Denise Toscani

Verified @unipr.it

42

Scopus Publications

Scopus Publications

  • Identification of PSMB4 and PSMD4 as novel target genes correlated with 1q21 amplification in patients with smoldering myeloma and multiple myeloma
    Jessica Burroughs Garcia, Paola Storti, Nicolas Thomas Iannozzi, Valentina Marchica, Luca Agnelli, Denise Toscani, Valentina Franceschi, Giannalisa Todaro, Gabriella Sammarelli, Laura Notarfranchi,et al.

    Ferrata Storti Foundation (Haematologica)
    Not available.

  • The impact of CD56 expression in smoldering myeloma patients on early progression
    Laura Notarfranchi, Roberta Segreto, Rosanna Vescovini, Anna Benedetta Dalla Palma, Valentina Marchica, Jessica Burroughs‐Garcia, Denise Toscani, Giannalisa Todaro, Vincenzo Raimondi, Nicolas Thomas Iannozzi,et al.

    Wiley
    Our study identified the absence of CD56 as a possible risk factor for early progression in SMM patients at 36 months. Furthermore, our results showed that the lack of CD56 expression could be a factor for a more aggressive disease regardless of the tumoral burden. For this reason, these patients should have a closer follow-up in the first years after the diagnosis of SMM and the expression of CD56 could potentially be used as a biomarker and could be incorporated in future prognostic or predictive scores. This article is protected by copyright. All rights reserved.

  • The Metabolic Features of Osteoblasts: Implications for Multiple Myeloma (MM) Bone Disease
    Oxana Lungu, Denise Toscani, Jessica Burroughs-Garcia, and Nicola Giuliani

    MDPI AG
    The study of osteoblast (OB) metabolism has recently received increased attention due to the considerable amount of energy used during the bone remodeling process. In addition to glucose, the main nutrient for the osteoblast lineages, recent data highlight the importance of amino acid and fatty acid metabolism in providing the fuel necessary for the proper functioning of OBs. Among the amino acids, it has been reported that OBs are largely dependent on glutamine (Gln) for their differentiation and activity. In this review, we describe the main metabolic pathways governing OBs’ fate and functions, both in physiological and pathological malignant conditions. In particular, we focus on multiple myeloma (MM) bone disease, which is characterized by a severe imbalance in OB differentiation due to the presence of malignant plasma cells into the bone microenvironment. Here, we describe the most important metabolic alterations involved in the inhibition of OB formation and activity in MM patients.

  • Molecular Features of the Mesenchymal and Osteoblastic Cells in Multiple Myeloma
    Nicolas Thomas Iannozzi, Valentina Marchica, Denise Toscani, Jessica Burroughs Garcìa, Nicola Giuliani, and Paola Storti

    MDPI AG
    Multiple myeloma (MM) is a monoclonal gammopathy characterized by biological heterogeneity and unregulated proliferation of plasma cells (PCs) in bone marrow (BM). MM is a multistep process based on genomic instability, epigenetic dysregulation and a tight cross-talk with the BM microenvironment that plays a pivotal role supporting the proliferation, survival, drug-resistance and homing of PCs. The BM microenvironment consists of a hematopoietic and a non-hematopoietic compartment, which cooperate to create a tumor environment. Among the non-hematopoietic component, mesenchymal stromal cells (MSCs) and osteoblasts (OBs) appear transcriptionally and functionally different in MM patients compared to healthy donors (HDs) and to patients with pre-malignant monoclonal gammopathies. Alterations of both MSCs and OBs underly the osteolytic lesions that characterize myeloma-associated bone disease. In this review, we will discuss the different characteristics of MSCs and OBs in MM patients, analyzing the transcriptome, the deregulated molecular pathways and the role performed by miRNAs and exosome in the pathophysiology of MM.

  • Metabolic features of myeloma cells in the context of bone microenvironment: Implication for the pathophysiology and clinic of myeloma bone disease
    Vincenzo Raimondi, Denise Toscani, Valentina Marchica, Jessica Burroughs-Garcia, Paola Storti, and Nicola Giuliani

    Frontiers Media SA
    Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of malignant plasma cells (PCs) into the bone marrow (BM). The complex interaction between the BM microenvironment and MM PCs can lead to severe impairment of bone remodeling. Indeed, the BM microenvironment exerts a critical role in the survival of malignant PCs. Growing evidence indicates that MM cells have several metabolic features including enhanced glycolysis and an increase in lactate production through the upregulation of glucose transporters and enzymes. More recently, it has been reported that MM cells arehighly glutamine addicted. Interestingly, these metabolic changes in MM cells may affect BM microenvironment cells by altering the differentiation process of osteoblasts from mesenchymal stromal cells. The identification of glutamine metabolism alterations in MM cells and bone microenvironment may provide a rationale to design new therapeutic approaches and diagnostic tools. The osteolytic lesions are the most frequent clinical features in MM patients, often characterized by pathological fractures and acute pain. The use of the newer imaging techniques such as Magnetic Resonance Imaging (MRI) and combined Positron Emission Tomography (PET) and Computerized Tomography (CT) has been introduced into clinical practice to better define the skeletal involvement. Currently, the PET/CT with 18F-fluorodeoxyglucose (FDG) is the diagnostic gold standard to detect active MM bone disease due to the high glycolytic activity of MM cells. However, new tracers are actively under investigation because a portion of MM patients remains negative at the skeletal level by 18F-FDG. In this review, we will summarize the existing knowledge on the metabolic alterations of MM cells considering their impact on the BM microenvironment cells and particularly in the subsequent formation of osteolytic bone lesions. Based on this, we will discuss the identification of possible new druggable targets and the use of novel metabolic targets for PET imaging in the detection of skeletal lesions, in the staging and treatment response of MM patients.

  • A personalized molecular approach in multiple myeloma: the possible use of RAF/RAS/MEK/ERK and BCL-2 inhibitors
    Vincenzo Raimondi, Nicolas Thomas Iannozzi, Jessica Burroughs-Garcìa, Denise Toscani, Paola Storti, and Nicola Giuliani

    Open Exploration Publishing
    Multiple myeloma (MM) is a blood cancer that derives from plasma cells (PCs), which will accumulate in the bone marrow (BM). Over time, several drugs have been developed to treat this disease that is still uncurable. The therapies used to treat the disease target immune activity, inhibit proteasome activity, and involve the use of monoclonal antibodies. However, MM is a highly heterogeneous disease, in fact, there are several mutations in signaling pathways that are particularly important for MM cell biology and that are possible therapeutic targets. Indeed, some studies suggest that MM is driven by mutations within the rat sarcoma virus (RAS) signaling cascade, which regulates cell survival and proliferation. The RAS/proto-oncogene, serine/threonine kinase (RAF)/mitogen-activated extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK signaling pathway is deregulated in several cancers, for which drugs have been developed to inhibit these pathways. In addition to the signaling pathways, the disease implements mechanisms to ensure the survival and consequently a high replicative capacity. This strategy consists in the deregulation of apoptosis. In particular, some cases of MM show overexpression of anti-apoptotic proteins belonging to the B cell lymphoma 2 (BCL-2) family that represent a possible druggable target. Venetoclax is an anti-BCL-2 molecule used in hematological malignancies that may be used in selected MM patients based on their molecular profile. We focused on the possible effects in MM of off-label drugs that are currently used for other cancers with the same molecular characteristics. Their use, combined with the current treatments, could be a good strategy against MM.

  • Immune response to SARS-CoV-2 mRNA vaccination and booster dose in patients with multiple myeloma and monoclonal gammopathies: impact of Omicron variant on the humoral response
    Paola Storti, Valentina Marchica, Rosanna Vescovini, Valentina Franceschi, Luca Russo, Laura Notarfranchi, Vincenzo Raimondi, Denise Toscani, Jessica Burroughs Garcia, Federica Costa,et al.

    Informa UK Limited
    ABSTRACT The humoral and cellular response to SARS-CoV-2 mRNA full vaccination and booster dose as well as the impact of the spike variants, including Omicron, are still unclear in patients with multiple myeloma (MM) and those with pre-malignant monoclonal gammopathies. In this study, involving 40 patients, we found that MM patients with relapsed-refractory disease (MMR) had reduced spike-specific antibody levels and neutralizing titers after SARS-CoV-2 vaccination. The five analyzed variants, remarkably Omicron, had a significant negative impact on the neutralizing ability of the vaccine-induced antibodies in all patients with MM and smoldering MM. Moreover, lower spike-specific IL-2-producing CD4+ T cells and reduced cytotoxic spike-specific IFN-γ and TNF-α-producing CD8+ T cells were found in MM patients as compared to patients with monoclonal gammopathy of undetermined significance. We found that a heterologous booster immunization improved SARS-CoV-2 spike humoral and cellular responses in newly diagnosed MM (MMD) patients and in most, but not all, MMR patients. After the booster dose, a significant increase of the neutralizing antibody titers against almost all the analyzed variants was achieved in MMD. However, in MMR patients, Omicron retained a negative impact on neutralizing ability, suggesting further approaches to potentiating the effectiveness of SARS-CoV-2 vaccination in these patients.

  • [<sup>18</sup>F](2S,4R)-4-Fluoroglutamine as a New Positron Emission Tomography Tracer in Myeloma
    Silvia Valtorta, Denise Toscani, Martina Chiu, Andrea Sartori, Angela Coliva, Arianna Brevi, Giuseppe Taurino, Matteo Grioni, Livia Ruffini, Federica Vacondio,et al.

    Frontiers Media SA
    The high glycolytic activity of multiple myeloma (MM) cells is the rationale for use of Positron Emission Tomography (PET) with 18F-fluorodeoxyglucose ([18F]FDG) to detect both bone marrow (BM) and extramedullary disease. However, new tracers are actively searched because [18F]FDG-PET has some limitations and there is a portion of MM patients who are negative. Glutamine (Gln) addiction has been recently described as a typical metabolic feature of MM cells. Yet, the possible exploitation of Gln as a PET tracer in MM has never been assessed so far and is investigated in this study in preclinical models. Firstly, we have synthesized enantiopure (2S,4R)-4-fluoroglutamine (4-FGln) and validated it as a Gln transport analogue in human MM cell lines, comparing its uptake with that of 3H-labelled Gln. We then radiosynthesized [18F]4-FGln, tested its uptake in two different in vivo murine MM models, and checked the effect of Bortezomib, a proteasome inhibitor currently used in the treatment of MM. Both [18F]4-FGln and [18F]FDG clearly identified the spleen as site of MM cell colonization in C57BL/6 mice, challenged with syngeneic Vk12598 cells and assessed by PET. NOD.SCID mice, subcutaneously injected with human MM JJN3 cells, showed high values of both [18F]4-FGln and [18F]FDG uptake. Bortezomib significantly reduced the uptake of both radiopharmaceuticals in comparison with vehicle at post treatment PET. However, a reduction of glutaminolytic, but not of glycolytic, tumor volume was evident in mice showing the highest response to Bortezomib. Our data indicate that [18F](2S,4R)-4-FGln is a new PET tracer in preclinical MM models, yielding a rationale to design studies in MM patients.

  • Role of 1q21 in multiple myeloma: From pathogenesis to possible therapeutic targets
    Jessica Burroughs Garcìa, Rosa Alba Eufemiese, Paola Storti, Gabriella Sammarelli, Luisa Craviotto, Giannalisa Todaro, Denise Toscani, Valentina Marchica, and Nicola Giuliani

    MDPI AG
    Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) in the bone marrow (BM). The amplification of 1q21 is one of the most common cytogenetic abnormalities occurring in around 40% of de novo patients and 70% of relapsed/refractory MM. Patients with this unfavorable cytogenetic abnormality are considered to be high risk with a poor response to standard therapies. The gene(s) driving amplification of the 1q21 amplicon has not been fully studied. A number of clear candidates are under investigation, and some of them (IL6R, ILF2, MCL-1, CKS1B and BCL9) have been recently proposed to be potential drivers of this region. However, much remains to be learned about the biology of the genes driving the disease progression in MM patients with 1q21 amp. Understanding the mechanisms of these genes is important for the development of effective targeted therapeutic approaches to treat these patients for whom effective therapies are currently lacking. In this paper, we review the current knowledge about the pathological features, the mechanism of 1q21 amplification, and the signal pathway of the most relevant candidate genes that have been suggested as possible therapeutic targets for the 1q21 amplicon.

  • The role of proteasome inhibitors in multiple myeloma bone disease and bone metastasis: Effects on osteoblasts and osteocytes
    Denise Toscani, Luisa Craviotto, and Nicola Giuliani

    MDPI AG
    The alterations of bone remodeling are typical of multiple myeloma (MM) patients where the uncoupled and unbalanced bone remodeling caused the onset of osteolytic lesions. Moreover, bone metastasis occurs in the majority of patients with breast and prostate cancer. Skeletal-related events negatively impact on quality of life by increasing the vulnerability to fractures. Several bone-targeting treatments have been developed to control bone pain and pathological fractures, including bisphosphonates and Denosumab. Nevertheless, these agents act by inhibiting osteoclast activity but do not improve bone formation. Proteasome inhibitors (PIs) have shown bone anabolic effects and encouraging results in stimulating osteoblast differentiation and bone healing. Among these, the first-in-class bortezomib and the second-generation PIs, carfilzomib, and ixazomib regulate the bone remodeling process by controlling the degradation of several bone proteins. PIs have been recently proven to also be efficacious in blocking MM-induced osteocyte death providing new possible therapeutic use in the management of bone loss. PIs have significant side effects that limit their use as bone anabolic strategy. Multiple alternative approaches have been made. The conjugation of PIs with bisphosphonates, which can target them to bone, showed good results in terms of bone anabolic activity. However, the clinical implications of these effects require further investigations.

  • PD-L1/PD-1 Pattern of Expression Within the Bone Marrow Immune Microenvironment in Smoldering Myeloma and Active Multiple Myeloma Patients
    Federica Costa, Rosanna Vescovini, Valentina Marchica, Paola Storti, Laura Notarfranchi, Benedetta Dalla Palma, Denise Toscani, Jessica Burroughs-Garcia, Maria Teresa Catarozzo, Gabriella Sammarelli,et al.

    Frontiers Media SA
    BackgroundThe PD-1/PD-L1 axis has recently emerged as an immune checkpoint that controls antitumor immune responses also in hematological malignancies. However, the use of anti-PD-L1/PD-1 antibodies in multiple myeloma (MM) patients still remains debated, at least in part because of discordant literature data on PD-L1/PD-1 expression by MM cells and bone marrow (BM) microenvironment cells. The unmet need to identify patients which could benefit from this therapeutic approach prompts us to evaluate the BM expression profile of PD-L1/PD-1 axis across the different stages of the monoclonal gammopathies.MethodsThe PD-L1/PD-1 axis was evaluated by flow cytometry in the BM samples of a total cohort of 141 patients with monoclonal gammopathies including 24 patients with Monoclonal Gammopathy of Undetermined Significance (MGUS), 38 patients with smoldering MM (SMM), and 79 patients with active MM, including either newly diagnosed or relapsed-refractory patients. Then, data were correlated with the main immunological and clinical features of the patients.ResultsFirst, we did not find any significant difference between MM and SMM patients in terms of PD-L1/PD-1 expression, on both BM myeloid (CD14+) and lymphoid subsets. On the other hand, PD-L1 expression by CD138+ MM cells was higher in both SMM and MM as compared to MGUS patients. Second, the analysis on the total cohort of MM and SMM patients revealed that PD-L1 is expressed at higher level in CD14+CD16+ non-classical monocytes compared with classical CD14+CD16− cells, independently from the stage of disease. Moreover, PD-L1 expression on CD14+ cells was inversely correlated with BM serum levels of the anti-tumoral cytokine, IL-27. Interestingly, relapsed MM patients showed an inverted CD4+/CD8+ ratio along with high levels of pro-tumoral IL-6 and a positive correlation between %CD14+PD-L1+ and %CD8+PD-1+ cells as compared to both SMM and newly diagnosed MM patients suggesting a highly compromised immune-compartment with low amount of CD4+ effector cells.ConclusionsOur data indicate that SMM and active MM patients share a similar PD-L1/PD-1 BM immune profile, suggesting that SMM patients could be an interesting target for PD-L1/PD-1 inhibition therapy, in light of their less compromised and more responsive immune-compartment.

  • Myeloma cells deplete bone marrow glutamine and inhibit osteoblast differentiation limiting asparagine availability
    Martina Chiu, Denise Toscani, Valentina Marchica, Giuseppe Taurino, Federica Costa, Massimiliano G. Bianchi, Roberta Andreoli, Valentina Franceschi, Paola Storti, Jessica Burroughs-Garcia,et al.

    MDPI AG
    Multiple myeloma (MM) cells consume huge amounts of glutamine and, as a consequence, the amino acid concentration is lower-than-normal in the bone marrow (BM) of MM patients. Here we show that MM-dependent glutamine depletion induces glutamine synthetase in stromal cells, as demonstrated in BM biopsies of MM patients, and reproduced in vitro by co-culturing human mesenchymal stromal cells (MSCs) with MM cells. Moreover, glutamine depletion hinders osteoblast differentiation of MSCs, which is also severely blunted by the spent, low-glutamine medium of MM cells, and rescued by glutamine restitution. Glutaminase and the concentrative glutamine transporter SNAT2 are induced during osteoblastogenesis in vivo and in vitro, and both needed for MSCs differentiation, pointing to enhanced the requirement for the amino acid. Osteoblastogenesis also triggers the induction of glutamine-dependent asparagine synthetase (ASNS), and, among non-essential amino acids, asparagine rescues differentiation of glutamine-starved MSCs, by restoring the transcriptional profiles of differentiating MSCs altered by glutamine starvation. Thus, reduced asparagine availability provides a mechanistic link between MM-dependent Gln depletion in BM and impairment of osteoblast differentiation. Inhibition of Gln metabolism in MM cells and supplementation of asparagine to stromal cells may, therefore, constitute novel approaches to prevent osteolytic lesions in MM.

  • Novel approaches to improve myeloma cell killing by monoclonal antibodies
    Paola Storti, Federica Costa, Valentina Marchica, Jessica Burroughs-Garcia, Benedetta dalla Palma, Denise Toscani, Rosa Alba Eufemiese, and Nicola Giuliani

    MDPI AG
    The monoclonal antibodies (mAbs) have significantly changed the treatment of multiple myeloma (MM) patients. However, despite their introduction, MM remains an incurable disease. The mAbs currently used for MM treatment were developed with different mechanisms of action able to target antigens, such as cluster of differentiation 38 (CD38) and SLAM family member 7 (SLAMF7) expressed by both, MM cells and the immune microenvironment cells. In this review, we focused on the mechanisms of action of the main mAbs approved for the therapy of MM, and on the possible novel approaches to improve MM cell killing by mAbs. Actually, the combination of anti-CD38 or anti-SLAMF7 mAbs with the immunomodulatory drugs significantly improved the clinical effect in MM patients. On the other hand, pre-clinical evidence indicates that different approaches may increase the efficacy of mAbs. The use of trans-retinoic acid, the cyclophosphamide or the combination of anti-CD47 and anti-CD137 mAbs have given the rationale to design these types of combinations therapies in MM patients in the future. In conclusion, a better understanding of the mechanism of action of the mAbs will allow us to develop novel therapeutic approaches to improve their response rate and to overcome their resistance in MM patients.

  • CD14<sup>+</sup>CD16<sup>+</sup> monocytes are involved in daratumumab-mediated myeloma cells killing and in anti-CD47 therapeutic strategy
    Paola Storti, Rosanna Vescovini, Federica Costa, Valentina Marchica, Denise Toscani, Benedetta Dalla Palma, Luisa Craviotto, Fabio Malavasi, and Nicola Giuliani

    Wiley
    A deep elucidation of the mechanisms of action of anti‐CD38 monoclonal antibodies (mAbs), such as daratumumab (DARA), is required to identify patients with multiple myeloma (MM) who are more responsive to this treatment. In the present study, an autologous ex vivo approach was established, focussing on the role of the monocytes in the anti CD38‐mediated killing of MM cells. In bone marrow (BM) samples from 29 patients with MM, we found that the ratio between monocytes (CD14+) and MM cells (CD138+) influences the response to DARA. Further, the exposure of the BM samples to DARA is followed by the formation of a CD138+CD14+ double‐positive (DP) population, that quantitatively correlates with the anti‐MM cells killing. These effects were dependent on the presence of a CD14+CD16+ monocyte subset and on high CD16 expression levels. Lastly, the addition of a mAb neutralising the CD47/signal‐regulatory protein α (SIRPα) axis was able to increase the killing mediated by DARA. The effects were observed only in coincidence with high CD14+:CD138+ ratio, with a significant presence of the DP population and were correlated with CD16 expression. In conclusion, the present study underlines the critical role of the CD16+ monocytes in DARA anti‐MM killing effects and gives a rationale to test the combination of an anti‐CD47 mAb with anti‐CD38 mAbs.

  • Bovine pestivirus is a new alternative virus for multiple myeloma oncolytic virotherapy
    Valentina Marchica, Valentina Franceschi, Rosanna Vescovini, Paola Storti, Emanuela Vicario, Denise Toscani, Alessia Zorzoli, Irma Airoldi, Benedetta Dalla Palma, Nicoletta Campanini,et al.

    Springer Science and Business Media LLC
    Abstract Background The oncolytic viruses have shown promising results for the treatment of multiple myeloma. However, the use of human viruses is limited by the patients’ antiviral immune response. In this study, we investigated an alternative oncolytic strategy using non-human pathogen viruses as the bovine viral diarrhea virus (BVDV) that were able to interact with CD46. Methods We treated several human myeloma cell lines and non-myeloma cell lines with BVDV to evaluate the expression of CD46 and to study the effect on cell viability by flow cytometry. The possible synergistic effect of bortezomib in combination with BVDV was also tested. Moreover, we infected the bone marrow mononuclear cells obtained from myeloma patients and we checked the BVDV effect on different cell populations, defined by CD138, CD14, CD3, CD19, and CD56 expression evaluated by flow cytometry. Finally, the in vivo BVDV effect was tested in NOD-SCID mice injected subcutaneously with myeloma cell lines. Results Human myeloma cells were selectively sensitive to BVDV treatment with an increase of cell death and, consequently, of apoptotic markers. Consistently, bone marrow mononuclear cells isolated from myeloma patients treated with BVDV, showed a significant selective decrease of the percentage of viable CD138+ cells. Interestingly, bortezomib pre-treatment significantly increased the cytotoxic effect of BVDV in myeloma cell lines with a synergistic effect. Finally, the in vitro data were confirmed in an in vivo myeloma mouse model showing that BVDV treatment significantly reduced the tumoral burden compared to the vehicle. Conclusions Overall, our data indicate, for the first time, a direct oncolytic effect of the BVDV in human myeloma cells suggesting its possible use as novel alternative anti-myeloma virotherapy strategy.

  • Novel targets for the treatment of relapsing multiple myeloma
    Nicola Giuliani, Fabrizio Accardi, Valentina Marchica, Benedetta Dalla Palma, Paola Storti, Denise Toscani, Emanuela Vicario, and Fabio Malavasi

    Informa UK Limited
    ABSTRACT Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.

  • Bone marrow CX3CL1/Fractalkine is a new player of the pro-angiogenic microenvironment in multiple myeloma patients
    Valentina Marchica, Denise Toscani, Anna Corcione, Marina Bolzoni, Paola Storti, Rosanna Vescovini, Elisa Ferretti, Benedetta Dalla Accardi, Emanuela Vicario, Fabrizio Accardi,et al.

    MDPI AG
    C-X3-C motif chemokine ligand 1 (CX3CL1)/fractalkine is a chemokine released after cleavage by two metalloproteases, ADAM metallopeptidase domain 10 (ADAM10) and ADAM metallopeptidase domain 17 (ADAM17), involved in inflammation and angiogenesis in the cancer microenvironment. The role of the CX3CL1/ C-X3-C motif chemokine receptor 1(CX3CR1) axis in the multiple myeloma (MM) microenvironment is still unknown. Firstly, we analyzed bone marrow (BM) plasma levels of CX3CL1 in 111 patients with plasma cell disorders including 70 with active MM, 25 with smoldering myeloma (SMM), and 16 with monoclonal gammopathy of undetermined significance (MGUS). We found that BM CX3CL1 levels were significantly increased in MM patients compared to SMM and MGUS and correlated with BM microvessel density. Secondly, we explored the source of CX3CL1 in MM and BM microenvironment cells. Primary CD138+ cells did not express CXC3L1 but up-regulated its production by endothelial cells (ECs) through the involvement of tumor necrosis factor alpha (TNFα). Lastly, we demonstrated the presence of CX3CR1 on BM CD14+CD16+ monocytes of MM patients and on ECs, but not on MM cells. The role of CX3CL1 in MM-induced angiogenesis was finally demonstrated in both in vivo chick embryo chorioallantoic membrane and in vitro angiogenesis assays. Our data indicate that CX3CL1, present at a high level in the BM of MM patients, is a new player of the MM microenvironment involved in MM-induced angiogenesis.

  • Multiple myeloma-derived exosomes are enriched of amphiregulin (AREG) and activate the epidermal growth factor pathway in the bone microenvironment leading to osteoclastogenesis
    Stefania Raimondo, Laura Saieva, Emanuela Vicario, Marzia Pucci, Denise Toscani, Mauro Manno, Samuele Raccosta, Nicola Giuliani, and Riccardo Alessandro

    Springer Science and Business Media LLC
    BackgroundMultiple myeloma (MM) is a clonal plasma cell malignancy associated with osteolytic bone disease. Recently, the role of MM-derived exosomes in the osteoclastogenesis has been demonstrated although the underlying mechanism is still unknown. Since exosomes-derived epidermal growth factor receptor ligands (EGFR) are involved in tumor-associated osteolysis, we hypothesize that the EGFR ligand amphiregulin (AREG) can be delivered by MM-derived exosomes and participate in MM-induced osteoclastogenesis.MethodsExosomes were isolated from the conditioned medium of MM1.S cell line and from bone marrow (BM) plasma samples of MM patients. The murine cell line RAW264.7 and primary human CD14+ cells were used as osteoclast (OC) sources.ResultsWe found that AREG was specifically enriched in exosomes from MM samples and that exosomes-derived AREG led to the activation of EGFR in pre-OC, as showed by the increase of mRNA expression of its downstream SNAIL in both RAW264.7 and CD14+ cells. The presence of neutralizing anti-AREG monoclonal antibody (mAb) reverted this effect. Consequently, we showed that the effect of MM-derived exosomes on osteoclast differentiation was inhibited by the pre-treatment of exosomes with anti-AREG mAb. In addition, we demonstrated the ability of MM-derived AREG-enriched exosomes to be internalized into human mesenchymal stromal cells (MSCs) blocking osteoblast (OB) differentiation, increasing MM cell adhesion and the release of the pro-osteoclastogenic cytokine interleukin-8 (IL8). Accordingly, anti-AREG mAb inhibited the release of IL8 by MSCs suggesting that both direct and indirect effects are responsible for AREG-enriched exosomes involvement on MM-induced osteoclastogenesis.ConclusionsIn conclusion, our data indicate that AREG is packed into MM-derived exosomes and implicated in OC differentiation through an indirect mechanism mediated by OBs.

  • The transcriptomic profile of CD138+ cells from patients with early progression from smoldering to active multiple myeloma remains substantially unchanged
    Paola Storti, Luca Agnelli, Benedetta dalla Palma, Katia Todoerti, Valentina Marchica, Fabrizio Accardi, Gabriella Sammarelli, Federica Deluca, Denise Toscani, Federica Costa,et al.

    Ferrata Storti Foundation (Haematologica)
    Smoldering myeloma (SMM) is a pre-malignant monoclonal gammopathy with an annual risk of about 10% of progressing to active multiple myeloma (MM). The International Myeloma Working Group (IMWG) has recently updated the diagnostic criteria for SMM. The previously defined “ultra high-risk” SMM (characterized by specific biomarkers associated with a greater than 80% risk of progression to symptomatic MM within 2 years) has now been incorporated in active MM. SMM is biologically heterogeneous and encompasses a condition with a very low rate of progression to symptomatic MM, behaving similarly to monoclonal gammopathy of uncertain significance, as well as a disease with acquired organ damage and progression to active MM within 5 years from diagnosis.

  • The link between bone microenvironment and immune cells in multiple myeloma: Emerging role of CD38
    Marina Bolzoni, Denise Toscani, Federica Costa, Emanuela Vicario, Franco Aversa, and Nicola Giuliani

    Elsevier BV
    The relationship between bone and immune cells is well established both in physiological and pathological conditions. Multiple myeloma (MM) is a plasma cell malignancy characterized by an increase of number and activity of osteoclasts (OCLs) and a decrease of osteoblasts (OBs). These events are responsible for bone lesions of MM patients. OCLs support MM cells survival in vitro and in vivo. Recently, the possible role of OCLs as immunosuppressive cells in the MM BM microenvironment has been underlined. OCLs protect MM cells against T cell-mediated cytotoxicity through the expression of several molecules including programmed death-ligand (PD-L) 1, galectin (Gal) 9, CD200, and indoleamine-2,3-dioxygenase (IDO). Among the molecules that could be involved in the link between immune-microenvironment and osteoclastogenesis the role of CD38 has been hypothesized. CD38 is a well-known adhesion molecule and an ectoenzyme highly expressed by MM cells. Moreover, CD38 is expressed by OCLs and at the surface level on OCL precursors. Targeting CD38 with monoclonal antibodies showed inhibition of both osteoclastogenesis and OCL-mediated suppression of T cell function. This review elucidates this evidence indicating that osteoclastogenesis affect MM immune-microenvironment being a potential target to improve anti-MM immunity and to ameliorate bone disease.

  • Bone marrow Dikkopf-1 levels are a new independent risk factor for progression in patients with smouldering myeloma
    Benedetta Dalla Palma, Valentina Marchica, Mario Pedrazzoni, Fabrizio Accardi, Laura Notarfranchi, Matteo Goldoni, Federica De Luca, Federica Costa, Paola Storti, Denise Toscani,et al.

    Wiley
    The diagnostic criteria for multiple myeloma (MM) have been recently updated, including patients previously defined highrisk smouldering MM (SMM) among those with active MM in the presence of biomarkers of malignancy (Rajkumar et al, 2014). Several models have recently been developed in order to stratify SMM patients based on their progression rate (Rajkumar et al, 2015) (Dhodapkar et al, 2014). Nevertheless, the definition of SMM still includes patients with different risk of progression to active MM; thus, new potential markers of progression need to be defined in order to identify those patients with higher risk for progression that could benefit from an early treatment including lenalidomide (Mateos et al, 2013). Recently, we analysed bone marrow (BM) levels of several cytokines and chemokines involved in MM-induced bone disease, finding that BM levels of Activin A, C-C motif chemokine ligand 20 (CCL20) and Dickkopf 1 (DKK-1) were significantly different among patients with monoclonal gammopathy of uncertain significance (MGUS), SMM and MM (Palma et al, 2016). Interestingly, of these cytokines, recent pre-clinical data indicate that DKK-1 is also involved in tumoural progression through the increase of the number of myeloid-derived suppressor cells (MDSCs) (D’Amico et al, 2016). Thus, in this study we focused on the potential role of these soluble factors in the progression of SMM patients. We identified 89 patients with a diagnosis of SMM according to the 2014 revised International Myeloma Working Group (IMWG) criteria (Rajkumar et al, 2014). Activin A, CCL20 and DKK-1 levels were evaluated on BM plasma using commercially available enzyme-linked immunosorbent assay (ELISA) kits. The main characteristics of the cohort of patients are reported in Table SI. Details on methods (study cohort, sample collection, cytogenetic and fluorescence in situ hybridisation analysis, ELISA assays and statistical analysis) are reported in Data S1. With a median follow-up of 43 months, 25 out of the 89 patients (28%) progressed to active MM. We firstly analysed the correlation between main clinical features in progressed and not progressed SMM patients, finding that, as previously reported (Kyle et al, 2007; Perez-Persona et al, 2007), percentage of BM plasma cells (BMPCs), entity of serum-M protein (considered either as continuous variables and ≥ vs. < 30 g/l) and immunoparesis were significantly correlated with progression to active MM (P < 0 0001, P < 0 0001, P = 0 022 and P = 0 005, respectively, by univariate Cox regression analysis). Serum free light chain (FLC) levels and FLC ratio were not available in all patients and consequently we did not find a statistically significant relationship with progression. However, two recent studies (Waxman et al, 2015; Sorrig et al, 2016) observed that an abnormal FLC ratio is not considerable as a stand-alone high risk factor for progression to active MM in SMM patients. Regarding the BM levels of the cytokines analysed, Activin A and CCL20 BM median levels were not different between the two populations (Table SII). On the other hand, DKK-1 BM levels were found to be statistically significant different among progressed versus not progressed patients (median levels: 1267 vs. 778 7 pg/ml) (Fig 1A); these results were confirmed by comparing DKK-1 levels normalized according to total protein concentration in each sample (DKK-1 BM median levels: 15 40 vs. 9 04 pg/mg, P < 0 0001). Univariate Cox regression analysis confirmed a statistically significant relationship between DKK-1 BM levels (expressed either as pg/ ml or pg/mg) and progression to active MM (P = 0 001 and P = 0 001 respectively) (Fig 1A and Table SII) but not with the presence of osteolysis at the progression (data not shown). Interestingly, we did not find a significant correlation between BM DKK-1 levels and the percentage of BMPCs infiltration (P = 0 112 by Spearman’s correlation; data not shown), suggesting that DKK-1 could derive not only from MM cells but also from the BM microenvironment. Consistent with this, D’Amico et al (2016) showed that stromaderived, rather than tumour-derived, DKK-1 targets betacatenin in MDSCs, exerting immunosuppressive effects during tumour progression.

  • Role of osteocytes in myeloma bone disease: Anti-sclerostin antibody as new therapeutic strategy
    Denise Toscani, Marina Bolzoni, Marzia Ferretti, Carla Palumbo, and Nicola Giuliani

    Frontiers Media SA
    Osteocytes are terminally differentiated cells of the osteoblast lineage. They are involved in the regulation of bone remodeling by increasing osteoclast formation or decreasing bone formation by the secretion of the osteoblast inhibitor sclerostin. Monoclonal antibody anti-sclerostin, Romosozumab, has been developed and tested in clinical trials in patients with osteoporosis. In the last years, the role of osteocytes in the development of osteolytic bone lesions that occurs in multiple myeloma, have been underlined. Myeloma cells increase osteocyte death through the up-regulation of both apoptosis and autophagy that, in turn, triggers osteoclast formation, and activity. When compared to healthy controls, myeloma patients with bone disease have higher osteocyte cell death, but the treatment with proteasome inhibitor bortezomib has been shown to maintain osteocyte viability. In preclinical mouse models of multiple myeloma, treatment with blocking anti-sclerostin antibody increased osteoblast numbers and bone formation rate reducing osteolytic bone lesions. Moreover, the combination of anti-sclerostin antibody and the osteoclast inhibitor zoledronic acid increased bone mass and fracture resistance synergistically. However, anti-sclerostin antibody did not affect tumor burden in vivo or the efficacy of anti-myeloma drugs in vitro. Nevertheless, the combination therapy of anti-sclerostin antibody and the proteasome inhibitor carfilzomib, displayed potent anti-myeloma activity as well as positive effects on bone disease in vivo. In conclusion, all these data suggest that osteocytes are involved in myeloma bone disease and may be considered a novel target for the use of antibody-mediated anti-sclerostin therapy also in multiple myeloma patients.

  • Growth factor independence 1 expression in myeloma cells enhances their growth, survival, and osteoclastogenesis
    Daniela N Petrusca, Denise Toscani, Feng-Ming Wang, Cheolkyu Park, Colin D Crean, Judith L Anderson, Silvia Marino, Khalid S Mohammad, Dan Zhou, Rebecca Silbermann,et al.

    Springer Science and Business Media LLC
    BackgroundIn spite of major advances in treatment, multiple myeloma (MM) is currently an incurable malignancy due to the emergence of drug-resistant clones. We previously showed that MM cells upregulate the transcriptional repressor, growth factor independence 1 (Gfi1), in bone marrow stromal cells (BMSCs) that induces prolonged inhibition of osteoblast differentiation. However, the role of Gfi1 in MM cells is unknown.MethodsHuman primary CD138+ and BMSC were purified from normal donors and MM patients’ bone marrow aspirates. Gfi1 knockdown and overexpressing cells were generated by lentiviral-mediated shRNA. Proliferation/apoptosis studies were done by flow cytometry, and protein levels were determined by Western blot and/or immunohistochemistry. An experimental MM mouse model was generated to investigate the effects of MM cells overexpressing Gfi1 on tumor burden and osteolysis in vivo.ResultsWe found that Gfi1 expression is increased in patient’s MM cells and MM cell lines and was further increased by co-culture with BMSC, IL-6, and sphingosine-1-phosphate. Modulation of Gfi1 in MM cells had major effects on their survival and growth. Knockdown of Gfi1 induced apoptosis in p53-wt, p53-mutant, and p53-deficient MM cells, while Gfi1 overexpression enhanced MM cell growth and protected MM cells from bortezomib-induced cell death. Gfi1 enhanced cell survival of p53-wt MM cells by binding to p53, thereby blocking binding to the promoters of the pro-apoptotic BAX and NOXA genes. Further, Gfi1-p53 binding could be blocked by HDAC inhibitors. Importantly, inoculation of MM cells overexpressing Gfi1 in mice induced increased bone destruction, increased osteoclast number and size, and enhanced tumor growth.ConclusionsThese results support that Gfi1 plays a key role in MM tumor growth, survival, and bone destruction and contributes to bortezomib resistance, suggesting that Gfi1 may be a novel therapeutic target for MM.

  • Microvesicles released from multiple myeloma cells are equipped with ectoenzymes belonging to canonical and non-canonical adenosinergic pathways and produce adenosine from ATP and NAD<sup>+</sup>
    F. Morandi, D. Marimpietri, A. L. Horenstein, M. Bolzoni, D. Toscani, F. Costa, B. Castella, A. C. Faini, M. Massaia, V. Pistoia,et al.

    Informa UK Limited
    ABSTRACT Multiple myeloma (MM) derives from malignant transformation of plasma cells (PC), which accumulate in the bone marrow (BM), where microenvironment supports tumor growth and inhibits anti-tumor immune responses. Adenosine (ADO), an immunosuppressive molecule, is produced within MM patients' BM by adenosinergic ectoenzymes, starting from ATP (CD39/CD73) or NAD+ [CD38/CD203a(PC-1)/CD73]. These ectoenzymes form a discontinuous network expressed by different BM cells. We investigated the expression and function of ectoenzymes on microvesicles (MVs) isolated from BM plasma samples of patients with MM, using asymptomatic forms of monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM (SMM) as controls. The percentage of MVs expressing ectoenzymes at high levels was higher when derived from MM patients than controls. BM CD138+ PC from MM patients expressed high levels of all ectoenzymes. Paired MVs samples confirmed a higher percentage of MVs with high ectoenzymes expression in MM patients than controls. Pooled MVs from MM patients or controls were tested for ADO production. The catabolism of ATP, NAD+, ADPR and AMP to ADO was higher in MVs from MM patients than in those from controls. In conclusion, our results confirmed the hypothesis that MVs in MM niche are main contributor of ADO production. The ability of MVs to reach biological fluids strongly support the view that MVs may assume diagnostic and pathogenetic roles.

  • Possible targets to treat myeloma-related osteoclastogenesis
    Marina Bolzoni, Denise Toscani, Paola Storti, Valentina Marchica, Federica Costa, and Nicola Giuliani

    Informa UK Limited
    ABSTRACT Introduction: Bone destruction is the hallmark of multiple myeloma (MM). About 80% of MM patients at diagnosis presents myeloma bone disease (MBD) leading to bone pain and pathological fractures, significantly affecting patients’ quality of life. Bisphosphonates are the treatment of choice for MBD, but osteolytic lesions remain a critical issue in the current management of MM patients. Several studies clarified the mechanisms involved in MM-induced osteoclast formation and activation, leading to the identification of new possible targets and the development of better bone-directed therapies, that are discussed in this review. Areas covered: This review summarizes the latest advances in the knowledge of the pathophysiology of the osteoclast formation and activation induced by MM cells, and the new therapeutic targets identified. Recently, neutralizing antibodies (i.e. denosumab, siltuximab, daratumumab), as well as recombinant fusion proteins, and receptor molecular inhibitors, have been developed to block these targets. Clinical trials testing their anti-MBD potential are ongoing. The emerging role of exosomes and microRNAs in the regulation of osteoclast differentiation has been also discussed. Expert commentary: Although further studies are needed to arrive at a clinical approving, the basis for the development of better bone-directed therapies has been established.