Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michele Moschetta is active.

Publication


Featured researches published by Michele Moschetta.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Engineered nanomedicine for myeloma and bone microenvironment targeting.

Archana Swami; Michaela R. Reagan; Pamela Basto; Yuji Mishima; Nazila Kamaly; Siobhan Glavey; Sufeng Zhang; Michele Moschetta; Dushanth Seevaratnam; Yong Zhang; Jinhe Liu; Masoumeh Memarzadeh; Jun Wu; Salomon Manier; Jinjun Shi; Nicolas Bertrand; Zhi Ning Lu; Kenichi Nagano; Roland Baron; Antonio Sacco; Aldo M. Roccaro; Omid C. Farokhzad; Irene M. Ghobrial

Significance Limited treatment options exist for cancer within the bone, as demonstrated by the inevitable, pernicious course of metastatic breast, prostate, and blood cancers. The difficulty of eliminating bone-residing cancer necessitates novel, alternative treatments to manipulate the tumor cells and their microenvironment, with minimal off-target effects. To this end, we engineered bone-homing, stealth nanoparticles to deliver anticancer, bone-stimulatory drugs, and demonstrated their utility with bortezomib (a model drug) and multiple myeloma (a model cancer). To test our hypothesis that increasing bone volume and strength inhibits tumor growth, mice were treated with these nanoparticles before being injected with cancer cells. Results demonstrated significantly slower myeloma growth and prolonged survival. Our research demonstrates the potential of bone-homing nanomedicine as an efficacious cancer treatment mechanism. Bone is a favorable microenvironment for tumor growth and a frequent destination for metastatic cancer cells. Targeting cancers within the bone marrow remains a crucial oncologic challenge due to issues of drug availability and microenvironment-induced resistance. Herein, we engineered bone-homing polymeric nanoparticles (NPs) for spatiotemporally controlled delivery of therapeutics to bone, which diminish off-target effects and increase local drug concentrations. The NPs consist of poly(d,l-lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and bisphosphonate (or alendronate, a targeting ligand). The engineered NPs were formulated by blending varying ratios of the synthesized polymers: PLGA-b-PEG and alendronate-conjugated polymer PLGA-b-PEG-Ald, which ensured long circulation and targeting capabilities, respectively. The bone-binding ability of Ald-PEG-PLGA NPs was investigated by hydroxyapatite binding assays and ex vivo imaging of adherence to bone fragments. In vivo biodistribution of fluorescently labeled NPs showed higher retention, accumulation, and bone homing of targeted Ald-PEG-PLGA NPs, compared with nontargeted PEG-PLGA NPs. A library of bortezomib-loaded NPs (bone-targeted Ald-Bort-NPs and nontargeted Bort-NPs) were developed and screened for optimal physiochemical properties, drug loading, and release profiles. Ald-Bort-NPs were tested for efficacy in mouse models of multiple myeloma (MM). Results demonstrated significantly enhanced survival and decreased tumor burden in mice pretreated with Ald-Bort-NPs versus Ald-Empty-NPs (no drug) or the free drug. We also observed that bortezomib, as a pretreatment regimen, modified the bone microenvironment and enhanced bone strength and volume. Our findings suggest that NP-based anticancer therapies with bone-targeting specificity comprise a clinically relevant method of drug delivery that can inhibit tumor progression in MM.


Blood | 2014

C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma

Aldo M. Roccaro; Antonio Sacco; Cristina Jiménez; Patricia Maiso; Michele Moschetta; Yuji Mishima; Yosra Aljawai; Ilyas Sahin; Michelle Kuhne; Pina M. Cardarelli; Lewis J. Cohen; Jesús F. San Miguel; Ramón García-Sanz; Irene M. Ghobrial

The C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in modulating cell trafficking in hematopoietic stem cells and clonal B cells. We screened 418 patients with B-cell lymphoproliferative disorders and described the presence of the C1013G/CXCR4 warts, hypogammaglobulinemia, infections, and myelokathexis-associated mutation in 28.2% (37/131) of patients with lymphoplasmacytic lymphoma (Waldenström macroglobulinemia [WM]), being either absent or present in only 7% of other B-cell lymphomas. In vivo functional characterization demonstrates its activating role in WM cells, as demonstrated by significant tumor proliferation and dissemination to extramedullary organs, leading to disease progression and decreased survival. The use of a monoclonal antibody anti-CXCR4 led to significant tumor reduction in a C1013G/CXCR4 WM model, whereas drug resistance was observed in mutated WM cells exposed to Brutons tyrosine kinase, mammalian target of rapamycin, and phosphatidylinositol 3-kinase inhibitors, but not proteasome inhibitors. These findings demonstrate that C1013G/CXCR4 is an activating mutation in WM and support its role as a critical regulator of WM molecular pathogenesis and as an important therapeutic target.


Immunological Reviews | 2015

Targeting the bone marrow microenvironment in multiple myeloma

Yawara Kawano; Michele Moschetta; Salomon Manier; Siobhan Glavey; Gullu Gorgun; Aldo M. Roccaro; Kenneth C. Anderson; Irene M. Ghobrial

Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow (BM). Despite the significant advances in treatment, MM is still a fatal malignancy. This is mainly due to the supportive role of the BM microenvironment in differentiation, migration, proliferation, survival, and drug resistance of the malignant plasma cells. The BM microenvironment is composed of a cellular compartment (stromal cells, osteoblasts, osteoclasts, endothelial cells, and immune cells) and a non‐cellular compartment. In this review, we discuss the interaction between the malignant plasma cell and the BM microenvironment and the strategy to target them.


Clinical Cancer Research | 2011

Lenalidomide restrains motility and overangiogenic potential of bone marrow endothelial cells in patients with active multiple myeloma

Annunziata De Luisi; Arianna Ferrucci; Addolorata Coluccia; Roberto Ria; Michele Moschetta; Emanuela de Luca; Luisa Pieroni; Michele Maffia; Andrea Urbani; Giulia Di Pietro; Attilio Guarini; Girolamo Ranieri; Paolo Ditonno; Simona Berardi; Antonella Caivano; Antonio Basile; Nicola Cascavilla; Silvana Capalbo; Giovanni Quarta; Franco Dammacco; Domenico Ribatti; Angelo Vacca

Purpose: To determine the in vivo and in vitro antiangiogenic power of lenalidomide, a “lead compound” of IMiD immunomodulatory drugs in bone marrow (BM) endothelial cells (EC) of patients with multiple myeloma (MM) in active phase (MMEC). Experimental Design: The antiangiogenic effect in vivo was studied using the chorioallantoic membrane (CAM) assay. Functional studies in vitro (angiogenesis, “wound” healing and chemotaxis, cell viability, adhesion, and apoptosis) were conducted in both primary MMECs and ECs of patients with monoclonal gammopathies (MGUS) of undetermined significance (MGEC) or healthy human umbilical vein endothelial cells (HUVEC). Real-time reverse transcriptase PCR, Western blotting, and differential proteomic analysis were used to correlate morphologic and biological EC features with the lenalidomide effects at the gene and protein levels. Results: Lenalidomide exerted a relevant antiangiogenic effect in vivo at 1.75 μmol/L, a dose reached in interstitial fluids of patients treated with 25 mg/d. In vitro, lenalidomide inhibited angiogenesis and migration of MMECs, but not of MGECs or control HUVECs, and had no effect on MMEC viability, apoptosis, or fibronectin- and vitronectin-mediated adhesion. Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways. Conclusions: This study provides information on the molecular mechanisms associated with the antimigratory and antiangiogenic effects of lenalidomide in primary MMECs, thus giving new avenues for effective endothelium-targeted therapies in MM. Clin Cancer Res; 17(7); 1935–46. ©2011 AACR.


Cancer Research | 2012

Paclitaxel Enhances Therapeutic Efficacy of the F8-IL2 Immunocytokine to EDA-Fibronectin–Positive Metastatic Human Melanoma Xenografts

Michele Moschetta; Francesca Pretto; Alexander Berndt; Kerstin Galler; Petra Richter; Andrea Bassi; Paolo Oliva; Edoardo Micotti; Giovanni Valbusa; Kathrin Schwager; Manuela Kaspar; Eveline Trachsel; Hartwig Kosmehl; Maria Rosa Bani; Dario Neri; Raffaella Giavazzi

The selective delivery of bioactive agents to tumors reduces toxicity and enhances the efficacy of anticancer therapies. In this study, we show that the antibody F8, which recognizes perivascular and stromal EDA-fibronectin (EDA-Fn), when conjugated to interleukin-2 (F8-IL2) can effectively inhibit the growth of EDA-Fn-expressing melanomas in combination with paclitaxel. We obtained curative effects with paclitaxel administered before the immunocytokine. Coadministration of paclitaxel increased the uptake of F8 in xenografted melanomas, enhancing tumor perfusion and permeability. Paclitaxel also boosted the recruitment of F8-IL2-induced natural killer (NK) cells to the tumor, suggesting a host response as part of the observed therapeutic benefit. In support of this likelihood, NK cell depletion impaired the antitumor effect of paclitaxel plus F8-IL2. Importantly, this combination reduced both the tumor burden and the number of pulmonary metastatic nodules. The combination did not cause cumulative toxicity. Together, our findings offer a preclinical proof that by acting on the tumor stroma paclitaxel potentiates the antitumor activity elicited by a targeted delivery of IL2, thereby supporting the use of immunochemotherapy in the treatment of metastatic melanoma.


European Journal of Cancer | 2010

Bortezomib and zoledronic acid on angiogenic and vasculogenic activities of bone marrow macrophages in patients with multiple myeloma

Michele Moschetta; Giulia Di Pietro; Roberto Ria; Antonio Gnoni; Giuseppe Mangialardi; Attilio Guarini; Paolo Ditonno; Pellegrino Musto; Fiorella D’Auria; Maria Rosaria Ricciardi; Franco Dammacco; Domenico Ribatti; Angelo Vacca

Bone marrow neovascularisation supports plasma cell tumour progression in patients with multiple myeloma (MM), and is partially sustained by bone marrow macrophages through their angiogenic and vasculogenic activities. As such, macrophages may be a target for antivascular treatment in MM. Here, we show that bortezomib (BZ) and zoledronic acid (ZOL) display distinct and synergistic inhibitory effects on cell proliferation, adhesion, migration and expression of angiogenic cytokines (i.e.: VEGF, bFGF, HGF and PDGF). Similar effects were found on capillarogenic organisation and expression of vascular markers in cells which became vasculogenic. VEGFR2 and ERK1/2 phosphoactivation as well as NF-kappaB activity were also inhibited. Overall these data provide evidence that the exposure of bone marrow macrophages in MM during the treatment with ZOL and BZ, alone and or in combination, impacts their angiogenic and vasculogenic properties, suggesting that these cells may be considered as a target of both drugs in MM patients.


Blood | 2014

The sialyltransferase ST3GAL6 influences homing and survival in multiple myeloma

Siobhan Glavey; Salomon Manier; Alessandro Natoni; Antonio Sacco; Michele Moschetta; Michaela R. Reagan; Laura Murillo; Ilyas Sahin; Ping Wu; Yuji Mishima; Yunyu Zhang; Weijia Zhang; Gareth J. Morgan; Lokesh Joshi; Aldo M. Roccaro; Irene M. Ghobrial; Michael E. O'Dwyer

Glycosylation is a stepwise procedure of covalent attachment of oligosaccharide chains to proteins or lipids, and alterations in this process, especially increased sialylation, have been associated with malignant transformation and metastasis. The role of altered sialylation in multiple myeloma (MM) cell trafficking has not been previously investigated. In the present study we identified high expression of β-galactoside α-2,3-sialyltransferase, ST3GAL6, in MM cell lines and patients. This gene plays a key role in selectin ligand synthesis in humans through the generation of functional sialyl Lewis X. In MRC IX patients, high expression of this gene is associated with inferior overall survival. In this study we demonstrate that knockdown of ST3GAL6 results in a significant reduction in levels of α-2,3-linked sialic acid on the surface of MM cells with an associated significant reduction in adhesion to MM bone marrow stromal cells and fibronectin along with reduced transendothelial migration in vitro. In support of our in vitro findings, we demonstrate significantly reduced homing and engraftment of ST3GAL6 knockdown MM cells to the bone marrow niche in vivo, along with decreased tumor burden and prolonged survival. This study points to the importance of altered glycosylation, particularly sialylation, in MM cell adhesion and migration.


Cell Reports | 2014

SDF-1 Inhibition Targets the Bone Marrow Niche for Cancer Therapy

Aldo M. Roccaro; Antonio Sacco; Werner Purschke; Michele Moschetta; Klaus Buchner; Christian Maasch; Dirk Zboralski; Stefan Zöllner; Stefan Vonhoff; Yuji Mishima; Patricia Maiso; Michaela R. Reagan; Silvia Lonardi; Marco Ungari; Fabio Facchetti; Dirk Eulberg; Anna Kruschinski; Axel Vater; Giuseppe Rossi; Sven Klussmann; Irene M. Ghobrial

Bone marrow (BM) metastasis remains one of the main causes of death associated with solid tumors as well as multiple myeloma (MM). Targeting the BM niche to prevent or modulate metastasis has not been successful to date. Here, we show that stromal cell-derived factor-1 (SDF-1/CXCL12) is highly expressed in active MM, as well as in BM sites of tumor metastasis and report on the discovery of the high-affinity anti-SDF-1 PEGylated mirror-image l-oligonucleotide (olaptesed-pegol). In vivo confocal imaging showed that SDF-1 levels are increased within MM cell-colonized BM areas. Using in vivo murine and xenograft mouse models, we document that in vivo SDF-1 neutralization within BM niches leads to a microenvironment that is less receptive for MM cells and reduces MM cell homing and growth, thereby inhibiting MM disease progression. Targeting of SDF-1 represents a valid strategy for preventing or disrupting colonization of the BM by MM cells.


Cancer Research | 2015

Metabolic Signature Identifies Novel Targets for Drug Resistance in Multiple Myeloma

Patricia Maiso; Daisy Huynh; Michele Moschetta; Antonio Sacco; Yosra Aljawai; Yuji Mishima; John M. Asara; Aldo M. Roccaro; Alec C. Kimmelman; Irene M. Ghobrial

Drug resistance remains a major clinical challenge for cancer treatment. Multiple myeloma is an incurable plasma cell cancer selectively localized in the bone marrow. The main cause of resistance in myeloma is the minimal residual disease cells that are resistant to the original therapy, including bortezomib treatment and high-dose melphalan in stem cell transplant. In this study, we demonstrate that altered tumor cell metabolism is essential for the regulation of drug resistance in multiple myeloma cells. We show the unprecedented role of the metabolic phenotype in inducing drug resistance through LDHA and HIF1A in multiple myeloma, and that specific inhibition of LDHA and HIF1A can restore sensitivity to therapeutic agents such as bortezomib and can also inhibit tumor growth induced by altered metabolism. Knockdown of LDHA can restore sensitivity of bortezomib resistance cell lines while gain-of-function studies using LDHA or HIF1A induced resistance in bortezomib-sensitive cell lines. Taken together, these data suggest that HIF1A and LDHA are important targets for hypoxia-driven drug resistance. Novel drugs that regulate metabolic pathways in multiple myeloma, specifically targeting LDHA, can be beneficial to inhibit tumor growth and overcome drug resistance.


British Journal of Pharmacology | 2014

Therapeutic targeting of the mTOR-signalling pathway in cancer: benefits and limitations

Michele Moschetta; Antonia Reale; Carolina Marasco; Angelo Vacca; Maria Rosaria Carratù

The mammalian target of rapamycin (mTOR) plays an important role in the regulation of protein translation, cell growth and metabolism. The mTOR protein forms two distinct multi‐subunit complexes: mTORC1 and mTORC2. The mTORC1 complex is activated by diverse stimuli, such as growth factors, nutrients, energy and stress signals; and essential signalling pathways, such as PI3K and MAPK, in order to control cell growth, proliferation and survival. mTORC1 also activates S6K1 and 4EBP1, which are involved in mRNA translation. The mTORC2 complex is resistant to rapamycin inhibitory activity and is generally insensitive to nutrient‐ and energy‐dependent signals. It activates PKC‐α and Akt and regulates the actin cytoskeleton. Deregulation of the mTOR‐signalling pathway (PI3K amplification/mutation, PTEN loss of function, Akt overexpression, and S6K1, 4EBP1 and eIF4E overexpression) is common in cancer, and alterations in components of the mTOR pathway have a major role in tumour progression. Therefore, mTOR is an appealing therapeutic target in many tumours. Here we summarize the upstream regulators and downstream effectors of the mTORC1 and mTORC2 pathways, the role of mTOR in cancer, and the potential therapeutic values and issues related to the novel agents targeting the mTOR‐signalling pathway.

Collaboration


Dive into the Michele Moschetta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge