Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Radiat Oncol ; 13(1): 89, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29747666

ABSTRACT

BACKGROUND: Resistance to radiotherapy is frequently encountered in patients with glioblastoma multiforme. It is caused at least partially by the high glutathione content in the tumour tissue. Therefore, the administration of the glutathione synthesis inhibitor Buthionine-SR-Sulfoximine (BSO) should increase survival time. METHODS: BSO was tested in combination with an experimental synchrotron-based treatment, microbeam radiation therapy (MRT), characterized by spatially and periodically alternating microscopic dose distribution. One hundred thousand F98 glioma cells were injected into the right cerebral hemisphere of adult male Fischer rats to generate an orthotopic small animal model of a highly malignant brain tumour in a very advanced stage. Therapy was scheduled for day 13 after tumour cell implantation. At this time, 12.5% of the animals had already died from their disease. The surviving 24 tumour-bearing animals were randomly distributed in three experimental groups: subjected to MRT alone (Group A), to MRT plus BSO (Group B) and tumour-bearing untreated controls (Group C). Thus, half of the irradiated animals received an injection of 100 µM BSO into the tumour two hours before radiotherapy. Additional tumour-free animals, mirroring the treatment of the tumour-bearing animals, were included in the experiment. MRT was administered in bi-directional mode with arrays of quasi-parallel beams crossing at the tumour location. The width of the microbeams was ≈28 µm with a center-to-center distance of ≈400 µm, a peak dose of 350 Gy, and a valley dose of 9 Gy in the normal tissue and 18 Gy at the tumour location; thus, the peak to valley dose ratio (PVDR) was 31. RESULTS: After tumour-cell implantation, otherwise untreated rats had a mean survival time of 15 days. Twenty days after implantation, 62.5% of the animals receiving MRT alone (group A) and 75% of the rats given MRT + BSO (group B) were still alive. Thirty days after implantation, survival was 12.5% in Group A and 62.5% in Group B. There were no survivors on or beyond day 35 in Group A, but 25% were still alive in Group B. Thus, rats which underwent MRT with adjuvant BSO injection experienced the largest survival gain. CONCLUSIONS: In this pilot project using an orthotopic small animal model of advanced malignant brain tumour, the injection of the glutathione inhibitor BSO with MRT significantly increased mean survival time.


Subject(s)
Brain Neoplasms/mortality , Buthionine Sulfoximine/pharmacology , Glioma/mortality , Glutathione/metabolism , Radiotherapy/methods , Synchrotrons , Animals , Antimetabolites/pharmacology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Combined Modality Therapy , Dose-Response Relationship, Radiation , Glioma/pathology , Glioma/therapy , Male , Pilot Projects , Rats , Rats, Inbred F344 , Survival Rate
2.
Sci Rep ; 6: 27250, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27264273

ABSTRACT

Synchrotron-generated X-ray (SRX) microbeams deposit high radiation doses to submillimetric targets whilst minimizing irradiation of neighboring healthy tissue. We developed a new radiosurgical method which demonstrably transects cortical brain tissue without affecting adjacent regions. We made such image-guided SRX microtransections in the left somatosensory cortex in a rat model of generalized epilepsy using high radiation doses (820 Gy) in thin (200 µm) parallel slices of tissue. This procedure, targeting the brain volume from which seizures arose, altered the abnormal neuronal activities for at least 9 weeks, as evidenced by a decrease of seizure power and coherence between tissue slices in comparison to the contralateral cortex. The brain tissue located between transections stayed histologically normal, while the irradiated micro-slices remained devoid of myelin and neurons two months after irradiation. This pre-clinical proof of concept highlights the translational potential of non-invasive SRX transections for treating epilepsies that are not eligible for resective surgery.


Subject(s)
Radiosurgery/instrumentation , Seizures/radiotherapy , Somatosensory Cortex/radiation effects , Animals , Disease Models, Animal , Humans , Rats , Seizures/physiopathology , Somatosensory Cortex/physiopathology , Synchrotrons
3.
Phys Med ; 31(6): 607-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25934524

ABSTRACT

Epilepsy is one of the most important neurological diseases. It concerns about 1% of the population worldwide. Despite the discovery of new molecules, one third of epileptic patients are resistant to anti-epileptic drugs and among them only a few can benefit from resective surgery. In this context, radiotherapy is an interesting alternative to the other treatments and several clinical devices exist (e.g., Gamma Knife(®)). The European Synchrotron Radiation Facility offers the possibility to develop new methods of radiosurgery and to study their antiepileptic effects. Here, we discuss several studies that we performed recently to test and try to understand the antiepileptic effects of X-ray synchrotron microbeams in different animal models of epilepsy. We showed a decrease of seizures after Interlaced Microbeam Radiotherapy (IntMRT) of the somatosensory cortex, known as the seizure generator, in a genetic model of absence epilepsy. These antiepileptic effects were stable over 4 months and with low tissular and functional side-effects. The irradiated pyramidal neurons still displayed their physiological activity but did not synchronize anymore. We also obtained a lasting suppression of seizures after IntMRT of the dorsal hippocampus in a mouse model of mesiotemporal lobe epilepsy. However, an important variability of antiepileptic efficiency was observed probably due to the small size of the targeted structure. Despite these encouraging proofs-of-concepts, there is now a need to adapt IntMRT to other models of epilepsy in rodents which are close to refractory forms of epilepsy in human patients and to implement this approach to non-human primates, before moving to clinical trials.


Subject(s)
Biological Clocks , Dose Fractionation, Radiation , Drug Resistant Epilepsy/physiopathology , Drug Resistant Epilepsy/surgery , Radiosurgery/instrumentation , Synchrotrons/instrumentation , Animals , Equipment Design , Feasibility Studies , Hippocampus/physiopathology , Hippocampus/radiation effects , Hippocampus/surgery , Humans , Mice , Mice, Inbred C57BL , Nerve Net/physiopathology , Nerve Net/surgery , Radiosurgery/methods , Radiotherapy, High-Energy/instrumentation , Radiotherapy, High-Energy/methods , Rats , Treatment Outcome
4.
Phys Med ; 31(6): 564-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25773883

ABSTRACT

Microbeam radiation therapy (MRT), a novel form of spatially fractionated radiotherapy (RT), uses arrays of synchrotron-generated X-ray microbeams (MB). MRT has been identified as a promising treatment concept that might be applied to patients with malignant central nervous system (CNS) tumours for whom, at the current stage of development, no satisfactory therapy is available yet. Preclinical experimental studies have shown that the CNS of healthy rodents and piglets can tolerate much higher radiation doses delivered by spatially separated MBs than those delivered by a single, uninterrupted, macroscopically wide beam. High-dose, high-precision radiotherapies such as MRT with reduced probabilities of normal tissue complications offer prospects of improved therapeutic ratios, as extensively demonstrated by results of experiments published by many international groups in the last two decades. The significance of developing MRT as a new RT approach cannot be understated. Up to 50% of cancer patients receive conventional RT, and any new treatment that provides better tumour control whilst preserving healthy tissue is likely to significantly improve patient outcomes.


Subject(s)
Brain Neoplasms/radiotherapy , Dose Fractionation, Radiation , Radiotherapy, High-Energy/instrumentation , Radiotherapy, High-Energy/methods , Synchrotrons/instrumentation , Animals , Equipment Design , Evidence-Based Medicine , Humans , Mice , Rats , Swine , Technology Assessment, Biomedical , Treatment Outcome
5.
Mutat Res ; 704(1-3): 160-6, 2010.
Article in English | MEDLINE | ID: mdl-20034592

ABSTRACT

Microbeam radiation therapy (MRT) uses highly collimated, quasi-parallel arrays of X-ray microbeams of 50-600keV, produced by third generation synchrotron sources, such as the European Synchrotron Radiation Facility (ESRF), in France. The main advantages of highly brilliant synchrotron sources are an extremely high dose rate and very small beam divergence. High dose rates are necessary to deliver therapeutic doses in microscopic volumes, to avoid spreading of the microbeams by cardiosynchronous movement of the tissues. The minimal beam divergence results in the advantage of steeper dose gradients delivered to a tumor target, thus achieving a higher dose deposition in the target volume in fractions of seconds, with a sharper penumbra than that produced in conventional radiotherapy. MRT research over the past 20 years has yielded many results from preclinical trials based on different animal models, including mice, rats, piglets and rabbits. Typically, MRT uses arrays of narrow ( approximately 25-100 microm wide) microplanar beams separated by wider (100-400 microm centre-to-centre) microplanar spaces. The height of these microbeams typically varies from 1 to 100 mm, depending on the target and the desired preselected field size to be irradiated. Peak entrance doses of several hundreds of Gy are surprisingly well tolerated by normal tissues, up to approximately 2 yr after irradiation, and at the same time show a preferential damage of malignant tumor tissues; these effects of MRT have now been extensively studied over nearly two decades. More recently, some biological in vivo effects of synchrotron X-ray beams in the millimeter range (0.68-0.95 mm, centre-to-centre distances 1.2-4 mm), which may differ to some extent from those of microscopic beams, have been followed up to approximately 7 months after irradiation. Comparisons between broad-beam irradiation and MRT indicate a higher tumor control for the same sparing of normal tissue in the latter, even if a substantial fraction of tumor cells are not receiving a radiotoxic level of radiation. The hypothesis of a selective radiovulnerability of the tumor vasculature versus normal blood vessels by MRT, and of the cellular and molecular mechanisms involved remains under investigation. The paper highlights the history of MRT including salient biological findings after microbeam irradiation with emphasis on the vascular components and the tolerance of the central nervous system. Details on experimental and theoretical dosimetry of microbeams, core issues and possible therapeutic applications of MRT are presented.


Subject(s)
Brain Neoplasms/radiotherapy , Brain/radiation effects , Radiotherapy/methods , Synchrotrons , Technology, Radiologic/methods , X-Rays , Animals , Blood Vessels/radiation effects , Brain Neoplasms/blood supply , Dose Fractionation, Radiation , History, 20th Century , History, 21st Century , Humans , Neoplasms/blood supply , Radiometry , Radiotherapy/instrumentation , Technology, Radiologic/history
6.
Rev Sci Instrum ; 80(7): 074301, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19655968

ABSTRACT

During the past decade microbeam radiation therapy has evolved from preclinical studies to a stage in which clinical trials can be planned, using spatially fractionated, highly collimated and high intensity beams like those generated at the x-ray ID17 beamline of the European Synchrotron Radiation Facility. The production of such microbeams typically between 25 and 100 microm full width at half maximum (FWHM) values and 100-400 microm center-to-center (c-t-c) spacings requires a multislit collimator either with fixed or adjustable microbeam width. The mechanical regularity of such devices is the most important property required to produce an array of identical microbeams. That ensures treatment reproducibility and reliable use of Monte Carlo-based treatment planning systems. New high precision wire cutting techniques allow the fabrication of these collimators made of tungsten carbide. We present a variable slit width collimator as well as a single slit device with a fixed setting of 50 microm FWHM and 400 microm c-t-c, both able to cover irradiation fields of 50 mm width, deemed to meet clinical requirements. Important improvements have reduced the standard deviation of 5.5 microm to less than 1 microm for a nominal FWHM value of 25 microm. The specifications of both devices, the methods used to measure these characteristics, and the results are presented.


Subject(s)
Microtechnology/instrumentation , Radiotherapy/instrumentation , Linear Models , Temperature
8.
Dev Med Child Neurol ; 49(8): 577-81, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17635201

ABSTRACT

Microbeam radiation therapy (MRT), a form of experimental radiosurgery of tumours using multiple parallel, planar, micrometres-wide, synchrotron-generated X-ray beams ('microbeams'), can safely deliver radiation doses to contiguous normal animal tissues that are much higher than the maximum doses tolerated by the same normal tissues of animals or patients from any standard millimetres-wide radiosurgical beam. An array of parallel microbeams, even in doses that cause little damage to radiosensitive developing tissues, for example, the chick chorioallantoic membrane, can inhibit growth or ablate some transplanted malignant tumours in rodents. The cerebella of 100 normal 20 to 38g suckling Sprague-Dawley rat pups and of 13 normal 5 to 12kg weanling Yorkshire piglets were irradiated with an array of parallel, synchrotron-wiggler-generated X-ray microbeams in doses overlapping the MRT-relevant range (about 50-600Gy) using the ID17 wiggler beamline tangential to the 6GeV electron synchrotron ring at the European Synchrotron Radiation Facility in Grenoble, France. Subsequent favourable development of most animals over at least 1 year suggests that MRT might be used to treat children's brain tumours with less risk to the development of the central nervous system than is presently the case when using wider beams.


Subject(s)
Brain Neoplasms/physiopathology , Brain Neoplasms/radiotherapy , X-Ray Therapy/methods , Animals , Dose-Response Relationship, Radiation , Humans , Neurologic Examination , X-Ray Therapy/adverse effects
9.
J Neurooncol ; 78(2): 135-43, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16598429

ABSTRACT

PURPOSE: Microbeam radiation therapy (MRT), a novel experimental radiosurgery that largely spares the developing CNS and other normal tissues, is tolerated well by developing animals and palliates advanced 9LGS tumors. This report, to our knowledge, is the first demonstration that gene-mediated immunotherapy (GMIMPR) enhances the efficacy of MRT for advanced 9LGS tumors. METHODS: Seventy-six male Fischer 344 rats were implanted ic with 10(4)9LGS cells on d0. By d14, the cells had generated approximately approximately 40 mm3 ic 9LGS tumours, experimental models for therapy of moderately aggressive human malignant astrocytomas. Each of the 14 untreated (control) rats died from a large (>100 mg) ic tumor before d29 (median, d21). On d14, the remaining 62 rats were given deliberately suboptimal microbeam radiation therapy (MRT) by a single lateral exposure of the tumor-bearing zone of the head to a 10.1 mm-wide, approximately approximately 11 mm-high array of 20-39 microm-wide, nearly parallel beams of synchrotron wiggler-generated radiation (mainly approximately 50-150 keV X-rays) that delivered 625 Gy peak skin doses at approximately approximately 211 microm ctc intervals in approximately approximately 300 ms either without additional treatments (MRT-only, 25 rats), with post-MRT GMIMPR (MRT+GMIMPR, 23 rats: multiple sc injections of irradiated (clonogenically-disabled) GM-CSF gene-transfected 9LGS cells), or with post-MRT IMPR (MRT+IMPR, 14 rats: multiple sc injections of irradiated (clonogenically-disabled) 9LGS cells. RESULTS: The median post-implantation survivals of rats in the MRT-only, MRT+GMIMPR and MRT+IMPR groups were over twice that of controls; further, approximately approximately 20% of rats in MRT-only and MRT+IMPR groups survived >1 yr with no obvious disabilities. Moreover, over 40% of MRT+GMIMPR rats survived >1 yr with no obvious disabilities, a significant (P<0.04) increase over the MRT-only and MRT+IMPR groups. SIGNIFICANCE: These data suggest that the combination of MRT+GMIMPR might be better than MRT only for unifocal CNS tumors, particularly in infants and young children.


Subject(s)
Brain Neoplasms/therapy , Gliosarcoma/therapy , Immunotherapy/methods , Radiosurgery/methods , Age Factors , Animals , Brain Neoplasms/immunology , Brain Neoplasms/surgery , Combined Modality Therapy/methods , Genetic Therapy , Gliosarcoma/immunology , Gliosarcoma/surgery , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunization , Male , Radiotherapy Dosage , Rats , Rats, Inbred F344 , Statistics, Nonparametric , Survival Analysis , Transfection
10.
Br J Radiol ; 79(937): 71-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16421408

ABSTRACT

Microbeam radiosurgery (MBRS), also referred to as microbeam radiation therapy (MRT), was tested at the European Synchrotron Radiation Facility (ESRF). The left tibiofibular thigh of a mouse bearing a subcutaneously (sc) implanted mouse model (SCCVII) of aggressive human squamous-cell carcinoma was irradiated in two orthogonal exposures with or without a 16 mm aluminium filter through a multislit collimator (MSC) by arrays of nearly parallel microbeams spaced 200 microm on centre (oc). The peak skin-entrance dose from each exposure was 442 Gy, 625 Gy, or 884 Gy from 35 microm wide beams or 442 Gy from 70 microm wide beams. The 442/35, 625/35, 884/35 and 442/70 MBRSs yielded 25 day, 29 day, 37 day and 35 day median survival times (MST) (post-irradiation), respectively, exceeding the 20 day MST from 35 Gy-irradiation of SCCVIIs with a seamless 100 kVp X-ray beam.


Subject(s)
Carcinoma, Squamous Cell/surgery , Radiosurgery/methods , Synchrotrons , Animals , Female , Humans , Lower Extremity , Mice , Mice, Inbred C3H , Models, Animal , Neoplasm Transplantation , Palliative Care , Survival Rate
11.
Med Phys ; 30(4): 583-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12722810

ABSTRACT

Preclinical experiments are carried out with approximately 20-30 microm wide, approximately 10 mm high parallel microbeams of hard, broad-"white"-spectrum x rays (approximately 50-600 keV) to investigate microbeam radiation therapy (MRT) of brain tumors in infants for whom other kinds of radiotherapy are inadequate and/or unsafe. Novel physical microdosimetry (implemented with MOSFET chips in the "edge-on" mode) and Monte Carlo computer-simulated dosimetry are described here for selected points in the peak and valley regions of a microbeam-irradiated tissue-equivalent phantom. Such microbeam irradiation causes minimal damage to normal tissues, possible because of rapid repair of their microscopic lesions. Radiation damage from an array of parallel microbeams tends to correlate with the range of peak-valley dose ratios (PVDR). This paper summarizes comparisons of our dosimetric MOSFET measurements with Monte Carlo calculations. Peak doses at depths <22 mm are 18% less than Monte Carlo values, whereas those depths >22 mm and valley doses at all depths investigated (2 mm-62 mm) are within 2-13% of the Monte Carlo values. These results lend credence to the use of MOSFET detector systems in edge-on mode for microplanar irradiation dosimetry.


Subject(s)
Brain Neoplasms/physiopathology , Brain Neoplasms/radiotherapy , Radiometry/instrumentation , Radiotherapy Planning, Computer-Assisted/instrumentation , Radiotherapy/instrumentation , Rhombencephalon/physiopathology , Transducers , Animals , Computer Simulation , Equipment Design , Equipment Failure Analysis , Europe , Humans , Infant, Newborn , Miniaturization , Models, Biological , Nuclear Medicine Department, Hospital , Radiometry/methods , Radiotherapy/methods , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted/methods , Reproducibility of Results , Semiconductors , Sensitivity and Specificity , Synchrotrons
12.
Eur J Nucl Med ; 28(7): 836-46, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11504080

ABSTRACT

Somatostatin receptors are known to be expressed in a large number of human tumours and represent the basis for in vivo tumour targeting. Stable somatostatin derivatives such as octreotide or lanreotide are the most frequently used radiopharmaceuticals acting through specific binding to somatostatin receptors; however, they do not bind with high affinity to all five receptor subtypes. Whereas the mRNAs for most receptor subtypes have been detected in tumours, it is in most cases unclear which of the receptor subtype proteins are expressed. Since in vitro receptor binding methods are close correlates and predictors of in vivo peptide receptor targeting, we took advantage of the recently developed subtype-selective analogues and evaluated approximately 200 tumours for their receptor subtype protein expression in specific binding assays using autoradiography with 125I-[Leu8, D-Trp22, Tyr25]-somatostatin-28 and displacement by subtype-selective analogues. The majority of the tested neuroblastomas, meningiomas, medulloblastomas, breast carcinomas, lymphomas, renal cell carcinomas, paragangliomas, small cell lung carcinomas and hepatocellular carcinomas predominantly expressed sst2. The prostate carcinomas and sarcomas preferentially expressed sstl, while a majority of inactive pituitary adenomas displayed sst3 and, to a lesser extent, sst2. Growth hormone-secreting pituitary adenomas preferentially expressed sst2 and sst5; gastroenteropancreatic tumours and phaeochromocytomas frequently displayed sst2 and/or sstl. Non-neoplastic human tissues such as vessels, nerve plexus, pancreatic islets, prostatic stroma, adrenal medulla, spleen and germinal centres of the lymphoid tissues preferentially expressed sst2. However, the human gastric mucosa predominantly expressed sst1 while colonic mucosa displayed sst2. Interestingly, a minority of tumours showed a strong 125I-[Leu8, D-Trp22, Tyr25]-somatostatin-28 binding, of which less than 50% could be displaced by the sum of the five subtype-selective analogues. This observation suggests the existence of an as yet unknown subtype in selected tumours. This study is the first report to analyse the somatostatin receptor subtype expression in tumours with binding methods. We conclude that sst2, with high affinity for current radiopharmaceuticals such as Octreoscan, is predominantly expressed in a majority of tumours. Fewer tumour types (sarcomas, prostate cancers, inactive pituitary adenomas) preferentially express another subtype. This information is of importance with regard to the clinical applications and development of somatostatin analogues with distinct receptor subtype selectivities.


Subject(s)
Iodine Radioisotopes , Ligands , Neoplasms/chemistry , Receptors, Somatostatin/analysis , Autoradiography , Humans , In Vitro Techniques , Tissue Distribution
13.
Blood ; 97(6): 1543-8, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238088

ABSTRACT

Lymph nodes with Hodgkin disease (HD) harbor few neoplastic cells in a marked leukocytic infiltrate. Since chemokines are likely to be involved in the recruitment of these leukocytes, the expression of potentially relevant chemokines and chemokine receptors were studied in lymph nodes from 24 patients with HD and in 5 control lymph nodes. The expression of regulated on activation, normal T cell expressed and secreted (RANTES), monocyte chemotactic protein (MCP)-1, macrophage inflammatory protein (MIP)-1alpha, and MIP-1beta was analyzed by in situ hybridization and that of CCR3 and CCR5 by immunohistochemistry and flow cytometry. It was found that, overall, the expression of all 4 chemokines was markedly enhanced, but the cellular source was different. RANTES was expressed almost exclusively by T cells whereas the expression of MCP-1, MIP-1alpha, and MIP-1beta was confined largely to macrophages. In control lymph nodes, chemokine expression was low, with the exception of MIP-1alpha in macrophages. CCR3 and CCR5 were highly expressed in T cells of HD involved but not of control lymph nodes. CCR3 was equally distributed in CD4+ and CD8+ cells, but CCR5 was associated largely with CD4+ cells. In HD lymph nodes, CCR3 and CCR5 were also expressed in B cells, which normally do not express these receptors. All these chemokines and receptors studied, by contrast, were absent in the neoplastic cells. It was concluded that chemokines are involved in the formation of the HD nonneoplastic leukocytic infiltrate. Expression of CCR3 and CCR5 appears to be characteristic of HD, but the roles of these receptors' up-regulation for the disease process remain unclear.


Subject(s)
Chemokines, CC/metabolism , Hodgkin Disease/metabolism , Leukocytes/cytology , Receptors, CCR5/metabolism , Receptors, Chemokine/metabolism , Adult , Cell Movement/immunology , Chemokines, CC/genetics , Female , Hodgkin Disease/pathology , Humans , Immunohistochemistry , Immunophenotyping , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Receptors, CCR3 , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured
14.
J Clin Endocrinol Metab ; 85(10): 3882-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061553

ABSTRACT

The distribution of the sst2A receptor was investigated, using immunohistochemistry, with the specific antipeptide antibody R2-88, in a total of 120 tumors of the nervous and the neuroendocrine systems, including small-cell lung carcinomas, medulloblastomas, neuroblastomas, pheochromocytomas, and paragangliomas. The great majority of the tumor samples, frozen or formalin-fixed, showed a positive immunohistochemical staining with R2-88, and an excellent correlation with receptor autoradiography using 125I[Tyr3]-octreotide. Whereas small-cell lung carcinomas and medulloblastomas had a predominantly plasma membrane staining, pheochromocytomas and neuroblastomas had variable ratios of cell surface and intracellular staining. Strikingly, a preferentially cytoplasmic staining was seen in tumors with a high level of somatostatin gene expression, whereas a more plasma membranous staining was seen in tumors lacking somatostatin messenger RNA. Specificity of both the plasma membrane and the cytoplasmic staining pattern was confirmed in immunoblots, which showed the immunoreactive receptor migrating as a characteristic 70-kDa broad band. In both immunohistochemical and immunoblotting experiments, staining was abolished by antibody blockade with 100 nM antigen peptide. These results describe, for the first time, the localization of the sst2A receptor protein in human small-cell lung carcinomas, medulloblastomas, neuroblastomas, and paragangliomas. Moreover, it is the first report investigating possible causes for distinct subcellular localizations of sst2A in human tissues. We show that the subcellular distribution of the receptor may be dependent on the surrounding somatostatin concentration, consistent with both the known effect of somatostatin to cause sst2A receptor internalization and an autocrine regulation of tumors by the peptide they produce. Moreover, our demonstration that the sst2A receptor can be identified in this group of tumors using simple immunohistochemical methods in formalin-fixed, paraffin-embedded material opens numerous diagnostic, therapeutic, and prognostic opportunities.


Subject(s)
Nervous System Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Somatostatin/metabolism , Subcellular Fractions/metabolism , Autoradiography , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Intracellular Membranes/metabolism , Membranes/metabolism , Octreotide , Somatostatin/biosynthesis
15.
Cell Mol Biol (Noisy-le-grand) ; 46(6): 1053-63, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10976863

ABSTRACT

The application of synchrotron radiation in medical research has become a mature field of research at synchrotron facilities worldwide. In the relatively short time that synchrotrons have been available to the scientific community, their characteristic beams of UV and X-ray radiation have been applied to virtually all areas of medical science which use ionizing radiation. The ability to tune intense monochromatic beams over wide energy ranges differentiates these sources from standard clinical and research tools. At the European Synchrotron Radiation Facility (Grenoble, France), a major research facility is operational on an advanced wiggler radiation beamport, ID17. The beamport is designed to carry out a broad range of research ranging from cell radiation biology to in vivo human studies. Medical imaging programs at ID17 include transvenous coronary angiography, computed tomography, mammography and bronchography. In addition, a major research program on microbeam radiation therapy is progressing. This paper will present a very brief overview of the beamline and the imaging and therapy programs.


Subject(s)
Synchrotrons/instrumentation , Animals , Coronary Angiography/instrumentation , Europe , Humans , Radiotherapy, High-Energy/instrumentation , Research , Tomography, X-Ray Computed/instrumentation , X-Ray Diffraction/instrumentation
16.
Med Phys ; 27(7): 1664-75, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10947271

ABSTRACT

Microbeam radiation therapy (MRT) is a currently experimental method of radiotherapy which is mediated by an array of parallel microbeams of synchrotron-wiggler-generated x-rays. Suitably selected, nominally supralethal doses of x-rays delivered to parallel microslices of tumor-bearing tissues in rats can be either palliative or curative while causing little or no serious damage to contiguous normal tissues. Although the pathogenesis of MRT-mediated tumor regression is not understood, as in all radiotherapy such understanding will be based ultimately on our understanding of the relationships among the following three factors: (1) microdosimetry, (2) damage to normal tissues, and (3) therapeutic efficacy. Although physical microdosimetry is feasible, published information on MRT microdosimetry to date is computational. This report describes Monte Carlo-based computational MRT microdosimetry using photon and/or electron scattering and photoionization cross-section data in the 1 eV through 100 GeV range distributed publicly by the U.S. Lawrence Livermore National Laboratory (LLNL) in the 1990s. These are compared with Monte Carlo-based microdosimetric computations using a code and physical data available in the 1980s. With the aim of using the PSI-version of GEANT Monte Carlo code for future macro- and micro/nano-dosimetric studies of Microbeam Radiation Therapy (MRT) a comparison of this code is made with the INHOM(EGS4) (version 1990), Dilmanian-CPE and Persliden-CPE Monte Carlo photon-electron codes (both version 1990) with which the absorbed dose distributions were calculated in 1990 and 1991 considering, (a) a single cylindrical microbeam, (b) multiple cylindrical microbeams in an orthogonal square bundle, and (c) multiple planar microbeams. It is shown that the PSI-version of GEANT can potentially deliver more accurate results (a) using presently the most advanced atomic data, and especially (b) employing "Single-collision" electron transport instead of only the "Condensed-history" electron transport as in code INHOM(EGS4). In contrast Dilmanian-CPE and Persliden-CPE codes deposit the electron energy locally instead of transporting it to the correct position.


Subject(s)
Radiotherapy/instrumentation , Radiotherapy/methods , Algorithms , Carbon , Dose-Response Relationship, Radiation , Electron Transport , Electrons/therapeutic use , Humans , Hydrogen , Ions , Monte Carlo Method , Neoplasms, Experimental/radiotherapy , Nitrogen , Oxygen , Phantoms, Imaging , Photons/therapeutic use , Radiometry/methods , Scattering, Radiation , Software , Water , X-Rays
17.
Hum Pathol ; 31(7): 854-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10923924

ABSTRACT

Follicular neoplasms of oncocytic type in the thyroid gland frequently cause diagnostic problems and prognostic uncertainties. To identify numerical chromosomal aberrations of possible pathogenetic importance, we determined chromosome copy numbers in situ in interphase nuclei of 31 oncocytic adenomas and 25 oncocytic carcinomas. Archival formaldehyde-fixed, paraffin-embedded tumor samples and normal control thyroid tissues were arranged in arrays and analyzed by fluorescence in situ hybridization (FISH). We used pericentromeric or locus specific probes for chromosomes 1, 7, 8, 9, 11, 12, 17, 18, 22, and X as well as for the oncogenes Her2/neu, cyclin D1, N-myc, and c-myc. The average number of aneusomies per nucleus was significantly higher in carcinomas than in adenomas, and in both, monosomies were more frequent than polysomies. Loss of chromosome 22 was found in 8 of 21 (38%) carcinomas; in 5 cases, it was associated with chromosome 2 monosomy. Conversely, chromosome 2 aberrations were not found in adenomas. Monosomies for chromosome 8 and X were detected in most adenomas and carcinomas. The most common gains in adenomas and carcinomas were for chromosome 7 (13.8% and 32.0% of the cases, respectively), chromosome 12 (9.6% and 12.0%), and chromosome 17 (19.3% and 32.0%). None of the adenomas with trisomy 17 was associated with gains for chromosomes 7 and 12. None of the analyzed oncogenes was found to be amplified by FISH analysis. Our results indicate that numerical chromosomal aberrations in oncocytic follicular tumors of the thyroid gland are common findings and suggest that different patterns of aberrations may occur in these neoplasms.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Chromosome Aberrations , Cytogenetics , Interphase , Thyroid Neoplasms/genetics , Adult , Aged , DNA Probes , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Oncogenes
18.
Clin Nephrol ; 54(1): 64-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10939759

ABSTRACT

The relationship between lipid abnormalities and the pathogenesis of renal disease is still unclear. Although most patients with primary hyperlipidemia do not develop renal function impairment, experimental and clinical data indicate a possible damaging effect of a disturbed lipid metabolism on the kidney. We report the case history of a patient with hyperlipidemia and mild nephropathy in which an accidentally removed kidney showed intrarenal arteriosclerosis which occured before the development of other cardiovascular risk factors, indicating that primary dyslipidemia induced nephroangiosclerosis.


Subject(s)
Hyperlipidemias/complications , Kidney Diseases/etiology , Arteriosclerosis/complications , Arteriosclerosis/pathology , Humans , Hypertension, Renal/drug therapy , Hypertension, Renal/etiology , Kidney/pathology , Kidney Diseases/pathology , Male , Middle Aged
19.
Cancer Res ; 60(11): 3105-12, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850463

ABSTRACT

The evaluation of peptide receptors in man is needed not only to discover the physiological target tissues of a given peptide but also to identify diseases with a sufficient receptor overexpression for diagnostic or therapeutic interventions. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) receptors have been evaluated in human tumors and in their tissues of origin using in vitro receptor autoradiography with 125I-VIP or 125I-acetyl-PACAP-27 in tissue sections. The VIP/PACAP receptor subtypes VPAC1, VPAC2, and PAC1 were evaluated in these tissues by determining the rank order of potencies of VIP and PACAP as well as VPAC1- and VPAC2-selective analogues. The VIP/PACAP receptors expressed in the great majority of the most frequently occurring human tumors, including breast (100% receptor incidence), prostate (100%), pancreas (65%), lung (58%), colon (96%), stomach (54%), liver (49%), and urinary bladder (100%) carcinomas as well as lymphomas (58%) and meningiomas (100%), are predominantly of the VPAC1 type. Their cells or tissues of origin, i.e., hepatocytes, breast lobules and ducts, urothelium, prostate glands, pancreatic ducts, lung acini, gastrointestinal mucosa, and lymphocytes, also predominantly express VPAC1. Leiomyomas predominantly express VPAC2 receptors, whereas paragangliomas, pheochromocytomas, and endometrial carcinomas preferentially express PAC1 receptors. Conversely, VPAC2 receptors are found mainly in smooth muscle (i.e., stomach), in vessels, and in stroma (e.g., of the prostate), whereas PAC1 receptors are present in the adrenal medulla and in some uterine glands. Whereas the very wide distribution of VIP/PACAP receptors in the normal human body is indicative of a key role of these peptides in human physiology, the high VIP/PACAP receptor expression in tumors may represent the molecular basis for clinical applications of VIP/PACAP such as in vivo scintigraphy and radiotherapy of tumors as well as VIP/PACAP analogue treatment for tumor growth inhibition.


Subject(s)
Neoplasms/metabolism , Receptors, Pituitary Hormone/biosynthesis , Receptors, Vasoactive Intestinal Peptide/biosynthesis , Adrenal Gland Neoplasms/metabolism , Adrenal Medulla/metabolism , Autoradiography , Breast/metabolism , Breast Neoplasms/metabolism , Dose-Response Relationship, Drug , Female , Gastric Mucosa/metabolism , Humans , Leiomyoma/metabolism , Lymph Nodes/metabolism , Male , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Peptides/metabolism , Pheochromocytoma/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Stomach Neoplasms/metabolism
20.
Int J Cancer ; 86(6): 777-81, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10842190

ABSTRACT

A variable fraction of anaplastic large-cell lymphomas (ALCLs) exhibits a t(2;5)(p23;q35) translocation that results in expression of the chimeric hyperphosphorylated protein NPM-ALK (p80). Tumor cells expressing NPM-ALK exhibit markedly enhanced proliferative activity, but comparative cellular kinetic studies on ALK(+) (ALK lymphomas) and ALK(-) lymphomas are lacking. The present study showed that ALK(+) lymphomas, detected with the monoclonal antibody ALKc (n = 17), had significantly higher average values for the proliferation-associated parameters mitotic index, ana/telophase index, growth index (x x mitotic index - apoptotic index, assuming x = 3), percentages of Ki-67(+) cells and fraction of cells expressing cyclin A or B or the cell cycle-regulatory protein p34(cdc2) than did ALK(-) ALCLs (n = 15). Whether this intense proliferative activity contributes to the good response to chemotherapy and favorable outcome of ALK(+) ALCLs remains to be assessed in a larger series of patients. Our findings support the notion that ALK(+) and ALK(-) ALCLs are 2 distinct disease entities.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/pathology , Protein-Tyrosine Kinases/analysis , Adolescent , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , Cell Division , Child , Cyclin A/analysis , Cyclin B/analysis , Female , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Middle Aged , Mitotic Index , Receptor Protein-Tyrosine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...