Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
J Control Release ; 296: 232-240, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30682443

ABSTRACT

Glucocorticoids are the cornerstone in the clinic for treatment of hematological malignancies, including multiple myeloma. Nevertheless, poor pharmacokinetic properties of glucocorticoids require high and frequent dosing with the off-target adverse effects defining the maximum dose. Recently, nanomedicine formulations of glucocorticoids have been developed that improve the pharmacokinetic profile, limit adverse effects and improve solid tumor accumulation. Multiple myeloma is a hematological malignancy characterized by uncontrolled growth of plasma cells. These tumors initiate increased angiogenesis and microvessel density in the bone marrow, which might be exploited using nanomedicines, such as liposomes. Nano-sized particles can accumulate as a result of the increased vascular leakiness at the bone marrow tumor lesions. Pre-clinical screening of novel anti-myeloma therapeutics in vivo requires a suitable animal model that represents key features of the disease. In this study, we show that fluorescently labeled long circulating liposomes were found in plasma up to 24 h after injection in an advanced human-mouse hybrid model of multiple myeloma. Besides the organs involved in clearance, liposomes were also found to accumulate in tumor bearing human-bone scaffolds. The therapeutic efficacy of liposomal dexamethasone phosphate was evaluated in this model showing strong tumor growth inhibition while free drug being ineffective at an equivalent dose (4 mg/kg) regimen. The liposomal formulation slightly reduced total body weight of myeloma-bearing mice during the course of treatment, which appeared reversible when treatment was stopped. Liposomal dexamethasone could be further developed as monotherapy or could fit in with existing therapy regimens to improve therapeutic outcomes for multiple myeloma.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Multiple Myeloma/drug therapy , Animals , Body Weight/drug effects , Bone and Bones/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Liposomes , Mice, Knockout , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tissue Distribution , Tumor Burden/drug effects
4.
Mol Ther ; 25(8): 1946-1958, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28506593

ABSTRACT

Chimeric antigen receptors (CARs) can effectively redirect cytotoxic T cells toward highly expressed surface antigens on tumor cells. The low expression of several tumor-associated antigens (TAAs) on normal tissues, however, hinders their safe targeting by CAR T cells due to on-target/off-tumor effects. Using the multiple myeloma (MM)-associated CD38 antigen as a model system, here, we present a rational approach for effective and tumor-selective targeting of such TAAs. Using "light-chain exchange" technology, we combined the heavy chains of two high-affinity CD38 antibodies with 176 germline light chains and generated ∼124 new antibodies with 10- to >1,000-fold lower affinities to CD38. After categorizing them into three distinct affinity classes, we incorporated the single-chain variable fragments of eight antibodies from each class into new CARs. T cells carrying these CD38-CARs were extensively evaluated for their on-tumor/off-tumor cytotoxicity as well as CD38-dependent proliferation and cytokine production. We identified CD38-CAR T cells of ∼1,000- fold reduced affinity, which optimally proliferated, produced Th1-like cytokines, and effectively lysed CD382+ MM cells, but spared CD38+ healthy hematopoietic cells in vitro and in vivo. Thus, this systematic approach is highly suitable for the generation of optimal CARs for effective and selective targeting of TAAs.


Subject(s)
ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase 1/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , ADP-ribosyl Cyclase 1/metabolism , Animals , Antibody Affinity/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Mice , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Protein Binding/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Xenograft Model Antitumor Assays
5.
Blood ; 128(25): 2949-2959, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27733356

ABSTRACT

To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Stem Cell Niche , Tissue Scaffolds/chemistry , Xenograft Model Antitumor Assays , Animals , Cell Self Renewal , Cell Separation , Clone Cells , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/cytology , Mice , Phenotype , Stromal Cells/pathology
7.
Haematologica ; 101(5): 616-25, 2016 05.
Article in English | MEDLINE | ID: mdl-26858358

ABSTRACT

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we transduced T cells from healthy donors and multiple myeloma patients. We then evaluated the preclinical efficacy and safety of the transduced T cells. Irrespective of the donor and antibody sequence, CD38-chimeric antigen receptor-transduced T cells proliferated, produced inflammatory cytokines and effectively lysed malignant cell lines and primary malignant cells from patients with acute myeloid leukemia and multi-drug resistant multiple myeloma in a cell-dose, and CD38-dependent manner, despite becoming CD38-negative during culture. CD38-chimeric antigen receptor-transduced T cells also displayed significant anti-tumor effects in a xenotransplant model, in which multiple myeloma tumors were grown in a human bone marrow-like microenvironment. CD38-chimeric antigen receptor-transduced T cells also appeared to lyse the CD38(+) fractions of CD34(+) hematopoietic progenitor cells, monocytes, natural killer cells, and to a lesser extent T and B cells but did not inhibit the outgrowth of progenitor cells into various myeloid lineages and, furthermore, were effectively controllable with a caspase-9-based suicide gene. These results signify the potential importance of CD38-chimeric antigen receptor-transduced T cells as therapeutic tools for CD38(+) malignancies and warrant further efforts to diminish the undesired effects of this immunotherapy using appropriate strategies.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Immunotherapy , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Animals , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Flow Cytometry , Gene Expression , Gene Transfer Techniques , Genes, Transgenic, Suicide , Hematopoietic Stem Cells/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Burden/genetics , Tumor Burden/immunology
8.
Cell Rep ; 14(2): 332-46, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26748712

ABSTRACT

Polycomb proteins are classical regulators of stem cell self-renewal and cell lineage commitment and are frequently deregulated in cancer. Here, we find that the non-canonical PRC1.1 complex, as identified by mass-spectrometry-based proteomics, is critically important for human leukemic stem cells. Downmodulation of PRC1.1 complex members, like the DNA-binding subunit KDM2B, strongly reduces cell proliferation in vitro and delays or even abrogates leukemogenesis in vivo in humanized xenograft models. PRC1.1 components are significantly overexpressed in primary AML CD34(+) cells. Besides a set of genes that is targeted by PRC1 and PRC2, ChIP-seq studies show that PRC1.1 also binds a distinct set of genes that are devoid of H3K27me3, suggesting a gene-regulatory role independent of PRC2. This set encompasses genes involved in metabolism, which have transcriptionally active chromatin profiles. These data indicate that PRC1.1 controls specific genes involved in unique cell biological processes required for leukemic cell viability.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Cell Differentiation , Cell Proliferation , Humans
9.
J Tissue Eng Regen Med ; 10(3): 233-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-23255230

ABSTRACT

The combination of scaffolds and mesenchymal stromal cells (MSCs) is a promising approach in bone tissue engineering (BTE). Knowledge on the survival, outgrowth and bone-forming capacity of MSCs in vivo is limited. Bioluminescence imaging (BLI), histomorphometry and immunohistochemistry were combined to study the fate of gene-marked goat and human MSCs (gMSCs, hMSCs) on scaffolds with different osteoinductive properties. Luciferase-GFP-labelled MSCs were seeded on hydroxyapatite (HA) or ß-tricalcium phosphate (TCP), cultured for 7 days in vitro in osteogenic medium, implanted subcutaneously in immunodeficient mice and monitored with BLI for 6 weeks. The constructs were retrieved and processed for histomorphometry and detection of luciferase-positive cells (LPCs). For gMSCs, BLI revealed doubling of signal after 1 week, declining to 60% of input after 3 weeks and remaining constant until week 6. hMSCs showed a constant decrease of BLI signal to 25% of input, indicating no further expansion. Bone formation of gMSCs was two-fold higher on TCP than HA. hMSCs and gMSCs control samples produced equal amounts of bone on TCP. Upon transduction, there was a four-fold reduction in bone formation compared with untransduced hMSCs, and no bone was formed on HA. LPCs were detected at day 14, but were much less frequent at day 42. Striking differences were observed in spatial distribution. MSCs in TCP were found to be aligned and interconnected on the surface but were scattered in an unstructured fashion in HA. In conclusion, the spatial distribution of MSCs on the scaffold is critical for cell-scaffold-based BTE.


Subject(s)
Calcium Phosphates/pharmacology , Durapatite/pharmacology , Mesenchymal Stem Cells/cytology , Animals , Cell Survival/drug effects , DNA/metabolism , Goats , Humans , Immunohistochemistry , Luciferases/metabolism , Luminescent Measurements , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Osteogenesis/drug effects , Tissue Scaffolds/chemistry
10.
Cancer Immunol Immunother ; 64(8): 951-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25920521

ABSTRACT

Immunotherapy with allogeneic natural killer (NK) cells offers therapeutic perspectives for multiple myeloma patients. Here, we aimed to refine NK cell therapy by evaluation of the relevance of HLA-class I and HLA-E for NK anti-myeloma reactivity. We show that HLA-class I was strongly expressed on the surface of patient-derived myeloma cells and on myeloma cell lines. HLA-E was highly expressed by primary myeloma cells but only marginally by cell lines. HLA-E(low) expression on U266 cells observed in vitro was strongly upregulated after in vivo (bone marrow) growth in RAG-2(-/-) γc(-/-) mice, suggesting that in vitro HLA-E levels poorly predict the in vivo situation. Concurrent analysis of inhibitory receptors (KIR2DL1, KIR2DL2/3, KIR3DL1 and NKG2A) and NK cell degranulation upon co-culture with myeloma cells revealed that KIR-ligand-mismatched NK cells degranulate more than matched subsets and that HLA-E abrogates degranulation of NKG2A+ subsets. Inhibition by HLA-class I and HLA-E was also observed with IL-2-activated NK cells and at low oxygen levels (0.6 %) mimicking hypoxic bone marrow niches where myeloma cells preferentially reside. Our study demonstrates that NKG2A-negative, KIR-ligand-mismatched NK cells are the most potent subset for clinical application. We envision that infusion of high numbers of this subclass will enhance clinical efficacy.


Subject(s)
Cell Separation/methods , Histocompatibility Antigens Class I/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , NK Cell Lectin-Like Receptor Subfamily C/immunology , Animals , Cell Degranulation , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , Interleukin-2/immunology , Mice , Mice, Knockout , Multiple Myeloma/immunology , Neoplasm Transplantation , Oxygen/metabolism , HLA-E Antigens
11.
MAbs ; 7(2): 311-21, 2015.
Article in English | MEDLINE | ID: mdl-25760767

ABSTRACT

Daratumumab (DARA) is a human CD38-specific IgG1 antibody that is in clinical development for the treatment of multiple myeloma (MM). The potential for IgG1 antibodies to induce macrophage-mediated phagocytosis, in combination with the known presence of macrophages in the tumor microenvironment in MM and other hematological tumors, led us to investigate the contribution of antibody-dependent, macrophage-mediated phagocytosis to DARA's mechanism of action. Live cell imaging revealed that DARA efficiently induced macrophage-mediated phagocytosis, in which individual macrophages rapidly and sequentially engulfed multiple tumor cells. DARA-dependent phagocytosis by mouse and human macrophages was also observed in an in vitro flow cytometry assay, using a range of MM and Burkitt's lymphoma cell lines. Phagocytosis contributed to DARA's anti-tumor activity in vivo, in both a subcutaneous and an intravenous leukemic xenograft mouse model. Finally, DARA was shown to induce macrophage-mediated phagocytosis of MM cells isolated from 11 of 12 MM patients that showed variable levels of CD38 expression. In summary, we demonstrate that phagocytosis is a fast, potent and clinically relevant mechanism of action that may contribute to the therapeutic activity of DARA in multiple myeloma and potentially other hematological tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Cytophagocytosis/drug effects , Lymphoma/drug therapy , Macrophages/immunology , Multiple Myeloma/drug therapy , Animals , Cell Line, Tumor , Humans , Lymphoma/immunology , Lymphoma/pathology , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 21(12): 2802-10, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25398450

ABSTRACT

PURPOSE: Novel therapeutic agents have significantly improved the survival of patients with multiple myeloma. Nonetheless, the prognosis of patients with multiple myeloma who become refractory to the novel agents lenalidomide and bortezomib is very poor, indicating the urgent need for new therapeutic options for these patients. The human CD38 monoclonal antibody daratumumab is being evaluated as a novel therapy for multiple myeloma. Prompted with the encouraging results of ongoing clinical phase I/II trials, we now addressed the potential value of daratumumab alone or in combination with lenalidomide or bortezomib for the treatment of lenalidomide- and bortezomib-refractory patients. EXPERIMENTAL DESIGN: In ex vivo assays, mainly evaluating antibody-dependent cell-mediated cytotoxicity, and in an in vivo xenograft mouse model, we evaluated daratumumab alone or in combination with lenalidomide or bortezomib as a potential therapy for lenalidomide- and bortezomib-refractory multiple myeloma patients. RESULTS: Daratumumab induced significant lysis of lenalidomide/bortezomib-resistant multiple myeloma cell lines and of primary multiple myeloma cells in the bone marrow mononuclear cells derived from lenalidomide- and/or bortezomib-refractory patients. In these assays, lenalidomide but not bortezomib, synergistically enhanced daratumumab-mediated multiple myeloma lysis through activation of natural killer cells. Finally, in an in vivo xenograft model, only the combination of daratumumab with lenalidomide effectively reduced the tumorigenic growth of primary multiple myeloma cells from a lenalidomide- and bortezomib-refractory patient. CONCLUSIONS: Our results provide the first preclinical evidence for the benefit of daratumumab plus lenalidomide combination for lenalidomide- and bortezomib-refractory patients.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Bortezomib/pharmacology , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Thalidomide/analogs & derivatives , Adult , Aged , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Bortezomib/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , Immunotherapy , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , Molecular Targeted Therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Thalidomide/administration & dosage , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
13.
Clin Cancer Res ; 20(6): 1542-54, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24486586

ABSTRACT

PURPOSE: MLN9708 (ixazomib citrate), which hydrolyzes to pharmacologically active MLN2238 (ixazomib), is a next-generation proteasome inhibitor with demonstrated preclinical and clinical antimyeloma activity, but yet with an unknown effect on myeloma bone disease. Here, we investigated its bone anabolic and antiresorptive effects in the myeloma setting and in comparison with bortezomib in preclinical models. EXPERIMENTAL DESIGN: The in vitro effect of MLN2238 was tested on osteoclasts and osteoclast precursors from healthy donors and patients with myeloma, and on osteoprogenitors derived from bone marrow mesenchymal stem cells also from both origins. We used an in vivo model of bone marrow-disseminated human myeloma to evaluate MLN2238 antimyeloma and bone activities. RESULTS: Clinically achievable concentrations of MLN2238 markedly inhibited in vitro osteoclastogenesis and osteoclast resorption; these effects involved blockade of RANKL (receptor activator of NF-κB ligand)-induced NF-κB activation, F-actin ring disruption, and diminished expression of αVß3 integrin. A similar range of MLN2238 concentrations promoted in vitro osteoblastogenesis and osteoblast activity (even in osteoprogenitors from patients with myeloma), partly mediated by activation of TCF/ß-catenin signaling and upregulation of the IRE1 component of the unfolded protein response. In a mouse model of bone marrow-disseminated human multiple myeloma, orally administered MLN2238 was equally effective as bortezomib to control tumor burden and also provided a marked benefit in associated bone disease (sustained by both bone anabolic and anticatabolic activities). CONCLUSION: Given favorable data on pharmacologic properties and emerging clinical safety profile of MLN9708, it is conceivable that this proteasome inhibitor may achieve bone beneficial effects in addition to its antimyeloma activity in patients with myeloma.


Subject(s)
Boron Compounds/pharmacology , Glycine/analogs & derivatives , Multiple Myeloma/metabolism , Osteoclasts/drug effects , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Bone Resorption/metabolism , Cell Line , Disease Models, Animal , Glycine/pharmacology , Humans , Immunoblotting , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Osteoclasts/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
14.
Stem Cells Dev ; 22(7): 1086-96, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23145933

ABSTRACT

Bone marrow (BM) stromal cells (MSCs), also known as mesenchymal stem cells, display a high degree of heterogeneity. To shed light on the causes of this heterogeneity, MSCs were collected from either human BM (n=5) or adipose tissue (AT) (n=5), and expanded using 2 different culture methods: one based on fetal calf serum, and one based on human platelet lysate. After initial expansion, MSCs were frozen, and the vials were transported to 3 different laboratories and grown for 1 passage using the same brand of culture plastic, medium, and supplements. Subsequently, the cells were harvested and assayed for their gene expression profile using the Affymetrix exon microarray platform. Based on gene expression profiles, the most discriminative feature was the anatomical harvesting site, followed by culture methodology. Remarkably, genes in the WNT pathway were expressed at higher levels in BM-derived MSCs than in AT-derived MSCs. Although differences were found between laboratories, cell culture location only slightly affects heterogeneity. Furthermore, individual donors contributed marginally to the observed differences in transcriptomes. Finally, BM-derived MSCs displayed the highest level of similarity, irrespective their culture conditions, when compared to AT-derived cells.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/cytology , Cell Culture Techniques , Gene Expression Profiling , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Adipose Tissue/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Humans , Mesenchymal Stem Cells/metabolism , Wnt Signaling Pathway/genetics
15.
Nat Cell Biol ; 14(10): 1099-1104, 2012 10.
Article in English | MEDLINE | ID: mdl-23000963

ABSTRACT

Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Lineage , Cells, Cultured , Gene Knock-In Techniques , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Organoids/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Wnt3A Protein/pharmacology
16.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653974

ABSTRACT

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Subject(s)
Hematopoietic Stem Cells/cytology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Stem Cell Niche/immunology , Transplantation Chimera/immunology , Tumor Microenvironment/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear Ossicles/cytology , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Mutant Strains , Neoplasm Transplantation , Osteolysis/immunology , Tissue Scaffolds , Transplantation, Heterologous
17.
Haematologica ; 96(11): 1653-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21828122

ABSTRACT

BACKGROUND: Multiple myeloma is a hematologic malignancy characterized by a clonal expansion of malignant plasma cells in the bone marrow, which is accompanied by the development of osteolytic lesions and/or diffuse osteopenia. The intricate bi-directional interaction with the bone marrow microenvironment plays a critical role in sustaining the growth and survival of myeloma cells during tumor progression. Identification and functional analysis of the (adhesion) molecules involved in this interaction will provide important insights into the pathogenesis of multiple myeloma. DESIGN AND METHODS: Multiple myeloma cell lines and patients' samples were analyzed for expression of the adhesion molecule N-cadherin by immunoblotting, flow cytometry, immunofluorescence microscopy, immunohistochemistry and expression microarray. In addition, by means of blocking antibodies and inducible RNA interference we studied the functional consequence of N-cadherin expression for the myeloma cells, by analysis of adhesion, migration and growth, and for the bone marrow microenvironment, by analysis of osteogenic differentiation. RESULTS: The malignant plasma cells in approximately half of the multiple myeloma patients, belonging to specific genetic subgroups, aberrantly expressed the homophilic adhesion molecule N-cad-herin. N-cadherin-mediated cell-substrate or homotypic cell-cell adhesion did not contribute to myeloma cell growth in vitro. However, N-cadherin directly mediated the bone marrow localization/retention of myeloma cells in vivo, and facilitated a close interaction between myeloma cells and N-cadherin-positive osteoblasts. Furthermore, this N-cadherin-mediated interaction contributed to the ability of myeloma cells to inhibit osteoblastogenesis. CONCLUSIONS: Taken together, our data show that myeloma cells frequently display aberrant expression of N-cadherin and that N-cadherin mediates the interaction of myeloma cells with the bone marrow microenvironment, in particular the osteoblasts. This N-cadherin-mediated interaction inhibits osteoblast differentiation and may play an important role in the pathogenesis of myeloma bone disease.


Subject(s)
Cadherins/metabolism , Cell Communication , Cell Differentiation , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Osteoblasts/metabolism , Tumor Microenvironment , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cadherins/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Osteoblasts/pathology
18.
Blood ; 115(3): 601-4, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19965677

ABSTRACT

Expression of the heparan sulfate proteoglycan syndecan-1 is a hallmark of both normal and multiple myeloma (MM) plasma cells. Syndecan-1 could affect plasma cell fate by strengthening integrin-mediated adhesion via its core protein and/or by accommodating and presenting soluble factors via its HS side chains. Here, we show that inducible RNAi-mediated knockdown of syndecan-1 in human MM cells leads to reduced growth rates and a strong increase of apoptosis. Importantly, knockdown of EXT1, a copolymerase critical for HS chain biosynthesis, had similar effects. Using an innovative myeloma xenotransplantation model in Rag-2(-/-)gamma(c)(-/-) mice, we demonstrate that induction of EXT1 knockdown in vivo dramatically suppresses the growth of bone marrow localized myeloma. Our findings provide direct evidence that the HS chains of syndecan-1 are crucial for the growth and survival of MM cells within the bone marrow environment, and indicate the HS biosynthesis machinery as a potential treatment target in MM.


Subject(s)
Cell Proliferation/drug effects , Heparitin Sulfate/physiology , Multiple Myeloma/pathology , N-Acetylglucosaminyltransferases/genetics , RNA, Small Interfering/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Doxycycline/administration & dosage , Drug Delivery Systems , Gene Targeting , Heparitin Sulfate/metabolism , Humans , Immunoglobulin gamma-Chains/genetics , Mice , Mice, Knockout , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/physiology , Syndecan-1/genetics , Syndecan-1/metabolism , Xenograft Model Antitumor Assays
19.
Tissue Eng Part A ; 15(12): 3741-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19519274

ABSTRACT

In tissue engineering, strategies are being developed to repair large bone defects by combining biomaterials and bone marrow-derived multipotent mesenchymal stromal cells (MSCs). For expansion of MSCs under good manufacturing practice conditions, human platelet lysate (PL) can serve as substitute for fetal bovine serum (FBS) in culture media. We compared the in vivo bone-forming capacity of passage 3 MSCs cultured with either PL or FBS for nine different human donors. We also tested the growth kinetics, antigen expression profile, and the multilineage differentiation capacity in vitro of these MSCs. The in vivo bone-forming capacity was determined by seeding culture-expanded MSCs onto biphasic calcium phosphate scaffolds. Hybrid constructs were implanted subcutaneously in nude mice, retrieved after 6 weeks, and analyzed using histomorphometry. PL-supplemented cultures resulted in significantly larger colonies, shorter culture time period, and higher population doublings between P1 and P3 compared to FBS-containing cultures. No differences were observed in antigen expression profiles or differentiation capacities into the osteoblastic, chondrogenic, and adipogenic lineages, qualitatively. In vivo bone formation with PL-supplemented cultures of MSCs was demonstrated in 9/9 donors versus 6/9 for FBS-supplemented cultures. These results warrant the use of PL for ex vivo expansion of human MSCs for bone tissue engineering applications.


Subject(s)
Blood Platelets/cytology , Cell Extracts/pharmacology , Mesoderm/cytology , Osteogenesis/drug effects , Serum/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Animals , Antigens/immunology , Blood Substitutes/pharmacology , Cattle , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Humans , Immunophenotyping , Kinetics , Mice , Stromal Cells/cytology
20.
J Orthop Res ; 26(7): 901-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18271011

ABSTRACT

Mesenchymal stem cells (MSCs) represent a powerful tool for applications in regenerative medicine. In this study, we used in vivo bioluminescence imaging to noninvasively investigate the fate and the contribution to bone formation of adult MSCs in tissue engineered constructs. Goat MSCs expressing GFP-luciferase were seeded on ceramic scaffolds and implanted subcutaneously in immune-deficient mice. The constructs were monitored weekly with bioluminescence imaging and were retrieved after 7 weeks to quantify bone formation by histomorphometry. With increasing amounts of seeded MSCs (from 0 to 1 x 10(6) MSC/scaffold), a cell-dose related increase in bioluminescence was observed at all time points, correlating with increased bone formation at 7 weeks. To investigate the relevance of MSC proliferation to bone deposition, cell-seeded scaffolds were irradiated. The irradiated cells were functional with respect to oxygen consumption but no increase in bioluminescence was observed in vivo, and only minimal bone was produced. Proliferating MSCs are likely required for initiation of bone formation in tissue engineered constructs in vivo. Bioluminescence is a useful tool to monitor cellular responses and predict bone formation in vivo.


Subject(s)
Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Tissue Engineering , Tissue Scaffolds , Animals , Cells, Cultured , Goats , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/radiation effects , Mice , Osteoclasts/cytology , Osteogenesis/radiation effects , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...