Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Lancet Oncol ; 25(4): 474-487, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38547892

ABSTRACT

BACKGROUND: The PI3K-mTOR pathway is frequently dysregulated in breast cancer. Combining an inhibitor targeting all class I PI3K isoforms and mTOR complex 1 (mTORC1)-mTOR complex 2 (mTORC2) with endocrine therapy and a CDK4/6 inhibitor might provide more effective tumour control than standard-of-care therapy. To evaluate this hypothesis, gedatolisib, a pan-PI3K-mTOR inhibitor, was assessed in a phase 1b trial combined with palbociclib and endocrine therapy in patients with hormone receptor-positive, HER2-negative, advanced breast cancer. Results from the dose expansion portion of this trial are reported herein. METHODS: This multicentre, open-label, phase 1b study recruited female patients aged at least 18 years from 17 sites across the USA with hormone-receptor-positive, HER2-negative, advanced breast cancer and an Eastern Cooperative Oncology Group performance status of 0-1. Four patient groups were studied in the dose expansion portion of the study: treatment-naive in the advanced setting (first line; group A), progression on 1-2 lines of endocrine therapy but CDK4/6 inhibitor-naive (group B); and one or more previous lines (second-line and higher) of therapy, including a CDK4/6 inhibitor (groups C and D). Gedatolisib 180 mg was administered intravenously weekly in 28-day treatment cycles for groups A-C, and on days 1, 8, and 15 for group D. Letrozole (group A), fulvestrant (groups B-D), and palbociclib (all groups) were administered at standard doses and schedules. The primary endpoint was investigator-assessed objective response rate per RECIST version 1.1 in the evaluable analysis set. This trial is completed and registered with ClinicalTrials.gov, NCT02684032. FINDINGS: Between Dec 19, 2017, and June 19, 2019, 103 female participants were enrolled in the dose expansion groups A (n=31), B (n=13), C (n=32), and D (n=27). Median follow-up was 16·6 months (IQR 5·7-48·4) for group A, 11·0 months (7·6-16·9) for group B, 3·6 months (1·8-7·5) for group C, and 9·4 months (5·3-16·7) for group D for the primary endpoint. Gedatolisib, palbociclib, and endocrine therapy induced an objective response in 23 (85·2%; 90% CI 69·2-94·8) of 27 evaluable first-line participants (group A). In the second-line and higher setting, an objective response was observed in eight (61·5%; 90% CI 35·5-83·4) of 13 evaluable group B participants, seven (25·0%; 12·4-41·9) of 28 evaluable group C participants, and 15 (55·6%; 38·2-72·0) of 27 evaluable group D participants; this included participants with both wild-type and mutated PIK3CA tumours. The most common grade 3-4 treatment-related adverse events were neutropenia (65 [63%] of 103), stomatitis (28 [27%]), and rash (21 [20%]). Grade 3-4 hyperglycaemia was reported in six (6%) participants. 23 (22%) of 103 participants had a treatment-related serious adverse event, and there were no treatment-related deaths. Nine (9%) participants discontinued treatment because of a treatment-emergent adverse event. INTERPRETATION: Gedatolisib plus palbociclib and endocrine therapy showed a promising objective response rate compared with the published results for standard-of-care therapies and had an acceptable safety profile. FUNDING: Pfizer and Celcuity.


Subject(s)
Breast Neoplasms , Morpholines , Piperazines , Pyridines , Triazines , Female , Humans , Adolescent , Adult , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Disease-Free Survival , Antineoplastic Combined Chemotherapy Protocols/adverse effects , TOR Serine-Threonine Kinases
3.
Br J Cancer ; 128(1): 30-41, 2023 01.
Article in English | MEDLINE | ID: mdl-36335217

ABSTRACT

BACKGROUND: This Phase 1b study (B2151002) evaluated the PI3K/mTOR inhibitor gedatolisib (PF-05212384) in combination with other anti-tumour agents in advanced solid tumours. METHODS: Patients with various malignancies were administered gedatolisib (90‒310 mg intravenously every week [QW]) plus docetaxel (arm A) or cisplatin (arm B) (each 75 mg/m2 intravenously Q3W) or dacomitinib (30 or 45 mg/day orally). The safety and tolerability of combination therapies were assessed during dose escalation; objective response (OR) and safety were assessed during dose expansion. RESULTS: Of 110 patients enrolled, 107 received gedatolisib combination treatment. Seven of 70 (10.0%) evaluable patients had dose-limiting toxicities; the most common was grade 3 oral mucositis (n = 3). Based upon reprioritisation of the sponsor's portfolio, dose expansion focused on arm B, gedatolisib (180 mg QW) plus cisplatin in patients (N = 22) with triple-negative breast cancer (TNBC). OR (95% CI) was achieved in four of ten patients in first-line (overall response rate 40.0% [12.2-73.8%]) and four of 12 in second/third-line (33.3% [9.9-65.1%]) settings. One patient in each TNBC arm (10%, first-line; 8.3%, second/third-line) achieved a complete response. CONCLUSIONS: Gedatolisib combination therapy showed an acceptable tolerability profile, with clinical activity at the recommended Phase 2 dose in patients with TNBC. CLINICAL TRIAL: ClinicalTrial.gov: NCT01920061.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Cisplatin/adverse effects , Triazines , Morpholines/therapeutic use , Antineoplastic Agents/adverse effects , Phosphoinositide-3 Kinase Inhibitors , Antineoplastic Combined Chemotherapy Protocols/adverse effects
4.
BMC Pulm Med ; 14: 3, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24405692

ABSTRACT

BACKGROUND: S-nitrosoglutathione (GSNO) serves as a reservoir for nitric oxide (NO) and thus is a key homeostatic regulator of airway smooth muscle tone and inflammation. Decreased levels of GSNO in the lungs of asthmatics have been attributed to increased GSNO catabolism via GSNO reductase (GSNOR) leading to loss of GSNO- and NO- mediated bronchodilatory and anti-inflammatory actions. GSNOR inhibition with the novel small molecule, N6022, was explored as a therapeutic approach in an experimental model of asthma. METHODS: Female BALB/c mice were sensitized and subsequently challenged with ovalbumin (OVA). Efficacy was determined by measuring both airway hyper-responsiveness (AHR) upon methacholine (MCh) challenge using whole body plethysmography and pulmonary eosinophilia by quantifying the numbers of these cells in the bronchoalveolar lavage fluid (BALF). Several other potential biomarkers of GSNOR inhibition were measured including levels of nitrite, cyclic guanosine monophosphate (cGMP), and inflammatory cytokines, as well as DNA binding activity of nuclear factor kappa B (NFκB). The dose response, onset of action, and duration of action of a single intravenous dose of N6022 given from 30 min to 48 h prior to MCh challenge were determined and compared to effects in mice not sensitized to OVA. The direct effect of N6022 on airway smooth muscle tone also was assessed in isolated rat tracheal rings. RESULTS: N6022 attenuated AHR (ED50 of 0.015 ± 0.002 mg/kg; Mean ± SEM) and eosinophilia. Effects were observed from 30 min to 48 h after treatment and were comparable to those achieved with three inhaled doses of ipratropium plus albuterol used as the positive control. N6022 increased BALF nitrite and plasma cGMP, while restoring BALF and plasma inflammatory markers toward baseline values. N6022 treatment also attenuated the OVA-induced increase in NFκB activation. In rat tracheal rings, N6022 decreased contractile responses to MCh. CONCLUSIONS: The significant bronchodilatory and anti-inflammatory actions of N6022 in the airways are consistent with restoration of GSNO levels through GSNOR inhibition. GSNOR inhibition may offer a therapeutic approach for the treatment of asthma and other inflammatory lung diseases. N6022 is currently being evaluated in clinical trials for the treatment of inflammatory lung disease.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Asthma/drug therapy , Benzamides/pharmacology , Benzamides/therapeutic use , Bronchoconstriction/drug effects , Inflammation/prevention & control , Pyrroles/pharmacology , Pyrroles/therapeutic use , Animals , Asthma/immunology , Asthma/physiopathology , Female , Mice , Mice, Inbred BALB C
5.
J Appl Physiol (1985) ; 114(6): 752-60, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23349456

ABSTRACT

Nitric oxide (NO) exerts a wide range of cellular effects in the cardiovascular system. NO is short lived, but S-nitrosoglutathione (GSNO) functions as a stable intracellular bioavailable NO pool. Accordingly, increased levels can facilitate NO-mediated processes, and conversely, catabolism of GSNO by the regulatory enzyme GSNO reductase (GSNOR) can impair these processes. Because dysregulated GSNOR can interfere with processes relevant to cardiovascular health, it follows that inhibition of GSNOR may be beneficial. However, the effect of GSNOR inhibition on vascular activity is unknown. To study the effects of GSNOR inhibition on endothelial function, we treated rats with a small-molecule inhibitor of GSNOR (N6338) that has vasodilatory effects on isolated aortic rings and assessed effects on arterial flow-mediated dilation (FMD), an NO-dependent process. GSNOR inhibition with a single intravenous dose of N6338 preserved FMD (15.3 ± 5.4 vs. 14.2 ± 6.3%, P = nonsignificant) under partial NO synthase inhibition that normally reduces FMD by roughly 50% (14.1 ± 2.9 vs. 7.6 ± 4.4%, P < 0.05). In hypertensive rats, daily oral administration of N6338 for 14 days reduced blood pressure (170.0 ± 5.3/122.7 ± 6.4 vs. 203.8 ± 1.9/143.7 ± 7.5 mmHg for vehicle, P < 0.001) and vascular resistance index (1.5 ± 0.4 vs. 3.2 ± 1.0 mmHg · min · l(-1) for vehicle, P < 0.001), and restored FMD from an initially impaired state (7.4 ± 1.7%, day 0) to a level (13.0 ± 3.1%, day 14, P < 0.001) similar to that observed in normotensive rats. N6338 also reversed the pathological kidney changes exhibited by the hypertensive rats. GSNOR inhibition preserves FMD under conditions of impaired NO production and protects against both microvascular and conduit artery dysfunction in a model of hypertension.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Antihypertensive Agents/pharmacology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Femoral Artery/drug effects , Hypertension/drug therapy , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Administration, Oral , Aldehyde Oxidoreductases/metabolism , Animals , Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/administration & dosage , Femoral Artery/enzymology , Femoral Artery/physiopathology , Humans , Hypertension/enzymology , Hypertension/etiology , Hypertension/pathology , Hypertension/physiopathology , Injections, Intravenous , Kidney/drug effects , Kidney/pathology , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , Sodium Chloride, Dietary , Time Factors , Vasodilator Agents/administration & dosage
6.
Bioorg Med Chem Lett ; 22(6): 2338-42, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22342142

ABSTRACT

The enzyme S-nitrosoglutathione reductase (GSNOR) is a member of the alcohol dehydrogenase family (ADH) that regulates the levels of S-nitrosothiols (SNOs) through catabolism of S-nitrosoglutathione (GSNO). GSNO and SNOs are implicated in the pathogenesis of many diseases including those in respiratory, gastrointestinal, and cardiovascular systems. The pyrrole based N6022 was recently identified as a potent, selective, reversible, and efficacious GSNOR inhibitor which is currently in clinical development for acute asthma. We describe here the synthesis and structure-activity relationships (SAR) of novel pyrrole based analogs of N6022 focusing on carboxamide modifications on the pendant N-phenyl moiety. We have identified potent and novel GSNOR inhibitors that demonstrate efficacy in an ovalbumin (OVA) induced asthma model in mice.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Anti-Asthmatic Agents/chemical synthesis , Asthma/drug therapy , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Pyrroles/chemical synthesis , Acute Disease , Aldehyde Oxidoreductases/metabolism , Animals , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/therapeutic use , Asthma/chemically induced , Asthma/enzymology , Benzamides/administration & dosage , Benzamides/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Humans , Male , Mice , Ovalbumin , Pyrroles/administration & dosage , Pyrroles/therapeutic use , S-Nitrosoglutathione/metabolism , S-Nitrosothiols/metabolism , Structure-Activity Relationship
7.
PLoS One ; 7(12): e52995, 2012.
Article in English | MEDLINE | ID: mdl-23285246

ABSTRACT

Endogenous S-nitrosothiols, including S-nitrosoglutathione (GSNO), mediate nitric oxide (NO)-based signaling, inflammatory responses, and smooth muscle function. Reduced GSNO levels have been implicated in several respiratory diseases, and inhibition of GSNO reductase, (GSNOR) the primary enzyme that metabolizes GSNO, represents a novel approach to treating inflammatory lung diseases. Recently, an association between decreased GSNOR expression and human lung cancer risk was proposed in part based on immunohistochemical staining using a polyclonal GSNOR antibody. GSNOR is an isozyme of the alcohol dehydrogenase (ADH) family, and we demonstrate that the antibody used in those studies cross reacts substantially with other ADH proteins and may not be an appropriate reagent. We evaluated human lung cancer tissue arrays using monoclonal antibodies highly specific for human GSNOR with minimal cross reactivity to other ADH proteins. We verified the presence of GSNOR in ≥85% of specimens examined, and extensive analysis of these samples demonstrated no difference in GSNOR protein expression between cancerous and normal lung tissues. Additionally, GSNOR and other ADH mRNA levels were evaluated quantitatively in lung cancer cDNA arrays by qPCR. Consistent with our immunohistochemical findings, GSNOR mRNA levels were not changed in lung cancer tissues, however the expression levels of other ADH genes were decreased. ADH IB mRNA levels were reduced (>10-fold) in 65% of the lung cancer cDNA specimens. We conclude that the previously reported results showed an incorrect association of GSNOR and human lung cancer risk, and a decrease in ADH IB, rather than GSNOR, correlates with human lung cancer.


Subject(s)
Alcohol Dehydrogenase/genetics , Lung Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Alcohol Dehydrogenase/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Antibodies, Monoclonal/metabolism , Case-Control Studies , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , RNA, Messenger/analysis , S-Nitrosoglutathione/metabolism
8.
Bioorg Med Chem Lett ; 21(19): 5849-53, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21855338

ABSTRACT

The pyrrole based N6022 was recently identified as a potent, selective, reversible, and efficacious S-nitrosoglutathione reductase (GSNOR) inhibitor and is currently undergoing clinical development for the treatment of acute asthma. GSNOR is a member of the alcohol dehydrogenase family (ADH) and regulates the levels of S-nitrosothiols (SNOs) through catabolism of S-nitrosoglutathione (GSNO). Reduced levels of GSNO, as well as other nitrosothiols (SNOs), have been implicated in the pathogenesis of many diseases including those of the respiratory, cardiovascular, and gastrointestinal systems. Preservation of endogenous SNOs through GSNOR inhibition presents a novel therapeutic approach with broad applicability. We describe here the synthesis and structure-activity relationships (SAR) of novel pyrrole based analogues of N6022 focusing on removal of cytochrome P450 inhibition activities. We identified potent and novel GSNOR inhibitors having reduced CYP inhibition activities and demonstrated efficacy in a mouse ovalbumin (OVA) model of asthma.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Benzamides/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Pyrroles/pharmacology , Animals , Asthma/drug therapy , Asthma/enzymology , Benzamides/chemistry , Benzamides/toxicity , Cytochrome P-450 Enzyme Inhibitors , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Humans , Imidazoles/pharmacokinetics , Imidazoles/toxicity , Lung/pathology , Lung/physiopathology , Mice , Molecular Structure , Molecular Targeted Therapy , No-Observed-Adverse-Effect Level , Pyrroles/chemistry , Pyrroles/toxicity , Receptors, Opioid, delta/metabolism , S-Nitrosoglutathione/metabolism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 21(12): 3671-5, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21570838

ABSTRACT

S-Nitrosoglutathione reductase (GSNOR) is a member of the alcohol dehydrogenase family (ADH) that regulates the levels of S-nitrosothiols (SNOs) through catabolism of S-nitrosoglutathione (GSNO). GSNO and SNOs are implicated in the pathogenesis of many diseases including those in respiratory, cardiovascular, and gastrointestinal systems. The pyrrole based N6022 was recently identified as a potent, selective, reversible, and efficacious GSNOR inhibitor which is currently undergoing clinical development. We describe here the synthesis and structure-activity relationships (SAR) of novel pyrrole based analogues of N6022 focusing on scaffold modification and propionic acid replacement. We identified equally potent and novel GSNOR inhibitors having pyrrole regioisomers as scaffolds using a structure based approach.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Benzamides/chemistry , Benzamides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Propionates/chemistry , Propionates/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Benzamides/chemical synthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Inhibitory Concentration 50 , Molecular Structure , Propionates/chemical synthesis , Pyrroles/chemical synthesis , Stereoisomerism , Structure-Activity Relationship
10.
ACS Med Chem Lett ; 2(5): 402-6, 2011 May 12.
Article in English | MEDLINE | ID: mdl-24900320

ABSTRACT

S-Nitrosoglutathione reductase (GSNOR) regulates S-nitrosothiols (SNOs) and nitric oxide (NO) in vivo through catabolism of S-nitrosoglutathione (GSNO). GSNOR and the anti-inflammatory and smooth muscle relaxant activities of SNOs, GSNO, and NO play significant roles in pulmonary, cardiovascular, and gastrointestinal function. In GSNOR knockout mice, basal airway tone is reduced and the response to challenge with bronchoconstrictors or airway allergens is attenuated. Consequently, GSNOR has emerged as an attractive therapeutic target for several clinically important human diseases. As such, small molecule inhibitors of GSNOR were developed. These GSNOR inhibitors were potent, selective, and efficacious in animal models of inflammatory disease characterized by reduced levels of GSNO and bioavailable NO. N6022, a potent and reversible GSNOR inhibitor, reduced bronchoconstriction and pulmonary inflammation in a mouse model of asthma and demonstrated an acceptable safety profile. N6022 is currently in clinical development as a potential agent for the treatment of acute asthma.

11.
Cancer Res ; 69(2): 510-7, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147564

ABSTRACT

The export protein CRM1 is required for the nuclear export of a wide variety of cancer-related "cargo" proteins including p53, c-Abl, and FOXO-3A. Leptomycin B (LMB) is a highly specific inhibitor of CRM1 with significant in vitro potency but limited in vivo efficacy due to toxicity. We now report a series of semisynthetic LMB derivatives showing substantially improved therapeutic windows. Exposure of cancer cells to these compounds leads to a rapid and prolonged block of nuclear export and apoptosis. In contrast to what is observed in cancer cells, these agents induce cell cycle arrest, but not apoptosis, in normal lung fibroblasts. These new nuclear export inhibitors (NEI) maintain the high potency of LMB, are up to 16-fold better tolerated than LMB in vivo, and show significant efficacy in multiple mouse xenograft models. These NEIs show the potential of CRM1 inhibitors as novel and potent anticancer agents.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/drug effects , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , HCT116 Cells , Humans , Lung/cytology , Lung/drug effects , Mice , Mice, Inbred C57BL , Mice, Nude , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Tumor Suppressor Protein p53/biosynthesis , Xenograft Model Antitumor Assays , Exportin 1 Protein
12.
Biochemistry ; 45(4): 1321-30, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16430229

ABSTRACT

The epothilones are a family of polyketide natural products that show a high potential as anticancer drugs. They are synthesized by the action of a hybrid nonribosomal peptide synthetase/polyketide synthase in the myxobacterium Sorangium cellulosum. In this work, the genes encoding the entire cluster,epoA, epoB, epoC, epoD, epoE, and epoF, were redesigned and synthesized to allow for expression in Escherichia coli. The expression of the largest of the proteins, EpoD, also required the protein be separated into two polypeptides with compatible module linkers. Using a combination of lowered temperature, chaperone coexpression, and alternative promoters, we succeeded in producing a soluble protein from all genes in the epothilone cluster. The entire synthetic epothilone cluster was then expressed in a strain of E. coli modified to enable polyketide biosynthesis, resulting in the production of epothilones C and D. Furthermore, feeding a thioester of the normal substrate for EpoD to cells expressing the epoD, epoE, and epoF genes also led to the production of epothilones C and D. The design of the synthetic epothilone genes together with E. coli expression provides the ideal platform for both the biochemical investigation of the epothilone PKS and the generation of novel biosynthetic epothilone analogues.


Subject(s)
Epothilones/biosynthesis , Escherichia coli/metabolism , Catalysis , Cysteamine/analogs & derivatives , Cysteamine/metabolism , Epothilones/metabolism , Escherichia coli/genetics , Methacrylates/metabolism , Peptide Chain Elongation, Translational/genetics , Peptide Chain Elongation, Translational/physiology , Peptide Synthases/genetics , Peptide Synthases/metabolism , Polyketide Synthases/biosynthesis , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Thiazoles/metabolism
13.
FEMS Yeast Res ; 6(1): 40-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16423069

ABSTRACT

Polyketides are a diverse group of natural products with significance in human and veterinary medicine. Because polyketides are structurally complex molecules and fermentation is the most commercially viable route of production, a generic heterologous host system for high-level polyketide production is desirable. Saccharomyces cerevisiae has been shown to be an excellent production host for a simple polyketide, yielding 1.7 g of 6-methylsalicylic acid per liter of culture in un-optimized shake-flask fermentations. However, a barrier to the heterologous production of more complex 'modular' polyketides in S. cerevisiae is the lack of required polyketide precursor pathways. In this work, we describe the introduction into S. cerevisiae of pathways for the production of methylmalonyl-coenzyme A (CoA), a precursor for complex polyketides, by both propionyl-CoA-dependent and propionyl-CoA-independent routes. Furthermore, we demonstrate that the methylmalonyl-CoA produced in the engineered yeast strains is used in vivo for the production of a polyketide product, a triketide lactone.


Subject(s)
Genetic Engineering/methods , Macrolides/metabolism , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Saccharomyces cerevisiae/genetics , Acyl Coenzyme A/metabolism , Gene Expression Regulation, Fungal , Macrolides/chemistry , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Saccharomyces cerevisiae/enzymology , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Streptomyces coelicolor/enzymology , Streptomyces coelicolor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...