Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Cancer Ther ; 22(6): 765-777, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37042205

ABSTRACT

MET, the cell-surface receptor for the hepatocyte growth factor/scatter factor, which is widely overexpressed in various solid cancer types, is an attractive target for the development of antibody-based therapeutics. BYON3521 is a novel site-specifically conjugated duocarmycin-based antibody-drug conjugate (ADC), comprising a humanized cysteine-engineered IgG1 monoclonal antibody with low pmol/L binding affinity towards both human and cynomolgus MET. In vitro studies showed that BYON3521 internalizes efficiently upon MET binding and induces both target- and bystander-mediated cell killing. BYON3521 showed good potency and full efficacy in MET-amplified and high MET-expressing cancer cell lines; in moderate and low MET-expressing cancer cell lines good potencies and partial efficacy were observed. In mouse xenograft models, BYON3521 showed significant antitumor activity upon single-dose administration in multiple non-MET-amplified tumor types with low, moderate, and high MET expression, including complete tumor remissions in models with moderate MET expression. In the repeat-dose Good Laboratory Practice (GLP) safety assessment in cynomolgus monkeys, BYON3521 was well tolerated and based on the observed toxicities and their reversibility, the highest non-severely toxic dose was set at 15 mg/kg. A human pharmacokinetics (PK) model was derived from the PK data from the cynomolgus safety assessments, and the minimal efficacious dose in humans is estimated to be in the range of 3 to 4 mg/kg. In all, our nonclinical data suggests that BYON3521 is a safe ADC with potential for clinical benefit in patients. A first-in-human dose-escalation study is currently ongoing to determine the maximum tolerated dose and recommended dose for expansion (NCT05323045).


Subject(s)
Antibodies, Monoclonal , Immunoconjugates , Animals , Humans , Mice , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Immunoglobulin G , Xenograft Model Antitumor Assays
2.
Bioconjug Chem ; 31(9): 2136-2146, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32697078

ABSTRACT

Engineering cysteines at specific sites in antibodies to create well-defined ADCs for the treatment of cancer is a promising approach to increase the therapeutic index and helps to streamline the manufacturing process. Here, we report the development of an in silico screening procedure to select for optimal sites in an antibody to which a hydrophobic linker-drug can be conjugated. Sites were identified inside the cavity that is naturally present in the Fab part of the antibody. Conjugating a linker-drug to these sites demonstrated the ability of the antibody to shield the hydrophobic character of the linker-drug while resulting ADCs maintained their cytotoxic potency in vitro. Comparison of site-specific ADCs versus randomly conjugated ADCs in an in vivo xenograft model revealed improved efficacy and exposure. We also report a selective reducing agent that is able to reduce the engineered cysteines while leaving the interchain disulfides in the oxidized state. This enables us to manufacture site-specific ADCs without introducing impurities associated with the conventional reduction/oxidation procedure for site-specific conjugation.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Cysteine/chemistry , Duocarmycins/analogs & derivatives , Immunoconjugates/chemistry , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Duocarmycins/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/therapeutic use , Immunoglobulin G/chemistry , Immunoglobulin G/therapeutic use , Mice , Models, Molecular , Neoplasms/drug therapy , Oxidation-Reduction
3.
Lab Anim ; 54(4): 365-372, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31366270

ABSTRACT

Chronic pain and subfertility are the main symptoms of concern in women with endometriosis. In order to find new therapeutic options to suppress the pain, translational animal models are indispensable. We have developed a new automated, experimental setup, with full consideration for animal wellbeing, to determine whether operant behaviour can reveal abdominal hyperalgesia in rats with surgically-induced endometriosis, in order to assess whether abdominal hyperalgesia affect behavioural parameters. Endometriosis was induced by transplantation of uterine fragments in the abdominal cavity. Control groups consisted of sham-operated rats and non-operated rats. We have developed an operant chamber (Skinnerbox) which includes a barrier. The rat can climb the barrier in order to reach the food pellet, increasing in this way the pressure to the abdomen. We show that endometriosis rats collect significantly less sugar pellets when compared with the control rats after the introduction of the barrier. In the Skinnerbox experiment, we showed that in a positive operant setting, the introduction of a barrier results in a contrast of operant behaviour of endometriosis rats and control groups, perchance as a result of abdominal discomfort/hyperalgesia due to surgically-induced endometriosis. This is a promising start for the further development of a refined animal model to monitor abdominal discomfort/hyperalgesia in rats with surgically-induced endometriosis.


Subject(s)
Abdomen/physiopathology , Endometriosis/complications , Hyperalgesia/physiopathology , Animals , Disease Models, Animal , Endometriosis/physiopathology , Female , Hyperalgesia/diagnosis , Rats , Rats, Wistar
4.
Mol Cancer Ther ; 17(11): 2389-2398, 2018 11.
Article in English | MEDLINE | ID: mdl-30093567

ABSTRACT

Carboxylesterase 1c (CES1c) is responsible for linker-drug instability and poor pharmacokinetics (PK) of several antibody-drug conjugates (ADC) in mice, but not in monkeys or humans. Preclinical development of these ADCs could be improved if the PK in mice would more closely resemble that of humans and is not affected by an enzyme that is irrelevant for humans. SYD985, a HER2-targeting ADC based on trastuzumab and linker-drug vc-seco-DUBA, is also sensitive to CES1c. In the present studies, we first focused on the interaction between CES1c and SYD985 by size- exclusion chromatography, Western blotting, and LC/MS-MS analysis, using recombinant CES1c and plasma samples. Intriguingly, CES1c activity not only results in release of the active toxin DUBA but also in formation of a covalent bond between CES1c and the linker of vc-seco-DUBA. Mass spectrometric studies enabled identification of the CES1c cleavage site on the linker-drug and the structure of the CES1c adduct. To assess the in vivo impact, CES1c-/- SCID mice were generated that showed stable PK for SYD985, comparable to that in monkeys and humans. Patient-derived xenograft (PDX) studies in these mice showed enhanced efficacy compared with PDX studies in CES1c+/+ mice and provided a more accurate prediction of clinical efficacy of SYD985, hence delivering better quality data. It seems reasonable to assume that CES1c-/- SCID mice can increase quality in ADC development much broader for all ADCs that carry linker-drugs susceptible to CES1c, without the need of chemically modifying the linker-drug to specifically increase PK in mice. Mol Cancer Ther; 17(11); 2389-98. ©2018 AACR.


Subject(s)
Carboxylesterase/deficiency , Immunoconjugates/pharmacology , Immunoconjugates/pharmacokinetics , Animals , Carboxylesterase/metabolism , Catalytic Domain , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Mice, Knockout , Mice, SCID , Peptides/chemistry , Rats, Wistar , Trastuzumab/chemistry , Treatment Outcome
5.
Reprod Sci ; 25(9): 1318-1329, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29962279

ABSTRACT

The pipelines of pharmaceutical companies are filled with thousands of promising new compounds for a plethora of indications. Yet, a close review of the drugs that have recently been in clinical trials quickly reveals that only a handful of drugs under evaluation in women with endometriosis can be genuinely qualified as truly innovative and breakthrough drugs. Why is there such an industry-wide lukewarm interest in drug research and development for endometriosis/adenomyosis? Why are pharmaceutical companies so reluctant to initiate programs or invest in academic research in endometriosis/adenomyosis? It is evident that a substantial part of the novel druggable targets originate from research in academia. However, only the pharmaceutical industry has the resources and expertise to bring drugs to patients. In other words, we are fully dependent on the pharmaceutical industry to bring new therapeutics to the market. The aim of this editorial is to make scientists from academia aware of the enormous complexity of the drug development process, the drivers that propel pharmaceutical companies to initiate new programs and to prioritize their portfolios, the value of intellectual property rights, and also about the importance of scientific rigor, predictive translational models, and biomarkers. At the same time, the pharmaceutical industry must be made aware of the enormous opportunity at hand, as the current patient population with endometriosis/adenomyosis is just the tip of the iceberg. We hope that the insights presented here will enable the endometriosis/adenomyosis research community to find ways to valorize their knowledge and attract the interest of the industry.


Subject(s)
Adenomyosis/drug therapy , Drug Industry , Endometriosis/drug therapy , Research , Female , Humans
6.
Hum Reprod Update ; 24(5): 577-598, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29893860

ABSTRACT

BACKGROUND: The drug research and development (R&D) for endometriosis/adenomyosis has been painfully slow. Most completed clinical trials on endometriosis did not publish their results, and presumably failed. While few published trials did report how they foundered, the reasons why they failed are often completely unclear. Surprisingly, there has been no open discussion on why these trials failed. If the causes for these failed trials remain unelucidated, mistakes made in these failed trials may be repeated in the future. Since failure can be infinitely more instructive and educational than success, elucidating the causes for failed clinical trials may yield a treasure trove for future drug R&D. Given our growing understanding of the natural history of ectopic endometrium, it is also important to make an inventory of biologicals/compounds that are currently under development to see where we stand and whether they would stand a better chance of gaining regulatory approval than their predecessors. OBJECTIVE AND RATIONALE: We provide an overview of all compounds under clinical investigation and in development in order to assess the evolution of R&D since the last inventory, reported in 2013. We also have attempted to analyse selected failed clinical trials in the context of published translational/preclinical research and our growing understanding of the natural history of endometriotic/adenomyotic lesions, in the hope that the lessons learned will steer investigators toward the right track in future drug R&D. SEARCH METHODS: We searched ClinicalTrials.gov and a database containing information on drugs gathered daily by Thomson Reuters from a wide range of sources (e.g. patent offices, biomedical literature, congresses, symposia, meetings, company information, regulatory information) for all therapeutic compounds that have undergone or are under clinical trials, or in the developmental stage, and then searched PubMed and Google to determine their publication status using trial identifiers. For trials that were completed at least 2 years ago and have, or have not, published their results, a PubMed search was performed using the name of the therapeutic that has been tested and 'endometriosis' or 'adenomyosis' to identify published preclinical studies prior to the launch of the trial. For those published trials, the cited preclinical studies were also retrieved and scrutinized. OUTCOMES: Despite repeated calls for more transparency, only a small fraction of completed trials on endometriosis has been published. A large number of 'novel' compounds under development are simply repurposed drugs, which seem to be ill-prepared to combat the fibroproliferative nature of endometriosis/adenomyosis. This sobering picture indicates an alarming innovation 'drought' in the drug R&D front, resulting in trickling drug pipelines. Some trials foundered owing to unanticipated serious side-effects, or because attempts were made to suppress a target that can be compensated for by redundant pathways, but many failed in efficacy, indicating that the translational value of the current models is seriously questionable. All existing animal models of endometriosis do not recapitulate the key features of human conditions. WIDER IMPLICATIONS: The glaring innovation drought in drug R&D for endometriosis/adenomyosis should sound alarms to all stake-holders. The failed clinical trials in endometriosis also indicate that some past research had serious deficiencies. In light of the recent understanding of the natural history of ectopic endometrium, it is perhaps time to shift the research paradigm and revamp our research focus and priorities.


Subject(s)
Adenomyosis/drug therapy , Clinical Trials as Topic , Drug Development , Endometriosis/drug therapy , Female , Hormone Antagonists/adverse effects , Hormone Antagonists/therapeutic use , Humans , Infliximab/adverse effects , Infliximab/therapeutic use , Oxazoles/adverse effects , Oxazoles/therapeutic use , Progesterone/agonists , Progesterone/antagonists & inhibitors , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/therapeutic use , Treatment Outcome
7.
Fertil Steril ; 107(3): 699-706.e6, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28259259

ABSTRACT

OBJECTIVE: To identify metabolites that are associated with and predict the presence of endometriosis. DESIGN: Metabolomics study using state-of-the-art mass spectrometry approaches. SETTING: University hospital and universities. PATIENT(S): Twenty-five women with laparoscopically confirmed endometriosis (cases) and 19 women with laparoscopically documented absence of endometriosis (controls). None of the women included in this study had received oral contraception or GnRH agonists for a minimum of 1 month before blood collection. INTERVENTION(S): Plasma collection. MAIN OUTCOME MEASURE(S): Metabolite profiles were generated and interrogated using multiple mass spectrometry methods, that is, high performance liquid chromatography coupled with negative mode electrospray ionization tandem mass spectrometry, UPLC-MS/MS, and ultra performance liquid chromatography-electroSpray ionization-quadrupole time-of-flight (UPLC-ESI-Q-TOF). Metabolite groups investigated included phospholipids, glycerophospholipids, ether-phospholipids, cholesterol-esters, triacylglycerol, sphingolipids, free fatty acids, steroids, eicosanoids, and acylcarnitines. RESULT(S): A panel of acylcarnitines predicted the presence of endometriosis with 88.9% specificity and 81.5% sensitivity in human plasma, with a positive predictive value of 75%. However, due to data limitations the outcome of the receiver operating characteristic curve analysis was not significant. CONCLUSION(S): A diagnostic model based on acylcarnitines has the potential to predict the presence and stage of endometriosis.


Subject(s)
Carnitine/analogs & derivatives , Endometriosis/blood , Lipids/blood , Metabolomics , Adult , Area Under Curve , Belgium , Biomarkers/blood , Carnitine/blood , Case-Control Studies , Chromatography, High Pressure Liquid , Endometriosis/diagnosis , Female , Hospitals, University , Humans , Laparoscopy , Metabolomics/methods , Pilot Projects , Predictive Value of Tests , ROC Curve , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
8.
Mol Cancer Ther ; 14(3): 692-703, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589493

ABSTRACT

SYD985 is a HER2-targeting antibody-drug conjugate (ADC) based on trastuzumab and vc-seco-DUBA, a cleavable linker-duocarmycin payload. To evaluate the therapeutic potential of this new ADC, mechanistic in vitro studies and in vivo patient-derived xenograft (PDX) studies were conducted to compare SYD985 head-to-head with T-DM1 (Kadcyla), another trastuzumab-based ADC. SYD985 and T-DM1 had similar binding affinities to HER2 and showed similar internalization. In vitro cytotoxicity assays showed similar potencies and efficacies in HER2 3+ cell lines, but in cell lines with low HER2 expression, SYD985 was 3- to 50-fold more potent than T-DM1. In contrast with T-DM1, SYD985 efficiently induced bystander killing in vitro in HER2-negative (HER2 0) cells mixed with HER2 3+, 2+, or 1+ cell lines. At pH conditions relevant for tumors, cathepsin-B cleavage studies showed efficient release of the active toxin by SYD985 but not by T-DM1. These in vitro data suggest that SYD985 might be a more potent ADC in HER2-expressing tumors in vivo, especially in low HER2-expressing and/or in heterogeneous tumors. In line with this, in vivo antitumor studies in breast cancer PDX models showed that SYD985 is very active in HER2 3+, 2+, and 1+ models, whereas T-DM1 only showed significant antitumor activity in HER2 3+ breast cancer PDX models. These properties of SYD985 may enable expansion of the target population to patients who have low HER2-expressing breast cancer, a patient population with still unmet high medical need.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Indoles/pharmacology , Receptor, ErbB-2/genetics , Animals , Cell Line, Tumor , Duocarmycins , Female , Humans , Mice , Mice, Nude , Pyrrolidinones/pharmacology , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays/methods
9.
Proc Natl Acad Sci U S A ; 111(46): 16514-9, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368192

ABSTRACT

It recently has been recognized that men develop colonic adenomas and carcinomas at an earlier age and at a higher rate than women. In the Apc(Pirc/+) (Pirc) rat model of early colonic cancer, this sex susceptibility was recapitulated, with male Pirc rats developing twice as many adenomas as females. Analysis of large datasets revealed that the Apc(Min/+) mouse also shows enhanced male susceptibility to adenomagenesis, but only in the colon. In addition, WT mice treated with injections of the carcinogen azoxymethane (AOM) showed increased numbers of colonic adenomas in males. The mechanism underlying these observations was investigated by manipulation of hormonal status. The preponderance of colonic adenomas in the Pirc rat model allowed a statistically significant investigation in vivo of the mechanism of sex hormone action on the development of colonic adenomas. Females depleted of endogenous hormones by ovariectomy did not exhibit a change in prevalence of adenomas, nor was any effect observed with replacement of one or a combination of female hormones. In contrast, depletion of male hormones by orchidectomy (castration) markedly protected the Pirc rat from adenoma development, whereas supplementation with testosterone reversed that effect. These observations were recapitulated in the AOM mouse model. Androgen receptor was undetectable in the colon or adenomas, making it likely that testosterone acts indirectly on the tumor lineage. Our findings suggest that indirect tumor-promoting effects of testosterone likely explain the disparity between the sexes in the development of colonic adenomas.


Subject(s)
Adenoma/epidemiology , Carcinogens/toxicity , Colonic Neoplasms/epidemiology , Dihydrotestosterone/toxicity , Gonadal Steroid Hormones/physiology , Neoplasms, Hormone-Dependent/epidemiology , Adenoma/chemically induced , Adenoma/physiopathology , Adenoma/prevention & control , Adenomatous Polyposis Coli/epidemiology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/physiopathology , Animals , Animals, Congenic , Azoxymethane/toxicity , Colonic Neoplasms/chemically induced , Colonic Neoplasms/physiopathology , Colonic Neoplasms/prevention & control , Disease Models, Animal , Estradiol/administration & dosage , Estradiol/pharmacology , Female , Genes, APC , Hormone Replacement Therapy , Humans , Male , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred C57BL , Mutation , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/prevention & control , Orchiectomy , Organ Specificity , Ovariectomy , Postmenopause , RNA, Messenger/analysis , Random Allocation , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Sex Distribution , Species Specificity
11.
PLoS One ; 6(7): e22620, 2011.
Article in English | MEDLINE | ID: mdl-21818351

ABSTRACT

Clinical data suggest that progestins have chemopreventive properties in the development of colorectal cancer. We set out to examine a potential protective effect of progestins and progesterone signaling on colon cancer development. In normal and neoplastic intestinal tissue, we found that the progesterone receptor (PR) is not expressed. Expression was confined to sporadic mesenchymal cells. To analyze the influence of systemic progesterone receptor signaling, we crossed mice that lacked the progesterone receptor (PRKO) to the Apc(Min/+) mouse, a model for spontaneous intestinal polyposis. PRKO-Apc(Min/+) mice exhibited no change in polyp number, size or localization compared to Apc(Min/+). To examine effects of progestins on the intestinal epithelium that are independent of the PR, we treated mice with MPA. We found no effects of either progesterone or MPA on gross intestinal morphology or epithelial proliferation. Also, in rats treated with MPA, injection with the carcinogen azoxymethane did not result in a difference in the number or size of aberrant crypt foci, a surrogate end-point for adenoma development. We conclude that expression of the progesterone receptor is limited to cells in the intestinal mesenchyme. We did not observe any effect of progesterone receptor signaling or of progestin treatment in rodent models of intestinal tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Progesterone/metabolism , Signal Transduction , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Colon/drug effects , Colon/metabolism , Colon/pathology , Disease Models, Animal , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Polyposis/metabolism , Intestinal Polyposis/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , Progestins/pharmacology , Rats , Receptors, Progesterone/metabolism , Signal Transduction/drug effects
12.
Fertil Steril ; 95(4): 1421-7, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316665

ABSTRACT

OBJECTIVE: To determine [1] expression levels of both DNA methyltransferases (DNMTs) and methyl-CpG-binding domain proteins (MBDs) in human endometrium throughout the menstrual cycle and in eutopic and ectopic endometrium of patients with endometriosis and [2] hormone responsiveness of DNMT and MBD expression in explant cultures of proliferative phase endometrium. DESIGN: In vitro study. SETTING: Academic medical center. PATIENT(S): Premenopausal women with and without endometriosis. INTERVENTION(S): Explant cultures of proliferative phase endometrium were treated with vehicle, 17ß-E(2), or a combination of E(2) and P (E(2) + P) for 24 hours. MAIN OUTCOME MEASURE(S): Expression levels of DNMT1, DNMT2, and DNMT3B and MBD1, MBD2, and MeCP2 with use of real-time quantitative polymerase chain reaction. RESULT(S): Expression levels of DNMT1 and MBD2 were significantly higher in secretory-phase endometrium compared with proliferative endometrium and menstrual endometrium. In explant cultures, treatment with E(2) + P resulted in significant up-regulation of DNMT1 and MBD2. Expression levels of several DNMTs and MBDs were significantly lower in endometriotic lesions compared with eutopic endometrium of women with endometriosis and disease-free controls. CONCLUSION(S): These findings suggest a role for DNMTs and MBDs in the growth and differentiation of the human endometrium and support the notion that endometriosis may be an epigenetic disease.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA-Binding Proteins/biosynthesis , Endometriosis/metabolism , Endometrium/metabolism , Transcription Factors/biosynthesis , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Endometriosis/enzymology , Endometriosis/genetics , Endometrium/enzymology , Endometrium/pathology , Female , Humans , Protein Structure, Tertiary/genetics , Transcription Factors/genetics , Up-Regulation/genetics
13.
Contraception ; 78(3): 257-65, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18692618

ABSTRACT

BACKGROUND: Fundamental and genetic differences between women in the endometrium may cause some to develop endometriosis, whereas others do not. Oral contraceptives (OC) may have an effect on the endometrium, rendering the development of endometriosis less likely. STUDY DESIGN: Endometrium from women using OC (OCE) and menstrual endometrium (ME) from normal cycling women were transplanted onto the chicken chorioallantoic membrane (CAM), and endometriosis-like lesion formation was evaluated. Microarray gene expression profiling was performed to identify differentially expressed genes in the endometrium from these groups. Microarray data were validated by real-time PCR. RESULTS: Less endometriosis-like lesions were formed after transplantation of OCE than after transplantation of ME (p<.05). Most of the differentially expressed genes belong to the TGFbeta superfamily. Real-time PCR validation revealed that inhibin betaA (INHBA) expression was significantly decreased in OCE as compared to ME. CONCLUSION: OC use affects the characteristics of endometrium, rendering it less potent to develop into endometriosis.


Subject(s)
Contraceptives, Oral, Hormonal/pharmacology , Endometriosis/prevention & control , Endometrium/drug effects , Adult , Animals , Case-Control Studies , Chickens , Chorioallantoic Membrane , Endometrium/transplantation , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Tissue Culture Techniques , Young Adult
14.
Mol Hum Reprod ; 14(5): 259-68, 2008 May.
Article in English | MEDLINE | ID: mdl-18430758

ABSTRACT

It is widely known that angiogenesis plays a key role in endometriotic lesion formation and development. Antiangiogenic treatments aimed at inhibiting new vessel formation have proven efficient in experimental models. However, as antiangiogenic strategies do not target pre-existing pericyte-protected vessels, they require chronic administration and are likely to be beneficial for early-stage disease only or to prevent recurrence after surgery. Moreover, they may have detrimental effects on reproductive function. Vascular-disrupting agents (VDAs) have emerged as a promising new tool for the treatment of tumors. VDAs target established blood vessels, resulting in tumor ischemia and necrosis. These agents may therefore be more efficient against advanced disease. Two major types of VDAs are being developed for cancer: ligand-directed VDAs using antibodies, peptides and growth factors to deliver toxic effectors to tumor endothelium; and small-molecule VDAs exploiting physiological differences between tumor and normal endothelium to induce acute vascular shutdown. The ongoing evolution in genomics and proteomics is revolutionizing the discovery of novel endothelial markers. Several studies suggest that the vasculature of endometriotic lesions may have particular pathophysiological properties, which could be exploited for the development of selective VDAs. The aim of this review is to explore the merits and limitations of vascular therapy for the treatment of endometriosis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endometriosis/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/adverse effects , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Disease Progression , Endometriosis/pathology , Endometrium/blood supply , Endometrium/drug effects , Endometrium/pathology , Female , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology
15.
Endocrinology ; 148(10): 5020-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17640988

ABSTRACT

Using cDNA microarray analysis, we identified SGK1 (serum- and glucocorticoid-regulated kinase 1) as a gene aberrantly expressed in midsecretory endometrium of women with unexplained infertility. SGK1 is a serine/threonine kinase involved primarily in epithelial ion transport and cell survival responses. Real-time quantitative PCR analysis of a larger, independent sample set timed to coincide with the period of uterine receptivity confirmed increased expression of SGK1 transcripts in infertile women compared with fertile controls. We further demonstrate that SGK1 expression is regulated by progesterone in human endometrium in vivo as well as in explant cultures. During the midsecretory phase of the cycle, SGK1 mRNA and protein were predominantly but not exclusively expressed in the luminal epithelium, and expression in this cellular compartment was higher in infertile women. In the stromal compartment, SGK1 expression was largely confined to decidualizing cells adjacent to the luminal epithelium. In primary culture, SGK1 was induced and phosphorylated upon decidualization of endometrial stromal cells in response to 8-bromo-cAMP and progestin treatment. Moreover, overexpression of SGK1 in decidualizing cells enhanced phosphorylation and cytoplasmic translocation of the forkhead transcription factor FOXO1 and inhibited the expression of PRL, a major decidual marker gene. Conversely, knockdown of endogenous SGK1 by small interfering RNA increased nuclear FOXO1 levels and enhanced PRL expression. The observation that SGK1 targets FOXO1 in differentiating human endometrium, together with its distinct temporal and spatial expression pattern and increased expression in infertile patients, suggest a major role for this kinase in early pregnancy events.


Subject(s)
Endometrium/enzymology , Fertility , Immediate-Early Proteins/metabolism , Infertility, Female/enzymology , Protein Serine-Threonine Kinases/metabolism , Adult , Cells, Cultured , Decidua/metabolism , Endometrium/metabolism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Humans , Menstrual Cycle , Microarray Analysis , Phosphorylation , Prolactin/metabolism , Stromal Cells/metabolism
16.
Gynecol Oncol ; 100(2): 397-404, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16271749

ABSTRACT

OBJECTIVE: To study alterations within the p53 pathway in relation to the development of recurrent stage I endometrioid endometrial carcinoma. METHODS: Paraffin-embedded tumor tissue of both primary and recurrent tumors from 44 patients with and 44 without recurrence was used for immunohistochemical analysis of TP53, hMdm2, P21(Waf1/Cip1) and M30. DNA was extracted, and mutation analysis of p53 (exon 5-8, 11) was performed by direct sequencing. RESULTS: TP53 overexpression was significantly associated with recurrent disease: Odds Ratio 3.8 (95% CI: 1.5-9.8). Overexpression of TP53 was associated with lower staining indices (SI:0-9) of both hMdm2 and P21 in tumors of patients with recurrence, compared to controls: 2.0 +/- 0.4 vs. 4.0 +/- 0.8 and 1.9 +/- 0.8 vs. 3.6 +/- 0.8, respectively. Eight p53 missense mutations were identified in six patients with recurrence and two controls. One nonsense mutation was found in a patient with recurrence and one deletion in a control patient. Only a minority of TP53 overexpression cases could be explained by the presence of these p53 mutations. CONCLUSION: TP53 overexpression was significantly predictive for recurrent endometrial carcinoma, and mostly not correlated with p53 mutations. Concomitant low hMdm2 and P21(Waf1/Cip1) expression in tumors with overexpressed TP53 suggests a dysfunctional TP53-P21(Waf1/Cip1) pathway.


Subject(s)
Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Tumor Suppressor Protein p53/biosynthesis , Aged , Aged, 80 and over , Apoptosis/genetics , Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Keratins/biosynthesis , Keratins/genetics , Middle Aged , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Paraffin Embedding , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics
18.
Cell Tissue Res ; 322(2): 299-311, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16082522

ABSTRACT

We have studied menstrual effluent in order to identify soluble menstrual factors that induce epithelial to mesenchymal transitions (EMT) in mesothelial cells. A variety of molecules, such as nitric oxide and its reaction products, proteases (i.e. matrix metalloproteinases, plasmin) and proteins and/or peptides (i.e. growth factors: b-fibroblast growth factor, epidermal growth factor, hepatocyte growth factor, transforming growth factor-beta; cytokines: interleukin 1 beta, tumour necrosis factor-alpha [TNF-alpha]) may be involved in this process. We have demonstrated that TNF-alpha is involved in EMT, whereas the other molecules are not. Biochemical analysis has shown that the inducing menstrual factors are heat-labile molecules, are uncharged at neutral pH, have a molecular weight between 50-70 kDa (or are bound in complexes of that size) and are eluted in the albumin fraction during gel filtration chromatography. Further analysis of this fraction by using proteomics and mass spectrometry has led to the identification of alpha-enolase and haemoglobin whose inhibition partially prevents EMT. When antibodies against TNF-alpha, alpha-enolase and haemoglobin are combined, EMT is almost completely inhibited. Thus, the candidates for soluble menstrual factors that induce mesothelial EMT are TNF-alpha, alpha-enolase and haemoglobin.


Subject(s)
Cell Differentiation/physiology , Endometrium/chemistry , Epithelial Cells/chemistry , Menstruation , Mesoderm/cytology , Cells, Cultured , Culture Media, Conditioned/chemistry , Endometrium/cytology , Epithelial Cells/cytology , Female , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Growth Substances/metabolism , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Mass Spectrometry , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Mesoderm/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Omentum/cytology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
19.
Fertil Steril ; 83(3): 793-5, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749522

ABSTRACT

A prospective study was performed to determine the effects of the angiostatic compounds anti-hVEGF antibody, TNP-470, endostatin, and anginex on the vascularization and on endometriosis-like lesion formation in the chicken chorioallantoic membrane model. Endometriosis-like lesion formation was significantly impaired after treatment with angiostatic agents, which was associated with decreased vessel densities in the surrounding chorioallantoic membrane and more necrosis in the endometriosis-like lesions.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Chorioallantoic Membrane/blood supply , Endometriosis/prevention & control , Neovascularization, Pathologic/drug therapy , Sesquiterpenes/pharmacology , Animals , Antibodies/pharmacology , Chick Embryo , Chickens , Chorioallantoic Membrane/pathology , Cyclohexanes , Endostatins/pharmacology , Female , Neovascularization, Pathologic/prevention & control , O-(Chloroacetylcarbamoyl)fumagillol , Vascular Endothelial Growth Factor A/immunology
20.
Proteomics ; 4(9): 2608-23, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15352236

ABSTRACT

Peritoneal endometriosis is the result of ectopic implantation and growth of endometrium tissue that has been regurgitated into the abdominal cavity during menstruation. We have previously shown that menstrual effluent induces epithelial to mesenchymal transitions (EMT) in mesothelial cells, which results in cell retraction and exposure of submesothelial extracellular matrix. Since endometrial tissue preferentially adheres to the extracellular matrix, adhesion of endometrial tissue to the peritoneum is facilitated. The EMT were shown to be associated with differential expression and phosphorylation of mesothelial proteins. Using radiolabeling and proteomics we detected changes in protein expression and phosphorylation that occur in mesothelial cells during the EMT process. The identity of 73 proteins, which were obtained from 324 analyzed spots, was confirmed. The expression of 35 proteins involved in organization of the cytoskeleton, signal transduction, regulation of the redox state, and production of ATP, was altered during the EMT process. Four of the identified proteins were differentially phosphorylated: annexin-1, an actin-binding protein and a substrate for receptor tyrosine kinases; tropomyosin-alpha, a regulator of actin filament stability and cell shape; elongation factor 1 delta; ATP synthase beta-chain. In conclusion, factors from menstrual effluent induce specific changes in the expression and phosphorylation status of structural, regulatory and metabolic proteins relevant to the complex process of EMT in mesothelial cells.


Subject(s)
Endometrium/physiology , Epithelial Cells/metabolism , Menstruation , Mesoderm/physiology , Proteome/analysis , Cells, Cultured , Culture Media/chemistry , Electrophoresis, Gel, Two-Dimensional , Endometriosis/pathology , Endometrium/cytology , Epithelial Cells/cytology , Female , Humans , Mesoderm/cytology , Phosphorus Radioisotopes , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...