Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
Int J Mol Sci ; 23(24)2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36555330

ABSTRACT

Multiple myeloma (MM) has a highly heterogeneous genetic background, which complicates its molecular tracking over time. Nevertheless, each MM patient's malignant plasma cells (PCs) share unique V(D)J rearranged sequences at immunoglobulin loci, which represent ideal disease biomarkers. Because the tumor-specific V(D)J sequence is highly expressed in bulk RNA in MM patients, we wondered whether it can be identified by single-cell RNA sequencing (scRNA-seq). To this end we analyzed CD138+ cells purified from bone marrow aspirates of 19 samples with PC dyscrasias by both a standard method based on bulk DNA and by an implementation of the standard 10x Genomics protocol to detect expressed V(D)J sequences. A dominant clonotype was easily identified in each sample, accounting on average for 83.65% of V(D)J-rearranged cells. Compared with standard methods, scRNA-seq analysis proved highly concordant and even more effective in identifying clonal productive rearrangements, by-passing limitations related to the misannealing of consensus primers in hypermutated regions. We next validated its accuracy to track 5 clonal cells with absolute sensitivity in a virtual sample containing 3180 polyclonal cells. This shows that single-cell V(D)J analysis may be used to find rare clonal cells, laying the foundations for functional single-cell dissection of minimal residual disease.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/pathology , Immunoglobulin Heavy Chains/genetics , V(D)J Recombination , Gene Rearrangement , Sequence Analysis, RNA
3.
Clin Cancer Res ; 27(23): 6479-6490, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34526359

ABSTRACT

PURPOSE: Multiple myeloma is a biologically heterogenous plasma-cell disorder. In this study, we aimed at dissecting the functional impact on transcriptome of gene mutations, copy-number abnormalities (CNA), and chromosomal rearrangements (CR). Moreover, we applied a geno-transcriptomic approach to identify specific biomarkers for personalized treatments. EXPERIMENTAL DESIGN: We analyzed 514 newly diagnosed patients from the IA12 release of the CoMMpass study, accounting for mutations in multiple myeloma driver genes, structural variants, copy-number segments, and raw-transcript counts. We performed an in silico drug sensitivity screen (DSS), interrogating the Cancer Dependency Map (DepMap) dataset after anchoring cell lines to primary tumor samples using the Celligner algorithm. RESULTS: Immunoglobulin translocations, hyperdiploidy and chr(1q)gain/amps were associated with the highest number of deregulated genes. Other CNAs and specific gene mutations had a lower but very distinct impact affecting specific pathways. Many recurrent genes showed a hotspot (HS)-specific effect. The clinical relevance of double-hit multiple myeloma found strong biological bases in our analysis. Biallelic deletions of tumor suppressors and chr(1q)-amplifications showed the greatest impact on gene expression, deregulating pathways related to cell cycle, proliferation, and expression of immunotherapy targets. Moreover, our in silico DSS showed that not only t(11;14) but also chr(1q)gain/amps and CYLD inactivation predicted differential expression of transcripts of the BCL2 axis and response to venetoclax. CONCLUSIONS: The multiple myeloma genomic architecture and transcriptome have a strict connection, led by CNAs and CRs. Gene mutations impacted especially with HS-mutations of oncogenes and biallelic tumor suppressor gene inactivation. Finally, a comprehensive geno-transcriptomic analysis allows the identification of specific deregulated pathways and candidate biomarkers for personalized treatments in multiple myeloma.


Subject(s)
Multiple Myeloma , Gene Expression Profiling , Genomics , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Oncogenes , Transcriptome
4.
Cancers (Basel) ; 13(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34282760

ABSTRACT

Smoldering multiple myeloma (SMM) is an asymptomatic disorder of clonal bone marrow (BM) plasma cells (PCs) in between the premalignant condition known as monoclonal gammopathy of undetermined significance and overt multiple myeloma (MM). It is characterized by a deep biological heterogeneity that is reflected in a markedly variable progression risk among patients. Recently proposed risk stratification models mainly rely on indirect markers of disease burden and are unable to identify cases in whom clonal PCs have already undergone the "malignant switch" but major clonal expansion has not occurred yet. In the last years, the application of next-generation sequencing (NGS) techniques has led to profound advances in the understanding of the molecular bases of SMM progression, and in all likelihood, it will contribute to the needed improvement of SMM prognostication. In this Review, we describe the recent advances in characterizing the genomic landscape of SMM and intrinsic determinants of its progression, highlighting their implications in terms of understanding of tumor evolution and prognostication. We also review the main studies investigating the role of the microenvironment in this early disease stage. Finally, we mention the results of the first randomized clinical trials and discuss the potential clinical translability of the genomic insights.

5.
Blood Cancer Discov ; 2(4): 338-353, 2021 07.
Article in English | MEDLINE | ID: mdl-34268498

ABSTRACT

The bone marrow (BM) microenvironment actively promotes multiple myeloma (MM) pathogenesis and therapies targeting both cancer cells and the niche are highly effective. We were interested in identifying novel signaling pathways supporting MM-BM crosstalk. Mutations in the transmembrane receptor Roundabout 1 (ROBO1) were recently identified in MM patients, however their functional consequences are uncertain. Through protein structure-function studies, we discovered that ROBO1 is necessary for MM adhesion to BM stromal and endothelial cells and ROBO1 knock out (KO) compromises BM homing and engraftment in a disseminated mouse model. ROBO1 KO significantly decreases MM proliferation in vitro and intra- and extramedullary tumor growth, in vivo. Mechanistically, ROBO1 C-terminus is cleaved in a ligand-independent fashion and is sufficient to promote MM proliferation. Viceversa, mutants lacking the cytoplasmic domain, including the human-derived G674* truncation, act dominantly negative. Interactomic and RNA sequencing studies suggest ROBO1 may be involved in RNA processing, supporting further studies.


Subject(s)
Bone Marrow , Multiple Myeloma , Nerve Tissue Proteins , Receptors, Immunologic , Animals , Bone Marrow/metabolism , Bone Marrow Cells , Endothelial Cells/metabolism , Humans , Mice , Multiple Myeloma/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Tumor Microenvironment/genetics , Roundabout Proteins
6.
Nat Med ; 27(4): 616-619, 2021 04.
Article in English | MEDLINE | ID: mdl-33619368

ABSTRACT

B cell maturation antigen (BCMA) is a target for various immunotherapies and a biomarker for tumor load in multiple myeloma (MM). We report a case of irreversible BCMA loss in a patient with MM who was enrolled in the KarMMa trial ( NCT03361748 ) and progressed after anti-BCMA CAR T cell therapy. We identified selection of a clone with homozygous deletion of TNFRSF17 (BCMA) as the underlying mechanism of immune escape. Furthermore, we found heterozygous TNFRSF17 loss or monosomy 16 in 37 out of 168 patients with MM, including 28 out of 33 patients with hyperhaploid MM who had not been previously treated with BCMA-targeting therapies, suggesting that heterozygous TNFRSF17 deletion at baseline could theoretically be a risk factor for BCMA loss after immunotherapy.


Subject(s)
B-Cell Maturation Antigen/genetics , Gene Deletion , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Aged , Antigens, Neoplasm/metabolism , DNA Copy Number Variations/genetics , Homozygote , Humans , Magnetic Resonance Imaging , Male , Multiple Myeloma/diagnostic imaging
7.
Haematologica ; 106(7): 1943-1956, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32354870

ABSTRACT

Interactions of malignant multiple myeloma (MM) plasma cells (MM-cells) with the microenvironment control MM-cell growth, survival, drug-resistance and dissemination. As in MM microvascular density increases in the bone marrow (BM), we investigated whether BM MM endothelial cells (MMECs) control disease progression via the junctional adhesion molecule A (JAM-A). Membrane and cytoplasmic JAM-A levels were upregulated in MMECs in 111 newly diagnosed (NDMM) and 201 relapsed-refractory (RRMM) patients compared to monoclonal gammopathy of undetermined significance (MGUS) and healthy controls. Elevated membrane expression of JAM-A on MMECs predicted poor clinical outcome. Mechanistically, addition of recombinant JAM-A to MMECs increased angiogenesis whereas its inhibition impaired angiogenesis and MM growth in 2D and 3D in vitro cell culture and chorioallantoic membrane-assays. To corroborate these findings, we treated MM bearing mice with JAM-A blocking mAb and demonstrated impaired MM progression corresponding to decreased MM-related vascularity. These findings support JAM-A as an important mediator of MM progression through facilitating MM-associated angiogenesis. Collectively, elevated JAM-A expression on bone marrow endothelial cells is an independent prognostic factor for patient survival in both NDMM and RRMM. Blocking JAM-A restricts angiogenesis in vitro, in embrio and in vivo and represents a suitable druggable molecule to halt neoangiogenesis and MM progression.


Subject(s)
Junctional Adhesion Molecule A , Multiple Myeloma , Animals , Bone Marrow , Ecosystem , Endothelial Cells , Homeostasis , Humans , Mice , Multiple Myeloma/drug therapy , Tumor Microenvironment
8.
Hemasphere ; 4(6): e502, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33283171

ABSTRACT

The knowledge of cancer origin and the subsequent tracking of disease evolution represent unmet needs that will soon be within clinical reach. This will provide the opportunity to improve patient's stratification and to personalize treatments based on cancer biology along its life history. In this review, we focus on the molecular pathogenesis of multiple myeloma (MM), a hematologic malignancy with a well-known multi-stage disease course, where such approach can sooner translate into a clinical benefit. We describe novel insights into modes and timing of disease initiation. We dissect the biology of the preclinical and pre-malignant phases, elucidating how knowledge of the genomics of the disease and the composition of the microenvironment allow stratification of patients based on risk of disease progression. Then, we explore cell-intrinsic and cell-extrinsic drivers of MM evolution to symptomatic disease. Finally, we discuss how this may relate to the development of refractory disease after treatment. By integrating an evolutionary view of myeloma biology with the recent acquisitions on its clonal heterogeneity, we envision a way to drive the clinical management of the disease based on its detailed biological features more than surrogates of disease burden.

9.
Blood ; 124(9): 1513-21, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24970933

ABSTRACT

Our knowledge of the genetic basis of myelodysplastic syndromes (MDS) and myelodysplastic/myeloproliferative neoplasms (MDS/MPN) has considerably improved. To define genotype/phenotype relationships of clinical relevance, we studied 308 patients with MDS, MDS/MPN, or acute myeloid leukemia evolving from MDS. Unsupervised statistical analysis, including the World Health Organization classification criteria and somatic mutations, showed that MDS associated with SF3B1-mutation (51 of 245 patients, 20.8%) is a distinct nosologic entity irrespective of current morphologic classification criteria. Conversely, MDS with ring sideroblasts with nonmutated SF3B1 segregated in different clusters with other MDS subtypes. Mutations of genes involved in DNA methylation, splicing factors other than SF3B1, and genes of the RAS pathway and cohesin complex were independently associated with multilineage dysplasia and identified a distinct subset (51 of 245 patients, 20.8%). No recurrent mutation pattern correlated with unilineage dysplasia without ring sideroblasts. Irrespective of driver somatic mutations, a threshold of 5% bone marrow blasts retained a significant discriminant value for identifying cases with clonal evolution. Comutation of TET2 and SRSF2 was highly predictive of a myeloid neoplasm characterized by myelodysplasia and monocytosis, including but not limited to, chronic myelomonocytic leukemia. These results serve as a proof of concept that a molecular classification of myeloid neoplasms is feasible.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic-Myeloproliferative Diseases/genetics , Adult , Aged , Aged, 80 and over , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Cohort Studies , Core Binding Factor Alpha 2 Subunit/genetics , DNA Methylation/genetics , Female , Genes, ras , Genetic Association Studies , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/pathology , Myelodysplastic-Myeloproliferative Diseases/classification , Myelodysplastic-Myeloproliferative Diseases/pathology , Myeloid Cells/pathology , Phosphoproteins/genetics , Prognosis , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/genetics , Cohesins
10.
Haematologica ; 98(3): 420-3, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23300182

ABSTRACT

Somatic mutations of the RNA splicing machinery have been recently identified in myelodysplastic syndromes. In particular, a strong association has been found between SF3B1 mutation and refractory anemia with ring sideroblasts, a condition characterized by ineffective erythropoiesis and parenchymal iron overload. We studied the relationship between SF3B1 mutation, erythroid activity and hepcidin levels in myelodysplastic syndrome patients. Erythroid activity was evaluated through the proportion of marrow erythroblasts, soluble transferrin receptor and serum growth differentiation factor 15. Significant relationships were found between SF3B1 mutation and marrow erythroblasts (P=0.001), soluble transferrin receptor (P=0.003) and serum growth differentiation factor 15 (P=0.033). Serum hepcidin varied considerably, and multivariable analysis showed that the hepcidin to ferritin ratio, a measure of adequacy of hepcidin levels relative to body iron stores, was inversely related to the SF3B1 mutation (P=0.013). These observations suggest that patients with SF3B1 mutation have inappropriately low hepcidin levels, which may explain their propensity to parenchymal iron loading.


Subject(s)
Hepcidins/blood , Mutation , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Aged , Aged, 80 and over , Alleles , Female , Ferritins/blood , Humans , Iron/metabolism , Male , Middle Aged , RNA Splicing Factors
11.
Am J Hematol ; 87(7): 746-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22565617

ABSTRACT

JAK2 (V617F) is associated with a genetic predisposition to its acquisition,as it is preferentially found in subjects with a common constitutional JAK2 haplotype known as 46/1 or GGCC. A recent study suggests that a genetic predisposition to acquisition of MPL mutation may exist in sporadic patients, since an association was found with the JAK2 46/1 haplotype. We genotyped 509 patients with myeloproliferative neoplasms (MPN), 7% of which carrying a somatic mutation of MPL Exon 10. We found that the JAK2 GGCC haplotype was closely associated with JAK2 (V617F) (OR 1.84, P < 0.001) but not with MPL mutations (OR 0.98), suggesting a different genetic background for these molecular lesions.


Subject(s)
Haplotypes , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Polymorphism, Single Nucleotide , Receptors, Thrombopoietin/genetics , Alleles , Amino Acid Substitution , Cohort Studies , Exons , Gene Frequency , Genetic Association Studies , Humans , Italy , Janus Kinase 2/metabolism , Myeloproliferative Disorders/metabolism , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Receptors, Thrombopoietin/metabolism , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/metabolism
12.
Blood ; 119(1): 188-91, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22072557

ABSTRACT

The somatically acquired V600E mutation of the BRAF gene has been recently described as a molecular marker of hairy cell leukemia (HCL). We developed an allele-specific PCR for this mutation and studied 62 patients with HCL, 1 with HCL variant, 91 with splenic marginal zone lymphoma, 29 with Waldenström macroglobulinemia, and 57 with B-cell chronic lymphoproliferative disorders. The BRAF V600E mutation was detected in all HCL cases and in only 2 of the remaining 178 patients. These 2 subjects had B-cell chronic lymphoproliferative disorders that did not fulfill the diagnostic criteria for HCL. Despite the positive PCR finding, the mutation could not be detected by Sanger sequencing in these 2 cases, suggesting that it was associated with a small subclone. We conclude that the BRAF V600E mutation is present in all patients with HCL and that, in combination with clinical and morphologic features, represents a reliable molecular marker for this condition.


Subject(s)
Leukemia, B-Cell/genetics , Leukemia, Hairy Cell/genetics , Lymphoma, B-Cell/genetics , Point Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Adult , DNA, Neoplasm/genetics , Exons/genetics , Female , Humans , Leukemia, B-Cell/pathology , Leukemia, Hairy Cell/pathology , Lymphoma, B-Cell/pathology , Male , Middle Aged , Polymerase Chain Reaction
13.
Blood ; 118(24): 6239-46, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21998214

ABSTRACT

In a previous study, we identified somatic mutations of SF3B1, a gene encoding a core component of RNA splicing machinery, in patients with myelodysplastic syndrome (MDS). Here, we define the clinical significance of these mutations in MDS and myelodysplastic/myeloproliferative neoplasms (MDS/MPN). The coding exons of SF3B1 were screened using massively parallel pyrosequencing in patients with MDS, MDS/MPN, or acute myeloid leukemia (AML) evolving from MDS. Somatic mutations of SF3B1 were found in 150 of 533 (28.1%) patients with MDS, 16 of 83 (19.3%) with MDS/MPN, and 2 of 38 (5.3%) with AML. There was a significant association of SF3B1 mutations with the presence of ring sideroblasts (P < .001) and of mutant allele burden with their proportion (P = .002). The mutant gene had a positive predictive value for ring sideroblasts of 97.7% (95% confidence interval, 93.5%-99.5%). In multivariate analysis including established risk factors, SF3B1 mutations were found to be independently associated with better overall survival (hazard ratio = 0.15, P = .025) and lower risk of evolution into AML (hazard ratio = 0.33, P = .049). The close association between SF3B1 mutations and disease phenotype with ring sideroblasts across MDS and MDS/MPN is consistent with a causal relationship. Furthermore, SF3B1 mutations are independent predictors of favorable clinical outcome, and their incorporation into stratification systems might improve risk assessment in MDS.


Subject(s)
Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Myelodysplastic-Myeloproliferative Diseases/genetics , Myelodysplastic-Myeloproliferative Diseases/physiopathology , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Aged , Alleles , Codon , DNA Mutational Analysis , Erythroblasts/pathology , Female , Follow-Up Studies , Genetic Association Studies , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/pathology , Prognosis , RNA Splicing Factors , Sex Characteristics , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...