Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Ultrasound Med ; 34(7): 1227-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26112625

RESUMEN

OBJECTIVES: To enhance the regional antitumor activity of the vascular-targeting agent truncated tissue factor (tTF)-NGR by combining the therapy with low-energy ultrasound (US) treatment. METHODS: For the in vitro US exposure of human umbilical vein endothelial cells (HUVECs), cells were put in the focus of a US transducer. For analysis of the US-induced phosphatidylserine (PS) surface concentration on HUVECs, flow cytometry was used. To demonstrate the differences in the procoagulatory efficacy of TF-derivative tTF-NGR on binding to HUVECs with a low versus high surface concentration of PS, we performed factor X activation assays. For low-energy US pretreatment, HT1080 fibrosarcoma xenotransplant-bearing nude mice were treated by tumor-regional US-mediated stimulation (ie, destruction) of microbubbles. The therapy cohorts received the tumor vessel-infarcting tTF-NGR protein with or without US pretreatment (5 minutes after US stimulation via intraperitoneal injection on 3 consecutive days). RESULTS: Combination therapy experiments with xenotransplant-bearing nude mice significantly increased the antitumor activity of tTF-NGR by regional low-energy US destruction of vascular microbubbles in tumor vessels shortly before application of tTF-NGR (P < .05). Mechanistic studies proved the upregulation of anionic PS on the outer leaflet of the lipid bilayer of endothelial cell membranes by low-energy US and a consecutive higher potential of these preapoptotic endothelial cells to activate coagulation via tTF-NGR and coagulation factor X as being a basis for this synergistic activity. CONCLUSIONS: Combining retargeted tTF to tumor vessels with proapoptotic stimuli for the tumor vascular endothelium increases the antitumor effects of tumor vascular infarction. Ultrasound treatment may thus be useful in this respect for regional tumor therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Fibrosarcoma/terapia , Infarto/etiología , Tromboplastina/farmacología , Terapia por Ultrasonido , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Endotelio Vascular , Femenino , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos , Microburbujas , Neovascularización Patológica/prevención & control
2.
Cancer Gene Ther ; 30(10): 1355-1368, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37391502

RESUMEN

To enhance the potency of chimeric antigen receptor (CAR) engineered T cells in solid cancers, we designed a novel cell-based combination strategy with an additional therapeutic mode of action. CAR T cells are used as micropharmacies to produce a targeted pro-coagulatory fusion protein, truncated tissue factor (tTF)-NGR, which exerts pro-coagulatory activity and hypoxia upon relocalization to the vascular endothelial cells that invade tumor tissues. Delivery by CAR T cells aimed to induce locoregional tumor vascular infarction for combined immune-mediated and hypoxic tumor cell death. Human T cells that were one-vector gene-modified to express a GD2-specific CAR along with CAR-inducible tTF-NGR exerted potent GD2-specific effector functions while secreting tTF-NGR that activates the extrinsic coagulation pathway in a strictly GD2-dependent manner. In murine models, the CAR T cells infiltrated GD2-positive tumor xenografts, secreted tTF-NGR into the tumor microenvironment and showed a trend towards superior therapeutic activity compared with control cells producing functionally inactive tTF-NGR. In vitro evidence supports a mechanism of hypoxia-mediated enhancement of T cell cytolytic activity. We conclude that combined CAR T cell targeting with an additional mechanism of antitumor action in a one-vector engineering strategy is a promising approach to be further developed for targeted treatment of solid cancers.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Linfocitos T , Células Endoteliales , Línea Celular Tumoral , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Muerte Celular , Hipoxia/metabolismo , Inmunoterapia Adoptiva , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias/terapia , Neoplasias/metabolismo
3.
Cancers (Basel) ; 13(11)2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-34200318

RESUMEN

Besides its central functional role in coagulation, TF has been described as being operational in the development of malignancies and is currently being studied as a possible therapeutic tool against cancer. One of the avenues being explored is retargeting TF or its truncated extracellular part (tTF) to the tumor vasculature to induce tumor vessel occlusion and tumor infarction. To this end, multiple structures on tumor vascular wall cells have been studied at which tTF has been aimed via antibodies, derivatives, or as bifunctional fusion protein through targeting peptides. Among these targets were vascular adhesion molecules, oncofetal variants of fibronectin, prostate-specific membrane antigens, vascular endothelial growth factor receptors and co-receptors, integrins, fibroblast activation proteins, NG2 proteoglycan, microthrombus-associated fibrin-fibronectin, and aminopeptidase N. Targeting was also attempted toward cellular membranes within an acidic milieu or toward necrotic tumor areas. tTF-NGR, targeting tTF primarily at aminopeptidase N on angiogenic endothelial cells, was the first drug candidate from this emerging class of coaguligands translated to clinical studies in cancer patients. Upon completion of a phase I study, tTF-NGR entered randomized studies in oncology to test the therapeutic impact of this novel therapeutic modality.

4.
Cancers (Basel) ; 12(12)2020 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-33256235

RESUMEN

BACKGROUND: CD-13 targeted tissue factor tTF-NGR is a fusion protein selectively inducing occlusion of tumor vasculature with resulting tumor infarction. Mechanistic and pharmacodynamic studies have shown broad anti-tumor therapeutic effects in xenograft models. METHODS: After successful Good Manufacturing Practice (GMP) production and before translation into clinical phase I, ICH S9 (S6) guideline-conforming animal safety, toxicology, and pharmacokinetic (PK) studies were requested by the federal drug authority in accordance with European and US regulations. RESULTS: These studies were performed in mice, rats, guinea pigs, and beagle dogs. Results of the recently completed clinical phase I trial in end-stage cancer patients showed only limited predictive value of these non-clinical studies for patient tolerability and safety in phase I. CONCLUSIONS: Although this experience cannot be generalized, alternative pathways with seamless clinical phase 0 microdosing-phase I dose escalation studies are endorsed for anticancer drug development and translation into the clinic.

5.
PLoS One ; 15(2): e0229271, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32084238

RESUMEN

BACKGROUND: Truncated tissue factor (tTF) retargeted by NGR-peptides to aminopeptidase N (CD13) in tumor vasculature is effective in experimental tumor therapy. tTF-NGR induces tumor growth inhibition in a variety of human tumor xenografts of different histology. To improve on the therapeutic efficacy we have combined tTF-NGR with radiotherapy. METHODS: Serum-stimulated human umbilical vein endothelial cells (HUVEC) and human HT1080 sarcoma cells were irradiated in vitro, and upregulated early-apoptotic phosphatidylserine (PS) on the cell surface was measured by standard flow cytometry. Increase of cellular procoagulant function in relation to irradiation and PS cell surface concentration was measured in a tTF-NGR-dependent Factor X activation assay. In vivo experiments with CD-1 athymic mice bearing human HT1080 sarcoma xenotransplants were performed to test the systemic therapeutic effects of tTF-NGR on tumor growth alone or in combination with regional tumor ionizing radiotherapy. RESULTS: As shown by flow cytometry with HUVEC and HT1080 sarcoma cells in vitro, irradiation with 4 and 6 Gy in the process of apoptosis induced upregulation of PS presence on the outer surface of both cell types. Proapoptotic HUVEC and HT1080 cells both showed significantly higher procoagulant efficacy on the basis of equimolar concentrations of tTF-NGR as measured by FX activation. This effect can be reverted by masking of PS with Annexin V. HT1080 human sarcoma xenografted tumors showed shrinkage induced by combined regional radiotherapy and systemic tTF-NGR as compared to growth inhibition achieved by either of the treatment modalities alone. CONCLUSIONS: Irradiation renders tumor and tumor vascular cells procoagulant by PS upregulation on their outer surface and radiotherapy can significantly improve the therapeutic antitumor efficacy of tTF-NGR in the xenograft model used. This synergistic effect will influence design of future clinical combination studies.


Asunto(s)
Antineoplásicos/farmacología , Antígenos CD13/metabolismo , Terapia Molecular Dirigida , Sarcoma/tratamiento farmacológico , Sarcoma/radioterapia , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/efectos de la radiación , Línea Celular Tumoral , Terapia Combinada , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Fosfatidilserinas/metabolismo , Sarcoma/metabolismo , Sarcoma/patología
6.
Leuk Res ; 32(6): 954-61, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18006056

RESUMEN

Vascular endothelial growth factor-C (VEGF-C) has been shown to promote survival and resistance to chemotherapy of AML-cells in vitro. We investigated the expression of VEGF-C/VEGFR-3 in the bone marrow and pretherapeutic plasma levels of VEGF-C in patients with newly diagnosed AML. Expression of VEGF-C/VEGFR-3 was significantly higher in AML patients than in controls, while circulating levels did not differ. However, VEGF-C/VEGFR-3 expression was not able to predict clinical outcome. In conclusion, AML is associated with an increased expression of VEGF-C/VEGFR-3. Although expression levels display no prognostic significance in our study, strategies targeting the VEGF-C/VEGFR-3-pathway might be a promising treatment approach.


Asunto(s)
Médula Ósea/metabolismo , Leucemia Mieloide Aguda/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Médula Ósea/patología , Estudios de Casos y Controles , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas para Inmunoenzimas , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Pronóstico , Inducción de Remisión , Tasa de Supervivencia
7.
Curr Drug Discov Technol ; 5(1): 1-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18537561

RESUMEN

Selective activation of blood coagulation in tumor vessels with subsequent thrombosis and tumor infarction is a promising strategy in cancer therapy. To this end, different fusion proteins consisting of the extracellular domain of tissue factor (truncated tissue factor, tTF) were fused to the peptides GRGDSP (abbr. RGD), GNGRAHA (abbr. NGR) or cyclic derivates of these peptides, which selectively target alpha(v)-integrins or aminopeptidase N (CD13), respectively. Rationale for this strategy is the fact that these surface receptors are preferentially expressed on tumor endothelial cells. The tTF constructs were expressed in Escherichia coli BL21 (DE3). The integrity of the fusion proteins was evaluated by SDS-PAGE, immunoblotting and mass spectrometry. The screening process for the activity contained coagulation assays as well as purified receptor binding assays. The fusion proteins which retained their thrombogenic and binding activity were evaluated further. In vivo studies in nude mice bearing established different malignant human tumors revealed that i.v. administration of tTF-RGD or tTF-NGR induced partial or complete thrombotic occlusion of tumor vessels, which was demonstrated by histological analysis. Furthermore, treatment studies showed that the targeted tTF fusion proteins but not untargeted tTF proteins induced significant tumor growth retardation in human adenocarcinoma of the breast in a nude mice model without apparent side effects such as thrombosis in liver, kidney, heart or lung at therapeutic dose levels. Finally, we illustrate the upscaling process of fusion protein fabrication in order to produce the amounts needed for clinical studies. Thus, generation and screening of active fusion proteins, which induce selective thrombosis in the tumor vasculature, may be a promising strategy for the development of new drugs as cancer therapeutics.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/farmacología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/tratamiento farmacológico , Inhibidores de la Angiogénesis/aislamiento & purificación , Animales , Secuencia de Bases , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/tratamiento farmacológico , Cromatografía Líquida de Alta Presión , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Células Endoteliales/efectos de los fármacos , Factor X/metabolismo , Femenino , Humanos , Integrina alfaVbeta3/genética , Integrinas/genética , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Ratones , Trasplante de Neoplasias , Receptores de Vitronectina/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Flujo Sanguíneo Regional , Estreptavidina/farmacología
8.
Lung Cancer ; 55(1): 53-60, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17067717

RESUMEN

To evaluate characteristics and prognostic impact of different structure types of intratumoural blood vessels, tissue samples of 72 patients with primary stages I and II non-small cell lung cancer (NSCLC) were analysed. Performing immunohistochemistry, 45 of 56 analysed tumours (80%) demonstrated an obvious alveolar vascular pattern with tight coverage with perivascular cells in at least parts of the sample. After an overall median follow-up of 139 months for surviving patients, tumours with an alveolar vascular pattern showed a significantly better overall survival (OS) compared to those with an entirely angiogenic vascular pattern (108 months versus 63 months; p<0.05). Furthermore, high expression of angiopoietin-1 (Ang-1) correlated with OS (p<0.05). In contrast, expression of Ang-2 or vascular endothelial growth factor was not significantly associated with survival. Collectively, alveolar vessel architecture and angiopoietin expression appear to be common phenomenons in early stage NSCLC and may serve as prognostic factors.


Asunto(s)
Vasos Sanguíneos/patología , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Neoplasias Pulmonares/irrigación sanguínea , Circulación Pulmonar , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Estadificación de Neoplasias , Pronóstico
9.
Haematologica ; 91(9): 1203-11, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16956819

RESUMEN

BACKGROUND AND OBJECTIVES: Angiopoietin-1 (Ang-1) and its natural antagonist angiopoietin-2 (Ang-2), both ligands for the receptor tyrosine kinase Tie2, are known to play an essential role in normal and pathological angiogenesis. DESIGN AND METHODS: We investigated the expression of Ang-1, Ang-2 and Tie2 by immunohistochemical analyses in bone marrow biopsies of 64 adult patients with newly diagnosed acute myeloid leukemia (AML) and correlated angiogenic factor expression with clinicopathological variables and long-term survival. RESULTS: Expression of Ang-2 was significantly higher in the bone marrow of AML patients than in 16 control patients. In contrast, the levels of Ang-1 expression in AML patients did not differ from those found in controls. Thus, we observed a reversal of the Ang-1 and Ang-2 expression balance in the neoplastic bone marrow. Furthermore, Tie2 was significantly overexpressed in leukemic blasts. Patients expressing high levels of Ang-2 had significantly longer overall survival than those with low Ang-2 levels (52.7 vs. 14.7 months). Multivariate analysis revealed that karyotype and Ang-2 expression were independent prognostic factors for overall survival (hazard ratio [CI]: 3.06 [1.39-6.70] and 0.31 [0.14-0.69], respectively). INTERPRETATION AND CONCLUSIONS: These data provide evidence that the alteration of angiopoietin balance in favor of Ang-2 may play a critical role in the pathophysiology of AML. Furthermore, high pre-therapeutic levels of Ang-2 in the bone marrow indicate a favorable prognosis in AML patients treated with polychemotherapy, although the mechanism is not yet known.


Asunto(s)
Angiopoyetinas/genética , Médula Ósea/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide/genética , Receptor TIE-2/genética , Enfermedad Aguda , Angiopoyetina 1/genética , Angiopoyetina 2/genética , Humanos
10.
Oncotarget ; 7(6): 6774-89, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26735180

RESUMEN

tTF-TAA and tTF-LTL are fusion proteins consisting of the extracellular domain of tissue factor (TF) and the peptides TAASGVRSMH and LTLRWVGLMS, respectively. These peptides represent ligands of NG2, a surface proteoglycan expressed on angiogenic pericytes and some tumor cells. Here we have expressed the model compound tTF-NGR, tTF-TAA, and tTF-LTL with different lengths in the TF domain in E. coli and used these fusion proteins for functional studies in anticancer therapy. We aimed to retarget TF to tumor vessels leading to tumor vessel infarction with two barriers of selectivity, a) the leaky endothelial lining in tumor vessels with the target NG2 being expressed on pericytes on the abluminal side of the endothelial cell barrier and b) the preferential expression of NG2 on angiogenic vessels such as in tumors. Chromatography-purified tTF-TAA showed identical Factor X (FX)-activating procoagulatory activity as the model compound tTF-NGR with Km values of approx. 0.15 nM in Michaelis-Menten kinetics. The procoagulatory activity of tTF-LTL varied with the chosen length of the TF part of the fusion protein. Flow cytometry revealed specific binding of tTF-TAA to NG2-expressing pericytes and tumor cells with low affinity and dissociation KD in the high nM range. In vivo and ex vivo fluorescence imaging of tumor xenograft-carrying animals and of the explanted tumors showed reduction of tumor blood flow upon tTF-TAA application. Therapeutic experiments showed a reproducible antitumor activity of tTF-TAA against NG2-expressing A549-tumor xenografts, however, with a rather small therapeutic window (active/toxic dose in mg/kg body weight).


Asunto(s)
Antígenos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Oligopéptidos/farmacología , Proteoglicanos/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Tromboplastina/farmacología , Animales , Antígenos/biosíntesis , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Proteoglicanos/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Oncotarget ; 7(50): 82458-82472, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27738341

RESUMEN

Truncated tissue factor (tTF), retargeted to tumor vasculature by GNGRAHA peptide (tTF-NGR), and doxorubicin have therapeutic activity against a variety of tumors. We report on combination experiments of both drugs using different schedules. We have tested fluorescence- and HPLC-based intratumoral pharmacokinetics of doxorubicin, flow cytometry for cellular phosphatidylserine (PS) expression, and tumor xenograft studies for showing in vivo apoptosis, proliferation decrease, and tumor shrinkage upon combination therapy with doxorubicin and induced tumor vascular infarction. tTF-NGR given before doxorubicin inhibits the uptake of the drug into human fibrosarcoma xenografts in vivo. Reverse sequence does not influence the uptake of doxorubicin into tumor, but significantly inhibits the late wash-out phase, thus entrapping doxorubicin in tumor tissue by vascular occlusion. Incubation of endothelial and tumor cells with doxorubicin in vitro increases PS concentrations in the outer layer of the cell membrane as a sign of early apoptosis. Cells expressing increased PS concentrations show comparatively higher procoagulatory efficacy on the basis of equimolar tTF-NGR present in the Factor X assay. Experiments using human M21 melanoma and HT1080 fibrosarcoma xenografts in athymic nude mice indeed show a combinatorial tumor growth inhibition applying doxorubicin and tTF-NGR in sequence over single drug treatment. Combination of cytotoxic drugs such as doxorubicin with tTF-NGR-induced tumor vessel infarction can improve pharmacodynamics of the drugs by new mechanisms, entrapping a cytotoxic molecule inside tumor tissue and reciprocally improving procoagulatory activity of tTF-NGR in the tumor vasculature via apoptosis induction in tumor endothelial and tumor cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Fibrosarcoma/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Neovascularización Patológica , Neoplasias Cutáneas/tratamiento farmacológico , Tromboplastina/farmacología , Animales , Antibióticos Antineoplásicos/farmacocinética , Coagulación Sanguínea/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacocinética , Femenino , Fibrosarcoma/sangre , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfatidilserinas/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tromboplastina/análogos & derivados , Tromboplastina/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Thromb Res ; 125 Suppl 2: S143-50, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20433995

RESUMEN

A variety of fusion proteins consisting of the extracellular domain of tissue factor (truncated tissue factor, tTF) fused to the peptides GRGDSP (abbr. RGD), GNGRAHA (abbr. NGR) or derivates of these peptides, have been synthesized. These binding motif peptides target av-integrins or aminopeptidase N (CD13), respectively, on tumor endothelial cells. After expression and deposition as inclusion bodies in Escherichia coli BL21 (DE3), the tTF-fusion proteins were refolded and purified in a multi-step chromatography process. The upscaling process of fusion protein synthesis in order to produce amounts needed for clinical studies is presented. The proteins retained their specific proteolytic ability to activate FX by FVIIa and were able to bind to endothelial cells in vitro. Western blot analysis, analytic chromatography, FX coagulation assay and in vivo experiments have been performed to test for the in vitro stability of the tTF-NGR protein after long-term incubation at 5 degrees C or 25 degrees C, respectively. In vivo xenograft studies in nude mice bearing different malignant human tumors (mammary carcinoma SKBR3, adenocarcinoma of the lung A549) revealed that intravenous or subcutaneous administration of tTF-NGR or -RGD fusion proteins, but not the tTF protein without binding motif, induced thrombosis of tumor vessels which led to significant tumor growth retardation or regression. The anti-vascular mechanism of the tTF fusion proteins was verified by the molecular imaging methods such as magnetic resonance imaging (MRI) and fluorescence reflectance imaging (FRI); MRI showed a reduction of the relative tumor blood volume (BV) and FRI the formation of fibrin in the tTF-fusion protein treated tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Tromboplastina/uso terapéutico , Animales , Antineoplásicos/farmacología , Vasos Sanguíneos/efectos de los fármacos , Escherichia coli/genética , Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias/patología , Oligopéptidos/genética , Oligopéptidos/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Tromboplastina/genética , Tromboplastina/farmacología
13.
Int J Oncol ; 37(6): 1389-97, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21042706

RESUMEN

tTF-NGR consists of the extracellular domain of the (truncated) tissue factor (tTF), a central molecule for coagulation in vivo, and the peptide GNGRAHA (NGR), a ligand of the surface protein aminopeptidase N (CD13). After deamidation of the NGR-peptide moiety, the fusion protein is also a ligand for integrin αvß3 (CD51/CD61). Both surface proteins are upregulated on endothelial cells of tumor vessels. tTF-NGR showed binding to specific binding sites on endothelial cells in vitro as shown by flow cytometry. Subcutaneous injection of tTF-NGR into athymic mice bearing human HT1080 fibrosarcoma tumors induced tumor growth retardation and delay. Contrast enhanced ultrasound detected a decrease in tumor blood flow in vivo after application of tTF-NGR. Histological analysis of the tumors revealed vascular disruption due to blood pooling and thrombotic occlusion of tumor vessels. Furthermore, a lack of resistance was shown by re-exposure of tumor-bearing mice to tTF-NGR after regrowth following a first cycle of treatment. However, after subcutaneous (s.c.) push injection with therapeutic doses (1-5 mg/kg bw) side effects have been observed, such as skin bleeding and reduced performance. Since lethality started within the therapeutic dose range (LD10 approximately 2 mg/kg bw) no safe therapeutic window could be found. Limiting toxicity was represented by thrombo-embolic events in major organ systems as demonstrated by histology. Thus, subcutaneous injection of tTF-NGR represents an active, but toxic application procedure and compares unfavourably to intravenous infusion.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Infarto/inducido químicamente , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Tromboplastina/administración & dosificación , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Línea Celular Tumoral , Células Cultivadas , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Oligopéptidos/efectos adversos , Oligopéptidos/química , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/química , Tromboplastina/efectos adversos , Tromboplastina/química , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Cell Sci ; 115(Pt 9): 1961-71, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11956327

RESUMEN

Emerging data suggest that urokinase-type plasminogen activator (UPA), beyond its role in pericellular proteolysis, may also act as a mitogen. We investigated the function of endogenous UPA in mediating the mitogenic effects of platelet-derived growth factor (PDGF) and basic fibroblast growth factor (bFGF) on human vascular smooth muscle cells (SMC). Growth-arrested SMC constitutively expressed UPA, but UPA expression and secretion increased several times upon stimulation with either PDGF or bFGF. Inhibition of endogenous UPA with a polyclonal antibody significantly reduced DNA synthesis and proliferation of PDGF or bFGF stimulated SMC, this effect already being evident when the cells entered S-phase. The proliferative activity of endogenous UPA was dependent on a functional catalytic domain as demonstrated by inhibition experiments with a specific monoclonal antibody (394OA) and p-aminobenzamidine, respectively. In contrast, neither plasmin generation nor binding of UPA to its receptor (CD87) were required for UPA-mediated mitogenic effects. The results demonstrate that endogenous UPA is not only overexpressed in SMC upon stimulation with PDGF/bFGF, but also mediates the mitogenic activity of the growth factors in a catalytic-domain-dependent manner. Specific inhibition of this UPA domain may represent an attractive target for pharmacological interventions in atherogenesis and restenosis after angioplasty.


Asunto(s)
Dominio Catalítico/fisiología , División Celular/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Arteriosclerosis/metabolismo , Arteriosclerosis/fisiopatología , Quinasa de la Caseína II , División Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/farmacología , Fibrinolisina/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/farmacología , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/metabolismo , Oclusión de Injerto Vascular/fisiopatología , Humanos , Hipertrofia/metabolismo , Hipertrofia/fisiopatología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/crecimiento & desarrollo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA