1
|
Colombo F, Guzzeloni V, Kizilirmak C, Brambilla F, Garcia-Manteiga JM, Tascini AS, Moalli F, Mercalli F, Ponzoni M, Mezzapelle R, Ferrarini M, Ferrero E, Visone R, Rasponi M, Bianchi ME, Zambrano S, Agresti A. In vitro models of the crosstalk between multiple myeloma and stromal cells recapitulate the mild NF-κB activation observed in vivo. Cell Death Dis 2024; 15:731. [PMID: 39370432 PMCID: PMC11456592 DOI: 10.1038/s41419-024-07038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 10/08/2024]
Abstract
Multiple myeloma (MM) is linked to chronic NF-κB activity in myeloma cells, but this activity is generally considered a cell-autonomous property of the cancer cells. The precise extent of NF-κB activation and the contributions of the physical microenvironment and of cell-to-cell communications remain largely unknown. By quantitative immunofluorescence, we found that NF-κB is mildly and heterogeneously activated in a fraction of MM cells in human BMs, while only a minority of MM cells shows a strong activation. To gain quantitative insights on NF-κB activation in living MM cells, we combined advanced live imaging of endogenous p65 Venus-knocked-in in MM.1S and HS-5 cell lines to model MM and mesenchymal stromal cells (MSCs), cell co-cultures, microfluidics and custom microbioreactors to mimic the 3D-interactions within the bone marrow (BM) microenvironment. We found that i) reciprocal MM-MSC paracrine crosstalk and cell-to-scaffold interactions shape the inflammatory response in the BM; ii) the pro-inflammatory cytokine IL-1β, abundant in MM patients' plasma, activates MSCs, whose paracrine signals are responsible for strong NF-κB activation in a minority of MM cells; iii) IL-1β, but not TNF-α, activates NF-κB in vivo in BM-engrafted MM cells, while its receptor inhibitor Anakinra reduces the global NF-κB activation. We propose that NF-κB activation in the BM of MM patients is mild, restricted to a minority of cells and modulated by the interplay of restraining physical microenvironmental cues and activating IL-1β-dependent stroma-to-MM crosstalk.
Collapse
Affiliation(s)
- Federica Colombo
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Virginia Guzzeloni
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Universita' Vita-Salute San Raffaele, Milan, Italy
| | - Cise Kizilirmak
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesca Brambilla
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Anna Sofia Tascini
- Universita' Vita-Salute San Raffaele, Milan, Italy
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Moalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | | | - Rosanna Mezzapelle
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Universita' Vita-Salute San Raffaele, Milan, Italy
| | - Marina Ferrarini
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Elisabetta Ferrero
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marco E Bianchi
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Universita' Vita-Salute San Raffaele, Milan, Italy
| | - Samuel Zambrano
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy.
- Universita' Vita-Salute San Raffaele, Milan, Italy.
| | - Alessandra Agresti
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy.
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
2
|
Liu TT, Chen YK, Adil M, Almehmadi M, Alshabrmi FM, Allahyani M, Alsaiari AA, Liu P, Khan MR, Peng Q. In Silico Identification of Natural Product-Based Inhibitors Targeting IL-1β/IL-1R Protein-Protein Interface. Molecules 2023; 28:4885. [PMID: 37446547 DOI: 10.3390/molecules28134885] [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: 05/08/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
IL-1β mediates inflammation and regulates immune responses, cell proliferation, and differentiation. Dysregulation of IL-1β is linked to inflammatory and autoimmune diseases. Elevated IL-1β levels are found in patients with severe COVID-19, indicating its excessive production may worsen the disease. Also, dry eye disease patients show high IL-1β levels in tears and conjunctival epithelium. Therefore, IL-1β signaling is a potential therapeutic targeting for COVID-19 and aforementioned diseases. No small-molecule IL-1β inhibitor is clinically approved despite efforts. Developing such inhibitors is highly desirable. Herein, a docking-based strategy was used to screen the TCM (Traditional Chinese Medicine) database to identify possible IL-1β inhibitors with desirable pharmacological characteristics by targeting the IL-1β/IL-1R interface. Primarily, the docking-based screening was performed by selecting the crucial residues of IL-1β interface to retrieve the potential compounds. Afterwards, the compounds were shortlisted on the basis of binding scores and significant interactions with the crucial residues of IL-1β. Further, to gain insights into the dynamic behavior of the protein-ligand interactions, MD simulations were performed. The analysis suggests that four selected compounds were stabilized in an IL-1β pocket, possibly blocking the formation of an IL-1β/IL-1R complex. This indicates their potential to interfere with the immune response, making them potential therapeutic agents to investigate further.
Collapse
Affiliation(s)
- Ting-Ting Liu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410200, China
| | - Yan-Kun Chen
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410200, China
| | - Muhammad Adil
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi 46000, Pakistan
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, 40-007 Katowice, Poland
| | - Mazen Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mamdouh Allahyani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Ahad Amer Alsaiari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Pei Liu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410200, China
| | - Muhammad Raheel Khan
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. Sklodowskiej-Curie 34, 41-819 Zabrze, Poland
- Department of Chemical Sciences, Joint Doctoral School, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Qinghua Peng
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410200, China
| |
Collapse
|
3
|
García-Sánchez D, González-González A, Alfonso-Fernández A, Del Dujo-Gutiérrez M, Pérez-Campo FM. Communication between bone marrow mesenchymal stem cells and multiple myeloma cells: Impact on disease progression. World J Stem Cells 2023; 15:421-437. [PMID: 37342223 PMCID: PMC10277973 DOI: 10.4252/wjsc.v15.i5.421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of immunoglobulin-secreting clonal plasma cells at the bone marrow (BM). The interaction between MM cells and the BM microenvironment, and specifically BM mesenchymal stem cells (BM-MSCs), has a key role in the pathophysiology of this disease. Multiple data support the idea that BM-MSCs not only enhance the proliferation and survival of MM cells but are also involved in the resistance of MM cells to certain drugs, aiding the progression of this hematological tumor. The relation of MM cells with the resident BM-MSCs is a two-way interaction. MM modulate the behavior of BM-MSCs altering their expression profile, proliferation rate, osteogenic potential, and expression of senescence markers. In turn, modified BM-MSCs can produce a set of cytokines that would modulate the BM microenvironment to favor disease progression. The interaction between MM cells and BM-MSCs can be mediated by the secretion of a variety of soluble factors and extracellular vesicles carrying microRNAs, long non-coding RNAs or other molecules. However, the communication between these two types of cells could also involve a direct physical interaction through adhesion molecules or tunneling nanotubes. Thus, understanding the way this communication works and developing strategies to interfere in the process, would preclude the expansion of the MM cells and might offer alternative treatments for this incurable disease.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Alberto González-González
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Ana Alfonso-Fernández
- Servicio de Traumatología y Cirugía Ortopédica, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Facultad de Medicina, Universidad de Cantabria, Santander 39008, Cantabria, Spain
| | - Mónica Del Dujo-Gutiérrez
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Flor M Pérez-Campo
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| |
Collapse
|
4
|
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: 2] [Impact Index Per Article: 0.7] [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
|
5
|
Lussier T, Schoebe N, Mai S. Risk Stratification and Treatment in Smoldering Multiple Myeloma. Cells 2021; 11:130. [PMID: 35011692 PMCID: PMC8750018 DOI: 10.3390/cells11010130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 11/30/2022] Open
Abstract
Smoldering multiple myeloma is a heterogeneous asymptomatic precursor to multiple myeloma. Since its identification in 1980, risk stratification models have been developed using two main stratification methods: clinical measurement-based and genetics-based. Clinical measurement models can be subdivided in three types: baseline measurements (performed at diagnosis), evolving measurements (performed over time during follow-up appointments), and imaging (for example, magnetic resonance imaging). Genetic approaches include gene expression profiling, DNA/RNA sequencing, and cytogenetics. It is important to accurately distinguish patients with indolent disease from those with aggressive disease, as clinical trials have shown that patients designated as "high-risk of progression" have improved outcomes when treated early. The risk stratification models, and clinical trials are discussed in this review.
Collapse
Affiliation(s)
- Tyler Lussier
- Department of Physiology and Pathophysiology, University of Manitoba, Cancer Care Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (T.L.); (N.S.)
- Faculté des Sciences, Université de Saint-Boniface, Winnipeg, MB R2H 0H7, Canada
| | - Natalie Schoebe
- Department of Physiology and Pathophysiology, University of Manitoba, Cancer Care Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (T.L.); (N.S.)
- Faculty of Biosciences, University of Heidelberg, 69117 Heidelberg, Germany
| | - Sabine Mai
- Department of Physiology and Pathophysiology, University of Manitoba, Cancer Care Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (T.L.); (N.S.)
| |
Collapse
|
6
|
Zhaoyun L, Rong F. Predictive Role of Immune Profiling for Survival of Multiple Myeloma Patients. Front Immunol 2021; 12:663748. [PMID: 34290698 PMCID: PMC8287504 DOI: 10.3389/fimmu.2021.663748] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/23/2021] [Indexed: 01/10/2023] Open
Abstract
Despite new efficacy drugs and cell therapy have been used for multiple myeloma (MM) patients, some patients will relapse over time. We wonder the immune system play a vital role as well as MM cell during the development of disease. It is clear that the characteristic of myeloma cell is associated with the survival of MM patients. However, the link between the immune profiling and the prognosis of the disease is still not entirely clear. As more study focus on the role of immunity on multiple myeloma pathogenesis. There are plenty of study about the predictive role of immunity on the survival of multiple myeloma patients. Up to mow, the majority reviews published have focused on the immunotherapy and immune pathogenesis. It is indispensable to overlook the predictive role of immunity on multiple myeloma patients. Here, we give a review of vital previous works and recent progress related to the predictive role of immune profiling on multiple myeloma, such as absolute lymphocyte count, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, lymphocytes and cytokines.
Collapse
Affiliation(s)
- Liu Zhaoyun
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu Rong
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
7
|
Rheumatologic diseases impact the risk of progression of MGUS to overt multiple myeloma. Blood Adv 2021; 5:1746-1754. [PMID: 33749761 DOI: 10.1182/bloodadvances.2020003193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/04/2021] [Indexed: 12/15/2022] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS), a premalignant condition, is associated with various chronic inflammatory rheumatic diseases (RDs) and is frequently observed as an incidental finding during routine work-up. The association of MGUS and chronic RDs is well established, but the impact of RDs on the risk of transformation into overt multiple myeloma (MM) has not been evaluated so far. MGUS patients diagnosed between January 2000 and August 2016 were identified and screened for concomitant RDs. RDs were grouped into antibody (Ab)-mediated RDs and non-Ab-mediated RDs (polymyalgia rheumatica, large-vessel giant cell arteritis, spondyloarthritis, and gout). Progression to MM was defined as a categorical (yes/no) or continuous time-dependent (time to progression) variable. Of 2935 MGUS patients, 255 (9%) had a concomitant RD. MGUS patients diagnosed with non-Ab-mediated RDs had a doubled risk of progression compared with those without a concomitant RD (hazard ratio, 2.1; 95% CI, 1.1-3.9; P = .02). These data translate into a 5-year risk of progression of 4% in MGUS patients without rheumatologic comorbidity, 10% in those with concomitant non-Ab-mediated RDS, and 2% in those with Ab-mediated RDs. By using the complex risk stratification model that includes myeloma protein (M-protein) concentration, immunoglobulin type, and level of free light chain ratio as variables, patients with non-Ab-mediated RDs (n = 57) had the highest risk for progression (hazard ratio, 6.8; 95% CI, 1.5-30.7; P = .01) compared with patients with Ab-mediated RDs (n = 77). Chronic inflammatory diseases have an impact on the risk of MGUS progressing into overt MM, with a doubled risk of transformation observed in patients with non-Ab-mediated RDs. Future research can elucidate whether comorbidities such as RDs should be included in currently applied prognostic MGUS scores.
Collapse
|
8
|
Melaiu O, Macauda A, Sainz J, Calvetti D, Facioni MS, Maccari G, Ter Horst R, Netea MG, Li Y, Grząśko N, Moreno V, Jurczyszyn A, Jerez A, Watek M, Varkonyi J, Garcia-Sanz R, Kruszewski M, Dudziński M, Kadar K, Jacobsen SEH, Mazur G, Andersen V, Rybicka M, Zawirska D, Raźny M, Zaucha JM, Ostrovsky O, Iskierka-Jazdzewska E, Reis RM, Stępień A, Beider K, Nagler A, Druzd-Sitek A, Marques H, Martìnez-Lopez J, Lesueur F, Avet-Loiseau H, Vangsted AJ, Krawczyk-Kulis M, Butrym A, Jamroziak K, Dumontet C, Vogel U, Rymko M, Pelosini M, Subocz E, Szombath G, Sarasquete ME, Silvestri R, Morani F, Landi S, Campa D, Canzian F, Gemignani F. Common gene variants within 3'-untranslated regions as modulators of multiple myeloma risk and survival. Int J Cancer 2021; 148:1887-1894. [PMID: 33152124 DOI: 10.1002/ijc.33377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/04/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
We evaluated the association between germline genetic variants located within the 3'-untranlsated region (polymorphic 3'UTR, ie, p3UTR) of candidate genes involved in multiple myeloma (MM). We performed a case-control study within the International Multiple Myeloma rESEarch (IMMEnSE) consortium, consisting of 3056 MM patients and 1960 controls recruited from eight countries. We selected p3UTR of six genes known to act in different pathways relevant in MM pathogenesis, namely KRAS (rs12587 and rs7973623), VEGFA (rs10434), SPP1 (rs1126772), IRF4 (rs12211228) and IL10 (rs3024496). We found that IL10-rs3024496 was associated with increased risk of developing MM and with a worse overall survival of MM patients. The variant allele was assayed in a vector expressing eGFP chimerized with the IL10 3'-UTR and it was found functionally active following transfection in human myeloma cells. In this experiment, the A-allele caused a lower expression of the reporter gene and this was also in agreement with the in vivo expression of mRNA measured in whole blood as reported in the GTEx portal. Overall, these data are suggestive of an effect of the IL10-rs3024496 SNP on the regulation of IL10 mRNA expression and it could have clinical implications for better characterization of MM patients in terms of prognosis.
Collapse
Affiliation(s)
| | - Angelica Macauda
- Department of Biology, University of Pisa, Pisa, Italy
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO. Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
- Hematology Department, Virgen de las Nieves University Hospital, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | - Diego Calvetti
- Department of Biology, University of Pisa, Pisa, Italy
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Victor Moreno
- Cancer Prevention and Control Program, Catalan Institute of Oncology (ICO), IDIBELL, CIBERESP and Department of Clinical Sciences, Faculty of Medicine, University of Barcelona. Hospitalet de Llobregat, Barcelona, Spain
| | - Artur Jurczyszyn
- Department of Hematology, Cracow University Hospital, Cracow, Poland
| | - Andrés Jerez
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, Murcia, Spain
| | | | - Judit Varkonyi
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Ramon Garcia-Sanz
- Department of Hematology, University Hospital of Salamanca, Instituto de Investigación Biomédica de Salamanca, IBSAL, Salamanca, Spain
| | | | - Marek Dudziński
- Department of Hematology, Specialist District Hospital, Rzeszow, Poland
| | - Katalin Kadar
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | | | - Grzegorz Mazur
- Department of Internal Diseases, Hypertension and Occupational Medicine, Medical University, Wroclaw, Poland
| | - Vibeke Andersen
- Research Unit of Molecular Diagnostics and Clinical Research, Laboratory Center, Hospital of Southern Jutland, Aabenraa, Denmark
- Institute of Regional Health Research, and Institute of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Malwina Rybicka
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Daria Zawirska
- Department of Hematology, Cracow University Hospital, Cracow, Poland
| | | | | | - Olga Ostrovsky
- Hematology Division Chaim Sheba Medical Center, Tel Hashomer, Israel
| | | | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Anna Stępień
- Laboratory of Clinical and Transplant Immunology and Genetics, Copernicus Memorial Hospital, Lodz, Poland
| | - Katia Beider
- Hematology Division Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Nagler
- Hematology Division Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Agnieszka Druzd-Sitek
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie Memorial Institute and Oncology Centre Warsaw, Warsaw, Poland
| | - Herlander Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquin Martìnez-Lopez
- Department of Hematology, Hospital Universitario 12 de Octubre, Complutense School of Medicine, CNIO, Madrid, Spain
| | - Fabienne Lesueur
- Institut Curie, Paris, France
- PSL Research University, Paris, France
- Inserm, Paris, France
- Mines Paris Tech, Fontainebleau, France
| | - Hervé Avet-Loiseau
- Hematology, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Annette Juul Vangsted
- Department of Haematology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Malgorzata Krawczyk-Kulis
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Aleksandra Butrym
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Medical University, Wrocław, Poland
| | - Krzysztof Jamroziak
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | | | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Marcin Rymko
- Department of Hematology, Copernicus Hospital, Torun, Poland
| | - Matteo Pelosini
- U.O. Dipartimentale di Ematologia, Azienda USL Toscana Nord Ovest, Livorno, Italy
| | - Edyta Subocz
- Department of Hematology, Military Institute of Medicine, Warsaw, Poland
| | - Gergely Szombath
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Maria Eugenia Sarasquete
- Department of Hematology, University Hospital of Salamanca, Instituto de Investigación Biomédica de Salamanca, IBSAL, Salamanca, Spain
| | | | | | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | |
Collapse
|
9
|
Takemori N, Ooi HK, Imai G, Hoshino K, Saio M. Possible mechanisms of action of clarithromycin and its clinical application as a repurposing drug for treating multiple myeloma. Ecancermedicalscience 2020; 14:1088. [PMID: 33014130 PMCID: PMC7498274 DOI: 10.3332/ecancer.2020.1088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Clarithromycin (CAM), a semisynthetic macrolide antibiotic, is a widely used antibacterial drug. Recently, the efficacy of CAM as an add-on drug for treating multiple myeloma (MM) has been noted. Its effect on treating MM has been confirmed in combination chemotherapies that include CAM. However, a single treatment of CAM has no efficacy for treating MM. Many myeloma growth factors (MGFs) including interleukin (IL)-6 are known to be closely involved in the development of MM. CAM has been shown to suppress many MGFs, particularly IL-6. The possible mechanisms of action of CAM in treating MM have been suggested to include its immunomodulatory effect, autophagy inhibition, reversibility of drug resistance, steroid-sparing/enhancing effect and suppression of MGFs. In addition, MM is characterised by uncontrolled cell growth of monoclonal immunoglobulin (Ig)-producing neoplastic plasma cells. Large quantities of unfolded or misfolded Ig production may trigger considerable endoplasmic reticulum stress. Thus, MM is originally a fragile neoplasm particularly susceptible to autophagy-, proteasome- and histone deacetylase 6-inhibitors. Taken together, CAM plays an important role in MM treatments through its synergistic mechanisms. In addition, CAM with its pleiotropic effects on cytokines including IL-6 and indirect antiviral effects might be worth a try for treating COVID-19.
Collapse
Affiliation(s)
- Nobuo Takemori
- Department of Internal Medicine, Division of Hematology, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
- https://orcid.org/0000-0001-9742-8385
| | - Hong-Kean Ooi
- Department of Veterinary Medicine, Azabu University, Fuchinobe 1-17-71, Sagamihara, Kanagawa 252-5201, Japan
| | - Goro Imai
- Department of Internal Medicine, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
| | - Kazuo Hoshino
- Department of Surgery, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
| | - Masanao Saio
- Laboratory of Histopathology & Cytopathology, Department of Laboratory Sciences, Gunma University, Graduate School of Health Sciences, 39-22, 3-chome, Showa-machi, Maebashi, Gunma 371-8514, Japan
| |
Collapse
|
10
|
Van Gorp H, Lamkanfi M. The emerging roles of inflammasome-dependent cytokines in cancer development. EMBO Rep 2019; 20:embr.201847575. [PMID: 31101676 DOI: 10.15252/embr.201847575] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/02/2019] [Accepted: 04/30/2019] [Indexed: 12/24/2022] Open
Abstract
In addition to the genomic alterations that occur in malignant cells, the immune system is increasingly appreciated as a critical axis that regulates the rise of neoplasms and the development of primary tumours and metastases. The interaction between inflammatory cell infiltrates and stromal cells in the tumour microenvironment is complex, with inflammation playing both pro- and anti-tumorigenic roles. Inflammasomes are intracellular multi-protein complexes that act as key signalling hubs of the innate immune system. They respond to cellular stress and trauma by promoting activation of caspase-1, a protease that induces a pro-inflammatory cell death mode termed pyroptosis along with the maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Here, we will briefly introduce inflammasome biology with a focus on the dual roles of inflammasome-produced cytokines in cancer development. Despite emerging insight that inflammasomes may promote and suppress cancer development according to the tumour stage and the tumour microenvironment, much remains to be uncovered. Further exploration of inflammasome biology in tumorigenesis should enable the development of novel immunotherapies for cancer patients.
Collapse
Affiliation(s)
- Hanne Van Gorp
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Center for Inflammation Research, VIB, Ghent, Belgium
| | - Mohamed Lamkanfi
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium .,Janssen Immunosciences, World Without Disease Accelerator, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| |
Collapse
|
11
|
Litmanovich A, Khazim K, Cohen I. The Role of Interleukin-1 in the Pathogenesis of Cancer and its Potential as a Therapeutic Target in Clinical Practice. Oncol Ther 2018; 6:109-127. [PMID: 32700032 PMCID: PMC7359982 DOI: 10.1007/s40487-018-0089-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
Interleukin-1 (IL-1) has long been known to be a key mediator of immunity and inflammation. Its dysregulation has been implicated in recent years in tumorigenesis and tumor progression, and its upregulation is thought to be associated with many tumors. Overexpression of the IL-1 agonists IL-1α and IL-1β has been shown to promote tumor invasiveness and metastasis by inducing the expression of angiogenic genes and growth factors. IL-1 blockers such as anakinra and canakinumab are already approved and widely used for the treatment of some autoimmune and autoinflammatory diseases and are currently being tested in preclinical and human clinical trials for cancer therapy. In this paper we review the most recent discoveries regarding the association between IL-1 dysregulation and cancer and present the novel IL-1 blockers currently being tested in cancer therapy and their corresponding clinical trials.
Collapse
Affiliation(s)
- Adi Litmanovich
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Khaled Khazim
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
- Department of Nephrology and Hypertension, Galilee Medical Center, Nahariya, Israel
| | - Idan Cohen
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
- Research Institute, Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
12
|
Abstract
Background Autoinflammatory diseases are distinct from autoimmune diseases. Whereas autoinflammatory diseases are due to dysfunctional T-cells and B-cells, autoinflammatory diseases are due to overproduction of macrophage cytokines particularly interleukin-1 beta (IL-1β). A causative role for IL-1 in autoinflammatory diseases is derived from clinical studies blocking the IL-1 receptor or neutralizing monoclonal antibodies or soluble receptors. Methods A review was performed of clinical trials in autoinflammatory diseases using the IL-1 receptor antagonist (anakinra), the soluble IL-1 receptor (rilonacept), antibodies to IL-1β (canakinumab, gevokizumab) and anti-IL-1α (xilonix). Findings Anakinra blocks the IL-1 Receptor type 1 (IL-1R1) and therefore blocks the activities of both IL-1α and IL-1β. Off-label use of anakinra is common for a broad spectrum of inflammatory diseases. Neutralization of IL-1β is used to treat hereditary autoinflammatory diseases but also atherosclerosis. Rilonacept reduces arterial wall inflammation in patients with chronic kidney disease. Neutralization of IL-1α has prolonged life in patients with advanced metastatic colorectal cancer. Compared to other cytokine blocking therapies, reducing the activities of IL-1 has an excellent safety record. Conclusions Blocking IL-1 therapies can be used to treat a wide-spectrum of acute and chronic inflammatory diseases.
Collapse
|
13
|
Abstract
Interleukin (IL)-1 is a pro-inflammatory cytokine that induces local and systemic inflammation aimed to eliminate microorganisms and tissue damage. However, an increasing number of clinical conditions have been identified in which IL-1 production is considered inappropriate and IL-1 is part of the disease etiology. In autoinflammatory diseases, gout, Schnitzler's syndrome, and adult-onset Still's disease, high levels of inappropriate IL-1 production have been shown to be a key process in the etiology of the disease. In these conditions, blocking IL-1 has proven very effective in clinical studies. In other diseases, IL-1 has shown to be present in disease process but is not the central driving force of inflammation. In these conditions, including type 1 and 2 diabetes mellitus, acute coronary syndrome, amyotrophic lateral sclerosis, and several neoplastic diseases, the benefits of IL-1 blockade are minimal or absent.
Collapse
|
14
|
Guo J, Zhang H, Xia J, Hou J, Wang Y, Yang T, Wang S, Zhang X, Chen X, Wu X. Interleukin-1β induces intercellular adhesion molecule-1 expression, thus enhancing the adhesion between mesenchymal stem cells and endothelial progenitor cells via the p38 MAPK signaling pathway. Int J Mol Med 2018; 41:1976-1982. [PMID: 29393395 PMCID: PMC5810197 DOI: 10.3892/ijmm.2018.3424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are an important component of stem-cell niches, which are able to promote the self-renewal and pluripotency of mesenchymal stem cells (MSCs). The biological functions of these two cell types is dependent on adhesion, and the adhesion between MSCs and EPCs is important due to their critical role in neovascularization and bone regeneration in tissue engineering. Intercellular adhesion molecule-1 (ICAM-1, also known as cluster of differentiation 54), is a member of the immunoglobulin supergene family, which functions in cell-cell and cell-matrix adhesive interactions. Compared with other adhesion molecules, ICAM-1 is expressed in hematopoietic and nonhematopoietic cells, and can mediate adhesive interactions. The present study aimed to investigate the importance of ICAM-1 in the adhesion of MSCs and EPCs, and demonstrated that adhesion between these cells could be regulated by interleukin (IL)-1β via the p38 mitogen-activated protein kinase pathway. In addition, the results confirmed that ICAM-1 served a critical role in regulation of adhesion between MSCs and EPCs. ELISA, cell immunofluorescence, western blot analysis and adhesion assay were used to confirm our theory from phenomenon to essence. The present study provided evidence to support and explain the adhesion between MSCs and EPCs. Furthermore, the present findings provide a theoretical basis for further stem-cell niche transplantation to increase understanding of the function of MSCs and the crosstalk between MSCs and EPCs in the stem-cell niche.
Collapse
Affiliation(s)
- Jun Guo
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Hongwei Zhang
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jie Xia
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jixue Hou
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Yixiao Wang
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Tao Yang
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Sibo Wang
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xuyong Zhang
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xuelin Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xiangwei Wu
- Department of General Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| |
Collapse
|
15
|
Inflammatory and Anti-Inflammatory Equilibrium, Proliferative and Antiproliferative Balance: The Role of Cytokines in Multiple Myeloma. Mediators Inflamm 2017; 2017:1852517. [PMID: 29089667 PMCID: PMC5635476 DOI: 10.1155/2017/1852517] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is typically exemplified by a desynchronized cytokine system with increased levels of inflammatory cytokines. We focused on the contrast between inflammatory and anti-inflammatory systems by assessing the role of cytokines and their influence on MM. The aim of this review is to summarize the available information to date concerning this equilibrium to provide an overview of the research exploring the roles of serum cytokines in MM. However, the association between MM and inflammatory cytokines appears to be inadequate, and other functions, such as pro-proliferative or antiproliferative effects, can assume the role of cytokines in the genesis and progression of MM. It is possible that inflammation, when guided by cancer-specific Th1 cells, may inhibit tumour onset and progression. In a Th1 microenvironment, proinflammatory cytokines (e.g., IL-6 and IL-1) may contribute to tumour eradication by attracting leucocytes from the circulation and by increasing CD4 + T cell activity. Hence, caution should be used when considering therapies that target factors with pro- or anti-inflammatory activity. Drugs that may reduce the tumour-suppressive Th1-driven inflammatory immune response should be avoided. A better understanding of the relationship between inflammation and myeloma will ensure more effective therapeutic interventions.
Collapse
|
16
|
Lust JA, Lacy MQ, Zeldenrust SR, Witzig TE, Moon-Tasson LL, Dinarello CA, Donovan KA. Reduction in C-reactive protein indicates successful targeting of the IL-1/IL-6 axis resulting in improved survival in early stage multiple myeloma. Am J Hematol 2016; 91:571-4. [PMID: 26945843 DOI: 10.1002/ajh.24352] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 11/08/2022]
Abstract
We report the long-term follow-up results of a phase II trial of IL-1 receptor antagonist and low-dose dexamethasone for early stage multiple myeloma (MM). Patients were eligible if they had smoldering multiple myeloma (SMM) or indolent multiple myeloma (IMM) without the need for immediate therapy. Forty seven patients were enrolled and subsequently treated with IL-1Ra; in 25/47 low-dose dexamethasone (20 mg weekly) was added. The primary endpoint was progression-free survival (PFS). In the clinical trial, three patients achieved a minor response (MR) to IL-1Ra alone; five patients a partial response (PR) and four patients an MR after addition of dexamethasone. Seven patients showed a decrease in the plasma cell labeling index (PCLI) which paralleled a decrease in the high sensitivity C-reactive protein (hs-CRP). The median PFS for the 47 patients was 1116 days (37.2 months). The median PFS for patients without (n = 22) and with (n = 25) a decrease in their baseline hs-CRP was 326 days (11 months) vs. 3139 days (104 months) respectively (P <0.0001). The median overall survival (OS) for the 47 patients was 3482 days (9.5 years). The median OS for patients without and with a decrease in their baseline hs-CRP was 2885 days (7.9 years) vs. median not reached, respectively (P = 0.001). In SMM/IMM patients at risk for progression to active myeloma, reduction in the hs-CRP indicates successful targeting of the IL-1/IL-6 axis resulting in improved PFS and OS. (Clinical Trials.gov Identifier: NCT00635154) Am. J. Hematol. 91:571-574, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- John A. Lust
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | - Martha Q. Lacy
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | | | | | | | - Charles A. Dinarello
- Department of Medicine; University of Colorado; 12700 East 19 Ave, B168 Aurora Colorado 80045
| | | |
Collapse
|
17
|
Ahn IE, Mailankody S, Korde N, Landgren O. Dilemmas in treating smoldering multiple myeloma. J Clin Oncol 2014; 33:115-23. [PMID: 25422486 DOI: 10.1200/jco.2014.56.4351] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Novel therapies hold promise for high-risk smoldering multiple myeloma (SMM). Recent studies suggest that modern combination approaches can be options for high-risk SMM to obtain deep molecular responses with favorable toxicity profiles. Although pioneering treatment trials based on small numbers of patients suggest progression-free and overall survival benefits, application of the data to real-life practice remains to be validated. Therapeutic modulation of disease tempo, disease burden, clonal evolution, and tumor microenvironment in SMM remains to be understood and calls for reliable biomarkers reflective of disease biology. Here, we review studies that open a new management platform for SMM, address ongoing dilemmas in practice and under investigation, and highlight emerging scientific questions in the era of SMM treatment.
Collapse
Affiliation(s)
- Inhye E Ahn
- Inhye Ahn and Sham Mailankody, the National Cancer Institute, National Institutes of Health, Bethesda, MD; Neha Korde and Ola Landgren, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sham Mailankody
- Inhye Ahn and Sham Mailankody, the National Cancer Institute, National Institutes of Health, Bethesda, MD; Neha Korde and Ola Landgren, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neha Korde
- Inhye Ahn and Sham Mailankody, the National Cancer Institute, National Institutes of Health, Bethesda, MD; Neha Korde and Ola Landgren, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ola Landgren
- Inhye Ahn and Sham Mailankody, the National Cancer Institute, National Institutes of Health, Bethesda, MD; Neha Korde and Ola Landgren, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
18
|
Hope C, Ollar SJ, Heninger E, Hebron E, Jensen JL, Kim J, Maroulakou I, Miyamoto S, Leith C, Yang DT, Callander N, Hematti P, Chesi M, Bergsagel PL, Asimakopoulos F. TPL2 kinase regulates the inflammatory milieu of the myeloma niche. Blood 2014; 123:3305-15. [PMID: 24723682 PMCID: PMC4046426 DOI: 10.1182/blood-2014-02-554071] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/07/2014] [Indexed: 02/06/2023] Open
Abstract
Targeted modulation of microenvironmental regulatory pathways may be essential to control myeloma and other genetically/clonally heterogeneous cancers. Here we report that human myeloma-associated monocytes/macrophages (MAM), but not myeloma plasma cells, constitute the predominant source of interleukin-1β (IL-1β), IL-10, and tumor necrosis factor-α at diagnosis, whereas IL-6 originates from stromal cells and macrophages. To dissect MAM activation/cytokine pathways, we analyzed Toll-like receptor (TLR) expression in human myeloma CD14(+) cells. We observed coregulation of TLR2 and TLR6 expression correlating with local processing of versican, a proteoglycan TLR2/6 agonist linked to carcinoma progression. Versican has not been mechanistically implicated in myeloma pathogenesis. We hypothesized that the most readily accessible target in the versican-TLR2/6 pathway would be the mitogen-activated protein 3 (MAP3) kinase, TPL2 (Cot/MAP3K8). Ablation of Tpl2 in the genetically engineered in vivo myeloma model, Vκ*MYC, led to prolonged disease latency associated with plasma cell growth defect. Tpl2 loss abrogated the "inflammatory switch" in MAM within nascent myeloma lesions and licensed macrophage repolarization in established tumors. MYC activation/expression in plasma cells was independent of Tpl2 activity. Pharmacologic TPL2 inhibition in human monocytes led to dose-dependent attenuation of IL-1β induction/secretion in response to TLR2 stimulation. Our results highlight a TLR2/6-dependent TPL2 pathway as novel therapeutic target acting nonautonomously through macrophages to control myeloma progression.
Collapse
Affiliation(s)
- Chelsea Hope
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Samuel J Ollar
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Erika Heninger
- University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Ellen Hebron
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Jeffrey L Jensen
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Jaehyup Kim
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Ioanna Maroulakou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Shigeki Miyamoto
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Oncology, University of Wisconsin-Madison, Madison, WI
| | - Catherine Leith
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; and
| | - David T Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; and
| | - Natalie Callander
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Peiman Hematti
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | | | | | - Fotis Asimakopoulos
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| |
Collapse
|
19
|
Goh AXH, Bertin-Maghit S, Ping Yeo S, Ho AWS, Derks H, Mortellaro A, Wang CI. A novel human anti-interleukin-1β neutralizing monoclonal antibody showing in vivo efficacy. MAbs 2014; 6:765-73. [PMID: 24671001 PMCID: PMC4011920 DOI: 10.4161/mabs.28614] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pro-inflammatory cytokine interleukin (IL)-1β is a clinical target in many conditions involving dysregulation of the immune system; therapeutics that block IL-1β have been approved to treat diseases such as rheumatoid arthritis (RA), neonatal onset multisystem inflammatory diseases, cryopyrin-associated periodic syndromes, active systemic juvenile idiopathic arthritis. Here, we report the generation and engineering of a new fully human antibody that binds tightly to IL-1β with a neutralization potency more than 10 times higher than that of the marketed antibody canakinumab. After affinity maturation, the derived antibody shows a>30-fold increased affinity to human IL-1β compared with its parent antibody. This anti-human IL-1β IgG also cross-reacts with mouse and monkey IL-1β, hence facilitating preclinical development. In a number of mouse models, this antibody efficiently reduced or abolished signs of disease associated with IL-1β pathology. Due to its high affinity for the cytokine and its potency both in vitro and in vivo, we propose that this novel fully human anti-IL-1β monoclonal antibody is a promising therapeutic candidate and a potential alternative to the current therapeutic arsenal.
Collapse
Affiliation(s)
- Angeline X H Goh
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Sebastien Bertin-Maghit
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Siok Ping Yeo
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Adrian W S Ho
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Heidi Derks
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Alessandra Mortellaro
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| | - Cheng-I Wang
- Singapore Immunology Network; Agency for Science; Technology and Research (A*STAR); Singapore
| |
Collapse
|
20
|
Kim CS, Kim JY, Kim HJ. The effects of a single bout pilates exercise on mRNA expression of bone metabolic cytokines in osteopenia women. J Exerc Nutrition Biochem 2014; 18:69-78. [PMID: 25566441 PMCID: PMC4241937 DOI: 10.5717/jenb.2014.18.1.69] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/03/2014] [Accepted: 02/08/2014] [Indexed: 11/09/2022] Open
Abstract
[Purpose] The purpose of this study was to examine the effect of a single bout pilates exercise on mRNA expression of bone metabolic cytokines in elderly osteopenia women. [Methods] We selected 11 people of elderly osteopenia women and loaded a single bout pilates exercise about RPE 10-14 level. The blood samples were collected before, immediately after and 60 minute after pilates exercise, then examined calcium metabolic markers in serum and extracted peripheral blood mononuclear cell (PBMC) from whole blood and confirmed mRNA expression of bone metabolic cytokines from PBMC. To clarify the changes during exercise, we designed repeated measure ANOVA as the control group to perform blood sampling without exercise. [Results] As a result, serum P showed significant interaction effect between group and time (p<.001), the pilates exercise group decreased about 9% at immediately after exercise and 13% during recovery after exercise (p<.05), while the control group showed a tendency to increase. Serum CK also showed a significant interaction between group and time (p<.05), the pilates group significantly increased at immediately after exercise and during recovery after exercise (p<.05) but the control group didn’t have changes. TNF-α and IL-6 mRNA expression in PBMC was significantly increased in the pilates group (p<.01, p<.05), although INF-γ mRNA expression didn’t show statistically significant difference, it tended to increase in the pilates group (NS). [Conclusion] These results suggested that a single bout pilates exercise of elderly osteopenia women cause hypophosphatemia with temporary muscle damage, and it leading high turnover bone metabolic state with to activate both of bone formation and bone resorption.
Collapse
Affiliation(s)
- Chang Sun Kim
- Department of Physical Education, Dongduk Women's University, Seoul, Korea
| | - Ji Yeon Kim
- Department of Physical Education, Dongduk Women's University, Seoul, Korea
| | - Hyo Jin Kim
- Department of Physical Education, Dongduk Women's University, Seoul, Korea
| |
Collapse
|
21
|
Tageja N, Manasanch EE, Korde N, Kwok M, Mailankody S, Bhutani M, Roschewski M, Landgren O. Smoldering multiple myeloma: present position and potential promises. Eur J Haematol 2013; 92:1-12. [PMID: 24112232 DOI: 10.1111/ejh.12205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2013] [Indexed: 12/24/2022]
Abstract
Since smoldering multiple myeloma (SMM) was first described over three decades ago based on a case series of six patients, its definition and our understanding of the entity have evolved considerably. The risk of progression to symptomatic myeloma (MM) varies greatly among individuals diagnosed with myeloma precursor disease. Epidemiologic, molecular, flow cytometric and radiological techniques have demonstrated that this transformation to MM from precursor states is not sudden but rather a continuous overlapping series of events with evidence of end-organ damage that could manifest in the earliest stages of disease. Contemporary antimyeloma therapies can yield rapid, deep, and durable responses with manageable toxicities, and molecular-cell-based measures are now available to rule out minimal residual disease. With this information, clinical studies with correlative measures can now be developed to test the fundamental hypothesis that intervention in early myeloma may provide a measurable clinical benefit to patients by either delaying progression or eradicating plasma cell clones.
Collapse
Affiliation(s)
- Nishant Tageja
- Multiple Myeloma Section, Metabolism Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Dinarello CA, Simon A, van der Meer JWM. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012; 11:633-52. [PMID: 22850787 PMCID: PMC3644509 DOI: 10.1038/nrd3800] [Citation(s) in RCA: 1306] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin-1 (IL-1) is a highly active pro-inflammatory cytokine that lowers pain thresholds and damages tissues. Monotherapy blocking IL-1 activity in autoinflammatory syndromes results in a rapid and sustained reduction in disease severity, including reversal of inflammation-mediated loss of sight, hearing and organ function. This approach can therefore be effective in treating common conditions such as post-infarction heart failure, and trials targeting a broad spectrum of new indications are underway. So far, three IL-1-targeted agents have been approved: the IL-1 receptor antagonist anakinra, the soluble decoy receptor rilonacept and the neutralizing monoclonal anti-IL-1β antibody canakinumab. In addition, a monoclonal antibody directed against the IL-1 receptor and a neutralizing anti-IL-1α antibody are in clinical trials.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045, USA.
| | | | | |
Collapse
|
23
|
Vangsted AJ, Nielsen KR, Klausen TW, Haukaas E, Tjønneland A, Vogel U. A functional polymorphism in the promoter region of the IL1B gene is associated with risk of multiple myeloma. Br J Haematol 2012; 158:515-8. [PMID: 22540426 DOI: 10.1111/j.1365-2141.2012.09141.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 03/23/2012] [Indexed: 11/29/2022]
Abstract
The cytokine interleukin-1β (IL1B) is important for anti-tumour immune response. Genetic variation may modify the expression of IL1B and thereby influence the risk of disease. We investigated genetic variations with functional importance in the IL1B and NFKB1 genes in 348 population-based samples of multiple myeloma (MM) and a random sample of 1700 individuals. Carriers of the variant T-allele IL1B C-3737T and carriers of the TGT haplotype were at lower risk of MM [relative risk (RR) 0·58 (95% confidence interval (CI) = 0·41-0·84) and RR 0·59 (95%CI 0·40-0·85), respectively]. No association with risk of MM was found for the NFKB1- 94 ins/del polymorphism.
Collapse
Affiliation(s)
- Annette J Vangsted
- Department of Haematology, Roskilde Hospital, Copenhagen University, Roskilde, Denmark.
| | | | | | | | | | | |
Collapse
|
24
|
Vangsted A, Klausen TW, Vogel U. Genetic variations in multiple myeloma II: association with effect of treatment. Eur J Haematol 2011; 88:93-117. [DOI: 10.1111/j.1600-0609.2011.01696.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Tsirakis G, Pappa CA, Kaparou M, Katsomitrou V, Hatzivasili A, Alegakis T, Xekalou A, Stathopoulos EN, Alexandrakis MG. Assessment of proliferating cell nuclear antigen and its relationship with proinflammatory cytokines and parameters of disease activity in multiple myeloma patients. Eur J Histochem 2011; 55:e21. [PMID: 22073368 PMCID: PMC3203474 DOI: 10.4081/ejh.2011.e21] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 12/02/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disease. Several proinflammatory cytokines produced by malignant plasma cells and bone marrow (BM) stromal cells are involved in the pathogenesis of the disease. We evaluated serum levels of the proinflammatory cytokines Interleukin-1β (IL-1β), Interleukin-6 (IL-6), Interleukin-8 (IL-8), macrophage inflammatory protein-1α (MIP-1α), in MM patients before treatment, and determined its significance in tumor progression. We also analyzed the correlation between measured parameters with proliferating cell nuclear antigen (PCNA). Forty-four MM patients and 20 healthy controls were studied. Serum levels of the proinflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA), whereas PCNA value in the BM was determined by immunohistochemistry staining. The mean concentrations of the measured cytokines were significantly different among the three stages of disease, with higher values in advanced disease stage. Furthermore, patients with MM had significantly higher serum levels of the measured cytokines than in controls. A positive correlation was found between IL-6 with IL-1β, IL-8 and MIP-1α. Similarly, IL-8 and MIP-1α were positively correlated with markers of disease activity such as β2 microglobulin and LDH. The proliferation index, determined by PCNA immunostaining, was higher in advanced disease stage. Furthermore PCNA value correlated significantly with β2 microglobulin, LDH and the levels of the measured cytokines. Our results showed that the proliferative activity, as measured with PCNA, increases in parallel with disease stage. The positive correlation between PCNA and other measured mediators supports the involvement of these factors in the biology of myeloma cell growth and can be used as markers of disease activity and as possible therapeutic targets.
Collapse
Affiliation(s)
- G Tsirakis
- Department of Haematology, University Hospital of Heraklion, Crete, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fuhler GM, Diks SH, Peppelenbosch MP, Kerr WG. Widespread deregulation of phosphorylation-based signaling pathways in multiple myeloma cells: opportunities for therapeutic intervention. Mol Med 2011; 17:790-8. [PMID: 21541441 DOI: 10.2119/molmed.2011.00013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 04/26/2011] [Indexed: 01/07/2023] Open
Abstract
Multiple myeloma (MM) is a neoplasm of plasma cell origin that is largely confined to the bone marrow (BM). Chromosomal translocations and other genetic events are known to contribute to deregulation of signaling pathways that lead to transformation of plasma cells and progression to malignancy. However, the tumor stroma may also provide trophic support and enhance resistance to therapy. Phosphorylation of proteins on tyrosine, serine and threonine residues plays a pivotal role in cell growth and survival. Therefore, knowing the status of phosphorylation-based signaling pathways in cells may provide key insights into how cell growth and survival is promoted in tumor cells. To provide a more comprehensive molecular analysis of signaling disruptions in MM, we conducted a kinome profile comparison of normal plasma cells and MM plasma cells as well as their surrounding cells from normal BM and diseased BM. Integrated pathway analysis of the profiles obtained reveals deregulation of multiple signaling pathways in MM cells but also in surrounding bone marrow blood cells compared to their normal counterparts. The deregulated kinase activities identified herein, which include the mTOR (mammalian target of rapamycin)/p70S6K and ERK1/2 (extracellular signal-regulated kinases 1 and 2) pathways, are potential novel molecular targets in this lethal disease.
Collapse
Affiliation(s)
- Gwenny Manel Fuhler
- Department of Gasteroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
27
|
Vangsted AJ, Klausen TW, Abildgaard N, Andersen NF, Gimsing P, Gregersen H, Nexø BA, Vogel U. Single nucleotide polymorphisms in the promoter region of the IL1B gene influence outcome in multiple myeloma patients treated with high-dose chemotherapy independently of relapse treatment with thalidomide and bortezomib. Ann Hematol 2011; 90:1173-81. [PMID: 21347685 DOI: 10.1007/s00277-011-1194-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 02/07/2011] [Indexed: 01/28/2023]
Abstract
Little is known on the impact of polymorphisms in the IL1B gene on outcome in multiple myeloma. In a population-based study of 348 Danish myeloma patients treated with high-dose treatment (HDT), 146 patients treated with INF-α maintenance treatment, and in 243 patients with relapse, we analysed the impact on outcome of HDT, INF-α maintenance treatment, and treatment with thalidomide and bortezomib at relapse, in relation to the major identified functional polymorphisms in the promoter region of IL1B. The wild-type C-allele of IL1B C-3737T and non-carriage of the IL1B promoter haplotype TGT (-3737(T), -1464(G) and -31(T)), giving high IL1B promoter activity, were associated with longer time-to-treatment failure (TTF) (HR, 1.4 (1.0-1.9) and 1.5 (1.1-2.0)) and overall survival (HR, 1.8 (1.2-2.6) and 1.6 (1.1-2.3)) after HDT. Among INF-α treated patients, a trend towards better TTF was found in patients carrying the wild-type C-allele of IL1B C-3737T (HR, 1.6 (1.1-2.4)). Furthermore, among INF-α treated patients, gene-gene interaction studies on IL1B C-3737T and NFКB1-94ins/del ATTG revealed a fourfold increase in TTF for homozygous carriers of wild-type alleles at both loci as compared to variant allele carriers at both loci. No relation to genotype and outcome was found for relapse patients treated with thalidomide or bortezomib. Our results indicate that a subpopulation of myeloma patients carrying the wild-type C-allele of IL1B C-3737T and non-carriers of the promoter haplotype TGT (-3737(T), -1464(G) and -31(T)) benefit from a better outcome of HDT and INF-α treatment, an effect that may be related to the NF-κB pathway.
Collapse
Affiliation(s)
- Annette J Vangsted
- Department of Oncology and Haematology, Roskilde Hospital, Copenhagen University, Køgevej 9-13, Roskilde, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Waxman AJ, Kuehl M, Balakumaran A, Weiss B, Landgren O. Smoldering (asymptomatic) multiple myeloma: revisiting the clinical dilemma and looking into the future. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2010; 10:248-57. [PMID: 20709660 PMCID: PMC6849209 DOI: 10.3816/clml.2010.n.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent studies show that multiple myeloma (MM) is consistently preceded by an asymptomatic precursor state. Smoldering MM (SMM) is a MM precursor defined by an M-protein concentration >or= 3 g/dL and/or >or= 10% bone marrow plasma cells, in the absence of end-organ damage. Compared with individuals diagnosed with monoclonal gammopathy of undetermined significance (MGUS), patients with SMM have a much higher annual risk of developing MM. However, based on clinical observations, the natural history of SMM varies greatly, from stable MGUS-like disease to highly progressive disease. Using conventional clinical markers, SMM patients can be stratified into 3 risk groups. Importantly, because of considerable molecular heterogeneity, we currently lack reliable markers to predict prognosis for individual SMM patients. Furthermore, until recently, potent drugs with reasonable toxicity profiles have not been available for the development of early MM treatment strategies. Consequently, current clinical guidelines emphasize the application of close clinical monitoring followed by treatment when the patient develops symptomatic MM. This review focuses on novel biomarkers, molecular profiles, and microenvironmental interactions of interest in myelomagenesis. We also discuss how the integration of novel biologic markers and clinical monitoring of SMM could facilitate the development of early treatment strategies for high-risk SMM patients in the future.
Collapse
Affiliation(s)
- Adam J. Waxman
- Medical Oncology Branch, National Cancer Institute,
Bethesda, MD
- Clinical Research Training Program, NIH Clinical Center,
Bethesda, MD
| | - Michael Kuehl
- The Genetics Branch, National Cancer Institute, Bethesda,
MD
| | - Arun Balakumaran
- National Institute of Dental and Craniofacial Research,
Bethesda, MD
| | - Brendan Weiss
- Medical Oncology Branch, National Cancer Institute,
Bethesda, MD
- Hematology-Oncology Service, Department of Medicine, Walter
Reed Army Medical Center, Washington, DC
| | - Ola Landgren
- Medical Oncology Branch, National Cancer Institute,
Bethesda, MD
| |
Collapse
|
29
|
Lemieux C, Cloutier I, Tanguay JF. Estrogen-induced gene expression in bone marrow c-kit+ stem cells and stromal cells: identification of specific biological processes involved in the functional organization of the stem cell niche. Stem Cells Dev 2009; 17:1153-63. [PMID: 19006456 DOI: 10.1089/scd.2007.0251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The recent interest in the role of bone marrow (BM)-derived endothelial progenitor cells (EPCs) and the benefits of estrogen on cardiovascular health brought us to evaluate if estrogen could affect cardiac repair more broadly by regulating biological processes involved in the functional organization of the BM stem cell (SC) niche. To assess such possibility, we evaluated gene expression profiles of BM c-kit+ SCs and CD44+ stromal cells (StroCs) after exposure to a physiological concentration of 17beta-estradiol (17betaE). Data analysis showed that 17betaE altered the expression (>1.5 fold) of 509 and 682 gene probes in c-kit+ SCs and CD44+ StroCs, respectively. Among them, 199 genes in c-kit+ SCs and 283 in CD44+ StroCs were associated to biological process categories of the Gene Ontology classification. Within processes highly regulated by 17betaE, we identified key factors involved in adhesion, migration, proteolysis, and signaling by which 17betaE influences physiological regulation of the functional organization of the SC niche. Together, our results demonstrate that estrogen benefits on cardiovascular health could involve other BM-derived cells than EPCs and that this capacity of estrogen to influence the physiology of the BM SC niche deserves to be investigated clinically.
Collapse
Affiliation(s)
- Caroline Lemieux
- Centre de Recherche, Institut de Cardiologie de Montréal, Montréal, Quebec, Canada
| | | | | |
Collapse
|
30
|
Lust JA, Lacy MQ, Zeldenrust SR, Dispenzieri A, Gertz MA, Witzig TE, Kumar S, Hayman SR, Russell SJ, Buadi FK, Geyer SM, Campbell ME, Kyle RA, Rajkumar SV, Greipp PR, Kline MP, Xiong Y, Moon-Tasson LL, Donovan KA. Induction of a chronic disease state in patients with smoldering or indolent multiple myeloma by targeting interleukin 1{beta}-induced interleukin 6 production and the myeloma proliferative component. Mayo Clin Proc 2009; 84:114-22. [PMID: 19181644 PMCID: PMC2664581 DOI: 10.4065/84.2.114] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To conduct in vitro studies as well as a phase 2 clinical trial in patients with smoldering or indolent multiple myeloma to determine if interleukin 1 (IL-1) inhibitors can delay or prevent active myeloma. PATIENTS AND METHODS Stromal cells were cocultured with IL-1beta-expressing myeloma cells in the presence of dexamethasone, IL-1 receptor antagonist (IL-1Ra), or both. Levels of interleukin 6 (IL-6) and of apoptosis were also quantified. Between November 19, 2002, and May 24, 2007, 47 patients were enrolled in the study and subsequently treated with IL-1Ra. In 25 (53%) of the 47 study patients, low-dose dexamethasone (20 mg/wk) was added. The primary end point was progression-free survival (PFS). RESULTS In vitro, IL-1Ra was superior to dexamethasone at inhibiting IL-6 production; maximal IL-6 inhibition and apoptosis induction were achieved by addition of both IL-1Ra and dexamethasone. In the clinical trial, 3 patients achieved a minor response to IL-1Ra alone; 5 patients achieved a partial response and 4 patients a minor response after addition of dexamethasone. Seven patients showed a decrease in the plasma cell labeling index that paralleled a decrease in high-sensitivity C-reactive protein (hs-CRP) levels. The median overall PFS was 37.5 months. The median PFS for patients without (n=12) or with (n=35) a greater than 15% decrease in 6-month vs baseline hs-CRP levels was 6 months and more than 3 years, respectively (P=.002). Disease stability was maintained in 8 patients who received therapy for more than 4 years. CONCLUSION In patients with smoldering or indolent multiple myeloma who were at risk of progression to active myeloma, treatment with IL-1 inhibitors decreased the myeloma proliferative rate and hs-CRP levels in those who responded, leading to a chronic disease state and an improved PFS. TRIAL REGISTRATION clinicaltrials.gov identifier: NCT00635154.
Collapse
Affiliation(s)
- John A Lust
- Division of Hematology, Mayo Clinic, Rochester, MN 55905.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
The polymorphism IL-1beta T-31C is associated with a longer overall survival in patients with multiple myeloma undergoing auto-SCT. Bone Marrow Transplant 2008; 43:539-45. [PMID: 18997828 DOI: 10.1038/bmt.2008.351] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Proinflammatory cytokines are suspected to play a role in the pathogenesis of multiple myeloma (MM). Therefore, it is possible that inborn genetic variations leading to a modified expression of these cytokines will influence the outcome for these patients. We investigated 348 MM patients undergoing high-dose melphalan treatment followed by Auto-SCT and examined the influence of single nucleotide polymorphisms (SNPs) in genes involved in the inflammatory response. We found that the polymorphism IL-1beta T-31C significantly influenced overall survival (OS; P=0.02) and that carriers of the variant C-allele had a significantly longer survival than homozygous wild-type allele TT-carriers (relative risk 0.6 (95% CI=0.5-0.9); P=0.008). The polymorphisms IL-6 G-174C, IL-10 C592A, PPARgamma2 Pro(12)Ala, COX-2 A-1195G, COX-2 T8473C and NFKB1 ins/del did not influence the OS in this group of patients. Furthermore, homozygous carriers of the variant allele of IL-1beta T-31C were at 1.37-fold (CI=1.05-1.80) increased risk of MM as compared with population-based controls (P=0.02). Our results indicate that IL-1beta is involved in the pathogenesis of MM.
Collapse
|
32
|
Sharma S, Nemeth E, Chen YH, Goodnough J, Huston A, Roodman GD, Ganz T, Lichtenstein A. Involvement of hepcidin in the anemia of multiple myeloma. Clin Cancer Res 2008; 14:3262-7. [PMID: 18519751 DOI: 10.1158/1078-0432.ccr-07-4153] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Hepcidin is a liver-produced peptide implicated in the anemia of inflammation. Because interleukin (IL)-6 is a potent inducer of hepcidin expression and its levels are elevated in multiple myeloma, we studied the role of hepcidin in the anemia of multiple myeloma. EXPERIMENTAL DESIGN Urinary hepcidin and serum levels of IL-6, ferritin, C-reactive protein, tumor necrosis factor-alpha, and IL-1 beta were studied in newly diagnosed myeloma patients. In vitro hepcidin induction assay was assessed by real-time PCR assay. RESULTS Pretreatment urinary hepcidin levels in 44 patients with stage III multiple myeloma were 3-fold greater than normal controls. In the subset of multiple myeloma patients without renal insufficiency (n = 27), a marked inverse correlation was seen between hemoglobin at diagnosis and urinary hepcidin level (P = 0.014) strongly supporting a causal relationship between up-regulated hepcidin expression and anemia. The urinary hepcidin also significantly (P < 0.05) correlated with serum ferritin and C-reactive protein, whereas its correlation with serum IL-6 levels was of borderline significance (P = 0.06). Sera from 14 multiple myeloma patients, with known elevated urinary hepcidin, significantly induced hepcidin mRNA in the Hep3B cells, whereas normal sera had no effect. For 10 patients, the ability of anti-IL-6 and anti-IL-6 receptor antibodies to prevent the serum-induced hepcidin RNA was tested. In 6 of these patients, hepcidin induction was abrogated by the anti-IL-6 antibodies, but in the other 4 patients, the neutralizing antibodies had no effect. CONCLUSIONS These results indicate hepcidin is up-regulated in multiple myeloma patients by both IL-6-dependent and IL-6-independent mechanisms and may play a role in the anemia of multiple myeloma.
Collapse
Affiliation(s)
- Sanjai Sharma
- West Los Angeles VA-University of California-Los Angeles Medical Center, Los Angeles, CA 90073, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Patients with monoclonal gammopathy of undetermined significance (MGUS) are at continuous risk of progression. Each year, 1% progress, usually to active multiple myeloma (MM).1 Such patients must be monitored for life. Asymptomatic smoldering multiple myeloma (SMM) has an even greater risk of progression to MM. Recently reported strategies improve our ability to estimate the risk of MM in these patients.
Collapse
|
34
|
Kline M, Donovan K, Wellik L, Lust C, Jin W, Moon-Tasson L, Xiong Y, Witzig TE, Kumar S, Rajkumar SV, Lust JA. Cytokine and chemokine profiles in multiple myeloma; significance of stromal interaction and correlation of IL-8 production with disease progression. Leuk Res 2007; 31:591-8. [PMID: 16879867 DOI: 10.1016/j.leukres.2006.06.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Revised: 06/12/2006] [Accepted: 06/13/2006] [Indexed: 11/24/2022]
Abstract
Multiple myeloma (MM) is a product of interactions between tumor plasma cells and multiple cell types native to the bone marrow (BM). We have used antibody array technology to examine the proteins produced by BM stromal cells in response to stimulation by BM taken from patients diagnosed with monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and MM. We observed increased production of the chemokine IL-8 by stromal cells co-cultured with supernatants from bone marrow cells of patients with active myeloma. IL-8 production is correlated with active disease and is dependent upon IL-1beta and NF-kappaB signaling. Consistent with the pro-angiogenic activity of IL-8, increased BM microvessel density (MVD) correlated with stimulation of stromal cell IL-8 production. In addition, the majority of MM cell lines and MM patient plasma cells were found to express IL-8 receptors CXCR1 and CXCR2. We conclude that stromal cell IL-8 production parallels MM disease activity, is IL-1beta induced, and correlates with bone marrow angiogenesis.
Collapse
Affiliation(s)
- Michael Kline
- Division of Hematology and Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Treatment of Newly Diagnosed Multiple Myeloma Based on Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART): Consensus Statement. Mayo Clin Proc 2007. [DOI: 10.1016/s0025-6196(11)61029-x] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Dispenzieri A, Rajkumar SV, Gertz MA, Fonseca R, Lacy MQ, Bergsagel PL, Kyle RA, Greipp PR, Witzig TE, Reeder CB, Lust JA, Russell SJ, Hayman SR, Roy V, Kumar S, Zeldenrust SR, Dalton RJ, Stewart AK. Treatment of newly diagnosed multiple myeloma based on Mayo Stratification of Myeloma and Risk-adapted Therapy (mSMART): consensus statement. Mayo Clin Proc 2007; 82:323-41. [PMID: 17352369 DOI: 10.4065/82.3.323] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple myeloma is a neoplastic plasma cell dyscrasia that on a yearly basis affects nearly 17,000 individuals and kills more than 11,000. Although no cure exists, many effective treatments are available that prolong survival and improve the quality of life of patients with this disease. The purpose of this consensus is to offer a simplified, evidence-based algorithm of decision making for patients with newly diagnosed myeloma. In cases in which evidence is lacking, our team of 18 Mayo Clinic myeloma experts reached a consensus on what therapy could generally be recommended. The focal point of our strategy revolves around risk stratification. Although a multitude of risk factors have been identified throughout the years, including age, tumor burden, renal function, lactate dehydrogenase, beta2-microglobulin, and serum albumin, our group has now recognized and endorsed a genetic stratification and patient functional status for treatment.
Collapse
Affiliation(s)
- Angela Dispenzieri
- Division of Hematology, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|