Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
PLoS One ; 19(3): e0298437, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38498459

RESUMEN

Ionizing radiation (IR) and oncolytic viruses are both used to treat cancer, and the effectiveness of both agents depends upon stimulating an immune response against the tumor. In this study we tested whether combining image guided ionizing radiation (IG-IR) with an oncolytic vaccinia virus (VACV) could yield a better therapeutic response than either treatment alone. ΔF4LΔJ2R VACV grew well on irradiated human and mouse breast cancer cells, and the virus can be combined with 4 or 8 Gy of IR to kill cells in an additive or weakly synergistic manner. To test efficacy in vivo we used immune competent mice bearing orthotopic TUBO mammary tumors. IG-IR worked well with 10 Gy producing 80% complete responses, but this was halved when the tumors were treated with VACV starting 2 days after IG-IR. VACV monotherapy was ineffective in this model. The antagonism was time dependent as waiting for 21 days after IG-IR eliminated the inhibitory effect but without yielding any further benefits over IR alone. In irradiated tumors, VACV replication was also lower, suggesting that irradiation created an environment that did not support infection as well in vivo as in vitro. A study of how four different treatment regimens affected the immune composition of the tumor microenvironment showed that treating irradiated tumors with VACV altered the immunological profiles in tumors exposed to IR or VACV alone. We detected more PD-1 and PD-L1 expression in tumors exposed to IR+VACV but adding an αPD-1 antibody to the protocol did not change the way VACV interferes with IG-IR therapy. VACV encodes many immunosuppressive gene products that may interfere with the ability of radiotherapy to induce an effective anti-tumor immune response through the release of danger-associated molecular patterns. These data suggest that infecting irradiated tumors with VACV, too soon after exposure, may interfere in the innate and linked adaptive immune responses that are triggered by radiotherapy to achieve a beneficial impact.


Asunto(s)
Neoplasias Mamarias Animales , Viroterapia Oncolítica , Virus Oncolíticos , Radioterapia Guiada por Imagen , Vaccinia , Humanos , Animales , Ratones , Virus Vaccinia/genética , Virus Oncolíticos/genética , Neoplasias Mamarias Animales/radioterapia , Inmunoterapia , Viroterapia Oncolítica/métodos , Microambiente Tumoral
2.
Clin Orthop Relat Res ; 479(1): 163-176, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32858719

RESUMEN

BACKGROUND: Recent advances in multidisciplinary treatments for various cancers have extended the survival period of patients with spinal metastases. Radiotherapy has been widely used to treat spinal metastases; nevertheless, long-term survivors sometimes undergo more surgical intervention after radiotherapy because of local tumor relapse. Generally, intradural invasion of a spinal tumor seldom occurs because the dura mater serves as a tissue barrier against tumor infiltration. However, after radiation exposure, some spinal tumors invade the dura mater, resulting in leptomeningeal dissemination, intraoperative dural injury, or postoperative local recurrence. The mechanisms of how radiation might affect the dura have not been well-studied. QUESTIONS/PURPOSES: To investigate how radiation affects the spinal meninges, we asked: (1) What is the effect of irradiation on the meningeal barrier's ability to protect against carcinoma infiltration? (2) What is the effect of irradiation on the meningeal barrier's ability to protect against sarcoma infiltration? (3) What is the effect of irradiation on dural microstructure observed by scanning electron microscopy (SEM)? (4) What is the effect of irradiation on dural microstructure observed by transmission electron microscopy (TEM)? METHODS: Eighty-four 10-week-old female ddY mice were randomly divided into eight groups: mouse mammary tumor (MMT) implantation 6 weeks after 0-Gy irradiation (nonirradiation) (n = 11), MMT implantation 6 weeks after 20-Gy irradiation (n = 10), MMT implantation 12 weeks after nonirradiation (n = 10), MMT implantation 12 weeks after 20-Gy irradiation (n = 11), mouse osteosarcoma (LM8) implantation 6 weeks after nonirradiation (n = 11), LM8 implantation 6 weeks after 20-Gy irradiation (n = 11), LM8 implantation 12 weeks after nonirradiation (n = 10), and LM8 implantation 12 weeks after 20-Gy irradiation (n = 10); female mice were used for a mammary tumor metastasis model and ddY mice, a closed-colony mice with genetic diversity, were selected to represent interhuman diversity. Mice in each group underwent surgery to generate a tumor-induced spinal cord compression model at either 6 weeks or 12 weeks after irradiation to assess changes in the meningeal barrier's ability to protect against tumor infiltration. During surgery, the mice were implanted with MMT (representative of a carcinoma) or LM8 tumor. When the mice became paraplegic because of spinal cord compression by the growing implanted tumor, they were euthanized and evaluated histologically. Four mice died from anesthesia and 10 mice per group were euthanized (MMT-implanted groups: MMT implantation occurred 6 weeks after nonirradiation [n = 10], 6 weeks after irradiation [n = 10], 12 weeks after nonirradiation [n = 10], and 12 weeks after irradiation [n = 10]; LM8-implanted groups: LM8 implantation performed 6 weeks after nonirradiation [n = 10], 6 weeks after irradiation [n = 10], 12 weeks after nonirradiation [n = 10], and 12 weeks after irradiation [n = 10]); 80 mice were evaluated. The spines of the euthanized mice were harvested; hematoxylin and eosin staining and Masson's trichrome staining slides were prepared for histologic assessment of each specimen. In the histologic assessment, intradural invasion of the implanted tumor was graded in each group by three observers blinded to the type of tumor, presence of irradiation, and the timing of the surgery. Grade 0 was defined as no intradural invasion with intact dura mater, Grade 1 was defined as intradural invasion with linear dural continuity, and Grade 2 was defined as intradural invasion with disruption of the dural continuity. Additionally, we euthanized 12 mice for a microstructural analysis of dura mater changes by two observers blinded to the presence of irradiation. Six mice (three mice in the 12 weeks after nonirradiation group and three mice in the 12 weeks after 20-Gy irradiation group) were quantitatively analyzed for defects on the dural surface with SEM. The other six mice (three mice in the 12 weeks after nonirradiation group and three mice in the 12 weeks after 20-Gy irradiation group) were analyzed for layer structure of collagen fibers constituting dura mater by TEM. In the SEM assessment, the number and size of defects on the dural surface on images (200 µm × 300 µm) at low magnification (× 2680) were evaluated. A total of 12 images (two per mouse) were evaluated for this assessment. The days from surgery to paraplegia were compared between each of the tumor groups using the Kruskal-Wallis test. The scores of intradural tumor invasion grades and the number of defects on dural surface per SEM image were compared between irradiation group and nonirradiation group using the Mann-Whitney U test. Interobserver reliabilities of assessing intradural tumor invasion grades and the number of dural defects on the dural surface were analyzed using Fleiss'κ coefficient. P values < 0.05 were considered statistically significant. RESULTS: There was no difference in the median (range) time to paraplegia among the MMT implantation 6 weeks after nonirradiation group, the 6 weeks after irradiation group, the 12 weeks after nonirradiation group, and the 12 weeks after irradiation group (16 days [14 to 17] versus 14 days [12 to 18] versus 16 days [14 to 17] versus 14 days [12 to 15]; χ2 = 4.7; p = 0.19). There was also no difference in the intradural invasion score between the MMT implantation 6 weeks after irradiation group and the 6 weeks after nonirradiation group (8 of 10 Grade 0 and 2 of 10 Grade 1 versus 10 of 10 Grade 0; p = 0.17). On the other hand, there was a higher intradural invasion score in the MMT implantation 12 weeks after irradiation group than the 12 weeks after nonirradiation group (5 of 10 Grade 0, 3 of 10 Grade 1 and 2 of 10 Grade 2 versus 10 of 10 Grade 0; p = 0.02). Interobserver reliability of assessing intradural tumor invasion grades in the MMT-implanted group was 0.94. There was no difference in the median (range) time to paraplegia among in the LM8 implantation 6 weeks after nonirradiation group, the 6 weeks after irradiation group, the 12 weeks after nonirradiation group, and the 12 weeks after irradiation group (12 days [9 to 13] versus 10 days [8 to 13] versus 11 days [8 to 13] versus 9 days [6 to 12]; χ2 = 2.4; p = 0.50). There was also no difference in the intradural invasion score between the LM8 implantation 6 weeks after irradiation group and the 6 weeks after nonirradiation group (7 of 10 Grade 0, 1 of 10 Grade 1 and 2 of 10 Grade 2 versus 8 of 10 Grade 0 and 2 of 10 Grade 1; p = 0.51), whereas there was a higher intradural invasion score in the LM8 implantation 12 weeks after irradiation group than the 12 weeks after nonirradiation group (3 of 10 Grade 0, 3 of 10 Grade 1 and 4 of 10 Grade 2 versus 8 of 10 Grade 0 and 2 of 10 Grade 1; p = 0.04). Interobserver reliability of assessing intradural tumor invasion grades in the LM8-implanted group was 0.93. In the microstructural analysis of the dura mater using SEM, irradiated mice had small defects on the dural surface at low magnification and degeneration of collagen fibers at high magnification. The median (range) number of defects on the dural surface per image in the irradiated mice was larger than that of nonirradiated mice (2 [1 to 3] versus 0; difference of medians, 2/image; p = 0.002) and the median size of defects was 60 µm (30 to 80). Interobserver reliability of assessing number of defects on the dural surface was 1.00. TEM revealed that nonirradiated mice demonstrated well-organized, multilayer structures, while irradiated mice demonstrated irregularly layered structures at low magnification. At high magnification, well-ordered cross-sections of collagen fibers were observed in the nonirradiated mice. However, disordered alignment of collagen fibers was observed in irradiated mice. CONCLUSION: Intradural tumor invasion and disruptions of the dural microstructure were observed in the meninges of mice after irradiation, indicating radiation-induced disruption of the meningeal barrier. CLINICAL RELEVANCE: We conclude that in this form of delivery, radiation is associated with disruption of the dural meningeal barrier, indicating a need to consider methods to avoid or limit Postradiation tumor relapse and spinal cord compression when treating spinal metastases so that patients do not experience intradural tumor invasion. Surgeons should be aware of the potential for intradural tumor invasion when they perform post-irradiation spinal surgery to minimize the risks for intraoperative dural injury and spinal cord injury. Further research in patients with irradiated spinal metastases is necessary to confirm that the same findings are observed in humans and to seek irradiation methods that prevent or minimize the disruption of meningeal barrier function.


Asunto(s)
Duramadre/efectos de la radiación , Neoplasias Mamarias Animales/radioterapia , Osteosarcoma/radioterapia , Compresión de la Médula Espinal/prevención & control , Médula Espinal/efectos de la radiación , Neoplasias de la Columna Vertebral/radioterapia , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Duramadre/ultraestructura , Femenino , Neoplasias Mamarias Animales/patología , Ratones , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Invasividad Neoplásica , Osteosarcoma/secundario , Paraplejía/etiología , Paraplejía/prevención & control , Radioterapia/efectos adversos , Médula Espinal/ultraestructura , Compresión de la Médula Espinal/etiología , Compresión de la Médula Espinal/patología , Neoplasias de la Columna Vertebral/complicaciones , Neoplasias de la Columna Vertebral/secundario , Factores de Tiempo
3.
Int J Radiat Oncol Biol Phys ; 109(3): 813-818, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33190969

RESUMEN

PURPOSE: Radiation therapy (RT) modulates the immune characteristics of the tumor microenvironment (TME). It is not known whether these effects are dependent on the type of RT used. METHODS AND MATERIALS: We evaluated the immunomodulatory effects of carbon-ion therapy (CiRT) compared with biologically equivalent doses of photon therapy (PhRT) on solid tumors. Orthotopic 4T1 mammary tumors in immunocompetent hosts were treated with CiRT or biologically equivalent doses of PhRT. Seventy-two hours after RT, tumors were harvested and the immune characteristics of the TME were quantified by flow cytometry and multiplex cytokine analyses. RESULTS: PhRT decreased the abundance of CD4+ and CD8+ T cells in the TME at all doses tested, with compensatory increases in proliferation. By contrast, CiRT did not significantly alter CD8+ T-cell infiltration. High-dose CiRT increased secretion of proinflammatory cytokines by tumor-infiltrating CD8+ T cells, including granzyme B, IL-2, and TNF-α, with no change in IFN-γ. Conversely, high-dose PhRT increased CD8+ T-cell secretion of IFN-γ only. At most of the doses studied, PhRT increased proliferation of immunosuppressive regulatory T cells; this was only seen with high-dose CiRT. Cytokine analyses of bulk dissociated tumors showed that CiRT significantly increased levels of IFN-γ, IL-2, and IL-1ß, whereas PhRT increased IL-6 levels alone. CONCLUSIONS: At low doses, lymphocytes differ in their sensitivity to CiRT compared with PhRT. Unlike PhRT, low-dose CiRT is generally lymphocyte-sparing. At higher doses, CiRT is a more potent inducer of proinflammatory cytokines and merits further study as a modulator of the immunologic characteristics of the TME.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Radioterapia de Iones Pesados , Neoplasias Mamarias Animales/radioterapia , Fotones/uso terapéutico , Microambiente Tumoral/efectos de la radiación , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Citometría de Flujo , Granzimas/metabolismo , Granzimas/efectos de la radiación , Inmunocompetencia , Interferón gamma/metabolismo , Interferón gamma/efectos de la radiación , Interleucina-1beta/metabolismo , Interleucina-1beta/efectos de la radiación , Interleucina-2/metabolismo , Interleucina-2/efectos de la radiación , Interleucina-6/metabolismo , Interleucina-6/efectos de la radiación , Neoplasias Mamarias Animales/inmunología , Ratones , Efectividad Biológica Relativa , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de la radiación , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/efectos de la radiación
4.
Nat Immunol ; 21(10): 1160-1171, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747819

RESUMEN

Autophagy supports both cellular and organismal homeostasis. However, whether autophagy should be inhibited or activated for cancer therapy remains unclear. Deletion of essential autophagy genes increased the sensitivity of mouse mammary carcinoma cells to radiation therapy in vitro and in vivo (in immunocompetent syngeneic hosts). Autophagy-deficient cells secreted increased amounts of type I interferon (IFN), which could be limited by CGAS or STING knockdown, mitochondrial DNA depletion or mitochondrial outer membrane permeabilization blockage via BCL2 overexpression or BAX deletion. In vivo, irradiated autophagy-incompetent mammary tumors elicited robust immunity, leading to improved control of distant nonirradiated lesions via systemic type I IFN signaling. Finally, a genetic signature of autophagy had negative prognostic value in patients with breast cancer, inversely correlating with mitochondrial abundance, type I IFN signaling and effector immunity. As clinically useful autophagy inhibitors are elusive, our findings suggest that mitochondrial outer membrane permeabilization may represent a valid target for boosting radiation therapy immunogenicity in patients with breast cancer.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Autofagia/genética , Neoplasias de la Mama/radioterapia , ADN Mitocondrial/genética , Neoplasias Mamarias Animales/radioterapia , Mitocondrias/metabolismo , Adulto , Anciano , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Humanos , Interferón Tipo I/metabolismo , Neoplasias Mamarias Animales/genética , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Pronóstico , Tolerancia a Radiación , Transducción de Señal , Análisis de Supervivencia
5.
Nano Lett ; 19(2): 805-815, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30592897

RESUMEN

Amplification of intracellular oxidative stress has been found to be an effective strategy to induce cancer cell death. To this end, we prepare a unique type of ultrasmall gallic acid-ferrous (GA-Fe(II)) nanocomplexes as the catalyst of Fenton reaction to enable persistent conversion of H2O2 to highly cytotoxic hydroxyl radicals (•OH). Then, both GA-Fe(II) and l-buthionine sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis, are coencapsulated within a stealth liposomal nanocarrier. Interestingly, the obtained BSO/GA-Fe(II)@liposome is able to efficiently amplify intracellular oxidative stress via increasing •OH generation and reducing GSH biosynthesis. After chelating with 99mTc4+ radioisotope, such BSO/GA-Fe(II)@liposome could be tracked under in vivo single-photon-emission-computed-tomography (SPECT) imaging, which illustrates the time-dependent tumor homing of such liposomal nanoparticles after intravenous injection. With GA-Fe(II)-mediated •OH production and BSO-mediated GSH depletion, treatment with such BSO/GA-Fe(II)@liposome would lead to dramatically enhanced intratumoral oxidative stresses, which then result in remarkably improved therapeutic efficacies of concurrently applied chemotherapy or radiotherapy. This work thus presents the concise fabrication of biocompatible BSO/GA-Fe(II)@liposome as an effective adjuvant nanomedicine to promote clinically used conventional cancer chemotherapy and radiotherapy, by greatly amplifying the intratumoral oxidative stress.


Asunto(s)
Butionina Sulfoximina/uso terapéutico , Compuestos Ferrosos/uso terapéutico , Ácido Gálico/uso terapéutico , Glutatión/antagonistas & inhibidores , Neoplasias Mamarias Animales/terapia , Estrés Oxidativo/efectos de los fármacos , Animales , Butionina Sulfoximina/administración & dosificación , Línea Celular Tumoral , Femenino , Compuestos Ferrosos/administración & dosificación , Ácido Gálico/administración & dosificación , Glutatión/metabolismo , Radical Hidroxilo/metabolismo , Liposomas/química , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Ratones , Ratones Endogámicos BALB C , Tomografía Computarizada de Emisión de Fotón Único
6.
Cancer Immunol Res ; 6(8): 910-920, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29907693

RESUMEN

Radiotherapy (RT) used at immunogenic doses leads to accumulation of cytosolic double-stranded DNA (dsDNA) in cancer cells, which activates type I IFN (IFN-I) via the cGAS/STING pathway. Cancer cell-derived IFN-I is required to recruit BATF3-dependent dendritic cells (DC) to poorly immunogenic tumors and trigger antitumor T-cell responses in combination with immune checkpoint blockade. We have previously demonstrated that the exonuclease TREX1 regulates radiation immunogenicity by degrading cytosolic dsDNA. Tumor-derived DNA can also activate cGAS/STING-mediated production of IFN-I by DCs infiltrating immunogenic tumors. However, how DNA from cancer cells is transferred to the cytoplasm of DCs remains unclear. Here, we showed that tumor-derived exosomes (TEX) produced by irradiated mouse breast cancer cells (RT-TEX) transfer dsDNA to DCs and stimulate DC upregulation of costimulatory molecules and STING-dependent activation of IFN-I. In vivo, RT-TEX elicited tumor-specific CD8+ T-cell responses and protected mice from tumor development significantly better than TEX from untreated cancer cells in a prophylactic vaccination experiment. We demonstrated that the IFN-stimulatory dsDNA cargo of RT-TEX is regulated by TREX1 expression in the parent cells. Overall, these results identify RT-TEX as a mechanism whereby IFN-stimulatory dsDNA is transferred from irradiated cancer cells to DCs. We have previously shown that the expression of TREX1 is dependent on the RT dose size. Thus, these data have important implications for the use of RT with immunotherapy. Cancer Immunol Res; 6(8); 910-20. ©2018 AACR.


Asunto(s)
ADN de Neoplasias/inmunología , Células Dendríticas/inmunología , Exodesoxirribonucleasas/inmunología , Exosomas/genética , Neoplasias Mamarias Animales/inmunología , Fosfoproteínas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Exosomas/inmunología , Femenino , Interferón Tipo I/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/prevención & control , Neoplasias Mamarias Animales/radioterapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/inmunología , Células Tumorales Cultivadas
7.
Mol Oncol ; 12(8): 1249-1263, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29738110

RESUMEN

We describe a cell damage-induced phenotype in mammary carcinoma cells involving acquisition of enhanced migratory and metastatic properties. Induction of this state by radiation required increased activity of the Ptgs2 gene product cyclooxygenase 2 (Cox2), secretion of its bioactive lipid product prostaglandin E2 (PGE2), and the activity of the PGE2 receptor EP4. Although largely transient, decaying to low levels in a few days to a week, this phenotype was cumulative with damage and levels of cell markers Sca-1 and ALDH1 increased with treatment dose. The Sca-1+ , metastatic phenotype was inhibited by both Cox2 inhibitors and PGE2 receptor antagonists, suggesting novel approaches to radiosensitization.


Asunto(s)
Antígenos Ly/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/radioterapia , Proteínas de la Membrana/genética , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígenos Ly/análisis , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Daño del ADN/efectos de la radiación , Femenino , Isoenzimas/análisis , Isoenzimas/genética , Neoplasias Mamarias Animales/patología , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Retinal-Deshidrogenasa/análisis , Retinal-Deshidrogenasa/genética
8.
Vet Comp Oncol ; 16(4): 497-504, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29806156

RESUMEN

In dogs, inflammatory mammary carcinoma is a clinicopathological entity characterized by rapid progression and aggressive behavior from onset of disease. Reported median survival time is short, with no effective treatment options. The aims of this prospective, noncontrolled clinical trial were to investigate outcome variables and safety profile of toceranib, thalidomide and piroxicam with or without hypofractionated radiation therapy in dogs with measurable histologically confirmed inflammatory mammary carcinoma that underwent a complete staging. Eighteen dogs were enrolled: 14 received medical treatment, and 4 were treated with hypofractionated radiation therapy and medical therapy. Overall, median time to progression was 34 days and median survival time was 109 days. In dogs treated with medical therapy, overall response rate was 21%, and clinical benefit rate (CBR) was 64%; median time to progression was 28 days and median survival time was 59 days. In dogs receiving medical therapy and undergoing radiation therapy, overall response rate and clinical benefit rate were 100%, with significantly longer time to progression (156 days) and survival time (180 days). Overall, treatment was well tolerated, with mild gastrointestinal and dermatological adverse events. Although the optimal treatment to this disease remains uncertain, the current approach consisting of systemic anti-angiogenic drugs with or without hypofractionated radiation therapy, provided clinical benefit in a significant proportion of dogs and should, therefore, be further explored.


Asunto(s)
Antineoplásicos/uso terapéutico , Enfermedades de los Perros/terapia , Indoles/uso terapéutico , Neoplasias Mamarias Animales/terapia , Piroxicam/uso terapéutico , Pirroles/uso terapéutico , Talidomida/uso terapéutico , Animales , Terapia Combinada/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/patología , Enfermedades de los Perros/radioterapia , Perros , Quimioterapia Combinada/veterinaria , Femenino , Indoles/administración & dosificación , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Estadificación de Neoplasias/veterinaria , Piroxicam/administración & dosificación , Pirroles/administración & dosificación , Hipofraccionamiento de la Dosis de Radiación , Talidomida/administración & dosificación , Resultado del Tratamiento
9.
Int J Radiat Oncol Biol Phys ; 100(3): 794-801, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29413289

RESUMEN

PURPOSE: This work investigates a new approach to enhance radiotherapy through a photo therapeutic agent activated by Cherenkov light produced from the megavoltage photon beam. The process is termed Radiotherapy Enhanced with Cherenkov photo-Activation (RECA). RECA is compatible with various photo-therapeutics, but here we focus on use with psoralen, an ultraviolet activated therapeutic with extensive history of application in superficial and extracorporeal settings. RECA has potential to extend the scope of psoralen treatments beyond superficial to deep seated lesions. METHODS AND MATERIALS: In vitro studies in B16 melanoma and 4T1 murine breast cancer cells were performed to investigate the potential of RT plus RECA versus RT alone for increasing cytotoxicity (local control) and increasing surface expression of major histocompatibility complex I (MHC I). The latter represents potential for immune response amplification (increased antigen presentation), which has been observed in other psoralen therapies. Cytotoxicity assays included luminescence and clonogenics. The MHC I assays were performed using flow cytometry. In addition, Cherenkov light intensity measurements were performed to investigate the possibility of increasing the Cherenkov light intensity per unit dose from clinical megavoltage beams, to maximize psoralen activation. RESULTS: Luminescence assays showed that RECA treatment (2 Gy at 6 MV) increased cytotoxicity by up to 20% and 9.5% for 4T1 and B16 cells, respectively, compared with radiation and psoralen alone (ie, Cherenkov light was blocked). Similarly, flow cytometry revealed median MHC I expression was significantly higher in RECA-treated cells, compared with those receiving radiation and psoralen alone (approximately 450% and 250% at 3 Gy and 6 Gy, respectively, P << .0001). Clonogenic assays of B16 cells at doses of 6 Gy and 12 Gy showed decreases in tumor cell viability of 7% (P = .017) and 36% (P = .006), respectively, when Cherenkov was present. CONCLUSION: This work demonstrates for the first time the potential for photo-activation of psoralen directly in situ, from Cherenkov light generated by a clinical megavoltage treatment beam.


Asunto(s)
Ficusina/uso terapéutico , Complejo Mayor de Histocompatibilidad , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/radioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia/métodos , Animales , Supervivencia Celular , Estudios de Factibilidad , Femenino , Mediciones Luminiscentes/métodos , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Radioterapia/métodos
10.
Acta Oncol ; 56(11): 1634-1638, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28838284

RESUMEN

INTRODUCTION: Vascular disrupting agents (VDAs) damage tumor vasculature and enhance tumor radiation response. In this pre-clinical study, we combined radiation with the leading VDA in clinical development, combretastatin A-4 phosphate (CA4P), and compared the effects seen in tumors and relevant normal tissues. MATERIAL AND METHODS: Radiation was applied locally to tissues in CDF1 mice to produce full radiation dose-response curves. CA4P (250 mg/kg) was intraperitoneally (i.p.) injected within 30 minutes after irradiating. Response of 200 mm3 foot implanted C3H mammary carcinomas was assessed using percent tumor control at 90 days. Normal tissue effects were evaluated using early responding skin (development of moist desquamation in the foot at 11-30 days), and late responding bladder (50% reduction in reservoir function estimated by cystometry up to 9 months after treatment), and lung (20% increase in ventilation rate measured by plethysmography within 9 months). A Chi-squared test was used for statistical comparisons (significance level of p < .05). RESULTS: The radiation dose controlling 50% of irradiated tumors was 52 Gy. This significantly decreased to 45 Gy with CA4P. The radiation doses inducing a change in skin, bladder and lung response in 50% of mice were 31 Gy, 14 Gy and 12 Gy, respectively. CA4P had no significant effect on the radiation response of any of these normal tissues. CONCLUSIONS: VDAs significantly enhance tumor radiation response, but had absolutely no effect on the radiation response of early or late responding normal tissues.


Asunto(s)
Bibencilos/farmacología , Pulmón/efectos de la radiación , Neoplasias Mamarias Animales/radioterapia , Neovascularización Patológica/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Piel/efectos de la radiación , Vejiga Urinaria/efectos de la radiación , Animales , Antineoplásicos Fitogénicos/farmacología , Femenino , Rayos gamma , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C3H , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
11.
Anticancer Res ; 37(4): 1723-1728, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28373434

RESUMEN

BACKGROUND: The lipophilic photosensitizer, TONS 501, is a novel porphyrin-derived methyl ester that was developed for photodynamic antimicrobial chemotherapy. This study developed a hydrophilic and anionic porphyrin salt of this compound (TONS 501-Na) for use in photodynamic therapy (PDT). This chlorin derivative is synthesized from the protoporphyrin IX dimethyl ester. MATERIALS AND METHODS: We investigated the in vitro cytotoxic effects of TONS 501-Na-mediated PDT on EMT6 mouse breast cancer cells. EMT6 cells were incubated with 0-100 µg/ml TONS 501-Na for 24 h prior to replacing the culture medium and exposing the cells to 6 mW/cm2 diode laser irradiation at 0-13 J/cm2 to induce PDT. Morphological changes and cell viability were evaluated 24 h after PDT. The percentages of apoptotic cells were evaluated 4 h and 24 h after PDT. RESULTS: The concentrations of TONS 501-Na that killed 50% of EMT6 cells after exposure to light doses of 0, 0.4, 3, 6, or 13 J/cm2 were 84.6, 33.2, 18, 8.2, and 2.2 µg/ml, respectively. Tumor cells exposed to PDT showed chromatin condensation and fragmentation. The percentages of apoptotic cells increased in a TONS 501-Na concentration-dependent manner in the PDT group, and were significantly higher than those in the control group or in cells treated with TONS 501-Na or laser irradiation alone. CONCLUSION: TONS 501-Na-mediated PDT induced mouse breast cancer cell death in a concentration-dependent manner. Future studies should evaluate the in vivo pharmacokinetics, tissue distribution, and photodynamic effects of TONS 501-Na.


Asunto(s)
Apoptosis/efectos de los fármacos , Luz , Neoplasias Mamarias Animales/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Porfirinas/química , Porfirinas/farmacología , Animales , Apoptosis/efectos de la radiación , Femenino , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Ratones , Células Tumorales Cultivadas
12.
Int J Radiat Biol ; 93(5): 507-516, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27935337

RESUMEN

PURPOSE: Radiotherapy increases the level of inflammatory cytokines, some of which are known to promote metastasis. In a mouse model of triple negative breast cancer (TNBC), we determined whether irradiation of the mammary tumor increases the level of key cytokines and favors the development of lung metastases. MATERIALS AND METHODS: D2A1 TNBC cells were implanted in the mammary glands of a Balb/c mouse and then 7 days old tumors were irradiated (4 × 6 Gy). The cytokines IL-1ß, IL-4, IL-6, IL-10, IL-17 and MIP-2 were quantified in plasma before, midway and after irradiation. The effect of tumor irradiation on the invasion of cancer cells, the number of circulating tumor cells (CTC) and lung metastases were also measured. RESULTS: TNBC tumor irradiation significantly increased the plasma level of IL-1ß, which was associated with a greater number of CTC (3.5-fold) and lung metastases (2.3-fold), compared to sham-irradiated animals. Enhancement of D2A1 cell invasion in mammary gland was associated with an increase of the matrix metalloproteinases-2 and -9 activity (MMP-2, -9). The ability of IL-1ß to stimulate the invasiveness of irradiated D2A1 cells was confirmed by in vitro invasion chamber assays. CONCLUSION: Irradiation targeting a D2A1 tumor and its microenvironment increased the level of the inflammatory cytokine IL-1ß and was associated with the promotion of cancer cell invasion and lung metastasis development.


Asunto(s)
Citocinas/inmunología , Interleucina-1beta/biosíntesis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/radioterapia , Animales , Línea Celular Tumoral , Femenino , Mediadores de Inflamación/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/radioterapia , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia , Dosificación Radioterapéutica
13.
Vet Comp Oncol ; 15(3): 932-951, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27076401

RESUMEN

We isolated 11 antibodies specific for canine CD138 (cCD138) to validate the interest of CD138 antigen targeting in dogs with spontaneous mammary carcinoma. The affinity of the monoclonal antibodies in the nanomolar range is suitable for immunohistochemistry and nuclear medicine applications. Four distinct epitopes were recognized on cCD138 by this panel of antibodies. CD138 expression in canine healthy tissues is comparable to that reported in humans. CD138 is frequently expressed in canine mammary carcinomas corresponding to the human triple negative breast cancer subtype, with cytoplasmic and membranous expression. In canine diffuse large B-cell lymphoma, CD138 expression is associated with the 'non-germinal center' phenotype corresponding to the most aggressive subtype in humans. This homology of CD138 expression between dogs and humans confirms the relevance of tumour-bearing dogs as spontaneous models for nuclear medicine applications, especially for the evaluation of new tumour targeting strategies for diagnosis by phenotypic imaging and radio-immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades de los Perros/radioterapia , Linfoma de Células B Grandes Difuso/veterinaria , Neoplasias Mamarias Animales/radioterapia , Radioinmunoterapia/veterinaria , Sindecano-1/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Enfermedades de los Perros/inmunología , Perros , Mapeo Epitopo/veterinaria , Femenino , Citometría de Flujo/veterinaria , Humanos , Hibridomas/inmunología , Linfoma de Células B Grandes Difuso/radioterapia , Ratones , Ratones Endogámicos BALB C , Radioinmunoterapia/métodos
14.
Radiat Prot Dosimetry ; 166(1-4): 369-73, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26246584

RESUMEN

An innovative molecule, GdBLDL, for boron neutron capture therapy (BNCT) has been developed and its effectiveness as a BNCT carrier is currently under evaluation using in vivo experiments on small animal tumour models. The molecule contains both (10)B (the most commonly used NCT agent) and (157)Gd nuclei. (157)Gd is the second most studied element to perform NCT, mainly thanks to its high cross section for the capture of low-energy neutrons. The main drawback of (157)Gd neutron capture reaction is the very short range and low-energy secondary charged particles (Auger electrons), which requires (157)Gd to be very close to the cellular DNA to have an appreciable biological effect. Treatment doses were calculated by Monte Carlo simulations to ensure the optimised tumour irradiation and the sparing of the healthy organs of the irradiated animals. The enhancement of the absorbed dose due to the simultaneous presence of (10)B and (157)Gd in the experimental set-up was calculated and the advantage introduced by the presence of (157)Gd was discussed.


Asunto(s)
Boro/uso terapéutico , Gadolinio/uso terapéutico , Neoplasias Mamarias Animales/radioterapia , Método de Montecarlo , Terapia por Captura de Neutrón , Planificación de la Radioterapia Asistida por Computador , Animales , Simulación por Computador , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Radiometría/métodos , Dosificación Radioterapéutica
15.
J Feline Med Surg ; 17(12): 1000-4, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25673018

RESUMEN

OBJECTIVES: The purpose of this study was to investigate the disease-free interval, survival time and adverse events of a combined treatment approach in cats with mammary malignant tumors using radical mastectomy and adjuvant mitoxantrone. METHODS: All cats underwent surgery to remove the mammary chain containing the tumors. A 3 cm margin was obtained around removed tumors. For staging purposes, regional inguinal lymphadenectomy was performed in all cases. After histopathology, cats were staged according to the World Health Organization's (WHO) staging system. Chemotherapy with mitoxantrone was started 15-30 days after surgery (6 mg/m(2) IV every 21 days for four cycles) with the objective of delaying metastasis. RESULTS: Three cats were intact, one cat was early spayed, four cats were late spayed and four cats were spayed at an unknown age. Based on the WHO's staging system, six cats were classified as stage I and six cats as stage III. The median disease-free interval and survival time were 360 and 480 days, respectively. Four (33%) cats received four doses of mitoxantrone, four (33%) cats received three doses and four (33%) cats received only one dose. The most frequent adverse effects of chemotherapy were azotemia, anorexia, leukopenia and vomiting. CONCLUSIONS AND RELEVANCE: Adjuvant mitoxantrone chemotherapy may be an option for feline mammary tumors. Further, sufficiently powered, randomized prospective trials are necessary to determine if mitoxantrone is superior, inferior or equivalent to doxorubicin in the adjuvant setting.


Asunto(s)
Antineoplásicos/administración & dosificación , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/radioterapia , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/radioterapia , Mitoxantrona/administración & dosificación , Animales , Antineoplásicos/efectos adversos , Gatos , Quimioterapia Adyuvante/veterinaria , Terapia Combinada/veterinaria , Supervivencia sin Enfermedad , Femenino , Mastectomía Radical/veterinaria , Mitoxantrona/efectos adversos , Estadificación de Neoplasias/veterinaria , Estudios Prospectivos
16.
Cancer Immunol Immunother ; 63(3): 259-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357146

RESUMEN

The need for an intact immune system for cancer radiation therapy to be effective suggests that radiation not only acts directly on the tumor but also indirectly, through the activation of host immune components. Recent studies demonstrated that endogenous type I interferons (type I IFNs) play a role in radiation-mediated anti-tumor immunity by enhancing the ability of dendritic cells to cross-prime CD8(+) T cells. However, it is still unclear to what extent endogenous type I IFNs contribute to the recruitment and function of CD8(+) T cells. Little is also known about the effects of type I IFNs on myeloid cells. In the current study, we demonstrate that type I and type II IFNs (IFN-γ) are both required for the increased production of CXCL10 (IP-10) chemokine by myeloid cells within the tumor after radiation treatment. Radiation-induced intratumoral IP-10 levels in turn correlate with tumor-infiltrating CD8(+) T cell numbers. Moreover, type I IFNs promote potent tumor-reactive CD8(+) T cells by directly affecting the phenotype, effector molecule production, and enhancing cytolytic activity. Using a unique inducible expression system to increase local levels of IFN-α exogenously, we show here that the capacity of radiation therapy to result in tumor control can be enhanced. Our preclinical approach to study the effects of local increase in IFN-α levels can be used to further optimize the combination therapy strategy in terms of dosing and scheduling, which may lead to better clinical outcome.


Asunto(s)
Linfocitos T CD8-positivos/efectos de la radiación , Interferón-alfa/metabolismo , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Células Mieloides/efectos de los fármacos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de la radiación , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/efectos de la radiación , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Interferón-alfa/genética , Interferón-alfa/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Recuento de Linfocitos , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Trasplante de Neoplasias
17.
J Exp Med ; 210(11): 2435-66, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24127486

RESUMEN

Rational combinatorial therapeutic strategies have proven beneficial for the management of cancer. Recent success of checkpoint blockade in highly immunogenic tumors has renewed interest in immunotherapy. Regulatory T (T reg) cells densely populate solid tumors, which may promote progression through suppressing anti-tumor immune responses. We investigated the role of T reg cells in murine mammary carcinogenesis using an orthotopic, polyoma middle-T antigen-driven model in Foxp3(DTR) knockin mice. T reg cell ablation resulted in significant determent of primary and metastatic tumor progression. Importantly, short-term ablation of T reg cells in advanced spontaneous tumors led to extensive apoptotic tumor cell death. This anti-tumor activity was dependent on IFN-γ and CD4(+) T cells but not on NK or CD8(+) T cells. Combination of T reg cell ablation with CTLA-4 or PD-1/PD-L1 blockade did not affect tumor growth or improve the therapeutic effect attained by T reg cell ablation alone. However, T reg cell targeting jointly with tumor irradiation significantly reduced tumor burden and improved overall survival. Together, our results demonstrate a major tumor-promoting role of T reg cells in an autochthonous model of tumorigenesis, and they reveal the potential therapeutic value of combining transient T reg cell ablation with radiotherapy for the management of poorly immunogenic, aggressive malignancies.


Asunto(s)
Depleción Linfocítica , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/radioterapia , Oncogenes/genética , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Puntos de Control del Ciclo Celular/efectos de la radiación , Muerte Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Progresión de la Enfermedad , Femenino , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de la radiación , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/patología , Ratones , Radiación Ionizante , Linfocitos T Reguladores/efectos de la radiación , Carga Tumoral/inmunología , Carga Tumoral/efectos de la radiación , Microambiente Tumoral/efectos de la radiación
18.
J Renin Angiotensin Aldosterone Syst ; 14(4): 330-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23132847

RESUMEN

BACKGROUND AND AIM: Cardiovascular complications are one limitation of breast cancer treatment. The aim of the current study was to investigate whether the renin-angiotensin related genes could be altered by chemotherapy and radiotherapy, using a rat model. METHODS: Female rats were divided into three groups: control, chemotherapy + irradiation (TC+IR) and irradiation (IR). Molecular analyses of the left ventricle were performed five months after the end of treatment. The analyses evaluated the changes in mRNA levels of some renin-angiotensin system (RAS) related genes: angiotensinogen, renin, angiotensin-converting enzyme (ACE) and angiotensin II type 1 receptor (AT1) and vascular endothelial growth factor (VEGF), which can be related to ACE production, by RT-PCR. RESULTS: Renin was only observed in treated groups, TC+IR and IR, compared with the control group. ACE and VEGF levels were decreased in TC+IR (p<0.001) and in IR (p<0.001), and AT1 mRNA was higher in groups TC+IR (p<0.01) and IR (p<0.05) compared with the control group. CONCLUSION: Chemotherapy and irradiation can induce significant changes in some RAS related genes. These alterations are important to understand the pathways and consequences beyond cardiotoxicity induced by breast cancer treatments.


Asunto(s)
Regulación de la Expresión Génica , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/radioterapia , Miocardio/metabolismo , Sistema Renina-Angiotensina/genética , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Femenino , Mastocitos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Renina/genética , Renina/metabolismo , Coloración y Etiquetado , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
PLoS One ; 7(10): e48049, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133545

RESUMEN

The primary tumor represents a potential source of antigens for priming immune responses for disseminated disease. Current means of debulking tumors involves the use of cytoreductive conditioning that impairs immune cells or removal by surgery. We hypothesized that activation of the immune system could occur through the localized release of tumor antigens and induction of tumor death due to physical disruption of tumor architecture and destruction of the primary tumor in situ. This was accomplished by intratumor injection of magneto-rheological fluid (MRF) consisting of iron microparticles, in Balb/c mice bearing orthotopic 4T1 breast cancer, followed by local application of a magnetic field resulting in immediate coalescence of the particles, tumor cell death, slower growth of primary tumors as well as decreased tumor progression in distant sites and metastatic spread. This treatment was associated with increased activation of DCs in the draining lymph nodes and recruitment of both DCs and CD8(+)T cells to the tumor. The particles remained within the tumor and no toxicities were observed. The immune induction observed was significantly greater compared to cryoablation. Further anti-tumor effects were observed when MRF/magnet therapy was combined with systemic low dose immunotherapy. Thus, mechanical disruption of the primary tumor with MRF/magnetic field application represents a novel means to induce systemic immune activation in cancer.


Asunto(s)
Antineoplásicos/farmacología , Hierro/farmacología , Neoplasias Mamarias Animales/radioterapia , Animales , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Progresión de la Enfermedad , Femenino , Citometría de Flujo/métodos , Sistema Inmunológico , Inmunoterapia/métodos , Campos Magnéticos , Magnetismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Necrosis , Metástasis de la Neoplasia , Neoplasias/inmunología , Células Madre
20.
Int J Radiat Biol ; 88(3): 277-85, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22111842

RESUMEN

PURPOSE: These studies explored questions related to the potential use of Laromustine in the treatment of solid tumors and in combination with radiotherapy. MATERIALS AND METHODS: The studies used mouse EMT6 cells (both parental and transfected with genes for O(6)-alkylguanine-DNA transferase [AGT]), repair-deficient human Fanconi Anemia C and Chinese hamster VC8 (BRCA2(-/-)) cells and corresponding control cells, and EMT6 tumors in mice assayed using cell survival and tumor growth assays. RESULTS: Hypoxia during Laromustine treatment did not protect EMT6 cells or human fibroblasts from this agent. Rapidly proliferating EMT6 cells were more sensitive than quiescent cultures. EMT6 cells expressing mouse or human AGT, which removes O(6)-alkyl groups from DNA guanine, thereby protecting against G-C crosslink formation, increased resistance to Laromustine. Crosslink-repair-deficient Fanconi Anemia C and VC8 cells were hypersensitive to Laromustine, confirming the importance of crosslinks as lethal lesions. In vitro, Laromustine and radiation produced additive toxicities to EMT6 cells. Studies using tumor cell survival and tumor growth assays showed effects of regimens combining Laromustine and radiation that were compatible with additive or subadditive interactions. CONCLUSIONS: The effects of Laromustine on solid tumors and with radiation are complex and are influenced by microenvironmental and proliferative heterogeneity within these malignancies.


Asunto(s)
Antineoplásicos/farmacología , Hidrazinas/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Sulfonamidas/farmacología , Animales , Antineoplásicos/uso terapéutico , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Terapia Combinada , Cricetinae , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Humanos , Hidrazinas/uso terapéutico , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Ratones , Neoplasias/metabolismo , Neoplasias/patología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Sulfonamidas/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...