Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Pharmaceuticals (Basel) ; 14(12)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34959631

ABSTRACT

PR-104 is a phosphate ester pre-prodrug that is converted in vivo to its cognate alcohol, PR-104A, a latent alkylator which forms potent cytotoxins upon bioreduction. Hypoxia selectivity results from one-electron nitro reduction of PR-104A, in which cytochrome P450 oxidoreductase (POR) plays an important role. However, PR-104A also undergoes 'off-target' two-electron reduction by human aldo-keto reductase 1C3 (AKR1C3), resulting in activation in oxygenated tissues. AKR1C3 expression in human myeloid progenitor cells probably accounts for the dose-limiting myelotoxicity of PR-104 documented in clinical trials, resulting in human PR-104A plasma exposure levels 3.4- to 9.6-fold lower than can be achieved in murine models. Structure-based design to eliminate AKR1C3 activation thus represents a strategy for restoring the therapeutic window of this class of agent in humans. Here, we identified SN29176, a PR-104A analogue resistant to human AKR1C3 activation. SN29176 retains hypoxia selectivity in vitro with aerobic/hypoxic IC50 ratios of 9 to 145, remains a substrate for POR and triggers γH2AX induction and cell cycle arrest in a comparable manner to PR-104A. SN35141, the soluble phosphate pre-prodrug of SN29176, exhibited superior hypoxic tumour log cell kill (>4.0) to PR-104 (2.5-3.7) in vivo at doses predicted to be achievable in humans. Orthologues of human AKR1C3 from mouse, rat and dog were incapable of reducing PR-104A, thus identifying an underlying cause for the discrepancy in PR-104 tolerance in pre-clinical models versus humans. In contrast, the macaque AKR1C3 gene orthologue was able to metabolise PR-104A, indicating that this species may be suitable for evaluating the toxicokinetics of PR-104 analogues for clinical development. We confirmed that SN29176 was not a substrate for AKR1C3 orthologues across all four pre-clinical species, demonstrating that this prodrug analogue class is suitable for further development. Based on these findings, a prodrug candidate was subsequently identified for clinical trials.

3.
Biochem Pharmacol ; 91(4): 436-46, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25130546

ABSTRACT

SN30000 is a second-generation benzotriazine-N-oxide hypoxia-activated prodrug scheduled for clinical trial. Previously we showed that covalent binding of the hypoxia probe EF5 predicts metabolic activation of SN30000 in a panel of cancer cell lines under anoxia, suggesting that they are activated by the same reductases. However the identity of these reductases is unknown. Here, we test whether forced expression of nine oxidoreductases with known or suspected roles in bioreductive prodrug metabolism (AKR1C3, CYB5R3, FDXR, MTRR, NDOR1, NOS2A, NQO1, NQO2 and POR) enhances oxic or anoxic reduction of SN30000 and EF5 by HCT116 cells. Covalent binding of (14)C-EF5 and reduction of SN30000 to its 1-oxide and nor-oxide metabolites was highly selective for anoxia in all lines, with significantly elevated anoxic metabolism of both compounds in lines over-expressing POR, MTRR, NOS2A or NDOR1. There was a strong correlation between EF5 binding and SN30000 metabolism under anoxia across the cell lines (R(2)=0.84, p=0.0001). Antiproliferative potency of SN30000 under anoxia was increased most strongly by overexpression of MTRR and POR. Transcript abundance in human tumours, evaluated using public domain mRNA expression data, was highest for MTRR, followed by POR, NOS2A and NDOR1, with little variation between tumour types. Immunostaining of tissue microarrays demonstrated variable MTRR protein expression across 517 human cancers with most displaying low expression. In conclusion, we have identified four diflavin reductases (POR, MTRR, NOS2A and NDOR1) capable of reducing both SN30000 and EF5, further supporting use of 2-nitroimidazole probes to predict the ability of hypoxic cells to activate SN30000.


Subject(s)
Cell Hypoxia , Cyclic N-Oxides/pharmacology , Oxidoreductases/metabolism , Prodrugs/pharmacology , Triazines/pharmacology , Electrons , Ferredoxin-NADP Reductase/metabolism , Flow Cytometry , HCT116 Cells , Humans
4.
Mol Pharmacol ; 81(1): 31-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21984255

ABSTRACT

The clinical agent PR-104 is converted systemically to PR-104A, a nitrogen mustard prodrug designed to target tumor hypoxia. Reductive activation of PR-104A is initiated by one-electron oxidoreductases in a process reversed by oxygen. The identity of these oxidoreductases is unknown, with the exception of cytochrome P450 reductase (POR). To identify other hypoxia-selective PR-104A reductases, nine candidate oxidoreductases were expressed in HCT116 cells. Increased PR-104A-cytotoxicity was observed in cells expressing methionine synthase reductase (MTRR), novel diflavin oxidoreductase 1 (NDOR1), and inducible nitric-oxide synthase (NOS2A), in addition to POR. Plasmid-based expression of these diflavin oxidoreductases also enhanced bioreductive metabolism of PR-104A in an anoxia-specific manner. Diflavin oxidoreductase-dependent PR-104A metabolism was suppressed >90% by pan-flavoenzyme inhibition with diphenyliodonium chloride. Antibodies were used to quantify endogenous POR, MTRR, NDOR1, and NOS2A expression in 23 human tumor cell lines; however, only POR protein was detectable and its expression correlated with anoxic PR-104A reduction (r(2) = 0.712). An anti-POR monoclonal antibody was used to probe expression using human tissue microarrays; 13 of 19 cancer types expressed detectable POR with 21% of cores (185 of 874) staining positive; this heterogeneity suggests that POR is a useful biomarker for PR-104A activation. Immunostaining for carbonic anhydrase 9 (CAIX), reportedly an endogenous marker of hypoxia, revealed only moderate coexpression (9.6%) of both CAIX and POR across a subset of five cancer types.


Subject(s)
Flavins/physiology , Flavoproteins/physiology , Nitrogen Mustard Compounds/metabolism , Oxidoreductases/physiology , Prodrugs/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Flavoproteins/metabolism , HCT116 Cells , Humans , Mice , Mice, Nude , NADPH-Ferrihemoprotein Reductase/physiology , Nitrogen Mustard Compounds/pharmacology , Oxidation-Reduction , Oxidoreductases/metabolism
5.
J Cutan Pathol ; 33(5): 383-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16640548

ABSTRACT

BACKGROUND: The most frequent molecular abnormality observed in dermatofibrosarcoma protuberans (DFSP) is the formation of a supernumerary ring chromosome or translocation resulting in fusion of the gene encoding the alpha-chain of type 1 collagen, COL1A1 from 17q22, to the platelet-derived growth factor beta-chain, PDGFB gene from 22q13. Rare cases documenting variant ring or marker chromosomes involving regions other than 17q22 and 22q13 have been reported. Further analysis in three of these cases demonstrated the presence of the COL1A1 and PDGFB genes. METHODS: We report a further case of DFSP with a rare variant ring chromosome. The tumor appeared to undergo accelerated growth during pregnancy, then metastasized following pregnancy. We describe the clinical, histological, immunohistochemical, and cytogenetic features. RESULTS: The metastatic tumor showed a variant r(17;?) chromosome. A locus-specific probe was required to demonstrate presence of the PDGFB gene within the ring, indicating cryptic molecular rearrangement between chromosomes 17 and 22, and recombination with an unknown chromosome. CONCLUSIONS: Cryptic rearrangement of chromosomes 17 and 22 should be suspected in variant ring chromosomes and translocations. Pregnancy may contribute to accelerated growth of DFSP, and delay in surgical resection should be avoided.


Subject(s)
Dermatofibrosarcoma/genetics , Dermatofibrosarcoma/pathology , Pregnancy Complications, Neoplastic/genetics , Ring Chromosomes , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Adult , Antineoplastic Agents/therapeutic use , Benzamides , Breast Neoplasms/drug therapy , Breast Neoplasms/secondary , Breast Neoplasms/surgery , Dermatofibrosarcoma/secondary , Female , Humans , Imatinib Mesylate , In Situ Hybridization , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mastectomy , Piperazines/therapeutic use , Pregnancy , Pregnancy Complications, Neoplastic/pathology , Pyrimidines/therapeutic use , Skin Neoplasms/surgery
6.
Ann Thorac Surg ; 75(6): 1971-3, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12822653

ABSTRACT

We report an unusual study of a 72-year-old woman who presented with a symptomatic space-occupying lesion in the left ventricle. The lesion was surgically excised with a good long-term result. It was histologicaly diagnosed as an inflammatory myofibroblastic tumor. Also known as an inflammatory pseudotumor, these tumors affecting the heart are extremely rare and occur mainly in children and adolescents. We review the literature and highlight the unique nature of the lesion.


Subject(s)
Heart Neoplasms/surgery , Heart Ventricles/surgery , Myxosarcoma/surgery , Neoplasms, Muscle Tissue/surgery , Papillary Muscles/surgery , Aged , Diagnosis, Differential , Female , Heart Neoplasms/diagnosis , Heart Neoplasms/pathology , Heart Ventricles/pathology , Humans , Magnetic Resonance Imaging , Myocardium/pathology , Myxosarcoma/pathology , Neoplasms, Muscle Tissue/pathology , Papillary Muscles/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...