Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 233
Filter
1.
J Transl Med ; 22(1): 548, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849800

ABSTRACT

BACKGROUND: Despite significant advancements in treatment strategies, multiple myeloma remains incurable. Additionally, there is a distinct lack of reliable biomarkers that can guide initial treatment decisions and help determine suitable replacement or adjuvant therapies when relapse ensues due to acquired drug resistance. METHODS: To define specific proteins and pathways involved in the progression of monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM), we have applied super-SILAC quantitative proteomic analysis to CD138 + plasma cells from 9 individuals with MGUS and 37 with MM. RESULTS: Unsupervised hierarchical clustering defined three groups: MGUS, MM, and MM with an MGUS-like proteome profile (ML) that may represent a group that has recently transformed to MM. Statistical analysis identified 866 differentially expressed proteins between MM and MGUS, and 189 between MM and ML, 177 of which were common between MGUS and ML. Progression from MGUS to MM is accompanied by upregulated EIF2 signaling, DNA repair, and proteins involved in translational quality control, whereas integrin- and actin cytoskeletal signaling and cell surface markers are downregulated. CONCLUSION: Compared to the premalignant plasma cells in MGUS, malignant MM cells apparently have mobilized several pathways that collectively contribute to ensure translational fidelity and to avoid proteotoxic stress, especially in the ER. The overall reduced expression of immunoglobulins and surface antigens contribute to this and may additionally mediate evasion from recognition by the immune apparatus. Our analyses identified a range of novel biomarkers with potential prognostic and therapeutic value, which will undergo further evaluation to determine their clinical significance.


Subject(s)
Disease Progression , Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Humans , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Monoclonal Gammopathy of Undetermined Significance/immunology , Proteomics , Male , Female , Protein Biosynthesis , Middle Aged , Aged , Cluster Analysis , Plasma Cells/immunology , Plasma Cells/pathology , Plasma Cells/metabolism , Signal Transduction , Proteome/metabolism , Quality Control
2.
JCI Insight ; 9(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38713510

ABSTRACT

Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.


Subject(s)
Disease Models, Animal , Interleukin-6 , Multiple Myeloma , Animals , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Humans , Mice , Interleukin-6/metabolism , Mice, Transgenic , Bortezomib/pharmacology , Bortezomib/therapeutic use , Male , Female , Plasma Cells/immunology , Monoclonal Gammopathy of Undetermined Significance/immunology , Monoclonal Gammopathy of Undetermined Significance/pathology
3.
Cancer Immunol Immunother ; 71(1): 177-187, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34061243

ABSTRACT

Tumour-associated macrophages (TAMs) support cancer cell survival and suppress anti-tumour immunity. Tumour infiltration by CD163pos TAMs is associated with poor outcome in several human malignancies, including multiple myeloma (MM). Signal transducer and activator of transcription 3 (STAT3) is over-activated in human cancers, and specifically within TAMs activation of STAT3 may induce an immunosuppressive (M2-like) phenotype. Therefore, STAT3-inhibition in TAMs may be a future therapeutic strategy.We investigated TAM markers CD163, CD206, and activated STAT3 (pSTAT3) in patients with MGUS (n = 32) and MM (n = 45), as well as healthy controls (HCs, n = 13).Blood levels of the macrophage biomarkers sCD163 and sCD206, and circulating cytokines, as well as bone marrow mRNA expression of CD163 and CD206, were generally increased in MGUS and MM patients, compared to HCs, but to highly similar levels. By immunohistochemistry, bone marrow levels of pSTAT3 were increased specifically within CD163pos cells in both MGUS and MM patients.In conclusion, macrophage-related inflammatory changes, including activation of STAT3, were present already at the MGUS stage, at similar levels as in MM. Specific increase in pSTAT3 levels within CD163pos cells supports that the CD163 scavenger receptor may be a useful target for future delivery of STAT3-inhibitory drugs to TAMs in MM patients.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Bone Marrow/metabolism , Macrophages/metabolism , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , Receptors, Cell Surface/biosynthesis , STAT3 Transcription Factor/biosynthesis , Aged , Bone Marrow Cells/metabolism , Case-Control Studies , Female , Humans , Immunosuppression Therapy , Immunosuppressive Agents , Inflammation , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/metabolism , Multiple Myeloma/metabolism , Phenotype , Phosphorylation , Prospective Studies
5.
Leukemia ; 36(1): 138-154, 2022 01.
Article in English | MEDLINE | ID: mdl-34290359

ABSTRACT

Immune profiling in patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and multiple myeloma (MM) provides the framework for developing novel immunotherapeutic strategies. Here, we demonstrate decreased CD4+ Th cells, increased Treg and G-type MDSC, and upregulation of immune checkpoints on effector/regulatory and CD138+ cells in MM patients, compared MGUS/SMM patients or healthy individuals. Among the checkpoints profiled, LAG3 was most highly expressed on proliferating CD4+ Th and CD8+ Tc cells in MM patients BMMC and PBMC. Treatment with antibody targeting LAG3 significantly enhanced T cells proliferation and activities against MM. XBP1/CD138/CS1-specific CTL generated in vitro displayed anti-MM activity, which was further enhanced following anti-LAG3 treatment, within the antigen-specific memory T cells. Treg and G-type MDSC weakly express LAG3 and were minimally impacted by anti-LAG3. CD138+ MM cells express GAL-3, a ligand for LAG3, and anti-GAL-3 treatment increased MM-specific responses, as observed for anti-LAG3. Finally, we demonstrate checkpoint inhibitor treatment evokes non-targeted checkpoints as a cause of resistance and propose combination therapeutic strategies to overcome this resistance. These studies identify and validate blockade of LAG3/GAL-3, alone or in combination with immune strategies including XBP1/CD138/CS1 multipeptide vaccination, to enhance anti-tumor responses and improve patient outcome in MM.


Subject(s)
Antigens, CD/chemistry , Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Immunosuppression Therapy/methods , Leukocytes, Mononuclear/immunology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , T-Lymphocytes, Cytotoxic/immunology , Apoptosis , Case-Control Studies , Cell Proliferation , Follow-Up Studies , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lymphocyte Activation , Monoclonal Gammopathy of Undetermined Significance/metabolism , Monoclonal Gammopathy of Undetermined Significance/pathology , Monoclonal Gammopathy of Undetermined Significance/therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Prognosis , Tumor Cells, Cultured , Lymphocyte Activation Gene 3 Protein
6.
Front Immunol ; 12: 792609, 2021.
Article in English | MEDLINE | ID: mdl-34880879

ABSTRACT

Background: We previously reported algorithms based on clinical parameters and plasma cell characteristics to identify patients with smoldering multiple myeloma (SMM) with higher risk of progressing who could benefit from early treatment. In this work, we analyzed differences in the immune bone marrow (BM) microenvironment in SMM to better understand the role of immune surveillance in disease progression and to identify immune biomarkers associated to higher risk of progression. Methods: Gene expression analysis of BM cells from 28 patients with SMM, 22 patients with monoclonal gammopathy of undetermined significance (MGUS) and 22 patients with symptomatic MM was performed by using Nanostring Technology. Results: BM cells in SMM compared to both MGUS and symptomatic MM showed upregulation of genes encoding for key molecules in cytotoxicity. However, some of these cytotoxic molecules positively correlated with inhibitory immune checkpoints, which may impair the effector function of BM cytotoxic cells. Analysis of 28 patients with SMM revealed 4 distinct clusters based on immune composition and activation markers. Patients in cluster 2 showed a significant increase in expression of cytotoxic molecules but also inhibitory immune checkpoints compared to cluster 3, suggesting the presence of cytotoxic cells with an exhausted phenotype. Accordingly, patients in cluster 3 had a significantly longer progression free survival. Finally, individual gene expression analysis showed that higher expression of TNF superfamily members (TNF, TNFAIP3, TNFRSF14) was associated with shorter progression free survival. Conclusions: Our results suggest that exhausted cytotoxic cells are associated to high-risk patients with SMM. Biomarkers overexpressed in patients with this immune gene profile in combination with clinical parameters and PC characterization may be useful to identify SMM patients with higher risk of progression.


Subject(s)
Bone Marrow/physiology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , Smoldering Multiple Myeloma/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinogenesis , Cellular Microenvironment , Cohort Studies , Cytotoxicity, Immunologic/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Proteins/genetics , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/mortality , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Phenotype , Smoldering Multiple Myeloma/genetics , Smoldering Multiple Myeloma/mortality , Survival Analysis , Tumor Necrosis Factor-alpha/genetics
7.
Sci Rep ; 11(1): 19266, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588555

ABSTRACT

Register-based studies indicate a possible association of monoclonal gammopathy of undetermined significance (MGUS) and prostate cancer (PCa). Aim of the present study was to investigate the relationship between MGUS and PCa considering potentially shared risk factors. Data from the prospective population-based Heinz Nixdorf Recall cohort study of 2.385 men (age 45-85) were analyzed. MGUS was determined at three points in time; cases of cancer were assessed annually. Potentially shared risk factors were assessed at baseline. Hazard ratios (HR), adjusted for age and educational attainment, and corresponding 95%-confidence intervals (95%-CI) were calculated. 157 cases of MGUS and 143 incident cases of PCa were detected. Of 19 participants diagnosed with both, MGUS and incident PCa, only in one case MGUS did not clearly occur before PCa. MGUS was associated with PCa presenting a HR of 2.00 (95%-CI: 1.23-3.25). Stratified by isotype, IgM-MGUS showed the strongest association with PCa. There was no relevant change of the effect estimate when adjusting for potentially shared risk factors. We were able to give supporting evidence for an association between MGUS and PCa and pointed out its temporality. There was no indication that the observed association is due to shared risk factors. The present study indicated that different isotypes of MGUS differ in the strength of the effect on PCa-risk. Based on these findings, future studies investigating the pathophysiological background of the association will be needed.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance/epidemiology , Prostatic Neoplasms/epidemiology , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/complications , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/immunology , Prospective Studies , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/immunology , Risk Assessment , Risk Factors
8.
Crit Rev Eukaryot Gene Expr ; 31(5): 21-26, 2021.
Article in English | MEDLINE | ID: mdl-34591386

ABSTRACT

FOXP3-expressing regulatory T-cells (Tregs), which suppress aberrant immune response against self-antigens, also suppress anti-tumor immune response. It has been shown that there is an increased proportion of Tregs in several different human malignancies, although the actual mechanism remains unclear. The research aims to explore the relationship between the number of Tregs and a predict prognosis in particular hematological diseases as monoclonal gammopathies of uncertain significance (MGUS). Tregs were evaluated by means of flow cytometry (CD4+CD25high/+ CD127low/-) in whole peripheral blood of 56 patients with MGUS to predict progression to overt multiple myeloma (MM). In two groups of patients, MGUS versus MGUS evolved to MM, we found a significative difference for the number of white blood cells, but not in terms of clinical and laboratory features evaluated at diagnosis. The study demonstrated the absence of a prognostic relevance of Tregs in MGUS. Nevertheless, their role in these disorders is still to be defined.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/diagnosis , Multiple Myeloma/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Cell Count , Disease Progression , Female , Flow Cytometry , Humans , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/pathology , Prognosis
9.
Front Immunol ; 12: 632564, 2021.
Article in English | MEDLINE | ID: mdl-33717170

ABSTRACT

Multiple myeloma (MM), a malignant neoplasm of plasma cells that reside in the bone marrow (BM), is universally preceded by a precursor state termed monoclonal gammopathy of undetermined significance (MGUS). Many individuals with MGUS never progress to MM or progress over many years. Therefore, MGUS provides a unique opportunity to surveil changes in the BM tumor microenvironment throughout disease progression. It is increasingly appreciated that MGUS cells carry many of the genetic changes found in MM. Prior studies have also shown that MGUS cells can be recognized by the immune system, leading to early changes in the BM immune environment compared to that of healthy individuals, including alterations in both innate and adaptive immunity. Progression to clinical MM is associated with attrition of T cells with stem memory-like features and instead accumulation of T cells with more terminally differentiated features. Recent clinical studies have suggested that early application of immune-modulatory drugs, which are known to activate both innate and adaptive immunity, can delay the progression to clinical MM. Understanding the biology of how the immune response and tumors coevolve over time is needed to develop novel immune-based approaches to achieve durable and effective prevention of clinical malignancy.


Subject(s)
Multiple Myeloma/immunology , Multiple Myeloma/prevention & control , Adaptive Immunity , Bone Marrow/immunology , Humans , Immune Checkpoint Proteins/immunology , Immunity, Innate , Immunologic Surveillance , Immunomodulation , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/genetics , Tumor Microenvironment
10.
Front Immunol ; 12: 620596, 2021.
Article in English | MEDLINE | ID: mdl-33708212

ABSTRACT

The classical paradigm of host-tumor interaction, i.e. elimination, equilibrium, and escape (EEE), is reflected in the clinical behavior of myeloma which progresses from the premalignant condition, Monoclonal Gammopathy of Unknown Significance (MGUS). Despite the role of other immune cells, CD4+ regulatory T cells (Treg) and cytotoxic CD8+ T cells have emerged as the dominant effectors of host control of the myeloma clone. Progression from MGUS to myeloma is associated with alterations in Tregs and terminal effector CD8+ T cells (TTE). These changes involve CD39 and CD69 expression, affecting the adenosine pathway and residency in the bone marrow (BM) microenvironment, together with oligoclonal expansion within CD8+ TTE cells. In this mini-review article, in the context of earlier data, we summarize our recent understanding of Treg involvement in the adenosine pathway, the significance of oligoclonal expansion within CD8+ TTE cells and BM-residency of CD8+ TTE cells in MGUS and newly diagnosed multiple myeloma patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , Precancerous Conditions/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carcinogenesis , Cell Proliferation , Humans , Lymphocyte Activation , Tumor Microenvironment
11.
Ann Hematol ; 100(4): 855-863, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33416902

ABSTRACT

Monoclonal gammopathy of undetermined significance (MGUS), precursor of multiple myeloma, is an asymptomatic plasma cell disorder that overproduces serum monoclonal protein. Older age, male sex, black race, and family history of MGUS increase the risk of MGUS, yet other risk factors are known. We systematically reviewed observational epidemiological studies that examined sociodemographic, clinical, and behavioral risk factors for the development of MGUS. The protocol for this study was registered on the PROSPERO registry for systematic reviews. We identified epidemiological studies from PubMed and Scopus. Articles were limited to those written in English and published before February 2019. Five case-control and three cohort studies were eligible for data extraction. Studies evaluating factors associated with MGUS risk are limited, with conflicting conclusions regarding risk associated with obesity. Despite the limited research, a significant elevated risk for being diagnosed with MGUS was associated with several specific prior infections, inflammatory disorders, and smoking. The sparse existing literature suggests an increased risk of MGUS associated with several risk factors related to immune function. Further research is needed to explore the potential mechanisms underlying the development of MGUS and to confirm risk factors, both modifiable and non-modifiable.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance/epidemiology , Age Factors , Aged , Antigens/immunology , Autoimmune Diseases/epidemiology , Case-Control Studies , Comorbidity , Diet , Europe/epidemiology , Female , Humans , Hypersensitivity/epidemiology , Infections/epidemiology , Inflammation/epidemiology , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/immunology , Obesity/epidemiology , Observational Studies as Topic , Risk Factors , Sex Factors , Smoking/epidemiology , Socioeconomic Factors , United States/epidemiology
12.
Pathol Res Pract ; 218: 153317, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360970

ABSTRACT

Monoclonal gammopathy of undetermined significance (MGUS) is a pre-malignant abnormality of plasma cells, with increased serum levels of immunoglobulins. Patients with MGUS may evolve to multiple myeloma through a multistep process including deregulated gene expression. microRNAs are small non-coding RNA molecules involved in post-transcriptional regulation of crucial biological processes, such as morphogenesis, cell differentiation, apoptosis, and cancer. This study aimed to evaluate microRNA expression on peripheral lymph-monocytes from MGUS subjects compared with healthy controls using qPCR arrays. Blood samples were collected by venipuncture from fifteen, newly diagnosed MGUS patients and fifteen healthy subjects. A further group (validation group) of six newly diagnosed MGUS patients and five healthy control were enrolled for the validation of miRNAs and their mRNAs target. The study was conducted performing miProfile miRNA qPCR arrays, followed by validation of miRNAs and related mRNA targets through RT-qPCR. The functional interaction between microRNAs and target gene were obtained by Ingenuity Pathways Analysis (IPA). IPA network analysis identified only molecules and relationships experimentally observed in peripheral lymphomonocytes. The following miRNAs :133a-3p, 16-5p, 291-3p, 23a-3p, 205-5p, 17-5p, 7a-5p, 221-3p, 30c-5p, 126a-3p,155-5p, let-7a-5p and 26a-5p, involved in the regulation of genes with a role in lymphocyte homeostasis, cell proliferation, apoptosis, and multiple myeloma (MM) progression, were differently expressed in MGUS with respect to healthy subjects. This miRNA signature and its relative targets could be considered for the formulation of new therapeutic strategies in the prophylaxis or treatment of monoclonal gammopathies.


Subject(s)
Gene Expression Profiling , Lymphocytes/immunology , MicroRNAs/genetics , Monoclonal Gammopathy of Undetermined Significance/genetics , Monocytes/immunology , Polymerase Chain Reaction , Transcriptome , Aged , Case-Control Studies , Female , Gene Regulatory Networks , Humans , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/immunology , Predictive Value of Tests
13.
Front Immunol ; 11: 602868, 2020.
Article in English | MEDLINE | ID: mdl-33343574

ABSTRACT

A highly recurrent somatic L265P mutation in the TIR domain of the signaling adapter MYD88 constitutively activates NF-κB. It occurs in nearly all human patients with Waldenström's macroglobulinemia (WM), a B cell malignancy caused by IgM-expressing cells. Here, we introduced an inducible leucine to proline point mutation into the mouse Myd88 locus, at the orthologous position L252P. When the mutation was introduced early during B cell development, B cells developed normally. However, IgM-expressing plasma cells accumulated with age in spleen and bone, leading to more than 20-fold elevated serum IgM titers. When introduced into germinal center B cells in the context of an immunization, the Myd88L252P mutation caused prolonged persistence of antigen-specific serum IgM and elevated numbers of antigen-specific IgM plasma cells. Myd88L252P-expressing B cells switched normally, but plasma cells expressing other immunoglobulin isotypes did not increase in numbers, implying that IgM expression may be required for the observed cellular expansion. In order to test whether the Myd88L252P mutation can cause clonal expansions, we introduced it into a small fraction of CD19-positive B cells. In this scenario, five out of five mice developed monoclonal IgM serum paraproteins accompanied by an expansion of clonally related plasma cells that expressed mostly hypermutated VDJ regions. Taken together, our data suggest that the Myd88L252P mutation is sufficient to promote aberrant survival and expansion of IgM-expressing plasma cells which in turn can cause IgM monoclonal gammopathy of undetermined significance (MGUS), the premalignant condition that precedes WM.


Subject(s)
B-Lymphocytes/metabolism , Gene Targeting , Immunoglobulin M/blood , Monoclonal Gammopathy of Undetermined Significance/genetics , Myeloid Differentiation Factor 88/genetics , Plasma Cells/metabolism , Point Mutation , Animals , B-Lymphocytes/immunology , Cell Proliferation , Cell Survival , Cells, Cultured , Genetic Predisposition to Disease , Immunoglobulin M/immunology , Lymphocyte Activation , Mice, Inbred C57BL , Monoclonal Gammopathy of Undetermined Significance/blood , Monoclonal Gammopathy of Undetermined Significance/immunology , Myeloid Differentiation Factor 88/metabolism , Paraproteins/metabolism , Phenotype , Plasma Cells/immunology
16.
Front Immunol ; 11: 854, 2020.
Article in English | MEDLINE | ID: mdl-32536913

ABSTRACT

Previous studies showed that monoclonal immunoglobulins G (IgGs) of "monoclonal gammopathy of undetermined significance" (MGUS) and myeloma were hyposialylated, thus presumably pro-inflammatory, and for about half of patients, the target of the monoclonal IgG was either a virus-Epstein-Barr virus (EBV), other herpes viruses, hepatitis C virus (HCV)-or a glucolipid, lysoglucosylceramide (LGL1), suggesting antigen-driven disease in these patients. In the present study, we show that monoclonal IgAs share these characteristics. We collected 35 sera of patients with a monoclonal IgA (6 MGUS, 29 myeloma), and we were able to purify 25 of the 35 monoclonal IgAs (6 MGUS, 19 myeloma). Monoclonal IgAs from MGUS and myeloma patients were significantly less sialylated than IgAs from healthy volunteers. When purified monoclonal IgAs were tested against infectious pathogens and LGL1, five myeloma patients had a monoclonal IgA that specifically recognized viral proteins: the core protein of HCV in one case, EBV nuclear antigen 1 (EBNA-1) in four cases (21.1% of IgA myeloma). Monoclonal IgAs from three myeloma patients reacted against LGL1. In summary, monoclonal IgAs are hyposialylated and as described for IgG myeloma, significant subsets (8/19, or 42%) of patients with IgA myeloma may have viral or self (LGL1) antigen-driven disease.


Subject(s)
Antibodies, Monoclonal/blood , Immunoglobulin A/blood , Monoclonal Gammopathy of Undetermined Significance/blood , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/blood , Multiple Myeloma/immunology , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Cohort Studies , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/immunology , Female , Glucosylceramides/immunology , Glycosylation , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Herpesvirus 4, Human/immunology , Humans , Immunoglobulin A/immunology , Male , Middle Aged , Viral Core Proteins/immunology
18.
Curr Hematol Malig Rep ; 15(2): 45-55, 2020 04.
Article in English | MEDLINE | ID: mdl-32222885

ABSTRACT

PURPOSE OF THE REVIEW: Monoclonal gammopathy of undetermined significance (MGUS) is a highly prevalent precursor condition in the general population, with an approximate 1% annual risk of progression to multiple myeloma (MM) or a related disorder. Our understanding of MGUS and its association with myriad clinical disorders, its progression to MM, and the genomic alterations in the setting of a conducive or permissive microenvironment has deepened considerably. RECENT FINDINGS: Data from gene expression profiling studies have underscored the heterogeneity in the risk of progression of MGUS to MM. Ongoing efforts are being directed toward precise identification of high-risk factors for progression and addressing the role of screening for MGUS in populations at higher risk of MGUS in order to diagnose this precursor condition early, and target modifiable risk factors. Ongoing clinical trials are assessing the role of therapeutic interventions to prevent MGUS from progressing. MGUS is a heterogeneous precursor condition with a risk for progression to symptomatic disease. Future directions are focusing on identifying high-risk populations of MGUS and smoldering multiple myeloma that may benefit with screening and/or early intervention.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Precancerous Conditions , Biomarkers, Tumor/genetics , Disease Progression , Genetic Predisposition to Disease , Humans , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/epidemiology , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/diagnosis , Multiple Myeloma/epidemiology , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Precancerous Conditions/diagnosis , Precancerous Conditions/epidemiology , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Risk Assessment , Risk Factors , Transcriptome , Tumor Microenvironment
19.
Sci Rep ; 10(1): 1983, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029833

ABSTRACT

To understand neutrophil impairment in the progression from MGUS through active MM, we investigated the function of mature, high-density neutrophils (HDNs), isolated from peripheral blood. In 7 MM, 3 MGUS and 3 healthy subjects by gene expression profile, we identified a total of 551 upregulated and 343 downregulated genes in MM-HDN, involved in chemokine signaling pathway and FC-gamma receptor mediated phagocytosis conveying in the activation of STAT proteins. In a series of 60 newly diagnosed MM and 30 MGUS patients, by flow-cytometry we found that HDN from MM, and to a lesser extend MGUS, had an up-regulation of the inducible FcγRI (also known as CD64) and a down-regulation of the constitutive FcγRIIIa (also known as CD16) together with a reduced phagocytic activity and oxidative burst, associated to increased immune-suppression that could be reverted by arginase inhibitors in co-culture with lymphocytes. In 43 consecutive newly-diagnosed MM patients, who received first-line treatment based on bortezomib, thalidomide and dexamethasone, high CD64 could identify at diagnosis patients with inferior median overall survival (39.5 versus 86.7 months, p = 0.04). Thus, HDNs are significantly different among healthy, MGUS and MM subjects. In both MGUS and MM neutrophils may play a role in supporting both the increased susceptibility to infection and the immunological dysfunction that leads to tumor progression.


Subject(s)
Disease Susceptibility/immunology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , Neutrophils/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Case-Control Studies , Disease Progression , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/drug therapy , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/mortality , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Signal Transduction/genetics , Tumor Escape/genetics
20.
Front Immunol ; 11: 613007, 2020.
Article in English | MEDLINE | ID: mdl-33488620

ABSTRACT

Background: The 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. Methods: The 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. Results: First, 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. Conclusions: Our 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.


Subject(s)
B7-H1 Antigen/immunology , Bone Marrow/immunology , Programmed Cell Death 1 Receptor/immunology , Smoldering Multiple Myeloma/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Interleukin-27/immunology , Interleukin-6/immunology , Lipopolysaccharide Receptors/immunology , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/immunology , Monocytes/immunology , Paraproteinemias/immunology , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...