1
|
Gardano L, Ferreira J, Le Roy C, Ledoux D, Varin-Blank N. The survival grip-how cell adhesion promotes tumor maintenance within the microenvironment. FEBS Lett 2024. [PMID: 39704141 DOI: 10.1002/1873-3468.15074] [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: 07/23/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Cell adhesion is warranted by proteins that are crucial for the maintenance of tissue integrity and homeostasis. Most of these proteins behave as receptors to link adhesion to the control of cell survival and their expression or regulation are often altered in cancers. B-cell malignancies do not evade this principle as they are sustained in relapsed niches by interacting with the microenvironment that includes cells and their secreted factors. Focusing on chronic lymphocytic leukemia and mantle cell lymphoma, this Review delves with the molecules involved in the dialog between the adhesion platforms and signaling pathways known to regulate both cell adhesion and survival. Current therapeutic strategies disrupt adhesive structures and compromise the microenvironment support to tumor cells, rendering them sensitive to immune recognition. The development of organ-on-chip and 3D culture systems, such as spheroids, have revealed the importance of mechanical cues in regulating signaling pathways to organize cell adhesion and survival. All these elements contribute to the elaboration of the crosstalk of lymphoma cells with the microenvironment and the education processes that allow the establishment of the supportive niche.
Collapse
Affiliation(s)
- Laura Gardano
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Jordan Ferreira
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Christine Le Roy
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Dominique Ledoux
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Nadine Varin-Blank
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
2
|
das Neves GM, Kagami LP, Battastini AMO, Figueiró F, Eifler-Lima VL. Targeting ecto-5'-nucleotidase: A comprehensive review into small molecule inhibitors and expression modulators. Eur J Med Chem 2023; 247:115052. [PMID: 36599229 DOI: 10.1016/j.ejmech.2022.115052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023]
Abstract
The purinergic signaling has drawn attention from academia and more recently from pharmaceutical industries as a potential therapeutic route for cancer treatment, since ATP may act as chemotactic agent and possess in vitro antineoplastic activity. On the other way, adenosine, produced in extracellular medium by ecto-5'-NT, acts as immunosuppressor and is related to neoangiogenesis, vasculogenesis and evasion to the immune system. Consequently, inhibitors of ecto-5'-NT may prevent tumor progression, reducing adenosine concentrations, preventing escape from the host's immune system and slowing cancer's growth. This review aims to highlight important biochemical and structural features of ecto-5'NT, highlight its expression profile in normal and cancer cell lines detailing compounds which may act as expression regulators and to review the several classes of ecto-5'NT inhibitors developed in the past 12 years, in order to build a general structure-activity relationship model to guide further compound design.
Collapse
Affiliation(s)
- Gustavo Machado das Neves
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Luciano Porto Kagami
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana Maria Oliveira Battastini
- Laboratório de Imunobioquímica do Câncer (LIBC), Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer (LIBC), Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vera Lucia Eifler-Lima
- Laboratório de Síntese Orgânica Medicinal (LaSOM), Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
3
|
Guo H, Yang J, Wang H, Liu X, Liu Y, Zhou K. Reshaping the tumor microenvironment: The versatility of immunomodulatory drugs in B-cell neoplasms. Front Immunol 2022; 13:1017990. [PMID: 36311747 PMCID: PMC9596992 DOI: 10.3389/fimmu.2022.1017990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) such as thalidomide, lenalidomide and pomalidomide are antitumor compounds that have direct tumoricidal activity and indirect effects mediated by multiple types of immune cells in the tumor microenvironment (TME). IMiDs have shown remarkable therapeutic efficacy in a set of B-cell neoplasms including multiple myeloma, B-cell lymphomas and chronic lymphocytic leukemia. More recently, the advent of immunotherapy has revolutionized the treatment of these B-cell neoplasms. However, the success of immunotherapy is restrained by immunosuppressive signals and dysfunctional immune cells in the TME. Due to the pleiotropic immunobiological properties, IMiDs have shown to generate synergetic effects in preclinical models when combined with monoclonal antibodies, immune checkpoint inhibitors or CAR-T cell therapy, some of which were successfully translated to the clinic and lead to improved responses for both first-line and relapsed/refractory settings. Mechanistically, despite cereblon (CRBN), an E3 ubiquitin ligase, is considered as considered as the major molecular target responsible for the antineoplastic activities of IMiDs, the exact mechanisms of action for IMiDs-based TME re-education remain largely unknown. This review presents an overview of IMiDs in regulation of immune cell function and their utilization in potentiating efficacy of immunotherapies across multiple types of B-cell neoplasms.
Collapse
Affiliation(s)
| | | | | | | | | | - Keshu Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
4
|
Fregnani A, Saggin L, Gianesin K, Quotti Tubi L, Carraro M, Barilà G, Scapinello G, Bonetto G, Pesavento M, Berno T, Branca A, Gurrieri C, Zambello R, Semenzato G, Trentin L, Manni S, Piazza F. CK1α/RUNX2 Axis in the Bone Marrow Microenvironment: A Novel Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14174173. [PMID: 36077711 PMCID: PMC9454895 DOI: 10.3390/cancers14174173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is an incurable disease for which novel therapeutic approaches targeting the malignant cells and the associated bone disease are urgently needed. CK1α is a protein kinase that plays a crucial role in the signaling network that sustains plasma cell (PC) survival and bone disease. This protein regulates Wnt/β-catenin signaling, which is fundamental for both MM cell survival and mesenchymal stromal cell (MSC) osteogenic differentiation. In this study, we investigated its involvement in MM–MSC cross-talk. We found that, by lowering CK1α expression levels in co-cultures of MM and MSC cells, expression of RUNX2—the master regulator of osteogenic differentiation—was regulated differently in the two cell types. Our data suggest the possibility of using a specific CK1α inhibitor as part of a novel therapeutic approach to selectively kill malignant PCs and overcome the blocking of osteogenic differentiation induced by MM cells in MSCs. Abstract Multiple myeloma (MM) is a malignant plasma cell (PC) neoplasm, which also displays pathological bone involvement. Clonal expansion of MM cells in the bone marrow causes a perturbation of bone homeostasis that culminates in MM-associated bone disease (MMABD). We previously demonstrated that the S/T kinase CK1α sustains MM cell survival through the activation of AKT and β-catenin signaling. CK1α is a negative regulator of the Wnt/β-catenin cascade, the activation of which promotes osteogenesis by directly stimulating the expression of RUNX2, the master gene regulator of osteoblastogenesis. In this study, we investigated the role of CK1α in the osteoblastogenic potential of mesenchymal stromal cells (MSCs) and its involvement in MM–MSC cross-talk. We found that CK1α silencing in in vitro co-cultures of MMs and MSCs modulated RUNX2 expression differently in PCs and in MSCs, mainly through the regulation of Wnt/β-catenin signaling. Our findings suggest that the CK1α/RUNX2 axis could be a potential therapeutic target for constraining malignant PC expansion and supporting the osteoblastic transcriptional program of MSCs, with potential for ameliorating MMABD. Moreover, considering that Lenalidomide treatment leads to MM cell death through Ikaros, Aiolos and CK1α proteasomal degradation, we examined its effects on the osteoblastogenic potential of MSC compartments.
Collapse
Affiliation(s)
- Anna Fregnani
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Lara Saggin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Ketty Gianesin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Laura Quotti Tubi
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Marco Carraro
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Gregorio Barilà
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Greta Scapinello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Giorgia Bonetto
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Maria Pesavento
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Tamara Berno
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Antonio Branca
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Carmela Gurrieri
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Renato Zambello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Gianpietro Semenzato
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Sabrina Manni
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| | - Francesco Piazza
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| |
Collapse
|
5
|
Ioannou N, Jain K, Ramsay AG. Immunomodulatory Drugs for the Treatment of B Cell Malignancies. Int J Mol Sci 2021; 22:8572. [PMID: 34445275 PMCID: PMC8395307 DOI: 10.3390/ijms22168572] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence suggests that the tumor microenvironment (TME) is involved in disease progression and drug resistance in B cell malignancies, by supporting tumor growth and facilitating the ability of malignant cells to avoid immune recognition. Immunomodulatory drugs (IMiDs) such as lenalidomide have some direct anti-tumor activity, but critically also target various cellular compartments of the TME including T cells, NK cells, and stromal cells, which interfere with pro-tumor signaling while activating anti-tumor immune responses. Lenalidomide has delivered favorable clinical outcomes as a single-agent, and in combination therapy leads to durable responses in chronic lymphocytic leukemia (CLL) and several non-Hodgkin lymphomas (NHLs) including follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), and mantle cell lymphoma (MCL). Recently, avadomide, a next generation cereblon E3 ligase modulator (CELMoD), has shown potent anti-tumor and TME immunomodulatory effects, as well as promising clinical efficacy in DLBCL. This review describes how the pleiotropic effects of IMiDs and CELMoDs could make them excellent candidates for combination therapy in the immuno-oncology era-a concept supported by preclinical data, as well as the recent approval of lenalidomide in combination with rituximab for the treatment of relapsed/refractory (R/R) FL.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/immunology
- Antineoplastic Agents/therapeutic use
- Enzyme Inhibitors/therapeutic use
- Humans
- Immunologic Factors/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/immunology
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/immunology
Collapse
Affiliation(s)
| | | | - Alan G. Ramsay
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK; (N.I.); (K.J.)
| |
Collapse
|
6
|
Mesenchymal Stem Cells in Aplastic Anemia and Myelodysplastic Syndromes: The "Seed and Soil" Crosstalk. Int J Mol Sci 2020; 21:ijms21155438. [PMID: 32751628 PMCID: PMC7432231 DOI: 10.3390/ijms21155438] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
There is growing interest in the contribution of the marrow niche to the pathogenesis of bone marrow failure syndromes, i.e., aplastic anemia (AA) and myelodysplastic syndromes (MDSs). In particular, mesenchymal stem cells (MSCs) are multipotent cells that contribute to the organization and function of the hematopoietic niche through their repopulating and supporting abilities, as well as immunomodulatory properties. The latter are of great interest in MDSs and, particularly, AA, where an immune attack against hematopoietic stem cells is the key pathogenic player. We, therefore, conducted Medline research, including all available evidence from the last 10 years concerning the role of MSCs in these two diseases. The data presented show that MSCs display morphologic, functional, and genetic alterations in AA and MDSs and contribute to immune imbalance, ineffective hematopoiesis, and leukemic evolution. Importantly, adoptive MSC infusion from healthy donors can be exploited to heal the "sick" niche, with even better outcomes if cotransplanted with allogeneic hematopoietic stem cells. Finally, future studies on MSCs and the whole microenvironment will further elucidate AA and MDS pathogenesis and possibly improve treatment.
Collapse
|
7
|
Pan Y, Zhao A, Zhong Z, Pan X, Cai S. Ganoderma spore lipid protects mouse bone marrow mesenchymal stem cells and hematopoiesis from the cytotoxicity of the chemotherapeutic agent. Biotechnol Prog 2019; 35:e2869. [PMID: 31207156 DOI: 10.1002/btpr.2869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
Cancer chemotherapeutic agents are frequently toxic to bone marrow and impair bone marrow functions. It is unclear whether ganoderma spore lipid (GSL) can protect bone marrow cells from the cytotoxicity of chemotherapy. To investigate the protective effects of GSL on bone marrow mesenchymal stem cells (MSCs) and hematopoiesis, we examined the effects of GSL on MSCs in vitro and hematopoiesis in vivo after treatment with the chemotherapeutic agent cyclophosphamide. MSCs and peripheral blood cells were isolated and counted from the bone marrow of normal mice were pre-treated with GSL before CTX treatment or co-treated with GSL and CTX, followed by examining the changes in phenotype, morphology, proliferation, apoptosis, and differentiation potentials. The results showed that GSL could reduce the CTX-induced changes in the phenotype of MSCs and maintain the elongated fibroblast-like morphology. MTT and annexin V/propidium iodide (PI) analyses found that GSL pre-treatment and co-treatment increased the proliferation and decreased the apoptosis in CTX-treated MSCs. Furthermore, GSL improved the osteogenic and adipogenic differentiation potentials of CTX-treated MSCs. In vivo, GSL treatment increased the number of peripheral blood cells including white blood cells (WBC) and platelets (PLT) in the CTX-treated mice and enhanced the in vitro formation of hematopoietic lineage colonies (erythrocyte colony forming unit, CFU-E; erythroid burst-forming units, BFU-E; and granulocyte macrophage colony-forming units, CFU-GM) from bone marrow cells in these mice. These findings suggest GSL could protect MSCs and hematopoiesis from the cytotoxicity of CTX and might become an effective adjuvant to attenuate side effects of chemotherapy during cancer treatment.
Collapse
Affiliation(s)
- Yu Pan
- Department of Trauma and Orthopedics, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen Baoan Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Andong Zhao
- Department of Trauma and Orthopedics, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen Baoan Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhiqiang Zhong
- Department of Oncology, The 1st Affiliate Hospital of Dalian Medical University, Dalian, China
| | - Xiaohua Pan
- Department of Trauma and Orthopedics, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen Baoan Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Sa Cai
- Department of Trauma and Orthopedics, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen Baoan Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
8
|
Götze KS, Platzbecker U. Old Dogs, New Tricks: Revisiting Immune Modulatory Approaches for Myelodysplastic Syndromes. Hemasphere 2018; 2:e162. [PMID: 31723800 PMCID: PMC6745963 DOI: 10.1097/hs9.0000000000000162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/17/2018] [Indexed: 01/21/2023] Open
Affiliation(s)
- Katharina S. Götze
- Department of Medicine III, Technische Universität München, Munich, Germany
- Deutsche MDS Gruppe (D-MDS)
- German Cancer Consortium (DKTK) and German Cancer Center (DKFZ) Germany, Partner site Munich
| | - Uwe Platzbecker
- Deutsche MDS Gruppe (D-MDS)
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
- EHA-SWG on MDS and EMSCO (www.emsco.eu)
| |
Collapse
|
9
|
Xu L, Hu YX, Li YC, Zhang L, Ai HX, Liu YF, Liu HS. In vitro DNA binding studies of lenalidomide using spectroscopic in combination with molecular docking techniques. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2017.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
10
|
Guo J, Fei C, Zhao Y, Zhao S, Zheng Q, Su J, Wu D, Li X, Chang C. Lenalidomide restores the osteogenic differentiation of bone marrow mesenchymal stem cells from multiple myeloma patients via deactivating Notch signaling pathway. Oncotarget 2017; 8:55405-55421. [PMID: 28903429 PMCID: PMC5589668 DOI: 10.18632/oncotarget.19265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/24/2017] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) always presents osteolytic bone lesions, resulting from the abnormal osteoblastic and osteoclastic function in patients. MM patients exhibit the impairment of osteogenic differentiation of BMMSCs (bone marrow mesenchymal stem cells) and osteoblast deficiency. Effects of the drug, lenalidomide on the osteoblastic functions and the involved mechanisms remain unexplored. In the present study, it is observed that the osteogenic differentiation of BMMSCs from MM patients (MM-MSCs) is impaired and activation of Notch signaling pathway in MM-MSCs is abnormal. Notch signaling activation inhibits BMMSCs osteogenesis. Knockdown of Notch1 expression and DAPT application reverse the osteogenic differentiation from MM-MSCs. Furthermore, it is shown that the gene expression of Notch signaling molecules, including receptors, ligands and downstream factors are significantly decreased in MM-MSCs following lenalidomide treatment, compared with non-treated MM-MSCs. Taken together, treatment with lenalidomide restores the osteogenic differentiation of MM-MSCs via deactivating Notch signaling pathway.
Collapse
Affiliation(s)
- Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chengming Fei
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Youshan Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Sida Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Qingqing Zheng
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jiying Su
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Dong Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
11
|
Schroeder T, Geyh S, Germing U, Haas R. Mesenchymal stromal cells in myeloid malignancies. Blood Res 2016; 51:225-232. [PMID: 28090484 PMCID: PMC5234241 DOI: 10.5045/br.2016.51.4.225] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal myeloid disorders characterized by hematopoietic insufficiency. As MDS and AML are considered to originate from genetic and molecular defects of hematopoietic stem and progenitor cells (HSPC), the main focus of research in this field has focused on the characterization of these cells. Recently, the contribution of BM microenvironment to the pathogenesis of myeloid malignancies, in particular MDS and AML has gained more interest. This is based on a better understanding of its physiological role in the regulation of hematopoiesis. Additionally, it was demonstrated as a ‘proof of principle’ that genetic disruption of cells of the mesenchymal or osteoblastic lineage can induce MDS, MPS or AML in mice. In this review, we summarize the current knowledge about the contribution of the BM microenvironment, in particular mesenchymal stromal cells (MSC) to the pathogenesis of AML and MDS. Furthermore, potential models integrating the BM microenvironment into the pathophysiology of these myeloid disorders are discussed. Finally, strategies to therapeutically exploit this knowledge and to interfere with the crosstalk between clonal hematopoietic cells and altered stem cell niches are introduced.
Collapse
Affiliation(s)
- Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Düesseldorf, Germany
| | - Stefanie Geyh
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Düesseldorf, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Düesseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Düesseldorf, Germany
| |
Collapse
|
12
|
Wilk CM, Heinzler N, Boquoi A, Cadeddu RP, Strapatsas T, Dienst A, Majidi F, Deenen R, Bruns I, Schroeder T, Köhrer K, Haas R, Kobbe G, Fenk R. Lenalidomide consolidation treatment in patients with multiple myeloma suppresses myelopoieses but spares erythropoiesis. Int J Cancer 2016; 139:2343-52. [PMID: 27389073 DOI: 10.1002/ijc.30257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 06/07/2016] [Indexed: 12/15/2022]
Abstract
New drugs for the treatment of multiple myeloma (MM) comprise immunomodulatory substances such as lenalidomide and related compounds. While lenalidomide has found its way into first-line treatment as well as into relapse therapy, little is known about lenalidomide effects on normal hematopoietic stem and progenitor cells (HSPCs). In this study, we investigated whether HSPCs are influenced by lenalidomide on a phenotypic, functional and gene expression level. For that purpose, samples from patients with MM were obtained who underwent equivalent first-line treatment including induction therapy, cytotoxic stem cell mobilization and high-dose melphalan therapy followed by autologous blood stem cell transplantation and a subsequent uniform lenalidomide consolidation treatment within a prospective clinical trial. We found that after six months of lenalidomide therapy, the number of CD34(+) HSPCs decreased. Additionally, lenalidomide affects the numerical composition of hematopoietic cells in the bone marrow while it does not affect long-term HSPC proliferation in vitro. We found a significant amplification of fetal hemoglobin (HbF) expression on a transcriptional level and can confirm a stimulated erythropoiesis on a phenotypic level. These effects were accompanied by silencing of the TGF-β signaling pathway on the gene expression and protein level that is known to be amplified in active MM. However, these pleiotropic effects gave no evidence for mutagenic potential. In conclusion, lenalidomide does not exert long-term effects on proliferation of HSPCs but instead promotes erythropoiesis by shifting hemoglobin expression toward HbF and by silencing the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Christian Matthias Wilk
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.,Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Niklas Heinzler
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Amelie Boquoi
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Ron-Patrick Cadeddu
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Tobias Strapatsas
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.,Division of Emergency Care, Helios Klinikum Wuppertal, Wuppertal, Germany
| | - Ariane Dienst
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Fatemeh Majidi
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - René Deenen
- Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Ingmar Bruns
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Karl Köhrer
- Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
13
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
14
|
Kim HY, Moon JY, Ryu H, Choi YS, Song IC, Lee HJ, Yun HJ, Kim S, Jo DY. Bortezomib inhibits the survival and proliferation of bone marrow stromal cells. Blood Res 2015; 50:87-96. [PMID: 26157778 PMCID: PMC4486164 DOI: 10.5045/br.2015.50.2.87] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/24/2015] [Accepted: 05/27/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Bortezomib is widely used for the treatment of multiple myeloma. Bone marrow stromal cells (BMSCs) endow myeloma cells with survival and growth advantages. However, the influence of bortezomib on BMSCs is not well elucidated. We examined the effects of bortezomib on the survival and growth of BMSCs in vitro. METHODS The effects of bortezomib on the survival and proliferation of the BMSC MS-5 cell line and on BMSCs obtained from healthy individuals (N=4) and newly diagnosed myeloma patients (N=5) were investigated in vitro. Transmembrane cell migration was evaluated using the Transwell system. A short interfering RNA strategy was used to knock down the expression of chemokine (CXC motif) ligand 12 (CXCL12) mRNA. To examine the effects of bortezomib-exposed BMSCs on the migration and localization of myeloma cells, MS-5 monolayers were treated with bortezomib for 24 hr, washed, and then overlaid with human RPMI8226 myeloma cells. RESULTS Bortezomib inhibited BMSC proliferation in a concentration-dependent manner, and induced cellular apoptosis. Bortezomib decreased CXCL12 production by BMSCs. Knockdown of CXCL12 mRNA in BMSCs revealed that CXCL12 served as an autocrine growth factor. Short-term bortezomib treatment of BMSC monolayers reduced the tendency of myeloma cells to locate to positions under the monolayers. CONCLUSION Bortezomib inhibits the survival and growth of BMSCs via downregulation of CXCL12, which may contribute to the clinical effects of this agent.
Collapse
Affiliation(s)
- Ha-Yon Kim
- Department of Drug Activity, New Drug Development Center, Medical Innovation Foundation, Osong, Korea
| | - Ji-Young Moon
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Haewon Ryu
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Yoon-Seok Choi
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Ik-Chan Song
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Hyo-Jin Lee
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Hwan-Jung Yun
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Samyong Kim
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Deog-Yeon Jo
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
15
|
Copland IB, Qayed M, Garcia MA, Galipeau J, Waller EK. Bone Marrow Mesenchymal Stromal Cells from Patients with Acute and Chronic Graft-versus-Host Disease Deploy Normal Phenotype, Differentiation Plasticity, and Immune-Suppressive Activity. Biol Blood Marrow Transplant 2015; 21:934-40. [DOI: 10.1016/j.bbmt.2015.01.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/13/2015] [Indexed: 01/15/2023]
|
16
|
The tumor microenvironment shapes hallmarks of mature B-cell malignancies. Oncogene 2015; 34:4673-82. [PMID: 25639873 DOI: 10.1038/onc.2014.403] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
B-cell tumorigenesis results from a host of known and unknown genetic anomalies, including non-random translocations of genes that normally function as determinants of cell proliferation or cell survival to regions juxtaposed to active immunoglobulin heavy chain enhancer elements, chromosomal aneuploidy, somatic mutations that further affect oncogenic signaling and loss of heterozygosity of tumor-suppressor genes. However, it is critical to recognize that even in the setting of a genetic disease, the B-cell/plasma cell tumor microenvironment (TME) contributes significantly to malignant transformation and pathogenesis. Over a decade ago, we proposed the concept of cell adhesion-mediated drug resistance to delineate a form of TME-mediated drug resistance that protects hematopoietic tumor cells from the initial effect of diverse therapies. In the interim, it has been increasingly appreciated that TME also contributes to tumor initiation and progression through sustained growth/proliferation, self-renewal capacity, immune evasion, migration and invasion as well as resistance to cell death in a host of B-cell malignancies, including mantle cell lymphoma, diffuse large B-cell lymphoma, Waldenstroms macroglobulinemia, chronic lymphocytic leukemia and multiple myeloma. Within this review, we propose that TME and the tumor co-evolve as a consequence of bidirectional signaling networks. As such, TME represents an important target and should be considered integral to tumor progression and drug response.
Collapse
|
17
|
DiNardo CD, Daver N, Jabbour E, Kadia T, Borthakur G, Konopleva M, Pemmaraju N, Yang H, Pierce S, Wierda W, Bueso-Ramos C, Patel KP, Cortes JE, Ravandi F, Kantarjian HM, Garcia-Manero G. Sequential azacitidine and lenalidomide in patients with high-risk myelodysplastic syndromes and acute myeloid leukaemia: a single-arm, phase 1/2 study. LANCET HAEMATOLOGY 2014; 2:e12-20. [PMID: 26687423 DOI: 10.1016/s2352-3026(14)00026-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/17/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND The standard of care for myelodysplastic syndromes is hypomethylating agents such as azacitidine. However, responses to azacitidine are generally temporary, and outcomes after hypomethylating agent failure are dismal. Therefore, the development of more effective treatments is crucial to improve outcomes in patients with myelodysplastic syndromes. We aimed to assess azacitidine and lenalidomide in patients with high-risk myelodysplastic syndromes and acute myeloid leukaemia. METHODS We did this single-arm phase 1/2 study at the University of Texas MD Anderson Cancer Center, TX, USA. Patients of any age were eligible for phase 1 and 2a if they had relapsed or refractory acute myeloid leukaemia or myelodysplastic syndrome with bone marrow blasts more than 10%. For phase 2b, eligible participants were previously untreated with myelodysplastic syndrome with an International Prognostic Scoring System (IPSS) score of intermediate-1 or higher with up to 30% blasts. All participants received 75 mg/m(2) azacitidine once a day for days 1-5 for each 28 day cycle. We gave patients oral lenalidomide for 5 or 10 days starting on day 6. We assessed seven lenalidomide doses in a 3 + 3 phase 1 design (n=28). The primary endpoint in phase 1 was the maximum tolerated dose, and the primary endpoint in phase 2 was overall survival. Outcome analyses were by intention to treat. This study is registered with ClinicalTrials.gov, number NCT01038635. FINDINGS Between Dec 30, 2009, and June, 17, 2013, we enrolled 88 patients (28 in phase 1 and 60 in phase 2). One patient unexpectedly died in the phase 1 study at the highest dose level, six more patients were recruited with no further serious adverse events. We recorded no dose-limiting toxic effects, and the maximum tolerated dose of lenalidomide in combination with azacitidine in patients with acute myeloid leukaemia and myelodysplastic syndrome was initially established at 50 mg per day for 10 days. In the first 20 patients in phase 2, we noted a high rate of myelosuppression and myelosuppression-related toxic effects; therefore, we amended the lenalidomide dose to 25 mg per day for 5 days. We also adjusted the inclusion criteria to include patients with less than 30% blasts to focus mainly on patients with myelodysplastic syndromes. Median overall survival was 75 weeks (IQR 25-not reached) for the 40 patients in phase 2b. The most common grade 3-4 adverse events overall were neutropenic fever (n=27) and pneumonia (n=18). INTERPRETATION We have identified a safe and active sequential treatment combination of azacitidine and lenalidomide for patient with myelodysplastic syndrome and have preliminary evidence that this dose is also safe for patients with acute myeloid leukaemia. FUNDING MD Anderson Cancer Center and Celgene.
Collapse
Affiliation(s)
- Courtney D DiNardo
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yang
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry Pierce
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Wierda
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Bueso-Ramos
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jorge E Cortes
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukaemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
18
|
Endothelium-mediated survival of leukemic cells and angiogenesis-related factors are affected by lenalidomide treatment in chronic lymphocytic leukemia. Exp Hematol 2014; 42:126-36.e1. [DOI: 10.1016/j.exphem.2013.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/27/2013] [Accepted: 10/27/2013] [Indexed: 12/12/2022]
|
19
|
Abstract
For many years there has been considerable disassociation between the understood biology of chronic lymphocytic leukemia (CLL) and the therapeutics used to treat this disease. With the introduction of the first targeted CD20 antibody rituximab and its addition to chemotherapy came the first observation that minimal residual disease-negative (MRD-negative) complete responses (CRs) could be obtained with dramatically improved progression-free survival and overall survival. This advance was soon to be surpassed by the introduction of therapeutics that target B-cell receptor (BCR) signaling. New data show that BCR-inhibiting agents are very active for the treatment of relapsed CLL, despite the lack of MRD-negative CR, with durability of response being considerably more impressive than previously observed with other agents not producing MRD-negative CRs. This perspective provides a view of where these agents may take us in the future as CLL therapy evolves with this exciting new class of drugs.
Collapse
|
20
|
Amé-Thomas P, Tarte K. The yin and the yang of follicular lymphoma cell niches: role of microenvironment heterogeneity and plasticity. Semin Cancer Biol 2013; 24:23-32. [PMID: 23978491 DOI: 10.1016/j.semcancer.2013.08.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/31/2013] [Accepted: 08/15/2013] [Indexed: 12/20/2022]
Abstract
Follicular lymphoma (FL) results from the malignant transformation of germinal center B cells and is characterized by recurrent genetic alterations providing a direct growth advantage or facilitating interaction with tumor microenvironment. In agreement, accumulating evidences suggest a dynamic bidirectional crosstalk between FL B cells and surrounding non-malignant cells within specialized tumor niches in both invaded lymph nodes and bone marrow. Infiltrating stromal cells, macrophages, and T/NK cell subsets either contribute to anti-tumor immune response, or conversely form a tumor supportive network promoting FL B cell survival, growth, and drug resistance. This review depicts the phenotypic heterogeneity and functional plasticity of the most important FL cell partners and describes their complex interplay. We also unravel how malignant B cells recruit and subvert accessory immune and stromal cells to trigger their polarization toward a supportive phenotype. Based on these observations, innovative therapeutic approaches have been recently proposed, in order to benefit from local anti-tumor immunity and/or to selectively target the protective cell niche.
Collapse
Affiliation(s)
- Patricia Amé-Thomas
- INSERM, UMR U917, Equipe Labellisée Ligue Contre le Cancer, Faculté de Médecine, Rennes, France; Université Rennes 1, Rennes, France; CHU de Rennes, Hôpital Pontchaillou, Service ITeCH, Pôle de Biologie, Rennes, France
| | - Karin Tarte
- INSERM, UMR U917, Equipe Labellisée Ligue Contre le Cancer, Faculté de Médecine, Rennes, France; Université Rennes 1, Rennes, France; CHU de Rennes, Hôpital Pontchaillou, Service ITeCH, Pôle de Biologie, Rennes, France; Etablissement Français du Sang Bretagne, Rennes, France.
| |
Collapse
|
21
|
Thalidomide corrects impaired mesenchymal stem cell function in inducing tolerogenic DCs in patients with immune thrombocytopenia. Blood 2013; 122:2074-82. [PMID: 23926306 DOI: 10.1182/blood-2013-03-491555] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Thalidomide (THD) is an immunomodulatory agent used to treat immune-mediated diseases. Immune thrombocytopenia (ITP) is an autoimmune disorder in which impaired mesenchymal stem cells (MSCs) are potentially involved. We demonstrated that MSCs in ITP patients had reduced proliferative capacity and lost their immunosuppressive function, which could be corrected with THD treatment. According to the gene profile, the downregulation of caspase-8 and caspase-10, and upregulation of oct3/4 and tgf-β1, may be associated with THD modulation. Dendritic cells (DCs) played an important role in mediating the inhibitory activity of MSCs. To study the functional alteration of DCs elicited by MSCs, we sorted DCs after incubation with MSCs and performed T-lymphocyte reaction assays. The THD-modulated MSCs from ITP patients induced mature DCs to become tolerogenic DCs, whereas unmodulated MSCs had no effect. The induction of tolerogenicity in DCs by MSCs was dependent on the expression of TIEG1 in DCs. The study reveals the inability of MSCs from ITP patients to induce tolerogenic ability in DCs. THD could restore the regulatory effect of MSCs on DCs. These findings will help us understand the pathogenesis of ITP, and with appropriate safeguards, THD may benefit patients with ITP.
Collapse
|
22
|
Ferrer RA, Wobus M, List C, Wehner R, Schönefeldt C, Brocard B, Mohr B, Rauner M, Schmitz M, Stiehler M, Ehninger G, Hofbauer LC, Bornhäuser M, Platzbecker U. Mesenchymal stromal cells from patients with myelodyplastic syndrome display distinct functional alterations that are modulated by lenalidomide. Haematologica 2013; 98:1677-85. [PMID: 23716561 DOI: 10.3324/haematol.2013.083972] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The contribution of the bone marrow microenvironment in myelodysplastic syndrome is controversial. We therefore analyzed the functional properties of primary mesenchymal stromal cells from patients with myelodysplastic syndrome in the presence or absence of lenalidomide. Compared to healthy controls, clonality and growth were reduced across all disease stages. Furthermore, differentiation defects and particular expression of adhesion and cell surface molecules (e.g. CD166, CD29, CD146) were detected. Interestingly, the levels of stromal derived factor 1-alpha in patients' cells culture supernatants were almost 2-fold lower (P<0.01) than those in controls and this was paralleled by a reduced induction of migration of CD34(+) hematopoietic cells. Co-cultures of mesenchymal stromal cells from patients with CD34(+) cells from healthy donors resulted in reduced numbers of cobblestone area-forming cells and fewer colony-forming units. Exposure of stromal cells from patients and controls to lenalidomide led to a further reduction of stromal derived factor 1-alpha secretion and cobblestone area formation, respectively. Moreover, lenalidomide pretreatment of mesenchymal stromal cells from patients with low but not high-risk myelodysplastic syndrome was able to rescue impaired erythroid and myeloid colony formation of early hematopoietic progenitors. In conclusion, our analyses support the notion that the stromal microenvironment is involved in the pathophysiology of myelodysplastic syndrome thus representing a potential target for therapeutic interventions.
Collapse
|
23
|
Platzbecker U, Germing U. Combination of azacitidine and lenalidomide in myelodysplastic syndromes or acute myeloid leukemia-a wise liaison? Leukemia 2013; 27:1813-9. [PMID: 23644421 DOI: 10.1038/leu.2013.140] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 04/15/2013] [Accepted: 04/17/2013] [Indexed: 01/19/2023]
Abstract
Treatment options for older patients with advanced myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) are limited and the prognosis remains poor, thereby warranting development of novel therapies. Aberrant epigenetic modifications, including altered DNA methylation, seem to contribute to the pathogenesis of these patients. In fact, hypomethylating agents (HMA) like azacitidine have been successfully used in clinical trials and achieved approval from health authorities. There is now growing evidence suggesting that the combination of drugs with different mechanisms of action might offer a potential benefit to these patients. This is especially done with the intention to synergize the positive effects of each drug on the defective hematopoiesis while sparing potential side effects and toxicities. Combination of HMA with histone deacetylase inhibitors, although mechanistically very tempting, have not yielded convincing improvement of the results in the majority of trials compared to single agent HMA treatment. Currently, combination therapies of azacitidine with lenalidomide appear to be promising thus making them an appealing option for treatment in these patients.
Collapse
|
24
|
Scherzed A, Hackenberg S, Froelich K, Rak K, Technau A, Radeloff A, Nöth U, Koehler C, Hagen R, Kleinsasser N. Effects of salinomycin on human bone marrow-derived mesenchymal stem cells in vitro. Toxicol Lett 2013; 218:207-14. [PMID: 23410960 DOI: 10.1016/j.toxlet.2013.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/29/2013] [Accepted: 02/01/2013] [Indexed: 01/22/2023]
Abstract
Various hypotheses on the origin of cancer stem cells (CSCs) exist, including that CSCs develop from transformed human bone marrow mesenchymal stem cells (hBMSC). Since the polyether antibiotic salinomycin selectively kills CSCs, the present study aims to elucidate the effects of salinomycin on normal hBMSC. The immunophenotype of hBMSC after salinomycin exposure was observed by flow cytometry. The multi-differentiation capacity of hBMSC was evaluated by Oil Red O and van Kossa staining. Cytotoxic effects of salinomycin were monitored by the [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] (MTT) assay. Furthermore, spheroid formation and migration capacity were assessed. There were no differences in the immunophenotype and multi-differentiation capacity of hBMSC induced by salinomycin treatment. Cytotoxic effects were observed at concentrations of 30 μM and above. Neither the migration capability nor the ability to form spheroids was affected. Essential functional properties of hBMSC were unaffected by salinomycin. However, dose-dependent cytotoxicity effects could be observed. Overall, low dose salinomycin showed no negative effects on hBMSC. Since mesenchymal stem cells from various sources respond differently, further in vitro studies are needed to clarify the effect of salinomycin on tissue-specific stem cells.
Collapse
Affiliation(s)
- A Scherzed
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mourcin F, Pangault C, Amin-Ali R, Amé-Thomas P, Tarte K. Stromal cell contribution to human follicular lymphoma pathogenesis. Front Immunol 2012; 3:280. [PMID: 22973275 PMCID: PMC3433684 DOI: 10.3389/fimmu.2012.00280] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/16/2012] [Indexed: 12/19/2022] Open
Abstract
Follicular lymphoma (FL) is the prototypical model of indolent B cell lymphoma displaying a strong dependence on a specialized cell microenvironment mimicking normal germinal center. Within malignant cell niches in invaded lymph nodes and bone marrow, external stimuli provided by infiltrating stromal cells make a pivotal contribution to disease development, progression, and drug resistance. The crosstalk between FL B cells and stromal cells is bidirectional, causing activation of both partners. In agreement, FL stromal cells exhibit specific phenotypic, transcriptomic, and functional properties. This review highlights the critical pathways involved in the direct tumor-promoting activity of stromal cells but also their role in the organization of FL cell niche through the recruitment of accessory immune cells and their polarization to a B cell supportive phenotype. Finally, deciphering the interplay between stromal cells and FL cells provides potential new therapeutic targets with the aim to mobilize malignant cells outside their protective microenvironment and increase their sensitivity to conventional treatment.
Collapse
|