Your search history is empty.
feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Die angezeigten Daten werden derzeit aktualisiert.
Export
  • 1
    Online Resource
    Online Resource
    Bentham Science Publishers Ltd. ; 2021
    In:  Current Medicinal Chemistry Vol. 28, No. 11 ( 2021-04), p. 2114-2136
    In: Current Medicinal Chemistry, Bentham Science Publishers Ltd., Vol. 28, No. 11 ( 2021-04), p. 2114-2136
    Abstract: The costs of developing, validating and buying new drugs are dramatically increasing. On the other hand, sobering economies have difficulties in sustaining their healthcare systems, particularly in countries with an elderly population requiring increasing welfare. This conundrum requires immediate action, and a possible option is to study the large, already present arsenal of drugs approved and to use them for innovative therapies. This possibility is particularly interesting in oncology, where the complexity of the cancer genome dictates in most patients a multistep therapeutic approach. In this review, we discuss a) Computational approaches; b) preclinical models; c) currently ongoing or already published clinical trials in the drug repurposing field in oncology; and d) drug repurposing to overcome resistance to previous therapies.
    Type of Medium: Online Resource
    ISSN: 0929-8673
    Language: English
    Publisher: Bentham Science Publishers Ltd.
    Publication Date: 2021
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2019
    In:  The Cancer Journal Vol. 25, No. 2 ( 2019-3), p. 82-87
    In: The Cancer Journal, Ovid Technologies (Wolters Kluwer Health), Vol. 25, No. 2 ( 2019-3), p. 82-87
    Abstract: The never-ending explosion in the cost of new oncology drugs is reducing in many countries the access to the most recent, effective anticancer therapies and represents a significant obstacle to the design and realization of combinatorial trials. Already approved, anticancer and nonanticancer drugs can be considered for in silico, preclinical, and clinical repurposing approaches and offer the significant advantages of a potentially cheaper, faster, and safer validation. This review discusses recent advances and challenges in the field.
    Type of Medium: Online Resource
    ISSN: 1540-336X , 1528-9117
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2019
    detail.hit.zdb_id: 2126320-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 18 ( 2017-09-15), p. 5169-5182
    Abstract: A cell population with progenitor-like phenotype (CD45-CD34+) resident in human white adipose tissue (WAT) is known to promote the progression of local and metastatic breast cancer and angiogenesis. However, the molecular mechanisms of the interaction have not been elucidated. In this study, we identified two proteins that were significantly upregulated in WAT-derived progenitors after coculture with breast cancer: granulocyte macrophage colony-stimulating factor (GM-CSF) and matrix metallopeptidase 9 (MMP9). These proteins were released by WAT progenitors in xenograft and transgenic breast cancer models. GM-CSF was identified as an upstream modulator. Breast cancer–derived GM-CSF induced GM-CSF and MMP9 release from WAT progenitors, and GM-CSF knockdown in breast cancer cells neutralized the protumorigenic activity of WAT progenitors in preclinical models. GM-CSF neutralization in diet-induced obese mice significantly reduced immunosuppression, intratumor vascularization, and local and metastatic breast cancer progression. Similarly, MMP9 inhibition reduced neoplastic angiogenesis and significantly decreased local and metastatic tumor growth. Combined GM-CSF neutralization and MMP9 inhibition synergistically reduced angiogenesis and tumor progression. High-dose metformin inhibited GM-CSF and MMP9 release from WAT progenitors in in vitro and xenograft models. In obese syngeneic mice, metformin treatment mimicked the effects observed with GM-CSF neutralization and MMP9 inhibition, suggesting these proteins as new targets for metformin. These findings support the hypothesis that GM-CSF and MMP9 promote the protumorigenic effect of WAT progenitors on local and metastatic breast cancer. Cancer Res; 77(18); 5169–82. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 900-900
    Abstract: The clinical success of cyclophosphamide (Cy)-based haploidentical stem cell transplants in hematological malignancies indicates that this drug has the potential to reorchestrate the immune system against cancer cells. Along a similar way, Cy is administered before the infusion of CAR-T cells to improve their clinical efficacy. We have previously found that daily, low/medium-dose Cy (20mg/Kg), in association with vinorelbine (V) was able to improve the preclinical efficacy of checkpoint inhibitors (CIs) anti-PD-1 and anti-PD-L1 in preclinical models of breast cancer and lymphoma (Orecchioni et al, 2018). Randomized clinical trials have recently indicated that the addition of CIs to other chemotherapy drugs such as taxanes (T), doxorubicin (D), or platinum (P) might be beneficial in triple negative breast cancer (TNBC) patients. Thus, we designed the present study in two orthotopic, immunocompetent, local and metastatic models of TNBC (CI-resistant 4T1 and EMT6, moderately responsive to CIs) to investigate whether Cy and V, in different schedules and doses, were more effective than T, D, or P. In vivo studies in local and metastatic models using both 4T1 and EMT6 TNBC cells clearly indicated that intermittent (ie every 6 days), medium-dosage (140 mg/Kg) Cy, was more effective than any other combinatorial regimens including CIs plus daily low/medium-dose Cy, or T, D, or P at their regular dosages. The association of V further increased the preclinical efficacy of intermittent Cy plus CIs, and abrogated tumor growth in both 4T1 and EMT6 models. In vivo studies with neutralizing monoclonal antibodies targeting in separate experiments T, B, NK, and myeloid cells demonstrated that CD3+CD4+, CD3+CD8+ T cells as well as CD11b+ monocyte/macrophage dendritic cells were crucial to abrogate tumor growth in TNBC-bearing mice treated with intermittent Cy, V and CIs. Single-cell transcriptome analysis of more than 60,000 intratumoral immune cells and flow cytometry studies in circulating and intratumoral subsets of immune cells indicated that V promoted the generation and maturation of myeloid APC cells, and that intermittent Cy generated new clones of tumor-infiltrating CD3+CD4+ and CD3+CD8+ TCR alpha beta cells. After treatment with intermittent Cy, V and CIs, intratumoral immune cells showed a unique gene signature of 42 genes (CD3D, Ptprc, CD69, Lat, Lck, CD2, CD28, CD27, and CD3E among others) suggestive of a change in morphology and behavior resulting from exposure to a mitogen, cytokine, chemokine, cellular ligand, or an antigen for which it is specific, as well as APC-to-T cell adhesion. Taken together, our data support the hypothesis that APC priming and T cell clonal replacement can significantly improve CI efficacy in vivo in TNBC models. We are further investigating the Cy-related mechanisms inducing intratumoral T-cell clonal replacement and how these T cells cooperate with V-induced APCs. Citation Format: Paolo Falvo, Stefania Orecchioni, Roman Hillje, Alessandro Raveane, Patrizia Mancuso, Chiara Camisaschi, Lucilla Luzi, Francesco Bertolini. Intermittent cyclophosphamide and vinorelbine reshape the immune cell environment, induce T cell clonal replacement and increase the efficacy of PD-1 inhibition in models of triple negative breast cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 900.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5214-5214
    Abstract: We have recently described that the human white adipose tissue (WAT) contains progenitors with cooperative roles in breast cancer (BC) angiogenesis, local and metastatic progression (Martin-Padura et al, 2012; Orecchioni et al, 2013). The biguanide metformin (met), commonly used for type 2 diabetes, might have activity against BC and we found it able to inhibit angiogenesis in vivo (Dallaglio et al, 2014; Orecchioni et al, 2014). We studied met and another biguanide, phenformin (phe), in vitro and in vivo in orthotopic NSG murine models of local and metastatic BC. As met is frequently administered with aspirin or atenolol in diabetic/obese patients, we studied in vitro and vivo their association. In vitro, biguanides activated AMPK, inhibited complex 1 of the respiratory chain and induced apoptosis of BC and WAT endothelial cells. Aspirin was synergistic with met and phe in inducing apoptosis of estrogen receptor+ BC cells. This synergistic effect was less evident in triple negative BC cells. In co-culture, biguanides significantly inhibited the production of several angiogenic proteins. In vivo, biguanides inhibited local and metastatic growth of triple negative and HER2+ BC in immune-competent and immune-deficient mice orthotopically injected with BC. Biguanides also inhibited local and metastatic BC growth in a genetically engineered model of HER2+ BC. In vivo, biguanides increased pimonidazole binding (but not HIF-1 expression) of WAT progenitors, reduced tumor microvessel density and impaired of the vascular pericyte/endothelial cell ratio, so that cancer vessels displayed a dysplastic phenotype. This effect was significantly increased by the addition of aspirin or atenolol. This was evident also when AMPK activation was assessed in combination therapy regimens. AMPK phosphorylation was significantly increased in BC cells treated with met+aspirin or phe+atenolol as compared to met or phe alone. In WAT progenitors, AMPK activation was enhanced only under met+aspirin and met+atenolol combinations. In immune-competent mice, met effect in BC models was significantly enhanced by the addition of atenolol or of aspirin. Phe was significantly more active than met both in vitro and in vivo. Considering their safety profile, biguanides (alone or in combination with aspirin or atenolol) deserve to be further investigated for BC prevention in high-risk subjects, in combination with chemo and/or targeted therapy and/or as post-therapy consolidation or maintenance therapy for the prevention of BC recurrence. Citation Format: Giovanna Talarico, Francesca Reggiani, Stefania Orecchioni, Patrizia Mancuso, Angelica Calleri, Giuliana Gregato, Valentina Labanca, Douglas M. Noonan, Katiuscia Dallaglio, Adriana Albini, Francesco Bertolini. Synergistic activity of aspirin, atenolol and metformin in the inhibition of angiogenesis, local and metastatic growth of breast cancer by targeting both neoplastic and microenvironment cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5214. doi:10.1158/1538-7445.AM2015-5214
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Stem Cells and Development, Mary Ann Liebert Inc, Vol. 27, No. 1 ( 2018-01), p. 23-34
    Type of Medium: Online Resource
    ISSN: 1547-3287 , 1557-8534
    Language: English
    Publisher: Mary Ann Liebert Inc
    Publication Date: 2018
    detail.hit.zdb_id: 2142305-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1712-1712
    Abstract: Checkpoint inhibitors (CIs) such as anti-PD-1 and anti PD-L1 have shown combinatorial clinical activity with chemotherapy in triple negative breast cancer (TNBC), but only in a minority of patients and only for a limited period of time. Moreover, it is currently unclear what chemotherapeutic drug is the most appropriate to combine with CIs in TNBC patients. We have investigated at the single cell level the transcriptome and the trajectories of more than 50,000 innate and adaptive intratumoral immune cells in two syngeneic, immune competent, orthotopic murine models of local and metastatic TNBC. Mice injected with 4T1 cells had a predominant lymphoid infiltrate, mice injected with EMT6 cells had a predominant myeloid infiltrate. Mice were treated with CIs and several different types of chemotherapeutics, alone or in combinations. In both models, capecitabine (alone or with CIs) was the less effective drug. Platinum, doxorubicin and taxanes showed synergy with CIs and had superimposable activity. Intermittent, medium dosage cyclophosphamide (CTX) plus vinorelbine and CIs was the most active combinatorial therapy (Falvo et al, Cancer Research 2021). Vinorelbine activated antigen presenting cells and CTX generated new T cell clones including stem cell-like TCF1+ CD8+ T cells. Treatments with most in vivo efficacy were associated to a decrease of regulatory T cells and of gamma delta T cells, which were found to have a pro-tumoral activity in these murine models, likely due to IL-17 expression in the neoplastic microenvironment. An increase of several different clusters of exhausted-like CD8+ T cells was observed in pre-clinical treatments with low efficacy; an opposite trend was found for several clusters of proliferative CD8+ T cells in treatments with high in vivo efficacy. Regarding macrophages, M2-like cells were enriched after treatments with low efficacy, while an opposite behaviour was found in M1-like macrophages. Interestingly, we observed a significant increase of an M1-like cluster with high expression of the Ly6c1/Ly6c2 gene in mice successfully treated with vinorelbine, CTX and CIs. For both cell lines the percentage of plasma B cells increased after in vivo treatments with high efficacy. In particular, the most effective treatment significantly increased the frequency of germinal B cells, which were absent in untreated tumors. These data can lead to new insights on the diagnosis and treatment of TNBC and to possible clinical applications. Citation Format: Laura Carpen, Paolo Falvo, Stefania Orecchioni, Giulia Mitola, Roman Hillje, Saveria Mazzara, Patrizia Mancuso, Stefano Pileri, Alessandro Raveane, Francesco Bertolini. A single-cell RNA atlas of innate and adaptive intratumoral immunity in triple negative breast cancer during chemo- and immunotherapies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1712.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 3374-3374
    Abstract: We recently described a human cell population with progenitor-like phenotype (CD45-CD34+), resident in the white adipose tissue (WAT) and able to promote local and metastatic breast cancer (BC) progression and angiogenesis (Orecchioni et al., 2013). The molecular mechanism involved in this interaction has been so far elusive. An extensive screening of candidate molecules related to angiogenesis, inflammation, motility and invasiveness revealed that two proteins are significantly up-regulated in WAT-derived progenitors following culture with BC cells: Granulocyte-macrophage colony-stimulating factor (GM-CSF) and Matrix metallopeptidase 9 (MMP9). In vivo, both proteins were overexpressed in orthotopic models of human BC co-injected with human WAT progenitors. The inhibition of GM-CSF by a monoclonal antibody in diet-induced, obese BC mice led to a reduced intratumor vascularization and a strong impairment of WAT immunosuppressive microenvironment, targeting mainly myeloid cells such as macrophages and myeloid derived suppressor cells (MDSCs) in peritumoral WAT. Circulating levels of monocytes and of CD4+CD25brightCD127low/neg T-regulatory cells (T-regs) were also reduced in treated mice. Similarly, soluble immunosuppressive factors, such as IL-10 and IL-5, and CD274 (PD-L1) were reduced in tumors and WAT collected from immune competent mice neutralized for GM-CSF, confirming the crucial role of the factor in promoting tumor immune escape. This resulted in a significantly reduced local BC growth and lower metastatic progression in vivo. All these findings challenge the clinical use of GM-CSF. In the same syngeneic model, MMP9 inhibition reduced neoplastic angiogenesis and significantly decreased local and metastatic tumor growth, without altering immune cells composition in tumor microenvironment. The combined inhibition of GM-CSF and MMP9 was synergic in impairing angiogenesis, local and metastatic BC growth in diet-induced obese orthotopic BC models, indicating a potential complementary role in tumor spread. As we recently reported that Metformin targets both BC cells and the neoplastic WAT environment (Orecchioni et al., 2015), we investigated Metformin effect over GM-CSF and MMP9 expression. Metformin inhibited GM-CSF and MMP9 up-release from WAT progenitors in vitro. Circulating GM-CSF was significantly impaired in BC xenografts administered with Metformin and MMP9 expression was also affected by the treatment. Collectively these results indicate GM-CSF and MMP9 as key candidates involved in the pro-tumorigenic effect of WAT progenitors on BC in a setting of obesity. The comparison between Metformin and GM-CSF/MMP9 specific inhibition is currently under investigation. Citation Format: Francesca Reggiani, Valentina Labanca, Giovanna Talarico, Stefania Orecchioni, Patrizia Mancuso, Francesco Bertolini. GM-CSF and MMP9 are key regulators of the effect of adipose progenitor cells over breast cancer onset and metastatic progression in obesity. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3374.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1010-1010
    Abstract: Epidemiological evidence has suggested that metformin, an anti-hyperglycemic agent commonly used in the treatment of type 2 diabetes, is a potential cancer preventive agent. Anti-angiogenesis represents a key mechanism in cancer prevention, a concept termed angioprevention. Since conflicting data concerning the anti-angiogenic action of metformin are emerging, we elucidate the effects of metformin, on endothelial and tumor cells as well as on angiogenesis using in vitro, in vivo and transcriptomic approaches. We show that metformin inhibits endothelial cell ability to organize into capillary-like networks; this effect is partially dependent on the energy sensor AMPK. Gene expression and proteins profiling revealed paradoxic effects on several angiogenesis associated factors. We found induction of VEGF, COX2 and CXCR4 at the mRNA level and down-regulation of ADAMTS1. Interestingly, antibody array analysis showed essentially opposite regulation of numerous angiogenesis-associated proteins in endothelial and breast cancer cells. We also show that endothelial production of cytochrome p450 family member CYP1B1 was up-regulated by tumor cell supernatants, while metformin blocked this effect by acting on AMPK. The metformin anti-angiogenic activity was exerted through inhibition of ERK1/2 activation, even in the presence of VEGF, while blocking AMPK activity abrogated this effect. Metformin inhibited angiogenesis induced by VEGF in matrigel pellets in vivo and contrasted the increase in microvessel density in obese mice on a high fat diet. Further, it down-regulated the number of endothelial precursor cells from white adipose tissue in obese mice. Our data show that metformin has an anti-angiogenic activity in vitro and in vivo, which is associated with a contradictory enhancement of chemokines and other inflammatory pro-angiogenic mediators, as well as a different regulation in endothelial and breast cancer cells. Citation Format: Antonino Bruno, Katiuscia Dallaglio, Anna Rita Cantelmo, Alessia I. Esposito, Luca Ruggiero, Stefania Orecchioni, Angelica Callieri, Francesco Bertolini, Ulrich Pfeffer, Douglas M. Noonan, Adriana Albini. Paradoxic effects of metformin on endothelial cells and angiogenesis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1010. doi:10.1158/1538-7445.AM2014-1010
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5212-5212
    Abstract: The white adipose tissue (WAT) promotes breast cancer (BC) progression with mechanisms that are only partially understood. We have found that WAT cells with progenitor-like phenotype (CD45-CD34+) are able to promote angiogenesis, motility, invasiveness, local and metastatic neoplastic progression in orthotopic murine models of triple negative BC (Martin-Padura et al., 2012; Orecchioni et al., 2013). Here we report the results of our screening for molecular signals involved in human WAT and BC interplay. Subpopulations of human WAT CD34+CD45- cells were cultured with several triple negative BC cell lines. An up-regulation of more than 10 proteins was detected in WAT- cells, suggesting a strong impact on their secretoma. In cellular supernatants two molecules were significantly up-regulated: GM-CSF and MMP9. Results were further confirmed by ELISA and western blotting using WAT cells from 10 different BC patients. GM-CSF was found to be 2 to 47 fold up-regulated both in transwell and in direct co-culture systems, thus suggesting a soluble factor implication. MMP9 showed up to 12 fold up-regulation in direct co-culture and a weak up-regulation in transwell system, suggesting a prevalent cell-to-cell mechanism. Quantitative RT-PCR revealed that WAT-progenitors were responsible for the observed up-regulation for both GM-CSF and MMP9, whereas BC cells did not change their basal expression levels of the two factors. Up-regulation was conserved among species both at RNA and protein levels. As we reported (Orecchioni et al., 2013) WAT CD45-CD34+ cells include two sub-populations of progenitors formally identified as mesenchymal (ASCs) and endothelial (EPCs). Both populations were able to contribute to GM-CSF and MMP9 up-release in a synergistic manner. In vivo studies in NODSCIDIL2Rgnull (NSG) mice orthotopic BC models confirmed the up-regulation of both factors: a significant increase of circulating human GM-CSF was detected in the plasma of mice injected with human WAT progenitors and BC compared to mice injected with BC alone. This up-regulation was statistically significant (p & lt;0.001) in the early phases of tumor growth. BC investigation revealed an increased expression of MMP9 in mice injected with WAT progenitors. Accordingly, the qRT-PCR on tumor tissues did not detect an up-regulation of the two factors, confirming that the observed up-regulation were due to WAT cells. As we recently published (Orecchioni et al, 2014), metformin can target in vitro and in vivo WAT progenitors. In this context, we found in vitro that metformin targeted GM-CSF and MMP9, significantly reducing mRNA in WAT progenitors and protein levels in co-culture systems. Moreover, in our NSG orthotopic BC models metformin significantly reduced BC-specific circulating human GM-CSF. We are currently investigating the effect of GM-CSF and MMP9 inhibition in vivo, alone or in combination with metformin. Citation Format: Francesca Reggiani, Patrizia Mancuso, Cristina Rabascio, Stefania Orecchioni, Giovanna Talarico, Cinzia Massaro, Valentina Labanca, Angelica Calleri, Francesco Bertolini. GM-CSF and MMP9, targets of metformin, are crucial mediators of the tumor-promoting role of adipose tissue cells in breast cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5212. doi:10.1158/1538-7445.AM2015-5212
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages