Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
2.
Med. clín (Ed. impr.) ; 161(5): 217-224, sept. 2023.
Article in Spanish | IBECS | ID: ibc-224739

ABSTRACT

La gammapatía monoclonal de significado incierto es una neoplasia de células plasmáticas premaligna, con una elevada prevalencia en la población mayor de 50 años, y un riesgo anual de progresión del 1%. Numerosos estudios recientes han permitido un avance en la compresión de la patogenia de estos trastornos y su riesgo de progresión a otras enfermedades. Los pacientes requieren un seguimiento de por vida, siendo fundamental un enfoque multidisciplinar y adaptado al riesgo. En los últimos años, cada vez se reconocen más entidades asociadas a la presencia de una paraproteína, conocidas como gammapatías monoclonales de significado clínico (AU)


Monoclonal gammopathy of uncertain significance is a premalignant plasma cell neoplasm with a high prevalence in the population over 50 years of age and an annual risk of progression of 1%. Multiple recent studies have led to advances in understanding both the pathogenesis of these disorders and their risk of progression to other diseases. Patients require lifelong follow-up, and a multidisciplinary and risk-adapted approach is essential. In recent years, an increasing number of entities associated with a paraprotein, known as clinically significant monoclonal gammopathies, have been recognized (AU)


Subject(s)
Humans , Genetic Predisposition to Disease , Paraproteinemias/diagnosis , Paraproteinemias/genetics , Paraproteinemias/etiology , Paraproteinemias/therapy , Mass Screening , Prognosis
3.
Front Immunol ; 14: 1203425, 2023.
Article in English | MEDLINE | ID: mdl-37520549

ABSTRACT

The adaptive immune system of jawed vertebrates generates a highly diverse repertoire of antibodies to meet the antigenic challenges of a constantly evolving biological ecosystem. Most of the diversity is generated by two mechanisms: V(D)J gene recombination and somatic hypermutation (SHM). SHM introduces changes in the variable domain of antibodies, mostly in the regions that form the paratope, yielding antibodies with higher antigen binding affinity. However, antigen recognition is only possible if the antibody folds into a stable functional conformation. Therefore, a key force determining the survival of B cell clones undergoing somatic hypermutation is the ability of the mutated heavy and light chains to efficiently fold and assemble into a functional antibody. The antibody is the structural context where the selection of the somatic mutations occurs, and where both the heavy and light chains benefit from protective mechanisms that counteract the potentially deleterious impact of the changes. However, in patients with monoclonal gammopathies, the proliferating plasma cell clone may overproduce the light chain, which is then secreted into the bloodstream. This places the light chain out of the protective context provided by the quaternary structure of the antibody, increasing the risk of misfolding and aggregation due to destabilizing somatic mutations. Light chain-derived (AL) amyloidosis, light chain deposition disease (LCDD), Fanconi syndrome, and myeloma (cast) nephropathy are a diverse group of diseases derived from the pathologic aggregation of light chains, in which somatic mutations are recognized to play a role. In this review, we address the mechanisms by which somatic mutations promote the misfolding and pathological aggregation of the light chains, with an emphasis on AL amyloidosis. We also analyze the contribution of the variable domain (VL) gene segments and somatic mutations on light chain cytotoxicity, organ tropism, and structure of the AL fibrils. Finally, we analyze the most recent advances in the development of computational algorithms to predict the role of somatic mutations in the cardiotoxicity of amyloidogenic light chains and discuss the challenges and perspectives that this approach faces.


Subject(s)
Multiple Myeloma , Paraproteinemias , Animals , Humans , Friends , Ecosystem , B-Lymphocytes , Paraproteinemias/genetics
4.
Am J Hematol ; 98(2): 264-271, 2023 02.
Article in English | MEDLINE | ID: mdl-36588407

ABSTRACT

Familial forms of monoclonal gammopathy, defined as multiple myeloma (MM) or Monoclonal Gammopathy of Undetermined Significance (MGUS), are relatively infrequent and most series reported in the literature describe a limited number of families. MM rarely occurs in a familial context. MGUS is observed much more commonly, which can in some cases evolve toward full-blown MM. Although recurrent cytogenetic abnormalities have been described in tumor cells of sporadic cases of MM, the pathogenesis of familial MM remains largely unexplained. In order to identify genetic factors predisposing to familial monoclonal gammopathy, the Intergroupe Francophone du Myélome identified 318 families with at least two confirmed cases of monoclonal gammopathy. There were 169 families with parent/child pairs and 164 families with cases in at least two siblings, compatible with an autosomal transmission. These familial cases were compared with sporadic cases who were matched for age at diagnosis, sex and immunoglobulin isotype, with 10 sporadic cases for each familial case. The gender distribution, age and immunoglobulin subtypes of familial cases were unremarkable in comparison to sporadic cases. With a median follow-up of 7.4 years after diagnosis, the percentage of MGUS cases having evolved to MM was 3%. The median overall survival of the 148 familial MM cases was longer than that of matched sporadic cases, with projected values of 7.6 and 16.1 years in patients older and younger than 65 years, respectively. These data suggest that familial cases of monoclonal gammopathy are similar to sporadic cases in terms of clinical presentation and carry a better prognosis.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Paraproteinemias , Child , Humans , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Paraproteinemias/genetics , Paraproteinemias/complications , Multiple Myeloma/pathology , Prognosis , Chromosome Aberrations
5.
Semin Oncol ; 49(6): 465-475, 2022 12.
Article in English | MEDLINE | ID: mdl-36503855

ABSTRACT

Clonal hematopoiesis of indeterminate potential (CHIP) is common both in healthy individuals and patients with hematological cancers. Recent studies have showed worse prognosis for patients with multiple myeloma (MM) and non-Hodgkin lymphoma undergoing stem cell transplant, that have concomitant presence of CHIP. Data regarding the clinical and biological role of CHIP in plasma cell dyscrasias (PCDs) is rapidly increasing. However, the prevalence and prognostic implication of CHIP in patients with MM outside of the transplant setting, and in those with other more indolent PCDs remains elusive. Here we explored the prevalence and clinical implications of CHIP detected through next-generation sequencing in 209 patients with PCDs including MM, light chain (AL) amyloidosis (ALA), monoclonal gammopathy of undetermined significance (MGUS), and smoldering multiple myeloma (SMM). To avoid attributing the mutations to the plasma cell clone, CHIP was defined as the presence of DNMT3A, TET2, or ASXL1 mutations in the peripheral blood or bone marrow (DTA-CH). The prevalence of DTA-CH was 19% in patients with PCDs, with no difference between each PCD. TET2 (23%) and DNMT3A (22%), were the most frequently mutated genes. DTA-CH correlated with older age in MM (P = .001) and MGUS/SMM (P = 0.0007), as well as with coronary artery disease or congestive heart failure in MM (P = .03). DTA-CH did not predict worse OS or PFS in either MM or ALA, nor it predict higher risk of progression to MM in patients with MGUS/SMM. Our results overall further elucidate the prevalence and mutational spectrum of CHIP in PCDs, providing more information regarding the clinical relevance of CHIP in this patient population.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Paraproteinemias , Smoldering Multiple Myeloma , Humans , Clonal Hematopoiesis , Prevalence , Paraproteinemias/genetics , Paraproteinemias/pathology , Mutation
6.
BMC Med Genomics ; 15(1): 203, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36138464

ABSTRACT

BACKGROUND: Next-generation sequencing (NGS) detects somatic mutations in a high proportion of plasma cell dyscrasias (PCD), but is currently not integrated into diagnostic routine. We correlated NGS data with degree of bone marrow (BM) involvement by cytomorphology (BMC), histopathology (BMH), and multiparameter flow cytometry (MFC) in 90 PCD patients. METHODS: Of the 90 patients the diagnoses comprised multiple myeloma (n = 77), MGUS (n = 7), AL-amyloidosis (n = 4) or solitary plasmocytoma (n = 2). The NGS panel included eight genes CCND1, DIS3, EGR1, FAM46C (TENT5C), FGFR3, PRDM1, TP53, TRAF3, and seven hotspots in BRAF, IDH1, IDH2, IRF4, KRAS, NRAS. RESULTS: Mutations were detected in 64/90 (71%) of cases. KRAS (29%), NRAS (16%) and DIS3 (16%) were most frequently mutated. At least one mutation/sample corresponded to a higher degree of BM involvement with a mean of 11% pathologic PC by MFC (range, 0.002-62%), and ~ 50% (3-100%) as defined by both BMC and BMH. CONCLUSIONS: The probability of detecting a mutation by NGS in the BM was highest in samples with > 10% clonal PC by MFC, or > 20% PC by BMC/ BMH. We propose further evaluation of these thresholds as a practical cut-off for processing of samples by NGS at initial PCD diagnosis.


Subject(s)
Paraproteinemias , Proto-Oncogene Proteins B-raf , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation , Paraproteinemias/genetics , Paraproteinemias/pathology , Plasma Cells/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , TNF Receptor-Associated Factor 3/genetics
8.
J Clin Lab Anal ; 36(4): e24287, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35170077

ABSTRACT

BACKGROUND: Study of the molecular biological characteristics of chronic neutrophilic leukemia complicated with plasma cell disorder (CNL-PCD) and lymphocytic proliferative disease (CNL-LPD). METHODS: The clinical data of a patient with chronic neutrophilic leukemia complicated with monoclonal gammopathy of undetermined significance (CNL-MGUS) in our hospital were reviewed, and the Chinese and/or English literature about CNL-PCD and CNL-LPD in PubMed and the Chinese database CNKI in the past 10 years was searched to analyze the molecular biological characteristics of this disease. RESULTS: A 73-year-old male had persistent leukocytosis for 18 months. The white blood cell count was 46.77 × 109/L and primarily composed of mature neutrophils; hemoglobin: 77 g/L; platelet count: 189 × 109/L. Serum immunofixation electrophoresis showed IgG-λ monoclonal M protein. A CT scan showed splenomegaly. Next-generation sequencing (NGS) showed that CSF3R T618I, ASXL1 and RUNX1 mutations were positive. It was diagnosed as CNL-MGUS. We summarized 10 cases of CNL-PCD and 1 case of CNL-LPD who underwent genetic mutation detection reported in the literature. The CSF3R mutational frequency (7/11, 63.6%) was lower than that of isolated CNL. The ASXL1 mutations were all positive (3/3), which may represent a poor prognostic factor. The SETBP1 mutation may promote the progression of CNL-PCD. We also found JAK2, RUNX1, NRAS, etc. in CNL-PCD. CONCLUSIONS: Chronic neutrophilic leukemia may be more inclined to coexist with plasma cell disorder. The CSF3R mutation in CNL-PCD is still the most common mutated gene compared with isolated CNL. Mutations in SETBP1 and ASXL1 may be poor prognostic factors for CNL-PCD.


Subject(s)
Leukemia, Neutrophilic, Chronic , Monoclonal Gammopathy of Undetermined Significance , Paraproteinemias , Aged , Core Binding Factor Alpha 2 Subunit/genetics , Humans , Leukemia, Neutrophilic, Chronic/complications , Leukemia, Neutrophilic, Chronic/diagnosis , Leukemia, Neutrophilic, Chronic/genetics , Male , Monoclonal Gammopathy of Undetermined Significance/complications , Monoclonal Gammopathy of Undetermined Significance/genetics , Mutation/genetics , Paraproteinemias/complications , Paraproteinemias/genetics
9.
Kidney Int ; 101(2): 331-337, 2022 02.
Article in English | MEDLINE | ID: mdl-34767830

ABSTRACT

The diagnostic approach of monoclonal gammopathy of renal significance is based on the detection of a monoclonal immunoglobulin in the blood and urine, and the identification of the underlying clone through bone marrow and/or peripheral blood cytologic and flow cytometry analysis. However, the monoclonal component and its corresponding clone may be undetectable using these routine techniques. Since clone identification is the cornerstone for guiding therapy and assessing disease response, more sensitive methods are required. We recently developed a high-throughput sequencing assay from bone marrow mRNA encoding immunoglobulins (RACE-RepSeq). This technique provides both full-length V(D)J region (variable, diversity and joining genes that generate unique receptors as antigen receptors) of the monoclonal immunoglobulin and the dominant immunoglobulin repertoire. This allows analysis of mutational patterns, immunoglobulin variable gene frequencies and diversity due to somatic hypermutation. Here, we evaluated the diagnostic performance of RACE-RepSeq in 16 patients with monoclonal-associated kidney lesions, and low serum monoclonal immunoglobulin and free light chain levels at diagnosis. Bone marrow immunohistochemical analysis was negative in all 11 patients so tested and 7 of 12 patients had no detectable clone matching the kidney deposits using flow cytometry analysis. By contrast, RACE-RepSeq detected a dominant clonal light chain sequence of matched isotype with respect to kidney deposits in all patients. Thus, high throughput mRNA sequencing appears highly sensitive to detect subtle clonal disorders in monoclonal gammopathy of renal significance and suggest this novel approach could help improve the management of this kidney disease.


Subject(s)
Kidney Diseases , Paraproteinemias , Humans , Immunoglobulin Light Chains , Kidney/pathology , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Kidney Diseases/therapy , Paraproteinemias/diagnosis , Paraproteinemias/genetics , Paraproteinemias/therapy , RNA
11.
Br J Haematol ; 190(6): 901-908, 2020 09.
Article in English | MEDLINE | ID: mdl-32712965

ABSTRACT

The relationship between chronic lymphocytic leukaemia (CLL) and qualitative/quantitative gammaglobulin abnormalities is well established. Nevertheless, in order to better understand this kind of connection, we examined 1505 patients with CLL and divided them into four subgroups on the basis of immunoglobulin (Ig) aberrations at diagnosis. A total of 73 (4·8%), 149 (10%), 200 (13·2%) and 1083 (72%) patients were identified with IgM monoclonal gammopathy (IgM/CLL), IgG monoclonal gammopathy (IgG/CLL), hypogammaglobulinaemia (hypo-γ) and normal Ig levels (γ-normal) respectively. IgM paraprotein was significantly associated with a more advanced Binet/Rai stage and del(17p)/TP53 mutation, while IgG abnormalities correlated with a higher occurrence of trisomy 12. Patients with any type of Ig abnormality had shorter treatment-free survival (TFS) but no significant impact affecting overall survival (OS) compared to those with normal Ig levels.


Subject(s)
Immunoglobulin G , Immunoglobulin M , Leukemia, Lymphocytic, Chronic, B-Cell , Neoplasm Proteins , Paraproteinemias , Adult , Aged , Aged, 80 and over , Chromosome Deletion , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 17/genetics , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunoglobulin M/blood , Immunoglobulin M/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Paraproteinemias/blood , Paraproteinemias/genetics , Paraproteinemias/mortality , Retrospective Studies , Smith-Magenis Syndrome/blood , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/mortality , Survival Rate , Trisomy , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
14.
Ann Hematol ; 99(4): 703-714, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32103323

ABSTRACT

Monoclonal gammopathy of renal significance (MGRS) is characterized by the nephrotoxic monoclonal immunoglobulin secreted by an otherwise asymptomatic or indolent B cell or plasma cell clone, without hematologic criteria for treatment. These MGRS-associated diseases can involve one or more renal compartments, including glomeruli, tubules, and vessels. Hydrophobic residue replacement, N-glycosylated, increase in isoelectric point in monoclonal immunoglobulin (MIg) causes it to transform from soluble form to tissue deposition, and consequently resulting in glomerular damage. In addition to MIg deposition, complement deposition is also found in C3 glomerulopathy with monoclonal glomerulopathy, which is caused by an abnormality of the alternative pathway and may involve multiple factors including complement component 3 nephritic factor, anti-complement factor auto-antibodies, or MIg which directly cleaves C3. Furthermore, inflammatory factors, growth factors, and virus infection may also participate in the development of the diseases. In this review, for the first time, we discussed current highlights in the mechanism of MGRS-related lesions.


Subject(s)
Antibodies, Monoclonal/metabolism , Kidney Diseases/etiology , Paraproteinemias/etiology , Paraproteins/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , Complement C3/metabolism , Complement C3 Nephritic Factor/metabolism , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3-C5 Convertases/metabolism , Complement Pathway, Alternative , Cryoglobulinemia/etiology , Cryoglobulinemia/metabolism , Glycosylation , Humans , Immunoglobulin Light-chain Amyloidosis/etiology , Immunoglobulin Light-chain Amyloidosis/metabolism , Inflammation Mediators/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Neoplasm Proteins/metabolism , Paraproteinemias/complications , Paraproteinemias/genetics , Paraproteinemias/metabolism , Protein Processing, Post-Translational , Transforming Growth Factor beta/metabolism
15.
Blood ; 135(14): 1101-1110, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32027747

ABSTRACT

Scleromyxedema is a rare skin and systemic mucinosis that is usually associated with monoclonal gammopathy (MG). In this French multicenter retrospective study of 33 patients, we investigated the clinical and therapeutic features of MG-associated scleromyxedema. Skin molecular signatures were analyzed using a transcriptomic approach. Skin symptoms included papular eruptions (100%), sclerodermoid features (91%), and leonine facies (39%). MG involved an immunoglobulin G isotype in all patients, with a predominant λ light chain (73%). Associated hematologic malignancies were diagnosed in 4 of 33 patients (12%) (smoldering myeloma, n = 2; chronic lymphoid leukemia, n = 1; and refractory cytopenia with multilineage dysplasia, n = 1). Carpal tunnel syndrome (33%), arthralgia (25%), and dermato-neuro syndrome (DNS) (18%) were the most common systemic complications. One patient with mucinous cardiopathy died of acute heart failure. High-dose IV immunoglobulin (HDIVig), alone or in combination with steroids, appeared to be quite effective in nonsevere cases (clinical complete response achieved in 13/31 patients). Plasma cell-directed therapies using lenalidomide and/or bortezomib with dexamethasone and HDIVig led to a significant improvement in severe cases (HDIVig refractory or cases with central nervous system or cardiac involvement). The emergency treatment of DNS with combined plasmapheresis, HDIVig, and high-dose corticosteroids induced the complete remission of neurological symptoms in 4 of 5 patients. Quantitative reverse-transcriptase polymerase chain reaction analysis of 6 scleromyxedema skin samples showed significantly higher profibrotic pathway levels (transforming growth factor ß and collagen-1) than in healthy skin. Prospective studies targeting plasma cell clones and/or fibrotic pathways are warranted for long-term scleromyxedema management.


Subject(s)
Paraproteinemias/complications , Paraproteinemias/therapy , Plasma Cells/pathology , Scleromyxedema/complications , Scleromyxedema/therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Dexamethasone/therapeutic use , Female , Glucocorticoids/therapeutic use , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Lenalidomide/therapeutic use , Male , Middle Aged , Paraproteinemias/genetics , Paraproteinemias/pathology , Plasma Cells/drug effects , Plasma Cells/metabolism , Plasmapheresis , Retrospective Studies , Scleromyxedema/genetics , Scleromyxedema/pathology , Skin/metabolism , Skin/pathology , Transcriptome
16.
Front Immunol ; 11: 607211, 2020.
Article in English | MEDLINE | ID: mdl-33384694

ABSTRACT

Factor H (FH), a member of the regulators-of-complement-activation (RCA) family of proteins, circulates in human plasma at concentrations of 180-420 mg/L where it controls the alternative pathway (AP) of complement in the fluid phase and on cell surfaces. When the regulatory function of FH is impaired, complement-mediated tissue injury and inflammation occur, leading to diseases such as atypical hemolytic uremic syndrome (a thrombotic microangiopathy or TMA), C3 glomerulopathy (C3G) and monoclonal gammopathy of renal significance (MGRS). A pathophysiological cause of compromised FH function is the development of autoantibodies to various domains of the FH protein. FH autoantibodies (FHAAs) are identified in 10.9% of patients with aHUS, 3.2% of patients with C3G, and rarely in patients with MGRS. The phenotypic variability of FHAA-mediated disease reflects both the complexity of FH and the epitope specificity of FHAA for select regions of the native protein. In this paper, we have characterized FHAA epitopes in a large cohort of patients diagnosed with TMA, C3G or MGRS. We explore the epitopes recognized by FHAAs in these diseases and the association of FHAAs with the genetic deletion of both copies of the CFHR1 gene to show how these disease phenotypes are associated with this diverse spectrum of autoantibodies.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/blood , Glomerulonephritis/immunology , Paraproteinemias/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/epidemiology , Atypical Hemolytic Uremic Syndrome/genetics , Biomarkers/blood , Child , Child, Preschool , Complement C3b Inactivator Proteins/genetics , Complement Factor H/immunology , Epitopes , Female , Gene Deletion , Genetic Predisposition to Disease , Glomerulonephritis/blood , Glomerulonephritis/epidemiology , Glomerulonephritis/genetics , Humans , Infant , Male , Middle Aged , Paraproteinemias/blood , Paraproteinemias/epidemiology , Paraproteinemias/genetics , Phenotype , Prevalence , Retrospective Studies , United States/epidemiology , Young Adult
17.
Rev. cuba. hematol. inmunol. hemoter ; 35(3): e1067, jul.-set. 2019.
Article in Spanish | CUMED, LILACS | ID: biblio-1093277

ABSTRACT

Introducción: El mieloma múltiple (MM) es una enfermedad que va precedida por una fase previa conocida como gammapatía monoclonal de significado incierto (GMSI); en esta última existen varias anormalidades citogenéticas, que permiten la progresión a MM, entre estas encontramos reordenamientos primarios del gen de la cadena pesada de la inmunoglobina (IGH), además de células hiperdiploides. Desarrollo: Las alteraciones cromosómicas en el MM se pueden clasificar en dos grupos principales: las que involucran las translocaciones del locus IGH ubicado en el cromosoma 14q32 y cuyos principales reordenamientos se dan entre las regiones cromosómicas 11q13, 16q23, 4p16.3, 6p21 y, un segundo grupo caracterizado por los desequilibrios genómicos. Los pacientes con translocaciones de la IGH, muestran un pronóstico diferente en dependencia del tipo de reordenamiento cromosómico. La t(4;14)(p16;q32) y t(14;16)(q32;q23) se asocian a un mal pronóstico, mientras que los pacientes con t(11;14) (q13;q32) tiene un buen pronóstico de la enfermedad en ausencia de otras anormalidades genéticas. En el grupo con desequilibrio genómico se encuentran deleciones, amplificaciones, y células con números anormales de cromosomas (hiperdiploidas y no hiperdiploides); casi siempre asociadas a mal pronóstico ya que muchas de estas alteraciones involucran perdida de material genómico relacionado con el control de ciclo celular y progresión de la enfermedad, como son las deleciones de los cromosomas 1,13 y 17. Los pacientes con trisomías de los cromosomas impares 1, 3, 5, 7, 9, 11, 15, 19,21 suelen tener un mejor pronóstico y una tasa mayor de sobrevivencia(AU)


Introduction: Multiple myeloma (MM) is a disease that is preceded by a previous phase known as monoclonal gammopathy of uncertain significance (MGUS); in this latter there are several cytogenetic abnormalities, which allow the progression to MM, among these we find primary rearrangements of the heavy chain gene of the immunoglobin (IGH), in addition to hyperdiploid cells. Development: Chromosomal alterations in MM can be classified into two main groups, those involving the translocations of the IGH locus located on chromosome 14q32 and whose main rearrangements occur between the chromosomal regions 11q13, 16q23, 4p16.3, 6p21, and a second group which is characterized by genomic imbalances. Patients with translocations of the IGH, show a different prognosis depending on the type of chromosomal rearrangement, the t(4; 14)(p16; q32) and t(14; 16)(q32; q23) are associated with a poor prognosis while patients with t(11; 14)(q13; q32) have a good prognosis of the disease in the absence of other genetic abnormalities. Within the genomic imbalances we find deletions, amplifications, and cells with abnormal numbers of chromosomes (hyperdiploids and not hyperdiploid), these almost always associated with poor prognosis since many of these alterations involve loss of genomic material related to cell cycle control and progression of the disease, such as deletions of chromosomes 1,13 and 17. Patients with trisomies of odd chromosomes 1, 3, 5, 7, 9, 11, 15, 19,21 usually have a better prognosis and a higher survival rate(AU)


Subject(s)
Humans , Male , Female , Prognosis , Multiple Myeloma/genetics , Multiple Myeloma/epidemiology , Paraproteinemias/genetics , Chromosome Deletion , Disease Progression , Cytogenetic Analysis/methods , Bortezomib/therapeutic use
18.
Blood Cancer J ; 9(9): 72, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31462637

ABSTRACT

Dysproteinemias progress through a series of clonal evolution events in the tumor cell along with the development of a progressively more "permissive" immune tumor microenvironment (iTME). Novel multiparametric cytometry approaches, such as cytometry by time-of-flight (CyTOF) combined with novel gating algorithms can rapidly characterize previously unknown phenotypes in the iTME of tumors and better capture its heterogeneity. Here, we used a 33-marker CyTOF panel to characterize the iTME of dysproteinemia patients (MGUS, multiple myeloma-MM, smoldering MM, and AL amyloidosis) at diagnosis and after standard of care first line therapies (triplet induction chemotherapy and autologous stem cell transplant-ASCT). We identify novel subsets, some of which are unique to the iTME and absent from matched peripheral blood samples, with potential roles in tumor immunosurveillance as well as tumor immune escape. We find that AL amyloidosis has a distinct iTME compared to other dysproteinemias with higher myeloid and "innate-like" T cell subset infiltration. We show that T cell immune senescence might be implicated in disease pathogenesis in patients with trisomies. Finally, we demonstrate that the early post-ASCT period is associated with an increase of senescent and exhausted subsets, which might have implications for the rational selection of post-ASCT therapies.


Subject(s)
Multiple Myeloma/genetics , Paraproteinemias/genetics , Adult , Female , Humans , Male , Multiple Myeloma/pathology , Paraproteinemias/pathology , T-Lymphocyte Subsets/pathology , Tumor Microenvironment , Young Adult
19.
BMC Cancer ; 19(1): 514, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142285

ABSTRACT

BACKGROUND: Crystal-storing histiocytosis (CSH) is a rare lesion characterized by sheets of crystal-laden non-neoplastic histiocytes. CSH shows a prominent association with lymphoproliferative disorders that express monoclonal immunoglobulins, mainly multiple myeloma (MM), lymphoplasmacytic lymphoma (LPL) and monoclonal gammopathy of undetermined significance (MGUS). However, no aggressive B cell lymphoma has been reported to be associated with CSH. CASE PRESENTATION: A 74-year-old Chinese woman presented with multiple subcutaneous masses, abdominal pain, and fever. An IgM kappa type of monoclonal gammopathy (MG) was noted by immunofixation performed on the patient's serum. Computed tomographic (CT) scan revealed subcutaneous masses on the left upper arm and at the waist and multiple low-density lesions in the spleen. Microscopically, sections of subcutaneous masses revealed sheets of large polygonal and spindle cells with abundant eosinophilic cytoplasm, round to ovoid eccentric nuclei, reticulate chromatin, and median nucleoli. Massive needle-shaped crystals were confined to the cytoplasm. Immunohistochemically, these crystal-containing cells were positive for CD68/PGM1, CD163, IgM, and Igκ. Meanwhile, the splenic tumour was diagnosed as diffuse large B-cell lymphoma (DLBCL), non-germinal-centre B (non-GCB) subtype (Hans algorithm). Immunohistochemistry for IgM was positive in the cytoplasm of some neoplastic cells. Immunoglobulin heavy chain (IgH) gene rearrangement was detected by PCR analysis of the subcutaneous mass and the splenic tumour. CONCLUSION: To the best of our knowledge, this is the first case of generalized CSH with DLBCL and MG. Although the rarity of CSH and separate locations of CSH and lymphoma led to a diagnostic dilemma, the presence of MG was a clue to appreciate the relation between CSH and DLBCL. This case stressed a full investigation into the underlying lymphoproliferative disorder for integrated diagnosis and correct treatments.


Subject(s)
Histiocytosis, Langerhans-Cell/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Paraproteinemias/diagnostic imaging , Aged , Female , Gene Rearrangement , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Paraproteinemias/genetics , Paraproteinemias/metabolism , Tomography, X-Ray Computed
20.
Blood ; 133(26): 2741-2744, 2019 06 27.
Article in English | MEDLINE | ID: mdl-30967366

ABSTRACT

We previously reported a new form of light chain deposition disease (LCDD) presenting as diffuse cystic lung disorder that differs from the usual systemic form with respect to patient age, the male/female ratio, the involved organs, and the hematologic characteristics. We also demonstrated that the light chains were produced by an intrapulmonary B-cell clone and that this clone shared a stereotyped antigen receptor IGHV4-34/IGKV1. However, we only analyzed 3 patients. We conducted a retrospective study including lung tissue samples from 24 patients with pulmonary LCDD (pLCDD) matched with samples from 13 patients with pulmonary κ light chain amyloidosis (pAL amyloidosis) used as controls. Mass spectrometry-based proteomics identified immunoglobulin κ peptides as the main protein component of the tissue deposits in all patients. Interestingly, in pLCDD, IGKV1 was the most common κ family detected (86.4%), and IGKV1-8 was overrepresented compared with pAL amyloidosis (75% vs 11.1%, P = .0033). Furthermore, IGKV1-8 was predominantly associated with a diffuse cystic pattern (94%) in pLCDD. In conclusion, the high frequency of IGKV1-8 usage in cystic pLCDD constitutes an additional feature arguing for a specific entity distinct from the systemic form that preferentially uses IGKV4-1.


Subject(s)
Immunoglobulin Light Chains/genetics , Lung Diseases/genetics , Paraproteinemias/genetics , Adult , Female , Humans , Lung Diseases/pathology , Male , Middle Aged , Paraproteinemias/pathology , Proteomics , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...