Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 349
Filter
1.
Dokl Biochem Biophys ; 516(1): 111-114, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38795244

ABSTRACT

Proton therapy can treat tumors located in radiation-sensitive tissues. This article demonstrates the possibility of enhancing the proton therapy with targeted gold nanoparticles that selectively recognize tumor cells. Au-PEG nanoparticles at concentrations above 25 mg/L and 4 Gy proton dose caused complete death of EMT6/P cells in vitro. Binary proton therapy using targeted Au-PEG-FA nanoparticles caused an 80% tumor growth inhibition effect in vivo. The use of targeted gold nanoparticles is promising for enhancing the proton irradiation effect on tumor cells and requires further research to increase the therapeutic index of the approach.


Subject(s)
Carcinoma, Ehrlich Tumor , Gold , Metal Nanoparticles , Proton Therapy , Gold/chemistry , Gold/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Proton Therapy/methods , Animals , Carcinoma, Ehrlich Tumor/radiotherapy , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/pathology , Mice , Cell Line, Tumor , Polyethylene Glycols/chemistry
2.
Bull Exp Biol Med ; 176(5): 626-630, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38730109

ABSTRACT

We studied the antitumor activity of the combined use of local proton irradiation in two modes (10 and 31 Gy) with preliminary intra-tumoral injection of two types of bismuth nanoparticles differing in surface coating: coated with the amphiphilic molecule Pluronic-F127 or Silane-PEG (5 kDa)-COOH polymer. Nanoparticles were used in doses of 0.75 and 1.5 mg/mouse. In two independent series on experimental tumor model (solid Ehrlich carcinoma), bismuth nanoparticles of both modifications injected directly into the tumor enhanced the antitumor effects of proton therapy. Moreover, the radiosensitizing effect of bismuth nanoparticles administered via this route increased with the increasing the doses of nanoparticles and the doses of radiation exposure. In our opinion, these promising data obtained for the first time extend the possibilities of treating malignant neoplasms.


Subject(s)
Bismuth , Carcinoma, Ehrlich Tumor , Poloxamer , Proton Therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/pathology , Animals , Bismuth/therapeutic use , Bismuth/chemistry , Mice , Proton Therapy/methods , Poloxamer/chemistry , Radiation-Sensitizing Agents/therapeutic use , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Polyethylene Glycols/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Nanoparticles/chemistry , Female
3.
Dokl Biochem Biophys ; 511(1): 151-155, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37833598

ABSTRACT

This study presents data on the growth rate and frequency of induction of the solid form of Ehrlich's ascites carcinoma (EAC) in mice in the short and long term after inoculation of ascitic cells irradiated ex vivo with a proton beam in the dose range of 30-150 Gy. It was shown that the growth rate of solid tumors after inoculation of irradiated cells ex vivo coincided with the growth of tumors in the control group. The frequency of tumor induction in mice after inoculation of EAC cells irradiated at a dose of 30 Gy was 80%, 60 Gy-60%, 90 Gy-25%, and 120 Gy-10%; at irradiation at a dose of 150 Gy, no tumors appeared during the entire observation period. Thus, we determined the dose of proton radiation required to eliminate tumor cells and/or signaling factors that can lead to the induction of tumor growth of EAC in mice.


Subject(s)
Carcinoma, Ehrlich Tumor , Mice , Animals , Carcinoma, Ehrlich Tumor/radiotherapy , Carcinoma, Ehrlich Tumor/pathology , Protons
4.
Dokl Biochem Biophys ; 513(Suppl 1): S30-S35, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38472666

ABSTRACT

The effect of carbon ions (12C) with the energy of 400 MeV/nucleon on the dynamics of induction and growth rate of solid tumors in mice under irradiation of Ehrlich ascites carcinoma cells (EAC) ex vivo at doses of 5-30 Gy relative to the action of equally effective doses of X-ray radiation was studied. The dynamics of tumor induction under the action of 12C and X-rays had a similar character and depended on the dose during 3 months of observation. The value of the latent period, both when irradiating cells with 12C and X-ray, increased with increasing dose, and the interval for tumor induction decreased. The rate of tumor growth after ex vivo irradiation of EAC cells was independent of either dose or type of radiation. The dose at which EAC tumors are not induced within 90 days was 30 Gy for carbon ions and 60 Gy for X-rays. The value of the relative biological effectiveness of carbon ions, calculated from an equally effective dose of 50% probability of tumors, was 2.59.


Subject(s)
Carcinoma, Ehrlich Tumor , Neoplasms , Animals , Mice , X-Rays , Ascites , Carbon , Carcinoma, Ehrlich Tumor/radiotherapy , Dose-Response Relationship, Radiation
5.
Appl Radiat Isot ; 187: 110305, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35688071

ABSTRACT

BACKGROUND: This work was carried out to compare the modifying roles of ascorbic and metformin during Ehrlich (ESC) tumor-bearing mice irradiation. METHODS: Fifty Swiss albino male mice were segmented into seven groups, including one control group and six Ehrlich induced tumors treated with ascorbic, ascorbic plus radiation, metformin, metformin plus radiation, and radiation only. Many tests, including behavioral, biochemical, immunohistochemistry, gene expression, DNA fragmentation, oxidative stress markers, and EPR, were performed to interrogate the modifying effects on tumor and liver tissues. RESULTS: Remarkable apoptosis was found in metformin irradiated animals compared to irradiated ascorbic counterparts. The irradiated metformin mice showed the greatest reduction in PCNA. There was a significant reduction of DNA fragmentation in the liver tissues of the irradiated metformin group. Irradiated metformin and irradiated ascorbic acid animals showed a reduced signal of ERK as well as c-Fos genes. There was a tendency of metformin and metformin irradiated animals to reduce MDA levels in liver tissues. ESC-bearing mice treated with ascorbic or metformin showed an improvement in the spontaneous alternation percentage (SAP%) and improved short-term memory. There was also an improvement in long memory tests. CONCLUSIONS: The study added more preclinical evidence on the utility of metformin in cancer treatment during radiotherapy. Metformin was shown to reduce lipid peroxidation in irradiated healthy tissues, increase tumor cytotoxicity, downregulate critical pathways involved in tumor progression and proliferation, and enhance tumor apoptosis. Controlled clinical trials using metformin are highly warranted.


Subject(s)
Carcinoma, Ehrlich Tumor , Metformin , Neoplasms , Animals , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Lipid Peroxidation/radiation effects , Metformin/therapeutic use , Mice
6.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502247

ABSTRACT

Previously, we showed that a nitric oxide synthase (NOS) inhibitor, compound T1023, induces transient hypoxia and prevents acute radiation syndrome (ARS) in mice. Significant efficacy (according to various tests, dose modifying factor (DMF)-1.6-1.9 against H-ARS/G-ARS) and safety in radioprotective doses (1/5-1/4 LD10) became the reason for testing its ability to prevent complications of tumor radiation therapy (RT). Research methods included studying T1023 effects on skin acute radiation reactions (RSR) in rats and mice without tumors and in tumor-bearing animals. The effects were evaluated using clinical, morphological and histological techniques as well as RTOG classification. T1023 administration prior to irradiation significantly limited the severity of acute RSR. This was due to a decrease in radiation alteration of the skin and underlying tissues, and the preservation of the functional activity of cell populations that are critical in the pathogenesis of radiation burn. The DMF values for T1023 for skin protection were 1.4-1.7. Moreover, its radioprotective effect was fully selective to normal tissues in RT models of solid tumors-T1023 reduced the severity of acute RSR and did not modify the antitumor effects of γ-radiation. The results indicate that T1023 can selectively protect the non-malignant tissues against γ-radiation due to hypoxic mechanism of action and potentiate opportunities of NOS inhibitors in RT complications prevention.


Subject(s)
Carcinoma, Ehrlich Tumor/radiotherapy , Gamma Rays/adverse effects , Nitric Oxide Synthase/antagonists & inhibitors , Radiation-Protective Agents/pharmacology , Radiodermatitis/drug therapy , Sarcoma, Experimental/radiotherapy , Thiourea/analogs & derivatives , Animals , Carcinoma, Ehrlich Tumor/pathology , Female , Male , Mice , Mice, Inbred ICR , Radiation Protection/methods , Radiodermatitis/etiology , Radiodermatitis/pathology , Rats , Rats, Sprague-Dawley , Sarcoma, Experimental/pathology , Thiourea/pharmacology
7.
Dokl Biochem Biophys ; 498(1): 159-164, 2021 May.
Article in English | MEDLINE | ID: mdl-34189642

ABSTRACT

The combined effect of the irradiation with a proton pencil scanning beam (PBS) at a total dose of 80 Gy and neutron radiation at a dose of 5 Gy on the growth of solid Ehrlich ascites carcinoma (EAC) and the remote effects in tumor-bearing mice was studied. Combined irradiation of mice with neutrons before and after irradiation with PBS, as well as irradiation only with PBS, effectively suppressed the growth of solid EAC within 1 month. In terms of the frequency and severity of radiation-induced skin reactions of mice observed 15-40 days after therapy, neutron irradiation after the irradiation with PBS showed better values of these parameters as compared to only PBS; however, exposure to neutrons before PBS was more damaging as compared to the other two options. It was also shown that the tumor relapse rate in the groups of animals with combined irradiation was higher, and the total lifespan was lower than the group of mice irradiated with PBS alone.


Subject(s)
Carcinoma, Ehrlich Tumor/radiotherapy , Mammary Neoplasms, Experimental/radiotherapy , Neutrons/therapeutic use , Proton Therapy , Animals , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Female , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Survival Rate , Treatment Outcome
8.
J Cancer Res Ther ; 17(1): 211-217, 2021.
Article in English | MEDLINE | ID: mdl-33723157

ABSTRACT

PURPOSE: The purpose of this study is to evaluate the effects of Vitamin E (VE) on the immune system and tumor growth during radiotherapy (RT) in mice model. METHODS: C57BL/6NCrSlc mice were randomly distributed in four groups (control, VE alone, RT alone, and VE + RT). In the VE and VE + RT groups, VE was administered in the diet at 500 mg/kg. Radiation was delivered at 2 Gy in a single fraction on the whole body or at 6 Gy in three fractions locally in the RT and VE + RT groups. Changes in leukocytes and T lymphocytes were counted and compared between the four groups. To evaluate the effects on tumor growth, Ehrlich carcinoma cells were injected into the thighs of mice, and tumor volumes and growth inhibition rates were compared. RESULTS: The number of leukocytes was increased in the VE group compared with that in the control group. The magnitude of leukocyte recovery after RT was also increased by VE. This change was affected largely by alterations in lymphocytes and monocytes rather than that in granulocytes. Both CD4+ and CD8+ T lymphocytes were positively affected by VE. The tumor growth was inhibited not only by RT but also by VE alone. If RT was delivered with VE, tumor growth was markedly inhibited. CONCLUSION: VE could increase the number of leukocytes, primarily lymphocytes, even after RT was delivered. VE also inhibited the tumor growth in addition to RT. Thus, VE may be a useful radioprotective supplement in radiotherapy without inducing tumor growth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Ehrlich Tumor/drug therapy , Radiotherapy/methods , Vitamin E/pharmacology , Animals , Antioxidants/pharmacology , Carcinoma, Ehrlich Tumor/immunology , Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/radiotherapy , Combined Modality Therapy , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Treatment Outcome
9.
Integr Cancer Ther ; 19: 1534735420950468, 2020.
Article in English | MEDLINE | ID: mdl-32783540

ABSTRACT

This study hypothesizes that, bromelain (BL) acts as radiosensitizer of tumor cells and that it protects normal cells from radiation effects. In vitro and in vivo studies have been carried out to prove that assumption. In vitro MTT cell proliferation assay has shown that the irradiated Ehrlich ascites carcinoma (EAC) cell line could be sensitized by BL pretreatment. In vivo: animals were randomly divided into 5 groups, Group 1: control (PBS i.p for 10 days), Group 2: Ehrlich solid tumor (EST) bearing mice, Group 3: EST + γ-radiation (fractionated dose, 1 Gy × 5), Group 4: EST + BL (6 mg/kg, i.p), daily for 10 days, Group 5: EST + BL for 10 days followed by γ-irradiation (1 Gy × 5). The size and weight of tumors in gamma-irradiated EST bearing mice treated with BL decreased significantly with a significant amelioration in the histopathological examination. Besides, BL mitigated the effect of γ-irradiation on the liver relative gene expression of poly ADP ribose polymerase-1 (PARP1), nuclear factor kappa activated B cells (NF-κB), and peroxisome proliferator-activated receptor α (PPAR-α), and it restored liver function via amelioration of paraoxonase1 (PON1) activity, reactive oxygen species (ROS) content, lipid peroxidation (LPO) and serum aspartate transaminase (AST), alanine transaminase (ALT), and albumin (ALB). It is concluded that BL can be considered as a radio-sensitizer and radio-protector, suggesting a possible role in reducing radiation exposure dose during radiotherapy.


Subject(s)
Ananas , Carcinoma, Ehrlich Tumor , Animals , Bromelains/pharmacology , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Gamma Rays , Lipid Peroxidation , Mice , Plant Extracts
10.
Integr Cancer Ther ; 19: 1534735420944476, 2020.
Article in English | MEDLINE | ID: mdl-32735464

ABSTRACT

Pathological angiogenesis and apoptosis evasion are common hallmarks of cancer. The present work was an endeavor to evaluate the influence of bee venom (BV) or its major constituent melittin (MEL) as antiapoptotic and angiogenic regulator modifier on the tumor growth and the cell sensitivity to ionizing radiation targeting the improvement of cancer therapeutic protocols. BV (0.56 mg/kg/day) and MEL (500 µg/kg body weight/day) were injected intraperitoneally to mice bearing 1 cm3 solid tumor of Ehrlich ascites carcinoma (EAC) for 21 consecutive days. Mice were whole-body exposed to 1 Gray (Gy) of γ-radiation (2 fractionated doses). Treatment with BV or MEL markedly suppresses the proliferation of tumor in EAC mice. The concentrations of m-RNA for angiogenic factors (TNF-α, VEGF) as well as MMPs 2 and 9 activities and NO concentration were significantly decreased, combined with improvements in apoptotic regulators (caspase-3 activity) and normal cells redox tone (catalase and free radicals content) compared with EAC mice. Moreover, the histopathological investigation confirms the improvement exerted by BV or MEL in the EAC mice group or EAC + R group. Exposure to γ-radiation sustained the modulatory effect of BV on tumor when compared with EAC + BV mice. Convincingly, the role of BV or MEL as a natural antiangiogenic in the biological sequelae after radiation exposure is verified. Hence, BV and its major constituent MEL might represent a potential therapeutic strategy for increasing the radiation response of solid tumors.


Subject(s)
Bee Venoms , Carcinoma, Ehrlich Tumor , Carcinoma , Animals , Apoptosis , Bee Venoms/pharmacology , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Melitten/pharmacology , Mice , Neovascularization, Pathologic/drug therapy
11.
Appl Radiat Isot ; 166: 109369, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32828009

ABSTRACT

Recently, pyrrolizine derivatives have been reported to possess numerous anticancer activities. In a previous study, (EZ)-6-((4-chlorobenzylidene)-amino)-7-cyano-N-(p-tolyl)-2,3-dihydro-1H-pyrrolizine carboxamide (EZPCA) compound was synthesized and the cytotoxic activity of EZPCA toward COX-2 enzyme (overexpressed in cancer cells) was reported. In order to assess the suitability of this compound as a promising pilot structure for in vivo applications, EZPCA was radiolabeled with radioiodine-131 (131I) and various factors affecting radiolabeling process were studied. Quality control studies of [131I]iodo-EZPCA were performed using paper chromatography and HPLC was used as a co-chromatographic technique for confirming the radiochemical yield. Biodistribution studies of [131I]iodo-EZPCA were undertaken in normal and tumor bearing mice. The radiochemical yield percentage of [131I]iodo-EZPCA was 94.20 ± 0.12%. The biodistribution results showed evident tumor uptake of [131I]iodo-EZPCA with promising target/non-target (T/NT) ratios. As a conclusion, these data suggest that [131I]iodo-EZPCA had high binding efficiency, high tumor uptake and sufficient stability to be used be used in diagnostic studies.


Subject(s)
Carcinoma, Ehrlich Tumor/radiotherapy , Iodine Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Carcinoma, Ehrlich Tumor/metabolism , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Female , HCT116 Cells , Hep G2 Cells , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Iodine Radioisotopes/chemistry , Iodine Radioisotopes/pharmacokinetics , Isotope Labeling , MCF-7 Cells , Mice , Molecular Docking Simulation , Pyrrolizidine Alkaloids/chemistry , Pyrrolizidine Alkaloids/pharmacokinetics , Pyrrolizidine Alkaloids/therapeutic use , Radiochemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics
12.
J Radiat Res ; 60(6): 747-758, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31504707

ABSTRACT

This study examines the ability of arabinoxylan rice bran (MGN-3/Biobran) to enhance the anti-cancer effects of fractionated X-ray irradiation of Ehrlich solid tumor-bearing mice. Swiss albino mice bearing tumors were exposed to the following: (i) Biobran treatment (40 mg/kg/day, intraperitoneal injections) beginning on day 11 post-tumor cell inoculation until day 30; (ii) ionizing radiation (Rad) 2 Gy at three consecutive doses on days 12, 14 and 16; or (iii) Biobran + Rad. Final tumor weight was suppressed by 46% for Biobran, 31% for Rad and 57% for the combined treatment (Biobran + Rad) relative to control untreated mice. Biobran and Rad also arrested the hypodiploid cells in the sub-G1-phase, signifying apoptosis by +102% and +85%, respectively, while the combined treatment induced apoptosis by +123%, with similar results in the degree of DNA fragmentation. Furthermore, Biobran + Rad upregulated the relative gene expression and protein level of p53 and Bax in tumor cells, down-regulated Bcl-2 expression, and increased the Bax/Bcl-2 ratio and caspase-3 activity, with the combined treatment greater than for either treatment alone. Additionally, the combined treatment modulated the decrease in body weight, the increase in liver and spleen weight, and the elevation of liver enzymes aspartate aminotransferase, alanine aminotransferase and gamma-glutamyl transferase to be within normal values. We conclude that Biobran enhances radiation therapy-induced tumor regression by potentiating apoptosis and minimizing toxicities related to radiation therapy, suggesting that Biobran may be useful in human cancer patients undergoing radiotherapy and warranting clinical trials.


Subject(s)
Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Xylans/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Body Weight/drug effects , Body Weight/radiation effects , Carcinoma, Ehrlich Tumor/genetics , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , DNA Damage , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Female , Gene Expression Regulation , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver/radiation effects , Mice , Organ Size/drug effects , Organ Size/radiation effects , Spleen/drug effects , Spleen/pathology , Spleen/radiation effects , Tumor Burden/drug effects , Tumor Burden/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , X-Rays , Xylans/pharmacology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Tumour Biol ; 40(3): 1010428317749676, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29587600

ABSTRACT

Guided treatments with nanoparticles and radiotherapy are a new approach in cancer therapy. This study evaluated the beneficial antitumor effects of γ-radiation together with gallium nanoparticles against solid Ehrlich carcinoma in female mice. Gallium nanoparticles were biologically synthesized using Lactobacillus helveticus cells. Transmission electron microscopy showed gallium nanoparticles with size range of 8-20 nm. In vitro study of gallium nanoparticles on MCF-7 revealed IC50 of 8.0 µg. Gallium nanoparticles (0.1 mg/kg body weight) were injected intraperitoneally daily on the seventh day of Ehrlich carcinoma cells inoculation. Whole-body γ-radiation was carried out at a single dose of 0.25 Gy on eighth day after tumor inoculation. Biochemical analysis showed that solid Ehrlich carcinoma induced a significant increase in alanine aminotransferase activity and creatinine level in serum, calcium, and iron concentrations in liver tissue compared to normal control. Treatment of Ehrlich carcinoma-bearing mice with gallium nanoparticles and/or low dose of γ-radiation exposure significantly reduced tumor volume, decreased alanine aminotransferase and creatinine levels in serum, increased lipid peroxidation, and decreased glutathione content as well as calcium and iron concentrations in liver and tumor tissues with intense DNA fragmentation accompanied compared to untreated tumor cells. Moreover, mitochondria in the treated groups displayed a significant increase in Na+/K+-ATPase, complexes II and III with significant reduction in CYP450 gene expression, which may indicate a synergistic effect of gallium nanoparticles and/or low dose of γ-radiation combination against Ehrlich carcinoma injury, and this results were well appreciated with the histopathological findings in the tumor tissue. We conclude that combined treatment of gallium nanoparticles and low dose of gamma-radiation resulted in suppressive induction of cytotoxic effects on cancer cells.


Subject(s)
Carcinoma, Ehrlich Tumor/radiotherapy , Gallium/therapeutic use , Gamma Rays/therapeutic use , Metal Nanoparticles/therapeutic use , Alanine Transaminase/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Creatinine/blood , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA Fragmentation/radiation effects , Female , Glutathione/metabolism , Humans , Iron/metabolism , Lipid Peroxidation/radiation effects , Liver/metabolism , MCF-7 Cells , Mice , Mitochondria/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Whole-Body Irradiation
14.
Tumour Biol ; 39(10): 1010428317728480, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29022496

ABSTRACT

Considerable attention has been paid to the introduction of novel naturally occurring plant-derived radiosensitizer compounds in order to augment the radiation efficacy and improve the treatment outcome of different tumors. This study was therefore undertaken to evaluate the antitumor, antiangiogeneic, and synergistic radiosensitizing effects of apigenin, a dietary flavonoid, and/or cryptotanshinone, a terpenoid isolated from the roots of Salvia miltiorrhiza, against the growth of solid Ehrlich carcinoma in female mice. Apigenin (50 mg/kg body weight) and/or cryptotanshinone (40 mg/kg body weight) was intraperitoneally (i.p.) injected into non-irradiated or γ-irradiated (6.5 Gy whole-body γ-irradiation) solid Ehrlich carcinoma-bearing mice for 30 consecutive days. Investigations included molecular targets involved in proliferation, inflammation, angiogenesis, and tumor invasiveness. Treatment with apigenin and/or cryptotanshinone significantly suppressed the growth of solid Ehrlich carcinoma tumors and demonstrated a synergistic radiosensitizing efficacy together with γ-irradiation. These effects were achieved through downregulating the expression of angiogenic and lymphangiogenic regulators, including signal transducer and activator of transcription 3, vascular endothelial growth factor C, and tumor necrosis factor alpha, suppressing matrix metalloproteinase-2 and -9 activities, which play a key role in tumor invasion and metastasis, and enhancing apoptosis via inducing cleaved caspase-3 and granzyme B levels. Histological findings of solid Ehrlich carcinoma tumors verified the recorded data. In conclusion, a synergistic radiosensitizing efficacy for apigenin and cryptotanshinone was demonstrated against Ehrlich carcinoma in the current in vivo murine model, representing therefore a potential therapeutic strategy for increasing the radiation response of solid tumors.


Subject(s)
Apigenin/administration & dosage , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Radiation-Sensitizing Agents/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Ehrlich Tumor/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Disease Models, Animal , Female , Gamma Rays , Humans , Mice , Phenanthrenes/administration & dosage , Whole-Body Irradiation
15.
Tumour Biol ; 37(2): 1825-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26318299

ABSTRACT

Combining chemotherapy with radiotherapy represents a key oncology strategy for a more comprehensive attack toward cancers and improves treatment outcome for various solid tumor malignancies. The present study aims to evaluate the synergistic antitumor effect of γ-radiation together with gallium trichloride (GaCl3) and/or doxorubicin (DOX) against solid Ehrlich carcinoma (EC) in female mice. GaCl3 (300 mg/kg body weight (b.w.)) was administered by gavages daily on the seventh day after tumor inoculation, while the cytotoxic drug DOX (4 mg/kg b.w.) was administered intraperitoneally once a week. Whole-body γ-radiation was carried out at a dose 2 Gy once a week. Biochemical analysis showed that solid EC induced a significant increase in malondialdehyde (MDA) content with a significant decrease in the antioxidant state (glutathione peroxidase (GPx) and catalase (CAT) activities) and depleted serum iron concentration compared to normal control. Moreover, a significant increase was observed in calcium level and caspase-3 concentrations in both serum and tumor homogenate respectively associated with a significant alteration in heart, liver, and kidney functions, as compared to control. Treatment of EC-bearing mice with GaCl3and/or DOX combined with γ-radiation exposure significantly reduced tumor volume and displayed a significant improvement in most studied markers which may indicate a synergistic effect of this combination against organ dysfunction and cellular injury. The histopathologically investigation showed that treatment of animals bearing EC with GaCl3and/or DOX with γ-radiation exposure showed shrinkage in tumor lesions and wide zones of apoptotic cells with signs of regenerations. It was concluded that the combination of GaCl3and/or DOX with γ-radiation exposure resulted in super-additive cytotoxic effects on treatment of cancer cells.


Subject(s)
Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/radiotherapy , Doxorubicin/pharmacology , Gallium/pharmacology , Gamma Rays/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Carcinoma, Ehrlich Tumor/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Female , Glutathione Peroxidase/metabolism , Malondialdehyde/metabolism , Mice
16.
Article in Russian | MEDLINE | ID: mdl-26595969

ABSTRACT

This paper summarizes the results of the application of thr magnetic fields for the treatment of experimental tumours, such as sarcoma M-1, alveolar liver cancer PC-1, and Erlich's carcinoma. The evidence of the anti-tumour action of both strong (1200 mTI) and weak (5 to 100 mTI) magnetic fields has been obtained. The author describes the modulating effect of the magnetic fields on the anti-tumour potency of photodynamic therapy and chemotherapy. The data concerning the impact of ferromagnetic hyperthermal therapy on the tumour growth and the survival rate among the tumour-bearing animals are presented.


Subject(s)
Carcinoma, Ehrlich Tumor/radiotherapy , Liver Neoplasms/radiotherapy , Magnetic Field Therapy , Sarcoma, Experimental/radiotherapy , Animals , Carcinoma, Ehrlich Tumor/drug therapy , Liver Neoplasms/drug therapy , Photochemotherapy , Rats , Sarcoma, Experimental/drug therapy
17.
Mol Pharmacol ; 88(4): 768-78, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26240287

ABSTRACT

Radiotherapy, a therapeutic modality of cancer treatment, nonselectively damages normal tissues as well as tumor tissues. The search is ongoing for therapeutic agents that selectively reduce radiation-induced normal tissue injury without reducing tumoricidal effect, thereby increasing the therapeutic ratio of radiation therapy. Our laboratory established 5-(4-methylpiperazin-1-yl)-2-[2'-(3,4-dimethoxyphenyl)-5'benzimidazolyl] benzimidazole (DMA) as noncytotoxic radioprotector in mammalian cells. DMA showed an excellent radioprotection in mice at single nontoxic oral dose by a dose-reduction factor of 1.28. An oxygen radical absorbing capacity assay confirmed its free-radical quenching ability. Single bolus dose and 28-days of repeated administration of DMA in mice for toxicity studies determined an LD50 of >2000 mg/kg body weight (bw) and 225 mg/kg bw, respectively, suggesting DMA is safe. Histopathology, biochemical parameters, and relative organ weight analysis revealed insignificant changes in the DMA-treated animals. The pharmacokinetic study of DMA at oral and intravenous doses showed its C(max) = 1 hour, bioavailability of 8.84%, elimination half-life of 4 hours, and an enterohepatic recirculation. Biodistribution study in mice with Ehrlich ascites tumors showed that (99m)Tc-DMA achieved its highest concentration in 1 hour and was retained up to 4 hours in the lungs, liver, kidneys, and spleen, and in a low concentration in the tumor, a solicited property of any radioprotector to protect normal cells over cancerous cells. We observed that the single-dose treatment of tumor-bearing mice with DMA 2 hours before 8 Gy total body irradiation showed an impressive rescue of radiation-induced morbidity in terms of weight loss and mortality without a change in tumor response.


Subject(s)
Benzimidazoles/pharmacokinetics , Benzimidazoles/toxicity , Piperazines/pharmacokinetics , Piperazines/toxicity , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/toxicity , Animals , Benzimidazoles/metabolism , Bisbenzimidazole/metabolism , Bisbenzimidazole/pharmacokinetics , Bisbenzimidazole/toxicity , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/radiotherapy , Dose-Response Relationship, Radiation , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Piperazines/metabolism , Radiation-Protective Agents/metabolism , Survival Rate/trends , Tissue Distribution/drug effects , Tissue Distribution/physiology
18.
PLoS One ; 9(9): e108131, 2014.
Article in English | MEDLINE | ID: mdl-25248151

ABSTRACT

Two-deoxy-D-glucose (2-DG), an inhibitor of glycolysis differentially enhances the radiation and chemotherapeutic drug induced cell death in cancer cells in vitro, while the local tumor control (tumor regression) following systemic administration of 2-DG and focal irradiation of the tumor results in both complete (cure) and partial response in a fraction of the tumor bearing mice. In the present studies, we investigated the effects of systemically administered 2-DG and focal irradiation of the tumor on the immune system in Ehrlich ascites tumor (EAT) bearing Strain "A" mice. Markers of different immune cells were analyzed by immune-flow cytometry and secretary cytokines by ELISA, besides monitoring tumor growth. Increase in the expression of innate (NK and monocytes) and adaptive CD4+cells, and a decrease in B cells (CD19) have been observed after the combined treatment, suggestive of activation of anti-tumor immune response. Interestingly, immature dendritic cells were found to be down regulated, while their functional markers CD86 and MHC II were up regulated in the remaining dendritic cells following the combination treatment. Similarly, decrease in the CD4(+) naïve cells with concomitant increase in activated CD4+ cells corroborated the immune activation. Further, a shift from Th2 and Th17 to Th1 besides a decrease in inflammatory cytokines was also observed in the animals showing complete response (cure; tumor free survival). This shift was also complimented by respective antibody class switching followed by the combined treatment. The immune activation or alteration in the homeostasis favoring antitumor immune response may be due to depletion in T regulatory cells (CD4(+)CD25(+)FoxP3(+)). Altogether, these results suggest that early differential immune activation is responsible for the heterogenous response to the combined treatment. Taken together, these studies for the first time provided insight into the additional mechanisms underlying radio-sensitization by 2-DG in vivo by unraveling its potential as an immune-modulator besides direct effects on the tumor.


Subject(s)
Carcinoma, Ehrlich Tumor/drug therapy , Deoxyglucose/pharmacology , Immunity/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Ehrlich Tumor/immunology , Carcinoma, Ehrlich Tumor/radiotherapy , Cytokines/metabolism , Deoxyglucose/therapeutic use , Glycolysis/drug effects , Lymphocyte Count , Male , Mice , Radiation-Sensitizing Agents/therapeutic use
19.
Int J Radiat Biol ; 89(8): 602-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23484905

ABSTRACT

PURPOSE: To test the participation of fatty acids (FA) in antitumor effects of extremely high-frequency electromagnetic radiation (EHF EMR), the changes in the FA composition in the thymus, liver, blood plasma, muscle tissue, and tumor tissue in mice with Ehrlich solid carcinoma exposed to EHF EMR were studied. MATERIALS AND METHODS: Normal and tumor-bearing mice were exposed to EHF EMR with effective parameters (42.2 GHz, 0.1 mW/cm2, 20 min daily during five consecutive days beginning the first day after the inoculation of tumor cells). Fatty acid composition of various organs and tissues of mice were determined using a gas chromatography. RESULTS: It was shown that the exposure of normal mice to EHF EMR or tumor growth significantly increased the content of monounsaturated FA (MUFA) and decreased the content of polyunsaturated FA (PUFA) in all tissues examined. Exposure of tumor-bearing mice to EHF EMR led to the recovery of FA composition in thymocytes to the state that is typical for normal animals. In other tissues of tumor-bearing mice, the exposure to EHF EMR did not induce considerable changes that would be significantly distinguished between disturbances caused by EHF EMR exposure or tumor growth separately. In tumor tissue which is characterized by elevated level of MUFA, the exposure to EHF EMR significantly decreased the summary content of MUFA and increased the summary content of PUFA. CONCLUSIONS: The recovery of the FA composition in thymocytes and the modification of the FA composition in the tumor under the influence of EHF EMR on tumor-bearing animals may have crucial importance for elucidating the mechanisms of antitumor effects of the electromagnetic radiation.


Subject(s)
Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/radiotherapy , Electromagnetic Radiation , Fatty Acids/chemistry , Thymocytes/metabolism , Thymocytes/radiation effects , Animals , Cell Proliferation/radiation effects , Male , Mice , Mice, Inbred BALB C , Organ Specificity
20.
Biofizika ; 58(5): 897-903, 2013.
Article in Russian | MEDLINE | ID: mdl-25481959

ABSTRACT

The effect of low-dose-rate red and near-infrared radiations from the matrix of light emitted diode (650 nm and 850 nm) and a He-Ne laser (633 nm) on activation of the reserve of a natural defense system in the mice exposed to radiation in vivo was studied by the level of reactive oxygen species (ROS) production in blood cells, the induction of cytogenetic adaptive response in bone marrow cells, thymus and spleen, and the rate of Ehrlich ascites carcinoma growth in a solid form. As a positive control animals were irradiated with X-rays by the scheme of the radiation-induced adaptive response (0.1 Gy + 1.5 Gy). The levels of ROS production was assessed in whole blood by luminol-dependent chemiluminescence, of cytogenetic damage--by the "micronucleus test" in the bone marrow, the weight of the thymus and spleen--by index of organ, and the rate of tumor growth--according to its size for 30 days after inoculation. Adaptogenic and anticarcinogenic effects of studied radiations were revealed. The values of these effects were not different from those in animals pre-irradiated with the X-rays. The relationship between the level of ROS production and adaptive response induction in the mice under the influence of non-ionizing radiation was first ascertained. The experimental data obtained may indicate a similar mechanism of induction of protective responses to ionizing and non-ionizing radiations in mice in vivo.


Subject(s)
Bone Marrow Cells/metabolism , Reactive Oxygen Species/metabolism , Spleen/metabolism , Animals , Bone Marrow Cells/radiation effects , Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/radiotherapy , Dose-Response Relationship, Radiation , Infrared Rays , Mice , Reactive Oxygen Species/radiation effects , Spleen/radiation effects , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...