Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Med ; 12(23): 21075-21096, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37997517

RESUMEN

BACKGROUND: Pediatric brain tumors (PBT) stand as the leading cause of cancer-related deaths in children. Chemoradiation protocols have improved survival rates, even for non-resectable tumors. Nonetheless, radiation therapy carries the risk of numerous adverse effects that can have long-lasting, detrimental effects on the quality of life for survivors. The pursuit of chemotherapeutics that could obviate the need for radiotherapy remains ongoing. Several anti-tumor agents, including sunitinib, valproic acid, carboplatin, and panobinostat, have shown effectiveness in various malignancies but have not proven effective in treating PBT. The presence of the blood-brain barrier (BBB) plays a pivotal role in maintaining suboptimal concentrations of anti-cancer drugs in the central nervous system (CNS). Ongoing research aims to modulate the integrity of the BBB to attain clinically effective drug concentrations in the CNS. However, current findings on the interaction of exogenous chemical agents with the BBB remain limited and do not provide a comprehensive explanation for the ineffectiveness of established anti-cancer drugs in PBT. METHODS: We conducted our search for chemotherapeutic agents associated with the blood-brain barrier (BBB) using the following keywords: Chemotherapy in Cancer, Chemotherapy in Brain Cancer, Chemotherapy in PBT, BBB Inhibition of Drugs into CNS, Suboptimal Concentration of CNS Drugs, PBT Drugs and BBB, and Potential PBT Drugs. We reviewed each relevant article before compiling the information in our manuscript. For the generation of figures, we utilized BioRender software. FOCUS: We focused our article search on chemical agents for PBT and subsequently investigated the role of the BBB in this context. Our search criteria included clinical trials, both randomized and non-randomized studies, preclinical research, review articles, and research papers. FINDING: Our research suggests that, despite the availability of potent chemotherapeutic agents for several types of cancer, the effectiveness of these chemical agents in treating PBT has not been comprehensively explored. Additionally, there is a scarcity of studies examining the role of the BBB in the suboptimal outcomes of PBT treatment, despite the effectiveness of these drugs for other types of tumors.


Asunto(s)
Antineoplásicos , Neoplasias Encefálicas , Niño , Humanos , Barrera Hematoencefálica , Calidad de Vida , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Antineoplásicos/uso terapéutico
2.
Cell Death Discov ; 9(1): 162, 2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37179350

RESUMEN

Metastatic breast cancer (mBC) is responsible for >90% of breast cancer-related deaths. Microtubule-targeting agents (MTAs) are the front-line treatment for mBC. However, the effectiveness of MTAs is frequently limited by the primary or acquired resistance. Furthermore, recurrent mBC derived from cancer cells that survived MTA treatment are typically more chemoresistant. The overall response rates for the second- and third-line MTAs in mBC patients previously treated with MTAs are 12-35%. Thus, there is an ongoing search for novel MTAs with a distinct mode of action that can circumvent chemoresistance mechanisms. Our results show that methyl N-(6-benzoyl-1H-benzimidazol-2-yl)carbamate (BCar), a microtubule-disrupting anthelmintic that binds to the colchicine binding site separate from the binding sites of clinically used MTAs, has the potential to treat MTA-resistant mBC. We have comprehensively evaluated the cellular effects of BCar in a panel of human breast cancer (BC) cell lines and normal breast cells. BCar effects on the clonogenic survival, cell cycle, apoptosis, autophagy, senescence, and mitotic catastrophe were measured. Approximately 25% of BCs harbor mutant p53. For this reason, the p53 status was included as a variable. The results show that BC cells are >10x more sensitive to BCar than normal mammary epithelial cells (HME). p53-mutant BC cells are significantly more sensitive to BCar treatment than p53 wild-type BC cells. Furthermore, BCar appears to kill BC cells primarily via either p53-dependent apoptosis or p53-independent mitotic catastrophe. When compared to docetaxel and vincristine, two clinical MTAs, BCar is fairly innocuous in HME cells, providing a much wider therapeutic window than docetaxel and vincristine. Together, the results strongly support the notion that BCar-based therapeutics may serve as a new line of MTAs for mBC treatment.

3.
J Pediatr Hematol Oncol ; 44(6): 293-304, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486544

RESUMEN

The physiological functions of butyrylcholinesterase (BChE) and its role in malignancy remain unexplained. Our studies in children newly diagnosed with neuroblastoma indicated that BChE expressions is proportional to MYCN amplification suggesting that pathogenesis of high-risk disease may be related to the persistent expression of abnormally high levels of tumor-associated BChE. BChE-deficient neuroblastoma cells (KO [knockout]) were produced from MYCN -amplified BE(2)-C cells (WT [wild-type]) by the CRISPR-Cas9 targeted disruption of the BCHE locus. KO cells have no detectable BChE activity. The compensatory acetylcholinesterase activity was not detected. The average population doubling time of KO cells is 47.0±2.4 hours, >2× longer than WT cells. Reduced proliferation rates of KO cells were accompanied by the loss of N-Myc protein and a significant deactivation of tyrosine kinase receptors associated with the aggressive neuroblastoma phenotype including Ros1, TrkB, and Ltk. Tumorigenicity of WT and KO cells in male mice was essentially identical. In contrast, KO xenografts in female mice were very small (0.37±0.10 g), ~3× smaller compared with WT xenografts (1.11±0.30 g). Unexpectedly, KO xenografts produced changes in plasma BChE similarly to WT tumors but lesser in magnitude. The disruption of BCHE locus in MYCN -amplified neuroblastoma cells decelerates proliferation and produces neuroblastoma cells that are less aggressive in female mice.


Asunto(s)
Butirilcolinesterasa , Neuroblastoma , Acetilcolinesterasa/genética , Animales , Butirilcolinesterasa/genética , Femenino , Humanos , Masculino , Ratones , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas
4.
Artículo en Inglés | MEDLINE | ID: mdl-32150284

RESUMEN

Neuroblastoma, the most common extracranial solid tumor in children, accounts for nearly 8% of childhood cancers in the United States. It is a disease with pronounced clinical and biological heterogeneities. The amplification of MYCN, whose key tumorigenic functions include the promotion of proliferation, facilitation of the cell's entry into the S phase, and prevention of cells from leaving the cell cycle, correlates with poor prognosis. Patients with a high proliferation index disease have low survival rates. Neuroblastoma is one of the most radioresponsive of all human tumors. To exploit this radiosensitivity, radioactive guanidine (R)-(-)-5-[125 I]iodo-3'-O-[2-(ε-guanidinohexanoyl)-2-phenylacetyl]-2'-deoxyuridine (9, GPAID) was designed. This compound enters neuroblastoma cells much like metaiodobenzylguanidine (MIBG). Additionally, it cotargets DNA of proliferating cells, an attribute especially advantageous in the treatment of MYCN-amplified tumors. GPAID was synthesized from the trimethylstannyl precursor with an average yield of >90% at the no-carrier-added specific activities. The norepinephrine transporter-aided delivery of GPAID to neuroblastoma cells was established in the competitive uptake studies with nonradioactive MIBG. The intracellular processing and DNA targeting properties were confirmed in the subcellular distribution experiments. Studies in a mouse model of neuroblastoma demonstrated the therapeutic potential of GPAID. The tin precursor of GPAID can be used to prepare compounds radiolabeled with single-photon emission computed tomography (SPECT)- and positron-emission tomography (PET)-compatible radionuclides. Accordingly, these reagents can function as theranostics useful in the individualized and comprehensive treatment strategies comprising treatment planning and the assessment of tumor responses as well as the targeted molecular radiotherapy employing treatment doses derived from the imaging data.

5.
Cancer Biother Radiopharm ; 35(6): 418-424, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32073902

RESUMEN

Osteosarcoma (OST) is the most common bone tumor in children and adolescents with a second peak of incidence in elderly adults usually diagnosed as secondary tumors in Paget's disease or irradiated bone. Subjects with metastatic disease or whose disease relapses after the initial therapy have a poor prognosis. Moreover, multifocal OST contains tumor-initiating cells that are resistant to chemotherapy. The use of aggressive therapies in an attempt to eradicate these cells can have long-term negative consequences in these vulnerable patient populations. 227Th-labeled molecular probes based on ligands to OST-associated receptors such as IGF-1R (insulin-like growth factor receptor 1), HER2 (human epidermal growth factor receptor 2), and PSMA (prostate-specific membrane antigen) are expected to detect and treat osseous and nonosseous sites of multifocal OST. Published reports indicate that 227Th has limited myelotoxicity, can be stably chelated to its carriers and, as it decays at targeted sites, 227Th produces 223Ra that is subsequently incorporated into the areas of increased osteoblastic activity, that is, osseous metastatic lesions. Linear energy transfer of α particles emitted by 227Th and its daughter 223Ra is within the range of the optimum relative biological effectiveness. The radiotoxicity of α particles is virtually independent of the phase in the cell cycle, oxygenation, and the dose rate. For these reasons, even resistant OST cells remain susceptible to killing by high-energy α particles, which can also kill adjacent quiescent OST cells or cells with low expression of targeted receptors. Systemic side effects are minimized by the limited range of these intense radiations. Quantitative single-photon emission computed tomography of 227Th and 223Ra is feasible. Additionally, the availability of radionuclide pairs, for example, 89Zr for positron emission tomography and 227Th for therapy, establish a strong basis for the theranostic use of 227Th in the individualized treatment of multifocal OST.


Asunto(s)
Partículas alfa/uso terapéutico , Neoplasias Óseas/radioterapia , Osteosarcoma/radioterapia , Radiofármacos/administración & dosificación , Nanomedicina Teranóstica/métodos , Adolescente , Antígenos de Superficie , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Niño , Relación Dosis-Respuesta en la Radiación , Portadores de Fármacos/química , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Humanos , Terapia Molecular Dirigida/efectos adversos , Terapia Molecular Dirigida/métodos , Nanopartículas/química , Osteosarcoma/mortalidad , Osteosarcoma/patología , Pronóstico , Radiofármacos/efectos adversos , Receptor ErbB-2/antagonistas & inhibidores , Receptor IGF Tipo 1/antagonistas & inhibidores , Torio/administración & dosificación , Torio/efectos adversos
6.
Drug Dev Res ; 81(1): 62-69, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31593323

RESUMEN

The role of theranostics in cancer management is growing so is the selection of vectors used to deliver these modalities to cancer cells. We describe biological evaluation of a novel theranostic agent targeted to microtubules. Methyl N-[5-(3'-[131 I]iodobenzoyl)-1H-benzimidazol-2-yl]carbamate (1) and methyl N-[5-(3'-[125 I]iodobenzoyl)-1H-benzimidazol-2-yl]carbamate (2) were synthesized from a common precursor 3'-stannylated derivative (4). Antiproliferative effects and radiotoxicity of 131 I-labeled ß-particle emitting 1 were examined in vitro in human neuroblastoma and glioblastoma cells lines. The therapeutic potential of 1 was also examined in a subcutaneous mouse model of human glioblastoma U-87 MG. Compound 1 at the extracellular radioactive concentration of 0.35 MBq/mL, easily achievable in vivo, kills >90% of neuroblastoma cells and >60% glioblastoma cells as measured in a clonogenic assay. D10 doses established for 1 indicate that as few as 3,000 decays are sufficient to kill 90% of BE(2)-C cells. Even U-87 MG cells, the least sensitive of the tested cell lines, require <20,000 decays of intracellular 131 I to reduce number of clonogenic cells by 90%. Biodistribution studies of 2 delivered either intratumorally or intraperitoneally show a similar tissue distribution for both routes of the drug administration. The whole body clearance half-lives were on average 6 hr. Intratumor administration of 1 produces significant tumor growth delay. After a single dose of 8.4 ± 0.3 MBq of compound 1, the tumor doubling times were 3.2 ± 0.1 and 7.9 ± 0.6 days in control and treated mice, respectively. Methyl N-[5-(3'-radiohalobenzoyl)-1H-benzimidazol-2-yl]carbamates have properties compatible with a theranostic approach to cancer management.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Carbamatos/administración & dosificación , Glioblastoma/radioterapia , Radioisótopos de Yodo/química , Animales , Carbamatos/química , Carbamatos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Humanos , Inyecciones Intraperitoneales , Ratones , Nanomedicina Teranóstica , Distribución Tisular , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Med Chem ; 63(5): 2051-2073, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-31268317

RESUMEN

High risk neuroblastoma often recurs, even with aggressive treatments. Clinical evidence suggests that proliferative activities are predictive of poor outcomes. This report describes syntheses, characterization, and biological properties of theranostic guanidines that target norepinephrine transporter and undergo intracellular processing, and subsequently their catabolites are efficiently incorporated into DNA of proliferating neuroblastoma cells. Radioactive guanidines are synthesized from 5-radioiodo-2'-deoxyuridine, a molecular radiotherapy platform with clinically proven minimal toxicities and DNA-targeting properties. The transport of radioactive guanidines into neuroblastoma cells is active as indicated by the competitive suppression of cellular uptake by meta-iodobenzylguanidine. The rate of intracellular processing and DNA uptake is influenced by the agent's catabolic stability and cell population doubling times. The radiotoxicity is directly proportional to DNA uptake and duration of exposure. Biodistribution of 5-[125I]iodo-3'-O-(ε-guanidinohexanoyl)-2'-deoxyuridine in a mouse neuroblastoma model shows significant tumor retention of radioactivity. Neuroblastoma xenografts regress in response to the clinically achievable doses of this agent.


Asunto(s)
ADN/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Guanidinas/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Nanomedicina Teranóstica/métodos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Guanidinas/administración & dosificación , Humanos , Masculino , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Cancer Biother Radiopharm ; 35(1): 16-25, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31687840

RESUMEN

Background: Resistance of cancer to chemo- and radiotherapy remains a major clinical problem. This study contributes to the ongoing search for agents that can bypass this resistance by developing a novel antimitotic theranostic. Materials and Methods: Methyl N-[5-(3'-iodobenzoyl)-1H-benzimidazol-2-yl]carbamates 1 and 2 were synthesized from a common precursor 3 or its 3'-stannylated derivative. The cytotoxicity of compound 1 was evaluated in several neuroblastoma and glioblastoma cell lines and in the NCI 60-cell assay. Biodistribution was conducted in mice after oral administration of compound 2 to determine tissue and brain uptake. Result: Lethal concentrations (LC50s) of compound 1 in neuroblastoma and glioblastoma are >15 × lower compared with compound 3, a drug currently tested in clinical studies in pediatric and adult brain tumors. Growth inhibition concentrations (GI50) are in the nanomolar range in 60 cancer cell lines. When compound 1 is combined with a 4-Gy dose of radiation, <0.5% of cells retain their reproductive integrity. Increased hydrophobicity of new agents greatly enhances their brain uptake after oral administration. Conclusions: Compound 1 is potently cytotoxic in a wide range of human cancer cell lines. Its structure allows incorporation of imaging and therapeutic radionuclides. It is therefore expected that compound 1 can be developed into a novel theranostic modality across a wide range of malignancies.


Asunto(s)
Antineoplásicos/uso terapéutico , Carbamatos/uso terapéutico , Animales , Antineoplásicos/farmacología , Carbamatos/farmacología , Línea Celular Tumoral , Humanos , Ratones , Relación Estructura-Actividad
9.
Artículo en Inglés | MEDLINE | ID: mdl-29707803

RESUMEN

Microtubules are a target for a broad spectrum of drugs used as chemotherapeutics to treat hematological malignancies and solid tumors. Most of these drugs have significant dose-limiting toxicities including peripheral neuropathies that can be debilitating and permanent. In an ongoing effort to develop safer and more effective drugs, benzimidazole-based compounds are being developed as replacement for vincristine and similar agents. In this report, we describe radiosyntheses of novel microtubule-targeting methyl N-[5-(3'-radiohalobenzoyl)-1H-benzimidazol-2-yl]carbamates 4 that are intended as potential imaging agents and molecular radiotherapeutics. 125 I- and 131 I-radiolabeled derivatives were prepared either by direct radioiodination of methyl N-(6-benzoyl-1H-benzimidazol-2-yl)carbamate 1 or radioiododestannylation of the corresponding stannane precursor 3. The direct radioiodination was conducted in a solution of 1 in triflic acid and produced after ~1 hour at elevated temperatures and HPLC purification on average 62% of the no-carrier added products 125 I-4 and 131 I-4. Radioiododestannylation of 3'-trimethylstannane 3 proceeded with ease at room temperature in the presence of H2 O2 as the oxidant and produced no-carrier-added 125 I-4 and 131 I-4 in high isolated yields, on average 85%. The radiohalodestannylation protocol is universal and can be applied to other radiohalides including 124 I to produce 124 I-4, a positron emission tomography agent, and 211 At to produce 211 At-4, an α-particle emitting radiotherapeutic.

10.
J Pediatr Hematol Oncol ; 39(4): 272-281, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28375942

RESUMEN

Blood-based biomarkers are important in the detection of the disease and in the assessment of responses to therapy. In this study, butyrylcholinesterase was evaluated as a potential biomarker in newly diagnosed neuroblastoma (NB) patients at diagnosis and longitudinally during treatment. Plasma butyrylcholinesterase activities in age-matched and sex-matched children were used as controls. Pretreatment butyrylcholinesterase levels in NB subjects are on an average 2 times lower than butyrylcholinesterase levels in healthy subjects. Significantly, butyrylcholinesterase activities are ∼40% lower in MYCN-amplified as compared with nonamplified disease. As the course of chemotherapy progresses, butyrylcholinesterase activities recover and normalize to control values. The evident response to treatment indicates that plasma butyrylcholinesterase is a good biomarker of tumor response to therapy. Depressed butyrylcholinesterase levels in NB subjects are not caused by hepatic deficits suggesting a specific role for butyrylcholinesterase in NB. Further examination of the mechanism of altered butyrylcholinesterase production require an animal model that best approximates human condition. Studies in mice show that murine NB allografts significantly reduce butyrylcholinesterase activity in plasma. This finding correlates with changes observed in NB patients. In contrast, human NB xenografts produce the opposite effect, that is, butyrylcholinesterase plasma levels rise as the xenograft size increases. In the absence of any liver damage, dissimilarities between butyrylcholinesterase production in murine and human NB models suggest species-specific signaling pathways. This disparity also suggests that human NB xenograft mouse models do not approximate the human disease.


Asunto(s)
Biomarcadores/sangre , Butirilcolinesterasa/sangre , Neuroblastoma/diagnóstico , Animales , Estudios de Casos y Controles , Preescolar , Modelos Animales de Enfermedad , Femenino , Xenoinjertos/normas , Humanos , Lactante , Masculino , Ratones , Neuroblastoma/sangre , Transducción de Señal , Especificidad de la Especie
11.
Prostate ; 75(1): 8-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283970

RESUMEN

BACKGROUND: The androgen receptor (AR) plays a dominant role in the pathogenesis of prostate cancer. 5-Radioiodo-3'-O-(17ß-succinyl-5α-androstan-3-one)-2'-deoxyuridin-5'-yl phosphate (RISAD-P) is an AR-targeting reagent developed for noninvasive assessment of AR and proliferative status of the AR-expressing tumors, and for molecular radiotherapy with Auger electron-emitting radionuclides. In this study, the preclinical toxicity and targeting potential of RISAD-P was evaluated. METHODS: Effects of nonradioactive ISAD-P and RISAD-P labeled with (123) I, (124) I, and (125) I were evaluated in male mice. Expanded-acute single dose toxicity studies, hematologic toxicity, liver and kidney function, pharmacokinetics, biodistribution, and imaging studies were conducted. Imaging and pilot therapy studies were conducted in transgenic mice. RESULTS: RISAD-P is not toxic at doses projected for clinical use. Its tissue distribution compares favorably with the distribution reported for (18) F-dihydrotestosterone derivatives. RISAD-P has excellent prostate cancer targeting properties. One hour after (125) IRISAD-P administration, nearly 10% of the injected dose is associated with prostate tumor. The tumor clearance is biphasic and plateaus between 24 and 48 hr post-injection. The estimated radiation doses calculated for 1 g tumor using the MIRD convention are well within the therapeutic range with values of 170, 250, 1,240 Gy × MBq(-1) × g(-1) for (125) I-, (123) I-, and (124) I-labeled RISAD-P, respectively. The transient uptake of radioactivity is observed in the genitourinary tract and stomach. Without the potassium iodide blockade, thyroid uptake is also observed. CONCLUSIONS: Biodistribution, toxicity, and radiation dosimetry studies suggest that RISAD-P holds characteristics of a promising candidate for imaging of AR expression and tumor proliferation, as well as molecular radiotherapy for metastatic or locally, regionally advanced prostate cancer.


Asunto(s)
Androstanoles/toxicidad , Nucleótidos de Desoxiuracil/toxicidad , Radioisótopos de Yodo , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/radioterapia , Receptores Androgénicos/metabolismo , Androstanoles/farmacocinética , Animales , Nucleótidos de Desoxiuracil/farmacocinética , Evaluación Preclínica de Medicamentos , Drogas en Investigación , Masculino , Ratones , Ratones Transgénicos , Proyectos Piloto , Radiofármacos , Dosificación Radioterapéutica , Distribución Tisular
12.
Prostate ; 74(16): 1634-46, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25214432

RESUMEN

BACKGROUND: The androgen receptor (AR) axis, the key growth and survival pathway in prostate cancer, remains a prime target for drug development. 5-Radioiodo-3'-O-(17ß-succinyl-5α-androstan-3-one)-2'-deoxyuridin-5'-yl phosphate (RISAD-P) is the AR-seeking reagent developed for noninvasive assessment of AR and proliferative status, and for molecular radiotherapy of prostate cancer with Auger electron-emitting radionuclides. METHODS: RISAD-P radiolabeled with 123I, 124I, and 125I were synthesized using a common stannylated precursor. The cellular uptake, subcellular distribution, and radiotoxicity of 123I-, 124I-, and (125) IRISAD-P were measured in LNCaP, DU145, and PC-3 cell lines expressing various levels of AR. RESULTS: The uptake of RISAD-P by prostate cancer cells is proportional to AR levels and independent of the radionuclide. The intracellular accumulation of radioactivity is directly proportional to the extracellular concentration of RISAD-P and the duration of exposure. Initially, RISAD-P is trapped in the cytoplasm. Within 24 hr, radioactivity is associated exclusively with DNA. The RISAD-P radiotoxicity is determined by the radionuclide; however, the cellular responses are directly proportional to the AR expression levels. LNCaP cells expressing high levels of AR are killed at the rate of up to 60% per day after a brief 1 hr RISAD-P treatment. For the first time, the AR expression in PC-3 and DU 145 cells, generally reported as AR-negative, was quantitated by the ultra sensitive RISAD-P-based method. CONCLUSIONS: RISAD-P is a theranostic drug, which targets AR. Its subcellular metabolite participates in DNA synthesis. RISAD-P is a promising candidate for imaging of the AR expression and tumor proliferation as well as molecular radiotherapy of prostate cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/radioterapia , ADN de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Receptores Androgénicos/metabolismo , Adenocarcinoma/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Técnicas In Vitro , Radioisótopos de Yodo , Masculino , Terapia Molecular Dirigida , Próstata/diagnóstico por imagen , Próstata/patología , Neoplasias de la Próstata/patología , Cintigrafía , Radioterapia Guiada por Imagen
13.
Int J Radiat Oncol Biol Phys ; 88(2): 404-411, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24315566

RESUMEN

BACKGROUND: Human liver has an unusual sensitivity to radiation that limits its use in cancer therapy or in preconditioning for hepatocyte transplantation. Because the characteristic veno-occlusive lesions of radiation-induced liver disease do not occur in rodents, there has been no experimental model to investigate the limits of safe radiation therapy or explore the pathogenesis of hepatic veno-occlusive disease. METHODS AND MATERIALS: We performed a dose-escalation study in a primate, the cynomolgus monkey, using hypofractionated stereotactic body radiotherapy in 13 animals. RESULTS: At doses ≥40 Gy, animals developed a systemic syndrome resembling human radiation-induced liver disease, consisting of decreased albumin, elevated alkaline phosphatase, loss of appetite, ascites, and normal bilirubin. Higher radiation doses were lethal, causing severe disease that required euthanasia approximately 10 weeks after radiation. Even at lower doses in which radiation-induced liver disease was mild or nonexistent, latent and significant injury to hepatocytes was demonstrated by asialoglycoprotein-mediated functional imaging. These monkeys developed hepatic failure with encephalopathy when they received parenteral nutrition containing high concentrations of glucose. Histologically, livers showed central obstruction via an unusual intimal swelling that progressed to central fibrosis. CONCLUSIONS: The cynomolgus monkey, as the first animal model of human veno-occlusive radiation-induced liver disease, provides a resource for characterizing the early changes and pathogenesis of venocclusion, for establishing nonlethal therapeutic dosages, and for examining experimental therapies to minimize radiation injury.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad Veno-Oclusiva Hepática/etiología , Hepatocitos/efectos de la radiación , Hígado/efectos de la radiación , Macaca fascicularis , Traumatismos Experimentales por Radiación/etiología , Alanina Transaminasa/análisis , Albúminas/análisis , Fosfatasa Alcalina/análisis , Animales , Peso Corporal/efectos de la radiación , Fraccionamiento de la Dosis de Radiación , Enfermedad Veno-Oclusiva Hepática/diagnóstico por imagen , Enfermedad Veno-Oclusiva Hepática/patología , Hepatocitos/diagnóstico por imagen , Hepatocitos/patología , Hígado/diagnóstico por imagen , Hígado/patología , Hígado/cirugía , Fallo Hepático Agudo/etiología , Masculino , Dosis de Radiación , Traumatismos Experimentales por Radiación/diagnóstico por imagen , Traumatismos Experimentales por Radiación/patología , Radiocirugia/efectos adversos , Retratamiento , Tomografía Computarizada de Emisión de Fotón Único/métodos
14.
J Med Chem ; 55(6): 2649-71, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22339166

RESUMEN

Targeted molecular radiotherapy opens unprecedented opportunities to eradicate cancer cells with minimal irradiation of normal tissues. Described in this study are radioactive cyclosaligenyl monophosphates designed to deliver lethal doses of radiation to cancer cells. These compounds can be radiolabeled with SPECT- and PET-compatible radionuclides as well as radionuclides suitable for Auger electron therapies. This characteristic provides an avenue for the personalized and comprehensive treatment strategy that comprises diagnostic imaging to identify sites of disease, followed by the targeted molecular radiotherapy based on the imaging results. The developed radiosynthetic methods produce no-carrier-added products with high radiochemical yield and purity. The interaction of these compounds with their target, butyrylcholinesterase, depends on the stereochemistry around the P atom. IC(50) values are in the nanomolar range. In vitro studies indicate that radiation doses delivered to the cell nucleus are sufficient to kill cells of several difficult to treat malignancies including glioblastoma and ovarian and colorectal cancers.


Asunto(s)
Neoplasias/radioterapia , Radiofármacos/síntesis química , Timidina Monofosfato/análogos & derivados , Timidina Monofosfato/síntesis química , Uridina Monofosfato/análogos & derivados , Uridina Monofosfato/síntesis química , Butirilcolinesterasa/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/farmacología , Neoplasias Colorrectales , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Glioblastoma , Humanos , Hidrólisis , Radioisótopos de Yodo , Terapia Molecular Dirigida , Neoplasias/diagnóstico por imagen , Neoplasias/enzimología , Neoplasias Ováricas , Cintigrafía , Radiofármacos/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Timidina Monofosfato/farmacología , Uridina Monofosfato/farmacología
15.
Immunotherapy ; 3(4): 491-4, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21463190

RESUMEN

Evaluation of: Boudousq V, Ricaud S, Garambois V et al.: Brief intraperitoneal radioimmunotherapy of small peritoneal carcinomatosis using high activities of noninternalizing (125)I-labeled monoclonal antibodies. J. Nucl. Med. 51, 1748-1755 (2010). Mesothelioma, pseudomyxoma peritonei, ovarian and colon cancers, and several other malignancies produce peritoneal carcinomatosis in their advanced stages. This condition is typically managed with curative intention by means of a radical cytoreductive surgery and intraperitoneal hyperthermic chemotherapy. The method has been shown to improve the survival of some patients with peritoneal dissemination. However, the intracavitary instillation of chemotherapy results in a nonuniform drug distribution and allows penetration of the drug only into the outermost layer of the cancer nodule. Authors of the current study demonstrate that (125)I-radiolabeled monoclonal antibody, 35A7, which recognizes anti-carcinoembryonic antigen, can be used effectively and safely with low toxicity in the therapy of small volume peritoneal carcinomatosis after cytoreductive surgery.

16.
Cancers (Basel) ; 3(2): 2501-15, 2011 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-24212821

RESUMEN

Pancreatic cancer does not respond to a single-agent imatinib therapy. Consequently, multimodality treatments are contemplated. Published data indicate that in colorectal cancer, imatinib and radioimmunotherapy synergize to delay tumor growth. In pancreatic cancer, the tumor response is additive. This disparity of outcomes merited further studies because interactions between these modalities depend on the imatinib-induced reduction of the tumor interstitial fluid pressure. The examination of human and murine PDGFr-ß/PDGF-B pathways in SW1990 pancreatic cancer xenografts revealed that the human branch is practically dormant in untreated tumors but the insult on the stromal component produces massive responses of human cancer cells. Inhibition of the stromal PDGFr-ß with imatinib activates human PDGFr-ß/PDGF-B signaling loop, silent in untreated xenografts, via an apparent paracrine rescue pathway. Responses are treatment- and time-dependent. Soon after treatment, levels of human PDGFr-ß, compared to untreated tumors, are 3.4×, 12.4×, and 5.7× higher in imatinib-, radioimmunotherapy + imatinib-, and radioimmunotherapy-treated tumors, respectively. A continuous 14-day irradiation of imatinib-treated xenografts reduces levels of PDGFr-ß and phosphorylated PDGFr-ß by 5.3× and 4×, compared to earlier times. Human PDGF-B is upregulated suggesting that the survival signaling via the autocrine pathway is also triggered after stromal injury. These findings indicate that therapies targeting pancreatic cancer stromal components may have unintended mitogenic effects and that these effects can be reversed when imatinib is used in conjunction with radioimmunotherapy.

17.
J Med Chem ; 52(16): 5124-43, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19653647

RESUMEN

High levels of androgen receptor (AR) are often indicative of recurrent, advanced, or metastatic cancers. These conditions are also characterized by a high proliferative fraction. 5-Radioiodo-3'-O-(17beta-succinyl-5alpha-androstan-3-one)-2'-deoxyuridine 8 and 5-radioiodo-3'-O-(17beta-succinyl-5alpha-androstan-3-one)-2'-deoxyuridin-5'-yl monophosphate 13 target AR. They are also degraded intracellularly to 5-radioiodo-2'-deoxyuridine 1 and its monophosphate 20, respectively, which can participate in the DNA synthesis. Both drugs were prepared at the no-carrier-added level. Precursors and methods are readily adaptable to radiolabeling with various radiohalides suitable for SPECT and PET imaging, as well as endoradiotherapy. In vitro and in vivo studies confirm the AR-dependent interactions. Both drugs bind to sex hormone binding globulin. This binding significantly improves their stability in serum. Biodistribution and imaging studies show preferential uptake and retention of 8 and 13 in ip xenografts of human ovarian adenocarcinoma cells NIH:OVCAR-3, which overexpress AR. When these drugs are administered at therapeutic dose levels, a significant tumor growth arrest is observed.


Asunto(s)
Androstanoles/síntesis química , Nucleótidos de Desoxiuracil/síntesis química , Desoxiuridina/análogos & derivados , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Radiofármacos/síntesis química , Receptores Androgénicos/metabolismo , Androstanoles/química , Androstanoles/farmacocinética , Animales , Proteínas Sanguíneas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Nucleótidos de Desoxiuracil/química , Nucleótidos de Desoxiuracil/farmacocinética , Desoxiuridina/síntesis química , Desoxiuridina/química , Desoxiuridina/farmacocinética , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Radioisótopos de Yodo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Hormono-Dependientes/metabolismo , Unión Proteica , Conejos , Radiofármacos/química , Radiofármacos/farmacocinética , Suero , Globulina de Unión a Hormona Sexual/química , Trasplante Heterólogo
18.
Med Chem ; 5(2): 171-81, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19275716

RESUMEN

Diagnostic agents enabling characterization of multidrug resistance (MDR) in tumors can aid in the selection of chemotherapy regimens. We report here synthesis and evaluation of radiopharmaceuticals based on the second-generation MDR-reversing drug MS-209. 5-[3-{4-(2-Phenyl-2-(4'-[(125)I]iodo-phenyl)acetyl)piperazin-1-yl}-2-hydroxypropoxy]quino-line (17) was prepared from the 4'-tributylstannyl precursor (16) in >95% radiochemical yield. (16) was synthesized in a six-step process with the overall yield of 25%. In vitro studies were conducted in MES-SA (drug-sensitive) and MES-SA/Dx5 (MDR) human uterine sarcoma cell lines. In vivo studies were performed in athymic mice bearing MES-SA and MES-SA/Dx5 xenografts. The uptake of (17) is higher in MES-SA than MES-SA/Dx5 cells. The uptake and efflux of (17) depend on temperature and concentration, and indicate active transport mechanism(s). Incubation of drug sensitive MES-SA cells with verapamil or (15), a nonradioactive analog of (17), alters the cellular retention of radioactivity only marginally. However, MES-SA/Dx5 cells retain approximately 12% more of (17) when incubated with 10 muM verapamil. The addition of (15) or high concentrations of (17) also increase the uptake of (17) in MES-SA/Dx5 up to 200%, depending on the concentration and temperature. The dependence of (17) uptake on the MDR status is also evident in the ex vivo binding studies. In vivo tests in mice xenografted simultaneously with both tumor cell lines indicate distinct pharmacokinetics for each tumor. The absorption half-life in MES-SA/Dx5 xenograft is approximately 10x shorter and the mean residence time approximately 50% shorter compared to MES-SA xenograft in the same mouse. Radioiodinated derivatives of MS-209 appear to be good indicators of multidrug resistance.


Asunto(s)
Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Neoplasias/diagnóstico por imagen , Cintigrafía/métodos , Radiofármacos/análisis , Radiofármacos/farmacocinética , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Radioisótopos de Yodo/química , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias/metabolismo , Quinolinas/química , Quinolinas/farmacología , Radiofármacos/síntesis química , Radiofármacos/metabolismo , Temperatura , Factores de Tiempo , Distribución Tisular , Verapamilo/farmacología
20.
Int J Radiat Oncol Biol Phys ; 72(3): 918-26, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19014781

RESUMEN

PURPOSE: To determine the late effects of radiotherapy (RT) on vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR2), and osteopontin (OPN) expression in cancer and stromal cells. METHODS AND MATERIALS: LS174T xenografted athymic mice were used as a tumor model. Radiation was delivered in two equivalent fractionation schemes: 5 x 7 Gy and 1 x 20 Gy, the latter at two dose rates. RESULTS: Tumor growth arrest was similar in all treatment groups, with the exception of a better response of small-size tumors in the 5 x 7-Gy group. The host VEGF and OPN levels were directly proportional to the tumor doubling time and were independent of the fractionation scheme. The host and cancer cell VEGFR2 levels in tumor were also directly related to the tumor response to RT. CONCLUSION: Upregulated VEGFR2 in cancer cells suggest paracrine signaling in the VEGFR2 pathway of cancer cells as the factor contributing to RT failure. The transient activation of the host VEGF/VEGFR2 pathway in tumor supports the model of angiogenic regeneration and suggests that radiation-induced upregulation of VEGF, VEGFR2, and downstream proteins might contribute to RT failure by escalating the rate of vascular repair. Coexpression of host OPN and VEGF, two factors closely associated with angiogenesis, indicate that OPN can serve as a surrogate marker of tumor recovery after RT. Taken together, these results strongly support the notion that to achieve optimal therapeutic outcomes, the scheduling of RT and antiangiogenic therapies will require patient-specific post-treatment monitoring of the VEGF/VEGFR2 pathway and that tumor-associated OPN can serve as an indicator of tumor regrowth.


Asunto(s)
División Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Neoplasias/genética , Osteopontina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Adenocarcinoma , Animales , División Celular/efectos de la radiación , Neoplasias Colorrectales , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias/patología , Neoplasias/radioterapia , Osteopontina/efectos de la radiación , ARN Mensajero/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/radioterapia , Células del Estroma/citología , Células del Estroma/fisiología , Células del Estroma/efectos de la radiación , Factor A de Crecimiento Endotelial Vascular/efectos de la radiación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA