Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Clin Lab ; 69(7)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37436385

ABSTRACT

BACKGROUND: The aim of the study was to investigate the clinical and laboratory characteristics of IgM primary plasma cell leukemia. METHODS: We retrospectively analyzed a case of clinical and laboratory characteristics of IgM primary plasma cell leukemia and review related literature of patient with primary plasma cell leukemia. RESULTS: Laboratory tests: Alanine aminotransferase 128 U/L, Aspartate aminotransferase 245 U/L, Globulin 47.8 g/L, Lactate dehydrogenase 1,114 U/L, Creatinine 111.7 mol/L, Serum calcium 2.47 mmol/L, ß2 microglobulin 8.52 µg/mL, Immunoglobulin G 31.41 g/L, D-dimer 2.34 mg/L, Prothrombin time 13.6 seconds Fibrinogen 2 g/L, White blood cell 7.38 x 109/L, Red blood cell 3.46 x 1012/L, Hemoglobin 115 g/L, Platelet 7 x 109/L, and 12% Primitive naive cells can be seen in peripheral blood smear. Bone marrow smear: Accounted for 52% of original cells, the cell size, shape is irregular, the edge is not neat, the cell quality is rich, stained gray blue, cytoplasmic staining uneven, some devouring blood cells can be seen in the cytoplasm or unknown substance, the nucleus shape is irregular, visible distortion and fold, is visible on the part of the nuclei cavitation sample inclusions, chromatin is meticulous, partly visible large nucleoli. Flow cytometry results showed abnormal cell group held 23.85% of nuclear cells, expression of CD38, CD138, CD117, cKappa, partly CD20, weak expressing CD45, not express CD27, CD19, CD56, CD200, CD81, cLambda. It was a monoclonal plasma cell with an abnormal phenotype, consistent with a plasma cell tumor. Immunofixation electrophoresis results showed that the serum M protein was 22.80 g/L, which was IgG-κ type, the serum free KAP light chain was 232.69 mg/L, the serum free LAM light chain was 5.37 mg/L, and the rFLC (κ-FLC: λFLC) was 43.33. The diagnosis was primary plasmacytic leukemia of light chain type. CONCLUSIONS: Primary plasma cell leukemia (pPCL) is a rare and highly aggressive plasma cell malignancy. Laboratory staff should pay more attention to and recognize the pleomorphic morphology of neoplastic plasma cells, which can enable timely clinical development of bone marrow smear, biopsy, flow cytometry, and cytogenetic tests providing help in early diagnosis and treatment.


Subject(s)
Leukemia, Plasma Cell , Neoplasms, Plasma Cell , Humans , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/pathology , Retrospective Studies , Antigens, CD19 , Immunoglobulin M
2.
Mol Ther ; 29(2): 645-657, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33278564

ABSTRACT

Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized the clinical treatment of hematological malignancies due to the prominent anti-tumor effects. B cell maturation antigen (BCMA) CAR-T cells have demonstrated promising effects in patients with relapsed/refractory multiple myeloma. However, the dynamics of CAR-T cell proliferation and cytotoxicity in clinical patients remains unexplored. Here, we longitudinally profiled the transcriptomes of 55,488 T cells including CAR-T products, CAR-T cells, and endogenous T cells at the peak and remission phases in a plasma cell leukemia (PCL) patient treated with BCMA CAR-T cells by single-cell transcriptomic analysis. Our results showed distinct CAR-T and endogenous T cell subsets indicating stage-specific expression in proliferation, cytotoxicity, and intercellular signaling pathways. Furthermore, we found that CAR-T cells at peak phase gradually convert to a highly cytotoxic state from a highly proliferative state along a development trajectory. Moreover, re-analysis of a single cell study from CD8+ CD19 CAR-T confirmed our findings. These commonalities suggest conserved mechanisms for CAR-T treatment across hematological malignancies. Taken together, our current study provides insight into CAR-T cell dynamics during CAR-T therapy and proves that both BCMA CAR-T and CD19 CAR-T have similar transcriptional characteristics, especially at the CAR-T peak phase.


Subject(s)
B-Cell Maturation Antigen/immunology , Immunotherapy, Adoptive , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/therapy , Transcriptome , Antigens, CD19/immunology , Drug Resistance, Neoplasm , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy, Adoptive/methods , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recurrence , Single-Cell Analysis/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
3.
Br J Cancer ; 124(11): 1843-1853, 2021 05.
Article in English | MEDLINE | ID: mdl-33785876

ABSTRACT

BACKGROUND: Secondary plasma cell leukaemia (sPCL) is an aggressive form of multiple myeloma (MM), but the mechanism underlying MM progresses into PCL remains unknown. METHODS: Gene expression profiling of MM patients and PCL patients was analysed to identify the molecular differences between the two diseases. Cox survival regression and Kaplan-Meier analysis were performed to illustrate the impact of integrin subunit alpha 6 (ITGA6) on prognosis of MM. Invasion assays were performed to assess whether ITGA6 regulated the progression of MM to PCL. RESULTS: Gene expression profiling analyses showed that cell metastasis pathways were enriched in PCL and ITGA6 was differentially expressed between PCL and MM. ITGA6 expression was an independent prognostic factor for event-free survival (EFS) and overall survival (OS) of MM patients. Moreover, the stratification ability of the International Staging System (ISS) of MM was improved when including ITGA6 expression. Functional studies uncovered that increased ITGA6 reduced the myeloma cell invasion. Additionally, low expression of ITGA6 resulted from epigenetic downregulating of its anti-sense non-coding RNA, ITGA6-AS1. CONCLUSION: Our data reveal that ITGA6 gradually decreases during plasma cell dyscrasias progression and low expression of ITGA6 contributes to myeloma metastasis. Moreover, ITGA6 abundance might help develop MM prognostic stratification.


Subject(s)
Integrin alpha6/genetics , Leukemia, Plasma Cell/genetics , Multiple Myeloma/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Down-Regulation/genetics , Down-Regulation/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha6/physiology , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/mortality , Leukemia, Plasma Cell/pathology , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis
4.
Ann Hematol ; 100(2): 437-443, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33392702

ABSTRACT

We analyzed variations in terms of chromosomal abnormalities (CA) by fluorescence in situ hybridization (FISH) analysis on purified bone marrow plasma cells throughout the progression from monoclonal gammopathy of undetermined significance/smoldering multiple myeloma (MGUS/SMM) to newly diagnosed MM/plasma cell leukemia (NDMM/PCL) at diagnosis and from diagnostic samples to progressive disease. High risk was defined by the presence of at least del(17p), t(4;14), and/or t(14;16). 1p/1q detection (in the standard FISH panel from 2012 onward) was not available for all patients. We analyzed 139 MM/PCL diagnostic samples from 144 patients, with a median follow-up of 71 months: high-risk CA at diagnosis (MGUS/SMM or NDMM) was present in 28% of samples, whereas 37-39% showed high-risk CA at relapse. In 115 patients with NDMM who evolved to relapsed/refractory MM, we identified 3 different populations: (1) 31/115 patients (27%) with gain of new CA (del13, del17p, t(4;14), t(14;16) or 1q CA when available); (2) 10/115 (9%) patients with loss of a previously identified CA; and (3) 74 patients with no changes. The CA gain group showed a median overall survival of 66 months vs. 84 months in the third group (HR 0.56, 95% CI 0.34-0.92, p = 0.023). Clonal evolution occurs as disease progresses after different chemotherapy lines. Patients who acquired high-risk CA had the poorest prognosis. Our findings highlight the importance of performing FISH analysis both at diagnosis and at relapse.


Subject(s)
Chromosome Aberrations , Chromosomes, Human/genetics , Clonal Evolution , Leukemia, Plasma Cell , Monoclonal Gammopathy of Undetermined Significance , Smoldering Multiple Myeloma , Aged , Disease-Free Survival , Female , Follow-Up Studies , Humans , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/mortality , Longitudinal Studies , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/mortality , Retrospective Studies , Risk Factors , Smoldering Multiple Myeloma/genetics , Smoldering Multiple Myeloma/mortality , Survival Rate
5.
Rev Med Chil ; 147(1): 18-23, 2019.
Article in Spanish | MEDLINE | ID: mdl-30848760

ABSTRACT

BACKGROUND: Primary plasma cell leukemia (pPCL) is uncommon, aggressive and has a different biology than multiple myeloma (MM). AIM: To report the features of patients with pPCL. MATERIAL AND METHODS: Review of databases of the Hematology Department and the Hematology laboratory. RESULTS: Of 178 patients with monoclonal gammopathies, five (2.8%) patients aged 33 to 64 years (three females) had a pPCL. The mean hemoglobin was 7.3 g/dL, the mean white blood cell count was 52,500/mm3, with 58% plasma cells, and the mean platelet count was 83,600/mm3. The mean bone marrow infiltration was 89%, LDH was 2,003 IU/L, serum calcium was 13 mg/dL, and creatinine 1.5 mg/dL. Two patients had bone lesions. Three were IgG, one IgA lambda and one lambda light chain. CD20 was positive in one, CD56 was negative in all and CD117 was negative in 3 cases. By conventional cytogenetic analysis, two had a complex karyotype. By Fluorescence in situ Hybridization, one was positive for TP53 and another for t (11; 14). One patient did not receive any treatment, three patients received VTD PACE and one CTD. None underwent transplant. Three patients are alive. The mean survival was 14 months. CONCLUSIONS: These patients with pPCL were younger and had a more aggressive clinical outcome than in multiple myeloma.


Subject(s)
Leukemia, Plasma Cell/epidemiology , Leukemia, Plasma Cell/genetics , Adult , Blood Cell Count , Calcium/blood , Chile/epidemiology , Creatinine/blood , Cytogenetic Analysis , Female , Flow Cytometry/methods , Humans , In Situ Hybridization, Fluorescence , Leukemia, Plasma Cell/pathology , Leukemia, Plasma Cell/therapy , Male , Middle Aged , Paraproteinemias/epidemiology , Paraproteinemias/genetics , Paraproteinemias/pathology , Retrospective Studies , Survival Analysis , Treatment Outcome
7.
Eur J Haematol ; 100(2): 215-217, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29064593

ABSTRACT

Primary plasma cell leukemia (pPCL) is the most aggressive form of the plasma cell (PC) malignancy, multiple myeloma (MM). It has been commonly associated with the presence of a chromosome translocation involving the immunoglobulin heavy chain (IgH) locus on 14q32, that is t (11;14). Results from early phase clinical trials utilizing the selective Bcl-2 inhibitor, venetoclax, as a single agent in patients with relapsed MM have had remarkable efficacy among patients with t (11;14) abnormality. The present case demonstrates the ability of a combination regimen incorporating Bcl-2 inhibition with daratumumab, bortezomib, venetoclax, and dexamethasone to induce a rapid and very deep hematologic response in a pPCL patient with t (11;14), even in a setting of very refractory disease. This case highlights the need to further study Bcl-2 inhibition-based therapy as an option for therapy in patients with pPCL with t (11;14).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/genetics , Translocation, Genetic , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Bone Marrow/pathology , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Immunoglobulin kappa-Chains/blood , Leukemia, Plasma Cell/diagnosis , Middle Aged , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Retreatment , Sulfonamides/administration & dosage , Treatment Outcome
9.
Eur J Haematol ; 95(1): 16-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25778450

ABSTRACT

Plasma cell leukemia (PCL) is a very aggressive and rare form of malignant monoclonal gammopathy characterized by the presence of plasmocytes in peripheral blood. It is classified as primary PCL occuring 'de novo', or as secondary PCL in patients with relapsed/refractory multiple myeloma. Primary PCL is a distinct clinicopathological entity from myeloma with different cytogenetic abnormalities and molecular findings, which are usually found only in advanced multiple myeloma. The clinical course is aggressive with short remissions and reduced overall survival. The diagnostic criteria are based on the percentage (>20%) and absolute number (2 × 10(9) /L) of plasma cells in peripheral blood. After establishing diagnosis, induction therapy should begin promptly which is aimed to rapid disease control and to minimize the risk of early death. Intensive chemotherapy regimens and bortezomib-based regimens, followed by high-dose therapy with autologous stem cell transplantation, are recommended. Allogeneic transplantation can be considered in younger patients. This article reviews recent knowledge of this hematological malignancy that is associated with a very poor prognosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/therapeutic use , Chromosome Aberrations , Leukemia, Plasma Cell/therapy , Multiple Myeloma/therapy , Stem Cell Transplantation , Antigens, CD/genetics , Antigens, CD/metabolism , Disease Progression , Gene Expression , Humans , Induction Chemotherapy/methods , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/mortality , Leukemia, Plasma Cell/pathology , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Plasma Cells/pathology , Prognosis , Survival Analysis , Transplantation, Autologous , Transplantation, Homologous
10.
Int J Mol Sci ; 16(8): 17514-34, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26263974

ABSTRACT

Primary plasma cell leukemia (pPCL) is a rare and aggressive variant of multiple myeloma (MM) which may represent a valid model for high-risk MM. This disease is associated with a very poor prognosis, and unfortunately, it has not significantly improved during the last three decades. New high-throughput technologies have allowed a better understanding of the molecular basis of this disease and moved toward risk stratification, providing insights for targeted therapy studies. This knowledge, added to the pharmacogenetic profile of new and old agents in the analysis of efficacy and safety, could contribute to help clinical decisions move toward a precision medicine and a better clinical outcome for these patients. In this review, we describe the available literature concerning the genomic characterization and pharmacogenetics of plasma cell leukemia (PCL).


Subject(s)
Leukemia, Plasma Cell/drug therapy , Leukemia, Plasma Cell/genetics , Neoplasm Proteins/genetics , Pharmacogenetics , Antineoplastic Agents/therapeutic use , Humans , Leukemia, Plasma Cell/classification , Molecular Targeted Therapy , Precision Medicine , Prognosis , Treatment Outcome
11.
Blood ; 120(5): 927-8, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22859708

ABSTRACT

In this issue of Blood, Keats et al,(1) Egan et al,(2) and Walker et al(3) provide a genome-wide snapshot of the clonal landscape in multiple myeloma(MM)illustrating the complexity of the evolutionary process and the dynamics of clonal evolution over time.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 4 , Clonal Evolution/genetics , DNA Copy Number Variations , Genes, Dominant/physiology , Genetic Heterogeneity , Leukemia, Plasma Cell/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Sequence Analysis, DNA , Translocation, Genetic/genetics , Animals , Female , Humans , Male
12.
Blood ; 120(5): 1060-6, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22529291

ABSTRACT

The longitudinal evolution of a myeloma genome from diagnosis to plasma cell leukemia has not previously been reported. We used whole-genome sequencing (WGS) on 4 purified tumor samples and patient germline DNA drawn over a 5-year period in a t(4;14) multiple myeloma patient. Tumor samples were acquired at diagnosis, first relapse, second relapse, and end-stage secondary plasma cell leukemia (sPCL). In addition to the t(4;14), all tumor time points also shared 10 common single-nucleotide variants (SNVs) on WGS comprising shared initiating events. Interestingly, we observed genomic sequence variants that waxed and waned with time in progressive tumors, suggesting the presence of multiple independent, yet related, clones at diagnosis that rose and fell in dominance. Five newly acquired SNVs, including truncating mutations of RB1 and ZKSCAN3, were observed only in the final sPCL sample suggesting leukemic transformation events. This longitudinal WGS characterization of the natural history of a high-risk myeloma patient demonstrated tumor heterogeneity at diagnosis with shifting dominance of tumor clones over time and has also identified potential mutations contributing to myelomagenesis as well as transformation from myeloma to overt extramedullary disease such as sPCL.


Subject(s)
Cell Transformation, Neoplastic/genetics , Clonal Evolution/genetics , Leukemia, Plasma Cell/genetics , Multiple Myeloma/genetics , Sequence Analysis, DNA , Aged , Chromosome Aberrations , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 4 , Clonal Evolution/physiology , Comparative Genomic Hybridization , Disease Progression , Female , Follow-Up Studies , Genome, Human/genetics , Humans , Leukemia, Plasma Cell/diagnosis , Multiple Myeloma/diagnosis , Recurrence , Sequence Analysis, DNA/methods
13.
Am J Hematol ; 94(1): E35-E37, 2019 01.
Article in English | MEDLINE | ID: mdl-30370955
14.
Genomics ; 102(4): 243-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23831116

ABSTRACT

The objective of this study was to describe co-expression correlations of cell cycle regulatory genes in multiple myeloma (MM) and plasma cell leukemia (PCL). Our results highlight the presence of dynamic equilibrium between co-expression of activator and inhibitor gene sets. Moreover inhibitor set is more sensitive to the activator changes, not vice versa. We have shown that CDKN2A expression is associated with short-term survival in newly diagnosed MM patients (survival was 30.3 ± 3.9 months for 'low' expressed and 7.5 ± 5.6 months for 'high' expressed group, p<0.0001). Moreover low-expression CDKN2A group showed time-to-progression benefit in newly diagnosed patients (remission was 20.8 ± 3.6 months for 'low' and 8.4 ± 2.7 months for 'high' expressed group, p<0.0001) as well as in whole studied cohort of MM patients (remission was 20.8 ± 2.8 months for 'low' and 9.8 ± 1.1 months for 'high' expressed group, p<0.0001). The overexpression of inhibitors can be explained as a compensatory reaction to growing "oncogenic stress".


Subject(s)
Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, cdc , Leukemia, Plasma Cell/genetics , Multiple Myeloma/genetics , Aged , Aged, 80 and over , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Leukemia, Plasma Cell/diagnosis , Male , Middle Aged , Multiple Myeloma/diagnosis , Prognosis , Survival Analysis , Time Factors , Tumor Cells, Cultured
15.
J Hematop ; 17(2): 117-119, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38580782

ABSTRACT

A 71-year-old female with relapsed IgA lambda myeloma developed progressive cytopenia. The peripheral blood film showed 5% blastoid cells. Flow cytometry analysis was indicative of plasma cells. The bone marrow smear was packed with plasmablasts. Target CD138-cell FISH and molecular karyotyping identified a complex genome. NGS identified high-risk mutations. Bone marrow histology confirmed myeloma with no evidence of acute leukaemia. The patient was diagnosed with plasmablastic progression of myeloma and secondary PCL. Secondary PCL patients have a poor prognosis. It is essential to recognize this subtype and explore a novel treatment approach.


Subject(s)
Leukemia, Plasma Cell , Plasma Cells , Humans , Female , Aged , Leukemia, Plasma Cell/pathology , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/diagnosis , Plasma Cells/pathology , Multiple Myeloma/pathology , Multiple Myeloma/diagnosis , Mutation
18.
Haematologica ; 98(10): 1586-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23716545

ABSTRACT

Multiple myeloma can be categorized into hyperdiploid or non-hyperdiploid myeloma based on the number of chromosomes found in the tumor clone. Among the non-hyperdiploid myelomas, the hypodiploid subtype has the most aggressive clinical phenotype, but the genetic differences between groups are not completely defined. In order to understand the genetic background of hypodiploid multiple myeloma better, we compared the genomic (array-based comparative genomic hybridization) and transcriptomic (gene expression profiling) background of 49 patients with hypodiploid myeloma with 50 other non-hyperdiploid and 125 hyperdiploid myeloma patients. There were significant chromosomal and gene expression differences between hyperdiploid patients and non-hyperdiploid and hypodiploid patients. Non-hyperdiploid and hypodiploid patients shared most of the chromosomal abnormalities; nevertheless a subset of these abnormalities, such as monosomies 13, 14 and 22, was markedly increased in hypodiploid patients. Furthermore, deletions of 1p, 12p, 16q and 17p, all associated with poor outcome or progression in multiple myeloma, were significantly enriched in hypodiploid patients. Molecular risk-stratification indices reinforce the worse prognosis associated with hypodiploid multiple myeloma compared with non-hyperdiploid multiple myeloma. Gene expression profiling clustered hypodiploid and non-hyperdiploid subgroups closer than hyperdiploid myeloma but also highlighted the up-regulation of CCND2, WHSC1/MMSET and FGFR3 in the hypodiploid subtype. In summary, hypodiploid multiple myeloma is genetically similar to non-hyperdiploid multiple myeloma but characterized by a higher prevalence of genetic alterations associated with poor outcome and disease progression. It is provocative to hypothesize that hypodiploid multiple myeloma is an advanced stage of non-hyperdiploid multiple myeloma.


Subject(s)
Biomarkers, Tumor/genetics , Diploidy , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/genetics , Cohort Studies , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics
19.
Am J Hematol ; 88(1): 16-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23044976

ABSTRACT

Primary plasma cell leukemia (pPCL) is a rare, yet aggressive form of de novo plasma cell tumor, distinct from secondary PCL (sPCL) which represents a leukemic transformation of pre-existing multiple myeloma (MM). Herein, we performed a comprehensive molecular analysis of a prospective series of pPCLs by means of FISH, single nucleotide polymorphism (SNP) array and gene expression profiling (GEP). IGH@ translocations were identified in 87% of pPCL cases, with prevalence of t(11;14) (40%) and t(14;16) (30.5%), whereas the most frequent numerical alterations involved 1p (38%), 1q (48%), 6q (29%), 8p (42%), 13q (74%), 14q (71%), 16q (53%), and 17p (35%). We identified a minimal biallelic deletion (1.5 Mb) in 8p21.2 encompassing the PPP2R2A gene, belonging to a family of putative tumor suppressors and found to be significantly down-regulated in deleted cases. Mutations of TP53 were identified in four cases, all but one associated with a monoallelic deletion of the gene, whereas activating mutations of the BRAF oncogene occurred in one case and were absent in N- and K-RAS. To evaluate the influence of allelic imbalances in transcriptional expression we performed an integrated genomic analysis with GEP data, showing a significant dosage effect of genes involved in transcription, translation, methyltransferase activity, apoptosis as well as Wnt and NF-kB signaling pathways. Overall, we provide a compendium of genomic alterations in a prospective series of pPCLs which may contribute to improve our understanding of the pathogenesis of this aggressive form of plasma cell dyscrasia and the mechanisms of tumor progression in MM.


Subject(s)
Allelic Imbalance , Gene Expression Regulation, Leukemic , Leukemia, Plasma Cell/genetics , Leukemia, Plasma Cell/metabolism , Neoplasm Proteins/biosynthesis , Polymorphism, Single Nucleotide , Transcription, Genetic , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Female , Follow-Up Studies , Gene Dosage , Gene Expression Profiling , Genome-Wide Association Study , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Wnt Signaling Pathway/genetics
20.
Hematology ; 28(1): 2254556, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37732631

ABSTRACT

BACKGROUND: Plasma cell leukemia (PCL) is a rare and aggressive plasma cell disorder, exhibiting a more unfavorable prognosis than multiple myeloma. PCL is classified into pPCL and sPCL. Recently, the IMWG has recommended new PCL definition criteria, which require the presence of ≥5% circulating plasma cells in peripheral blood smears. Due to its low incidence, research on pPCL and sPCL is limited. METHODS: We conducted a retrospective study and analyzed clinical and cytogenetic data of pPCL and sPCL patients. Overall survival (OS) and progression-free survival (PFS) were assessed by the Kaplan-Meier method, and survival distributions were compared using the log-rank test. RESULTS: This is a small cohort comprising 23 pPCL and 9 sPCL patients. Notably, sPCL patients showed a higher incidence of extramedullary infiltration and a higher percentage of bone marrow plasma cells (p = 0.015 and 0.025, respectively). Although no significant difference was found between the two groups in OS and PFS, a trend emerged suggesting a superior survival outcome for pPCL patients, with a higher cumulative 1-year PFS rate (38.3% vs. 13.3%) and a lower early mortality rate (mortality rate at 3 months: 15% vs. 33%). We also suggested that pPCL patients carrying t(11;14) may have a longer median survival time than individuals with other cytogenetic abnormalities, but this was not confirmed due to the small sample size. CONCLUSION: Our study revealed clinical and cytogenetic features of pPCL and sPCL patients according to the new diagnostic criteria. The findings suggested a generally better prognosis for pPCL than sPCL and the likelihood of t(11;14) translocation acting as a favorable prognostic factor in pPCL. It is important to note that our study had a limited sample size, which may lead to bias. We hope well-designed studies can be conducted to provide more results.


Subject(s)
Leukemia, Plasma Cell , Multiple Myeloma , Humans , Leukemia, Plasma Cell/diagnosis , Leukemia, Plasma Cell/genetics , Retrospective Studies , Cytogenetics , Cytogenetic Analysis
SELECTION OF CITATIONS
SEARCH DETAIL