Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.881
Filter
1.
Clin Exp Med ; 24(1): 118, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833040

ABSTRACT

Vitamin D acts through the vitamin D receptor (VDR), and vitamin D level decreases in multiple myeloma (MM) patients. Single nucleotide polymorphisms in VDR alter its functions to affect the vitamin D status. This raises the question of whether VDR gene polymorphisms are associated with MM risk, which has been investigated in case‒control studies, but the results have been inconsistent. This meta-analysis aimed to investigate the relationship between VDR gene polymorphisms and MM risk. The PubMed, Web of Science, Medline, Embase, Chinese National Knowledge Infrastructure (CNKI), Chinese Scientific Journal (VIP), Wanfang Databases (WANFANG) were searched from inception to June 1, 2023, without language restriction or publication preference. Pooled odds ratio (OR) and 95% confidence interval (CI) for each variable were calculated. Leave-one-out sensitivity analysis was performed to determine the source of heterogeneity. Publication bias was assessed using Begg' and Egger's tests, and the trim-and-fill method was used to compensate for publication bias. The correlation meta-analysis was conducted using Comprehensive Meta-Analysis 3.0 and STATA 12.0 software. All the included studies were based on Asian populations and involved four VDR gene polymorphisms, TaqI (rs731236), ApaI (rs7975232), BsmI (rs1544410) and FokI (rs2228570). The results showed that TaqI (C vs. T: OR = 1.487, 95% CI 1.052, 2.104, P = 0.025; CC + CT vs. TT: OR = 1.830, 95% CI 1.138, 2.944, P = 0.013), ApaI (T vs. G: OR = 1.292, 95% CI 1.101, 1.517, P = 0.002; TT vs. GG: OR = 1.600, 95% CI 1.106, 2.314, P = 0.013; TG vs. GG: OR 1.305, 95% CI 1.050, 1.622; P = 0.016; TT + TG vs. GG: OR = 1.353, 95% CI 1.103, 1.662, P = 0.004), BsmI (GG vs. AA: OR = 1.918, 95% CI 1.293, 2.844, P = 0.001; GA vs. AA: OR = 1.333, 95% CI 1.058, 1.679, P = 0.015; G vs. A: OR = 1.398, 95% CI 1.180, 1.657, P = 0.000; GG vs. AA + GA: OR = 1.686, 95% CI 1.174, 2.423, P = 0.005), and FokI (T vs. C: OR = 1.687, 95% CI 1.474, 1.931, P = 0.000; TT vs. CC: OR = 2.829, 95% CI 2.066, 3.872, P = 0.000; TC vs. CC: OR = 1.579, 95% CI 1.304, 1.913, P = 0.000, TT + TC vs. CC: OR = 1.771, 95% CI 1.477, 2.125, P = 0.000; TT vs. CC + TC: OR = 2.409, 95% CI 1.814, 3.200, P = 0.000) are associated with MM risk. VDR gene polymorphisms including ApaI, BsmI, TaqI, and FokI are associated with MM risk in Asian populations. Additional studies with large sample sizes and different ethnicities are needed.


Subject(s)
Genetic Predisposition to Disease , Multiple Myeloma , Polymorphism, Single Nucleotide , Receptors, Calcitriol , Receptors, Calcitriol/genetics , Multiple Myeloma/genetics , Humans , Asian People/genetics , Case-Control Studies , Odds Ratio
2.
Clin Exp Med ; 24(1): 95, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717497

ABSTRACT

The prognostication of survival trajectories in multiple myeloma (MM) patients presents a substantial clinical challenge. Leveraging transcriptomic and clinical profiles from an expansive cohort of 2,088 MM patients, sourced from the Gene Expression Omnibus and The Cancer Genome Atlas repositories, we applied a sophisticated nested lasso regression technique to construct a prognostic model predicated on 28 gene pairings intrinsic to cell death pathways, thereby deriving a quantifiable risk stratification metric. Employing a threshold of 0.15, we dichotomized the MM samples into discrete high-risk and low-risk categories. Notably, the delineated high-risk cohort exhibited a statistically significant diminution in survival duration, a finding which consistently replicated across both training and external validation datasets. The prognostic acumen of our cell death signature was further corroborated by TIME ROC analyses, with the model demonstrating robust performance, evidenced by AUC metrics consistently surpassing the 0.6 benchmark across the evaluated arrays. Further analytical rigor was applied through multivariate COX regression analyses, which ratified the cell death risk model as an independent prognostic determinant. In an innovative stratagem, we amalgamated this risk stratification with the established International Staging System (ISS), culminating in the genesis of a novel, refined ISS categorization. This tripartite classification system was subjected to comparative analysis against extant prognostic models, whereupon it manifested superior predictive precision, as reflected by an elevated C-index. In summation, our endeavors have yielded a clinically viable gene pairing model predicated on cellular mortality, which, when synthesized with the ISS, engenders an augmented prognostic tool that exhibits pronounced predictive prowess in the context of multiple myeloma.


Subject(s)
Cell Death , Multiple Myeloma , Multiple Myeloma/pathology , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Humans , Prognosis , Male , Female , Risk Assessment , Gene Expression Profiling , Middle Aged , Neoplasm Staging , Aged , Survival Analysis
3.
Zhonghua Xue Ye Xue Za Zhi ; 45(3): 303-307, 2024 Mar 14.
Article in Chinese | MEDLINE | ID: mdl-38716605

ABSTRACT

Multiple myeloma (MM) is a plasma cell neoplasm characterized by numerous chromosomal number and structural abnormalities, which are of great significance for risk stratification and response evaluation of MM patients. Optical genome mapping (OGM) is a novel technology that has the potential to resolve many of the issues and limitations associated with traditional cytogenetic methods. To date, the clinical utility of OGM has been validated in the fields of cancer, reproduction, and embryonic dysplasia, et al. In this study, we compared OGM to traditional techniques for the first time in five newly diagnosed MM patients, and evaluated the potential of OGM for detecting cytogenetic aberrations and its clinical application value in MM.


Subject(s)
Chromosome Aberrations , Multiple Myeloma , Humans , Multiple Myeloma/genetics , Multiple Myeloma/diagnosis , Chromosome Mapping , Middle Aged
4.
Hematology ; 29(1): 2352687, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38767507

ABSTRACT

OBJECTIVE: Examine Bach1 protein expression in bone marrow biopsy specimens obtained from newly diagnosed multiple myeloma (NDMM) and iron deficiency anemia (IDA) patients. Conduct a thorough analysis to explore the potential connection between Bach1 and the onset as well as treatment response of NDMM. METHODS: This study investigated Bach1 expression in bone marrow biopsy tissues from NDMM and IDA patients. Immunohistochemical staining and Image-pro Plus software were utilized for quantitatively obtaining the expression level of Bach1 protein. Arrange Bach1 expression levels from high to low, and use its median expression level as the threshold. Samples with Bach1 expression level above the median are categorized as the high-expression group, while those below the median are categorized as the low-expression group. Under this grouping, a detailed discussion was conducted to explore relationship of the Bach1 expression level with the patients' gender, ISS stage, and survival rate based on the Bortezomib (Btz) therapy. RESULTS: Our experiment indicates that the expression level of Bach1 in NDMM patients is significantly higher than in IDA patients. Furthermore, we discovered that patients in the high-expression group exhibit better prognosis compared to those in the low-expression group after Btz-treatment. Bioinformatics analysis further confirms this conclusion. CONCLUSION: By categorizing Bach1 expression level as high and low, our study offers a unique perspective on understanding the relationship between Bach1 and NDMM.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Multiple Myeloma , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Male , Female , Middle Aged , Aged , Prognosis , Adult , Anemia, Iron-Deficiency/metabolism , Bortezomib/therapeutic use
5.
Technol Cancer Res Treat ; 23: 15330338241252605, 2024.
Article in English | MEDLINE | ID: mdl-38759699

ABSTRACT

OBJECTIVE: 1q21 gain/Amp is one of the most common cytogenetic abnormalities. There are controversies about its effects on prognosis and may be associated with inferior outcomes in patients with newly diagnosed multiple myeloma (NDMM). To explore the optimal induction treatment, we analyzed and compared the efficacy of combinations of bortezomib-lenalidomide-dexamethasone (VRD) and only bortezomib-based triplet regimens without lenalidomide (only bortezomib-based) as induction therapy in patients with NDMM with 1q21 gain/Amp. METHODS: Seventy-six NDMM patients with 1q21 gain/Amp who were admitted to our center from 2016 to 2022 were retrospectively analyzed in this study. The progression and efficacy of the patients were observed. RESULTS: Within our study group, the overall survival rate stood at 75.0%, and the progression-free survival (PFS) rate reached 40.8% in NDMM patients with 1q21 gain/Amp. The best outcome assessment was that 17.1% achieved complete response (CR) and 44.7% achieved very good partial response (VGPR). Patients in the VRD group had a deeper response (VGPR: 63.6% vs 37.0%, P = 0.034), lower disease progression rate (31.8% vs 70.3%, P = 0.002), longer sustained remission (median 49.7 months vs 18.3 months, P = 0.030), and longer PFS (median 61.9 months vs 22.9 months, P = 0.032) than those treated with only bortezomib-based induction therapy. No significant differences were found among patients with partial response or better (86.4% vs 77.8%, P = 0.532) or CR (27.3% vs 13.0%, P = 0.180). Multivariate analysis showed that only bortezomib-based induction therapy (P = 0.003, HR 0.246, 95% CI 0.097-0.620), International Staging System stage III (P = 0.003, HR 3.844, 95% CI 1.588-9.308) and LMR <3.6 (P = 0.032, HR 0.491, 95% CI 0.257-0.940) were significantly associated with adverse PFS. CONCLUSIONS: When compared with the sequential administration of bortezomib and lenalidomide or only bortezomib-based protocols, NDMM patients with 1q21 gain/Amp may benefit more from VRD as initial treatments.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bortezomib , Chromosomes, Human, Pair 1 , Lenalidomide , Multiple Myeloma , Humans , Bortezomib/administration & dosage , Lenalidomide/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/genetics , Female , Male , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Aged , Chromosomes, Human, Pair 1/genetics , Adult , Retrospective Studies , Prognosis , Treatment Outcome , Chromosome Aberrations , Aged, 80 and over , Dexamethasone/administration & dosage
6.
Cell Commun Signal ; 22(1): 258, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711131

ABSTRACT

Although bortezomib (BTZ) is the cornerstone of anti-multiple myeloma (MM) therapy, the inevitable primary and secondary drug resistance still seriously affects the prognosis of patients. New treatment strategies are in need. Sodium-calcium exchanger 1 (NCX1) is a calcium-permeable ion transporter on the membrane, and our previous studies showed that low NCX1 confers inferior viability in MM cells and suppressed osteoclast differentiation. However, the effect of NCX1 on BTZ sensitivity of MM and its possible mechanism remain unclear. In this study, we investigated the effect of NCX1 on BTZ sensitivity in MM, focusing on cellular processes of autophagy and cell viability. Our results provide evidence that NCX1 expression correlates with MM disease progression and low NCX1 expression increases BTZ sensitivity. NCX1/Ca2+ triggered autophagic flux through non-canonical NFκB pathway in MM cells, leading to attenuated the sensitivity of BTZ. Knockdown or inhibition of NCX1 could potentiate the anti-MM activity of BTZ in vitro and vivo, and inhibition of autophagy sensitized NCX1-overexpressing MM cells to BTZ. In general, this work implicates NCX1 as a potential therapeutic target in MM with BTZ resistance and provides novel mechanistic insights into its vital role in combating BTZ resistance.


Subject(s)
Autophagy , Bortezomib , Multiple Myeloma , Sodium-Calcium Exchanger , Sodium-Calcium Exchanger/metabolism , Sodium-Calcium Exchanger/genetics , Humans , Autophagy/drug effects , Animals , Bortezomib/pharmacology , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/genetics , Cell Line, Tumor , Mice , Calcium/metabolism , Drug Resistance, Neoplasm/genetics , NF-kappa B/metabolism , Cell Survival/drug effects
7.
Expert Opin Biol Ther ; 24(5): 365-381, 2024 May.
Article in English | MEDLINE | ID: mdl-38757726

ABSTRACT

INTRODUCTION: Gain/amplification of 1q (+1q) represents one of the most prevalent cytogenetic abnormalities (CAs) observed in multiple myeloma (MM). Historical studies predating the advent of anti-CD38 monoclonal antibodies (moAbs) implicated + 1q in poor prognoses, prompting its integration into novel staging systems. However, with the emergence of daratumumab and isatuximab, two pivotal anti-CD38 moAbs, the landscape of MM therapy has undergone a profound transformation. AREAS COVERED: This review encompasses a comprehensive analysis of diverse study methodologies, including observational investigations, clinical trials, meta-analyses, and real-world database analyses. By synthesizing these data sources, we aim to provide an overview of the current understanding of + 1q in the context of anti-CD38 moAbs therapies. EXPERT OPINION: Despite the paucity of available data, evidence suggests a potential mitigating effect of daratumumab on the adverse prognostic implications of + 1q. However, this benefit seems to diminish in patients harboring ≥ 4 copies or with concurrent high-risk CAs. On the other hand, isatuximab demonstrated promising outcomes in the relapsed-refractory setting for + 1q MM patients. Nevertheless, direct comparison between the two compounds is currently challenging. The current evidence firmly supports the integration of anti-CD38 moAb-based therapies as the standard of care for + 1q patients, pending further elucidation.


Subject(s)
ADP-ribosyl Cyclase 1 , Antibodies, Monoclonal , Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/immunology , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/genetics , Antibodies, Monoclonal/therapeutic use , Chromosomes, Human, Pair 1/genetics , Membrane Glycoproteins , Antibodies, Monoclonal, Humanized
8.
J Cancer Res Clin Oncol ; 150(5): 239, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38713252

ABSTRACT

PURPOSE: Multiple myeloma (MM) is an incurable hematological malignancy characterized by clonal proliferation of malignant plasma B cells in bone marrow, and its pathogenesis remains unknown. The aim of this study was to determine the role of kinesin family member 22 (KIF22) in MM and elucidate its molecular mechanism. METHODS: The expression of KIF22 was detected in MM patients based upon the public datasets and clinical samples. Then, in vitro assays were performed to investigate the biological function of KIF22 in MM cell lines, and subcutaneous xenograft models in nude mice were conducted in vivo. Chromatin immunoprecipitation (ChIP) and luciferase reporter assay were used to determine the mechanism of KIF22-mediated regulation. RESULTS: The results demonstrated that the expression of KIF22 in MM patients was associated with several clinical features, including gender (P = 0.016), LDH (P < 0.001), ß2-MG (P = 0.003), percentage of tumor cells (BM) (P = 0.002) and poor prognosis (P < 0.0001). Furthermore, changing the expression of KIF22 mainly influenced the cell proliferation in vitro and tumor growth in vivo, and caused G2/M phase cell cycle dysfunction. Mechanically, KIF22 directly transcriptionally regulated cell division cycle 25C (CDC25C) by binding its promoter and indirectly influenced CDC25C expression by regulating the ERK pathway. KIF22 also regulated CDC25C/CDK1/cyclinB1 pathway. CONCLUSION: KIF22 could promote cell proliferation and cell cycle progression by transcriptionally regulating CDC25C and its downstream CDC25C/CDK1/cyclinB1 pathway to facilitate MM progression, which might be a potential therapeutic target in MM.


Subject(s)
CDC2 Protein Kinase , Cyclin B1 , DNA-Binding Proteins , Kinesins , Multiple Myeloma , cdc25 Phosphatases , Animals , Female , Humans , Male , Mice , Middle Aged , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/genetics , cdc25 Phosphatases/metabolism , cdc25 Phosphatases/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin B1/metabolism , Cyclin B1/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Kinesins/metabolism , Kinesins/genetics , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/genetics , Prognosis , Signal Transduction
9.
Nat Commun ; 15(1): 4139, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755155

ABSTRACT

The natural history of multiple myeloma is characterized by its localization to the bone marrow and its interaction with bone marrow stromal cells. The bone marrow stromal cells provide growth and survival signals, thereby promoting the development of drug resistance. Here, we show that the interaction between bone marrow stromal cells and myeloma cells (using human cell lines) induces chromatin remodeling of cis-regulatory elements and is associated with changes in the expression of genes involved in the cell migration and cytokine signaling. The expression of genes involved in these stromal interactions are observed in extramedullary disease in patients with myeloma and provides the rationale for survival of myeloma cells outside of the bone marrow microenvironment. Expression of these stromal interaction genes is also observed in a subset of patients with newly diagnosed myeloma and are akin to the transcriptional program of extramedullary disease. The presence of such adverse stromal interactions in newly diagnosed myeloma is associated with accelerated disease dissemination, predicts the early development of therapeutic resistance, and is of independent prognostic significance. These stromal cell induced transcriptomic and epigenomic changes both predict long-term outcomes and identify therapeutic targets in the tumor microenvironment for the development of novel therapeutic approaches.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells , Multiple Myeloma , Tumor Microenvironment , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Humans , Tumor Microenvironment/genetics , Cell Line, Tumor , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Transcription, Genetic , Bone Marrow Cells/metabolism , Cell Movement/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Female , Male
10.
Oncoimmunology ; 13(1): 2360275, 2024.
Article in English | MEDLINE | ID: mdl-38812570
11.
Cell Rep ; 43(5): 114211, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38722741

ABSTRACT

Multiple myeloma (MM) remains an incurable hematological malignancy demanding innovative therapeutic strategies. Targeting MYC, the notorious yet traditionally undruggable oncogene, presents an appealing avenue. Here, using a genome-scale CRISPR-Cas9 screen, we identify the WNK lysine-deficient protein kinase 1 (WNK1) as a regulator of MYC expression in MM cells. Genetic and pharmacological inhibition of WNK1 reduces MYC expression and, further, disrupts the MYC-dependent transcriptional program. Mechanistically, WNK1 inhibition attenuates the activity of the immunoglobulin heavy chain (IgH) enhancer, thus reducing MYC transcription when this locus is translocated near the MYC locus. WNK1 inhibition profoundly impacts MM cell behaviors, leading to growth inhibition, cell-cycle arrest, senescence, and apoptosis. Importantly, the WNK inhibitor WNK463 inhibits MM growth in primary patient samples as well as xenograft mouse models and exhibits synergistic effects with various anti-MM compounds. Collectively, our study uncovers WNK1 as a potential therapeutic target in MM.


Subject(s)
Multiple Myeloma , Proto-Oncogene Proteins c-myc , WNK Lysine-Deficient Protein Kinase 1 , Multiple Myeloma/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism , WNK Lysine-Deficient Protein Kinase 1/genetics , Humans , Animals , Mice , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Gene Expression Regulation, Neoplastic/drug effects , Immunoglobulin Heavy Chains/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Xenograft Model Antitumor Assays
12.
BMC Nephrol ; 25(1): 165, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755555

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is a malignant disorder characterized by monoclonal differentiated plasma cells. While it is more commonly diagnosed in elderly individuals, it can also affect younger populations, though with a lower incidence. CASE PRESENTATION: Here, we present the case of a 32-year-old woman diagnosed with IgA lambda MM. She presented with fatigue, nausea, acute kidney injury (AKI) with a rapid increase in creatinine, and anemia. A kidney biopsy was done to rule out a rapidly progressive glomerular disease and a diagnosis was thus reached. A genetic workup revealed t(14;16) translocation and an extra copy of TP53. The patient received aggressive intravenous steroids and intravenous fluid resuscitation, resulting in an improvement in renal function. Treatment with daratumumab in combination with bortezomib, thalidomide, and dexamethasone was initiated and well tolerated. Despite the generally poor prognosis of IgA MM, our case emphasizes the importance of considering MM in young patients with unexplained kidney injury. CONCLUSION: Early recognition and prompt intervention are essential in managing MM patients, especially in those with high-risk cytogenetic abnormalities. This case serves as a reminder for clinicians to maintain a high index of suspicion for MM, even in younger populations, when presented with unexplained kidney injury.


Subject(s)
Acute Kidney Injury , Multiple Myeloma , Proteinuria , Translocation, Genetic , Humans , Female , Adult , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Proteinuria/etiology , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Immunoglobulin A , Immunoglobulin lambda-Chains/genetics , Chromosomes, Human, Pair 14/genetics
13.
Nat Commun ; 15(1): 4144, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755140

ABSTRACT

Multiple Myeloma is an incurable plasma cell malignancy with a poor survival rate that is usually treated with immunomodulatory drugs (iMiDs) and proteosome inhibitors (PIs). The malignant plasma cells quickly become resistant to these agents causing relapse and uncontrolled growth of resistant clones. From whole genome sequencing (WGS) and RNA sequencing (RNA-seq) studies, different high-risk translocation, copy number, mutational, and transcriptional markers can be identified. One of these markers, PHF19, epigenetically regulates cell cycle and other processes and is already studied using RNA-seq. In this study, we generate a large (325,025 cells and 49 patients) single cell multi-omic dataset and jointly quantify ATAC- and RNA-seq for each cell and matched genomic profiles for each patient. We identify an association between one plasma cell subtype with myeloma progression that we call relapsed/refractory plasma cells (RRPCs). These cells are associated with chromosome 1q alterations, TP53 mutations, and higher expression of PHF19. We also identify downstream regulation of cell cycle inhibitors in these cells, possible regulation by the transcription factor (TF) PBX1 on chromosome 1q, and determine that PHF19 may be acting primarily through this subset of cells.


Subject(s)
Chromosomes, Human, Pair 1 , DNA-Binding Proteins , Multiple Myeloma , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/drug therapy , Humans , Chromosomes, Human, Pair 1/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Plasma Cells/metabolism , Mutation , Neoplasm Recurrence, Local/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Drug Resistance, Neoplasm/genetics , Gene Amplification
14.
Sci Rep ; 14(1): 11593, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773213

ABSTRACT

Multiple myeloma (MM) progression involves diminished tumor antigen presentation and an immunosuppressive microenvironment, characterized by diminished expression of major histocompatibility complexes (MHC) class I molecule and elevated programmed death ligand 1 (PDL1) in MM cells, along with an enriched population of regulatory T cells (Tregs). To investigate Treg's influence on MM cells, we established a co-culture system using Tregs from MM patients and the MM cell lines (MM.1S and SK-MM-1) in vitro and assessed the effects of intervening in the relevant pathways connecting Tregs and MM cells in vivo. In vitro, Tregs induced transforming growth factor beta-1 (TGF-ß1) production, downregulated MHC I members, and increased PDL1 expression in MM cells. Treg-derived TGF-ß1 suppressed the cGAS-STING pathway, contributing to the loss of MHC I molecule expression and PDL1 upregulation. Correspondingly, neutralizing TGF-ß1 or activating the cGAS-STING pathway restored MHC I and PDL1 expression, effectively countering the pro-tumorigenic effect of Tregs on MM cells in vivo. These data elucidated how Tregs influence tumor antigen presentation and immunosuppressive signal in MM cells, potentially providing therapeutic strategies, such as neutralizing TGF-ß1 or activating the cGAS-STING pathway, to address the immune escape and immunosuppressive dynamics in MM.


Subject(s)
B7-H1 Antigen , Histocompatibility Antigens Class I , Membrane Proteins , Multiple Myeloma , Nucleotidyltransferases , Signal Transduction , T-Lymphocytes, Regulatory , Transforming Growth Factor beta1 , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Multiple Myeloma/genetics , Transforming Growth Factor beta1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Animals , Down-Regulation , Mice , Female , Coculture Techniques , Male , Gene Expression Regulation, Neoplastic
15.
Nat Commun ; 15(1): 3844, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714690

ABSTRACT

Multiple myeloma (MM) is a heterogeneous disease characterized by frequent MYC translocations. Sporadic MYC activation in the germinal center of genetically engineered Vk*MYC mice is sufficient to induce plasma cell tumors in which a variety of secondary mutations are spontaneously acquired and selected over time. Analysis of 119 Vk*MYC myeloma reveals recurrent copy number alterations, structural variations, chromothripsis, driver mutations, apolipoprotein B mRNA-editing enzyme, catalytic polypeptide (APOBEC) mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identify frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC. In summary, here we credential the Vk*MYC mouse as a unique resource to explore MM genomic evolution and describe a fully annotated collection of diverse and immortalized murine MM tumors.


Subject(s)
Multiple Myeloma , Proto-Oncogene Proteins c-myc , Animals , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Humans , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Cell Transformation, Neoplastic/genetics , Mutation , Signal Transduction/genetics , Mice, Transgenic , NF-kappa B/metabolism , NF-kappa B/genetics , Mutagenesis, Insertional , DNA Copy Number Variations/genetics , Genomics/methods , Translocation, Genetic
17.
Int J Mol Sci ; 25(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38791247

ABSTRACT

Over the last decades, the survival of multiple myeloma (MM) patients has considerably improved. However, despite the availability of new treatments, most patients still relapse and become therapy-resistant at some point in the disease evolution. The mutation profile has an impact on MM patients' outcome, while typically evolving over time. Because of the patchy bone marrow (BM) infiltration pattern, the analysis of a single bone marrow sample can lead to an underestimation of the known genetic heterogeneity in MM. As a result, interest is shifting towards blood-derived liquid biopsies, which allow for a more comprehensive and non-invasive genetic interrogation without the discomfort of repeated BM aspirations. In this review, we compare the application potential for mutation profiling in MM of circulating-tumor-cell-derived DNA, cell-free DNA and extracellular-vesicle-derived DNA, while also addressing the challenges associated with their use.


Subject(s)
Multiple Myeloma , Mutation , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/diagnosis , Humans , Liquid Biopsy/methods , Circulating Tumor DNA/genetics , Cell-Free Nucleic Acids/genetics , Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism
18.
Int J Mol Sci ; 25(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38791307

ABSTRACT

Multiple myeloma (MM) is the second most common hematological malignancy, which remains incurable despite recent advances in treatment strategies. Like other forms of cancer, MM is characterized by genomic instability, caused by defects in DNA repair. Along with mutations in DNA repair genes and genotoxic drugs used to treat MM, non-canonical secondary DNA structures (four-stranded G-quadruplex structures) can affect accumulation of somatic mutations and chromosomal abnormalities in the tumor cells of MM patients. Here, we tested the hypothesis that G-quadruplex structures may influence the distribution of somatic mutations in the tumor cells of MM patients. We sequenced exomes of normal and tumor cells of 11 MM patients and analyzed the data for the presence of G4 context around points of somatic mutations. To identify molecular mechanisms that could affect mutational profile of tumors, we also analyzed mutational signatures in tumor cells as well as germline mutations for the presence of specific SNPs in DNA repair genes or in genes regulating G-quadruplex unwinding. In several patients, we found that sites of somatic mutations are frequently located in regions with G4 context. This pattern correlated with specific germline variants found in these patients. We discuss the possible implications of these variants for mutation accumulation and specificity in MM and propose that the extent of G4 context enrichment around somatic mutation sites may be a novel metric characterizing mutational processes in tumors.


Subject(s)
G-Quadruplexes , Multiple Myeloma , Mutation , Humans , Multiple Myeloma/genetics , Polymorphism, Single Nucleotide , DNA Repair/genetics , Genomic Instability
19.
Oncol Res ; 32(4): 753-768, 2024.
Article in English | MEDLINE | ID: mdl-38560563

ABSTRACT

Multiple myeloma (MM) is a hematologic malignancy notorious for its high relapse rate and development of drug resistance, in which cell adhesion-mediated drug resistance plays a critical role. This study integrated four RNA sequencing datasets (CoMMpass, GSE136337, GSE9782, and GSE2658) and focused on analyzing 1706 adhesion-related genes. Rigorous univariate Cox regression analysis identified 18 key prognosis-related genes, including KIF14, TROAP, FLNA, MSN, LGALS1, PECAM1, and ALCAM, which demonstrated the strongest associations with poor overall survival (OS) in MM patients. To comprehensively evaluate the impact of cell adhesion on MM prognosis, an adhesion-related risk score (ARRS) model was constructed using Lasso Cox regression analysis. The ARRS model emerged as an independent prognostic factor for predicting OS. Furthermore, our findings revealed that a heightened cell adhesion effect correlated with tumor resistance to DNA-damaging drugs, protein kinase inhibitors, and drugs targeting the PI3K/Akt/mTOR signaling pathway. Nevertheless, we identified promising drug candidates, such as tirofiban, pirenzepine, erlotinib, and bosutinib, which exhibit potential in reversing this resistance. In vitro, experiments employing NCIH929, RPMI8226, and AMO1 cell lines confirmed that MM cell lines with high ARRS exhibited poor sensitivity to the aforementioned candidate drugs. By employing siRNA-mediated knockdown of the key ARRS model gene KIF14, we observed suppressed proliferation of NCIH929 cells, along with decreased adhesion to BMSCs and fibronectin. This study presents compelling evidence establishing cell adhesion as a significant prognostic factor in MM. Additionally, potential molecular mechanisms underlying adhesion-related resistance are proposed, along with viable strategies to overcome such resistance. These findings provide a solid scientific foundation for facilitating clinically stratified treatment of MM.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Cell Adhesion/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Neoplasm Recurrence, Local
20.
Cancer Immunol Res ; 12(4): 385-386, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38562081

ABSTRACT

All chimeric antigen receptor (CAR) T-cell products currently approved by the FDA are autologous, which poses several challenges for widespread use. In this issue, Degagné and colleagues present their preclinical research on creating off-the-shelf CAR T cells for multiple myeloma. They utilized the CRISPR/Cas12a genome editing platform and gene knock-in techniques to eliminate alloreactivity and decrease susceptibility to natural killer (NK)-cell elimination. This work has led to an ongoing phase I trial of off-the-shelf CAR T cells for multiple myeloma treatment. See related article by Degagné et al., p. 462 (2).


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Immunotherapy, Adoptive/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...