1
|
Knop S, Szarejko M, Grząśko N, Bringhen S, Trautmann‐Grill K, Jurczyszyn A, Vacca A, Khandanpour C, Gamberi B, Pour L, Iversen KF, Stumpp MT, Suter C, Dawson KM, Zitt C, Legenne P, Stavropoulou V, Fey MF, Leupin N, Goldschmidt H. A phase 1b/2 study evaluating efficacy and safety of MP0250, a designed ankyrin repeat protein (DARPin) simultaneously targeting vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF), in combination with bortezomib and dexamethasone, in patients with relapsed or refractory multiple myeloma. EJHAEM 2024; 5:940-950. [PMID: 39415900 PMCID: PMC11474421 DOI: 10.1002/jha2.968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 10/19/2024]
Abstract
MP0250 is a designed ankyrin repeat protein that specifically inhibits both vascular endothelial growth factor A (VEGF-A) and hepatocyte growth factor (HGF), aiming at potentiating cancer therapy by disrupting the tumour microenvironment. Encouraging results from a phase 1 trial of MP0250 in patients with solid tumours prompted further investigation in multiple myeloma (MM) as both MP0250 targets are reported to be drivers of MM pathogenesis. In this open-label, single-arm phase 1b/2 study (NCT03136653) in patients with proteasome inhibitor- and/or immunomodulatory drug-relapsed or refractory MM, MP0250 was administered every 3 weeks with standard bortezomib/dexamethasone regimen. Thirty-three patients received at least one dose of MP0250. The most frequent treatment-related adverse events were arterial hypertension (58.1%), thrombocytopenia (32.3%), proteinuria (29.0%) and peripheral oedema (19.4%). Of the 28 patients evaluable for response (median age: 60 [range 44-75]), nine achieved at least partial response, corresponding to an overall response rate of 32.1% (95% confidence interval [CI]: 17.9%, 50.7%), with a median duration of response of 8 months (95% CI 5-NR). An additional three patients achieved minimal response and nine stable diseases as the best overall response. Overall median progression-free survival was 4.2 months (95% CI 1.9-7.1). These findings are in line with the results of recent trials testing new agents on comparable patient cohorts and provide initial evidence of clinical benefit for patients with refractory/relapsed MM treated with MP0250 in combination with bortezomib/dexamethasone. Further clinical evaluation in the emerging MM treatment landscape would be required to confirm the clinical potential of MP0250.
Collapse
Affiliation(s)
- Stefan Knop
- Universitätsklinikum WürzburgWürzburgGermany
| | | | - Norbert Grząśko
- Department of Experimental HematooncologyMedical University of Lublin and Centrum Onkologii Ziemi LubelskiejLublinPoland
| | - Sara Bringhen
- SSD Clinical Trial in Oncoematologia e Myeloma, Dipartimento di OncologiaAzienda Ospedaliera‐Universitaria Città della Salute e della Scienza di TorinoTorinoItaly
| | | | - Artur Jurczyszyn
- Plasma Cell Dyscrasias Center, Department of HematologyJagiellonian University Medical CollegeKrakowPoland
| | - Angelo Vacca
- Department of Precision and Regenerative Medicine and Ionian Area Unit of Medicina Interna “Guido Baccelli”University of Bari Aldo MoroAzienda PoliclinicoBariItaly
| | - Cyrus Khandanpour
- Universitätsklinikum Münster, Münster, Germany and University Hospital Schleswig‐Holstein Campus LübeckUniversity Cancer Center Schleswig‐Holstein, and University of LübeckLübeckGermany
| | | | | | | | | | - Cosima Suter
- Molecular Partners AGZurich‐SchlierenSwitzerland
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Marcoux P, Imeri J, Desterke C, Latsis T, Chaker D, Hugues P, Griscelli AB, Turhan AG. Impact of the overexpression of the tyrosine kinase receptor RET in the hematopoietic potential of induced pluripotent stem cells (iPSCs). Cytotherapy 2024; 26:63-72. [PMID: 37921725 DOI: 10.1016/j.jcyt.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Previous studies have suggested that the tyrosine kinase receptor RET plays a significant role in the hematopoietic potential in mice and could also be used to expand cord-blood derived hematopoietic stem cells (HSCs). The role of RET in human iPSC-derived hematopoiesis has not been tested so far. METHODS To test the implication of RET on the hematopoietic potential of iPSCs, we activated its pathway with the lentiviral overexpression of RETWT or RETC634Y mutation in normal iPSCs. An iPSC derived from a patient harboring the RETC634Y mutation (iRETC634Y) and its CRISPR-corrected isogenic control iPSC (iRETCTRL) were also used. The hematopoietic potential was tested using 2D cultures and evaluated regarding the phenotype and the clonogenic potential of generated cells. RESULTS Hematopoietic differentiation from iPSCs with RET overexpression (WT or C634Y) led to a significant reduction in the number and in the clonogenic potential of primitive hematopoietic cells (CD34+/CD38-/CD49f+) as compared to control iPSCs. Similarly, the hematopoietic potential of iRETC634Y was reduced as compared to iRETCTRL. Transcriptomic analyses revealed a specific activated expression profile for iRETC634Y compared to its control with evidence of overexpression of genes which are part of the MAPK network with negative hematopoietic regulator activities. CONCLUSION RET activation in iPSCs is associated with an inhibitory activity in iPSC-derived hematopoiesis, potentially related to MAPK activation.
Collapse
Affiliation(s)
- Paul Marcoux
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Jusuf Imeri
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Christophe Desterke
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | | | - Diana Chaker
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France
| | - Patricia Hugues
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Annelise Bennaceur Griscelli
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France; Department of Hematology, APHP Paris Saclay, Hôpital Bicetre, Le Kremlin Bicetre France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France; Department of Hematology, APHP Paris Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Ali G Turhan
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France; Department of Hematology, APHP Paris Saclay, Hôpital Bicetre, Le Kremlin Bicetre France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France; Department of Hematology, APHP Paris Saclay, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
3
|
Sharma NS, Choudhary B. Good Cop, Bad Cop: Profiling the Immune Landscape in Multiple Myeloma. Biomolecules 2023; 13:1629. [PMID: 38002311 PMCID: PMC10669790 DOI: 10.3390/biom13111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple myeloma (MM) is a dyscrasia of plasma cells (PCs) characterized by abnormal immunoglobulin (Ig) production. The disease remains incurable due to a multitude of mutations and structural abnormalities in MM cells, coupled with a favorable microenvironment and immune suppression that eventually contribute to the development of drug resistance. The bone marrow microenvironment (BMME) is composed of a cellular component comprising stromal cells, endothelial cells, osteoclasts, osteoblasts, and immune cells, and a non-cellular component made of the extracellular matrix (ECM) and the liquid milieu, which contains cytokines, growth factors, and chemokines. The bone marrow stromal cells (BMSCs) are involved in the adhesion of MM cells, promote the growth, proliferation, invasion, and drug resistance of MM cells, and are also crucial in angiogenesis and the formation of lytic bone lesions. Classical immunophenotyping in combination with advanced immune profiling using single-cell sequencing technologies has enabled immune cell-specific gene expression analysis in MM to further elucidate the roles of specific immune cell fractions from peripheral blood and bone marrow (BM) in myelomagenesis and progression, immune evasion and exhaustion mechanisms, and development of drug resistance and relapse. The review describes the role of BMME components in MM development and ongoing clinical trials using immunotherapeutic approaches.
Collapse
Affiliation(s)
- Niyati Seshagiri Sharma
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Electronic City, Bengaluru 560100, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Electronic City, Bengaluru 560100, India
| |
Collapse
|
4
|
Li Z, Liu Y, Huang Y, Tan Q, Mei H, Zhu G, Liu K, Yang G. Circ_0000888 regulates osteogenic differentiation of periosteal mesenchymal stem cells in congenital pseudarthrosis of the tibia. iScience 2023; 26:107923. [PMID: 37810257 PMCID: PMC10551655 DOI: 10.1016/j.isci.2023.107923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/02/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a refractory condition characterized by the decreased osteogenic ability in tibial pseudarthrosis repair. Periosteal mesenchymal stem cells (PMSCs) are multipotent cells involved in bone formation regulation. However, the mechanisms underlying its role in CPT remain unclear. In this study, we observed downregulation of circ_0000888 and pleiotrophin (PTN), as well as upregulation of miR-338-3p in CPT derived PMSCs (CPT-dPMSCs). Our results demonstrated that circ_0000888 and PTN likely enhanced the viability, proliferation, and osteogenic ability of PMSCs, while miR-338-3p had the opposite effect. Further analysis confirmed the regulatory relationship circ_0000888 suppressed the activity of miR-338-3p and upregulated the expression of its downstream target PTN by binding to miR-338-3p, consequently promoting the viability and osteogenic differentiation ability of CPT-dPMSCs. Our findings unveil an unexpected link between circ_0000888/miR-338-3p/PTN in promoting osteogenic ability and indicate the potential pathogenic mechanisms of CPT.
Collapse
Affiliation(s)
- Zhuoyang Li
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
- Department of Orthopedics, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yaoxi Liu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Yiyong Huang
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Qian Tan
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Haibo Mei
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Guanghui Zhu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Kun Liu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ge Yang
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| |
Collapse
|
5
|
Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh SCA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Res 2023; 11:15. [PMID: 36918531 PMCID: PMC10014945 DOI: 10.1038/s41413-023-00249-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Prior research establishing that bone interacts in coordination with the bone marrow microenvironment (BMME) to regulate hematopoietic homeostasis was largely based on analyses of individual bone-associated cell populations. Recent advances in intravital imaging has suggested that the expansion of hematopoietic stem cells (HSCs) and acute myeloid leukemia cells is restricted to bone marrow microdomains during a distinct stage of bone remodeling. These findings indicate that dynamic bone remodeling likely imposes additional heterogeneity within the BMME to yield differential clonal responses. A holistic understanding of the role of bone remodeling in regulating the stem cell niche and how these interactions are altered in age-related hematological malignancies will be critical to the development of novel interventions. To advance this understanding, herein, we provide a synopsis of the cellular and molecular constituents that participate in bone turnover and their known connections to the hematopoietic compartment. Specifically, we elaborate on the coupling between bone remodeling and the BMME in homeostasis and age-related hematological malignancies and after treatment with bone-targeting approaches. We then discuss unresolved questions and ambiguities that remain in the field.
Collapse
Affiliation(s)
- Hengwei Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jane L Liesveld
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Endocrinology/Metabolism, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea C Lipe
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy/Immunology/Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
6
|
Lungu O, Toscani D, Burroughs-Garcia J, Giuliani N. The Metabolic Features of Osteoblasts: Implications for Multiple Myeloma (MM) Bone Disease. Int J Mol Sci 2023; 24:ijms24054893. [PMID: 36902326 PMCID: PMC10003241 DOI: 10.3390/ijms24054893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The study of osteoblast (OB) metabolism has recently received increased attention due to the considerable amount of energy used during the bone remodeling process. In addition to glucose, the main nutrient for the osteoblast lineages, recent data highlight the importance of amino acid and fatty acid metabolism in providing the fuel necessary for the proper functioning of OBs. Among the amino acids, it has been reported that OBs are largely dependent on glutamine (Gln) for their differentiation and activity. In this review, we describe the main metabolic pathways governing OBs' fate and functions, both in physiological and pathological malignant conditions. In particular, we focus on multiple myeloma (MM) bone disease, which is characterized by a severe imbalance in OB differentiation due to the presence of malignant plasma cells into the bone microenvironment. Here, we describe the most important metabolic alterations involved in the inhibition of OB formation and activity in MM patients.
Collapse
Affiliation(s)
- Oxana Lungu
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Hematology, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy
- Correspondence:
| |
Collapse
|
7
|
Molecular Features of the Mesenchymal and Osteoblastic Cells in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232415448. [PMID: 36555090 PMCID: PMC9779562 DOI: 10.3390/ijms232415448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a monoclonal gammopathy characterized by biological heterogeneity and unregulated proliferation of plasma cells (PCs) in bone marrow (BM). MM is a multistep process based on genomic instability, epigenetic dysregulation and a tight cross-talk with the BM microenvironment that plays a pivotal role supporting the proliferation, survival, drug-resistance and homing of PCs. The BM microenvironment consists of a hematopoietic and a non-hematopoietic compartment, which cooperate to create a tumor environment. Among the non-hematopoietic component, mesenchymal stromal cells (MSCs) and osteoblasts (OBs) appear transcriptionally and functionally different in MM patients compared to healthy donors (HDs) and to patients with pre-malignant monoclonal gammopathies. Alterations of both MSCs and OBs underly the osteolytic lesions that characterize myeloma-associated bone disease. In this review, we will discuss the different characteristics of MSCs and OBs in MM patients, analyzing the transcriptome, the deregulated molecular pathways and the role performed by miRNAs and exosome in the pathophysiology of MM.
Collapse
|
8
|
Zhu S, Xing C, Zhang G, Peng H, Wang Z. CC1007, a small molecular compound, suppresses multiple myeloma via upregulation of Nur77. Bioorg Chem 2022; 129:106217. [PMID: 36283176 DOI: 10.1016/j.bioorg.2022.106217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/20/2022] [Accepted: 10/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is a hematological malignancy of plasma cells characterized by the production of monoclonal immunoglobulin protein. Despite significant advances in the treatment of MM, it remains an incurable disorder owing to its resistance to chemotherapy and refractory nature. Inhibitors of histone deacetylases (HDACIs) have been identified as promising therapeutic drugs for cancer treatment. At present, numerous HDACIs are under study for the treatment of MM in monotherapy or in conjunction with other agents. OBJECTIVES In the present study, we investigated the anti-MM effect of CC1007, which was designed to indirectly inhibit class IIa HDACs by binding to myocyte enhancer factor-2 (MEF2) and blocking the targets regulated by the HDAC-MEF2 complex. DESIGN The effect of CC1007 on human MM cell lines, namely U266 and MM1.S, and CD138+ cells collected from the bone marrow of patients with MM was evaluated. METHODS The cells were subjected to growth-inhibition assay, apoptosis assay, cell cycle analysis, real-time PCR, western blotting, immunofluorescence, co-immunoprecipitation, ChIP assay, and siRNA transfection. Statistical differences were compared using two-tailed t tests or one-way analysis of variance followed by the Bonferroni post hoc test. RESULTS CC1007 inhibited the proliferation of MM cell lines and primary MM cells and induced their apoptosis and cell cycle arrest. Furthermore, CC1007 decreased the expression of MEF2C and HDAC7, thereby disturbing their interaction and promoting the overexpression of Nur77, a target of MEF2C. The overexpression of Nur77 and its translocation from the nucleus to the cytoplasm resulted in its binding to B-cell lymphoma 2 on the mitochondrial surface, thereby inducing the release of cytochrome C and activating the mitochondrial apoptotic pathway. CONCLUSIONS Since CC1007 demonstrates remarkable anti-MM effect on MM cells, it may be a promising drug for the treatment of MM.
Collapse
Affiliation(s)
- Shicong Zhu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Molecular Hematology, Central South University, Changsha, China
| | - Cheng Xing
- Institute of Molecular Hematology, Central South University, Changsha, China; Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Guangsen Zhang
- Institute of Molecular Hematology, Central South University, Changsha, China; Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Hongling Peng
- Institute of Molecular Hematology, Central South University, Changsha, China; Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Zhihua Wang
- Institute of Molecular Hematology, Central South University, Changsha, China; Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China.
| |
Collapse
|
9
|
Raimondi V, Toscani D, Marchica V, Burroughs-Garcia J, Storti P, Giuliani N. Metabolic features of myeloma cells in the context of bone microenvironment: Implication for the pathophysiology and clinic of myeloma bone disease. Front Oncol 2022; 12:1015402. [PMID: 36313705 PMCID: PMC9608343 DOI: 10.3389/fonc.2022.1015402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of malignant plasma cells (PCs) into the bone marrow (BM). The complex interaction between the BM microenvironment and MM PCs can lead to severe impairment of bone remodeling. Indeed, the BM microenvironment exerts a critical role in the survival of malignant PCs. Growing evidence indicates that MM cells have several metabolic features including enhanced glycolysis and an increase in lactate production through the upregulation of glucose transporters and enzymes. More recently, it has been reported that MM cells arehighly glutamine addicted. Interestingly, these metabolic changes in MM cells may affect BM microenvironment cells by altering the differentiation process of osteoblasts from mesenchymal stromal cells. The identification of glutamine metabolism alterations in MM cells and bone microenvironment may provide a rationale to design new therapeutic approaches and diagnostic tools. The osteolytic lesions are the most frequent clinical features in MM patients, often characterized by pathological fractures and acute pain. The use of the newer imaging techniques such as Magnetic Resonance Imaging (MRI) and combined Positron Emission Tomography (PET) and Computerized Tomography (CT) has been introduced into clinical practice to better define the skeletal involvement. Currently, the PET/CT with 18F-fluorodeoxyglucose (FDG) is the diagnostic gold standard to detect active MM bone disease due to the high glycolytic activity of MM cells. However, new tracers are actively under investigation because a portion of MM patients remains negative at the skeletal level by 18F-FDG. In this review, we will summarize the existing knowledge on the metabolic alterations of MM cells considering their impact on the BM microenvironment cells and particularly in the subsequent formation of osteolytic bone lesions. Based on this, we will discuss the identification of possible new druggable targets and the use of novel metabolic targets for PET imaging in the detection of skeletal lesions, in the staging and treatment response of MM patients.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- *Correspondence: Paola Storti, ; Nicola Giuliani,
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Hematology, “Azienda Ospedaliero-Universitaria di Parma”, Parma, Italy
- *Correspondence: Paola Storti, ; Nicola Giuliani,
| |
Collapse
|
10
|
Analysis of Serum Proteome after Treatment of Osteoporosis with Anabolic or Antiresorptive Drugs. Metabolites 2022; 12:metabo12050399. [PMID: 35629903 PMCID: PMC9143104 DOI: 10.3390/metabo12050399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
The aim of the study was to explore new markers in serum proteome associated with the response to antiosteoporosis drugs, namely teriparatide and denosumab. We obtained serum samples from 14 patients with osteoporosis, both at baseline and after 6 months of treatment with teriparatide (n = 10) or denosumab (n = 4). Samples were analyzed by nanoliquid chromatography coupled to high-resolution mass spectrometry on a QTOF 5600 (SCIEX) apparatus. The spectrometry data were analyzed with Mascot against the UniProtKB base and then several quality-control filters were applied for the identification of peptides (false discovery rate, FDR q < 0.02) and their quantification (FDR q < 0.05). In the group treated with teriparatide, 28 proteins were identified with significant differences before and after treatment. A pathway analysis by using the Reactome database revealed significant enrichment in the Insulin Like Growth Factor 1 (IGF-I) (FDR q 4 × 10−2) and innate immune system (FDR q 2 × 10−3) pathways. Among patients treated with denosumab, we observed significant differences in the levels of 10 proteins, which were also enriched in the pathways related to the innate immune system (FDR q 3 × 10−2). These results suggest that the innate immune system may be involved in the response to antiosteoporosis drugs.
Collapse
|
11
|
Uhl C, Nyirenda T, Siegel DS, Lee WY, Zilberberg J. Natural killer cells activity against multiple myeloma cells is modulated by osteoblast-induced IL-6 and IL-10 production. Heliyon 2022; 8:e09167. [PMID: 35846441 PMCID: PMC9280577 DOI: 10.1016/j.heliyon.2022.e09167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/29/2021] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Background Natural killer (NK) cells are part of the innate arm of the immune system; as such NK cells can be activated rapidly to target virus-infected cells and tumor cells without prior sensitization. The human NK-92MI cell line is among the most widely used NK cell in preclinical research studies and has also been approved for clinical applications. Previous studies have shown that osteoblasts (OSB) confer drug resistance in multiple myeloma (MM) and other cancers that metastasize to the bone marrow. Aim We evaluated here how OSB, which are bone forming cells and a key cellular component of the bone marrow microenvironment, modulate the cytotoxic activity of NK-92MI cells against the MM.1S multiple myeloma cell line. Methods The osteoblastic niche was recapitulated with either the osteoblastic cell line hFOB 1.19 (hFOB) or primary osteoblasts (P-OSB) derived from surgical resections. Time-lapse imaging was utilized to quantify changes in MM.1S cell viability under different conditions, including: (1) Co-culture of MM.1S with NK92MI cells, (2) triple-culture of hFOB or P-OSB with MM.1S and NK-92MI, and (3) MM.1S or NK-92MI cells primed with OSB-derived supernatant. Cytokine analysis was conducted to quantify potential secreted factors associated with the protective effects of OSB. Results The physical presence of OSB hindered the activity of NK-92MI cells, resulting in the increased viability of MM.1S compared to co-cultures which lacked OSB. This observation was accompanied by reduced perforin and granzyme A secretion from NK-92MI cells. Contact of OSB and NK-92MI cells also induced interleukin 6 (IL-6) and interleukin 10 (IL-10) production; two cytokines which are known to impair the NK cell immunity against MM and other cancers. OSB supernatant also conferred cytoprotection to MM.1S, suggesting a dual mechanism by which OSB may modulate both NK and MM cells. Conclusions We demonstrated here that OSB can negatively impact the activity of NK cells against MM. As NK cells and their chimeric antigen receptor-modified versions become more widely used in the clinic, our results suggest that understanding the role of OSB as potential immunoregulators of the NK cell-mediated cytotoxic response in the bone marrow tumor microenvironment may provide new opportunities for enhancing the effectiveness of this potent immunotherapeutic approach.
Collapse
|
12
|
Heider M, Nickel K, Högner M, Bassermann F. Multiple Myeloma: Molecular Pathogenesis and Disease Evolution. Oncol Res Treat 2021; 44:672-681. [PMID: 34749378 DOI: 10.1159/000520312] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Multiple myeloma is the second most common hematologic malignancy, which to date remains incurable despite advances in treatment strategies including the use of novel substances such as proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. SUMMARY The bone marrow-based disease is preceded by the 2 sequential premalignant conditions: monoclonal gammo-pathy of undetermined significance and smoldering myeloma. Plasma cell leukemia and extramedullary disease occur, when malignant clones lose their dependency on the bone marrow. Key genetic features of these plasma cell dyscrasias include chromosomal aberrations such as translocations and hyperdiploidy, which occur during error-prone physiologic processes in B-cell development. Next-generation sequencing studies have identified mutations in major oncogenic pathways and tumor suppressors, which contribute to the pathogenesis of multiple myeloma and have revealed insights into the clonal evolution of the disease, particularly along different lines of therapy. More recently, the importance of epigenetic alterations and the role of the bone marrow microenvironment, including immune and osteogenic cells, have become evident. Key Messages: We herein review the current knowledge of the pathogenesis of multiple myeloma, which is crucial for the development of novel targeted therapeutic strategies. These can contribute to the endeavor to make multiple myeloma a curable disease.
Collapse
Affiliation(s)
- Michael Heider
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Katharina Nickel
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marion Högner
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
Gastelum G, Veena M, Lyons K, Lamb C, Jacobs N, Yamada A, Baibussinov A, Sarafyan M, Shamis R, Kraut J, Frost P. Can Targeting Hypoxia-Mediated Acidification of the Bone Marrow Microenvironment Kill Myeloma Tumor Cells? Front Oncol 2021; 11:703878. [PMID: 34350119 PMCID: PMC8327776 DOI: 10.3389/fonc.2021.703878] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is an incurable cancer arising from malignant plasma cells that engraft in the bone marrow (BM). The physiology of these cancer cells within the BM microenvironment (TME) plays a critical role in MM development. These processes may be similar to what has been observed in the TME of other (non-hematological) solid tumors. It has been long reported that within the BM, vascular endothelial growth factor (VEGF), increased angiogenesis and microvessel density, and activation of hypoxia-induced transcription factors (HIF) are correlated with MM progression but despite a great deal of effort and some modest preclinical success the overall clinical efficacy of using anti-angiogenic and hypoxia-targeting strategies, has been limited. This review will explore the hypothesis that the TME of MM engrafted in the BM is distinctly different from non-hematological-derived solid tumors calling into question how effective these strategies may be against MM. We further identify other hypoxia-mediated effectors, such as hypoxia-mediated acidification of the TME, oxygen-dependent metabolic changes, and the generation of reactive oxygen species (ROS), that may prove to be more effective targets against MM.
Collapse
Affiliation(s)
- Gilberto Gastelum
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Research, Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, United States
| | - Mysore Veena
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Research, Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, United States
| | - Kylee Lyons
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher Lamb
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicole Jacobs
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexandra Yamada
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alisher Baibussinov
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Martin Sarafyan
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rebeka Shamis
- Department of Research, Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, United States
| | - Jeffry Kraut
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Research, Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, United States
| | - Patrick Frost
- Department of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Research, Greater Los Angeles Veterans Administration Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
14
|
Da Vià MC, Ziccheddu B, Maeda A, Bagnoli F, Perrone G, Bolli N. A Journey Through Myeloma Evolution: From the Normal Plasma Cell to Disease Complexity. Hemasphere 2020; 4:e502. [PMID: 33283171 PMCID: PMC7710229 DOI: 10.1097/hs9.0000000000000502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The knowledge of cancer origin and the subsequent tracking of disease evolution represent unmet needs that will soon be within clinical reach. This will provide the opportunity to improve patient's stratification and to personalize treatments based on cancer biology along its life history. In this review, we focus on the molecular pathogenesis of multiple myeloma (MM), a hematologic malignancy with a well-known multi-stage disease course, where such approach can sooner translate into a clinical benefit. We describe novel insights into modes and timing of disease initiation. We dissect the biology of the preclinical and pre-malignant phases, elucidating how knowledge of the genomics of the disease and the composition of the microenvironment allow stratification of patients based on risk of disease progression. Then, we explore cell-intrinsic and cell-extrinsic drivers of MM evolution to symptomatic disease. Finally, we discuss how this may relate to the development of refractory disease after treatment. By integrating an evolutionary view of myeloma biology with the recent acquisitions on its clonal heterogeneity, we envision a way to drive the clinical management of the disease based on its detailed biological features more than surrogates of disease burden.
Collapse
Affiliation(s)
- Matteo C. Da Vià
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Bachisio Ziccheddu
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Akihiro Maeda
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Filippo Bagnoli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Perrone
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
15
|
Myeloma Cells Deplete Bone Marrow Glutamine and Inhibit Osteoblast Differentiation Limiting Asparagine Availability. Cancers (Basel) 2020; 12:cancers12113267. [PMID: 33167336 PMCID: PMC7694402 DOI: 10.3390/cancers12113267] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Osteolytic bone lesions represent an important clinical feature of multiple myeloma (MM). MM cells metabolize very high amounts of glutamine (Gln) and significantly lower Gln in the bone marrow. In this contribution we demonstrate that MM-dependent Gln depletion impairs the differentiation of bone marrow mesenchymal stromal cells into osteoblasts, the cells that form new bone tissue. We also found that osteoblast differentiation is associated with increased expression of glutaminase, the main enzyme that metabolizes Gln, SNAT2, a transporter able to accumulate Gln into the cells, and asparagine synthetase, the enzyme that uses Gln to obtain asparagine (Asn). Asn rescued osteoblast differentiation of Gln-starved mesenchymal stromal cells. These results demonstrate that MM cells impair osteoblast differentiation, hindering mesenchymal Asn synthesis through Gln depletion. Besides providing a metabolic mechanism underlying osteolytic lesions in MM, these results suggest that Asn supplementation may prevent bone disease in MM patients. Abstract Multiple myeloma (MM) cells consume huge amounts of glutamine and, as a consequence, the amino acid concentration is lower-than-normal in the bone marrow (BM) of MM patients. Here we show that MM-dependent glutamine depletion induces glutamine synthetase in stromal cells, as demonstrated in BM biopsies of MM patients, and reproduced in vitro by co-culturing human mesenchymal stromal cells (MSCs) with MM cells. Moreover, glutamine depletion hinders osteoblast differentiation of MSCs, which is also severely blunted by the spent, low-glutamine medium of MM cells, and rescued by glutamine restitution. Glutaminase and the concentrative glutamine transporter SNAT2 are induced during osteoblastogenesis in vivo and in vitro, and both needed for MSCs differentiation, pointing to enhanced the requirement for the amino acid. Osteoblastogenesis also triggers the induction of glutamine-dependent asparagine synthetase (ASNS), and, among non-essential amino acids, asparagine rescues differentiation of glutamine-starved MSCs, by restoring the transcriptional profiles of differentiating MSCs altered by glutamine starvation. Thus, reduced asparagine availability provides a mechanistic link between MM-dependent Gln depletion in BM and impairment of osteoblast differentiation. Inhibition of Gln metabolism in MM cells and supplementation of asparagine to stromal cells may, therefore, constitute novel approaches to prevent osteolytic lesions in MM.
Collapse
|
16
|
Multiple Myeloma as a Bone Disease? The Tissue Disruption-Induced Cell Stochasticity (TiDiS) Theory. Cancers (Basel) 2020; 12:cancers12082158. [PMID: 32759688 PMCID: PMC7463431 DOI: 10.3390/cancers12082158] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022] Open
Abstract
The standard model of multiple myeloma (MM) relies on genetic instability in the normal counterparts of MM cells. MM-induced lytic bone lesions are considered as end organ damages. However, bone is a tissue of significance in MM and bone changes could be at the origin/facilitate the emergence of MM. We propose the tissue disruption-induced cell stochasticity (TiDiS) theory for MM oncogenesis that integrates disruption of the microenvironment, differentiation, and genetic alterations. It starts with the observation that the bone marrow endosteal niche controls differentiation. As decrease in cellular stochasticity occurs thanks to cellular interactions in differentiating cells, the initiating role of bone disruption would be in the increase of cellular stochasticity. Thus, in the context of polyclonal activation of B cells, memory B cells and plasmablasts would compete for localizing in endosteal niches with the risk that some cells cannot fully differentiate if they cannot reside in the niche because of a disrupted microenvironment. Therefore, they would remain in an unstable state with residual proliferation, with the risk that subclones may transform into malignant cells. Finally, diagnostic and therapeutic perspectives are provided.
Collapse
|
17
|
Bone, a Secondary Growth Site of Breast and Prostate Carcinomas: Role of Osteocytes. Cancers (Basel) 2020; 12:cancers12071812. [PMID: 32640686 PMCID: PMC7408809 DOI: 10.3390/cancers12071812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is the primarily preferred site for breast and prostate cancer to metastasize. Bone metastases are responsible for most deaths related to breast and prostate cancer. The bone's particular microenvironment makes it conducive for the growth of cancer cells. Studies on bone metastasis have focused on the interaction between cancer cells and the bone microenvironment. Osteocytes, the most common cell type of bone tissue, have received little attention in bone metastasis, although they are master signal sensors, integrators, and skeleton transducers. They play an important role in regulating bone mass by acting on both osteoblasts and osteoclasts, through the release of proteins such as sclerostin, Dickkopf-1 (DKK-1), and fibroblast growth factor 23 (FGF23). Osteocytes have been extensively re-evaluated, in light of their multiple functions: with different experimental approaches, it has been shown that, indeed, osteocytes are actively involved in the colonization of bone tissue by cancer cells. The present review focuses on recent research on the role that osteocytes play in bone metastasis of breast and prostate cancers. Moreover, the studies here summarized open up perspectives for new therapeutic approaches focused on modulating the activity of osteocytes to improve the condition of the bone metastatic patients. A better understanding of the complex interactions between cancer cells and bone-resident cells is indispensable for identifying potential therapeutic targets to stop tumor progression and prevent bone metastases.
Collapse
|
18
|
Raimondo S, Urzì O, Conigliaro A, Lo Bosco G, Parisi S, Carlisi M, Siragusa S, Raimondi L, De Luca A, Giavaresi G, Alessandro R. Extracellular Vesicle microRNAs Contribute to the Osteogenic Inhibition of Mesenchymal Stem Cells in Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12020449. [PMID: 32075123 PMCID: PMC7072478 DOI: 10.3390/cancers12020449] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
Osteolytic bone disease is the major complication associated with the progression of multiple myeloma (MM). Recently, extracellular vesicles (EVs) have emerged as mediators of MM-associated bone disease by inhibiting the osteogenic differentiation of human mesenchymal stem cells (hMSCs). Here, we investigated a correlation between the EV-mediated osteogenic inhibition and MM vesicle content, focusing on miRNAs. By the use of a MicroRNA Card, we identified a pool of miRNAs, highly expressed in EVs, from MM cell line (MM1.S EVs), expression of which was confirmed in EVs from bone marrow (BM) plasma of patients affected by smoldering myeloma (SMM) and MM. Notably, we found that miR-129-5p, which targets different osteoblast (OBs) differentiation markers, is enriched in MM-EVs compared to SMM-EVs, thus suggesting a selective packaging correlated with pathological grade. We found that miR-129-5p can be transported to hMSCs by MM-EVs and, by the use of miRNA mimics, we investigated its role in recipient cells. Our data demonstrated that the increase of miR-129-5p levels in hMSCs under osteoblastic differentiation stimuli inhibited the expression of the transcription factor Sp1, previously described as a positive modulator of osteoblastic differentiation, and of its target the Alkaline phosphatase (ALPL), thus identifying miR-129-5p among the players of vesicle-mediated bone disease.
Collapse
Affiliation(s)
- Stefania Raimondo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (O.U.); (A.C.); (S.P.)
| | - Ornella Urzì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (O.U.); (A.C.); (S.P.)
| | - Alice Conigliaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (O.U.); (A.C.); (S.P.)
| | - Giosuè Lo Bosco
- Department of Mathematics and Computer Science, University of Palermo, 90133 Palermo, Italy;
- Department of Sciences for technological innovation, Euro-Mediterranean Institute of Science and Technology, 90133 Palermo, Italy
| | - Sofia Parisi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (O.U.); (A.C.); (S.P.)
| | - Melania Carlisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Haematology Unit, University of Palermo, 90133 Palermo, Italy; (M.C.); (S.S.)
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Haematology Unit, University of Palermo, 90133 Palermo, Italy; (M.C.); (S.S.)
| | - Lavinia Raimondi
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.R.); (A.D.L.); (G.G.)
| | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.R.); (A.D.L.); (G.G.)
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.R.); (A.D.L.); (G.G.)
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (S.R.); (O.U.); (A.C.); (S.P.)
- Correspondence:
| |
Collapse
|
19
|
Li R, Lin S, Zhu M, Deng Y, Chen X, Wei K, Xu J, Li G, Bian L. Synthetic presentation of noncanonical Wnt5a motif promotes mechanosensing-dependent differentiation of stem cells and regeneration. SCIENCE ADVANCES 2019; 5:eaaw3896. [PMID: 31663014 PMCID: PMC6795506 DOI: 10.1126/sciadv.aaw3896] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 09/25/2019] [Indexed: 05/30/2023]
Abstract
Noncanonical Wnt signaling in stem cells is essential to numerous developmental events. However, no prior studies have capitalized on the osteoinductive potential of noncanonical Wnt ligands to functionalize biomaterials in enhancing the osteogenesis and associated skeleton formation. Here, we investigated the efficacy of the functionalization of biomaterials with a synthetic Wnt5a mimetic ligand (Foxy5 peptide) to promote the mechanosensing and osteogenesis of human mesenchymal stem cells by activating noncanonical Wnt signaling. Our findings showed that the immobilized Wnt5a mimetic ligand activated noncanonical Wnt signaling via the up-regulation of Disheveled 2 and downstream RhoA-ROCK signaling, leading to enhanced intracellular calcium level, F-actin stability, actomyosin contractility, and cell adhesion structure development. This enhanced mechanotransduction in stem cells promoted the in vitro osteogenic lineage commitment and the in vivo healing of rat calvarial defects. Our work provides valuable guidance for the developmentally inspired design of biomaterials for a wide array of therapeutic applications.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Sien Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Meiling Zhu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Xiaoyu Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Kongchang Wei
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, Lerchenfeldstrasse 5, CH-9014 St. Gallen, Switzerland
| | - Jianbin Xu
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P. R. China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, P. R. China
- Center of Novel Biomaterials, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077 Hong Kong, P.R. China
| |
Collapse
|
20
|
Giuliani N, Accardi F, Marchica V, Dalla Palma B, Storti P, Toscani D, Vicario E, Malavasi F. Novel targets for the treatment of relapsing multiple myeloma. Expert Rev Hematol 2019; 12:481-496. [PMID: 31125526 DOI: 10.1080/17474086.2019.1624158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.
Collapse
Affiliation(s)
- Nicola Giuliani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabrizio Accardi
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Valentina Marchica
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | | | - Paola Storti
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Denise Toscani
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Emanuela Vicario
- a Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Fabio Malavasi
- b Department of Medical Science , University of Turin , Turin , Italy
| |
Collapse
|
21
|
Faraahi Z, Baud'huin M, Croucher PI, Eaton C, Lawson MA. Sostdc1: A soluble BMP and Wnt antagonist that is induced by the interaction between myeloma cells and osteoblast lineage cells. Bone 2019; 122:82-92. [PMID: 30776499 PMCID: PMC6458996 DOI: 10.1016/j.bone.2019.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 01/01/2023]
Abstract
Multiple myeloma (MM) is characterised by destructive lytic bone disease, caused by induction of bone resorption and impaired bone formation. Our understanding of the molecular mechanisms responsible for osteoblast suppression, are limited. Using the 5T2MM murine model of MM we have previously shown that suppression of the activity of a known inhibitor of bone formation Dikkopf-1 (Dkk1) prevents the development of lytic bone disease. Here we have demonstrated that another potential inhibitor of bone formation, sclerostin domain containing 1 (Sostdc1) is expressed at low levels in MM and osteoblast lineage cells when these cells are grown separately in cell culture but its expression is significantly induced in both cell types when these cells are in contact. The distribution of Sostdc1 staining in bones infiltrated with 5TGM1 myeloma cells in vivo suggested its presence in both myeloma and osteoblast lineage populations when in close proximity. We have also shown that recombinant Sostdc1 inhibits both bone morphogenic proteins (BMP2 and 7) and Wnt signalling in primary osteoblasts and suppresses differentiation of these cells. Together, these findings suggest that Sostdc1 expression in 5TGM1-infiltrated bones as a result of the interaction between myeloma and osteoblast lineage populations, could result in suppression of osteoblast differentiation.
Collapse
Affiliation(s)
- Z Faraahi
- Institute for Cancer Sciences, University of Manchester, UK
| | | | - P I Croucher
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - C Eaton
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK
| | - M A Lawson
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.
| |
Collapse
|
22
|
Monoclonal gammopathy of undetermined significance. Blood 2019; 133:2484-2494. [PMID: 31010848 DOI: 10.1182/blood.2019846782] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS) is a premalignant plasma cell dyscrasia that consistently precedes multiple myeloma (MM) with a 1% risk of progression per year. Recent advances have improved understanding of the complex genetic and immunologic factors that permit progression from the aberrant plasma cell clone to MGUS and overt MM. Additional evidence supports bidirectional interaction of MGUS cells with surrounding cells in the bone marrow niche that regulates malignant transformation. However, there are no robust prognostic biomarkers. Herein we review the current body of literature on the biology of MGUS and provide a rationale for the improved identification of high-risk MGUS patients who may be appropriate for novel clinical interventions to prevent progression or eradicate premalignant clones prior to the development of overt MM.
Collapse
|
23
|
Yurova KA, Khaziakhmatova OG, Melashchenko ES, Malashchenko VV, Shunkin EO, Shupletsova VV, Ivanov PA, Khlusov IA, Litvinova LS. Cellular and Molecular Basis of Osteoblastic and Vascular Niches in the Processes of Hematopoiesis and Bone Remodeling (A Short Review of Modern Views). Curr Pharm Des 2019; 25:663-669. [PMID: 30931856 DOI: 10.2174/1381612825666190329153626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023]
Abstract
In evolutionary processes, human bone marrow has formed as an organ depot of various types of cells that arise from hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Vital HSC activity is controlled through molecular interactions with the niche microenvironment. The review describes current views on the formation of key molecular and cellular components of the HSC niche, which ensure maintenance of home ostasis in stem cell niches, obtained from studies of their role in regulating the proliferation and differentiation of HSCs, including the physiological, reparative and pathological remodeling of bone tissue. Due to rapid developments in biotechnology, tissue bioengineering, and regenerative medicine, information can be useful for developing biomimetic and bioinspired materials and implants that provide an effective bone/bone marrow recovery process after injuries and, to a greater extent, diseases of various etiologies.
Collapse
Affiliation(s)
- Kristina A Yurova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Olga G Khaziakhmatova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Elena S Melashchenko
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Vladimir V Malashchenko
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Egor O Shunkin
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Valeria V Shupletsova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation.,Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Pavel A Ivanov
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Igor A Khlusov
- Morphology and General Pathology Department, Siberian State Medical University, Tomsk, Russian Federation.,Research School of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Tomskaya oblast, Russian Federation
| | - Larisa S Litvinova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| |
Collapse
|
24
|
Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9010174] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies.
Collapse
|
25
|
Zhang L, Lei Q, Wang H, Xu C, Liu T, Kong F, Yang C, Yan G, Sun L, Zhao A, Chen W, Hu Y, Xie H, Cao Y, Fu F, Yuan G, Chen Z, Guo AY, Li Q. Tumor-derived extracellular vesicles inhibit osteogenesis and exacerbate myeloma bone disease. Am J Cancer Res 2019; 9:196-209. [PMID: 30662562 PMCID: PMC6332790 DOI: 10.7150/thno.27550] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Background: As a hallmark driver of multiple myeloma (MM), MM bone disease (MBD) is unique in that it is characterized by severely impaired osteoblast activity resulting from blocked osteogenesis in bone marrow-derived mesenchymal stem cells (BM-MSCs). The mechanisms underlying this preferential blockade are incompletely understood. Methods: miRNA expression of MM cell-derived extracellular vesicles (MM-EVs) was detected by RNA sequencing. MM-EVs impaired osteogenesis and exacerbated MBD were in vitro and in vivo validated by histochemical staining, qPCR and micro-CT. We additionally examined the correlation between CD138+ circulating EVs (cirEVs) count and bone lesion in de novo MM patients. Results: Here, by sequencing and bioinformatics analysis, we found that MM-EVs were enriched in various molecules negatively regulating osteogenesis. We experimentally verified that MM-EVs inhibited BM-MSC osteogenesis, induced elevated expression of miR-103a-3p inhibiting osteogenesis in BM-MSCs, and increased cell viability and interleukin-6 secretion in MM cells. In a mouse model, MM-EVs that were injected into the marrow space of the left tibia led to impaired osteogenesis and exacerbated MBD and MM progression. Furthermore, the levels of CD138+ cirEVs in the peripheral blood were positively correlated with the number of MM bone lesions in MM patients. Conclusions: These findings suggest that MM-EVs play a pivotal role in the development of severely impaired osteoblast activity, which represents a novel biomarker for the precise diagnosis of MBD and a compelling rationale for exploring MM-EVs as a therapeutic target.
Collapse
|
26
|
Horenstein AL, Morandi F, Bracci C, Pistoia V, Malavasi F. Functional insights into nucleotide-metabolizing ectoenzymes expressed by bone marrow-resident cells in patients with multiple myeloma. Immunol Lett 2018; 205:40-50. [PMID: 30447309 DOI: 10.1016/j.imlet.2018.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022]
Abstract
Human myeloma cells grow in a hypoxic acidic niche in the bone marrow. Cross talk among cellular components of this closed niche generates extracellular adenosine, which promotes tumor cell survival. This is achieved through the binding of adenosine to purinergic receptors into complexes that function as an autocrine/paracrine signal factor with immune regulatory activities that i) down-regulate the functions of most immune effector cells and ii) enhance the activity of cells that suppress anti-tumor immune responses, thus facilitating the escape of malignant myeloma cells from immune surveillance. Here we review recent findings confirming that the dominant phenotype for survival of tumor cells is that where the malignant cells have been metabolically reprogrammed for the generation of lactic acidosis in the bone marrow niche. Adenosine triphosphate and nicotinamide-adenine dinucleotide extruded from tumor cells, along with cyclic adenosine monophosphate, are the main intracellular energetic/messenger molecules that serve as leading substrates in the extracellular space for membrane-bound ectonucleotidases metabolizing purine nucleotides to signaling adenosine. Within this mechanistic framework, the adenosinergic substrate conversion can vary significantly according to the metabolic environment. Indeed, the neoplastic expansion of plasma cells exploits both enzymatic networks and hypoxic acidic conditions for migrating and homing to a protected niche and for evading the immune response. The expression of multiple specific adenosine receptors in the niche completes the profile of a complex regulatory framework whose signals modify multiple myeloma and host immune responses.
Collapse
Affiliation(s)
- A L Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy.
| | - F Morandi
- Stem Cell Laboratory and Cell Therapy Center, Istituto Giannina Gaslini, Genova, Italy
| | - C Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy
| | - V Pistoia
- Immunology Area, Pediatric Hospital Bambino Gesù, Rome, Italy
| | - F Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Italy; CeRMS, University of Torino, Torino, Italy
| |
Collapse
|
27
|
Toscani D, Bolzoni M, Ferretti M, Palumbo C, Giuliani N. Role of Osteocytes in Myeloma Bone Disease: Anti-sclerostin Antibody as New Therapeutic Strategy. Front Immunol 2018; 9:2467. [PMID: 30410490 PMCID: PMC6209728 DOI: 10.3389/fimmu.2018.02467] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/05/2018] [Indexed: 11/16/2022] Open
Abstract
Osteocytes are terminally differentiated cells of the osteoblast lineage. They are involved in the regulation of bone remodeling by increasing osteoclast formation or decreasing bone formation by the secretion of the osteoblast inhibitor sclerostin. Monoclonal antibody anti-sclerostin, Romosozumab, has been developed and tested in clinical trials in patients with osteoporosis. In the last years, the role of osteocytes in the development of osteolytic bone lesions that occurs in multiple myeloma, have been underlined. Myeloma cells increase osteocyte death through the up-regulation of both apoptosis and autophagy that, in turn, triggers osteoclast formation, and activity. When compared to healthy controls, myeloma patients with bone disease have higher osteocyte cell death, but the treatment with proteasome inhibitor bortezomib has been shown to maintain osteocyte viability. In preclinical mouse models of multiple myeloma, treatment with blocking anti-sclerostin antibody increased osteoblast numbers and bone formation rate reducing osteolytic bone lesions. Moreover, the combination of anti-sclerostin antibody and the osteoclast inhibitor zoledronic acid increased bone mass and fracture resistance synergistically. However, anti-sclerostin antibody did not affect tumor burden in vivo or the efficacy of anti-myeloma drugs in vitro. Nevertheless, the combination therapy of anti-sclerostin antibody and the proteasome inhibitor carfilzomib, displayed potent anti-myeloma activity as well as positive effects on bone disease in vivo. In conclusion, all these data suggest that osteocytes are involved in myeloma bone disease and may be considered a novel target for the use of antibody-mediated anti-sclerostin therapy also in multiple myeloma patients.
Collapse
Affiliation(s)
- Denise Toscani
- Department Medicine and Surgery, University of Parma, Parma, Italy
| | - Marina Bolzoni
- Department Medicine and Surgery, University of Parma, Parma, Italy
| | - Marzia Ferretti
- Department of Biomedical, Metabolic and Neural Sciences, Human Morphology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Carla Palumbo
- Department of Biomedical, Metabolic and Neural Sciences, Human Morphology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicola Giuliani
- Department Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
28
|
Capp JP, Bataille R. Multiple Myeloma Exemplifies a Model of Cancer Based on Tissue Disruption as the Initiator Event. Front Oncol 2018; 8:355. [PMID: 30250824 PMCID: PMC6140628 DOI: 10.3389/fonc.2018.00355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022] Open
Abstract
The standard model of multiple myeloma (MM) oncogenesis is based on the genetic instability of MM cells and presents its evolution as the emergence of clones with more and more aggressive genotypes, giving them surviving and proliferating advantage. The micro-environment has a passive role. In contrast, many works have shown that the progression of MM is also characterized by the selection of clones with extended phenotypes able to destroy bone trabeculae, suggesting a major role for early micro-environmental disruption. We present a model of MM oncogenesis in which genetic instability is the consequence of the disruption of normal interactions between plasma cells and their environment, the bone remodeling compartment. These interactions, which normally ensure the stability of the genotypes and phenotypes of normal plasma cells could be disrupted by many factors as soon as the early steps of the disease (MGUS, pre-MGUS states). Therapeutical implications of the model are presented.
Collapse
Affiliation(s)
- Jean-Pascal Capp
- LISBP, UMR CNRS 5504, UMR INRA 792, INSA Toulouse, University of Toulouse, Toulouse, France
| | - Régis Bataille
- Faculty of Medecine, University of Angers, Angers, France
| |
Collapse
|
29
|
Zhao W, Zhang X, Zang L, Zhao P, Chen Y, Wang X. ILK promotes angiogenic activity of mesenchymal stem cells in multiple myeloma. Oncol Lett 2018; 16:1101-1106. [PMID: 29963187 DOI: 10.3892/ol.2018.8711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 02/06/2018] [Indexed: 01/19/2023] Open
Abstract
Angiogenic activity in solid tumors has been demonstrated to promote metastasis through the activation of certain proteins involved in the epithelial-mesenchymal transition-associated process. The molecular mechanism underlying multiple myeloma-induced angiogenesis involves angiogenic cytokines by plasma cells as well as their induction within the microenvironment. Integrin-linked kinase (ILK) is a highly evolutionarily conserved intracellular protein that was originally identified as an integrin-interacting protein, and extensive genetic and biochemical studies have identified ILK expression to be vital during tumor-driven angiogenesis. In the present study, it was identified that angiogenic factors were upregulated in mesenchymal stem cells (MSCs) that were co-cultured with multiple myeloma cell lines. It was also revealed that upregulated ILK expression significantly promoted the capillary-formation ability of MSCs. The concentrations of angiogenic factors were significantly decreased compared with non-targeting siRNA-transfected and control MSCs. MSCs may participate in inducing the angiogenic response in multiple myeloma depending on ILK expression.
Collapse
Affiliation(s)
- Weipeng Zhao
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xiaoying Zhang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Li Zang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Pan Zhao
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Yafang Chen
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xiaofang Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| |
Collapse
|
30
|
Braham MV, Ahlfeld T, Akkineni AR, Minnema MC, Dhert WJ, Öner FC, Robin C, Lode A, Gelinsky M, Alblas J. Endosteal and Perivascular Subniches in a 3D Bone Marrow Model for Multiple Myeloma. Tissue Eng Part C Methods 2018; 24:300-312. [DOI: 10.1089/ten.tec.2017.0467] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Maaike V.J. Braham
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tilman Ahlfeld
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - A. Rahul Akkineni
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Monique C. Minnema
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Dresden, Germany
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Bolzoni M, Toscani D, Costa F, Vicario E, Aversa F, Giuliani N. The link between bone microenvironment and immune cells in multiple myeloma: Emerging role of CD38. Immunol Lett 2018; 205:65-70. [PMID: 29702149 DOI: 10.1016/j.imlet.2018.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/30/2022]
Abstract
The relationship between bone and immune cells is well established both in physiological and pathological conditions. Multiple myeloma (MM) is a plasma cell malignancy characterized by an increase of number and activity of osteoclasts (OCLs) and a decrease of osteoblasts (OBs). These events are responsible for bone lesions of MM patients. OCLs support MM cells survival in vitro and in vivo. Recently, the possible role of OCLs as immunosuppressive cells in the MM BM microenvironment has been underlined. OCLs protect MM cells against T cell-mediated cytotoxicity through the expression of several molecules including programmed death-ligand (PD-L) 1, galectin (Gal) 9, CD200, and indoleamine-2,3-dioxygenase (IDO). Among the molecules that could be involved in the link between immune-microenvironment and osteoclastogenesis the role of CD38 has been hypothesized. CD38 is a well-known adhesion molecule and an ectoenzyme highly expressed by MM cells. Moreover, CD38 is expressed by OCLs and at the surface level on OCL precursors. Targeting CD38 with monoclonal antibodies showed inhibition of both osteoclastogenesis and OCL-mediated suppression of T cell function. This review elucidates this evidence indicating that osteoclastogenesis affect MM immune-microenvironment being a potential target to improve anti-MM immunity and to ameliorate bone disease.
Collapse
Affiliation(s)
- Marina Bolzoni
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Denise Toscani
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Federica Costa
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Emanuela Vicario
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Biopathology and Medical Biotechnologies, Biology and Genetic Section, University of Palermo, 90133 Palermo, Italy
| | - Franco Aversa
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", 43126 Parma, Italy
| | - Nicola Giuliani
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", 43126 Parma, Italy.
| |
Collapse
|
32
|
Braham MVJ, Minnema MC, Aarts T, Sebestyen Z, Straetemans T, Vyborova A, Kuball J, Öner FC, Robin C, Alblas J. Cellular immunotherapy on primary multiple myeloma expanded in a 3D bone marrow niche model. Oncoimmunology 2018; 7:e1434465. [PMID: 29872571 PMCID: PMC5980416 DOI: 10.1080/2162402x.2018.1434465] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 12/01/2022] Open
Abstract
Bone marrow niches support multiple myeloma, providing signals and cell-cell interactions essential for disease progression. A 3D bone marrow niche model was developed, in which supportive multipotent mesenchymal stromal cells and their osteogenic derivatives were co-cultured with endothelial progenitor cells. These co-cultured cells formed networks within the 3D culture, facilitating the survival and proliferation of primary CD138+ myeloma cells for up to 28 days. During this culture, no genetic drift was observed within the genomic profile of the primary myeloma cells, indicating a stable outgrowth of the cultured CD138+ population. The 3D bone marrow niche model enabled testing of a novel class of engineered immune cells, so called TEGs (αβT cells engineered to express a defined γδTCR) on primary myeloma cells. TEGs were engineered and tested from both healthy donors and myeloma patients. The added TEGs were capable of migrating through the 3D culture, exerting a killing response towards the primary myeloma cells in 6 out of 8 donor samples after both 24 and 48 hours. Such a killing response was not observed when adding mock transduced T cells. No differences were observed comparing allogeneic and autologous therapy. The supporting stromal microenvironment was unaffected in all conditions after 48 hours. When adding TEG therapy, the 3D model surpassed 2D models in many aspects by enabling analyses of specific homing, and both on- and off-target effects, preparing the ground for the clinical testing of TEGs. The model allows studying novel immunotherapies, therapy resistance mechanisms and possible side-effects for this incurable disease.
Collapse
Affiliation(s)
- Maaike V. J. Braham
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Monique C. Minnema
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Tineke Aarts
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zsolt Sebestyen
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Trudy Straetemans
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna Vyborova
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jurgen Kuball
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
33
|
Ghobrial IM, Detappe A, Anderson KC, Steensma DP. The bone-marrow niche in MDS and MGUS: implications for AML and MM. Nat Rev Clin Oncol 2018; 15:219-233. [PMID: 29311715 DOI: 10.1038/nrclinonc.2017.197] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Several haematological malignancies, including multiple myeloma (MM) and acute myeloid leukaemia (AML), have well-defined precursor states that precede the development of overt cancer. MM is almost always preceded by monoclonal gammopathy of undetermined significance (MGUS), and at least a quarter of all patients with myelodysplastic syndromes (MDS) have disease that evolves into AML. In turn, MDS are frequently anteceded by clonal haematopoiesis of indeterminate potential (CHIP). The acquisition of additional genetic and epigenetic alterations over time clearly influences the increasingly unstable and aggressive behaviour of neoplastic haematopoietic clones; however, perturbations in the bone-marrow microenvironment are increasingly recognized to have key roles in initiating and supporting oncogenesis. In this Review, we focus on the concept that the haematopoietic neoplasia-microenvironment relationship is an intimate rapport between two partners, provide an overview of the evidence supporting a role for the bone-marrow niche in promoting neoplasia, and discuss the potential for niche-specific therapeutic targets.
Collapse
Affiliation(s)
- Irene M Ghobrial
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Alexandre Detappe
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Kenneth C Anderson
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
34
|
Yang Y, Wang B. PTH1R-CaSR Cross Talk: New Treatment Options for Breast Cancer Osteolytic Bone Metastases. Int J Endocrinol 2018; 2018:7120979. [PMID: 30151009 PMCID: PMC6087585 DOI: 10.1155/2018/7120979] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/14/2018] [Indexed: 12/11/2022] Open
Abstract
Metastatic breast cancer (BrCa) is currently incurable despite great improvements in treatment of primary BrCa. The incidence of skeletal metastases in advanced BrCa occurs up to 70%. Recent findings have established that the distribution of BrCa metastases to the skeleton is not a random process but due to the favorable microenvironment for tumor invasion and growth. The complex interplay among BrCa cells, stromal/osteoblastic cells, and osteoclasts in the osseous microenvironment creates a bone-tumor vicious cycle (a feed-forward loop) that results in excessive bone destruction and progressive tumor growth. Both the type 1 PTH receptor (PTH1R) and extracellular calcium-sensing receptor (CaSR) participate in the vicious cycle and influence the skeletal metastatic niche. Thus, this review focuses on how the PTH1R and CaSR signaling pathways interact and contribute to the pathogenesis of BrCa bone metastases. The effects of intermittent PTH and allosteric modulators of CaSR for the use of bone-anabolic agents and prevention of BrCa bone metastases constitute a proof of principle for therapeutic consideration. Understanding the interplay between PTH1R and CaSR signaling in the development of BrCa bone metastases could lead to a novel therapeutic approach to control both osteolysis and tumor burden in the bone.
Collapse
Affiliation(s)
- Yanmei Yang
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bin Wang
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
35
|
Heusschen R, Muller J, Binsfeld M, Marty C, Plougonven E, Dubois S, Mahli N, Moermans K, Carmeliet G, Léonard A, Baron F, Beguin Y, Menu E, Cohen-Solal M, Caers J. SRC kinase inhibition with saracatinib limits the development of osteolytic bone disease in multiple myeloma. Oncotarget 2017; 7:30712-29. [PMID: 27095574 PMCID: PMC5058712 DOI: 10.18632/oncotarget.8750] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/31/2016] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM)-associated osteolytic bone disease is a major cause of morbidity and mortality in MM patients and the development of new therapeutic strategies is of great interest. The proto-oncogene SRC is an attractive target for such a strategy. In the current study, we investigated the effect of treatment with the SRC inhibitor saracatinib (AZD0530) on osteoclast and osteoblast differentiation and function, and on the development of MM and its associated bone disease in the 5TGM.1 and 5T2MM murine MM models. In vitro data showed an inhibitory effect of saracatinib on osteoclast differentiation, polarization and resorptive function. In osteoblasts, collagen deposition and matrix mineralization were affected by saracatinib. MM cell proliferation and tumor burden remained unaltered following saracatinib treatment and we could not detect any synergistic effects with drugs that are part of standard care in MM. We observed a marked reduction of bone loss after treatment of MM-bearing mice with saracatinib as reflected by a restoration of trabecular bone parameters to levels observed in naive control mice. Histomorphometric analyses support that this occurs through an inhibition of bone resorption. In conclusion, these data further establish SRC inhibition as a promising therapeutic approach for the treatment of MM-associated osteolytic bone disease.
Collapse
Affiliation(s)
- Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Joséphine Muller
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Caroline Marty
- INSERM-UMR-1132, Hôpital Lariboisière and Université Paris Diderot, Paris, France
| | - Erwan Plougonven
- Department of Chemical Engineering, PEPs (Products, Environments, Processes), University of Liège, Liège, Belgium
| | - Sophie Dubois
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Nadia Mahli
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Angélique Léonard
- Department of Chemical Engineering, PEPs (Products, Environments, Processes), University of Liège, Liège, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Martine Cohen-Solal
- INSERM-UMR-1132, Hôpital Lariboisière and Université Paris Diderot, Paris, France
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| |
Collapse
|
36
|
Mohammadi S, Nikbakht M, Sajjadi SM, Rad F, Chahardouli B, Sabour Takanlu J, Rostami S, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Reciprocal Interactions of Leukemic Cells with Bone Marrow Stromal Cells Promote Enrichment of Leukemic Stem Cell Compartments in Response to Curcumin and Daunorubicin. Asian Pac J Cancer Prev 2017; 18:831-840. [PMID: 28441794 PMCID: PMC5464507 DOI: 10.22034/apjcp.2017.18.3.831] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A predominant challenge in developing curative leukemia therapy is interactions of leukemic cells with the bone marrow stromal microenvironment. We aimed to investigate the role of stromal cells, such as bone marrow mesenchymal stromal cells (BMSCs) and osteoblasts (OBs), in curcumin (CUR) and daunorubicin (DNR) induced apoptosis of acute myeloid leukemia (AML) cells. We used KG1 and U937 as leukemia cell line models and treated them with CUR and DNR. The cells were then co-cultured with BMSCs or a combination of BMSCs and OBs as feeders. After 24 hours of co-culture, BMSCs or OBs were sorted and separated from the leukemia cells and apoptosis levels were analyzed by annexin/propidium iodide (PI) staining on flow cytometry. Potentially involved molecular pathways were analyzed at gene and protein levels by Real time PCR and western blotting, respectively. The results showed AML cells co-cultured with BMSCs plus OBs to be more resistant to drug induced-apoptosis compared to co-culture with BMSCs alone or without co-culture. Expression levels of OPN, CXCL-12, IL-6, STAT-3 and VCAM-1 were also significantly up-regulated in OBs and AML cells, at both mRNA and protein levels after co-culture, with concurrent enrichment of CD34+ AML cells. Our data showed, in a stromal cell niche-based model, that OBs revoke the influence of BMSCs on leukemic cells and promote enrichment of both CD34+ and CD34- leukemic stem cell (LSC) compartments in response to CUR and DNR. Up-regulation of OPN, CXCL-12, IL-6, STAT-3 and VCAM-1 in OBs and AML cells in co-culture might be part of molecular mechanisms that block CUR or CUR+DNR-induced apoptosis and promote enrichment of CD34+ and CD34- LSCs.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Aberrantly expressed LGR4 empowers Wnt signaling in multiple myeloma by hijacking osteoblast-derived R-spondins. Proc Natl Acad Sci U S A 2016; 114:376-381. [PMID: 28028233 DOI: 10.1073/pnas.1618650114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The unrestrained growth of tumor cells is generally attributed to mutations in essential growth control genes, but tumor cells are also affected by, or even addicted to, signals from the microenvironment. As therapeutic targets, these extrinsic signals may be equally significant as mutated oncogenes. In multiple myeloma (MM), a plasma cell malignancy, most tumors display hallmarks of active Wnt signaling but lack activating Wnt-pathway mutations, suggesting activation by autocrine Wnt ligands and/or paracrine Wnts emanating from the bone marrow (BM) niche. Here, we report a pivotal role for the R-spondin/leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis in driving aberrant Wnt/β-catenin signaling in MM. We show that LGR4 is expressed by MM plasma cells, but not by normal plasma cells or B cells. This aberrant LGR4 expression is driven by IL-6/STAT3 signaling and allows MM cells to hijack R-spondins produced by (pre)osteoblasts in the BM niche, resulting in Wnt (co)receptor stabilization and a dramatically increased sensitivity to auto- and paracrine Wnts. Our study identifies aberrant R-spondin/LGR4 signaling with consequent deregulation of Wnt (co)receptor turnover as a driver of oncogenic Wnt/β-catenin signaling in MM cells. These results advocate targeting of the LGR4/R-spondin interaction as a therapeutic strategy in MM.
Collapse
|
38
|
Franqui-Machin R, Wendlandt EB, Janz S, Zhan F, Tricot G. Cancer stem cells are the cause of drug resistance in multiple myeloma: fact or fiction? Oncotarget 2016; 6:40496-506. [PMID: 26415231 PMCID: PMC4747348 DOI: 10.18632/oncotarget.5800] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/12/2015] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) remains a largely incurable, genetically heterogeneous plasma-cell malignancy that contains – just like many other cancers – a small fraction of clonogenic stem cell-like cells that exhibit pronounced self-renewal and differentiation capacities, but also pronounced drug resistance. These MM stem cells (MMSCs) are a controversial but highly significant issue in myeloma research because, in our opinion, they are at the root of the failure of anti-neoplastic chemotherapies to transform myeloma to a manageable chronic disease. Several markers including CD138−, ALDH1+ and SP have been used to identify MMSCs; however, no single marker is reliable for the isolation of MMSC. Nonetheless, it is now known that MMSCs depend on self-renewal and pro-survival pathways, such as AKT, Wnt/β-catenin, Notch and Hedgehog, which can be targeted with novel drugs that have shown promise in pre-clinical and clinical trials. Here, we review the pathways of myeloma “stemness”, the interactions with the bone marrow microenvironment that promote drug resistance, and the obstacles that must be overcome to eradicate MMSCs and make myeloma a curable disease.
Collapse
Affiliation(s)
- Reinaldo Franqui-Machin
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Erik B Wendlandt
- Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Siegfried Janz
- Department of Pathology, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Fenghuang Zhan
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Guido Tricot
- Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Trotter TN, Gibson JT, Sherpa TL, Gowda PS, Peker D, Yang Y. Adipocyte-Lineage Cells Support Growth and Dissemination of Multiple Myeloma in Bone. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3054-3063. [PMID: 27648615 DOI: 10.1016/j.ajpath.2016.07.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023]
Abstract
Multiple myeloma (MM) cells reside in the bone marrow microenvironment and form complicated interactions with nonneoplastic, resident stromal cells. We previously found that aggressive MM cells shift osteoblast progenitors toward adipogenesis. In addition, adipocytes are among the most common cell types in the adult skeleton; both mature adipocytes and preadipocytes serve as endocrine cells that secrete a number of soluble molecules into the microenvironment. Therefore, we used a combination of in vivo and in vitro methods to test the hypothesis that an increase in adipocyte lineage cells feeds back to promote MM progression. The results of this study revealed that bone marrow from patients with MM indeed contains increased preadipocytes and significantly larger mature adipocytes than normal bone marrow. We also found that preadipocytes and mature adipocytes secrete many molecules important for supporting MM cells in the bone marrow and directly recruit MM cells through both monocyte chemotactic protein-1 and stromal cell-derived factor-1α. Co-culture experiments found that preadipocytes activate Wnt signaling and decrease cleaved caspase-3, whereas mature adipocytes activate ERK signaling in MM cells. Furthermore, mature adipocyte conditioned medium promotes MM growth, whereas co-culture with preadipocytes results in enhanced MM cell chemotaxis in vitro and increased tumor growth in bone in vivo. Combined, these data reveal the importance of preadipocytes and mature adipocytes on MM progression and represent a unique target in the bone marrow microenvironment.
Collapse
Affiliation(s)
- Timothy N Trotter
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Justin T Gibson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tshering Lama Sherpa
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pramod S Gowda
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Deniz Peker
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yang Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
40
|
Rurali E, Bassetti B, Perrucci GL, Zanobini M, Malafronte C, Achilli F, Gambini E. BM ageing: Implication for cell therapy with EPCs. Mech Ageing Dev 2016; 159:4-13. [PMID: 27045606 DOI: 10.1016/j.mad.2016.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/22/2016] [Accepted: 04/01/2016] [Indexed: 12/17/2022]
Abstract
The bone marrow (BM) is a well-recognized source of stem/progenitor cells for cell therapy in cardiovascular diseases (CVDs). Preclinical and clinical studies suggest that endothelial progenitor cells (EPCs) contribute to reparative process of vascular endothelium and participate in angiogenesis. As for all organs and cells across the lifespan, BM and EPCs are negatively impacted by ageing due to microenvironment modifications and EPC progressive dysfunctions. The encouraging results in terms of neovascularization observed in young animals after EPC administration were mitigated in aged patients treated for ischemic CVDs. The limited efficacy of EPC-based therapy in clinical setting might be ascribed at least partly to ageing. In this review, we comprehensively discussed the age-related changes of BM and EPCs and their implication for cardiovascular cell-therapies. Finally, we examined alternative approaches under investigation to enhance EPC potency.
Collapse
Affiliation(s)
- Erica Rurali
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Marco Zanobini
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Felice Achilli
- Cardiology Department, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
41
|
New approaches to targeting the bone marrow microenvironment in multiple myeloma. Curr Opin Pharmacol 2016; 28:43-9. [PMID: 27018230 DOI: 10.1016/j.coph.2016.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 01/17/2023]
Abstract
Multiple myeloma is a tumour with a remarkably destructive effect on its host organ, the bone marrow. Through expression or secretion of adhesion molecules, growth factors, exosomes, miRNAs, chemokines and inhibitors, the tumour substantially alters its microenvironment, promoting both tumour survival and osteolytic bone disease. This altered niche is ideally suited to the sustenance of its proliferating compartment and the protection and immune evasion of its dormant, drug resistant fraction. The possibility of deepening response to a drug treatment regime, maintaining remission or even eradicating resistant stem cells by pharmacologically manipulating the tumour's interactions with this niche is a major driving force in current myeloma research. Examples of promising therapies include CXCR4 antagonists, RANKL inhibitors, HIF1α pathway inhibitors, and inhibitors of Notch, Wnt and TGFβ family pathways.
Collapse
|
42
|
Muz B, Ghazarian RN, Ou M, Luderer MJ, Kusdono HD, Azab AK. Spotlight on ixazomib: potential in the treatment of multiple myeloma. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:217-26. [PMID: 26811670 PMCID: PMC4714737 DOI: 10.2147/dddt.s93602] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite the significant therapeutic advances achieved with proteasome inhibitors (PIs) such as bortezomib and carfilzomib in prolonging the survival of patients with multiple myeloma, the development of drug resistance, peripheral neuropathy, and pharmacokinetic limitations continue to pose major challenges when using these compounds. Ixazomib is a second-generation PI with improved activity over other PIs. Unlike bortezomib and carfilzomib, which are administered by injection, ixazomib is the first oral PI approved by US Food and Drug Administration. This review discusses the biochemical properties, mechanisms of action, preclinical efficacy, and clinical trial results leading to the US Food and Drug Administration approval of ixazomib.
Collapse
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Rachel Nicole Ghazarian
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, St Louis College of Pharmacy, St Louis, MO, USA
| | - Monica Ou
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA; Department of Biology, St Louis University, St Louis, MO, USA
| | - Micah John Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Hubert Daniel Kusdono
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, St Louis College of Pharmacy, St Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St Louis School of Medicine, St Louis, MO, USA
| |
Collapse
|
43
|
Bouyssou JMC, Ghobrial IM, Roccaro AM. Targeting SDF-1 in multiple myeloma tumor microenvironment. Cancer Lett 2015; 380:315-8. [PMID: 26655999 DOI: 10.1016/j.canlet.2015.11.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a type of B-cell malignancy that remains incurable to date. The bone marrow (BM) microenvironment plays a crucial role in MM progression. The chemokine SDF-1 (CXCL12) is an important actor of the BM microenvironment that has the ability to regulate numerous processes related to its malignant transformation during MM development. The activity of SDF-1 is mainly mediated by its specific receptor CXCR4, which is expressed at the surface of MM cells and various other BM cell types. Current treatments available for MM patients mainly target tumor cells but have limited effects on the BM microenvironment. In this context, SDF-1 and CXCR4 represent ideal targets for the normalization of the MM-supportive BM microenvironment. The present review focuses on the activity of SDF-1 in the MM BM microenvironment and the current efforts carried out to target the SDF-1/CXCR4 axis for treatment of MM.
Collapse
Affiliation(s)
- Juliette M C Bouyssou
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA; INSERM UMR 1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Paris, France
| | - Irene M Ghobrial
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA
| | - Aldo M Roccaro
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood 2015; 125:3049-58. [PMID: 25838343 DOI: 10.1182/blood-2014-11-568881] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
Over the past 4 decades, basic research has provided crucial information regarding the cellular and molecular biology of cancer. In particular, the relevance of cancer microenvironment (including both cellular and noncellular elements) and the concept of clonal evolution and heterogeneity have emerged as important in cancer pathogenesis, immunologic escape, and resistance to therapy. Multiple myeloma (MM), a cancer of terminally differentiated plasma cells, is emblematic of the impact of cancer microenvironment and the role of clonal evolution. Although genetic and epigenetic aberrations occur in MM and evolve over time under the pressure of exogenous stimuli, they are also largely present in premalignant plasma cell dyscrasia such as monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM), suggesting that genetic mutations alone are necessary, but not sufficient, for myeloma transformation. The role of bone marrow microenvironment in mediating survival, proliferation, and resistance to therapy in myeloma is well established; and although an appealing speculation, its role in fostering the evolution of MGUS or SMM into MM is yet to be proven. In this review, we discuss MM pathogenesis with a particular emphasis on the role of bone marrow microenvironment.
Collapse
|