GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 22, No. 21 ( 2021-10-28), p. 11639-
    Abstract: Polyphenols from olive oil are endowed with several biological activities. Chemical modifications have been recently applied to these compounds to improve their therapeutic activity in different pathological settings, including cancer. Herein, we describe the in vitro effects on multiple myeloma (MM) cells of oleil hydroxytyrosol (HTOL), a synthetic fatty ester of natural hydroxytyrosol with oleic acid. HTOL reduced the viability of various human MM cell lines (HMCLs), even when co-cultured with bone marrow stromal cells, triggering ER stress, UPR and apoptosis, while it was not cytotoxic against healthy peripheral blood mononuclear cells or B lymphocytes. Whole-transcriptome profiling of HTOL-treated MM cells, coupled with protein expression analyses, indicate that HTOL antagonizes key survival pathways for malignant plasma cells, including the undruggable IRF4–c-MYC oncogenic axis. Accordingly, c-MYC gain- and loss-of-function strategies demonstrate that HTOL anti-tumor activity was, at least in part, due to c-MYC targeting. Taken together, these findings underscore the anti-MM potential of HTOL, providing the molecular framework for further investigation of HTOL-based treatments as novel anti-cancer agents.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Experimental & Clinical Cancer Research, Springer Science and Business Media LLC, Vol. 42, No. 1 ( 2023-03-27)
    Abstract: Multiple myeloma (MM) is a hematologic malignancy characterized by high genomic instability, and telomere dysfunction is an important cause of acquired genomic alterations. Telomeric repeat-containing RNA (TERRA) transcripts are long non-coding RNAs involved in telomere stability through the interaction with shelterin complex. Dysregulation of TERRAs has been reported across several cancer types. We recently identified a small molecule, hit 17, which stabilizes the secondary structure of TERRA. In this study, we investigated in vitro and in vivo anti-MM activities of hit 17. Methods Anti-proliferative activity of hit 17 was evaluated in different MM cell lines by cell proliferation assay, and the apoptotic process was analyzed by flow cytometry. Gene and protein expressions were detected by RT-qPCR and western blotting, respectively. Microarray analysis was used to analyze the transcriptome profile. The effect of hit 17 on telomeric structure was evaluated by chromatin immunoprecipitation. Further evaluation in vivo was proceeded upon NCI-H929 and AMO-1 xenograft models. Results TERRA G4 stabilization induced in vitro dissociation of telomeric repeat‐binding factor 2 (TRF2) from telomeres leading to the activation of ATM-dependent DNA damage response, cell cycle arrest, proliferation block, and apoptotic death in MM cell lines. In addition, up-regulation of TERRA transcription was observed upon DNA damage and TRF2 loss. Transcriptome analysis followed by gene set enrichment analysis (GSEA) confirmed the involvement of the above-mentioned processes and other pathways such as E2F, MYC, oxidative phosphorylation, and DNA repair genes as early events following hit 17-induced TERRA stabilization. Moreover, hit 17 exerted anti-tumor activity against MM xenograft models. Conclusion Our findings provide evidence that targeting TERRA by hit 17 could represent a promising strategy for a novel therapeutic approach to MM.
    Type of Medium: Online Resource
    ISSN: 1756-9966
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2430698-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Translational Medicine, Springer Science and Business Media LLC, Vol. 20, No. 1 ( 2022-10-22)
    Abstract: DNA ligases are crucial for DNA repair and cell replication since they catalyze the final steps in which DNA breaks are joined. DNA Ligase III (LIG3) exerts a pivotal role in Alternative-Non-Homologous End Joining Repair (Alt-NHEJ), an error-prone DNA repair pathway often up-regulated in genomically unstable cancer, such as Multiple Myeloma (MM). Based on the three-dimensional (3D) LIG3 structure, we performed a computational screening to identify LIG3-targeting natural compounds as potential candidates to counteract Alt-NHEJ activity in MM. Methods Virtual screening was conducted by interrogating the Phenol Explorer database. Validation of binding to LIG3 recombinant protein was performed by Saturation Transfer Difference (STD)—nuclear magnetic resonance (NMR) experiments. Cell viability was analyzed by Cell Titer-Glo assay; apoptosis was evaluated by flow cytometric analysis following Annexin V-7AAD staining. Alt-NHEJ repair modulation was evaluated using plasmid re-joining assay and Cytoscan HD. DNA Damage Response protein levels were analyzed by Western blot of whole and fractionated protein extracts and immunofluorescence analysis. The mitochondrial DNA (mtDNA) copy number was determined by qPCR. In vivo activity was evaluated in NOD-SCID mice subcutaneously engrafted with MM cells. Results Here, we provide evidence that a natural flavonoid Rhamnetin (RHM), selected by a computational approach, counteracts LIG3 activity and killed Alt-NHEJ-dependent MM cells. Indeed, Nuclear Magnetic Resonance (NMR) showed binding of RHM to LIG3 protein and functional experiments revealed that RHM interferes with LIG3-driven nuclear and mitochondrial DNA repair, leading to significant anti-MM activity in vitro and in vivo. Conclusion Taken together, our findings provide proof of concept that RHM targets LIG3 addiction in MM and may represent therefore a novel promising anti-tumor natural agent to be investigated in an early clinical setting.
    Type of Medium: Online Resource
    ISSN: 1479-5876
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2118570-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 2 ( 2021-02), p. e002026-
    Abstract: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease with a poor cure rate for relapsed/resistant patients. Due to the lack of T-cell restricted targetable antigens, effective immune-therapeutics are not presently available and the treatment of chemo-refractory T-ALL is still an unmet clinical need. To develop novel immune-therapy for T-ALL, we generated an afucosylated monoclonal antibody (mAb) (ahuUMG1) and two different bispecific T-cell engagers (BTCEs) against UMG1, a unique CD43-epitope highly and selectively expressed by T-ALL cells from pediatric and adult patients. Methods UMG1 expression was assessed by immunohistochemistry (IHC) on a wide panel of normal tissue microarrays (TMAs), and by flow cytometry on healthy peripheral blood/bone marrow-derived cells, on 10 different T-ALL cell lines, and on 110 T-ALL primary patient-derived cells. CD43-UMG1 binding site was defined through a peptide microarray scanning. ahuUMG1 was generated by Genetic Glyco-Engineering technology from a novel humanized mAb directed against UMG1 (huUMG1). BTCEs were generated as IgG1-(scFv) 2 constructs with bivalent (2+2) or monovalent (2+1) CD3ε arms. Antibody dependent cellular cytotoxicity (ADCC), antibody dependent cellular phagocytosis (ADCP) and redirected T-cell cytotoxicity assays were analysed by flow cytometry. In vivo antitumor activity of ahUMG1 and UMG1-BTCEs was investigated in NSG mice against subcutaneous and orthotopic xenografts of human T-ALL. Results Among 110 T-ALL patient-derived samples, 53 (48.1%) stained positive (24% of TI/TII, 82% of TIII and 42.8% of TIV). Importantly, no expression of UMG1-epitope was found in normal tissues/cells, excluding cortical thymocytes and a minority ( 〈 5%) of peripheral blood T lymphocytes. ahUMG1 induced strong ADCC and ADCP on T-ALL cells in vitro, which translated in antitumor activity in vivo and significantly extended survival of treated mice. Both UMG1-BTCEs demonstrated highly effective killing activity against T-ALL cells in vitro. We demonstrated that this effect was specifically exerted by engaged activated T cells. Moreover, UMG1-BTCEs effectively antagonized tumor growth at concentrations 〉 2 log lower as compared with ahuUMG1, with significant mice survival advantage in different T-ALL models in vivo. Conclusion Altogether our findings, including the safe UMG1-epitope expression profile, provide a framework for the clinical development of these innovative immune-therapeutics for this still orphan disease.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Experimental Hematology & Oncology, Springer Science and Business Media LLC, Vol. 11, No. 1 ( 2022-09-12)
    Abstract: Activating G protein-coupled estrogen receptor 1 (GPER1) is an attractive therapeutic strategy for treating a variety of human diseases including cancer. Here, we show that GPER1 is significantly upregulated in tumor cells from different cohorts of Waldenström Macroglobulinemia (WM) patients compared to normal B cells. Using the clinically applicable GPER1-selective small-molecule agonist G-1 (also named Tespria), we found that pharmacological activation of GPER1 leads to G2/M cell cycle arrest and apoptosis both in vitro and in vivo in animal models, even in the context of the protective bone marrow milieu . Activation of GPER1 triggered the TP53 pathway, which remains actionable during WM progression. Thus, this study identifies a novel therapeutic target in WM and paves the way for the clinical development of the GPER1 agonist G-1.
    Type of Medium: Online Resource
    ISSN: 2162-3619
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2669066-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2827-2827
    Abstract: Alternative Non Homologous End Joining DNA repair as therapeutic target in Multiple MyelomaDaniele Caracciolo, Martina Montesano, Emanuela Altomare, Grazia Consolo, Nicola Amodio, Maria Teresa Di Martino, Marco Rossi, Cirino Botta, Eugenio Morelli, Giada Juli, Pierosandro Tagliaferri, Pierfrancesco Tassone. UMG of Catanzaro, Catanzaro, ItalyBackground- Multiple Myeloma (MM) is a hematologic malignancy strongly characterized by genomic instability. We previously demonstrated that LIG3-dependent Alt-NHEJ repair is significantly involved in the genomic instability which promote survival and drug resistance of MM cells. Based on this rationale, we aimed to elucidate the efficacy of available Alt-NHEJ pathway inhibitors as new therapeutic agents in MM. Materials and methods- Cell proliferation and apoptosis were evaluated with CellTiter-Glo assay and Annexin V staining. Alt-NHEJ repair was evaluated using EJ2-GFP and EJ5-GFP plasmids. LIG3, LIG1, PARP1, Caspase 3 and p-H2AX, levels were analyzed by Western blot of whole protein extracts. In vivo anti-MM activity was evaluated in NOD-SCID mice bearing subcutaneous AMO-1 Bortezomib resistant (ABZB) xenografts, daily treated with Olaparib via oral gavage. Results- By interrogating public available datasets, we found that higher mRNA expression of Alt-NHEJ core components was correlated with shorter survival of MM patients. Indeed, plasmid-based reporter assays confirmed a prevalent activation of Alt-NHEJ repair in MM cells as compared to normal lymphoblastoid cell lines, except for U266 cells which instead displayed a prevalence of Canonical-NHEJ repair activity. Next, we attempted to evaluate the effects of available Alt-NHEJ inhibitors on MM cell survival. Consistently, L67 (LIG3-LIG1 inhibitor) or Olaparib (PARP-inhibitor) induced a significant reduction of proliferation and clonogenic cell growth of different MM cell lines at low micromolar concentrations, while no significant effects on survival were observed in U226 as well as in normal lymphoblastoid cells. Importantly L67 or Olaparib impaired viability of MM cell lines or primary plasmacells co-cultured on stromal cells, thus overcoming the supporting role played by MM-microenvironment on plasma-cells survival. As result of Alt-NHEJ repair inhibition, anti-proliferative effects were correlated to increase of DNA double-strand breaks (DSBs), cell cycle arrest and finally apoptosis. Notably, we observed that Bortezomib resistant AMO-1 derivative cells (ABZB) exhibited significantly increased sensitivity to a combination of L67 and Olaparib that increased the number of DSBs. Moreover, Alt-NHEJ inhibitors synergistically sensitized primary cells as well as MM cell lines to Bortezomib. Finally, to demonstrate the in vivo relevance of our findings, we showed that clinically available Parp-inhibitor Olaparib, exerted a significant anti-MM activity on ABZB cells injected in immunocompromised mice. Conclusion- Taken together, our findings indicate that MM cells are addicted to Alt-NHEJ repair parthway, which therefore represent a novel druggable target pathway in MM. Citation Format: Daniele Caracciolo, Martina Montesano, Emanuela Altomare, Grazia Consolo, Nicola Amodio, Maria Teresa Di Martino, Marco Rossi, Cirino Botta, Eugenio Morelli, Giada Juli, Pierosandro Tagliaferri, Pierfrancesco Tassone. Alternative non-homologous end joining DNA repair as therapeutic target in multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2827.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2550-2550
    Abstract: Dysregulation of non-coding RNAs features prominently in the biology of multiple myeloma (MM). However, if short non-coding RNAs have been extensively studied in this malignancy, the role of long non-coding RNAs remains to be elucidated. A custom annotation pipeline of microarray data investigating lncRNA expression in plasma cells from 20 MGUS, 33 smoldering MM, 170 MM, 36 plasma cell leukemia patients, and 9 healthy donors, indicates overexpression of the lncRNA MALAT1 during MM progression. On this finding, we sought to investigate the functional significance of MALAT1 expression in MM by the use of 16-mer LNA-gapmeR synthetic oligonucleotides, that trigger specific RNAse H-dependent degradation of MALAT1. Transfected or gymnotic (naked) anti-MALAT1 gapmeRs decreased MM patients (n=3) and cell lines growth (n=7), triggering ER stress and apoptosis, while spared healthy peripheral blood mononuclear cells. Microarray gene profiling indicates that MALAT1 depletion was associated with modulation of various gene sets, and specifically had a negative impact on genes encoding for 20S proteasome β subunits, such as PSMβ2/4/5 and 7, whose overexpression is linked to proteasome impairment in cancer cells. Down-modulation of PSMβs after anti-MALAT1 gapmeRs exposure was confirmed at protein level and was associated to reduced trypsine, chymotrypsine and caspase-like proteasomal activities and to the accumulation of poly-ubiquitylated species. Consistently, anti-MALAT1 gapmeRs enhanced the in vitro anti-MM activity of the proteasome inhibitor bortezomib in bortezomib-sensitive and resistant MM cells. In vivo, i.p. administration of naked anti-MALAT1 gapmeRs (10 and 25 mg/kg) inhibited the growth of bortezomib-resistant MM xenografts, with no evidence of organ toxicity. Mechanistically, anti-MALAT1-induced PSMβs down-modulation could be ascribed to the inhibition of the transcription factors NRF1-2, which act as positive regulators of PSMSβs transcription. On the other hand, siRNA or pharmacologic inhibition of NRF1 by all-trans retinoic acid decreased promoter activity and expression of MALAT1, indicating the occurrence of a feedback loop involving MALAT1 and NRF1. Collectively, our data indicate that MALAT1-targeting represents a promising strategy to dampen aberrant proteasome activity in MM cells, and provide the preclinical rationale to use oligonucleotide therapeutics as anti-MALAT1 gapmeRs for MM treatment. Citation Format: Maria Angelica Stamato, Nicola Amodio, Eugenio Morelli, Giada Juli, Martina Manzoni, Maria Eugenia Gallo Cantafio, Lavinia Biamonte, Elisa Taiana, Annamaria Gullà, Emanuela Altomare, Francesca Scionti, Daniele Caracciolo, Cirino Botta, Maria Cucè, Mariamena Arbitrio, Maria Teresa Di Martino, Marco Rossi, Antonino Neri, Pierosandro Tagliaferri, Pierfrancesco Tassone. Anti-MALAT1 synthetic oligonucleotides target the proteasome and exert anti-multiple myeloma activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2550. doi:10.1158/1538-7445.AM2017-2550
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancers, MDPI AG, Vol. 14, No. 12 ( 2022-06-11), p. 2886-
    Abstract: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy burdened by poor prognosis. While huge progress of immunotherapy has recently improved the outcome of B-cell malignancies, the lack of tumor-restricted T-cell antigens still hampers its progress in T-ALL. Therefore, innovative immunotherapeutic agents are eagerly awaited. To this end, we generated a novel asymmetric (2 + 1) bispecific T-cell engager (BTCE) targeting CD1a and CD3ε (CD1a x CD3ε) starting from the development of a novel mAb named UMG2. UMG2 mAb reacts against CD1a, a glycoprotein highly expressed by cortical T-ALL cells. Importantly, no UMG2 binding was found on normal T-cells. CD1a x CD3ε induced high T-cell mediated cytotoxicity against CD1a+ T-ALL cells in vitro, as demonstrated by the concentration-dependent increase of T-cell proliferation, degranulation, induction of cell surface activation markers, and secretion of pro-inflammatory cytokines. Most importantly, in a PBMC-reconstituted NGS mouse model bearing human T-ALL, CD1a x CD3ε significantly inhibited the growth of human T-ALL xenografts, translating into a significant survival advantage of treated animals. In conclusion, CD1a x CD3ε is a novel BTCE highly active against CD1a-expressing cortical-derived T-ALL cells suitable for clinical development as an effective therapeutic option for this rare and aggressive disease.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancers, MDPI AG, Vol. 15, No. 6 ( 2023-03-08), p. 1647-
    Abstract: Sarcomas are heterogeneous malignancies with limited therapeutic options and a poor prognosis. We developed an innovative immunotherapeutic agent, a first-in-class Pronectin™-based Bispecific T-Cell Engager (pAXL×CD3ε), for the targeting of AXL, a TAM family tyrosine kinase receptor highly expressed in sarcomas. AXL expression was first analyzed by flow cytometry, qRT-PCR, and Western blot on a panel of sarcoma cell lines. The T-cell-mediated pAXL×CD3ε cytotoxicity against sarcoma cells was investigated by flow cytometry, luminescence assay, and fluorescent microscopy imaging. The activation and degranulation of T cells induced by pAXL×CD3ε were evaluated by flow cytometry. The antitumor activity induced by pAXL×CD3ε in combination with trabectedin was also investigated. In vivo activity studies of pAXL×CD3ε were performed in immunocompromised mice (NSG), engrafted with human sarcoma cells and reconstituted with human peripheral blood mononuclear cells from healthy donors. Most sarcoma cells showed high expression of AXL. pAXL×CD3ε triggered T-lymphocyte activation and induced dose-dependent T-cell-mediated cytotoxicity. The combination of pAXL×CD3ε with trabectedin increased cytotoxicity. pAXL×CD3ε inhibited the in vivo growth of human sarcoma xenografts, increasing the survival of treated mice. Our data demonstrate the antitumor efficacy of pAXL×CD3ε against sarcoma cells, providing a translational framework for the clinical development of pAXL×CD3ε in the treatment of human sarcomas, aggressive and still-incurable malignancies.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2023
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Archiv der Pharmazie, Wiley, Vol. 356, No. 8 ( 2023-08)
    Abstract: Nowadays, RNA is an attractive target for the design of new small molecules with different pharmacological activities. Among several RNA molecules, long noncoding RNAs (lncRNAs) are extensively reported to be involved in cancer pathogenesis. In particular, the overexpression of lncRNA metastasis‐associated lung adenocarcinoma transcript 1 ( MALAT1) plays an important role in the development of multiple myeloma (MM). Starting from the crystallographic structure of the triple‐helical stability element at the 3'‐end of MALAT1 , we performed a structure‐based virtual screening of a large commercial database, previously filtered according to the drug‐like properties. After a thermodynamic analysis, we selected five compounds for the in vitro assays. Compound M5 , characterized by a diazaindene scaffold, emerged as the most promising molecule enabling the destabilization of the MALAT1 triplex structure and antiproliferative activity on in vitro models of MM. M5 is proposed as a lead compound to be further optimized for improving its affinity toward MALAT1 .
    Type of Medium: Online Resource
    ISSN: 0365-6233 , 1521-4184
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 1496815-0
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...