabolism , Animals , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Energy Metabolism/drug effects , Female , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Magnetic Resonance Spectroscopy , Male , Mice, Nude , Monocarboxylic Acid Transporters/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/pathology , Symporters/metabolism
9.
NMR Biomed ; 28(3): 395-403, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25702942

ABSTRACT

We seek to exploit the natural tendency of melanomas and other tumors to convert glucose to lactate as a method for the selective intracellular acidification of cancer cells and for the potentiation of the activity of nitrogen-mustard antineoplastic agents. We performed this study to evaluate whether the induction of hyperglycemia (26 mM) could enhance the effects of lonidamine (LND, 100 mg/kg; intraperitoneally) on the induction of intracellular acidification, bioenergetic decline and potentiation of the activity of melphalan (LPAM) against DB-1 melanoma xenografts in mice. Intracellular pH (pHi ), extracellular pH (pHe ) and bioenergetics (ß-nucleoside triphosphate to inorganic phosphate ratio, ß-NTP/Pi) were reduced by 0.7 units (p < 0.001), 0.3 units (p > 0.05) and 51.4% (p < 0.05), respectively. The therapeutic response to LPAM (7.5 mg/kg; intravenously) + LND (100 mg/kg; intraperitoneally) was reduced by about a factor of three under hyperglycemic conditions relative to normoglycemia, producing a growth delay of 7.76 days (tumor doubling time, 5.31 days; cell kill, 64%) compared with LND alone of 1.70 days and LPAM alone of 0.29 days. Under normoglycemic conditions, LND plus LPAM produced a growth delay of 17.75 days, corresponding to a cell kill of 90% at the same dose for each of these agents. The decrease in tumor cell kill under hyperglycemic conditions correlates with an increase in tumor ATP levels resulting from increased glycolytic activity. However, hyperglycemia substantially increases lactic acid production in tumors by a factor of approximately six (p < 0.05), but hyperglycemia did not increase the effects of LND on acidification of the tumor, most probably because of the strong buffering action of carbon dioxide (the pKa of carbonic acid is 6.4). Therefore, this study demonstrates that the addition of glucose during treatment with LND diminishes the activity of this agent.


Subject(s)
Acids/metabolism , Energy Metabolism/drug effects , Hyperglycemia/complications , Indazoles/pharmacology , Melanoma/metabolism , Melphalan/pharmacology , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Magnetic Resonance Spectroscopy , Male , Melanoma/complications , Melanoma/pathology , Mice, Nude , Organ Specificity
10.
Int J Hyperthermia ; 30(1): 75-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24295212

ABSTRACT

PURPOSE: This study tested the ability of lonidamine (LND), a clinically applicable inhibitor of monocarboxylate transporters (MCT), to thermally sensitise human melanoma cells cultured at a tumour-like extracellular pH (pHe) 6.7. MATERIALS AND METHODS: Human melanoma DB-1 cells cultured at pHe 6.7 and pHe 7.3 were exposed to 150 µM LND for 3 h, beginning 1 h prior to heating at 42 °C (2 h). Intracellular pH (pHi) was determined using 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) and whole spectrum analysis. Levels of heat shock proteins (HSPs) were determined by immunoblot analysis. Cell survival was determined by colony formation. RESULTS: Treatment with LND at pHe 6.7 reduced pHi to 6.30 ± 0.21, reduced thermal induction of HSPs, and sensitised cells growing at pHe 6.7 to 42 °C. When LND was combined with an acute acidification from pHe 6.7 to pHe 6.5, pHi was reduced to 6.09 ± 0.26, and additional sensitisation was observed. LND had negligible effects on cells cultured at pH 7.3. CONCLUSIONS: The results show that LND can reduce pHi in human melanoma cells cultured at a tumour-like low pHe so that the 42 °C induction of HSPs are abrogated and the cells are sensitised to thermal therapy. Cells cultured at a normal tissue-like pHe 7.3 were not sensitised to 42 °C by LND. These findings support the strategy that human melanoma cells growing in an acidic environment can be sensitised to thermal therapy in vivo by exposure to an MCT inhibitor such as LND.


Subject(s)
Indazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Hot Temperature , Humans , Hydrogen-Ion Concentration , Melanoma , Molecular Chaperones , Monocarboxylic Acid Transporters/antagonists & inhibitors
11.
NMR Biomed ; 26(1): 98-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22745015

ABSTRACT

In vivo (31) P MRS demonstrates that human melanoma xenografts in immunosuppressed mice treated with lonidamine (LND, 100 mg/kg intraperitoneally) exhibit a decrease in intracellular pH (pH(i) ) from 6.90 ± 0.05 to 6.33 ± 0.10 (p < 0.001), a slight decrease in extracellular pH (pH(e) ) from 7.00 ± 0.04 to 6.80 ± 0.07 (p > 0.05) and a monotonic decline in bioenergetics (nucleoside triphosphate/inorganic phosphate) of 66.8 ± 5.7% (p < 0.001) relative to the baseline level. Both bioenergetics and pH(i) decreases were sustained for at least 3 h following LND treatment. Liver exhibited a transient intracellular acidification by 0.2 ± 0.1 pH units (p > 0.05) at 20 min post-LND, with no significant change in pH(e) and a small transient decrease in bioenergetics (32.9 ± 10.6%, p > 0.05) at 40 min post-LND. No changes in pH(i) or adenosine triphosphate/inorganic phosphate were detected in the brain (pH(i) , bioenergetics; p > 0.1) or skeletal muscle (pH(i) , pH(e) , bioenergetics; p > 0.1) for at least 120 min post-LND. Steady-state tumor lactate monitored by (1) H MRS with a selective multiquantum pulse sequence with Hadamard localization increased approximately three-fold (p = 0.009). Treatment with LND increased the systemic melanoma response to melphalan (LPAM; 7.5 mg/kg intravenously), producing a growth delay of 19.9 ± 2.0 days (tumor doubling time, 6.15 ± 0.31 days; log(10) cell kill, 0.975 ± 0.110; cell kill, 89.4 ± 2.2%) compared with LND alone of 1.1 ± 0.1 days and LPAM alone of 4.0 ± 0.0 days. The study demonstrates that the effects of LND on tumor pH(i) and bioenergetics may sensitize melanoma to pH-dependent therapeutics, such as chemotherapy with alkylating agents or hyperthermia.


Subject(s)
Hydrogen-Ion Concentration/drug effects , Indazoles/administration & dosage , Magnetic Resonance Spectroscopy/methods , Melanoma/chemistry , Melanoma/drug therapy , Melphalan/administration & dosage , Animals , Antineoplastic Agents, Alkylating , Cell Line, Tumor , Drug Synergism , Energy Metabolism/drug effects , Melanoma/physiopathology , Mice , Phosphorus Radioisotopes , Protons , Treatment Outcome
12.
Pharmaceutics ; 15(4)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37111787

ABSTRACT

Tumor hypoxia (oxygen deficiency) is a major contributor to radiotherapy resistance. Ultrasound-sensitive microbubbles containing oxygen have been explored as a mechanism for overcoming tumor hypoxia locally prior to radiotherapy. Previously, our group demonstrated the ability to encapsulate and deliver a pharmacological inhibitor of tumor mitochondrial respiration (lonidamine (LND)), which resulted in ultrasound-sensitive microbubbles loaded with O2 and LND providing prolonged oxygenation relative to oxygenated microbubbles alone. This follow-up study aimed to evaluate the therapeutic response to radiation following the administration of oxygen microbubbles combined with tumor mitochondrial respiration inhibitors in a head and neck squamous cell carcinoma (HNSCC) tumor model. The influences of different radiation dose rates and treatment combinations were also explored. The results demonstrated that the co-delivery of O2 and LND successfully sensitized HNSCC tumors to radiation, and this was also enhanced with oral metformin, significantly slowing tumor growth relative to unsensitized controls (p < 0.01). Microbubble sensitization was also shown to improve overall animal survival. Importantly, effects were found to be radiation dose-rate-dependent, reflecting the transient nature of tumor oxygenation.

13.
Int J Pharm ; 625: 122072, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-35932933

ABSTRACT

Prior work has shown that microbubble-assisted delivery of oxygen improves tumor oxygenation and radiosensitivity, albeit over a limited duration. Lonidamine (LND) has been investigated because of its ability to stimulate glycolysis, lactate production, inhibit mitochondrial respiration, and inhibit oxygen consumption rates in tumors but suffers from poor bioavailability. The goal of this work was to characterize LND-loaded oxygen microbubbles and assess their ability to oxygenate a human head and neck squamous cell carcinoma (HNSCC) tumor model, while also assessing LND biodistribution. In tumors treated with surfactant-shelled microbubbles with oxygen core (SE61O2) and ultrasound, pO2 levels increased to a peak 19.5 ± 9.7 mmHg, 50 s after injection and returning to baseline after 120 s. In comparison, in tumors treated with SE61O2/LND and ultrasound, pO2 levels showed a peak increase of 29.0 ± 8.3 mmHg, which was achieved 70 s after injection returning to baseline after 300 s (p < 0.001). The co-delivery of O2andLNDvia SE61 also showed an improvement of LND biodistribution in both plasma and tumor tissues (p < 0.001). In summary, ultrasound-sensitive microbubbles loaded with O2 and LND provided prolonged oxygenation relative to oxygenated microbubbles alone, as well as provided an ability to locally deliver LND, making them more appropriate for clinical translation.


Subject(s)
Microbubbles , Neoplasms , Humans , Indazoles , Oxygen , Tissue Distribution
14.
Adv Exp Med Biol ; 701: 69-73, 2011.
Article in English | MEDLINE | ID: mdl-21445771

ABSTRACT

Previously we reported that three imaging methods, dynamic contrast enhanced magnetic resonance imaging (DCE-MRI), T1(ρ)-MRI, and ultralow temperature NADH/flavoprotein fluorescence imaging (redox scanning), could differentiate the less metastatic human melanoma cell line A375P from a more metastatic line C8161 growing as mouse xenografts in nude mice (Li LZ et al. Adv. Exp. Med. Biol., 2007, 599:67-78; PNAS, 2009, 106:6608-6613). The more metastatic C8161 tumor was characterized by less blood perfusion/permeability, a more oxidized mitochondrial redox state in the tumor core, and a smaller T1(ρ) relaxation time constant averaged across the entire tumor section. In the current study, we have further probed the bioenergetic status and tissue microenvironment of these tumors by applying whole tumor phosphorous magnetic resonance spectroscopy ((31)P-MRS) to these two xenografts in a vertical bore 9.4-T Varian magnet. The phosphomonoester (PME)/ßNTP ratio and intracellular pH value (pHi) were determined. The phosphomonoester (PME)/ßNTP was higher in the more metastatic C8161 tumors (n=4) than in the less metastatic A375P tumors (n=4) (p < 0.1). No significant difference between the pHi of C8161 and A375P was observed.


Subject(s)
Diagnostic Imaging , Magnetic Resonance Spectroscopy , Melanoma/pathology , Phosphorus Isotopes , Animals , Contrast Media , Humans , Male , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Metastasis , Oxidation-Reduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Ultrasound Med Biol ; 47(6): 1465-1474, 2021 06.
Article in English | MEDLINE | ID: mdl-33653626

ABSTRACT

Radiation therapy (RT) causes DNA damage through ionization, leading to double-strand breaks. In addition, it generates reactive oxygen species (ROS), which are toxic to tumor cells and the vasculature. However, hypoxic regions in the tumor have been shown to not only decrease treatment response but also increase the likelihood of recurrence and metastasis. Ultrasound-sensitive micro-bubbles are emerging as a useful diagnostic and therapeutic tool within RT. Contrast-enhanced ultrasound (CEUS) has shown great promise in early prediction of tumor response to RT. Ultrasound-triggered micro-bubble cavitation has also been shown to induce bio-effects that can sensitize angiogenic tumor vessels to RT. Additionally, ultrasound can trigger the release of drugs from micro-bubble carriers via localized micro-bubble destruction. This approach has numerous applications in RT, including targeted oxygen delivery before radiotherapy. Furthermore, micro-bubbles can be used to locally create ROS without radiation. Sonodynamic therapy uses focused ultrasound and a sonosensitizer to selectively produce ROS in the tumor region and has been explored as a treatment option for cancer. This review summarizes emerging applications of ultrasound contrast agents in RT and ROS augmentation.


Subject(s)
Contrast Media , Microbubbles , Neoplasms/radiotherapy , Drug Delivery Systems , Humans , Radiotherapy/methods , Ultrasonography
16.
Sci Rep ; 8(1): 14654, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279592

ABSTRACT

Lonidamine (LND), a metabolic modulator, sensitizes DB-1 human melanoma to doxorubicin (DOX) chemotherapy by acidifying and de-energizing the tumor. This report compares the effects of LND on two human melanoma lines, DB-1 and WM983B, which exhibit different metabolic properties. Using liquid chromatography mass spectrometry and Seahorse analysis, we show that DB-1 was more glycolytic than WM983B in vitro. 31P magnetic resonance spectroscopy (MRS) indicates that LND (100 mg/kg, i.p.) induces similar selective acidification and de-energization of WM983B xenografts in immunosuppressed mice. Over three hours, intracellular pH (pHi) of WM983B decreased from 6.91 ± 0.03 to 6.59 ± 0.10 (p = 0.03), whereas extracellular pH (pHe) of this tumor changed from 7.03 ± 0.05 to 6.89 ± 0.06 (p = 0.19). A decline in bioenergetics (ß-NTP/Pi) of 55 ± 5.0% (p = 0.03) accompanied the decline in pHi of WM983B. Using 1H MRS with a selective multiquantum pulse sequence and Hadamard localization, we show that LND induced a significant increase in tumor lactate levels (p < 0.01). LND pre-treatment followed by DOX (10 mg/kg, i.v.) produced a growth delay of 13.7 days in WM983B (p < 0.01 versus control), a growth delay significantly smaller than the 25.4 days that occurred with DB-1 (p = 0.03 versus WM983B). Differences in relative levels of glycolysis may produce differential therapeutic responses of DB-1 and WM983B melanomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Indazoles/pharmacology , Melanoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Doxorubicin/therapeutic use , Drug Synergism , Glucose/analysis , Glucose/metabolism , Glycolysis/drug effects , Humans , Hydrogen-Ion Concentration , Indazoles/therapeutic use , Lactic Acid/analysis , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Melanoma/pathology , Mice , Mice, Nude , Oxygen/analysis , Oxygen/metabolism , Oxygen Consumption/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
17.
Int J Radiat Oncol Biol Phys ; 101(1): 88-96, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29477294

ABSTRACT

PURPOSE: Much of the volume of solid tumors typically exists in a chronically hypoxic microenvironment that has been shown to result in both chemotherapy and radiation therapy resistance. The purpose of this study was to use localized microbubble delivery to overcome hypoxia prior to therapy. MATERIALS AND METHODS: In this study, surfactant-shelled oxygen microbubbles were fabricated and injected intravenously to locally elevate tumor oxygen levels when triggered by noninvasive ultrasound in mice with human breast cancer tumors. Changes in oxygen and sensitivity to radiation therapy were then measured. RESULTS: In this work, we show that oxygen-filled microbubbles successfully and consistently increase breast tumor oxygenation levels in a murine model by 20 mmHg, significantly more than control injections of saline solution or untriggered oxygen microbubbles (P < .001). Using photoacoustic imaging, we also show that oxygen delivery is independent of hemoglobin transport, enabling oxygen delivery to avascular regions of the tumor. Finally, we show that overcoming hypoxia by this method immediately prior to radiation therapy nearly triples radiosensitivity. This improvement in radiosensitivity results in roughly 30 days of improved tumor control, providing statistically significant improvements in tumor growth and animal survival (P < .03). CONCLUSIONS: Our findings demonstrate the potential advantages of ultrasound-triggered oxygen delivery to solid tumors and warrant future efforts into clinical translation of the microbubble platform.


Subject(s)
Microbubbles , Oxygen/administration & dosage , Radiation Tolerance , Triple Negative Breast Neoplasms/radiotherapy , Tumor Hypoxia/radiation effects , Animals , Cell Line, Tumor , Female , Humans , Injections, Intravenous , Mice , Mice, Nude , Oxygen/metabolism , Oxygen Consumption , Partial Pressure , Random Allocation , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Ultrasonic Therapy/methods
18.
Cancer Lett ; 411: 100-105, 2017 12 28.
Article in English | MEDLINE | ID: mdl-28969964

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide and the fastest growing malignancy in the United States. With a 5-year survival rate below 12%, effective therapies for HCC are needed. Current treatments for HCC include microwave and radiofrequency ablation, high intensity focused ultrasound, liver transplant, surgical resection, and localized embolizations. However, each of these approaches has some limitation, making it imperative to develop improved methods for sensitizing tumors prior to therapy. We hypothesized that the use of ultrasound-triggered microbubble destruction (UTMD), which sensitizes tumors to radiotherapy by inducing vascular endothelial cell apoptosis, will selectively sensitize malignant tissue to radiotherapy and improve outcomes. To test this, 18 nude rats were inoculated in the right liver lobe with Hu7.5 HCC cells and after tumor formation, received 5 Gy radiotherapy, UTMD, or UTMD prior to radiotherapy. Compared to radiotherapy alone, there was a 170% reduction in tumor growth 7 days post treatment and a 3.2X improvement in median survival time when radiotherapy was combined with UTMD. These results indicate that UTMD is an effective adjunct when combined with radiotherapy to treat HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Microbubbles/therapeutic use , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/radiotherapy , Cell Line, Tumor , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , Neovascularization, Pathologic/therapy , Random Allocation , Rats , Rats, Nude , Xenograft Model Antitumor Assays
19.
Anticancer Res ; 37(7): 3413-3421, 2017 07.
Article in English | MEDLINE | ID: mdl-28668829

ABSTRACT

BACKGROUND/AIM: Since temozolomide (TMZ) is activated under alkaline conditions, we expected lonidamine (LND) to have no effect or perhaps diminish its activity, but initial results suggest it may actually enhance either or both short- and long-term activity of TMZ in melanoma xenografts. MATERIALS AND METHODS: Cohorts of 5 mice with subcutaneous xenografts ~5 mm in diameter were treated with saline (control (CTRL)), LND only, TMZ only or LND followed by TMZ at t=40 min (time required for maximal tumor acidification). RESULTS: Mean tumor volume for LND+TMZ for the period between 6 and 26 days was reduced compared to TMZ alone (repeated measures ANOVA F (1, 8), p=0.006), suggesting a pronounced impact of LND on this phenomenon. TMZ and LND+TMZ produced median growth delays of 82 and 106 days, respectively. CONCLUSION: The use of TMZ alone and in combination with LND deserves further investigation in treatment of melanoma and other malignancies.


Subject(s)
Dacarbazine/analogs & derivatives , Indazoles/administration & dosage , Melanoma/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/administration & dosage , Drug Synergism , Heterografts/drug effects , Humans , Mice , Mice, Nude , Temozolomide , Transplantation, Heterologous/methods
20.
PLoS One ; 11(6): e0157125, 2016.
Article in English | MEDLINE | ID: mdl-27285585

ABSTRACT

Previous NMR studies demonstrated that lonidamine (LND) selectively diminishes the intracellular pH (pHi) of DB-1 melanoma and mouse xenografts of a variety of other prevalent human cancers while decreasing their bioenergetic status (tumor ßNTP/Pi ratio) and enhancing the activities of melphalan and doxorubicin in these cancer models. Since melphalan and doxorubicin are highly toxic agents, we have examined three other nitrogen (N)-mustards, chlorambucil, cyclophosphamide and bendamustine, to determine if they exhibit similar potentiation by LND. As single agents LND, melphalan and these N-mustards exhibited the following activities in DB-1 melanoma xenografts; LND: 100% tumor surviving fraction (SF); chlorambucil: 100% SF; cyclophosphamide: 100% SF; bendamustine: 79% SF; melphalan: 41% SF. When combined with LND administered 40 min prior to administration of the N-mustard (to maximize intracellular acidification) the following responses were obtained; chlorambucil: 62% SF; cyclophosphamide: 42% SF; bendamustine: 36% SF; melphalan: 10% SF. The effect of LND on the activities of these N-mustards is generally attributed to acid stabilization of the aziridinium active intermediate, acid inhibition of glutathione-S-transferase, which acts as a scavenger of aziridinium, and acid inhibition of DNA repair by O6-alkyltransferase. Depletion of ATP by LND may also decrease multidrug resistance and increase tumor response. At similar maximum tolerated doses, our data indicate that melphalan is the most effective N-mustard in combination with LND when treating DB-1 melanoma in mice, but the choice of N-mustard for coadministration with LND will also depend on the relative toxicities of these agents, and remains to be determined.


Subject(s)
Alkylating Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indazoles/pharmacology , Mechlorethamine/pharmacology , Melanoma/pathology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chlorambucil/pharmacology , Cyclophosphamide/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Humans , Male , Melanoma/drug therapy , Melphalan/pharmacology , Mice , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL