Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Helena J. Tutill is active.

Publication


Featured researches published by Helena J. Tutill.


Endocrine-related Cancer | 2008

Design and validation of specific inhibitors of 17β-hydroxysteroid dehydrogenases for therapeutic application in breast and prostate cancer, and in endometriosis

Joanna M. Day; Helena J. Tutill; Atul Purohit; Michael J. Reed

17beta-Hydroxysteroid dehydrogenases (17beta-HSDs) are enzymes that are responsible for reduction or oxidation of hormones, fatty acids and bile acids in vivo, regulating the amount of the active form that is available to bind to its cognate receptor. All require NAD(P)(H) for activity. Fifteen 17beta-HSDs have been identified to date, and with one exception, 17beta-HSD type 5 (17beta-HSD5), an aldo-keto reductase, they are all short-chain dehydrogenases/reductases, although overall homology between the enzymes is low. Although named as 17beta-HSDs, reflecting the major redox activity at the 17beta-position of the steroid, the activities of these 15 enzymes vary, with several of the 17beta-HSDs able to reduce and/or oxidise multiple substrates at various positions. These activities are involved in the progression of a number of diseases, including those related to steroid metabolism. Despite the success of inhibitors of steroidogenic enzymes in the clinic, such as those of aromatase and steroid sulphatase, the development of inhibitors of 17beta-HSDs is at a relatively early stage, as at present none have yet reached clinical trials. However, many groups are now working on inhibitors specific for several of these enzymes for the treatment of steroid-dependent diseases, including breast and prostate cancer, and endometriosis, with demonstrable efficacy in in vivo disease models. In this review, the recent advances in the validation of these enzymes as targets for the treatment of these diseases, with emphasis on 17beta-HSD1, 3 and 5, the development of specific inhibitors, the models used for their evaluation, and their progress towards the clinic will be discussed.


International Journal of Cancer | 2008

17β‐hydroxysteroid dehydrogenase Type 1, and not Type 12, is a target for endocrine therapy of hormone‐dependent breast cancer

Joanna M. Day; Paul A. Foster; Helena J. Tutill; Michael F.C. Parsons; Simon P. Newman; Surinder K. Chander; Gillian M. Allan; Harshani R. Lawrence; Nigel Vicker; Barry V. L. Potter; Michael J. Reed; Atul Purohit

Oestradiol (E2) stimulates the growth of hormone‐dependent breast cancer. 17β‐hydroxysteroid dehydrogenases (17β‐HSDs) catalyse the pre‐receptor activation/inactivation of hormones and other substrates. 17β‐HSD1 converts oestrone (E1) to active E2, but it has recently been suggested that another 17β‐HSD, 17β‐HSD12, may be the major enzyme that catalyses this reaction in women. Here we demonstrate that it is 17β‐HSD1 which is important for E2 production and report the inhibition of E1‐stimulated breast tumor growth by STX1040, a non‐oestrogenic selective inhibitor of 17β‐HSD1, using a novel murine model. 17β‐HSD1 and 17β‐HSD12 mRNA and protein expression, and E2 production, were assayed in wild type breast cancer cell lines and in cells after siRNA and cDNA transfection. Although 17β‐HSD12 was highly expressed in breast cancer cell lines, only 17β‐HSD1 efficiently catalysed E2 formation. The effect of STX1040 on the proliferation of E1‐stimulated T47D breast cancer cells was determined in vitro and in vivo. Cells inoculated into ovariectomised nude mice were stimulated using 0.05 or 0.1 μg E1 (s.c.) daily, and on day 35 the mice were dosed additionally with 20 mg/kg STX1040 s.c. daily for 28 days. STX1040 inhibited E1‐stimulated proliferation of T47D cells in vitro and significantly decreased tumor volumes and plasma E2 levels in vivo. In conclusion, a model was developed to study the inhibition of the major oestrogenic 17β‐HSD, 17β‐HSD1, in breast cancer. Both E2 production and tumor growth were inhibited by STX1040, suggesting that 17β‐HSD1 inhibitors such as STX1040 may provide a novel treatment for hormone‐dependent breast cancer.


Cancer Research | 2006

The role of 17β-hydroxysteroid dehydrogenases in modulating the activity of 2-methoxyestradiol in breast cancer cells

Simon P. Newman; Christopher R. Ireson; Helena J. Tutill; Joanna M. Day; Michael Parsons; Mathew P. Leese; Barry V. L. Potter; Michael J. Reed; Atul Purohit

The bis-sulfamoylated derivative of 2-methoxyestradiol (2-MeOE2), 2-methoxyestradiol-3,17-O,O-bis-sulfamate (2-MeOE2bisMATE), has shown potent antiproliferative and antiangiogenic activity in vitro and inhibits tumor growth in vivo. 2-MeOE2bisMATE is bioavailable, in contrast to 2-MeOE2 that has poor bioavailability. In this study, we have examined the role of 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 2 in the metabolism of 2-MeOE2. In MDA-MB-231 cells, which express high levels of 17beta-HSD type 2, and in MCF-7 cells transfected with 17beta-HSD type 2, high-performance liquid chromatography analysis showed that a significant proportion of 2-MeOE2 was metabolized to inactive 2-methoxyestrone. Furthermore, MCF-7 cells transfected with 17beta-HSD type 2 were protected from the cytotoxic effects of 2-MeOE2. In contrast, no significant metabolism of 2-MeOE2bisMATE was detected in transfected cells and 17beta-HSD type 2 transfection did not offer protection against 2-MeOE2bisMATE cytotoxicity. This study may go some way to explaining the poor bioavailability of 2-MeOE2, as the gastrointestinal mucosa expresses high levels of 17beta-HSD type 2. In addition, this study shows the value of synthesizing sulfamoylated derivatives of 2-MeOE2 with C17-position modifications as these compounds have improved bioavailability and potency both in vitro and in vivo.


Molecular and Cellular Endocrinology | 2006

17β-Hydroxysteroid dehydrogenase Type 1 and Type 2: Association between mRNA expression and activity in cell lines

Joanna M. Day; Helena J. Tutill; Simon P. Newman; Atul Purohit; Harshani R. Lawrence; Nigel Vicker; Barry V. L. Potter; Michael J. Reed

17Beta-hydroxysteroid dehydrogenases (17beta-HSDs) are a family of enzymes that regulate steroid availability within a tissue by catalysing the interconversion of active and inactive forms. Type 1 is up-regulated in many breast tumours, and is responsible for the reduction of oestrone to active oestradiol which stimulates cell proliferation within the tumour. Type 2 oxidises many active steroids to their inactive forms, including oestradiol to oestrone. In this study, we have compared the mRNA expression and enzyme activities of Type 1 and Type 2 in MCF-7, MDA-MB-231, T47D, JEG3 and 293-EBNA cell lines. Also studied were two cell lines stably expressing transfected Type 1 cDNA. RT-PCR indicated that little Type 1 mRNA is expressed in two of the breast cancer cell lines, MCF-7 and MDA-MB-231, and in 293-EBNA cells, but that expression is much higher in the T47D breast cancer cell line, and in the choriocarcinoma cell line, JEG3. However, a higher level of expression of Type 1 is seen in the transfected cell lines MCF-7.8H and 293-EBNA[His617beta-HSD1]. Activity assays show that there is high association between mRNA expression and enzyme activity. Assays indicate that, with the exception of MDA-MB-231 cells, Type 2 activity is low in these lines. The study of the basal activities of these enzymes will be used in future studies investigating the regulation of the enzymes by endogenous and exogenous factors. An understanding of their regulation in both healthy and malignant tissues may lead to future therapeutic intervention at the regulatory level.


Bioorganic & Medicinal Chemistry | 2008

Novel inhibitors of 17beta-hydroxysteroid dehydrogenase type 1: templates for design.

Gillian M. Allan; Nigel Vicker; Harshani R. Lawrence; Helena J. Tutill; Joanna M. Day; Marion Huchet; Eric Ferrandis; Michael J. Reed; Atul Purohit; Barry V. L. Potter

The 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyze the interconversion between the oxidized and reduced forms of androgens and estrogens at the 17 position. The 17beta-HSD type 1 enzyme (17beta-HSD1) catalyzes the reduction of estrone (E1) to estradiol and is expressed in malignant breast cells. Inhibitors of this enzyme thus have potential as treatments for hormone dependent breast cancer. Syntheses and biological evaluation of novel non-steroidal inhibitors designed to mimic the E1 template are reported using information from potent steroidal inhibitors. Of the templates investigated biphenyl ethanone was promising and led to inhibitors with IC(50) values in the low micromolar range.


ChemMedChem | 2006

Focused libraries of 16-substituted estrone derivatives and modified E-ring steroids : Inhibitors of 17ß-hydroxysteroid dehydrogenase type 1

Nigel Vicker; Harshani R. Lawrence; Gillian M. Allan; Christian Bubert; Andrew Smith; Helena J. Tutill; Atul Purohit; Joanna M. Day; Mary F. Mahon; Michael J. Reed; Barry V. L. Potter

17β‐Hydroxysteroid dehydrogenase type 1 (17β‐HSD1), an oxidoreductase which has a preferential reductive activity using NADPH as cofactor, converts estrone to estradiol and is expressed in many steroidogenic tissues including breast and in malignant breast cells. As estradiol stimulates the growth and development of hormone‐dependent breast cancer, inhibition of the final step of its synthesis is an attractive target for the treatment of this disease. The parallel synthesis of novel focused libraries of 16‐substituted estrone derivatives and modified E‐ring pyrazole steroids as new potent 17β‐HSD1 inhibitors is described. Substituted 3‐O‐sulfamoylated estrone derivatives were used as templates and were immobilised on 2‐chlorotrityl chloride resin to give resin‐bound scaffolds with a multi‐detachable linker. Novel focused libraries of 16‐substituted estrone derivatives and new modified E‐ring steroids were assembled from these immobilised templates using solid‐phase organic synthesis and solution‐phase methodologies. Among the derivatives synthesised, the most potent 17β‐HSD1 inhibitors were 25 and 26 with IC50 values in T‐47D human breast cancer cells of 27 and 165 nm, respectively. Parallel synthesis resulting in a library of C5′‐linked amides from the pyrazole E‐ring led to the identification of 62 with an IC50 value of 700 nM. These potent inhibitors of 17β‐HSD1 have a 2‐ethyl substituent which will decrease their estrogenic potential. Several novel 17β‐HSD1 inhibitors emerged from these libraries and these provide direction for further template exploration in this area. A new efficient diastereoselective synthesis of 25 has also been developed to facilitate supply for in vivo evaluation, and an X‐ray crystal structure of this inhibitor is presented.


Annals of the New York Academy of Sciences | 2009

The development of steroid sulfatase inhibitors for hormone-dependent cancer therapy.

Joanna M. Day; Atul Purohit; Helena J. Tutill; Paul A. Foster; L. W. Lawrence Woo; Barry V. L. Potter; Michael J. Reed

Steroid sulfatase (STS) regulates the hydrolysis of steroid sulfates to their unconjugated forms. Estrone sulfate and dehydroepiandrosterone sulfate can be hydrolyzed by STS to estrone and dehydroepiandrosterone, respectively, with these steroids being the precursors for the synthesis of more biologically active estrogens or androgens. A number of potent STS inhibitors have now been developed including STX64, which entered a phase I trial for the treatment of postmenopausal women with advanced metastatic hormone‐dependent breast cancer. The results from this phase I trial were encouraging, suggesting that STS inhibitors may also have a role in the treatment of other hormone‐dependent cancers including those of the endometrium, ovary, and prostate. In this paper the potential use of STS inhibitors to treat these hormone‐dependent cancers is reviewed. In addition, results from in vitro studies show that Ishikawa endometrial cancer cells, OVCAR‐3 ovarian cancer cells, and LNCaP prostate cancer cells all possess significant STS activity. Furthermore, STS activity in these cells can be almost completely inhibited by STX64 or the second‐generation STS inhibitor, STX213. Results from these investigations therefore suggest that STS inhibitors could have therapeutic potential for the treatment of a range of hormone‐dependent cancers.


Molecular and Cellular Endocrinology | 2006

Novel, potent inhibitors of 17β-hydroxysteroid dehydrogenase type 1

Gillian M. Allan; Christian Bubert; Nigel Vicker; Andrew Smith; Helena J. Tutill; Atul Purohit; Michael J. Reed; Barry V. L. Potter

Many breast tumours are hormone-responsive and rely on estrogens for their sustained growth and development. The enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) is primarily responsible for the conversion of estrone (E1) into the most potent of the human estrogens 17beta-estradiol (E2). Here we report the syntheses, inhibitory activities and docking studies for a novel series of pyrazole amides which have been discovered with the aim of probing the structure activity relationships (SAR) for such a template and of using this template to mimic the potent inhibitor 1 (Fig. 1). Amides containing an aromatic pyridyl moiety have been found to give the best inhibition, indicating that the pyridyl group interacts beneficially in the active site. This work has shown that extension from this position on the pyrazole template is well tolerated and the optimization of such systems is under investigation.


Molecular and Cellular Endocrinology | 2006

The regulation and inhibition of 17β-hydroxysteroid dehydrogenase in breast cancer

Atul Purohit; Helena J. Tutill; Joanna M. Day; Surinder K. Chander; Harshani R. Lawrence; Gillian M. Allan; Delphine S. Fischer; Nigel Vicker; Simon P. Newman; Barry V. L. Potter; Michael J. Reed

17Beta-hydroxysteroid dehydrogenase Type 1 (17beta-HSD1) has a pivotal role in regulating the synthesis of oestradiol (E2) within breast tumours. In whole body studies in postmenopausal women with breast cancer the conversion of oestrone (E1) to E2 (4.4+/-1.1%) was much lower than the inactivation of E2 to E1 (17.3+/-5.0%). In contrast, an examination of in vivo oestrogen metabolism within breast tumours revealed that whereas little metabolism of E2 occurred, E1 was converted to E2 to a much greater extent in malignant (48+/-14%) than in normal (19+/-6%) breast tissue. Findings from these studies originally suggested that oestrogen metabolism within breast tumours may differ from the mainly oxidative direction found in most other body tissues and that the activity of 17beta-HSD1 might be regulated by tumour-derived factors. Several growth factors (e.g. IGF-I, IGF-II) and cytokines (e.g. IL-6, TNFalpha) have now been identified which can markedly stimulate the activity of 17beta-HSD1 and such a mechanism may account for the high concentrations of E2 found in most breast tumours. Cells of the immune system, which can infiltrate breast tumours, are thought to be a major source of the growth factors and cytokines which can modulate 17beta-HSD1 activity. Given the central role that 17beta-HSD1 has in regulating breast tumour E2 concentrations the development of potent inhibitors of this enzyme has recently attracted considerable attention. Our initial studies in this area explored the use of derivatives of E1 as inhibitors, with 2-ethyl- and 2-methoxy E1 being found to inhibit 17beta-HSD1 activity in T-47D breast cancer cells by 96+/-2 and 91+/-1% respectively at 10 microM, but with a lack of specificity. Using the E1 scaffold a number of potent, selective 17beta-HSD1 inhibitors have now been identified including E1- and 2-ethyl-E1 containing a side chain with a m-pyridylmethylamidomethyl functionality extending from the 16beta position of the steroid nucleus. At 10 microM these compounds both inhibited 17beta-HSD1 activity by >90%, however some inhibition of 17beta-HSD2 activity was exhibited by the E1 derivative (25%) but not the 2-ethyl analogue. It is now apparent that 17beta-HSD1 activity contributes to the high E2 concentrations found in most breast tumours. The identification of potent, selective novel 17beta-HSD1 inhibitors will allow their efficacy to be tested in in vitro and in vivo studies.


Molecular and Cellular Endocrinology | 2009

The design of novel 17β-hydroxysteroid dehydrogenase type 3 inhibitors

Nigel Vicker; Christopher M. Sharland; Wesley Heaton; Ana Maria Ramos Gonzalez; Helen Victoria Bailey; Andrew Smith; Jeremy S. Springall; Joanna M. Day; Helena J. Tutill; Michael J. Reed; Atul Purohit; Barry V. L. Potter

17beta-Hydroxysteroid dehydrogenase type 3 (17beta-HSD3) is expressed at high levels in the testes and seminal vesicles but has also been shown to be present in prostate tissue, suggesting its potential involvement in both gonadal and non-gonadal testosterone biosynthesis. The role of 17beta-HSD3 in testosterone biosynthesis makes this enzyme an attractive molecular target for small molecule inhibitors for the treatment of prostate cancer. Here we report the design of selective inhibitors of 17beta-HSD3 as potential anti-cancer agents. Due to 17beta-HSD3 being a membrane-bound protein a crystal structure is not yet available. A homology model of 17beta-HSD3 has been built to aid structure-based drug design. This model has been used with docking studies to identify a series of lead compounds that may give an insight as to how inhibitors interact with the active site. Compound 1 was identified as a potent selective inhibitor of 17beta-HSD3 with an IC(50)=700nM resulting in the discovery of a novel lead series for further optimisation. Using our homology model as a tool for inhibitor design compound 5 was discovered as a novel potent and selective inhibitor of 17beta-HSD3 with an IC(50) approximately 200nM.

Collaboration


Dive into the Helena J. Tutill's collaboration.

Top Co-Authors

Avatar

Atul Purohit

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge