Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Rep Med ; 5(7): 101619, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38897206

ABSTRACT

Liver-directed adeno-associated viral (AAV) vector-mediated homology-independent targeted integration (AAV-HITI) by CRISPR-Cas9 at the highly transcribed albumin locus is under investigation to provide sustained transgene expression following neonatal treatment. We show that targeting the 3' end of the albumin locus results in productive integration in about 15% of mouse hepatocytes achieving therapeutic levels of systemic proteins in two mouse models of inherited diseases. We demonstrate that full-length HITI donor DNA is preferentially integrated upon nuclease cleavage and that, despite partial AAV genome integrations in the target locus, no gross chromosomal rearrangements or insertions/deletions at off-target sites are found. In line with this, no evidence of hepatocellular carcinoma is observed within the 1-year follow-up. Finally, AAV-HITI is effective at vector doses considered safe if directly translated to humans providing therapeutic efficacy in the adult liver in addition to newborn. Overall, our data support the development of this liver-directed AAV-based knockin strategy.


Subject(s)
Dependovirus , Disease Models, Animal , Genetic Vectors , Liver , Animals , Dependovirus/genetics , Liver/metabolism , Liver/pathology , Mice , Genetic Vectors/genetics , Hepatocytes/metabolism , Humans , Virus Integration/genetics , CRISPR-Cas Systems/genetics , Transgenes , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Mice, Inbred C57BL , Albumins/genetics , Albumins/metabolism
2.
Mol Ther Methods Clin Dev ; 28: 396-411, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36910588

ABSTRACT

Gene therapy of Usher syndrome type 1B (USH1B) due to mutations in the large Myosin VIIA (MYO7A) gene is limited by the packaging capacity of adeno-associated viral (AAV) vectors. To overcome this, we have previously developed dual AAV8 vectors which encode human MYO7A (dual AAV8.MYO7A). Here we show that subretinal administration of 1.37E+9 to 1.37E+10 genome copies of a good-manufacturing-practice-like lot of dual AAV8.MYO7A improves the retinal defects of a mouse model of USH1B. The same lot was used in non-human primates at doses 1.6× and 4.3× the highest dose proposed for the clinical trial which was based on mouse efficacy data. Long-lasting alterations in retinal function and morphology were observed following subretinal administration of dual AAV8.MYO7A at the high dose. These findings were modest and improved over time in the low-dose group, as also observed in other studies involving the use of AAV8 in non-human primates and humans. Biodistribution and shedding studies confirmed the presence of vector DNA mainly in the visual pathway. Accordingly, we detected human MYO7A mRNA expression predominantly in the retina. Overall, these studies pave the way for the clinical translation of subretinal administration of dual AAV vectors in USH1B subjects.

3.
Nat Commun ; 13(1): 1963, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35414130

ABSTRACT

Challenges to the widespread application of gene therapy with adeno-associated viral (AAV) vectors include dominant conditions due to gain-of-function mutations which require allele-specific knockout, as well as long-term transgene expression from proliferating tissues, which is hampered by AAV DNA episomal status. To overcome these challenges, we used CRISPR/Cas9-mediated homology-independent targeted integration (HITI) in retina and liver as paradigmatic target tissues. We show that AAV-HITI targets photoreceptors of both mouse and pig retina, and this results in significant improvements to retinal morphology and function in mice with autosomal dominant retinitis pigmentosa. In addition, we show that neonatal systemic AAV-HITI delivery achieves stable liver transgene expression and phenotypic improvement in a mouse model of a severe lysosomal storage disease. We also show that HITI applications predominantly result in on-target editing. These results lay the groundwork for the application of AAV-HITI for the treatment of diseases affecting various organs.


Subject(s)
Dependovirus , Gene Editing , Animals , CRISPR-Cas Systems , Dependovirus/genetics , Gene Editing/methods , Genetic Vectors/genetics , Liver , Mice , Retina/metabolism , Swine
4.
NEJM Evid ; 1(7): EVIDoa2200052, 2022 07.
Article in English | MEDLINE | ID: mdl-38319253

ABSTRACT

BACKGROUND: Mucopolysaccharidosis type VI (MPS VI) is an inherited multisystem lysosomal disorder due to arylsulfatase B (ARSB) deficiency that leads to widespread accumulation of glycosaminoglycans (GAG), which are excreted in increased amounts in urine. MPS VI is characterized by progressive dysostosis multiplex, connective tissue and cardiac involvement, and hepatosplenomegaly. Enzyme replacement therapy (ERT) is available but requires life-long and costly intravenous infusions; moreover, it has limited efficacy on diseased skeleton and cardiac valves, compromised pulmonary function, and corneal opacities. METHODS: We enrolled nine patients with MPS VI 4 years of age or older in a phase 1/2 open-label gene therapy study. After ERT was interrupted, patients each received a single intravenous infusion of an adeno-associated viral vector serotype 8 expressing ARSB. Participants were sequentially enrolled in one of three dose cohorts: low (three patients), intermediate (two patients), or high (four patients). The primary outcome was safety; biochemical and clinical end points were secondary outcomes. RESULTS: The infusions occurred without severe adverse events attributable to the vector, meeting the prespecified end point. Participants in the low and intermediate dose cohorts displayed stable serum ARSB of approximately 20% of the mean healthy value but returned to ERT by 14 months after gene therapy because of increased urinary GAG. Participants in the high-dose cohort had sustained serum ARSB of 30% to 100% of the mean healthy value and a modest urinary GAG increase that did not reach a concentration at which ERT reintroduction was needed. In the high-dose group, there was no clinical deterioration for up to 2 years after gene therapy. CONCLUSIONS: Liver-directed gene therapy for participants with MPS VI did not have a dose-limiting side-effect and adverse event profile; high-dose treatment resulted in ARSB expression over at least 24 months with preliminary evidence of disease stabilization. (Funded by the Telethon Foundation ETS, the European Commission Seventh Framework Programme, and the Isaac Foundation; ClinicalTrials.gov number, NCT03173521; EudraCT number, 2016-002328-10.)


Subject(s)
Dependovirus , Genetic Therapy , Mucopolysaccharidosis VI , N-Acetylgalactosamine-4-Sulfatase , Humans , Genetic Therapy/methods , Mucopolysaccharidosis VI/therapy , Mucopolysaccharidosis VI/genetics , Mucopolysaccharidosis VI/urine , Male , Female , Dependovirus/genetics , Child , Adolescent , Child, Preschool , N-Acetylgalactosamine-4-Sulfatase/genetics , Adult , Young Adult , Genetic Vectors/administration & dosage , Liver/metabolism , Liver/pathology , Enzyme Replacement Therapy/methods
5.
Mol Ther Methods Clin Dev ; 20: 247-257, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33473358

ABSTRACT

Adeno-associated viral (AAV) vectors have emerged as the preferred platform for in vivo gene transfer because of their combined efficacy and safety. However, insertional mutagenesis with the subsequent development of hepatocellular carcinomas (HCCs) has been recurrently noted in newborn mice treated with high doses of AAV, and more recently, the association of wild-type AAV integrations in a subset of human HCCs has been documented. Here, we address, in a comprehensive, prospective study, the long-term risk of tumorigenicity in young adult mice following delivery of single-stranded AAVs targeting liver. HCC incidence in mice treated with therapeutic and reporter AAVs was low, in contrast to what has been previously documented in mice treated as newborns with higher doses of AAV. Specifically, HCCs developed in 6 out 76 of AAV-treated mice, and a pathogenic integration of AAV was found in only one tumor. Also, no evidence of liver tumorigenesis was found in juvenile AAV-treated mucopolysaccharidosis type VI (MPS VI) cats followed as long as 8 years after vector administration. Together, our results support the low risk of tumorigenesis associated with AAV-mediated gene transfer targeting juvenile/young adult livers, although constant monitoring of subjects enrolled in AAV clinical trial is advisable.

6.
Hum Gene Ther ; 30(10): 1204-1210, 2019 10.
Article in English | MEDLINE | ID: mdl-31517544

ABSTRACT

Inborn errors of metabolism (IEM) are disorders affecting human biochemical pathways and represent attractive targets for gene therapy because of their severity, high overall prevalence, lack of effective treatments, and possibility of early diagnosis through newborn screening. The liver is a central organ involved in several metabolic reactions and is a favorite target for gene therapy in many IEM. Adeno-associated virus (AAV) vectors have emerged in the last years as the preferred vectors for in vivo gene delivery. Gene replacement strategies are aimed either at correcting liver disease or providing a source for production and secretion of the lacking enzyme for cross-correction of other tissues. A number of preclinical studies have been conducted in the last years and, for several diseases, gene therapy has reached the clinical stage, with a growing number of ongoing clinical trials. Moreover, recent applications of genome editing to the field of inherited metabolic diseases have further expanded potential therapeutic possibilities. This review describes relevant clinical gene therapy studies for IEM with particular attention to current obstacles and drawbacks.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Liver Diseases/therapy , Metabolism, Inborn Errors/therapy , Animals , Clinical Trials as Topic , Dependovirus/genetics , Dependovirus/metabolism , Gene Transfer Techniques , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lentivirus/genetics , Lentivirus/metabolism , Liver/metabolism , Liver/pathology , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/pathology , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology
7.
Mol Ther Methods Clin Dev ; 6: 143-158, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28932756

ABSTRACT

In vivo gene therapy with adeno-associated viral (AAV) vectors is safe and effective in humans. We recently demonstrated that AAV8-mediated liver gene transfer is effective in animal models of mucopolysaccharidosis type VI (MPS VI), a rare lysosomal storage disease that is caused by arylsulfatase B (ARSB) deficiency. In preparing for a first-in-human trial, we performed non-clinical studies to assess the safety of intravenous administrations of AAV2/8.TBG.hARSB produced under good manufacturing practice-like conditions. No toxicity was observed in AAV-treated mice, except for a transient increase in alanine aminotransferase in females and thyroid epithelial hypertrophy. AAV2/8.TBG.hARSB biodistribution and expression confirmed the liver as the main site of both infection and transduction. Shedding and breeding studies suggest that the risk of both horizontal and germline transmission is minimal. An AAV dose-response study in MPS VI mice was performed to define the range of doses to be used in the clinical study. Overall, these data support the non-clinical safety and efficacy of AAV2/8.TBG.hARSB and pave the way for a phase I/II clinical trial based on intravascular infusions of AAV8 in patients with MPS VI.

8.
Mol Ther ; 24(12): 2054-2063, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27658524

ABSTRACT

Enzyme replacement therapy (ERT) is the standard of care for several lysosomal storage diseases (LSDs). ERT, however, requires multiple and costly administrations and has limited efficacy. We recently showed that a single high dose administration of adeno-associated viral vector serotype 8 (AAV2/8) is at least as effective as weekly ERT in a mouse model of mucopolysaccharidosis type VI (MPS VI). However, systemic administration of high doses of AAV might result in both cell-mediated immune responses and insertional mutagenesis. Here we evaluated whether the combination of low doses of AAV2/8 with a less frequent (monthly) than canonical (weekly) ERT schedule may be as effective as the single treatments at high doses or frequent regimen. A greater reduction of both urinary glycosaminoglycans, considered a sensitive biomarker of therapeutic efficacy, and storage in the myocardium and heart valves was observed in mice receiving the combined than the single therapies. Importantly, these levels of correction were similar to those we obtained in a previous study following either high doses of AAV2/8 or weekly ERT. Our data show that low-dose gene therapy can be used as a means to rarify ERT administration, thus reducing both the risks and costs associated with either therapies.


Subject(s)
Combined Modality Therapy/methods , Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Lysosomal Storage Diseases/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Glycosaminoglycans/urine , Humans , Lysosomal Storage Diseases/urine , Mice , Treatment Outcome
9.
Hum Gene Ther ; 26(3): 145-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25654180

ABSTRACT

Recombinant vectors based on adeno-associated virus serotype 8 (AAV8) have been successfully used in the clinic and hold great promise for liver-directed gene therapy. Preexisting immunity against AAV8 or the development of antibodies against the therapeutic transgene product might negatively affect the outcomes of gene therapy. In the prospect of an AAV8-mediated, liver-directed gene therapy clinical trial for mucopolysaccharidosis VI (MPS VI), a lysosomal storage disorder caused by arylsulfatase B (ARSB) deficiency, we investigated in a multiethnic cohort of MPS VI patients the prevalence of neutralizing antibodies (Nab) to AAV8 and the presence of ARSB cross-reactive immunologic material (CRIM), which will either affect the efficacy of gene transfer or the duration of phenotypic correction. Thirty-six MPS VI subjects included in the study harbored 45 (62.5%) missense, 13 (18%) nonsense, 9 (12.5%) frameshift (2 insertions and 7 deletions), and 5 (7%) splicing ARSB mutations. The detection of ARSB protein in 24 patients out of 34 (71%) was predicted by the type of mutations. Preexisting Nab to AAV8 were undetectable in 19/33 (58%) analyzed patients. Twelve out of 31 patients (39%) tested were both negative for Nab to AAV8 and CRIM-positive. In conclusion, this study allows estimating the number of MPS VI patients eligible for a gene therapy trial by intravenous injections of AAV8.


Subject(s)
Antibodies, Neutralizing/blood , Dependovirus/immunology , Genetic Therapy/methods , Mucopolysaccharidosis VI/immunology , N-Acetylgalactosamine-4-Sulfatase/blood , Patient Selection , Cohort Studies , Cross Reactions , DNA Mutational Analysis , Dependovirus/genetics , Genetic Therapy/standards , Humans , Italy , Mucopolysaccharidosis VI/therapy , Mutation/genetics , N-Acetylgalactosamine-4-Sulfatase/genetics , Netherlands , Turkey
10.
Hum Gene Ther ; 25(7): 609-18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24725025

ABSTRACT

Enzyme replacement therapy (ERT) has become the standard of care for several lysosomal storage disorders (LSDs). Despite ERT's undisputed efficacy, the requirement for multiple and costly administrations as well as ERT's limited improvement of some LSD manifestations prompts the search for better therapies. Using a mouse model of mucopolysaccharidosis VI, we compared the efficacy of a single intravascular administration of an adeno-associated viral vector targeting liver to weekly infusions of human recombinant enzyme at the same doses used in mucopolysaccharidosis VI patients. While gene therapy results in increased and stable levels of circulating enzyme up to 1 year after vector administration, ERT has typical peak-and-drop serum kinetics. Both therapies similarly reduced glycosaminoglycan levels in urine and tissues including heart valves and myocardium, with gene therapy improving skeletal skull abnormalities slightly better, although not significantly, than ERT. Both therapies seem to similarly improve animal motor performance, with gene therapy possibly associated with less animal distress. Thus, a single vector administration that converts liver into a factory organ for systemic secretion of therapeutic proteins is at least as effective as ERT in a mouse model of LSD, potentially eliminating problems with compliance and costs. Only testing in humans will prove whether this holds true in a clinical setting.


Subject(s)
Dependovirus , Genetic Therapy/methods , Genetic Vectors , Lysosomal Storage Diseases/therapy , N-Acetylgalactosamine-4-Sulfatase , Animals , Disease Models, Animal , Glycosaminoglycans/urine , Humans , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomal Storage Diseases/urine , Mice , Myocardium/metabolism , Myocardium/pathology , N-Acetylgalactosamine-4-Sulfatase/biosynthesis , N-Acetylgalactosamine-4-Sulfatase/genetics
11.
Hum Gene Ther ; 24(2): 163-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23194248

ABSTRACT

Liver gene transfer with adeno-associated viral (AAV) 2/8 vectors is being considered for therapy of systemic diseases like mucopolysaccharidosis type VI (MPS VI), a lysosomal storage disease due to deficiency of arylsulfatase B (ARSB). We have previously reported that liver gene transfer with AAV2/8 results in sustained yet variable expression of ARSB. We hypothesized that the variability we observed could be due to pre-existing immunity to wild-type AAV8. To test this, we compared the levels of AAV2/8-mediated transduction in MPS VI cats with and without pre-existing immunity to AAV8. In addition, since levels of lysosomal enzymes as low as 5% of normal are expected to be therapeutic, we evaluated the impact of pre-existing immunity on MPS VI phenotypic rescue. AAV2/8 administration to MPS VI cats without pre-existing neutralizing antibodies to AAV8 resulted in consistent and dose-dependent expression of ARSB, urinary glycosaminoglycan (GAG) reduction, and femur length amelioration. Conversely, animals with pre-existing immunity to AAV8 showed low levels of ARSB expression and limited phenotypic improvement. Our data support the use of AAV2/8-mediated gene transfer for MPS VI and other systemic diseases, and highlight that pre-existing immunity to AAV8 should be considered in determining subject eligibility for therapy.


Subject(s)
Dependovirus/immunology , Genetic Therapy/methods , Mucopolysaccharidosis VI/therapy , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cats , Dependovirus/genetics , Dose-Response Relationship, Drug , Enzyme Activation , Femur/anatomy & histology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Glycosaminoglycans/urine , Green Fluorescent Proteins/metabolism , Liver/enzymology , Mucopolysaccharidosis VI/immunology , N-Acetylgalactosamine-4-Sulfatase/blood , N-Acetylgalactosamine-4-Sulfatase/genetics , Phenotype
12.
Oncol Lett ; 4(5): 1077-1081, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23162655

ABSTRACT

Metformin, a derivative of biguanide, is a first-line therapy for type 2 diabetes mellitus. Since the drug has been shown to significantly reduce the risk of various cancers and cancer mortality in diabetic patients, it is being considered as a potential anticancer therapeutic or preventive agent. In cellular models, metformin inhibits the growth of many types of cancer cells; however, its effects on glioblastoma multi-forme (GBM) are not well characterized. Here, we analyzed the effects of metformin on the growth and migration of LN18 and LN229 GBM cells cultured under basal conditions or exposed to leptin, a cytokine that has recently been implicated in GBM development. We found that 2-16 mM metformin reduced basal and leptin-stimulated growth of GBM cells in a dose-dependent manner. Furthermore, the drug significantly inhibited the migration of GBM cells. The action of metformin was mediated through the upregulation of its main signaling molecule, the adenosine monophosphate-activated protein kinase (AMPK), as well as through the downregulation of the signal transducer and activator of transcription 3 (STAT3) and the Akt/PKB serine/threonine protein kinase. In leptin-treated cells, the drug reversed the effects of the cytokine on the AMPK and STAT3 pathways, but modulated Akt activity in a cell-dependent manner. Our results suggest that metformin or similar AMPK-targeting agents with optimized blood-brain-barrier penetrability could be developed as potential treatments of GBM and could be used in conjunction with other target drugs such as leptin receptor antagonists.

13.
BMC Cancer ; 11: 303, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21771332

ABSTRACT

BACKGROUND: Leptin is a pleiotropic hormone whose mitogenic and angiogenic activity has been implicated in the development and progression of several malignancies, including brain tumors. In human brain cancer, especially in glioblastoma multiforme (GBM), leptin and its receptor (ObR) are overexpressed relative to normal tissue. Until present, the potential of intratumoral leptin to exert proangiogenic effects on endothelial cells has not been addressed. Using in vitro models, we investigated if GBM can express leptin, if leptin can affect angiogenic and mitogenic potential of endothelial cells, and if its action can be inhibited with specific ObR antagonists. Leptin effects were compared with that induced by the best-characterized angiogenic regulator, VEGF. RESULTS: We found that GBM cell lines LN18 and LN229 express leptin mRNA and LN18 cells secrete detectable amounts of leptin protein. Both lines also expressed and secreted VEGF. The conditioned medium (CM) of LN18 and LN 229 cultures as well as 200 ng/mL pure leptin or 50 ng/mL pure VEGF stimulated proliferation of human umbilical vein endothelial cells (HUVEC) at 24 h of treatment. Mitogenic effects of CM were ~2-fold greater than that of pure growth factors. Furthermore, CM treatment of HUVEC for 24 h increased tube formation by ~5.5-fold, while leptin increased tube formation by ~ 80% and VEGF by ~60% at 8 h. The mitogenic and angiogenic effects of both CM were blocked by Aca 1, a peptide ObR antagonist, and by SU1498, which inhibits the VEGF receptor. The best anti-angiogenic and cytostatic effects of Aca1 were obtained with 10 nM and 25 nM, respectively, while for SU1498, the best growth and angiogenic inhibition was observed at 5 µM. The combination of 5 µM SU1498 and Aca1 at 25 nM (growth inhibition) or at 10 nM (reduction of tube formation) produced superior effects compared with single agent treatments. CONCLUSIONS: Our data provide the first evidence that LN18 and LN 229 human GBM cells express leptin mRNA and might produce biologically active leptin, which can stimulate tube formation and enhance proliferation of endothelial cells. Furthermore, we demonstrate for the first time that a peptide ObR antagonist inhibits proangiogenic and growth effects of leptin on endothelial cells, and that the pharmacological potential of this compound might be combined with drugs targeting the VEGF pathway.


Subject(s)
Endothelial Cells/drug effects , Glioblastoma/metabolism , Leptin/pharmacology , Neoplasm Proteins/pharmacology , Neovascularization, Pathologic/etiology , Angiogenesis Inhibitors/pharmacology , Cell Division/drug effects , Cell Line, Tumor/metabolism , Cells, Cultured/drug effects , Cinnamates/pharmacology , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Neoplastic , Glioblastoma/blood supply , Humans , In Vitro Techniques , Leptin/biosynthesis , Leptin/genetics , Morphogenesis/drug effects , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/physiopathology , Oligopeptides/pharmacology , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Leptin/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Umbilical Cord , Up-Regulation , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
14.
Eur J Cancer ; 47(10): 1578-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21353530

ABSTRACT

Triple-negative breast cancers, which represent 10-20% of all mammary tumours, are characterised by the aggressive phenotype, are often found in younger women and have been associated with poor prognosis. Obesity increases the risk for triple-negative breast cancer development. Because triple-negative breast cancer patients are unresponsive to current targeted therapies and other treatment options are only partially effective, new pharmacological modalities are urgently needed. Here we examined if the leptin (obesity hormone) receptor is a viable target for the treatment of this cancer subtype. In human triple-negative breast cancer tissues, the leptin receptor was expressed in 92% (64/69) and leptin in 86% (59/69) of cases. In a model triple-negative breast cancer cell line MDA-MB-231, the leptin receptor antagonist peptide Allo-aca inhibited leptin-induced proliferation at 50 pM concentration. In an MDA-MB-231 orthotopic mouse xenograft model, Allo-aca administered subcutaneously significantly extended the average survival time from 15.4 days (untreated controls) to 24 and 28.1 days at 0.1 and 1mg/kg/day doses, respectively. In parallel, conventional treatment with 1mg/kg/day intraperitoneal cisplatin prolonged the average survival time to 18.6 days, while administration of 20mg/kg/day oral Tamoxifen (negative control) had no significant survival effects relative to controls. In normal CD-1 mice, Allo-aca produced no systemic toxicity up to the highest studied subcutaneous bolus dose of 50mg/kg, while, as expected, it induced a modest 6-10% body weight increase. Our results indicate that leptin receptor antagonists could become attractive options for triple-negative breast cancer treatment, especially in the obese patient population.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Leptin/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Infusions, Subcutaneous , Leptin/metabolism , Mice , Neoplasm Transplantation , Obesity/metabolism , Peptides/chemistry , Receptor, ErbB-2/chemistry , Tamoxifen/pharmacology , Time Factors , Treatment Outcome
15.
Endocr Relat Cancer ; 17(3): 823-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20603394

ABSTRACT

The incidence of colon cancer has increased in developed countries, possibly due to sedentary lifestyle and high caloric diet. Experimental and epidemiological evidence suggests a link between colon cancer development and adipose tissue-derived circulating hormones. Leptin, a pluripotent cytokine secreted by adipocytes, is a key regulator of appetite and energy balance acting in the brain. On the other hand, leptin also controls many physiological and pathological processes in peripheral organs. Recent studies in colon cancer cell lines and human tumors suggested that leptin and its receptor (ObR) are implicated in colon carcinogenesis, and may serve as new biomarkers and pharmacological targets. Here, we explored, for the first time, whether leptin can affect the biology of colorectal tumor stem cells (CTSCs). We found that our previously established and characterized CTSC clones express ObR and respond to leptin with cell proliferation, activation of the extracellular signal-related kinase (ERK)1/2 and AKT signaling pathways, enhanced growth in soft agar, and improved sphere formation associated with E-cadherin overexpression. Moreover, leptin counteracted cytotoxic effects of 5-fluorouracil, a common colon cancer therapeutic agent. These results suggest that obesity and increased leptin levels might promote colorectal cancer by increasing growth and survival of CTSCs.


Subject(s)
Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Fluorouracil/pharmacology , Leptin/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Cell Adhesion , Cell Movement , Colony-Forming Units Assay , Colorectal Neoplasms/drug therapy , Humans , Neoplastic Stem Cells/drug effects , Obesity , Receptors, Leptin/metabolism , Signal Transduction
16.
J Cell Physiol ; 224(1): 242-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20232316

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate the expression of different genes, including genes involved in cancer progression. A functional link between hypoxia, a key feature of the tumor microenvironment, and miRNA expression has been documented. We investigated whether and how miR-20b can regulate the expression of vascular endothelial growth factor (VEGF) in MCF-7 breast cancer cells under normoxic and hypoxia-mimicking conditions (CoCl(2) exposure). Using immunoblotting, ELISA, and quantitative real-time PCR, we demonstrated that miR-20b decreased VEGF protein levels at 4 and 24 h following CoCl(2) treatment, and VEGF mRNA at 4 h of treatment. In addition, miR-20b reduced VEGF protein expression in untreated cells. Next, we investigated the molecular mechanism by which pre-miR-20b can affect VEGF transcription, focusing on hypoxia inducible factor 1 (HIF-1) and signal transducer and activator of transcription 3 (STAT3), transcriptional inducers of VEGF and putative targets of miR-20b. Downregulation of VEGF mRNA by miR-20b under a 4 h of CoCl(2) treatment was associated with reduced levels of nuclear HIF-1 alpha subunit and STAT3. Chromatin immunoprecipitation (ChIP) assays revealed that HIF-1 alpha, but not STAT3, was recruited to the VEGF promoter following the 4 h of CoCl(2) treatment. This effect was inhibited by transfection of cells with pre-miR-20b. In addition, using siRNA knockdown, we demonstrated that the presence of STAT3 is necessary for CoCl(2)-mediated HIF-1 alpha nuclear accumulation and recruitment on VEGF promoter. In summary, this report demonstrates, for the first time, that the VEGF expression in breast cancer cells is mediated by HIF-1 and STAT3 in a miR-20b-dependent manner.


Subject(s)
Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cobalt/pharmacology , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promoter Regions, Genetic , RNA Interference , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Transfection , Vascular Endothelial Growth Factor A/genetics
17.
Brain Pathol ; 20(2): 481-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19775291

ABSTRACT

Although leptin and its receptor (ObR) have emerged as important cancer biomarkers, the role of the leptin system in brain tumor development remains unknown. We screened 87 human brain tumor biopsies using immunohistochemistry and detected leptin and ObR in 55.2% and 60.9% cases, respectively. In contrast, leptin and ObR were absent in 14 samples of normal brain tissue. The presence of leptin correlated with ObR with overall concordance 80.5%. The leptin/ObR system was highly expressed in glioblastomas and anaplastic astrocytomas, while lower expression of both markers was noted in low-grade astrocytomas and gangliogliomas. The association between leptin/ObR and the degree of tumor malignancy was highly significant (P < 0.001). Using double immunofluorescence of glioblastoma tissues, we found co-expression of leptin with ObR and with the proliferation marker Ki-67 in 87% and 64% of cells, respectively. The leptin/ObR-positive tissues also expressed activated forms of STAT3 and Akt. In line with this finding, ObR-positive glioblastoma cells responded to leptin with cell growth and induction of the STAT3 and Akt pathways as well as inactivation of the cell cycle suppressor Rb. In summary, our data demonstrate that the leptin/ObR system is expressed in malignant brain tumors and might be involved in tumor progression.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Leptin/metabolism , Receptors, Leptin/metabolism , Blotting, Western , Brain/metabolism , Brain/pathology , Cell Count , Cell Line, Tumor , Cell Proliferation , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Oncogene Protein v-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
18.
J Cell Physiol ; 221(1): 189-94, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19492417

ABSTRACT

Both leptin and vascular endothelial growth factor (VEGF) are growth and angiogenic cytokines that are upregulated in different types of cancer and have been implicated in neoplastic progression. Here we investigated the molecular mechanism by which leptin and VEGF expression are regulated in colon cancer by epidermal growth factor (EGF). In colon cancer cell line HT-29, EGF induced the binding of signal transducer and activator transcription 3 (STAT3) to STAT3 consensus motifs within the VEGF and leptin promoters and stimulated leptin and VEGF mRNA and protein synthesis. All these EGF effects were significantly blocked when HT-29 cells were treated with an inhibitor of the phosphoinositide 3-kinase (PI3K) pathway, LY294002, or with small interfering RNA (siRNA) targeting STAT3. Thus, our study identified the EGF/PI3K/STAT3 signaling as an essential pathway regulating VEGF and leptin expression in EGF-responsive colon cancer cells. This suggests that STAT3 pathways might constitute attractive pharmaceutical targets in colon cancer patients where anti-EGF receptor drugs are ineffective.


Subject(s)
Colorectal Neoplasms/enzymology , Epidermal Growth Factor/metabolism , Leptin/metabolism , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , HT29 Cells , Humans , Leptin/genetics , Neovascularization, Physiologic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL