Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Vaccine ; 41(37): 5383-5391, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37468389

ABSTRACT

The viral safety of biological products is ensured by tests throughout the production chain, and, for certain products, by steps in the manufacturing process enabling the elimination or inactivation of viruses. Current testing programs include sample inoculation in animals and embryonic eggs. Following the 3Rs principles of replacement, reduction, and refinement of animal-use methods, such techniques are intended to be replaced not only for ethical reasons but also because of their inherent technical limitations, their long turnaround times, and their limits in virus detection. Therefore, we have compared the limit and range of sensitivity of in vivo tests used for viral testing of cells with a transcriptomic assay based on Next Generation Sequencing (NGS). Cell cultures were infected with a panel of nine (9) viruses, among them only five (5) were detected, with variable sensitivity, by in vivo tests. The transcriptomic assay was able to detect one (1) infected cell among 103 to 107 non-infected cells for all viruses assessed, including those not detected by the conventional in vivo tests. Here we show that NGS extends the breath of detection of viral contaminants compared to traditional testing. Collectively, these results support the replacement of the conventional in vivo tests by an NGS-based transcriptomic assay for virus safety testing of cell substrates.


Subject(s)
Biological Products , Viruses , Animals , Transcriptome , High-Throughput Nucleotide Sequencing , Viruses/genetics , Cell Culture Techniques
3.
Article in English | MEDLINE | ID: mdl-34569920

ABSTRACT

A strict anaerobic, Gram-stain-positive rod-shaped bacterium, designated PTT, was isolated from the faecal material of a painted turtle (Chrysemys picta). Based on a comparative 16S rRNA gene sequence analysis, the isolate was assigned to Clostridium sensu stricto with the highest sequence similarities to Clostridium moniliforme (97.4 %), Clostridium sardiniense (97.2 %) and the misclassified organism Eubacterium multiforme (97.1 %). The predominant cellular fatty acids of strain PTT were C14 : 0, C16 : 0 and an unidentified product with an equivalent chain length of 14.969. The G+C content determined from the genome was 28.8 mol%. The fermentation end products from glucose were acetate and butyrate with no alcohols detected and trace amounts of CO2 and H2 also detected; no respiratory quinones were detected. Based on biochemical, phylogenetic, genotypic and chemotaxonomic criteria, the isolate represents a novel species of the genus Clostridium for which the name Clostridium chrysemydis sp. nov. is proposed. The type strain is strain PTT (=CCUG 74180T=ATCC TSD-219T).


Subject(s)
Turtles , Animals , Bacterial Typing Techniques , Base Composition , Clostridium/genetics , DNA, Bacterial/genetics , Eubacterium , Fatty Acids/chemistry , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
4.
Clin Imaging ; 79: 273-277, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34171595

ABSTRACT

PURPOSE: Emergency laparotomy has a high reported thirty-day mortality, ranging from 11 to 15%. Current peri-operative risk assessment tools may poorly estimate the risk of perioperative mortality. We sought to determine if CT-determined sarcopenia may be a useful predictor of post-operative outcomes in patients undergoing an emergency laparotomy. METHODS: A retrospective review of a prospectively maintained database of consecutive adult patients who underwent an emergency laparotomy at our institution was performed. Post-operative mortality (90-day mortality), admission to HDU or ICU and APACHE-II scores were recorded for these patients. Sarcopenia was calculated by determining psoas area and density at the level of the third lumbar vertebra. The lowest quartile was determined to be sarcopenic. Univariate statistical analysis investigated associations between sarcopenia and these outcome measures. RESULTS: Eighty patients were included in the study following application of exclusion criteria. Thirty-eight were male. The 90-day mortality rate was 11%. Compared to their non-sarcopenic counterparts, sarcopenic patients were significantly more likely to have died by 90 days post-operatively (χ2 = 9.51, p = 0.002) and to require admission to either the HDU or ICU in the post-operative period (χ2 = 10.62, p = 0.001). CONCLUSIONS: CT determined sarcopenia is significantly associated with 90-day mortality and post-operative admission to HDU or ICU in patients undergoing an emergency laparotomy. The future development of a validated scoring tool incorporating sarcopenia could help to better select out those patients who will require higher levels of post-operative care as well as identifying those for whom surgery may not confer a survival benefit and be deemed futile.


Subject(s)
Sarcopenia , Adult , Emergencies , Humans , Laparotomy , Male , Retrospective Studies , Sarcopenia/diagnostic imaging , Tomography, X-Ray Computed
5.
Leuk Res ; 36(11): 1422-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22906694

ABSTRACT

Preclinical and clinical studies have shown that proteasome inhibitors (PIs) have anti-MM activity in combination with dexamethasone or lenalidomide. However, no data exists on the anti-MM effects of combinations involving the PI delanzomib with dexamethasone and/or lenalidomide. Herein, we show that delanzomib in combination with dexamethasone and/or lenalidomide results in superior tumor reduction and extended tumor growth delays when compared to vehicle alone, these drugs alone, or the doublet of dexamethasone and lenalidomide. The favorable results obtained from the three xenograft studies suggest that delanzomib in combination with dexamethasone and lenalidomide should be explored for the treatment of MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Multiple Myeloma/drug therapy , Threonine/analogs & derivatives , Animals , Cell Line, Tumor , Dexamethasone/administration & dosage , Humans , Lenalidomide , Mice , Mice, SCID , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Threonine/administration & dosage , Xenograft Model Antitumor Assays
6.
Mol Cancer Ther ; 11(4): 930-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22319199

ABSTRACT

Mutations in the BRAF gene have been identified in approximately 7% of cancers, including 60% to 70% of melanomas, 29% to 83% of papillary thyroid carcinomas, 4% to 16% colorectal cancers, and a lesser extent in serous ovarian and non-small cell lung cancers. The V600E mutation is found in the vast majority of cases and is an activating mutation, conferring transforming and immortalization potential to cells. CEP-32496 is a potent BRAF inhibitor in an in vitro binding assay for mutated BRAF(V600E) (K(d) BRAF(V600E) = 14 nmol/L) and in a mitogen-activated protein (MAP)/extracellular signal-regulated (ER) kinase (MEK) phosphorylation (pMEK) inhibition assay in human melanoma (A375) and colorectal cancer (Colo-205) cell lines (IC(50) = 78 and 60 nmol/L). In vitro, CEP-32496 has multikinase binding activity at other cancer targets of interest; however, it exhibits selective cellular cytotoxicity for BRAF(V600E) versus wild-type cells. CEP-32496 is orally bioavailable in multiple preclinical species (>95% in rats, dogs, and monkeys) and has single oral dose pharmacodynamic inhibition (10-55 mg/kg) of both pMEK and pERK in BRAF(V600E) colon carcinoma xenografts in nude mice. Sustained tumor stasis and regressions are observed with oral administration (30-100 mg/kg twice daily) against BRAF(V600E) melanoma and colon carcinoma xenografts, with no adverse effects. Little or no epithelial hyperplasia was observed in rodents and primates with prolonged oral administration and sustained exposure. CEP-32496 benchmarks favorably with respect to other kinase inhibitors, including RAF-265 (phase I), sorafenib, (approved), and vemurafenib (PLX4032/RG7204, approved). CEP-32496 represents a novel and pharmacologically active BRAF inhibitor with a favorable side effect profile currently in clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Quinazolines/pharmacology , Administration, Oral , Animals , Cell Line, Tumor , Cell Proliferation , Dogs , Drug Screening Assays, Antitumor , Humans , Macaca fascicularis , Male , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Quinazolines/pharmacokinetics , Rats , Rats, Sprague-Dawley
7.
J Med Chem ; 55(2): 903-13, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22148921

ABSTRACT

A substantial body of evidence supports the utility of antiangiogenesis inhibitors as a strategy to block or attenuate tumor-induced angiogenesis and inhibition of primary and metastatic tumor growth in a variety of solid and hematopoietic tumors. Given the requirement of tumors for different cytokine and growth factors at distinct stages of their growth and dissemination, optimal antiangiogenic therapy necessitates inhibition of multiple, complementary, and nonredundant angiogenic targets. 11-(2-Methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5,4-a]pyrrolo[3,4-c]carbazol-4-one (11b, CEP-11981) is a potent orally active inhibitor of multiple targets (TIE-2, VEGF-R1, 2, and 3, and FGF-R1) having essential and nonredundant roles in tumor angiogenesis and vascular maintenance. Outlined in this article are the design strategy, synthesis, and biochemical and pharmacological profile for 11b, which completed Phase I clinical assessing safety and pharmacokinetics allowing for the initiation of proof of concept studies.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Carbazoles/chemical synthesis , Indazoles/chemical synthesis , Receptor, TIE-2/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Biological Availability , Carbazoles/pharmacokinetics , Carbazoles/pharmacology , Humans , Indazoles/pharmacokinetics , Indazoles/pharmacology , Macaca fascicularis , Male , Mice , Mice, Nude , Models, Molecular , Neovascularization, Physiologic/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, TIE-2/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Xenograft Model Antitumor Assays
8.
Bioorg Med Chem Lett ; 18(7): 2368-72, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18343109

ABSTRACT

Orally bioavailable, dual inhibitors of TIE-2/VEGF-R2 were identified by elaborating the C3/N13 SAR around a fused pyrrolodihydroindazolocarbazole scaffold. Analogs bearing a C3-thiophencarbonyl group were evaluated in enzymatic and cellular biochemical assays; two orally bioavailable analogs were further profiled in functional assays and found to inhibit microvessel growth in rat aortic explant cultures and inhibit Ang-1-stimulated chemotaxis of HUVECs.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Aorta/drug effects , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacokinetics , Indazoles/pharmacology , Indoles/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Umbilical Veins/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Acylation , Angiogenesis Inhibitors/chemical synthesis , Angiotensin I , Animals , Aorta/cytology , Aorta/metabolism , Biological Availability , Cells, Cultured , Chemotaxis/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Inhibitors/chemical synthesis , Indazoles/chemical synthesis , Indoles/chemical synthesis , Models, Chemical , Rats , Structure-Activity Relationship , Thiophenes/chemistry , Umbilical Veins/cytology , Umbilical Veins/metabolism
9.
Blood ; 111(5): 2765-75, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18057228

ABSTRACT

Modulating protein ubiquitination via proteasome inhibition represents a promising target for cancer therapy, because of the higher sensitivity of cancer cells to the cytotoxic effects of proteasome inhibition. Here we show that CEP-18770 is a novel orally-active inhibitor of the chymotrypsin-like activity of the proteasome that down-modulates the nuclear factor-kappaB (NF-kappaB) activity and the expression of several NF-kappaB downstream effectors. CEP-18770 induces apoptotic cell death in multiple myeloma (MM) cell lines and in primary purified CD138-positive explant cultures from untreated and bortezomib-treated MM patients. In vitro, CEP-18770 has a strong antiangiogenic activity and potently represses RANKL-induced osteoclastogenesis. Importantly, CEP-18770 exhibits a favorable cytotoxicity profile toward normal human epithelial cells, bone marrow progenitors, and bone marrow-derived stromal cells. Intravenous and oral administration of CEP-18770 resulted in a more sustained pharmacodynamic inhibition of proteasome activity in tumors relative to normal tissues, complete tumor regression of MM xenografts and improved overall median survival in a systemic model of human MM. Collectively, these findings provide evidence for the utility of CEP-18770 as a novel orally active proteasome inhibitor with a favorable tumor selectivity profile for the treatment of MM and other malignancies responsive to proteasome inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Proteasome Inhibitors , Pyrazines/pharmacology , Threonine/analogs & derivatives , Administration, Oral , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Boronic Acids/administration & dosage , Boronic Acids/chemistry , Boronic Acids/therapeutic use , Bortezomib , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Nude , Multiple Myeloma/pathology , NF-kappa B/antagonists & inhibitors , Neoplasms/pathology , Osteogenesis/drug effects , Pyrazines/administration & dosage , Pyrazines/therapeutic use , RANK Ligand/pharmacology , Threonine/administration & dosage , Threonine/chemistry , Threonine/pharmacology , Threonine/therapeutic use , Treatment Outcome , Ubiquitin/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
Soc Sci Med ; 64(2): 272-82, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050056

ABSTRACT

In recent years, there has been a rise in the creation of DNA databases promising a range of health benefits to individuals and populations. This development has been accompanied by an interest in, and concern for the ethical, legal and social aspects of such collections. In terms of policy solutions, much of the focus of these debates has been on issues of consent, confidentiality and research governance. However, there are broader concerns, such as those associated with commercialisation, which cannot be adequately addressed by these foci. In this article, we focus on the health-wealth benefits that DNA databases promise by considering the views of 10 focus groups on Generation Scotland, Scotland's first national genetic database. As in previous studies, our qualitative research on public/s and stakeholders' views of DNA databases show the prospect of utilising donated samples and information derived for wealth-related ends (i.e. for private profit), irrespective of whether there is an associated health-related benefit, arouses considerable reaction. While health-wealth benefits are not mutually exclusive ideals, the tendency has been to cast 'public' benefits as exclusively health-related, while 'private' commercial benefits for funders and/or researchers are held out as a necessary pay-off. We argue for a less polarised approach that reconsiders what is meant by 'public benefits' and questions the exclusivity of commercial interests. We believe accommodation can be achieved via the mobilisation of a grass roots solution known as 'benefit-sharing' or a 'profit pay-off'. We propose a sociologically informed model that has a pragmatic, legal framework, which responds seriously to public concerns.


Subject(s)
Commerce , Databases, Genetic , Genetic Research , Drug Industry , Focus Groups , Humans , Referral and Consultation , Scotland
12.
Mol Cancer Ther ; 5(7): 1744-53, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891460

ABSTRACT

CEP-7055, a fully synthetic, orally active N,N-dimethylglycine ester of CEP-5214, a C3-(isopropylmethoxy)-fused pyrrolocarbazole with potent pan-vascular endothelial growth factor receptor (VEGFR) kinase inhibitory activity, has recently completed phase I clinical trials in cancer patients. These studies evaluated the antitumor efficacy of CEP-7055 using orthotopic models of glioblastoma and colon carcinoma in combination with temozolomide, and irinotecan and oxaliplatin, respectively, for their effects on primary and metastatic tumor burden and median survival. Chronic administration of CEP-7055 (23.8 mg/kg/dose) and temozolomide resulted in improvement of median survival of nude mice bearing orthotopic human glioblastoma xenografts compared with temozolomide alone (261 versus 192 days, respectively; P < or = 0.02). Reductions in neurologic dysfunction, brain edema, hemorrhage, and intratumoral microvessel density (CD34 staining) were observed in glioma-bearing mice receiving CEP-7055 alone, temozolomide alone, and the combination of CEP-7055 and temozolomide relative to vehicle and to temozolomide monotherapy. The administration of CEP-7055 in combination with irinotecan (20 mg/kg/dose i.p. x 5 days), and to a lesser degree with oxaliplatin (10 mg/kg/dose i.v.), showed reductions on primary colon carcinoma and hepatic metastatic burden in the CT-26 tumor model relative to that achieved by irinotecan and oxaliplatin monotherapy. These data show the significant efficacy and tolerability of optimal efficacious doses of CEP-7055 when given in combination with temozolomide and irinotecan relative to monotherapy with these cytotoxic agents in preclinical orthotopic glioma and colon carcinoma models and lend support for the use of these treatment regimens in a clinical setting in patients with glioblastoma and colon carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carbazoles/therapeutic use , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Glioblastoma/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Administration, Oral , Animals , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carbazoles/administration & dosage , Carcinoma/pathology , Colonic Neoplasms/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Glioblastoma/pathology , Humans , Irinotecan , Mice , Mice, Nude , Neoplasm Metastasis , Temozolomide , Xenograft Model Antitumor Assays
13.
Cancer Res ; 64(3): 910-9, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871820

ABSTRACT

Tumor-associated angiogenesis is critical for tumor growth and metastasis and is controlled by various pro- and antiangiogenic factors. The Eph family of receptor tyrosine kinases has emerged as one of the pivotal regulators of angiogenesis. Here we report that interfering with EphA signaling resulted in a pronounced inhibition of angiogenesis in ex vivo and in vivo model systems. Administration of EphA2/Fc soluble receptors inhibited, in a dose-dependent manner, microvessel formation in rat aortic ring assay, with inhibition reaching 76% at the highest dose of 5000 ng/ml. These results were further confirmed in vivo in a porcine aortic endothelial cell-vascular endothelial growth factor (VEGF)/basic fibroblast growth factor Matrigel plug assay, in which administration of EphA2/Fc soluble receptors resulted in 81% inhibition of neovascularization. The additive effects of simultaneous inhibition of VEGF receptor 2 and EphA signaling pathways in aortic ring assay and antiangiogenic efficacy of EphA2/Fc soluble receptors against VEGF/basic fibroblast growth factor-mediated neovascularization in vivo indicated a critical and nonredundant role for EphA signaling in angiogenesis. Furthermore, in two independent experiments, we demonstrated that EphA2/Fc soluble receptors strongly (by approximately 50% versus controls) suppressed growth of ASPC-1 human pancreatic tumor s.c. xenografts. Inhibition of tumor growth was due to decreased proliferation of tumor cells. In an orthotopic pancreatic ductal adenocarcinoma model in mice, suppression of EphA signaling by i.p. administration of EphA2/Fc (30 micro g/dose, three times a week for 56 days) profoundly inhibited the growth of primary tumors and the development of peritoneal, lymphatic, and hepatic metastases. These data demonstrate a critical role of EphA signaling in tumor growth and metastasis and provide a strong rationale for targeting EphA2 receptors for anticancer therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Receptor, EphA2/antagonists & inhibitors , Receptors, Fc/administration & dosage , Animals , Aorta/drug effects , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/metabolism , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Rats , Rats, Sprague-Dawley , Receptor, EphA2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
14.
Cancer Res ; 63(18): 5978-91, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522925

ABSTRACT

Inhibition of the vascular endothelial growth factor VEGF-VEGF receptor (VEGF-R) kinase axes in the tumor angiogenic cascade is a promising therapeutic strategy in oncology. CEP-7055 is the fully synthetic orally active N,N-dimethyl glycine ester of CEP-5214, a C3-(isopropylmethoxy) fused pyrrolocarbazole with potent pan-VEGF-R kinase inhibitory activity. CEP-5214 demonstrates IC(50) values of 18 nM, 12 nM, and 17 nM against human VEGF-R2/KDR kinase, VEGF-R1/FLT-1 kinase, and VEGF-R3/FLT-4 kinase, respectively, in biochemical kinase assays. CEP-5214 inhibited VEGF-stimulated VEGF-R2/KDR autophosphorylation in human umbilical vein endothelial cells (HUVECs) with an IC (50) of approximately 10 nM and demonstrated an equivalent inhibition of murine FLK-1 autophosphorylation in transformed SVR endothelial cells. Evaluation of the antiangiogenic activity of CEP-5214 revealed a dose-related inhibition of microvessel growth ex vivo in rat aortic ring explant cultures and in vitro on HUVEC capillary-tube formation on Matrigel at low nanomolar concentrations. The antiangiogenic activity of CEP-5214 in these bioassays was observed in the absence of apparent cytotoxicity. Single-dose p.o. or s.c. administration of CEP-7055 or CEP-5214 to CD-1 mice at 23.8 mg/kg/dose b.i.d. resulted in a reversible inhibition of VEGF-R2/FLK-1 phosphorylation in murine lung tissues. Administration p.o. of CEP-7055 at 2.57 to 23.8 mg/kg/dose b.i.d. resulted in dose-related reductions in neovascularization in vivo in porcine aortic endothelial cell (PAEC)-VEGF/basic fibroblast growth factor-Matrigel implants in nude mice (maximum, 82% inhibition), significant reductions in granuloma formation (30%) and granuloma vascularity (42%) in a murine chronic inflammation-induced angiogenesis model, and significant and sustained (6 h) inhibition of VEGF-induced plasma extravasation in rats, with an ED(50) of 20 mg/kg/dose. Chronic p.o. administration of CEP-7055 at doses of 11.9 to 23.8 mg/kg/dose b.i.d. resulted in significant inhibition (50-90% maximum inhibition relative to controls) in the growth of a variety of established murine and human s.c. tumor xenografts in nude mice, including A375 melanomas, U251MG and U87MG glioblastomas, CALU-6 lung carcinoma, ASPC-1 pancreatic carcinoma, HT-29 and HCT-116 colon carcinomas, MCF-7 breast carcinomas, and SVR angiosarcomas. Significant antitumor efficacy was observed similarly against orthotopically implanted LNCaP human prostate carcinomas in male nude mice and orthotopically implanted renal carcinoma (RENCA) tumors in BALB/c mice, in terms of a significant reduction in the metastatic score and the extent of pulmonary metastases. These antitumor responses were associated with marked increases in tumor apoptosis, and significant reductions in intratumoral microvessel density (CD34 and Factor VIII staining) of 22-38% relative to controls depending on the specific tumor xenograft. The antitumor efficacy of chronic CEP-7055 administration was independent of initial tumor volume (in the ASPC-1 pancreatic carcinoma model) and reversible on withdrawal of treatment. Chronic p.o. administration of CEP-7055 in preclinical efficacy studies for periods of up to 65 days was well tolerated with no apparent toxicity or significant morbidity. Orally administered CEP-7055 has entered Phase I clinical trials in cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Neoplasms, Experimental/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , In Vitro Techniques , Male , Mice , Mice, Nude , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Phosphorylation , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
15.
Mol Cancer Ther ; 2(4): 371-82, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12700281

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1) is a nuclear zinc finger DNA-binding protein that is implicated in the repair of DNA damage. Inhibition of PARP-1 through genetic knockouts causes cells to become hypersensitive to various chemotherapeutic agents. We tested the chemopotentiating ability of the PARP-1 inhibitor, CEP-6800, when used in combination with temozolomide (TMZ), irinotecan (camptothecin or SN38), and cisplatin against U251MG glioblastoma, HT29 colon carcinoma, and Calu-6 non-small cell lung carcinoma xenografts and cell lines, respectively. Exposure of tumor cells to TMZ, camptothecin (or SN38), and cisplatin before, or in the presence of, CEP-6800 significantly increased the onset and the magnitude of DNA damage, the duration for cells to effect repair, and the onset, duration, or fraction of cells arrested at the G(2)/M boundary. In addition, in vivo biochemical efficacy studies with CEP-6800 showed that it was able to attenuate irinotecan- and TMZ-induced poly(ADP-ribose) accumulation in LoVo and HT29 xenografts, respectively. Treatment of CEP 6800 (30 mg/kg) with TMZ (17 and 34 mg/kg) resulted in 100% complete regression of U251MG tumors by day 28 versus 60% complete regression caused by TMZ alone. CEP-6800 (30 mg/kg) in combination with irinotecan (10 mg/kg) resulted in a 60% inhibition of HT29 tumor growth versus irinotecan alone by day 33. The combination therapy of cisplatin (5 mg/kg) with CEP-6800 (30 mg/kg) caused a 35% reduction in Calu-6 tumor growth versus cisplatin alone by day 28. These data suggest that CEP-6800 could be used as a chemopotentiating agent with a variety of clinically effective chemotherapeutic agents.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cisplatin/pharmacology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Division , Cell Line , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Drug , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Irinotecan , Kinetics , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Models, Chemical , Neoplasm Transplantation , Temozolomide , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...