1
|
Maura F, Coffey DG, Stein CK, Braggio E, Ziccheddu B, Sharik ME, Du MT, Tafoya Alvarado Y, Shi CX, Zhu YX, Meermeier EW, Morgan GJ, Landgren O, Bergsagel PL, Chesi M. The genomic landscape of Vk*MYC myeloma highlights shared pathways of transformation between mice and humans. Nat Commun 2024; 15:3844. [PMID: 38714690 PMCID: PMC11076575 DOI: 10.1038/s41467-024-48091-w] [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: 07/12/2023] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
Multiple myeloma (MM) is a heterogeneous disease characterized by frequent MYC translocations. Sporadic MYC activation in the germinal center of genetically engineered Vk*MYC mice is sufficient to induce plasma cell tumors in which a variety of secondary mutations are spontaneously acquired and selected over time. Analysis of 119 Vk*MYC myeloma reveals recurrent copy number alterations, structural variations, chromothripsis, driver mutations, apolipoprotein B mRNA-editing enzyme, catalytic polypeptide (APOBEC) mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identify frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC. In summary, here we credential the Vk*MYC mouse as a unique resource to explore MM genomic evolution and describe a fully annotated collection of diverse and immortalized murine MM tumors.
Collapse
Affiliation(s)
| | - David G Coffey
- Division of Myeloma, University of Miami, Miami, FL, USA
| | - Caleb K Stein
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Esteban Braggio
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Meaghen E Sharik
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Megan T Du
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yuliza Tafoya Alvarado
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Chang-Xin Shi
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yuan Xiao Zhu
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Erin W Meermeier
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Gareth J Morgan
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Ola Landgren
- Division of Myeloma, University of Miami, Miami, FL, USA
| | - P Leif Bergsagel
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Marta Chesi
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA.
| |
Collapse
|
2
|
Maura F, Coffey DG, Stein CK, Braggio E, Ziccheddu B, Sharik ME, Du M, Alvarado YT, Shi CX, Zhu YX, Meermeier EW, Morgan GJ, Landgren O, Leif Bergsagel P, Chesi M. The Vk*MYC Mouse Model recapitulates human multiple myeloma evolution and genomic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550482. [PMID: 37546905 PMCID: PMC10402028 DOI: 10.1101/2023.07.25.550482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Despite advancements in profiling multiple myeloma (MM) and its precursor conditions, there is limited information on mechanisms underlying disease progression. Clincal efforts designed to deconvolute such mechanisms are challenged by the long lead time between monoclonal gammopathy and its transformation to MM. MM mouse models represent an opportunity to overcome this temporal limitation. Here, we profile the genomic landscape of 118 genetically engineered Vk*MYC MM and reveal that it recapitulates the genomic heterogenenity and life history of human MM. We observed recurrent copy number alterations, structural variations, chromothripsis, driver mutations, APOBEC mutational activity, and a progressive decrease in immunoglobulin transcription that inversely correlates with proliferation. Moreover, we identified frequent insertional mutagenesis by endogenous retro-elements as a murine specific mechanism to activate NF-kB and IL6 signaling pathways shared with human MM. Despite the increased genomic complexity associated with progression, advanced tumors remain dependent on MYC expression, that drives the progression of monoclonal gammopathy to MM.
Collapse
|
3
|
Cencini E, Sicuranza A, Ciofini S, Fabbri A, Bocchia M, Gozzetti A. Tumor-Associated Macrophages in Multiple Myeloma: Key Role in Disease Biology and Potential Therapeutic Implications. Curr Oncol 2023; 30:6111-6133. [PMID: 37504315 PMCID: PMC10378698 DOI: 10.3390/curroncol30070455] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple myeloma (MM) is characterized by multiple relapse and, despite the introduction of novel therapies, the disease becomes ultimately drug-resistant. The tumor microenvironment (TME) within the bone marrow niche includes dendritic cells, T-cytotoxic, T-helper, reactive B-lymphoid cells and macrophages, with a complex cross-talk between these cells and the MM tumor cells. Tumor-associated macrophages (TAM) have an important role in the MM pathogenesis, since they could promote plasma cells proliferation and angiogenesis, further supporting MM immune evasion and progression. TAM are polarized towards M1 (classically activated, antitumor activity) and M2 (alternatively activated, pro-tumor activity) subtypes. Many studies demonstrated a correlation between TAM, disease progression, drug-resistance and reduced survival in lymphoproliferative neoplasms, including MM. MM plasma cells in vitro could favor an M2 TAM polarization. Moreover, a possible correlation between the pro-tumor effect of M2 TAM and a reduced sensitivity to proteasome inhibitors and immunomodulatory drugs was hypothesized. Several clinical studies confirmed CD68/CD163 double-positive M2 TAM were associated with increased microvessel density, chemoresistance and reduced survival, independently of the MM stage. This review provided an overview of the biology and clinical relevance of TAM in MM, as well as a comprehensive evaluation of a potential TAM-targeted immunotherapy.
Collapse
Affiliation(s)
- Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Anna Sicuranza
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Sara Ciofini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alessandro Gozzetti
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| |
Collapse
|
4
|
Moscvin M, Evans B, Bianchi G. Dissecting molecular mechanisms of immune microenvironment dysfunction in multiple myeloma and precursor conditions. JOURNAL OF CANCER METASTASIS AND TREATMENT 2023; 9:17. [PMID: 38213954 PMCID: PMC10783205 DOI: 10.20517/2394-4722.2022.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Multiple myeloma (MM) is a disease of clonally differentiated plasma cells. MM is almost always preceded by precursor conditions, monoclonal gammopathy of unknown significance (MGUS), and smoldering MM (SMM) through largely unknown molecular events. Genetic alterations of the malignant plasma cells play a critical role in patient clinical outcomes. Del(17p), t(4;14), and additional chromosomal alterations such as del(1p32), gain(1q) and MYC translocations are involved in active MM evolution. Interestingly, these genetic alterations appear strikingly similar in transformed plasma cell (PC) clones from MGUS, SMM, and MM stages. Recent studies show that effectors of the innate and adaptive immune response show marked dysfunction and skewing towards a tolerant environment that favors disease progression. The MM myeloid compartment is characterized by myeloid-derived suppressor cells (MDSCs), dendritic cells as well as M2-like phenotype macrophages that promote immune evasion. Major deregulations are found in the lymphoid compartment as well, with skewing towards immune tolerant Th17 and Treg and inhibition of CD8+ cytotoxic and CD4+ activated effector T cells. In summary, this review will provide an overview of the complex cross-talk between MM plasma cells and immune cells in the microenvironment and the molecular mechanisms promoting progression from precursor states to full-blown myeloma.
Collapse
Affiliation(s)
- Maria Moscvin
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Evans
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
| | - Giada Bianchi
- Department of Medicine, Division of Hematology, Brigham and Womens Hospital, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Lourenço D, Lopes R, Pestana C, Queirós AC, João C, Carneiro EA. Patient-Derived Multiple Myeloma 3D Models for Personalized Medicine-Are We There Yet? Int J Mol Sci 2022; 23:12888. [PMID: 36361677 PMCID: PMC9657251 DOI: 10.3390/ijms232112888] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/03/2023] Open
Abstract
Despite the wide variety of existing therapies, multiple myeloma (MM) remains a disease with dismal prognosis. Choosing the right treatment for each patient remains one of the major challenges. A new approach being explored is the use of ex vivo models for personalized medicine. Two-dimensional culture or animal models often fail to predict clinical outcomes. Three-dimensional ex vivo models using patients' bone marrow (BM) cells may better reproduce the complexity and heterogeneity of the BM microenvironment. Here, we review the strengths and limitations of currently existing patient-derived ex vivo three-dimensional MM models. We analyze their biochemical and biophysical properties, molecular and cellular characteristics, as well as their potential for drug testing and identification of disease biomarkers. Furthermore, we discuss the remaining challenges and give some insight on how to achieve a more biomimetic and accurate MM BM model. Overall, there is still a need for standardized culture methods and refined readout techniques. Including both myeloma and other cells of the BM microenvironment in a simple and reproducible three-dimensional scaffold is the key to faithfully mapping and examining the relationship between these players in MM. This will allow a patient-personalized profile, providing a powerful tool for clinical and research applications.
Collapse
Affiliation(s)
- Diana Lourenço
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Lopes
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Centre of Statistics and Its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana C. Queirós
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Cristina João
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| |
Collapse
|
6
|
Yue Y, Cao Y, Wang F, Zhang N, Qi Z, Mao X, Guo S, Li F, Guo Y, Lin Y, Dong W, Huang Y, Gu W. Bortezomib-resistant multiple myeloma patient-derived xenograft is sensitive to anti-CD47 therapy. Leuk Res 2022; 122:106949. [PMID: 36113267 DOI: 10.1016/j.leukres.2022.106949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy due to its frequent drug resistance and relapse. Cluster of Differentiation 47 (CD47) is reported to be highly expressed on MM cells, suggesting that the blockade of CD47 signaling pathway could be a potential therapeutic candidate for MM. In this study, we developed a bortezomib-resistant myeloma patient-derived xenograft (PDX) from an extramedullary pleural effusion myeloma patient sample. Notably, anti-CD47 antibody treatments significantly inhibited tumor growth not only in MM cell line-derived models, including MM.1S and NCI-H929, but also in the bortezomib-resistant MM PDX model. Flow cytometric data showed that anti-CD47 therapy promoted the polarization of tumor-associated macrophages from an M2- to an M1-like phenotype. In addition, anti-CD47 therapy decreased the expression of pro-angiogenic factors, increased the expression of anti-angiogenic factors, and improved tumor vascular function, suggesting that anti-CD47 therapy induces tumor vascular normalization. Taken together, these data show that anti-CD47 antibody therapy reconditions the tumor immune microenvironment and inhibits the tumor growth of bortezomib-resistant myeloma PDX. Our findings suggest that CD47 is a potential new target to treat bortezomib-resistant MM.
Collapse
Affiliation(s)
- Yanhua Yue
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Fei Wang
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Naidong Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Ziwei Qi
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Xunyuan Mao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Shuxin Guo
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Feng Li
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yanting Guo
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yan Lin
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Weimin Dong
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yuhui Huang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China.
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China.
| |
Collapse
|
7
|
Yi Z, Ma T, Liu J, Tie W, Li Y, Bai J, Li L, Zhang L. The yin–yang effects of immunity: From monoclonal gammopathy of undetermined significance to multiple myeloma. Front Immunol 2022; 13:925266. [PMID: 35958625 PMCID: PMC9357873 DOI: 10.3389/fimmu.2022.925266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/30/2022] [Indexed: 01/10/2023] Open
Abstract
Multiple myeloma (MM) is the third most common malignant neoplasm of the hematological system. It often develops from monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) precursor states. In this process, the immune microenvironment interacts with the MM cells to exert yin and yang effects, promoting tumor progression on the one hand and inhibiting it on the other. Despite significant therapeutic advances, MM remains incurable, and the main reason for this may be related to the complex and variable immune microenvironment. Therefore, it is crucial to investigate the dynamic relationship between the immune microenvironment and tumors, to elucidate the molecular mechanisms of different factors in the microenvironment, and to develop novel therapeutic agents targeting the immune microenvironment of MM. In this paper, we review the latest research progress and describe the dual influences of the immune microenvironment on the development and progression of MM from the perspective of immune cells and molecules.
Collapse
Affiliation(s)
- Zhigang Yi
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Pediatric Orthopedics and Pediatrics Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenting Tie
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanhong Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jun Bai
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| |
Collapse
|
8
|
Petrusca DN, Lee KP, Galson DL. Role of Sphingolipids in Multiple Myeloma Progression, Drug Resistance, and Their Potential as Therapeutic Targets. Front Oncol 2022; 12:925807. [PMID: 35756630 PMCID: PMC9213658 DOI: 10.3389/fonc.2022.925807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is an incapacitating hematological malignancy characterized by accumulation of cancerous plasma cells in the bone marrow (BM) and production of an abnormal monoclonal protein (M-protein). The BM microenvironment has a key role in myeloma development by facilitating the growth of the aberrant plasma cells, which eventually interfere with the homeostasis of the bone cells, exacerbating osteolysis and inhibiting osteoblast differentiation. Recent recognition that metabolic reprograming has a major role in tumor growth and adaptation to specific changes in the microenvironmental niche have led to consideration of the role of sphingolipids and the enzymes that control their biosynthesis and degradation as critical mediators of cancer since these bioactive lipids have been directly linked to the control of cell growth, proliferation, and apoptosis, among other cellular functions. In this review, we present the recent progress of the research investigating the biological implications of sphingolipid metabolism alterations in the regulation of myeloma development and its progression from the pre-malignant stage and discuss the roles of sphingolipids in in MM migration and adhesion, survival and proliferation, as well as angiogenesis and invasion. We introduce the current knowledge regarding the role of sphingolipids as mediators of the immune response and drug-resistance in MM and tackle the new developments suggesting the manipulation of the sphingolipid network as a novel therapeutic direction for MM.
Collapse
Affiliation(s)
- Daniela N Petrusca
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kelvin P Lee
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Deborah L Galson
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, McGowan Institute for Regenerative Medicine, HCC Research Pavilion, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Sun J, Park C, Guenthner N, Gurley S, Zhang L, Lubben B, Adebayo O, Bash H, Chen Y, Maksimos M, Muz B, Azab AK. Tumor-associated macrophages in multiple myeloma: advances in biology and therapy. J Immunother Cancer 2022; 10:e003975. [PMID: 35428704 PMCID: PMC9014078 DOI: 10.1136/jitc-2021-003975] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a cancer of plasma cells in the bone marrow (BM) and represents the second most common hematological malignancy in the world. The MM tumor microenvironment (TME) within the BM niche consists of a wide range of elements which play important roles in supporting MM disease progression, survival, proliferation, angiogenesis, as well as drug resistance. Together, the TME fosters an immunosuppressive environment in which immune recognition and response are repressed. Macrophages are a central player in the immune system with diverse functions, and it has been long established that macrophages play a critical role in both inducing direct and indirect immune responses in cancer. Tumor-associated macrophages (TAMs) are a major population of cells in the tumor site. Rather than contributing to the immune response against tumor cells, TAMs in many cancers are found to exhibit protumor properties including supporting chemoresistance, tumor proliferation and survival, angiogenesis, immunosuppression, and metastasis. Targeting TAM represents a novel strategy for cancer immunotherapy, which has potential to indirectly stimulate cytotoxic T cell activation and recruitment, and synergize with checkpoint inhibitors and chemotherapies. In this review, we will provide an updated and comprehensive overview into the current knowledge on the roles of TAMs in MM, as well as the therapeutic targets that are being explored as macrophage-targeted immunotherapy, which may hold key to future therapeutics against MM.
Collapse
Affiliation(s)
- Jennifer Sun
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| | - Chaelee Park
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Nicole Guenthner
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Shannon Gurley
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Luna Zhang
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| | - Berit Lubben
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Ola Adebayo
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Hannah Bash
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Yixuan Chen
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Mina Maksimos
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Cencini E, Fabbri A, Sicuranza A, Gozzetti A, Bocchia M. The Role of Tumor-Associated Macrophages in Hematologic Malignancies. Cancers (Basel) 2021; 13:cancers13143597. [PMID: 34298810 PMCID: PMC8304632 DOI: 10.3390/cancers13143597] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Tumor-associated macrophages (TAM) represent a leading component of the tumor microenvironment in hematologic malignancies. TAM could display antitumor activity or, conversely, could contribute to tumor growth and survival, depending on their polarization. TAM are polarized towards form M1, with a pro-inflammatory phenotype and an antineoplastic activity, or M2, with an alternately activated phenotype, associated with a poor outcome in patients presenting with leukemia, lymphoma or multiple myeloma. The molecular mechanisms of TAM in different types of hematologic malignancies are different due to the peculiar microenvironment of each disease. TAM could contribute to tumor progression, reduced apoptosis and angiogenesis; a different TAM polarization could explain a reduced treatment response in patients with a similar disease subtype. The aim of our review is to better define the role of TAM in patients with leukemia, lymphoma or multiple myeloma. Finally, we would like to focus on TAM as a possible target for antineoplastic therapy. Abstract The tumor microenvironment includes dendritic cells, T-cytotoxic, T-helper, reactive B-lymphoid cells and macrophages; these reactive cells could interplay with malignant cells and promote tumor growth and survival. Among its cellular components, tumor-associated macrophages (TAM) represent a component of the innate immune system and play an important role, especially in hematologic malignancies. Depending on the stimuli that trigger their activation, TAM are polarized towards form M1, contributing to antitumor responses, or M2, associated with tumor progression. Many studies demonstrated a correlation between TAM, disease progression and the patient’s outcome in lymphoproliferative neoplasms, such as Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL), even if with conflicting results. A critical hurdle to overcome is surely represented by the heterogeneity in the choice of the optimal markers and methods used for TAM analysis (gene-expression profile vs. immunohistochemistry, CD163vs. CD68vs. CD163/CD68 double-positive cells). TAM have been recently linked to the development and progression of multiple myeloma and leukemia, with a critical role in the homing of malignant cells, drug resistance, immune suppression and angiogenesis. As such, this review will summarize the role of TAM in different hematologic malignancies, focusing on the complex interplay between TAM and tumor cells, the prognostic value of TAM and the possible TAM-targeted therapeutic strategies.
Collapse
|
11
|
Macrophages in multiple myeloma: key roles and therapeutic strategies. Cancer Metastasis Rev 2021; 40:273-284. [PMID: 33404860 DOI: 10.1007/s10555-020-09943-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Macrophages are a vital component of the tumour microenvironment and crucial mediators of tumour progression. In the last decade, significant strides have been made in understanding the crucial functional roles played by macrophages in the development of the plasma cell (PC) malignancy, multiple myeloma (MM). Whilst the interaction between MM PC and stromal cells within the bone marrow (BM) microenvironment has been extensively studied, we are only just starting to appreciate the multifaceted roles played by macrophages in disease progression. Accumulating evidence demonstrates that macrophage infiltration is associated with poor overall survival in MM. Indeed, macrophages influence numerous pathways critical for the initiation and progression of MM, including homing of malignant cells to BM, tumour cell growth and survival, drug resistance, angiogenesis and immune suppression. As such, therapeutic strategies aimed at targeting macrophages within the BM niche have promise in the clinical setting. This review will discuss the functions elicited by macrophages throughout different stages of MM and provide a comprehensive evaluation of potential macrophage-targeted therapies.
Collapse
|
12
|
Engineering a Humanised Niche to Support Human Haematopoiesis in Mice: Novel Opportunities in Modelling Cancer. Cancers (Basel) 2020; 12:cancers12082205. [PMID: 32781703 PMCID: PMC7466161 DOI: 10.3390/cancers12082205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/18/2022] Open
Abstract
Despite the bone marrow microenvironment being widely recognised as a key player in cancer research, the current animal models that represent a human haematopoietic system lack the contribution of the humanised marrow microenvironment. Here we describe a murine model that relies on the combination of an orthotopic humanised tissue-engineered bone construct (ohTEBC) with patient-specific bone marrow (BM) cells to create a humanised bone marrow (hBM) niche capable of supporting the engraftment of human haematopoietic cells. Results showed that this model supports the engraftment of human CD34+ cells from a healthy BM with human haematopoietic cells migrating into the mouse BM, human BM compartment, spleen and peripheral blood. We compared these results with the engraftment capacity of human CD34+ cells obtained from patients with multiple myeloma (MM). We demonstrated that CD34+ cells derived from a diseased BM had a reduced engraftment potential compared to healthy patients and that a higher cell dose is required to achieve engraftment of human haematopoietic cells in peripheral blood. Finally, we observed that hematopoietic cells obtained from the mobilised peripheral blood of patients yields a higher number of CD34+, overcoming this problem. In conclusion, this humanised mouse model has potential as a unique and patient-specific pre-clinical platform for the study of tumour–microenvironment interactions, including human bone and haematopoietic cells, and could, in the future, serve as a drug testing platform.
Collapse
|
13
|
Ria R, Melaccio A, Racanelli V, Vacca A. Anti-VEGF Drugs in the Treatment of Multiple Myeloma Patients. J Clin Med 2020; 9:E1765. [PMID: 32517267 PMCID: PMC7355441 DOI: 10.3390/jcm9061765] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
The interaction between the bone marrow microenvironment and plasma cells plays an essential role in multiple myeloma progression and drug resistance. The vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway in vascular endothelial cells activates and promotes angiogenesis. Moreover, VEGF activates and promotes vasculogenesis and vasculogenic mimicry when it interacts with VEGF receptors expressed in precursor cells and inflammatory cells, respectively. In myeloma bone marrow, VEGF and VEGF receptor expression are upregulated and hyperactive in the stromal and tumor cells. It has been demonstrated that several antiangiogenic agents can effectively target VEGF-related pathways in the preclinical phase. However, they are not successful in treating multiple myeloma, probably due to the vicarious action of other cytokines and signaling pathways. Thus, the simultaneous blocking of multiple cytokine pathways, including the VEGF/VEGFR pathway, may represent a valid strategy to treat multiple myeloma. This review aims to summarize recent advances in understanding the role of the VEGF/VEGFR pathway in multiple myeloma, and mainly focuses on the transcription pathway and on strategies that target this pathway.
Collapse
Affiliation(s)
- Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (V.R.); (A.V.)
| | | | | | | |
Collapse
|
14
|
Microbiota-Propelled T Helper 17 Cells in Inflammatory Diseases and Cancer. Microbiol Mol Biol Rev 2020; 84:84/2/e00064-19. [PMID: 32132244 DOI: 10.1128/mmbr.00064-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Technologies allowing genetic sequencing of the human microbiome are opening new realms to discovery. The host microbiota substantially impacts immune responses both in immune-mediated inflammatory diseases (IMIDs) and in tumors affecting tissues beyond skin and mucosae. However, a mechanistic link between host microbiota and cancer or IMIDs has not been well established. Here, we propose T helper 17 (TH17) lymphocytes as the connecting factor between host microbiota and rheumatoid or psoriatic arthritides, multiple sclerosis, breast or ovarian cancer, and multiple myeloma. We theorize that similar mechanisms favor the expansion of gut-borne TH17 cells and their deployment at the site of inflammation in extraborder IMIDs and tumors, where TH17 cells are driving forces. Thus, from a pathogenic standpoint, tumors may share mechanistic routes with IMIDs. A review of similarities and divergences in microbiota-TH17 cell interactions in IMIDs and cancer sheds light on previously ignored pathways in either one of the two groups of pathologies and identifies novel therapeutic avenues.
Collapse
|
15
|
Chesi M, Stein CK, Garbitt VM, Sharik ME, Asmann YW, Bergsagel M, Riggs DL, Welsh SJ, Meermeier EW, Kumar SK, Braggio E, Bergsagel PL. Monosomic loss of MIR15A/MIR16-1 is a driver of multiple myeloma proliferation and disease progression. Blood Cancer Discov 2020; 1:68-81. [PMID: 32954360 DOI: 10.1158/0008-5472.bcd-19-0068] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The most common genetic abnormality in multiple myeloma (MM) is the deletion of chromosome 13, seen in almost half of newly diagnosed patients. Unlike chronic lymphocytic leukemia, where a recurrent minimally deleted region including MIR15A/MIR16-1 has been mapped, the deletions in MM predominantly involve the entire chromosome and no specific driver gene has been identified. Additional candidate loci include RB1 and DIS3, but while biallelic deletion of RB1 is associated with disease progression, DIS3 is a common essential gene and complete inactivation is not observed. The Vk*MYC transgenic mouse model of MM spontaneously acquires del(14), syntenic to human chromosome 13, and Rb1 complete inactivation, but not Dis3 mutations. Taking advantage of this model, we explored the role in MM initiation and progression of two candidate loci on chromosome 13: RB1 and MIR15A/MIR16-1. Monoallelic deletion of Mir15a/Mir16-1 but not Rb1 was sufficient to accelerate the development of monoclonal gammopathy in wildtype mice, and the progression of MM in Vk*MYC mice, resulting in increased expression of Mir15a/Mir16-1 target genes and plasma cell proliferation, which was similarly observed in patients with MM.
Collapse
Affiliation(s)
- Marta Chesi
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Caleb K Stein
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Victoria M Garbitt
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Meaghen E Sharik
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Yan W Asmann
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | - Matteo Bergsagel
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Daniel L Riggs
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Seth J Welsh
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Erin W Meermeier
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - Shaji K Kumar
- Division of Hematology and Medical Oncology, Mayo Clinic, Rochester, MN
| | - Esteban Braggio
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| | - P Leif Bergsagel
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
16
|
Romano A, Parrinello NL, Simeon V, Puglisi F, La Cava P, Bellofiore C, Giallongo C, Camiolo G, D'Auria F, Grieco V, Larocca F, Barbato A, Cambria D, La Spina E, Tibullo D, Palumbo GA, Conticello C, Musto P, Di Raimondo F. High-density neutrophils in MGUS and multiple myeloma are dysfunctional and immune-suppressive due to increased STAT3 downstream signaling. Sci Rep 2020; 10:1983. [PMID: 32029833 PMCID: PMC7005058 DOI: 10.1038/s41598-020-58859-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
To understand neutrophil impairment in the progression from MGUS through active MM, we investigated the function of mature, high-density neutrophils (HDNs), isolated from peripheral blood. In 7 MM, 3 MGUS and 3 healthy subjects by gene expression profile, we identified a total of 551 upregulated and 343 downregulated genes in MM-HDN, involved in chemokine signaling pathway and FC-gamma receptor mediated phagocytosis conveying in the activation of STAT proteins. In a series of 60 newly diagnosed MM and 30 MGUS patients, by flow-cytometry we found that HDN from MM, and to a lesser extend MGUS, had an up-regulation of the inducible FcγRI (also known as CD64) and a down-regulation of the constitutive FcγRIIIa (also known as CD16) together with a reduced phagocytic activity and oxidative burst, associated to increased immune-suppression that could be reverted by arginase inhibitors in co-culture with lymphocytes. In 43 consecutive newly-diagnosed MM patients, who received first-line treatment based on bortezomib, thalidomide and dexamethasone, high CD64 could identify at diagnosis patients with inferior median overall survival (39.5 versus 86.7 months, p = 0.04). Thus, HDNs are significantly different among healthy, MGUS and MM subjects. In both MGUS and MM neutrophils may play a role in supporting both the increased susceptibility to infection and the immunological dysfunction that leads to tumor progression.
Collapse
Affiliation(s)
- A Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - N L Parrinello
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| | - V Simeon
- Laboratory of Pre-Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture (Pz), Potenza, Italy
- Department of Mental Health and Preventive Medicine, Medical Statistics Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - F Puglisi
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - P La Cava
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - C Bellofiore
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - C Giallongo
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - G Camiolo
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - F D'Auria
- Laboratory of Pre-Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture (Pz), Potenza, Italy
| | - V Grieco
- Laboratory of Pre-Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture (Pz), Potenza, Italy
| | - F Larocca
- Laboratory of Pre-Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture (Pz), Potenza, Italy
| | - A Barbato
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - D Cambria
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - E La Spina
- Biometec, Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, Catania, Italy
| | - D Tibullo
- Biometec, Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, Catania, Italy
| | - G A Palumbo
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| | - C Conticello
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy
| | - P Musto
- Laboratory of Pre-Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture (Pz), Potenza, Italy
- Chair and Unit of Hematology and Stem Cell Transplantation, Aldo Moro University, Bari, Italy
| | - F Di Raimondo
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy.
- Division of Hematology, Azienda Ospedaliera Policlinico e Vittorio Emanuele di Catania, Catania, Italy.
- Department of Mental Health and Preventive Medicine, Medical Statistics Unit, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
17
|
Immunotherapeutics in Multiple Myeloma: How Can Translational Mouse Models Help? JOURNAL OF ONCOLOGY 2019; 2019:2186494. [PMID: 31093282 PMCID: PMC6481018 DOI: 10.1155/2019/2186494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/04/2019] [Indexed: 12/30/2022]
Abstract
Multiple myeloma (MM) is usually diagnosed in older adults at the time of immunosenescence, a collection of age-related changes in the immune system that contribute to increased susceptibility to infection and cancer. The MM tumor microenvironment and cumulative chemotherapies also add to defects in immunity over the course of disease. In this review we discuss how mouse models have furthered our understanding of the immune defects caused by MM and enabled immunotherapeutics to progress to clinical trials, but also question the validity of using immunodeficient models for these purposes. Immunocompetent models, in particular the 5T series and Vk⁎MYC models, are increasingly being utilized in preclinical studies and are adding to our knowledge of not only the adaptive immune system but also how the innate system might be enhanced in anti-MM activity. Finally we discuss the concept of immune profiling to target patients who might benefit the most from immunotherapeutics, and the use of humanized mice and 3D culture systems for personalized medicine.
Collapse
|
18
|
Calcinotto A, Brevi A, Chesi M, Ferrarese R, Garcia Perez L, Grioni M, Kumar S, Garbitt VM, Sharik ME, Henderson KJ, Tonon G, Tomura M, Miwa Y, Esplugues E, Flavell RA, Huber S, Canducci F, Rajkumar VS, Bergsagel PL, Bellone M. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat Commun 2018; 9:4832. [PMID: 30510245 PMCID: PMC6277390 DOI: 10.1038/s41467-018-07305-8] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/15/2018] [Indexed: 12/31/2022] Open
Abstract
The gut microbiota has been causally linked to cancer, yet how intestinal microbes influence progression of extramucosal tumors is poorly understood. Here we provide evidence implying that Prevotella heparinolytica promotes the differentiation of Th17 cells colonizing the gut and migrating to the bone marrow (BM) of transgenic Vk*MYC mice, where they favor progression of multiple myeloma (MM). Lack of IL-17 in Vk*MYC mice, or disturbance of their microbiome delayed MM appearance. Similarly, in smoldering MM patients, higher levels of BM IL-17 predicted faster disease progression. IL-17 induced STAT3 phosphorylation in murine plasma cells, and activated eosinophils. Treatment of Vk*MYC mice with antibodies blocking IL-17, IL-17RA, and IL-5 reduced BM accumulation of Th17 cells and eosinophils and delayed disease progression. Thus, in Vk*MYC mice, commensal bacteria appear to unleash a paracrine signaling network between adaptive and innate immunity that accelerates progression to MM, and can be targeted by already available therapies.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Arianna Brevi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Roberto Ferrarese
- Laboratory of Microbiology, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Laura Garcia Perez
- Molekulare Immunologie und Gastroenterologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Matteo Grioni
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - Victoria M Garbitt
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Meaghen E Sharik
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | | | - Giovanni Tonon
- Division of Molecular Oncology, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Michio Tomura
- Faculty of Pharmacy, Osaka Ohtani University, Osaka, 584-8540, Japan
| | | | - Enric Esplugues
- Department of Immunobiology, School of Medicine, and Howard Hughes Medical Institute Yale University, New Haven, 06520, USA
| | - Richard A Flavell
- Department of Immunobiology, School of Medicine, and Howard Hughes Medical Institute Yale University, New Haven, 06520, USA
| | - Samuel Huber
- Molekulare Immunologie und Gastroenterologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Filippo Canducci
- Laboratory of Microbiology, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, 21100, Italy
| | - Vincent S Rajkumar
- Division of Hematology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - P Leif Bergsagel
- Comprehensive Cancer Center, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Matteo Bellone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy.
| |
Collapse
|
19
|
Castella B, Melaccio A, Foglietta M, Riganti C, Massaia M. Vγ9Vδ2 T Cells as Strategic Weapons to Improve the Potency of Immune Checkpoint Blockade and Immune Interventions in Human Myeloma. Front Oncol 2018; 8:508. [PMID: 30460198 PMCID: PMC6232124 DOI: 10.3389/fonc.2018.00508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
The advent of immune checkpoint (ICP) blockade has introduced an unprecedented paradigm shift in the treatment of cancer. Though very promising, there is still a substantial proportion of patients who do not respond or develop resistance to ICP blockade. In vitro and in vivo models are eagerly needed to identify mechanisms to maximize the immune potency of ICP blockade and overcome primary and acquired resistance to ICP blockade. Vγ9Vδ2 T cells isolated from the bone marrow (BM) from multiple myeloma (MM) are excellent tools to investigate the mechanisms of resistance to PD-1 blockade and to decipher the network of mutual interactions between PD-1 and the immune suppressive tumor microenvironment (TME). Vγ9Vδ2 T cells can easily be interrogated to dissect the progressive immune competence impairment generated in the TME by the long-lasting exposure to myeloma cellss. BM MM Vγ9Vδ2 T cells are PD-1+ and anergic to phosphoantigen (pAg) stimulation; notably, single agent PD-1 blockade is insufficient to fully recover their anti-tumor activity in vitro indicating that additional players are involved in the anergy of Vγ9Vδ2 T cells. In this mini-review we will discuss the value of Vγ9Vδ2 T cells as investigational tools to improve the potency of ICP blockade and immune interventions in MM.
Collapse
Affiliation(s)
- Barbara Castella
- Laboratorio di Immunologia dei Tumori del Sangue, Centro Interdipartimentale di Ricerca in Biologia Molecolare, Università degli Studi di Torino, Turin, Italy
| | - Assunta Melaccio
- Dipartimento di Scienze Biomediche ed Oncologia Umana, Sezione di Medicina Interna ed Oncologia, Università degli studi di Bari "A. Moro", Bari, Italy
| | - Myriam Foglietta
- Laboratorio di Immunologia dei Tumori del Sangue, Centro Interdipartimentale di Ricerca in Biologia Molecolare, Università degli Studi di Torino, Turin, Italy.,SC Ematologia, AO S.Croce e Carle, Cuneo, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università degli Studi di Torino, Turin, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue, Centro Interdipartimentale di Ricerca in Biologia Molecolare, Università degli Studi di Torino, Turin, Italy.,SC Ematologia, AO S.Croce e Carle, Cuneo, Italy
| |
Collapse
|
20
|
van Nieuwenhuijzen N, Spaan I, Raymakers R, Peperzak V. From MGUS to Multiple Myeloma, a Paradigm for Clonal Evolution of Premalignant Cells. Cancer Res 2018; 78:2449-2456. [PMID: 29703720 DOI: 10.1158/0008-5472.can-17-3115] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/16/2018] [Accepted: 03/16/2018] [Indexed: 11/16/2022]
Abstract
Multiple myeloma (MM) is a treatable, but incurable, malignancy of plasma cells (PC) in the bone marrow (BM). It represents the final stage in a continuum of PC dyscrasias and is consistently preceded by a premalignant phase termed monoclonal gammopathy of undetermined significance (MGUS). The existence of this well-defined premalignant phase provides the opportunity to study clonal evolution of a premalignant condition into overt cancer. Unraveling the mechanisms of malignant transformation of PC could enable early identification of MGUS patients at high risk of progression and may point to novel therapeutic targets, thereby possibly delaying or preventing malignant transformation. The MGUS-to-MM progression requires multiple genomic events and the establishment of a permissive BM microenvironment, although it is generally not clear if the various microenvironmental events are causes or consequences of disease progression. Advances in gene-sequencing techniques and the use of serial paired analyses have allowed for a more specific identification of driver lesions. The challenge in cancer biology is to identify and target those lesions that confer selective advantage and thereby drive evolution of a premalignant clone. Here, we review recent advances in the understanding of malignant transformation of MGUS to MM. Cancer Res; 78(10); 2449-56. ©2018 AACR.
Collapse
Affiliation(s)
- Niels van Nieuwenhuijzen
- Laboratory of Translational Immunology, University Medical Center, Utrecht, the Netherlands.,Department of Hematology, University Medical Center, Utrecht, the Netherlands
| | - Ingrid Spaan
- Laboratory of Translational Immunology, University Medical Center, Utrecht, the Netherlands
| | - Reinier Raymakers
- Department of Hematology, University Medical Center, Utrecht, the Netherlands
| | - Victor Peperzak
- Laboratory of Translational Immunology, University Medical Center, Utrecht, the Netherlands.
| |
Collapse
|
21
|
Nakamura K, Kassem S, Cleynen A, Chrétien ML, Guillerey C, Putz EM, Bald T, Förster I, Vuckovic S, Hill GR, Masters SL, Chesi M, Bergsagel PL, Avet-Loiseau H, Martinet L, Smyth MJ. Dysregulated IL-18 Is a Key Driver of Immunosuppression and a Possible Therapeutic Target in the Multiple Myeloma Microenvironment. Cancer Cell 2018; 33:634-648.e5. [PMID: 29551594 DOI: 10.1016/j.ccell.2018.02.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/27/2017] [Accepted: 02/13/2018] [Indexed: 02/06/2023]
Abstract
Tumor-promoting inflammation and avoiding immune destruction are hallmarks of cancer. Here, we demonstrate that the pro-inflammatory cytokine interleukin (IL)-18 is critically involved in these hallmarks in multiple myeloma (MM). Mice deficient for IL-18 were remarkably protected from Vk∗MYC MM progression in a CD8+ T cell-dependent manner. The MM-niche-derived IL-18 drove generation of myeloid-derived suppressor cells (MDSCs), leading to accelerated disease progression. A global transcriptome analysis of the immune microenvironment in 73 MM patients strongly supported the negative impact of IL-18-driven MDSCs on T cell responses. Strikingly, high levels of bone marrow plasma IL-18 were associated with poor overall survival in MM patients. Furthermore, our preclinical studies suggested that IL-18 could be a potential therapeutic target in MM.
Collapse
Affiliation(s)
- Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4006 QLD, Australia
| | - Sahar Kassem
- Cancer Research Center of Toulouse, INSERM UMR 1037, 2 av Hubert Curien, 31037 Toulouse, France
| | - Alice Cleynen
- Institut Montpelliérain Alexander Grothendieck, CNRS, University Montpellier, 34095 Montpellier, France
| | - Marie-Lorraine Chrétien
- Cancer Research Center of Toulouse, INSERM UMR 1037, 2 av Hubert Curien, 31037 Toulouse, France; University Hospital, Dijon, 21000 Dijon, France
| | - Camille Guillerey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4006 QLD, Australia; School of Medicine, University of Queensland, St Lucia 4006 QLD, Australia
| | - Eva Maria Putz
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4006 QLD, Australia
| | - Tobias Bald
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4006 QLD, Australia
| | - Irmgard Förster
- Immunology and Environment, LIMES Institute, University of Bonn, 53115 Bonn, Germany
| | - Slavica Vuckovic
- School of Medicine, University of Queensland, St Lucia 4006 QLD, Australia; Department of Bone Marrow Transplantation, QIMR Berghofer Medical Research Institute, Herston, 4006 QLD, Australia
| | - Geoffrey R Hill
- Department of Bone Marrow Transplantation, QIMR Berghofer Medical Research Institute, Herston, 4006 QLD, Australia
| | - Seth L Masters
- Division of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Marta Chesi
- Mayo Clinic in Arizona, Phoenix, AZ 85054, USA
| | | | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM UMR 1037, 2 av Hubert Curien, 31037 Toulouse, France; Institut Universitaire du Cancer, Toulouse 31100, France
| | - Ludovic Martinet
- Cancer Research Center of Toulouse, INSERM UMR 1037, 2 av Hubert Curien, 31037 Toulouse, France.
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, 4006 QLD, Australia; School of Medicine, University of Queensland, St Lucia 4006 QLD, Australia.
| |
Collapse
|
22
|
Di Nicola M, Apetoh L, Bellone M, Colombo MP, Dotti G, Ferrone S, Muscolini M, Hiscott J, Anichini A, Pupa SM, Braud FD, Del Vecchio M. Innovative Therapy, Monoclonal Antibodies and Beyond. Cytokine Growth Factor Rev 2017; 38:1-9. [PMID: 29029813 DOI: 10.1016/j.cytogfr.2017.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
Abstract
The seventh Edition of "Innovative Therapy, Monoclonal Antibodies and Beyond" Meeting took place in Milan, Italy, on January 27, 2017. The two sessions of the meeting were focused on: 1) Preclinical assays and novel biotargets; and 2) monoclonal antibodies, cell therapies and targeted molecules. Between these two sessions, a lecture entitled "HLA-antigens modulation and response to immune checkpoint inhibitor immunotherapy" was also presented. Despite the impressive successes in cancer immunotherapy in recent years, the response to immune based interventions occurs only in a minority of patients (∼20%). Several basic and translational mechanisms of resistance to immune checkpoint blockers (ICBs) were discussed during the meeting: 1. the impact of tumor microenvironment on the activity of immune system; 2. strategies to inhibit the cross-talk between extracellular matrix and myeloid-derived suppressor cells (MDSC) in the preclinical setting; 3. microRNA expression as a biomarker and as a target of therapy in non-small cell lung cancer (NSCLC); 4. the significance of complement activation pathways in response to immune checkpoint inhibitors; 5. the immunosuppressive activity of the microbiota by inducing IL-17 producing cells; and 6. modulation of HLA antigens as possible markers of response to ICB therapy. In order to overcome the deficiency in active anti-tumor T cells, several clinically applicable combination strategies were also discussed: 1. strategies to enhance the anticancer effects of immunogenic cell death inducing-chemotherapy; 2. the use of CAR T-cells in solid tumors; 3. the use of combination strategies involving oncolytic viruses and ICBs; 4. combinations of new ICBs with anti-PD-1/CTLA-4 therapy; and 4. combinations of targeted therapies and ICBs in melanoma. Overall, this conference emphasized the many novel strategies that are being investigated to improve the overall patient response to cancer immunotherapy. Optimization of biomarkers to accurately select patients who will respond to immunotherapy, coupled with combination strategies to improve long term patient survival remain critical challenges in the immuno-oncology field.
Collapse
Affiliation(s)
- M Di Nicola
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Milan, Italy; Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - L Apetoh
- INSERM, U1231, Dijon, France; 4Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - M Bellone
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - M P Colombo
- Centre Georges François Leclerc, Dijon, France
| | - G Dotti
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - S Ferrone
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - M Muscolini
- Laboratorio Pasteur, Istituto Pasteur-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - J Hiscott
- Laboratorio Pasteur, Istituto Pasteur-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - A Anichini
- Human Tumor Immunobiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S M Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F de Braud
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - M Del Vecchio
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| |
Collapse
|
23
|
Endothelial progenitor cells in multiple myeloma neovascularization: a brick to the wall. Angiogenesis 2017; 20:443-462. [PMID: 28840415 DOI: 10.1007/s10456-017-9571-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 07/31/2017] [Indexed: 12/12/2022]
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion of plasma cells in the bone marrow that leads to events such as bone destruction, anaemia and renal failure. Despite the several therapeutic options available, there is still no effective cure, and the standard survival is up to 4 years. The evolution from the asymptomatic stage of monoclonal gammopathy of undetermined significance to MM and the progression of the disease itself are related to cellular and molecular alterations in the bone marrow microenvironment, including the development of the vasculature. Post-natal vasculogenesis is characterized by the recruitment to the tumour vasculature of bone marrow progenitors, known as endothelial progenitor cells (EPCs), which incorporate newly forming blood vessels and differentiate into endothelial cells. Several processes related to EPCs, such as recruitment, mobilization, adhesion and differentiation, are tightly controlled by cells and molecules in the bone marrow microenvironment. In this review, the bone marrow microenvironment and the mechanisms associated to the development of the neovasculature promoted by EPCs are discussed in detail in both a non-pathological scenario and in MM. The latest developments in therapy targeting the vasculature and EPCs in MM are also highlighted. The identification and characterization of the pathways relevant to the complex setting of MM are of utter importance to identify not only biomarkers for an early diagnosis and disease progression monitoring, but also to reveal intervention targets for more effective therapy directed to cancer cells and the endothelial mediators relevant to neovasculature development.
Collapse
|
24
|
De Beule N, De Veirman K, Maes K, De Bruyne E, Menu E, Breckpot K, De Raeve H, Van Rampelbergh R, Van Ginderachter JA, Schots R, Van Valckenborgh E, Vanderkerken K. Tumour-associated macrophage-mediated survival of myeloma cells through STAT3 activation. J Pathol 2017; 241:534-546. [PMID: 27976373 DOI: 10.1002/path.4860] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/04/2016] [Accepted: 11/24/2016] [Indexed: 12/26/2022]
Abstract
Overcoming drug resistance is one of the greatest challenges in the treatment of multiple myeloma (MM). The interaction of myeloma cells with the bone marrow (BM) microenvironment is a major factor contributing to drug resistance. Tumour-associated macrophages (TAMs) with different polarization states constitute an important component of this microenvironment. Previous studies have revealed a role of TAMs in MM cell survival and drug resistance; however, the impact of macrophage polarization (anti-tumoural 'M1' versus pro-tumoural 'M2'-like phenotype) in this process has not yet been described. Here, the presence of TAMs was confirmed in BM sections from MM patients, both at diagnosis and relapse, with two M2 markers, CD163 and CD206. By following different TAM subpopulations during disease progression in the syngeneic murine 5T33MM model, we demonstrated a decrease in the number of inflammatory monocytes and an increase in the number of M2-oriented TAMs in BM. Co-culture experiments demonstrated that macrophages provide a survival benefit to myeloma cells that is maintained after treatment with several classes of anti-myeloma agent (melphalan and bortezomib); the greatest effect was observed with M2-polarized macrophages. The pro-survival effect was associated with activation of the STAT3 pathway in 5T33MM cells, less cleavage of caspase-3, and thus less apoptosis. AZD1480, an ATP-competitive JAK2 inhibitor, abrogated the observed TAM-mediated MM cell survival, and partially inhibited resistance to bortezomib. Despite having only a small quantitative impact on myeloid cells in vivo, AZD1480 treatment alone and in combination with bortezomib significantly reduced tumour load. In conclusion, M2 TAMs are present in the MM microenvironment, and contribute to MM cell survival and protection from drug-induced apoptosis. As a result of TAM-induced activation of the STAT3 pathway, 5T33MM cells are sensitized to AZD1480 treatment. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nathan De Beule
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Technology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hendrik De Raeve
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, VIB Inflammation Research Centre, Ghent, Belgium
| | - Rik Schots
- Department of Clinical Haematology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Laboratory of Haematology and Immunology, Myeloma Centre Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
25
|
A gene expression inflammatory signature specifically predicts multiple myeloma evolution and patients survival. Blood Cancer J 2016; 6:e511. [PMID: 27983725 PMCID: PMC5223153 DOI: 10.1038/bcj.2016.118] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/28/2016] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma (MM) is closely dependent on cross-talk between malignant plasma cells and cellular components of the inflammatory/immunosuppressive bone marrow milieu, which promotes disease progression, drug resistance, neo-angiogenesis, bone destruction and immune-impairment. We investigated the relevance of inflammatory genes in predicting disease evolution and patient survival. A bioinformatics study by Ingenuity Pathway Analysis on gene expression profiling dataset of monoclonal gammopathy of undetermined significance, smoldering and symptomatic-MM, identified inflammatory and cytokine/chemokine pathways as the most progressively affected during disease evolution. We then selected 20 candidate genes involved in B-cell inflammation and we investigated their role in predicting clinical outcome, through univariate and multivariate analyses (log-rank test, logistic regression and Cox-regression model). We defined an 8-genes signature (IL8, IL10, IL17A, CCL3, CCL5, VEGFA, EBI3 and NOS2) identifying each condition (MGUS/smoldering/symptomatic-MM) with 84% accuracy. Moreover, six genes (IFNG, IL2, LTA, CCL2, VEGFA, CCL3) were found independently correlated with patients' survival. Patients whose MM cells expressed high levels of Th1 cytokines (IFNG/LTA/IL2/CCL2) and low levels of CCL3 and VEGFA, experienced the longest survival. On these six genes, we built a prognostic risk score that was validated in three additional independent datasets. In this study, we provide proof-of-concept that inflammation has a critical role in MM patient progression and survival. The inflammatory-gene prognostic signature validated in different datasets clearly indicates novel opportunities for personalized anti-MM treatment.
Collapse
|
26
|
Cooke RE, Gherardin NA, Harrison SJ, Quach H, Godfrey DI, Prince M, Koldej R, Ritchie DS. Spontaneous onset and transplant models of the Vk*MYC mouse show immunological sequelae comparable to human multiple myeloma. J Transl Med 2016; 14:259. [PMID: 27599546 PMCID: PMC5011922 DOI: 10.1186/s12967-016-0994-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
Background The Vk*MYC transgenic and transplant mouse models of multiple myeloma (MM) are well established as a research tool for anti-myeloma drug discovery. However, little is known of the immune response in these models. Understanding the immunological relevance of these models is of increasing importance as immunotherapeutic drugs are developed against MM. Methods We set out to examine how cellular immunity is affected in Vk*MYC mouse models and compare that to the immunology of patients with newly diagnosed and relapsed/refractory MM. Results We found that there were significant immunological responses in mice developing either spontaneous (transgenic) or transplanted MM as a consequence of the degree of tumor burden. Particularly striking were the profound B cell lymphopenia and the expansion of CD8+ effector memory T cells within the lymphocyte population that progressively developed with advancing disease burden, mirroring changes seen in human MM. High disease burden was also associated with increased inflammatory cytokine production by T lymphocytes, which is more fitting with relapsed/refractory MM in humans. Conclusions These findings have important implications for the application of this mouse model in the development of MM immunotherapies. Trial registration LitVacc ANZCTR trial ID ACTRN12613000344796; RevLite ANZCTR trial ID NCT00482261
Collapse
Affiliation(s)
- Rachel E Cooke
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, 3010, Australia. .,Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia. .,Department of Medicine, University of Melbourne, Melbourne, Australia.
| | - Nicholas A Gherardin
- Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, 3010, Australia
| | - Simon J Harrison
- Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Hang Quach
- Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Miles Prince
- Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Rachel Koldej
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, 3010, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - David S Ritchie
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, 3010, Australia.,Cancer Immunology Program, Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, VIC, 3002, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| |
Collapse
|
27
|
Targeting vasculogenesis to prevent progression in multiple myeloma. Leukemia 2016; 30:1103-15. [DOI: 10.1038/leu.2016.3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/09/2015] [Accepted: 12/16/2015] [Indexed: 12/17/2022]
|
28
|
Xiao J, Mao Y, Zhou Y, Xiang K. Clinicopathological significance of NGX6 expression in breast cancer and its relationship to angiogenesis. Int J Clin Exp Med 2015; 8:22198-22203. [PMID: 26885195 PMCID: PMC4729981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 12/05/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE The aim was to explore clinicopathological significance of Nasopharyngeal carcinoma-associated gene 6 (NGX6) expression in breast cancer and its relationship to angiogenesis. METHODS Clinicopathological feature of 168 patients with breast cancer were analyzed. NGX6 expression and microvessel density (MVD) in tumor tissue were measured using immunohistochemistry methods. The association of NGX6 expression with MVD and clinicopathological features was assessed. RESULTS Among 168 cases of breast cancer, NGX6 positive expression were found in 92 (54.8%) cases and NGX6 negative expression were found in 76 (45.2%) cases. Incidence rate of large size tumor, high tumor node metastasis (TNM) stage and lymph node metastasis in NGX6 negative expression group were higher than NGX6 positive expression group in breast cancer (P=0.003, 0.007, and 0.003, respectively). MVD in NGX6 negative expression group is 22.5±4.8, MVD in NGX6 positive expression group is 15.2±4.2. MVD in the NGX6 negative expression group was significantly higher than the NGX6 positive expression group (P<0.05). CONCLUSION The expression of NGX6 was closely associated with tumor size, TNM stage, lymph node metastasis and MVD. NGX6 is involved in metastasis and angiogenesis activity of breast cancer. The study may provide a theoretical basis for anti-angiogenic therapy of breast cancer.
Collapse
Affiliation(s)
- Jidong Xiao
- Department of Diagnostic Ultrasound, Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan Province, China
| | - Yuyao Mao
- Department of Diagnostic Ultrasound, Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan Province, China
| | - Yuanquan Zhou
- Department of Diagnostic Ultrasound, Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan Province, China
| | - Kaimin Xiang
- Department of General Surgery, Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan Province, China
| |
Collapse
|