Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Clin Exp Med ; 24(1): 7, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38240866

ABSTRACT

Bone metastases (BM) are a serious cancer complication, potentially causing substantial morbidity. Among the clinical issues related to BM, there is the lack of specific tools for early diagnosis and prognosis. We explored whether combining bone turnover markers (BTM) with dual-energy X-ray absorptiometry (DXA) assessment could identify early BM progression and risk of skeletal-related events (SREs) during zoledronate treatment. Before the initiation of zoledronate (T0) and after six months of treatment (T1), serum levels of five BTM were measured, and patients (N = 47) underwent DXA evaluation. Standard radiological imaging was performed to assess bone tumor response to medical anti-cancer treatment. High tumor burden in bone correlated with higher serum CTX (p = 0.007) and NTX (p = 0.005) at baseline. Low concentrations of OPG at T0 predicted BM progression with a sensitivity and specificity of 63% and 77%, respectively, when a cutoff of 5.2 pmol/l was used; such a predictive meaning was stronger in patients with lytic BM (sensitivity: 88%, specificity: 80%; p = 0.0006). As for the risk of SREs, we observed an association between low baseline OC (p = 0.04) and OPG (p = 0.08) and the onset of any-time SREs, whereas an increase in OPG over time was associated with reduced risk of on-study events (p = 0.03). Moreover, a statistically significant correlation emerged between low baseline lumbar T-score and femur BMD and on-study SREs (p < 0.001 in both instances). These findings suggest that addition of DXA to BTM dosage could help stratifying the risk of SREs at the time of BM diagnosis but does not enhance our capability of detecting bone progression, during zoledronate treatment.


Subject(s)
Bone Neoplasms , Humans , Zoledronic Acid/therapeutic use , Absorptiometry, Photon , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/drug therapy , Prognosis , Bone Remodeling/physiology
2.
Breast ; 66: 157-161, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36283134

ABSTRACT

BACKGROUND: Gastric pH changes by proton-pump-inhibitors (PPIs) were found to affect progression-free survival (PFS) in metastatic breast cancer (mBC) patients treated with palbociclib. The current study was aimed at investigating whether the same effect could occur in patients treated with ribociclib. PATIENTS AND METHODS: Patients with hormone-positive/HER-2-negative mBC candidates for first-line treatment with ribociclib were enrolled in this retrospective-cohort study. Patients were classified as "no concomitant PPIs" or "concomitant PPIs"; PPI administration covered the entire or not less than 2/3 of treatment with ribociclib. All clinical interventions were made according to clinical practice. RESULTS: A total of 128 patients were consecutively enrolled in the study; 78 belonged to the "no concomitant PPIs" group and 50 to the "concomitant PPIs" group. One hundred and six patients were endocrine-sensitive and received ribociclib and letrozole, while 22 were endocrine-resistant and were treated with ribociclib and fulvestrant. The most prescribed PPI was lansoprazole. According to PFS, patients taking PPIs had a PFS almost superimposable to those assuming ribociclib and endocrine therapy alone (35.3 vs. 49.2 months, p = 0.594). No difference in PFS was observed in estrogen-sensitive or estrogen-resistant mBC in the presence or absence of concomitant PPI treatment (p = 0.852). No correlation with adverse events was found including grade>2 hematological toxicities. CONCLUSIONS: The present study supports the hypothesis that the concomitant use of PPIs does not compromise the efficacy of ribociclib in a real-life setting.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Proton Pump Inhibitors/therapeutic use , Cohort Studies , Retrospective Studies , Aminopyridines , Estrogens/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Front Immunol ; 12: 697298, 2021.
Article in English | MEDLINE | ID: mdl-34858389

ABSTRACT

Introduction: Bone metastases (BMs) are a negative prognostic factor in patients with non-small cell lung cancer (NSCLC). Although immune-checkpoint inhibitors (ICIs) have dramatically changed the therapeutic landscape of NSCLC, little information is available on BMs from NSCLC treated with ICIs alone or in association with bone-targeted therapy (BTT) such as zoledronate or denosumab. Methods: From 2014 to 2020, 111 of the 142 patients with BMs secondary to NSCLC extrapolated from the prospective multicenter Italian BM Database were eligible for analysis. Information on blood count, comorbidities, and toxicity was retrospectively collected. The neutrophil-to-lymphocyte ratio (NLR) pre- and post-treatment was calculated. Survival was analyzed using the Kaplan-Meier method, with statistical significance of survival differences assessed using the log-rank test. Results: Median age was 66 (range, 42-84) years. Performance status (PS) Eastern Cooperative Oncology Group (ECOG) was 0-1 in 79/111 patients. The majority of patients (89.2%) had adenocarcinoma histology. At a median follow-up of 47.4 months, median progression-free (mPFS) and overall survival (mOS) was 4.9 (95%CI, 2.8-10.0) and 11.9 (95%CI, 8.2-14.4) months, respectively. Forty-six (43.4%) patients with BM NSCLC underwent first- or further-line therapy with ICIs: 28 (60.8%) received nivolumab, 9 (19.6%) pembrolizumab, and 9 (19.6%) atezolizumab. Of the 46 patients treated with ICIs, 30 (65.2%) underwent BTT: 24 (80.0%) with zoledronate and 6 (20.0%) with denosumab. The ICI-alone group had an mOS of 15.8 months [95%CI, 8.2-not evaluable (NE)] vs. 21.8 months (95%CI, 14.5-not evaluable) for the ICI plus BTT group and 7.5 (95%CI, 6.1-10.9) months for the group receiving other treatments (p < 0.001). NLR ≤5 had a positive impact on OS. Conclusion: BTT appears to have a synergistic effect when used in combination with ICIs, improving patient survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Bone and Bones/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Denosumab/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lymphocytes/immunology , Neutrophils/immunology , Zoledronic Acid/therapeutic use , Adult , Aged , Aged, 80 and over , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Drug Synergism , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Survival Analysis
4.
Oncotarget ; 9(38): 25355-25382, 2018 May 18.
Article in English | MEDLINE | ID: mdl-29861877

ABSTRACT

In the "precision medicine" era, chemotherapy still remains the backbone for the treatment of many cancers, but no affordable predictors of response to the chemodrugs are available in clinical practice. Single nucleotide polymorphisms (SNPs) are gene sequence variations occurring in more than 1% of the full population, and account for approximately 80% of inter-individual genomic heterogeneity. A number of studies have investigated the predictive role of SNPs of genes enrolled in both pharmacodynamics and pharmacokinetics of chemotherapeutics, but the clinical implementation of related results has been modest so far. Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. While formal recommendations for routine testing of these SNPs cannot be drawn at this moment, therapeutic decisions may indeed benefit of germline genomic information, when available. Here, we summarize the clinical impact of germline genomic variants on the efficacy and toxicity of major chemodrugs, with the aim to facilitate the therapeutic expectance of clinicians in the odiern quicksand field of complex molecular biology concepts and controversial trial data interpretation.

5.
Oncol Lett ; 14(5): 5671-5680, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29113194

ABSTRACT

Immunotherapy is one of the most recent systemic treatments to emerge for use in oncology, and is based on the blocking of inhibitory immune checkpoints to potentiate the immune response to cancer. The anti-cytotoxic T lymphocyte-associated antigen-4 antibody ipilimumab and anti-programmed cell death protein 1 antibodies, including nivolumab and pembrolizumab, are currently available and widely used, and other immune-inhibiting antibodies are now under intensive investigation. These antibodies have shown efficacy in a growing number of tumor types, following initial observations of their notable effects in melanoma treatment. Despite the efficacy of these antibodies, their novel mechanisms of action are also associated with a new class of side effects called immune-related adverse events (IRAEs). These side effects do not share a common pathophysiology with other anticancer treatments and, therefore, they often require specific therapies. When detected early and correctly treated, IRAEs are reversible; however, they can become severe and life-threatening if underestimated or inappropriately treated. This review aims to revisit the pathogenesis of IRAEs, with attention to gastrointestinal manifestations, since these are common and potentially dangerous complications of immunotherapy and represent a major cause of treatment discontinuation. Recommendations and guidelines for the management of IRAEs are also presented, in order to provide a clear and applicable algorithm for use by clinicians.

6.
Ann Ital Chir ; 88: 553-556, 2017.
Article in English | MEDLINE | ID: mdl-28944768

ABSTRACT

OBJECTIVE: Malignant melanomas presenting with unknown primaries are uncommon. In the majority of cases metastases of occult melanoma were detected in skin or in lymph nodes. Melanoma can rarely occur as a primary or metastatic intramammary tumor. CASE REPORT: We report the case of a 58-year-old Caucasian woman who came to our department with a voluminous mass in her right breast. Histopathological examination found metastasis of epithelioid melanoma with unknown primary lesion. Our patient underwent a radical enlarged mastectomy, but due to the extension a radical removal was not possible. DISCUSSION: In 2.2% of cases, melanoma may present with a metastasis without an identifiable primary lesion; this case should be considered a stage IV melanoma (Tx; N1; M1) due to the extension of the lesion and the infiltration of adjacent structures. CONCLUSIONS: In literature, the presence of a breast metastasis of melanoma with unknown primary origin was reported just in one case. The execution of histopathological analysis is mandatory for a correct differential diagnosis with primary carcinoma of the breast. Palliative metastasectomy should be discussed with multidisciplinary melanoma board. KEY WORDS: Breast metastases, Metastatic melanoma, Unknown primary site.


Subject(s)
Breast Neoplasms/secondary , Melanoma/secondary , Neoplasms, Unknown Primary , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/diagnosis , Combined Modality Therapy , Diagnosis, Differential , Female , Humans , Imidazoles/administration & dosage , Mastectomy , Melanoma/diagnosis , Melanoma/drug therapy , Melanoma/surgery , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Oximes/administration & dosage , Palliative Care , Pyridones/administration & dosage , Pyrimidinones/administration & dosage
7.
Exp Hematol ; 47: 48-53, 2017 03.
Article in English | MEDLINE | ID: mdl-27765615

ABSTRACT

Interleukin-17A (IL-17A) promotes the osteoclast (OC)-like differentiation of dendritic cells (DCs) in multiple myeloma (MM) and contributes to the pathogenesis of myeloma bone disease (MBD). In our study, everolimus (EVR) abrogated the in vitro OC-like activity of DCs from 12 MM patients significantly. Exploring the EVR effects, we found that the inhibition of the osteoerosive activity of OC-DCs was mostly due to the blockade of signals driven by the IL-17A receptor toward the CCAAT/enhancer-binding protein beta/musculoaponeurotic fibrosarcoma oncogene homolog B axis Therefore, MM patients with MBD would probably benefit from mammalian target of rapamycin inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transdifferentiation/drug effects , Dendritic Cells/metabolism , Everolimus/pharmacology , Interleukin-17/metabolism , Multiple Myeloma/metabolism , Osteoclasts/metabolism , Antineoplastic Agents/therapeutic use , Cell Adhesion/drug effects , Cell Survival/drug effects , Cell Transdifferentiation/genetics , Dendritic Cells/pathology , Everolimus/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology
8.
Br J Haematol ; 173(1): 59-69, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26728969

ABSTRACT

Cilengitide (CLG) is an inhibitor of both αv ß3 and αv ß5 integrins, with a defined anti-tumour effect in glioblastoma. Pre-clinical studies demonstrate its ability to restrain the bone resorbing property of metastatic osteotropic tumours and we have previously shown that the disablement of αv ß3 in multiple myeloma (MM) plasma cells results in exhaustion of their in vitro osteoclast (OC)-like activity on bone substrate. Here, we investigated the effect of CLG on this functional property of MM cells. Both αv ß3 and αv ß5 were measured on primary marrow MM cells from 19 patients, and the effect of CLG on proliferation, apoptosis and adhesion was investigated in parallel with MM cell lines and OCs from healthy donors. In addition, the effect of CLG on the capability of malignant plasma cells to produce erosive lacunae on calcium phosphate was explored in relation to the activation of intracellular kinases of molecular pathways of both integrins. Ultrastructural microscopy was used to evaluate the morphological changes in MM cells due to the effect of CLG on cell adhesion. The data from our study demonstrate that CLG restrains the bone resorbing function of MM cells by disabling their adhesion properties. Further investigations in pre-clinical studies of osteotropic tumours are warranted.


Subject(s)
Bone Resorption/metabolism , Cell Proliferation/drug effects , Multiple Myeloma/metabolism , Osteoclasts/metabolism , Plasma Cells/metabolism , Snake Venoms/pharmacology , Apoptosis/drug effects , Bone Resorption/pathology , Cell Adhesion/drug effects , Female , Humans , Integrin alphaVbeta3/metabolism , Male , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Osteoclasts/pathology , Plasma Cells/pathology , Receptors, Vitronectin/metabolism , Tumor Cells, Cultured
9.
Expert Rev Clin Immunol ; 12(1): 79-89, 2016.
Article in English | MEDLINE | ID: mdl-26505837

ABSTRACT

The crosstalk of melanoma cells with components of the microenvironment promotes malignant cell proliferation and spread to distant tissues. Although the major pathogenetic events have already been elucidated, the mechanisms that drive the metastatic behavior of tumor cells are still undefined. MicroRNAs (miRNAs) are small non-coding RNAs that control post-transcriptional gene expression through interconnected kinases upstream of functional genes involved in tumor progression. Here, we review the biological relevance of melanoma-related miRNAs and focus on their potential role in propagating signals that may cause tumor microenvironment rearrangements, as well as disablement of the immune system and melanoma cell proliferation.


Subject(s)
Melanoma/genetics , MicroRNAs/genetics , Protein Kinases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Neoplasm Metastasis , Protein Kinases/genetics , Tumor Escape , Tumor Microenvironment
10.
Neuroendocrinology ; 103(3-4): 321-34, 2016.
Article in English | MEDLINE | ID: mdl-26227818

ABSTRACT

BACKGROUND: Neuroendocrine tumors (NETs) metastasize to the bone. However, the incidence, clinical features, management and pathogenesis of bone involvement in NET patients have been poorly investigated. METHODS: We reviewed all published reports of histologically confirmed bone metastatic NETs and explored clinical, radiological, prognostic and therapeutic characteristics in a population of 152 patients. We then evaluated immunohistochemical expression of a panel of eight epithelial-mesenchymal transition (EMT)-related factors including SNAIL, TGF-ß1, CTGF, IL-11, PTHrP, EpCAM, CXCR4 and RANK in an independent cohort of 44 archival primary NETs. Biomarker expression was correlated with clinicopathological variables, including skeletal involvement, and tested for survival prediction. RESULTS: We found that 55% of NET patients with bone metastases were male, with a median age of 55 years at diagnosis. Metastases were restricted to the skeleton in 34% of the NET population, and axial and osteoblastic lesions were prevalent. NETs differently expressed proteins involved in EMT activation. High CXCR4 (p < 0.0001) and low TGF-ß1 levels (p = 0.0015) were significantly associated with increased risk of skeletal metastases, suggesting that EMT is implicated in NET osteotropism. By applying an algorithm measuring distinct immunohistochemical predictors of osteotropism on primary tumors, we were able to identify NET patients with bone metastases with a sensitivity and specificity of 91 and 100%, respectively (p < 0.0001). Patients whose primary tumors expressed CTGF (p = 0.0007) as well as the truncated form of EpCAM (p = 0.06) showed shorter survival. CONCLUSION: Although underestimated, bone metastases are a prominent feature of NETs, and the tumor expression of EMT markers at diagnosis may predict concurrent or subsequent skeleton colonization.


Subject(s)
Bone Neoplasms/secondary , Epithelial-Mesenchymal Transition/physiology , Neuroendocrine Tumors/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Algorithms , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cohort Studies , Databases, Bibliographic/statistics & numerical data , Female , Humans , Male , Middle Aged , PubMed/statistics & numerical data , Receptors, CXCR4/metabolism , Sensitivity and Specificity , Statistics, Nonparametric , Transforming Growth Factor beta1/metabolism , Young Adult
11.
Crit Rev Oncol Hematol ; 96(1): 183-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26126493

ABSTRACT

The interplay of cancer cells and accessory cells within the microenvironment drives signals regulating the proliferation, migration and skeleton colonization. Osteotropism of tumor cells depends on chemokine activation, production of soluble factors and defective gene expression that cooperate within the metastatic niche to the bone resorbing functions of osteoclasts. Adhesion of cancer cells to the extracellular matrix is regulated by integrins as αvß3 that enhances their invasiveness, pro-tumor angiogenesis and skeleton invasion. Therefore, αvß3 signaling is implicated in enhancing osteotropism of breast and prostate cancers as well as of multiple myeloma. Targeting of αvß3 has been adopted to restrain the tumor progression in several cancer models leading to improvement of overall survival as effect of the reduction of both tumor burden and osteotropism by malignant cells. Here, we review both the role of αvß3 in malignant osteoclastogenesis and its potential targeting to restrain the bone colonization by skeleton invading cancers.


Subject(s)
Bone Neoplasms/secondary , Integrin alphaVbeta3/physiology , Bone Neoplasms/etiology , Bone Neoplasms/pathology , Cell Proliferation , Cell Survival , Epithelial-Mesenchymal Transition , Humans , Integrin alphaVbeta3/chemistry , Male , Neoplastic Stem Cells/physiology , Osteoclasts/physiology , Osteogenesis , Signal Transduction/physiology
14.
Expert Rev Clin Immunol ; 10(10): 1395-404, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25225774

ABSTRACT

Melanoma is an immunogenic cancer that overcomes the control of the immune system through the production of tolerogenic cytokines and growth factors in the microenvironment. In melanoma, dendritic cells (DC) show severe alterations in maturation, cross-priming and antigenic presentation, while other accessory cells infiltrating the tumor milieu also suppress DCs through the activation of the STAT pathway by IL-10 and IL-6. Novel immunotherapy strategies blocking cytotoxic T-lymphocyte antigen (CTLA-4) are successful in advanced disease, while melanoma cells carrying the BRAF(V600E) mutation further reinforce the immune suppression by activating MAPKs. Here, we review the major mechanisms involved in the cross-talk between melanoma cells and the immune system as well as the issue of defects in DCs in relation to novel studies aimed at restoring their anti-tumor activity.


Subject(s)
Dendritic Cells/immunology , Melanoma/immunology , Tumor Escape/immunology , Animals , Humans
15.
Exp Hematol ; 42(10): 897-908.e1, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24971696

ABSTRACT

Autophagy occurs in tumor cells acquiring cytotoxic drug resistance and its activation may impair their susceptibility to apoptosis in response to apoptogen agents. We investigated the pro-apoptotic effect of dexamethasone (Dex) on MM cell lines (U266, INA-6, LR5-8226, LIG, and MCC2) and primary malignant plasma cells from naïve and refractory/relapsed patients. We evaluated the transcriptional and ultrastructural events leading to autophagy by measuring Beclin-1 and p62 levels and transmission electronic microscopy. Autophagy was inhibited by hydroxychloroquine (HCQ), whereas the ability of Dex-resistant MM cells to recover the susceptibility to apoptosis was measured. A direct relationship between autophagy and Beclin-1 or LC3/Atg8 levels was observed, whereas their mRNAs were inversely correlated to p62 expression. Starvation strongly activated autophagy by inducing cellular, transcriptional, and ultrastructural modifications that were reversed by HCQ. Taken together, these data suggest that autophagy is a potential mechanism leading to drug resistance in MM, and suggest Beclin-1 and p62 as early markers of cell susceptibility to apoptosis. The combination of HCQ with novel agents may thus be considered to improve the therapeutic response in relapsed/resistant MM patients.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis Regulatory Proteins/physiology , Autophagy/physiology , Membrane Proteins/physiology , Multiple Myeloma/pathology , Neoplasm Proteins/physiology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Autophagy-Related Protein 8 Family , Beclin-1 , Cell Line, Tumor , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hydroxychloroquine/pharmacology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Sequestosome-1 Protein , Transcription, Genetic
16.
J Bone Miner Res ; 29(1): 55-66, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23787729

ABSTRACT

To promote their survival and progression in the skeleton, osteotropic malignancies of breast, lung, and prostate produce parathyroid hormone-related protein (PTHrP), which induces hypercalcemia. PTHrP serum elevations have also been described in multiple myeloma (MM), although their role is not well defined. When we investigated MM cells from patients and cell lines, we found that PTHrP and its receptor (PTH-R1) are highly expressed, and that PTHrP is secreted both as a full-length molecule and as small subunits. Among these subunits, the mid-region, including the nuclear localization sequence (NLS), exerted a proliferative effect because it was accumulated in nuclei of MM cells surviving in starvation conditions. This was confirmed by increased transcription of several genes enrolled in proliferation and apoptosis control. PTHrP was also found to stimulate PTH-R1 in MM cells. PTH-R1's selective activation by the full-length PTHrP molecule or the NH2 -terminal fragment resulted in a significant increase of intracellular Ca(2+) influx, cyclic adenosine monophosphate (cAMP) content, and expression of receptor activator of NF-κB ligand (RANKL) and monocyte chemoattractant protein-1 (MCP-1). Our data definitely clarify the role of PTHrP in MM. The PTHrP peptide is functionally secreted by malignant plasma cells and contributes to MM tumor biology and progression, both by intracrine maintenance of cell proliferation in stress conditions and by autocrine or paracrine stimulation of PTH-R1, which in turn reinforces the production of osteoclastogenic factors. © 2014 American Society for Bone and Mineral Research.


Subject(s)
Multiple Myeloma/metabolism , Parathyroid Hormone-Related Protein/biosynthesis , Plasma Cells/metabolism , Receptor, Parathyroid Hormone, Type 1/biosynthesis , Cell Line, Tumor , Cell Proliferation , Chemokine CCL2/biosynthesis , Cyclic AMP/metabolism , Disease Progression , Humans , Peptide Fragments/biosynthesis , Peptide Fragments/pharmacology , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Receptor, Parathyroid Hormone, Type 1/metabolism
17.
Br J Haematol ; 161(6): 821-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23594390

ABSTRACT

Interleukin 17A (IL17A), a cytokine involved in allergy, inflammation and osteoclastogenesis, was investigated in multiple myeloma (MM) to assess its role in the osteoclast (OC)-like activity of marrow immature dendritic cells (iDCs). Comparing nine MM patients with control subjects affected by monoclonal gammopathy of undetermined significance, we found high IL17A expression in the marrow plasma of MM patients in parallel with its deposits within the stromal matrix. Increased expression of the IL17A receptor (IL17RA) was also found in primary myeloma iDCs, which underwent OC-like transdifferentiation after IL17A stimulation. To assess the role of IL17A, we measured the activity of the IL17/IL17RA pathway in IL17A-transdifferentiated iDCs and the expression of functional OC genes by Western blotting and real-time polymerase chain reaction. These cells showed increased RNA transcription of genes enrolled in the maturation of OCs, while NFATC1 and FOS were induced by IL17A, independently of NFKB1 phosphorylation. Moreover, the concurrent phosphorylation of the Lip isoform of CEBPB and the down-regulation of MAFB supported the activation of IL17RA pathway in OC-like transdifferentiated iDCs that was apparently unrelated to TNFRSF11A signalling. These data emphasize the involvement of iDCs in MM hyperactive osteoclastogenesis and suggest that their bone resorption activity is also regulated, at least in vitro, by IL17RA.


Subject(s)
Cell Transdifferentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Interleukin-17/pharmacology , Multiple Myeloma/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Resorption/genetics , Cell Transdifferentiation/genetics , Dendritic Cells/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin-17/metabolism , Multiple Myeloma/genetics , Osteoclasts/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism
18.
Oncologist ; 16(7): 1040-8, 2011.
Article in English | MEDLINE | ID: mdl-21659611

ABSTRACT

The crosstalk of myeloma cells with accessory cells drives the expansion of malignant plasma cell clones and the hyperactivation of osteoclastogenesis that occurs in multiple myeloma (MM). These reciprocal interactions promote defective dendritic cell (DC) function in terms of antigen processing, clearance of tumor cells, and efficacy of the immune response. Thus, myeloma cells exert immune suppression that explains, at least in part, the failure of therapeutic approaches, including DC vaccination. Impairment of DCs depends on high bone marrow levels of cytokines and adhesion molecules that affect both maturation and expression of costimulatory molecules by DCs. Moreover, DCs share with osteoclasts (OCs) a common ontogenetic derivation from the monocyte lineage, and thus may undergo OC-like transdifferentiation both in vitro and in vivo. Immature DCs (iDCs) induce clonogenic growth of malignant plasma cells while displaying OC-like features, including the ability to resorb bone tissue once cultured with myeloma cells. This OC-like transdifferentiation of iDCs is dependent on the activation of both the receptor activator of nuclear factor κB (RANK)-RANK ligand (RANK-L) and CD47-thrombospondin (TSP)-I axes, although interleukin 17-producing T helper-17 clones within the bone microenvironment may also take part in this function. Therefore, iDCs allied with malignant plasma cells contribute to MM osteoclastogenesis, although other molecules released by tumor cells may independently contribute to the bone-resorbing machinery.


Subject(s)
Dendritic Cells/pathology , Multiple Myeloma/pathology , Plasma Cells/pathology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Dendritic Cells/immunology , Disease Progression , Humans , Multiple Myeloma/immunology , Plasma Cells/immunology
19.
Recenti Prog Med ; 102(2): 88-95, 2011 Feb.
Article in Italian | MEDLINE | ID: mdl-21513124

ABSTRACT

In tumors characterized by a high osteotropism, such as multiple myeloma, the measurement of bone metabolism markers is helpful in monitoring both severity and prognosis of the skeletal disease. Here, we review the pathophysiology of these markers including tartrate resistant acid phosphatase (TRAcP), which appears highly specific and closely related to the extent of myeloma bone lesions.


Subject(s)
Acid Phosphatase/metabolism , Biomarkers, Tumor/metabolism , Bone Neoplasms/enzymology , Isoenzymes/metabolism , Multiple Myeloma/enzymology , Acid Phosphatase/blood , Biomarkers, Tumor/blood , Bone Neoplasms/blood , Bone Neoplasms/physiopathology , Humans , Isoenzymes/blood , Multiple Myeloma/blood , Multiple Myeloma/physiopathology , Tartrate-Resistant Acid Phosphatase
20.
J Biomed Biotechnol ; 2010: 457146, 2010.
Article in English | MEDLINE | ID: mdl-20671931

ABSTRACT

Lupus nephritis (LN) occurs in more than one-third of patients with systemic lupus erythematosus. Its pathogenesis is mostly attributable to the glomerular deposition of immune complexes and overproduction of T helper- (Th-) 1 cytokines. In this context, the high glomerular expression of IL-12 and IL-18 exerts a major pathogenetic role. These cytokines are locally produced by both macrophages and dendritic cells (DCs) which attract other inflammatory cells leading to maintenance of the kidney inflammation. However, other populations including T-cells and B-cells are integral for the development and worsening of renal damage. T-cells include many pathogenetic subsets, and the activation of Th-17 in keeping with defective T-regulatory (Treg) cell function regards as further event contributing to the glomerular damage. These populations also activate B-cells to produce nephritogenic auto-antibodies. Thus, LN includes a complex pathogenetic mechanism that involves different players and the evaluation of their activity may provide an effective tool for monitoring the onset of the disease.


Subject(s)
Cytokines/biosynthesis , Disease Progression , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Dendritic Cells/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...