Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cancers (Basel) ; 15(18)2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37760488

ABSTRACT

Multiple myeloma (MM) induces dysfunctional bone marrow (BM) mesenchymal cells and neoangiogenesis. Pericytes and smooth muscle cells (SMCs) could detach from vessels and become cancer-associated fibroblasts. We found that the pericyte and SMC marker endothelin receptor type A (EDNRA) is overexpressed in whole MM bone biopsies; we sought to characterize its expression. EDNRA expression gradually increased with disease progression. High-risk MM patients had higher EDNRA expression than low-risk MM patients and EDNRA expression was highest in focal lesions. High EDNRA expression was associated with high expression of pericyte markers (e.g., RGS5, POSTN, and CD146) and the angiogenic marker FLT1. A single-cell analysis of unexpanded BM mesenchymal cells detected EDNRA expression in a subset of cells that coexpressed mesenchymal cell markers and had higher expression of proliferation genes. Immunohistochemistry revealed that the number of EDNRA+ cells in the interstitial BM increased as MM progressed; EDNRA+ cells were prevalent in areas near the MM focal growth. EDNRA+ cells were detached from CD34+ angiogenic cells and coexpressed RGS5 and periostin. Therefore, they likely originated from pericytes or SMCs. These findings identify a novel microenvironmental biomarker in MM and suggest that the presence of detached EDNRA+ cells indicates disrupted vasculature and increased angiogenesis.

2.
Leuk Res ; 133: 107355, 2023 10.
Article in English | MEDLINE | ID: mdl-37499483

ABSTRACT

Bone marrow mesenchymal stem cells (MSCs) may have contrasting impacts on the progression of multiple myeloma (MM). Priming normal MSCs, by culturing them with MM cells, mimics the MSC-induced MM growth. We studied the contrasting effects of conditioned medium (CM) from unprimed or primed MSCs on growth of MM cells from newly diagnosed cases. We elucidated potential molecular pathways using global gene expression profiling and focused on the role of the mTOR2 component, RICTOR, as a novel mediator of dormancy in MM. Primed MSCs CM consistently increased proportions of proliferating cells and supported MM growth in 3-day (n = 20) and 10-day (n = 12) cultures, effects that were partially mediated through the IGF1 axis. In contrast, unprimed MSCs CM inhibited growth of MM cells in cases mainly from stages I/II MM. The genes most overexpressed in MM cells treated with primed MSCs CM were associated with cell cycle, DNA-damage repair, and proliferation; genes most overexpressed in MM cells treated with unprimed MSCs CM were associated with dormancy pathways including RICTOR (mTOR2 pathway), CXCR4, and BCL2. RICTOR protein level was induced by unprimed MSCs CM and was lower in KI67+ proliferating MM cells treated with primed MSCs CM. RICTOR was underexpressed in clinical relapse samples compared with baseline samples of the same patients. Inhibiting RICTOR expression in primary MM cells promoted their growth, and enforced expression of RICTOR in MM cell lines inhibited their growth. Our findings suggest that, after prolonged interactions with MM cells, bone marrow MSCs shift from MM-repressive to MM-permissive. AVAILABILITY OF DATA AND MATERIALS: Our institutional GEP data of MM cells from newly diagnosed patients used to show RICTOR expression have been deposited at Gene Expression Omnibus (GEO: GSE2658, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE2658).


Subject(s)
Mesenchymal Stem Cells , Multiple Myeloma , Humans , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplasm Recurrence, Local/metabolism , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Gene Expression Profiling , Cell Proliferation
3.
Genome Res ; 32(7): 1343-1354, 2022 07.
Article in English | MEDLINE | ID: mdl-34933939

ABSTRACT

Chromosomal translocations are important drivers of haematological malignancies whereby proto-oncogenes are activated by juxtaposition with enhancers, often called enhancer hijacking We analyzed the epigenomic consequences of rearrangements between the super-enhancers of the immunoglobulin heavy locus (IGH) and proto-oncogene CCND1 that are common in B cell malignancies. By integrating BLUEPRINT epigenomic data with DNA breakpoint detection, we characterized the normal chromatin landscape of the human IGH locus and its dynamics after pathological genomic rearrangement. We detected an H3K4me3 broad domain (BD) within the IGH locus of healthy B cells that was absent in samples with IGH-CCND1 translocations. The appearance of H3K4me3-BD over CCND1 in the latter was associated with overexpression and extensive chromatin accessibility of its gene body. We observed similar cancer-specific H3K4me3-BDs associated with hijacking of super-enhancers of other common oncogenes in B cell (MAF, MYC, and FGFR3/NSD2) and T cell malignancies (LMO2, TLX3, and TAL1). Our analysis suggests that H3K4me3-BDs can be created by super-enhancers and supports the new concept of epigenomic translocation, in which the relocation of H3K4me3-BDs from cell identity genes to oncogenes accompanies the translocation of super-enhancers.


Subject(s)
Epigenomics , Translocation, Genetic , Chromatin/genetics , Histones , Humans , Oncogenes
4.
Curr Res Transl Med ; 69(3): 103290, 2021 07.
Article in English | MEDLINE | ID: mdl-33894670

ABSTRACT

Epigenetic deregulation is increasingly recognized as a contributing pathological factor in multiple myeloma (MM). In particular tri-methylation of H3 lysine 27 (H3K27me3), which is catalyzed by PHD finger protein 19 (PHF19), a subunit of the Polycomb Repressive Complex 2 (PRC2), has recently shown to be a crucial mediator of MM tumorigenicity. Overexpression of PHF19 in MM has been associated with worse clinical outcome. Yet, while there is mounting evidence that PHF19 overexpression plays a crucial role in MM carcinogenesis downstream mechanisms remain to be elucidated. In the current study we use a functional knock down (KD) of PHF19 to investigate the biological role of PHF19 and show that PHF19KD leads to decreased tumor growth in vitro and in vivo. Expression of major cancer players such as bcl2, myc and EGR1 were decreased upon PHF19KD further underscoring the role of PHF19 in MM biology. Additionally, our results highlighted the prognostic impact of PHF19 overexpression, which was significantly associated with worse survival. Overall, our study underscores the premise that targeting the PHF19-PRC2 complex would open up avenues for novel MM therapies.


Subject(s)
Multiple Myeloma , Cell Proliferation , DNA-Binding Proteins , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Polycomb Repressive Complex 2 , Transcription Factors/genetics
5.
Haematologica ; 105(4): 1055-1066, 2020 04.
Article in English | MEDLINE | ID: mdl-31221783

ABSTRACT

MYC is a widely acting transcription factor and its deregulation is a crucial event in many human cancers. MYC is important biologically and clinically in multiple myeloma, but the mechanisms underlying its dysregulation are poorly understood. We show that MYC rearrangements are present in 36.0% of newly diagnosed myeloma patients, as detected in the largest set of next generation sequencing data to date (n=1,267). Rearrangements were complex and associated with increased expression of MYC and PVT1, but not other genes at 8q24. The highest effect on gene expression was detected in cases where the MYC locus is juxtaposed next to super-enhancers associated with genes such as IGH, IGK, IGL, TXNDC5/BMP6, FAM46C and FOXO3 We identified three hotspots of recombination at 8q24, one of which is enriched for IGH-MYC translocations. Breakpoint analysis indicates primary myeloma rearrangements involving the IGH locus occur through non-homologous end joining, whereas secondary MYC rearrangements occur through microhomology-mediated end joining. This mechanism is different to lymphomas, where non-homologous end joining generates MYC rearrangements. Rearrangements resulted in overexpression of key genes and chromatin immunoprecipitation-sequencing identified that HK2, a member of the glucose metabolism pathway, is directly over-expressed through binding of MYC at its promoter.


Subject(s)
Genes, myc , Multiple Myeloma , RNA, Long Noncoding/genetics , Genes, Immunoglobulin Heavy Chain , Humans , In Situ Hybridization, Fluorescence , Multiple Myeloma/genetics , Protein Disulfide-Isomerases , Translocation, Genetic
6.
Br J Haematol ; 184(4): 578-593, 2019 02.
Article in English | MEDLINE | ID: mdl-30408155

ABSTRACT

Recent studies suggest that multiple myeloma (MM) induces proliferation and expansion of bone marrow (BM) mesenchymal stem cells (MSCs), but others showed that MM cells induce MSC senescence. To clarify the interaction between MM and MSCs, we exploited our established MSC gene signature to identify gene expression changes in myeloma MSCs and associated functional differences. Single MSCs from patients with MM had changes in expression of genes associated with cellular proliferation and senescence and a higher proportion of senescent cells and lower proliferative potential than those from age-matched healthy donors. Single MSCs from both sources heterogeneously express MSC genes associated with adipogenesis and osteoblastogenesis. We identified the gene encoding insulin-like growth factor-binding protein 2 (IGFBP2), an MSC gene commonly altered in high risk MM, as under-expressed. Morphologically, IGFBP2+ cells are underrepresented in MM BM compared to smouldering MM. Strong IGFBP2 and adiponectin co-expression was detected in a subset of small adipocytes. Co-culturing normal MSCs with myeloma cells suppressed MSC differentiation to adipocytes and osteoblasts, and reduced expression of IGFBP2 and adiponectin. Recombinant IGFBP2 blocked IGF1-mediated myeloma cell growth. Our data demonstrate that myeloma MSCs are less proliferative and that IGFBP2+ small adipocytes are a distinct mesenchymal cell population suppressed by myeloma.


Subject(s)
Adipocytes , Bone Marrow , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Mesenchymal Stem Cells , Multiple Myeloma , Neoplasm Proteins/biosynthesis , Adipocytes/metabolism , Adipocytes/pathology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Risk Factors
7.
Blood ; 132(1): 59-66, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29784643

ABSTRACT

Spatial intratumor heterogeneity is frequently seen in multiple myeloma (MM) and poses a significant challenge for risk classifiers, which rely on tumor samples from the iliac crest. Because biopsy-based assessment of multiple skeletal sites is difficult, alternative strategies for risk stratification are required. Recently, the size of focal lesions (FLs) was shown to be a surrogate marker for spatial heterogeneity, suggesting that data from medical imaging could be used to improve risk stratification approaches. Here, we investigated the prognostic value of FL size in 404 transplant-eligible, newly diagnosed MM patients. Using diffusion-weighted magnetic resonance imaging with background suppression, we identified the presence of multiple large FLs as a strong prognostic factor. Patients with at least 3 large FLs with a product of the perpendicular diameters >5 cm2 were associated with poor progression-free survival (PFS) and overall survival (OS; median, 2.3 and 3.6 years, respectively). This pattern, seen in 13.8% of patients, was independent of the Revised International Staging System (RISS), gene expression profiling (GEP)-based risk score, gain(1q), or extramedullary disease (hazard ratio, 2.7 and 2.2 for PFS and OS in multivariate analysis, respectively). The number of FLs lost its negative impact on outcome after adjusting for FL size. In conclusion, the presence of at least 3 large FL is a feature of high risk, which can be used to refine the diagnosis of this type of disease behavior and as an entry criterion for risk-stratified trials.


Subject(s)
Diffusion Magnetic Resonance Imaging , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/mortality , Adult , Disease-Free Survival , Female , Humans , Male , Middle Aged , Risk Assessment , Risk Factors , Survival Rate
8.
Clin Cancer Res ; 24(12): 2913-2919, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29563136

ABSTRACT

Purpose: Mesenchymal stem cells (MSC) are an essential component of the bone marrow microenvironment and have shown to support cancer evolution in multiple myeloma. Despite the increasing evidence that multiple myeloma MSCs differ from their healthy counterparts, little knowledge exists as to whether MSCs independently influence disease outcome. The aim of this study was to determine the importance of MSCs in disease progression and outcome in multiple myeloma.Experimental Design: To determine the impact of MSCs on multiple myeloma outcome in an in vivo system, we first identified genes from cultured MSCs that were specific to MSC expression and were not or minimally expressed in plasma cells (PC) or other cells present in bone marrow aspirates. We then applied this MSC gene signature to whole bone marrow biopsies of multiple myeloma patients compared with healthy controls and determined MSC expression scores specific to multiple myeloma and predictive of outcome.Results: We show that multiple myeloma MSC gene expression signatures can differentiate multiple myeloma from monoclonal gammopathy and smoldering multiple myeloma (SMM) as well as from healthy controls and treated multiple myeloma patients who have achieved a complete remission. We identified a prognostic gene score based on three MSC specific genes, COL4A1, NPR3 and ITGBL1, that was able to predict progression-free survival in multiple myeloma patients and progression into multiple myeloma from SMM.Conclusions: Our findings show that progression of multiple myeloma and of SMM into multiple myeloma does not rely solely on intrinsic PC factors, but is independently affected by the biology of the surrounding microenvironment. Clin Cancer Res; 24(12); 2913-9. ©2018 AACR.


Subject(s)
Biomarkers, Tumor , Gene Expression , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Biopsy , Bone Marrow/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/mortality , Multiple Myeloma/diagnosis , Prognosis , Survival Analysis , Transcriptome
9.
Clin Lymphoma Myeloma Leuk ; 18(1): 38-43, 2018 01.
Article in English | MEDLINE | ID: mdl-29066162

ABSTRACT

The identity of so-called malignant stem cells in multiple myeloma has long been controversial. However, it is now appreciated that a small population of myeloma cells has a stem-like capacity for self-renewal and resides within the population of recognizable myeloma plasma cells, although whether these cells are quiescent or proliferative remains unresolved. It is also now accepted that subpopulations of myeloma plasma cells alternate bidirectionally and dynamically between immature and mature phenotypes, and both subpopulations are capable of clonogenicity in vitro and in animal models. Mounting evidence suggests that in multiple myeloma, small populations of quiescent myeloma stem cells behave as tumor-initiating cells as a result of their interaction with the endosteal bone marrow niche. However, a different, small population of proliferative myeloma stem cells produces the tumor bulk, and this population is likely responsible for the emergence of evolved subclones. A cellular-based plasticity model in which 2 states of stem-like cell compartments-quiescent and proliferative-exist simultaneously and are regulated dynamically by the microenvironment, epigenetics, and genetic aberrations may explain various clinical scenarios and the lack of consensus among some reported studies.


Subject(s)
Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Cell Differentiation , Humans
12.
Blood ; 130(1): 30-34, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28432222

ABSTRACT

18F-Fluorodeoxyglucose (FDG)-positron emission tomography (PET) and diffusion-weighted magnetic resonance imaging with background signal suppression (DWIBS) are 2 powerful functional imaging modalities in the evaluation of malignant plasma cell (PC) disease multiple myeloma (MM). Preliminary observations have suggested that MM patients with extensive disease according to DWIBS may be reported as being disease-free on FDG-PET ("PET false-negative"). The aim of this study was to describe the proportion of PET false-negativity in a representative set of 227 newly diagnosed MM patients with simultaneous assessment of FDG-PET and DWIBS, and to identify tumor-intrinsic features associated with this pattern. We found the incidence of PET false-negativity to be 11%. Neither tumor load-associated parameters, such as degree of bone marrow PC infiltration, nor the PC proliferation rate were associated with this subset. However, the gene coding for hexokinase-2, which catalyzes the first step of glycolysis, was significantly lower expressed in PET false-negative cases (5.3-fold change, P < .001) which provides a mechanistic explanation for this feature. In conclusion, we demonstrate a relevant number of patients with FDG-PET false-negative MM and a strong association between hexokinase-2 expression and this negativity: a finding which may also be relevant for clinical imaging of other hematological cancers.


Subject(s)
Fluorodeoxyglucose F18/administration & dosage , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hexokinase/biosynthesis , Multiple Myeloma , Neoplasm Proteins/biosynthesis , Positron-Emission Tomography , False Positive Reactions , Female , Humans , Male , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/enzymology
13.
J Bone Miner Res ; 32(6): 1261-1266, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28240368

ABSTRACT

Osteolytic bone lesions are a hallmark of multiple myeloma (MM) bone disease. Bone destruction is associated with severely imbalanced bone remodeling, secondary to increased osteoclastogenesis and significant osteoblast suppression. Lytic lesions of the pelvis are relatively common in MM patients and are known to contribute to the increased morbidity because of the high risk of fracture, which frequently demands extensive surgical intervention. After observing unexpected radiological improvement in serial large pelvic CT assessment in a patient treated in a total therapy protocol, the radiographic changes of pelvic osteolytic lesions by PET/CT scanning in patients who received Total Therapy 4 (TT4) treatment for myeloma were retrospectively analyzed. Sixty-two (62) patients with lytic pelvic lesions >1 cm in diameter were identified at baseline PET/CT scanning. Follow-up CT studies showed that 27 of 62 patients (43%) with large baseline pelvic lesions achieved significant reaccumulation of radiodense mineralization at the lytic cortical site. The average size of lytic lesions in which remineralization occurred was 4 cm (range, 1.3 to 10 cm). This study clearly demonstrates that mineral deposition in large pelvic lesions occurs in a significant proportion of MM patients treated with TT4, potentially affecting patient outcomes, quality of life, and future treatment strategies. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Calcification, Physiologic , Multiple Myeloma/physiopathology , Multiple Myeloma/therapy , Pelvis/pathology , Pelvis/physiopathology , Adult , Aged , Alkaline Phosphatase/metabolism , Female , Humans , Imaging, Three-Dimensional , Male , Middle Aged , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/pathology , Pelvis/diagnostic imaging , Tomography, X-Ray Computed
14.
Clin Cancer Res ; 23(11): 2665-2672, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27810902

ABSTRACT

Purpose: To determine whether a reduction in the intensity of Total Therapy (TT) reduces toxicity and maintains efficacy.Experimental Design: A total of 289 patients with gene expression profiling (GEP70)-defined low-risk multiple myeloma were randomized between a standard arm (TT4-S) and a light arm (TT4-L). TT4-L employed one instead of two inductions and consolidations. To compensate for potential loss of efficacy of TT4-L, bortezomib and thalidomide were added to fractionated melphalan 50 mg/m2/d for 4 days.Results: Grade ≥3 toxicities and treatment-related mortalities were not reduced in TT4-L. Complete response (CR) rates were virtually identical (P = 0.2; TT4-S, 59%; TT4-L, 61% at 2 years), although CR duration was superior with TT4-S (P = 0.05; TT4-S, 87%; TT4-L, 81% at 2 years). With a median follow-up of 4.5 years, there was no difference in overall survival (OS) and progression-free survival (PFS). Whereas metaphase cytogenetic abnormalities (CAs) tended to be an adverse feature in TT4-S, as with predecessor TT trials, the reverse applied to TT4-L. Employing historical TT3a as training and TT3b as test set, 51 gene probes (GEP51) significantly differentiated the presence and absence of CA (q < 0.0001), seven of which function in DNA replication, recombination, and repair. Applying the GEP51 model to clinical outcomes, OS and PFS were significantly inferior with GEP51/CA in TT4-S; such a difference was not observed in TT4-L.Conclusions: We identified a prognostic CA-linked GEP51 signature, the adversity of which could be overcome by potentially synergizing anti-multiple myeloma effects of melphalan and bortezomib. These exploratory findings require confirmation in a prospective randomized trial. Clin Cancer Res; 23(11); 2665-72. ©2016 AACR.


Subject(s)
Bortezomib/administration & dosage , Melphalan/administration & dosage , Multiple Myeloma/drug therapy , Thalidomide/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/adverse effects , Chromosome Aberrations/drug effects , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Melphalan/adverse effects , Metaphase/drug effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Prognosis , Prospective Studies , Thalidomide/adverse effects
15.
Immunotherapy ; 8(3): 367-84, 2016.
Article in English | MEDLINE | ID: mdl-26888183

ABSTRACT

Multiple myeloma is a plasma cell malignancy that is characterized by refractory and relapsing course of disease. Despite the introduction of high-dose chemotherapy in combination with autologous stem cell transplantation and innovative agents such as proteasome inhibitors and immunomodulatory drugs, achieving cure in multiple myeloma is a challenging endeavor. In the last couple of years, enormous advances were made in implementing monoclonal antibody therapy in multiple myeloma. A large number of preclinical and clinical studies have been introduced successfully, demonstrating a safe and efficient administration of monoclonal antibodies in multiple myeloma. In particular, the application of monoclonal antibodies in combination with immunomodulatory drugs, proteasome inhibitors, corticosteroids or conventional chemotherapy seem to be promising and will expand the treatment arsenal for patients with multiple myeloma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunologic Factors/therapeutic use , Immunotherapy/methods , Multiple Myeloma/therapy , Proteasome Inhibitors/therapeutic use , Animals , Clinical Trials as Topic , Combined Modality Therapy , Disease Models, Animal , Humans , Immunotherapy/trends , Multiple Myeloma/immunology
16.
Mol Cancer Ther ; 15(2): 241-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26719576

ABSTRACT

PARP1/2 are required for single-strand break repair, and their inhibition causes DNA replication fork collapse and double-strand break (DSB) formation. These DSBs are primarily repaired via homologous recombination (HR), a high-fidelity repair pathway. Should HR be deficient, DSBs may be repaired via error-prone nonhomologous end-joining mechanisms, or may persist, ultimately resulting in cell death. The combined disruption of PARP and HR activities thus produces synthetic lethality. Multiple myeloma cells are characterized by chromosomal instability and pervasive DNA damage, implicating aberrant DNA repair. Cyclin-dependent kinases (CDK), upstream modulators of HR, are dysregulated in multiple myeloma. Here, we show that a CDK inhibitor, dinaciclib, impairs HR repair and sensitizes multiple myeloma cells to the PARP1/2 inhibitor ABT-888. Dinaciclib abolishes ABT-888-induced BRCA1 and RAD51 foci and potentiates DNA damage, indicated by increased γH2AX foci. Dinaciclib treatment reduces expression of HR repair genes, including Rad51, and blocks BRCA1 phosphorylation, a modification required for HR repair, thus inhibiting HR repair of chromosome DSBs. Cotreatment with dinaciclib and ABT-888 in vitro resulted in synthetic lethality of multiple myeloma cells, but not normal CD19(+) B cells, and slowed growth of multiple myeloma xenografts in SCID mice almost two-fold. These findings support combining dinaciclib with PARP inhibitors for multiple myeloma therapy. Mol Cancer Ther; 15(2); 241-50. ©2015 AACR.


Subject(s)
Benzimidazoles/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cyclin-Dependent Kinases/antagonists & inhibitors , Homologous Recombination/drug effects , Multiple Myeloma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Pyridinium Compounds/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzimidazoles/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indolizines , Mice , Mice, SCID , Multiple Myeloma/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , Xenograft Model Antitumor Assays
17.
BMC Cancer ; 15: 864, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26545722

ABSTRACT

BACKGROUND: Human primary myeloma (MM) cells do not survive in culture; current in vitro and in vivo systems for growing these cells are limited to coculture with a specific bone marrow (BM) cell type or growth in an immunodeficient animal model. The purpose of the study is to establish an interactive healthy donor whole BM based culture system capable of maintaining prolonged survival of primary MM cells. This normal BM (NBM) coculture system is different from using autologous BM that is already affected by the disease. METHODS: Whole BM from healthy donors was cultured in medium supplemented with BM serum from MM patients for 7 days, followed by 7 days of coculture with CD138-selected primary MM cells or MM cell lines. MM cells in the coculture were quantified using flow cytometry or bioluminescence of luciferase-expressing MM cells. T-cell cytokine array and proteomics were performed to identify secreted factors. RESULTS: NBM is composed of adherent and nonadherent compartments containing typical hematopoietic and mesenchymal cells. MM cells, or a subset of MM cells, from all examined cases survived and grew in this system, regardless of the MM cells' molecular risk or subtype, and growth was comparable to coculture with individual stromal cell types. Adherent and nonadherent compartments supported MM growth, and this support required patient serum for optimal growth. Increased levels of MM growth factors IL-6 and IL-10 along with MM clinical markers B2M and LDHA were detected in supernatants from the NBM coculture than from the BM cultured alone. Levels of extracellular matrix factors (e.g., MMP1, HMCN1, COL3A1, ACAN) and immunomodulatory factors (e.g., IFI16, LILRB4, PTPN6, AZGP1) were changed in the coculture system. The NBM system protected MM cells from dexamethasone but not bortezomib, and effects of lenalidomide varied. CONCLUSIONS: The NBM system demonstrates the ability of primary MM plasma cells to interact with and to survive in coculture with healthy adult BM. This model is suitable for studying MM-microenvironment interactions, particularly at the early stage of engagement in new BM niches, and for characterizing MM cell subpopulations capable of long-term survival through secretion of extracellular matrix and immune-related factors.


Subject(s)
Bone Marrow Cells/metabolism , Cell Communication , Coculture Techniques , Multiple Myeloma/metabolism , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Communication/drug effects , Cell Line , Cell Survival , Cytokines/biosynthesis , Humans , Immunohistochemistry , Immunophenotyping , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/pathology , Osteoclasts/metabolism , Proteome , Proteomics/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment
18.
Haematologica ; 100(9): 1214-21, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26022710

ABSTRACT

Multiple myeloma is preceded by an asymptomatic phase, comprising monoclonal gammopathy of uncertain significance and smoldering myeloma. Compared to the former, smoldering myeloma has a higher and non-uniform rate of progression to clinical myeloma, reflecting a subset of patients with higher risk. We evaluated the gene expression profile of smoldering myeloma plasma cells among 105 patients enrolled in a prospective observational trial at our institution, with a view to identifying a high-risk signature. Baseline clinical, bone marrow, cytogenetic and radiologic data were evaluated for their potential to predict time to therapy for symptomatic myeloma. A gene signature derived from four genes, at an optimal binary cut-point of 9.28, identified 14 patients (13%) with a 2-year therapy risk of 85.7%. Conversely, a low four-gene score (< 9.28) combined with baseline monoclonal protein < 3 g/dL and albumin ≥ 3.5 g/dL identified 61 patients with low-risk smoldering myeloma with a 5.0% chance of progression at 2 years. The top 40 probe sets showed concordance with indices of chromosome instability. These data demonstrate high discriminatory power of a gene-based assay and suggest a role for dysregulation of mitotic checkpoints in the context of genomic instability as a hallmark of high-risk smoldering myeloma.


Subject(s)
Genes, Neoplasm , Genomic Instability , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Aged , Aged, 80 and over , Bone Marrow/pathology , Female , Humans , Male , Multiple Myeloma/pathology , Prospective Studies
19.
Cancer Biol Ther ; 16(6): 976-86, 2015.
Article in English | MEDLINE | ID: mdl-25996477

ABSTRACT

RAD51-mediated recombinational repair is elevated in multiple myeloma (MM) and predicts poor prognosis. RAD51 has been targeted to selectively sensitize and/or kill tumor cells. Here, we employed a peptide nucleic acid (PNA) to inhibit RAD51 expression in MM cells. We constructed a PNA complementary to a unique segment of the RAD51 gene promoter, spanning the transcription start site, and conjugated it to a nuclear localization signal (PKKKRKV) to enhance cellular uptake and nuclear delivery without transfection reagents. This synthetic construct, (PNArad51_nls), significantly reduced RAD51 transcripts in MM cells, and markedly reduced the number and intensity of de novo and melphalan-induced nuclear RAD51 foci, while increasing the level of melphalan-induced γH2AX foci. Melphalan alone markedly induced the expression of 5 other genes involved in homologous-recombination repair, yet suppression of RAD51 by PNArad51_nls was sufficient to synergize with melphalan, producing significant synthetic lethality of MM cells in vitro. In a SCID-rab mouse model mimicking the MM bone marrow microenvironment, treatment with PNArad51_nls ± melphalan significantly suppressed tumor growth after 2 weeks, whereas melphalan plus control PNArad4µ_nls was ineffectual. This study highlights the importance of RAD51 in myeloma growth and is the first to demonstrate that anti-RAD51 PNA can potentiate conventional MM chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Melphalan/pharmacology , Multiple Myeloma/metabolism , Peptide Nucleic Acids/pharmacology , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Animals , Cell Line, Tumor , DNA Damage/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Rad51 Recombinase/genetics , S Phase/drug effects , Transcription, Genetic , Xenograft Model Antitumor Assays
20.
J Immunother ; 38(1): 24-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25415285

ABSTRACT

Highly activated/expanded natural killer (NK) cells can be generated by stimulation with the human leukocyte antigen-deficient cell line K562, genetically modified to express 41BB-ligand and membrane-bound interleukin (IL)15. We tested the safety, persistence, and activity of expanded NK cells generated from myeloma patients (auto-NK) or haploidentical family donors (allo-NK) in heavily pretreated patients with high-risk relapsing myeloma. The preparative regimen comprised bortezomib only or bortezomib and immunosuppression with cyclophosphamide, dexamethasone, and fludarabine. NK cells were shipped overnight either cryopreserved or fresh. In 8 patients, up to 1×108 NK cells/kg were infused on day 0 and followed by daily administrations of IL2. Significant in vivo expansion was observed only in the 5 patients receiving fresh products, peaking at or near day 7, with the highest NK-cell counts in 2 subjects who received cells produced in a high concentration of IL2 (500 U/mL). Seven days after infusion, donor NK cells comprised >90% of circulating leukocytes in fresh allo-NK cell recipients, and cytolytic activity against allogeneic myeloma targets was retained in vitro. Among the 7 evaluable patients, there were no serious adverse events that could be related to NK-cell infusion. One patient had a partial response and in another the tempo of disease progression decreased; neither patient required further therapy for 6 months. In the 5 remaining patients, disease progression was not affected by NK-cell infusion. In conclusion, infusion of large numbers of expanded NK cells was feasible and safe; infusing fresh cells was critical to their expansion in vivo.


Subject(s)
Cell Proliferation , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , Aged , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Killer Cells, Natural/immunology , Male , Middle Aged , Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/therapy , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...