1
|
Koumpis E, Papoudou-Bai A, Papathanasiou K, Kolettas E, Kanavaros P, Hatzimichael E. Unraveling the Immune Microenvironment in Diffuse Large B-Cell Lymphoma: Prognostic and Potential Therapeutic Implications. Curr Issues Mol Biol 2024; 46:7048-7064. [PMID: 39057061 PMCID: PMC11276293 DOI: 10.3390/cimb46070420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is a multifaceted condition characterized by significant diversity in its molecular and pathological subtypes and clinical manifestation. Despite the progress made in the treatment of DLBCL through the development of novel drugs, an estimated one-third of patients encounter relapse or acquire refractory disease. The tumor microenvironment (TME) of DLBCL, a complex network consisting of cellular and noncellular components that engage in interactions with the tumor, is a parameter that is gaining increasing attention. The TME comprises both the immune and nonimmune microenvironments. The immune microenvironment comprises natural killer (NK) cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), neutrophils, myeloid-derived suppressor cells (MDSCs), and T and B lymphocytes. The nonimmune microenvironment consists of the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), mesenchymal stromal cells, and other molecules that are secreted. Despite ongoing research, the exact impact of these components and their interaction on the progression of the disease remains elusive. A comprehensive review of significant discoveries concerning the cellular and noncellular constituents, molecular characteristics, and treatment response and prognosis of the TME in DLBCL, as well as the potential targeting of the TME with novel therapeutic approaches, is provided in this article.
Collapse
Affiliation(s)
- Epameinondas Koumpis
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| | - Alexandra Papoudou-Bai
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece;
| | - Konstantina Papathanasiou
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| | - Evangelos Kolettas
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 110 Ioannina, Greece;
- Biomedical Research Institute, Foundation for Research and Technology, 45 110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 110 Ioannina, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| |
Collapse
|
2
|
Gu A, Li J, Qiu S, Hao S, Yue ZY, Zhai S, Li MY, Liu Y. Pancreatic cancer environment: from patient-derived models to single-cell omics. Mol Omics 2024; 20:220-233. [PMID: 38414408 DOI: 10.1039/d3mo00250k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Pancreatic cancer (PC) is a highly malignant cancer characterized by poor prognosis, high heterogeneity, and intricate heterocellular systems. Selecting an appropriate experimental model for studying its progression and treatment is crucial. Patient-derived models provide a more accurate representation of tumor heterogeneity and complexity compared to cell line-derived models. This review initially presents relevant patient-derived models, including patient-derived xenografts (PDXs), patient-derived organoids (PDOs), and patient-derived explants (PDEs), which are essential for studying cell communication and pancreatic cancer progression. We have emphasized the utilization of these models in comprehending intricate intercellular communication, drug responsiveness, mechanisms underlying tumor growth, expediting drug discovery, and enabling personalized medical approaches. Additionally, we have comprehensively summarized single-cell analyses of these models to enhance comprehension of intercellular communication among tumor cells, drug response mechanisms, and individual patient sensitivities.
Collapse
Affiliation(s)
- Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Shimei Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - Shenglin Hao
- Department of Functional Neurosurgery, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| |
Collapse
|
3
|
Bao E, Zhou Y, He S, Tang J, He Y, Zhu M, Cheng C, Wang Y. RING box protein-1(RBX1), a key component of SCF E3 ligase, induced multiple myeloma cell drug-resistance though suppressing p27. Cancer Biol Ther 2023; 24:2231670. [PMID: 37639640 PMCID: PMC10464534 DOI: 10.1080/15384047.2023.2231670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 02/18/2023] [Accepted: 04/27/2023] [Indexed: 08/31/2023] Open
Abstract
Multiple myeloma (MM) is a clonal disease of plasma cells that remains, for the most part, incurable despite the advent of several novel therapeutics. The elevated expression of p27 and its association with cell-cycle arrest is speculated to be one of the major mechanisms by which MM cells escape the cytotoxic effects of therapeutic agents. In this study, we demonstrated that RBX1 silencing could inhibit MM cell growth and promote cell drug resistance. RBX1 directly interacted with and triggered the ubiquitination and degradation of p27, ultimately causing p27 reduction. Additionally, cell growth and apoptosis analysis indicated that the role of RBX1 in regulating myeloma cell proliferation and drug resistance resulted from p27 accumulation, which occurred in a Thr187 phosphorylation-dependent manner. Furthermore, the cell-cycle analysis demonstrated that RBX1 overexpression induced cells to enter the cell cycle (S-phase) and partially inhibited chemotherapeutic drugs-mediated cell cycle arrest. Notably, the forced expression of RBX1 also inhibited the cell adhesion-mediated elevation of p27 and induced the accumulation of adherent cells in apoptosis, especially the proteolytic cleavage of caspase-3. Additionally, RBX1 knockdown significantly inhibited myeloma development in SCID-Hu mice and in a human MM xenotransplant model. Overall, these in vitro and in vivo experiments indicated that the RBX1-p27 axis could be a central molecular mechanism by which RBX1 functions as a tumor promoter and stimulates cell growth in chemotherapeutic drugs treated MM cells.
Collapse
Affiliation(s)
- Enfang Bao
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu Province, People’s Republic of China
| | - Yu Zhou
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu Province, People’s Republic of China
| | - Song He
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, People’s Republic of China
| | - Jie Tang
- Department of Pathology, Liyang People’s Hospital, Liyang, Jiangsu, China
| | - Yunhua He
- Department of Oncology, Nantong Tongzhou People’s Hospital, Nantong, Jiangsu Province, People’s Republic of China
| | - Mengyuan Zhu
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu Province, People’s Republic of China
| | - Chun Cheng
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu Province, People’s Republic of China
| | - Yuchan Wang
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu Province, People’s Republic of China
| |
Collapse
|
4
|
Li G, Feng M, Zhang Z, Liu J, Zhang H. BACH1 Loss Exerts Antitumor Effects on Mantle Cell Lymphoma Cells via Inducing a Tumor-Intrinsic Innate Immune Response and Cell-Cycle Arrest. Mol Cancer Res 2023; 21:1274-1287. [PMID: 37713314 DOI: 10.1158/1541-7786.mcr-23-0424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/31/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023]
Abstract
BTB and CNC homology 1 (BACH1) is a transcription repressor that regulates multiple physiological processes, including intracellular heme homeostasis and immune responses. Increasing lines of evidence indicate that BACH1 reshapes metastasis and metabolism of human solid tumors. However, its potential roles in mantle cell lymphoma (MCL) remain largely unknown. Here, we found that silencing BACH1 in MCL cells induced markedly cell-cycle arrest and cell apoptosis, whereas overexpression of BACH1 exhibited the opposite patterns. Increased BACH1 levels not only promoted tumor growth and dispersal in xenografts, but also conferred a long-term poor prognosis in patients with MCL. Interestingly, RNA sequencing analysis revealed noncanonical function of BACH1 in regulation of type I interferon (IFNI) response, DNA replication and repair, and cell cycle. Mechanistically, zinc finger and BTB domain containing 20 (ZBTB20) and HMG-box transcription factor 1 (HBP1) were for the first time identified as two novel downstream targets repressed by BACH1 in MCL cells. Further double-knockdown functional assays confirmed that loss of BACH1 induced ZBTB20-mediated IFNα production and HBP1-mediated cell-cycle arrest, indicating that BACH1-centered regulatory network may be a novel targetable vulnerability in MCL cells. IMPLICATIONS BACH1 serves as a pleotropic regulator of tumor-intrinsic innate immune response and cell-cycle progression, disruption of which may offer a promising therapeutic strategy for MCL treatment.
Collapse
Affiliation(s)
- Guilan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Min Feng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Ziting Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Jiangyuan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China
| |
Collapse
|
5
|
Burger KL, Fernandez MR, Meads MB, Sudalagunta P, Oliveira PS, Renatino Canevarolo R, Alugubelli RR, Tungsevik A, De Avila G, Silva M, Graeter AI, Dai HA, Vincelette ND, Prabhu A, Magaletti D, Yang C, Li W, Kulkarni A, Hampton O, Koomen JM, Roush WR, Monastyrskyi A, Berglund AE, Silva AS, Cleveland JL, Shain KH. CK1δ and CK1ε Signaling Sustains Mitochondrial Metabolism and Cell Survival in Multiple Myeloma. Cancer Res 2023; 83:3901-3919. [PMID: 37702657 PMCID: PMC10690099 DOI: 10.1158/0008-5472.can-22-2350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 06/09/2023] [Accepted: 09/08/2023] [Indexed: 09/14/2023]
Abstract
Multiple myeloma remains an incurable malignancy due to acquisition of intrinsic programs that drive therapy resistance. Here we report that casein kinase-1δ (CK1δ) and CK1ε are therapeutic targets in multiple myeloma that are necessary to sustain mitochondrial metabolism. Specifically, the dual CK1δ/CK1ε inhibitor SR-3029 had potent in vivo and ex vivo anti-multiple myeloma activity, including against primary multiple myeloma patient specimens. RNA sequencing (RNA-seq) and metabolic analyses revealed inhibiting CK1δ/CK1ε disables multiple myeloma metabolism by suppressing genes involved in oxidative phosphorylation (OxPhos), reducing citric acid cycle intermediates, and suppressing complexes I and IV of the electron transport chain. Finally, sensitivity of multiple myeloma patient specimens to SR-3029 correlated with elevated expression of mitochondrial genes, and RNA-seq from 687 multiple myeloma patient samples revealed that increased CSNK1D, CSNK1E, and OxPhos genes correlate with disease progression and inferior outcomes. Thus, increases in mitochondrial metabolism are a hallmark of multiple myeloma progression that can be disabled by targeting CK1δ/CK1ε. SIGNIFICANCE CK1δ and CK1ε are attractive therapeutic targets in multiple myeloma whose expression increases with disease progression and connote poor outcomes, and that are necessary to sustain expression of genes directing OxPhos.
Collapse
Affiliation(s)
- Karen L. Burger
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mario R. Fernandez
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark B. Meads
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Praneeth Sudalagunta
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Paula S. Oliveira
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Rafael Renatino Canevarolo
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | - Alexandre Tungsevik
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Gabe De Avila
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Maria Silva
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Allison I. Graeter
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | - Nicole D. Vincelette
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Antony Prabhu
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Dario Magaletti
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Chunying Yang
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Weimin Li
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | | | - John M. Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | - Andrii Monastyrskyi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Ariosto S. Silva
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John L. Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kenneth H. Shain
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
6
|
Han B, Yim J, Lim S, Na S, Lee C, Kim TM, Paik JH, Kim S, Jeon YK. Prognostic Impact of the Immunoscore Based on Whole-Slide Image Analysis of CD3+ Tumor-Infiltrating Lymphocytes in Diffuse Large B-Cell Lymphoma. Mod Pathol 2023; 36:100224. [PMID: 37257823 DOI: 10.1016/j.modpat.2023.100224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/28/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
An Immunoscore based on tumor-infiltrating T-cell density was validated as a prognostic factor in patients with solid tumors. However, the potential utility of the Immunoscore in predicting the prognosis of patients with diffuse large B-cell lymphoma (DLBCL) is unclear. Here, the prognostic value of an Immunoscore based on tumor-infiltrating CD3+ T-cell density was evaluated in 104 patients with DLBCL who underwent R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone) therapy. Digitally scanned whole-slide images were analyzed using Aperio ImageScope software. CD3+ cell densities in the whole tumor area were quantitated using 3 different methods, including number of CD3+ cells/area (mm2), ratio of CD3+ cells to total cells, and ratio of CD3+ cells to CD20+ cells. There was a high concordance among the 3 methods. Patients with low CD3+ cell density had an elevated serum lactate dehydrogenase level and a high Ki-67 proliferation index (all, P < .05). Patients with low CD3+ cell density, according to all 3 methods, had worse overall survival (OS) and worse progression-free survival (P < .05, all). They also had poor OS, independent of MYC/BCL2 double expression (DE) status, Eastern Cooperative Oncology Group performance status, or Ann Arbor stage (all, P < .05). These results were validated using 2 publicly available data sets. In both validation cohorts, patients with low CD3E mRNA expression had an elevated serum lactate dehydrogenase level, extranodal site involvement, and DE status (P < .05). They also had worse progression-free survival (P = .067 and P = .002, respectively) and OS (both P < .05). A low CD3E mRNA level was predictive of poor OS, independent of DE status. An Immunoscore based on whole-slide image analysis of CD3+ T-cell infiltration was sufficient to predict survival in patients with DLBCL. Low CD3+ cell density was a poor prognostic factor, independent of other prognostic parameters and DE status.
Collapse
Affiliation(s)
- Bogyeong Han
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeemin Yim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sojung Lim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sei Na
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Ho Paik
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Sehui Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Korea University Guro Hospital, Seoul, Republic of Korea.
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Henderson F, Brem S, Hussain J, Buch L, Maloney E, Singhal S, Lee JYK. Second window indocyanine green localizes CNS lymphoma in real time in the operating room: report of two cases. Br J Neurosurg 2023; 37:619-623. [PMID: 32009484 PMCID: PMC10997215 DOI: 10.1080/02688697.2020.1716945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
Abstract
Intraoperative distinction of lesional tissue versus normal brain parenchyma can be difficult in neurosurgical oncology procedures. We report the successful, real-time visualization of central nervous system (CNS) lymphoma using the 'Second Window Indocyanine Green' (SWIG) method for two patients who underwent craniotomy for pathology that was determined to be large B cell lymphoma. Indocyanine green (ICG), when administered intravenously the day prior to cranial surgery, is a re-purposed fluorophore that may afford safe, immediate visual confirmation of on-target tissue resection, thereby providing a valuable adjunct to intraoperative navigation and decreasing reliance on frozen pathology analysis. These first reported cases of SWIG for lymphoma in the CNS indicate that further study of fluorophores to improve biopsy targeting and yield is warranted.
Collapse
Affiliation(s)
- Fraser Henderson
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Steven Brem
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jasmin Hussain
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Love Buch
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Eileen Maloney
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Till KJ, Abdullah M, Alnassfan T, Janet GZ, Marks T, Coma S, Weaver DT, Pachter JA, Pettitt AR, Slupsky JR. Roles of PI3Kγ and PI3Kδ in mantle cell lymphoma proliferation and migration contributing to efficacy of the PI3Kγ/δ inhibitor duvelisib. Sci Rep 2023; 13:3793. [PMID: 36882482 PMCID: PMC9992372 DOI: 10.1038/s41598-023-30148-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma that is incurable with existing therapies, and therefore presents a significant unmet clinical need. The ability of this disease to overcome therapy, including those that target the B cell receptor pathway which has a pathogenic role in MCL, highlights the need to develop new treatment strategies. Herein, we demonstrate that a distinguishing feature of lymph node resident MCL cells is the expression of phosphatidylinositol 3-kinase γ (PI3Kγ), a PI3K isoform that is not highly expressed in other B cells or B-cell malignancies. By exploring the role of PI3K in MCL using different PI3K isoform inhibitors, we provide evidence that duvelisib, a dual PI3Kδ/γ inhibitor, has a greater effect than PI3Kδ- and PI3Kγ-selective inhibitors in blocking the proliferation of primary MCL cells and MCL cell lines, and in inhibiting tumour growth in a mouse xenograft model. In addition, we demonstrated that PI3Kδ/γ signalling is critical for migration of primary MCL cells and cell lines. Our data indicates that aberrant expression of PI3Kγ is a critical feature of MCL pathogenesis. Thus, we suggest that the dual PI3Kδ/γ duvelisib would be effective for the treatment of mantle cell lymphoma.
Collapse
Affiliation(s)
- Kathleen J Till
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.
| | - Mariah Abdullah
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Tahera Alnassfan
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Gallardo Zapata Janet
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Thomas Marks
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Silvia Coma
- Verastem Oncology, 117 Kendrick St #500, Needham, MA, 02494, USA
| | - David T Weaver
- Verastem Oncology, 117 Kendrick St #500, Needham, MA, 02494, USA
| | | | - Andrew R Pettitt
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| |
Collapse
|
9
|
Cariello M, Squilla A, Piacente M, Venutolo G, Fasano A. Drug Resistance: The Role of Exosomal miRNA in the Microenvironment of Hematopoietic Tumors. Molecules 2022; 28:molecules28010116. [PMID: 36615316 PMCID: PMC9821808 DOI: 10.3390/molecules28010116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, have an important role thanks to their ability to communicate and exchange information between tumor cells and the tumor microenvironment (TME), and have also been associated with communicating anti-cancer drug resistance (DR). The increase in proliferation of cancer cells alters oxygen levels, which causes hypoxia and results in a release of exosomes by the cancer cells. In this review, the results of studies examining the role of exosomal miRNA in DR, and their mechanism, are discussed in detail in hematological tumors: leukemia, lymphoma, and multiple myeloma. In conclusion, we underline the exosome's function as a possible drug delivery vehicle by understanding its cargo. Engineered exosomes can be used to be more specific for personalized therapy.
Collapse
Affiliation(s)
- Mariaconcetta Cariello
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 84125 Salerno, Italy
| | - Angela Squilla
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 84125 Salerno, Italy
| | - Martina Piacente
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 84125 Salerno, Italy
| | - Giorgia Venutolo
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 84125 Salerno, Italy
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 84125 Salerno, Italy
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA 02114, USA
- Correspondence: ; Tel.: +1-617-724-4604
| |
Collapse
|
10
|
Jakša R, Karolová J, Svatoň M, Kazantsev D, Grajciarová M, Pokorná E, Tonar Z, Klánová M, Winkowska L, Maláriková D, Vočková P, Forsterová K, Renešová N, Dolníková A, Nožičková K, Dundr P, Froňková E, Trněný M, Klener P. Complex genetic and histopathological study of 15 patient-derived xenografts of aggressive lymphomas. J Transl Med 2022; 102:957-965. [PMID: 36775424 PMCID: PMC9420679 DOI: 10.1038/s41374-022-00784-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 11/14/2022] Open
Abstract
Non-Hodgkin lymphomas (NHL) represent the most common hematologic malignancies. Patient-derived xenografts (PDXs) are used for various aspects of translational research including preclinical in vivo validation of experimental treatment approaches. While it was repeatedly demonstrated that PDXs keep majority of somatic mutations with the primary lymphoma samples, from which they were derived, the composition of PDX tumor microenvironment (TME) has not been extensively studied. We carried out a comparative genetic and histopathological study of 15 PDX models derived from patients with various types of NHL including diffuse large B-cell lymphoma (DLBCL; n = 7), Burkitt lymphoma (BL; n = 1), mantle cell lymphoma (MCL; n = 2), and peripheral T-cell lymphomas (PTCL; n = 5). Whole exome sequencing (WES) of the PDXs and primary lymphoma cells was implemented in 13 out of 15 cases with available DNA samples. Standard immunohistochemistry (IHC) was used to analyze the composition of PDX TME. WES data confirmed that PDXs maintained the genetic heterogeneity with the original primary lymphoma cells. In contrast, IHC analysis revealed the following recurrently observed alterations in the composition of PDX tumors: more blastoid lymphoma cell morphology, increased proliferation rate, lack of non-malignant cellular components including T cells and (human or murine) macrophages, and significantly lower intratumoral microvessel density and microvessel area composed of murine vessels. In addition, PDX tumors derived from T-NHL displayed additional differences compared to the primary lymphoma samples including markedly lower desmoplasia (i.e., the extent of both reticular and collagen fibrosis), loss of expression of cytotoxic granules (i.e., perforin, TIA, granzyme B), or loss of expression of T-cell specific antigens (i.e., CD3, CD4, CD8). Our data suggest that despite keeping the same genetic profiles, PDX models of aggressive NHL do not recapitulate the microenvironmental heterogeneity of the original lymphomas. These findings have implications on the relevance of PDX models in the context of preclinical research.
Collapse
Affiliation(s)
- Radek Jakša
- Institute of Pathology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jana Karolová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michael Svatoň
- CLIP- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Dmitry Kazantsev
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Grajciarová
- Department of Histology and Embryology and Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Eva Pokorná
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zbyněk Tonar
- Department of Histology and Embryology and Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Magdalena Klánová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Diana Maláriková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petra Vočková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristina Forsterová
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicol Renešová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alexandra Dolníková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristýna Nožičková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Froňková
- CLIP- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Marek Trněný
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
11
|
Smith AL, Eiken AP, Skupa SA, Moore DY, Umeta LT, Smith LM, Lyden ER, D’Angelo CR, Kallam A, Vose JM, Kutateladze TG, El-Gamal D. A Novel Triple-Action Inhibitor Targeting B-Cell Receptor Signaling and BRD4 Demonstrates Preclinical Activity in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:6712. [PMID: 35743155 PMCID: PMC9224275 DOI: 10.3390/ijms23126712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL) results from intrinsic genetic defects and complex microenvironment stimuli that fuel CLL cell growth through an array of survival signaling pathways. Novel small-molecule agents targeting the B-cell receptor pathway and anti-apoptotic proteins alone or in combination have revolutionized the management of CLL, yet combination therapy carries significant toxicity and CLL remains incurable due to residual disease and relapse. Single-molecule inhibitors that can target multiple disease-driving factors are thus an attractive approach to combat both drug resistance and combination-therapy-related toxicities. We demonstrate that SRX3305, a novel small-molecule BTK/PI3K/BRD4 inhibitor that targets three distinctive facets of CLL biology, attenuates CLL cell proliferation and promotes apoptosis in a dose-dependent fashion. SRX3305 also inhibits the activation-induced proliferation of primary CLL cells in vitro and effectively blocks microenvironment-mediated survival signals, including stromal cell contact. Furthermore, SRX3305 blocks CLL cell migration toward CXCL-12 and CXCL-13, which are major chemokines involved in CLL cell homing and retention in microenvironment niches. Importantly, SRX3305 maintains its anti-tumor effects in ibrutinib-resistant CLL cells. Collectively, this study establishes the preclinical efficacy of SRX3305 in CLL, providing significant rationale for its development as a therapeutic agent for CLL and related disorders.
Collapse
Affiliation(s)
- Audrey L. Smith
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Alexandria P. Eiken
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Dalia Y. Moore
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lelisse T. Umeta
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lynette M. Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Elizabeth R. Lyden
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Avyakta Kallam
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Julie M. Vose
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Tatiana G. Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| |
Collapse
|
12
|
Tao J, Srinivasan V, Yi X, Zhao Y, Zhang H, Lin X, Zhou X, Boyce BF, Villalta PW, Ebetino FH, Ho KK, Boeckman RK, Xing L. Bone-Targeted Bortezomib Inhibits Bortezomib-Resistant Multiple Myeloma in Mice by Providing Higher Levels of Bortezomib in Bone. J Bone Miner Res 2022; 37:629-642. [PMID: 34970782 PMCID: PMC9018514 DOI: 10.1002/jbmr.4496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 11/10/2022]
Abstract
Limited treatment options exist for cancer within the bone, as demonstrated by the inevitable, pernicious course of metastatic and blood cancers. The difficulty of eliminating bone-residing cancer, especially drug-resistant cancer, necessitates novel, alternative treatments to manipulate tumor cells and their microenvironment, with minimal off-target effects. To this end, bone-targeted conjugate (BP-Btz) was generated by linking bortezomib (Btz, an anticancer, bone-stimulatory drug) to a bisphosphonate (BP, a targeting ligand) through a cleavable linker that enables spatiotemporally controlled delivery of Btz to bone under acidic conditions for treating multiple myeloma (MM). Three conjugates with different linkers were developed and screened for best efficacy in mouse model of MM. Results demonstrated that the lead candidate BP-Btz with optimal linker could overcome Btz resistance, reduced tumor burden, bone destruction, or tumor metastasis more effectively than BP or free Btz without thrombocytopenia and neurotoxicity in mice bearing myeloma. Furthermore, pharmacokinetic and pharmacodynamic studies showed that BP-Btz bound to bone matrix, released Btz in acidic conditions, and had a higher local concentration and longer half-life than Btz in bone. Our findings suggest the potential of bone-targeted Btz conjugate as an efficacious Btz-resistant MM treatment mechanism. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Jianguo Tao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Venkat Srinivasan
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | - Xiangjiao Yi
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Yingchun Zhao
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xichao Zhou
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Peter W Villalta
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Frank H Ebetino
- Department of Chemistry, University of Rochester, Rochester, NY, USA.,BioVinc, Pasadena, CA, USA
| | - Koc Kan Ho
- Ionova Life Science Co., Ltd, Shenzhen, China
| | - Robert K Boeckman
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
13
|
Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers (Basel) 2022; 14:1681. [PMID: 35406451 PMCID: PMC8996887 DOI: 10.3390/cancers14071681] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Our body is constantly exposed to pathogens or external threats, but with the immune response that our body can develop, we can fight off and defeat possible attacks or infections. Nevertheless, sometimes this threat comes from an internal factor. Situations such as the existence of a tumour also cause our immune system (IS) to be put on alert. Indeed, the link between immunology and cancer is evident these days, with IS being used as one of the important targets for treating cancer. Our IS is able to eliminate those abnormal or damaged cells found in our body, preventing the uncontrolled proliferation of tumour cells that can lead to cancer. However, in several cases, tumour cells can escape from the IS. It has been observed that immune cells, the extracellular matrix, blood vessels, fat cells and various molecules could support tumour growth and development. Thus, the developing tumour receives structural support, irrigation and energy, among other resources, making its survival and progression possible. All these components that accompany and help the tumour to survive and to grow are called the tumour microenvironment (TME). Given the importance of its presence in the tumour development process, this review will focus on one of the components of the TME: immune cells. Immune cells can support anti-tumour immune response protecting us against tumour cells; nevertheless, they can also behave as pro-tumoural cells, thus promoting tumour progression and survival. In this review, the anti-tumour and pro-tumour immunity of several immune cells will be discussed. In addition, the TME influence on this dual effect will be also analysed.
Collapse
Affiliation(s)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, 30120 Murcia, Spain;
| |
Collapse
|
14
|
Chen W, Liang W, He Y, Liu C, Chen H, Lv P, Yao Y, Zhou H. Immune microenvironment-related gene mapping predicts immunochemotherapy response and prognosis in diffuse large B-cell lymphoma. Med Oncol 2022; 39:44. [PMID: 35092504 DOI: 10.1007/s12032-021-01642-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin's lymphoma (NHL). The R-CHOP immunochemotherapy regimen is the first-line treatment option for DLBCL patients and has greatly improved the prognosis of DLBCL, making it a curable disease. However, drug resistance or relapse is the main challenge for current DLBCL treatment. Studies have shown that the tumor microenvironment plays an important role in the onset, development, and responsiveness to drugs in DLBCL. Here, we used the CIBERSORT algorithm to resolve the composition of the immune microenvironment of 471 DLBCL patients from the GEO database. We found that activated memory CD4+ T cells and γδ T cells were significantly associated with immunochemotherapy response. Weighted gene co-expression networks (WGCNA) were constructed using differentially expressed genes from immunochemotherapy responders and non-responders. The module most associated with these two types of T cells was defined as hub module. Enrichment analysis of the hub module showed that baseline immune status was significantly stronger in responders than in non-responders. A protein-protein interaction (PPI) network was constructed for hub module to identify hub genes. After survival analysis, five prognosis-related genes (CD3G, CD3D, GNB4, FCHO2, GPR183) were identified and all these genes were significantly negatively associated with PD1. Using our own patient cohort, we validated the efficacy of CD3G and CD3D in predicting immunochemotherapy response. Our study showed that CD3G, CD3D, GNB4, FCHO2, and GPR183 are involved in the regulation of the immune microenvironment of DLBCL. They can be used as biomarkers for predicting immunochemotherapy response and potential therapeutic targets in DLBCL.
Collapse
Affiliation(s)
- Wanjun Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weijie Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongjian He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chixiang Liu
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongtian Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Piao Lv
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Yao
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huayou Zhou
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Abstract
ABSTRACT Multiple myeloma is a hematological malignancy of differentiated B cells that resides primarily in bone marrow niches. Its interaction with the microenvironment is known to provide a survival advantage and plays an important role in drug resistance. Despite the increased efficacy of new treatment drugs, clinical results oftentimes fall short of in vitro observations, and this disease remains incurable. Conventional 2-dimensional cultures used to perform chemosensitivity assays and the established multiple myeloma cells lines commonly used do not replicate the conditions seen in vivo. This review presents various 3-dimensional culture platforms for myeloma that attempt to overcome this obstacle by incorporating aspects of the tumor microenvironment. The unique features of each model and contributions they have provided in personalized medicine, tumor physiology, and chemosensitivity assays will be summarized.
Collapse
|
16
|
Li P, Yuan J, Ahmed FS, McHenry A, Fu K, Yu G, Cheng H, Xu ML, Rimm DL, Pan Z. High Counts of CD68+ and CD163+ Macrophages in Mantle Cell Lymphoma Are Associated With Inferior Prognosis. Front Oncol 2021; 11:701492. [PMID: 34527580 PMCID: PMC8435777 DOI: 10.3389/fonc.2021.701492] [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: 04/28/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
Background Lymphoma-associated macrophages (LAMs) are key components in the lymphoma microenvironment, which may impact disease progression and response to therapy. There are two major subtypes of LAMs, CD68+ M1 and CD163+ M2. M2 LAMs can be transformed from M1 LAMs, particularly in certain diffuse large B-cell lymphomas (DLBCL). While mantle cell lymphoma (MCL) is well-known to contain frequent epithelioid macrophages, LAM characterization within MCL has not been fully described. Herein we evaluate the immunophenotypic subclassification, the expression of immune checkpoint molecule PD-L1, and the prognostic impact of LAMs in MCL. Materials and Methods A total of 82 MCL cases were collected and a tissue microarray block was constructed. Immunohistochemical staining was performed using CD68 and CD163, and the positive cells were recorded manually in four representative 400× fields for each case. Multiplexed quantitative immunofluorescence assays were carried out to determine PD-L1 expression on CD68+ M1 LAMs and CD163+ M2 LAMs. In addition, we assessed Ki67 proliferation rate of MCL by an automated method using the QuPath digital imaging analysis. The cut-off points of optimal separation of overall survival (OS) were analyzed using the X-Tile software, the SPSS version 26 was used to construct survival curves, and the log-rank test was performed to calculate the p-values. Results MCL had a much higher count of M1 LAMs than M2 LAMs with a CD68:CD163 ratio of 3:1. Both M1 and M2 LAMs were increased in MCL cases with high Ki67 proliferation rates (>30%), in contrast to those with low Ki67 (<30%). Increased number of M1 or M2 LAMs in MCL was associated with an inferior OS. Moreover, high expression of PD-L1 on M1 LAMs had a slightly better OS than the cases with low PD-L1 expression, whereas low expression of PD-L1 on M2 LAMs had a slightly improved OS, although both were not statistically significant. Conclusions In contrast to DLBCL, MCL had a significantly lower rate of M1 to M2 polarization, and the high levels of M1 and M2 LAMs were associated with poor OS. Furthermore, differential PD-L1 expressions on LAMs may partially explain the different functions of tumor-suppressing or tumor-promoting of M1 and M2 LAMs, respectively.
Collapse
Affiliation(s)
- Philippa Li
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Ji Yuan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fahad Shabbir Ahmed
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States.,Department of Pathology, Wayne State University, Detroit, MI, United States
| | - Austin McHenry
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Kai Fu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pathology and Laboratory Medicine, Roswell Park Cancer Center, Buffalo, NY, United States
| | - Guohua Yu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pathology, Yantai Yuhuangding Hospital, Yantai, China
| | - Hongxia Cheng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mina L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Zenggang Pan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
17
|
Pediatric Mixed-Phenotype Acute Leukemia: What's New? Cancers (Basel) 2021; 13:cancers13184658. [PMID: 34572885 PMCID: PMC8469808 DOI: 10.3390/cancers13184658] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Pediatric mixed-phenotype leukemia is a rare form of blood cancer in children. In this review, we cover both the evolution of treatment over the past several years and outline new emerging concepts in this disease. Abstract Mixed-phenotype acute leukemias (MPAL) are rare in children and often lack consensus on optimal management. This review examines the current controversies and emerging paradigms in the management of pediatric MPAL. We examine risk stratification, outcomes of recent retrospective and prospective collaborative trials, and the role of transplantation and precision genomics, and outline emerging targets and concepts in this rare entity.
Collapse
|
18
|
Zheng W, Lin Q, Issah MA, Liao Z, Shen J. Identification of PLA2G7 as a novel biomarker of diffuse large B cell lymphoma. BMC Cancer 2021; 21:927. [PMID: 34404374 PMCID: PMC8369790 DOI: 10.1186/s12885-021-08660-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 08/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma is the most common form of non-Hodgkin lymphoma globally, and patients with relapsed or refractory DLBCL typically experience poor long-term outcomes. METHODS Differentially expressed genes associated with DLBCL were identified using two GEO datasets in an effort to detect novel diagnostic or prognostic biomarkers of this cancer type, after which receiver operating characteristic curve analyses were conducted. Genes associated with DLBCL patient prognosis were additionally identified via WCGNA analyses of the TCGA database. The expression of PLA2G7 in DLBCL patient clinical samples was further assessed, and the functional role of this gene in DLBCL was assessed through in vitro and bioinformatics analyses. RESULTS DLBCL-related DEGs were found to be most closely associated with immune responses, cell proliferation, and angiogenesis. WCGNA analyses revealed that PLA2G7 exhibited prognostic value in DLBCL patients, and the upregulation of this gene in DLBCL patient samples was subsequently validated. PLA2G7 was also found to be closely linked to tumor microenvironmental composition such that DLBCL patients expressing higher levels of this gene exhibited high local monocyte and gamma delta T cell levels. In vitro experiments also revealed that knocking down PLA2G7 expression was sufficient to impair the migration and proliferation of DLBCL cells while promoting their apoptotic death. Furthmore, the specific inhibitor of PLA2G7, darapladib, could noticeably restrained the DLBCL cell viability and induced apoptosis. CONCLUSIONS PLA2G7 may represent an important diagnostic, prognostic, or therapeutic biomarker in patients with DLBCL.
Collapse
Affiliation(s)
- Weili Zheng
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory on Hematology; Fujian Medical University Union Hospital, Fuzhou, China
| | - Qiaochu Lin
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory on Hematology; Fujian Medical University Union Hospital, Fuzhou, China
| | - Mohammed Awal Issah
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory on Hematology; Fujian Medical University Union Hospital, Fuzhou, China
| | - Ziyuan Liao
- Meng Chao Hepatobiliary Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Jianzhen Shen
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory on Hematology; Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
19
|
Xu T, Chai J, Wang K, Jia Q, Liu Y, Wang Y, Xu J, Yu K, Zhao D, Ma J, Fan L, Yan Q, Guo S, Chen G, Chen Q, Xiao H, Liu F, Qi C, Liang R, Li M, Wang Z. Tumor Immune Microenvironment Components and Checkpoint Molecules in Anaplastic Variant of Diffuse Large B-Cell Lymphoma. Front Oncol 2021; 11:638154. [PMID: 34221962 PMCID: PMC8242181 DOI: 10.3389/fonc.2021.638154] [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: 12/05/2020] [Accepted: 05/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background Anaplastic diffuse large B-cell lymphoma(A-DLBCL) is a rare morphological subtype characterized by the presence of polygonal, bizarre-shaped tumor cells. Our previous research found that A-DLBCL displays many genetic alterations and biological features that differ greatly from those of ordinary DLBCL. However, the status of tumor immune microenvironment components and checkpoint molecules in A-DLBCL remains unclear. Methods Thirty A-DLBCL patients were enrolled to study tumor immune microenvironment components and checkpoint molecules and their associations with clinicopathological features and prognosis. Results Patients with A-DLBCL presented higher expression of PD-L1 (40% vs 10%, P=0.004) than patients with ordinary DLBCL. FISH analysis showed that extra copies of PD-L1 were more frequent in A-DLBCL cases than in ordinary DLBCL cases (23.3% vs 4.0%, P=0.001). The numbers of PD-1+ TILs (tumor infiltrating lymphocytes) and CD8+T cells were significantly lower in A-DLBCL versus ordinary DLBCL. In contrast, the numbers of GATA3+ Th2 cells, FOXP3+ Tregs and CD33+ myeloid-derived suppressor cells (MDSCs) were significantly higher in A-DLBCL than in ordinary DLBCL. The associations between clinicopathological features and tumor immune microenvironment cell frequency were analyzed in A-DLBCL patients. Briefly, the number of PD-1+ TILs was lower and the number of CD33+ MDSCs was higher in patients with mutated TP53 compared to those with wild-type TP53. The number of FOXP3+ Tregs was much lower in patients with a noncomplete response (CR) to chemotherapy than in those with a complete response. The number of CD8+ T cells showed a decreasing trend in patients with high International Prognostic Index (IPI) scores and in those with concurrent MYC and BCL2 and/or BCL6 abnormalities. Univariate survival analysis showed that patients with PD-L1+, mPD-L1+(PD-L1+ nonmalignant stromal cells) or mPD-L1+ status had a significantly poorer overall survival (OS) than those with PD-L1- status. An increase in the number of CD3+ T cells, FOXP3+ Treg cells and T-bet+ Th1 cells was significantly associated with prolonged OS in patients with A-DLBCL. Conclusion Our study suggests that A-DLBCL displays a distinct pattern of tumor immune microenvironment components and checkpoint molecules that distinguish it from ordinary DLBCL. The analysis of tumor immune microenvironment components and checkpoint molecules could help in predicting the prognosis of A-DLBCL patients and determining therapeutic strategies targeting the tumor immune microenvironment.
Collapse
Affiliation(s)
- Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Jia Chai
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Kaijing Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qingge Jia
- Xi'an International Medical Center, Northwest University, Xi'an, China
| | - Yixiong Liu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Junpeng Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Kangjie Yu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Danhui Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Jing Ma
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Linni Fan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qingguo Yan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Shuangping Guo
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Gang Chen
- Department of Pathology, Fujian Cancer Hospital, Fuzhou, China
| | - Qiongrong Chen
- Department of Pathology, Hubei Cancer Hospital, Wuhan, China
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Fang Liu
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Chubo Qi
- Department of Pathology, Hubei Cancer Hospital, Wuhan, China
| | - Rong Liang
- Department of Hematology, People's Liberation Army Centre for Hematologic Disorders, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Zhe Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| |
Collapse
|
20
|
Sadeghi L, Wright AP. Migration and Adhesion of B-Lymphocytes to Specific Microenvironments in Mantle Cell Lymphoma: Interplay between Signaling Pathways and the Epigenetic Landscape. Int J Mol Sci 2021; 22:6247. [PMID: 34200679 PMCID: PMC8228059 DOI: 10.3390/ijms22126247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Lymphocyte migration to and sequestration in specific microenvironments plays a crucial role in their differentiation and survival. Lymphocyte trafficking and homing are tightly regulated by signaling pathways and is mediated by cytokines, chemokines, cytokine/chemokine receptors and adhesion molecules. The production of cytokines and chemokines is largely controlled by transcription factors in the context of a specific epigenetic landscape. These regulatory factors are strongly interconnected, and they influence the gene expression pattern in lymphocytes, promoting processes such as cell survival. The epigenetic status of the genome plays a key role in regulating gene expression during many key biological processes, and it is becoming more evident that dysregulation of epigenetic mechanisms contributes to cancer initiation, progression and drug resistance. Here, we review the signaling pathways that regulate lymphoma cell migration and adhesion with a focus on Mantle cell lymphoma and highlight the fundamental role of epigenetic mechanisms in integrating signals at the level of gene expression throughout the genome.
Collapse
Affiliation(s)
- Laia Sadeghi
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden;
| | | |
Collapse
|
21
|
Zhong W, Liu X, Zhu Z, Li Q, Li K. High levels of Tim-3 +Foxp3 +Treg cells in the tumor microenvironment is a prognostic indicator of poor survival of diffuse large B cell lymphoma patients. Int Immunopharmacol 2021; 96:107662. [PMID: 33864956 DOI: 10.1016/j.intimp.2021.107662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/27/2022]
Abstract
Foxp3+Treg cells display phenotypic and functional heterogeneity, which express high levels of T cell immunoglobulin and mucin-domain containing-3 (Tim-3) in the tumor microenvironment (TME) of colorectal and lung cancer. High abundance of Tim-3+Foxp3+Treg (TFT) cells are associated with poor prognosis in these patients. However, the expression patterns and roles of TFT cells in TME of diffuse large B cell lymphoma (DLBCL) remain to be established. Double immunofluorescence and flow cytometry analyses were employed to investigate TFT cell enrichment in paraffin-embedded fresh tumor tissues from patients with DLBCL. Spearman's or Pearson's correlation and Kaplan-Meier survival analyses were further applied to decide the prognostic value of TFT cell levels in DLBCL. The IL-10-secreting function of TFT cells in vitro was examined via flow cytometry and ELISA. Our results showed for the first time that TFT cells are highly enriched in TME of DLBCL patients and associated with predictions of poor prognoses. TFT cell-induced secretion of IL-10 in the TME was suppressed by an anti-Tim-3 antibody in vitro. In conclusion, high abundance of TFT cells in the TME is predictive of poor outcomes of DLBCL. TFT cells promote DLBCL development partly by secreting IL-10 in the TME. Anti-Tim-3 antibodies (that block IL-10 secretion) may present an effective therapeutic agent for DLBCL.
Collapse
Affiliation(s)
- Weijie Zhong
- Department of Geriatrics, Hematology & Oncology Ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Xiudan Liu
- Department of Medical Ultrasound, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Zhigang Zhu
- Department of Geriatrics, Hematology & Oncology Ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Qingshan Li
- Department of Hematology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong 510220, China.
| | - Kangbao Li
- Department of Geriatrics, Gastroenterology Ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China.
| |
Collapse
|
22
|
Sun S, Tao J, Sedghizadeh PP, Cherian P, Junka AF, Sodagar E, Xing L, Boeckman RK, Srinivasan V, Yao Z, Boyce BF, Lipe B, Neighbors JD, Russell RGG, McKenna CE, Ebetino FH. Bisphosphonates for delivering drugs to bone. Br J Pharmacol 2021; 178:2008-2025. [PMID: 32876338 DOI: 10.1111/bph.15251] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
Advances in the design of potential bone-selective drugs for the treatment of various bone-related diseases are creating exciting new directions for multiple unmet medical needs. For bone-related cancers, off-target/non-bone toxicities with current drugs represent a significant barrier to the quality of life of affected patients. For bone infections and osteomyelitis, bacterial biofilms on infected bones limit the efficacy of antibiotics because it is hard to access the bacteria with current approaches. Promising new experimental approaches to therapy, based on bone-targeting of drugs, have been used in animal models of these conditions and demonstrate improved efficacy and safety. The success of these drug-design strategies bodes well for the development of therapies with improved efficacy for the treatment of diseases affecting the skeleton. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.
Collapse
Affiliation(s)
| | - Jianguo Tao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Parish P Sedghizadeh
- Center for Biofilms, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | | | - Adam F Junka
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw; Wroclaw Research Centre EIT, Wroclaw, Poland
| | - Esmat Sodagar
- Center for Biofilms, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Robert K Boeckman
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | | | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea Lipe
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jeffrey D Neighbors
- BioVinc, Pasadena, CA, USA.,Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - R Graham G Russell
- The Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK.,Department of Oncology and Metabolism, The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, California, USA
| | - Frank H Ebetino
- BioVinc, Pasadena, CA, USA.,Department of Chemistry, University of Rochester, Rochester, NY, USA.,Department of Oncology and Metabolism, The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
23
|
Zhang K, Roy NK, Vicioso Y, Woo J, Beck R, de Lima M, Caimi P, Feinberg D, Parameswaran R. BAFF receptor antibody for mantle cell lymphoma therapy. Oncoimmunology 2021; 10:1893501. [PMID: 33747637 PMCID: PMC7939563 DOI: 10.1080/2162402x.2021.1893501] [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] [Indexed: 11/26/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive form of B cell non-Hodgkin’s lymphoma and remains incurable under current treatment modalities. One of the main reasons for treatment failure is the development of drug resistance. Accumulating evidence suggests that B cell activating factor (BAFF) and BAFF receptor (BAFF-R) play an important role in the proliferation and survival of malignant B cells. High serum BAFF levels are often correlated with poor drug response and relapse in MCL patients. Our study shows that BAFF-R is expressed on both MCL patient cells and cell lines. BAFF-R knockdown leads to MCL cell death showing the importance of BAFF-R signaling in MCL survival. Moderate knockdown of BAFF-R in MCL cells did not affect its viability, but sensitized them to cytarabine treatment in vitro and in vivo, with prolonged mice survival. Anti-BAFF-R antibody treatment promoted drug-induced MCL cell death. Conversely, the addition of recombinant BAFF (rhBAFF) to MCL cells protected them from cytarabine-induced apoptosis. We tested the efficacy of a humanized defucosylated ADCC optimized anti-BAFF-R antibody in killing MCL. Our data show both in vitro and in vivo efficacy of this antibody for MCL therapy. To conclude, our data indicate that BAFF/BAFF-R signaling is crucial for survival and involved in drug resistance of MCL. Targeting BAFF-R using BAFF-R antibody might be a promising therapeutical strategy to treat MCL patients resistant to chemotherapy.
Collapse
Affiliation(s)
- Keman Zhang
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nand K Roy
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yorleny Vicioso
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Janghee Woo
- Novartis Institute for BioMedical Research, Translational Clinical Oncology, NJ, USA
| | - Rose Beck
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Marcos de Lima
- Hematology and Oncology, University Hospitals, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Paolo Caimi
- Hematology and Oncology, University Hospitals, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daniel Feinberg
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Reshmi Parameswaran
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Potential Role of microRNAs in inducing Drug Resistance in Patients with Multiple Myeloma. Cells 2021; 10:cells10020448. [PMID: 33672466 PMCID: PMC7923438 DOI: 10.3390/cells10020448] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis for newly diagnosed subjects with multiple myeloma (MM) has significantly progressed in recent years. However, most MM patients relapse and after several salvage therapies, the onset of multidrug resistance provokes the occurrence of a refractory disease. A continuous and bidirectional exchange of information takes place between the cells of the microenvironment and neoplastic cells to solicit the demands of cancer cells. Among the molecules serving as messengers, there are microRNAs (miRNA), a family of small noncoding RNAs that regulate gene expression. Numerous miRNAs are associated with drug resistance, also in MM, and the modulation of their expression or activity might be explored to reverse it. In this review we report the most recent studies concerning the relationship between miRNAs and chemoresistance to the most frequently used drugs, such as proteasome inhibitors, steroids, alkylating agents and immunomodulators. The experimental use of antagomirs or miRNA mimics have successfully been proven to counteract chemoresistance and display synergistic effects with antimyeloma drugs which could represent a fundamental moment to overcome resistance in MM treatment.
Collapse
|
25
|
Follicular Lymphoma Microenvironment: An Intricate Network Ready for Therapeutic Intervention. Cancers (Basel) 2021; 13:cancers13040641. [PMID: 33562694 PMCID: PMC7915642 DOI: 10.3390/cancers13040641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Follicular Lymphoma (FL), the most common indolent non-Hodgkin's B cell lymphoma, is a paradigm of the immune microenvironment's contribution to disease onset, progression, and heterogeneity. Over the last few years, state-of-the-art technologies, including whole-exome sequencing, single-cell RNA sequencing, and mass cytometry, have precisely dissected the specific cellular phenotypes present in the FL microenvironment network and their role in the disease. In this already complex picture, the presence of recurring mutations, including KMT2D, CREBBP, EZH2, and TNFRSF14, have a prominent contributory role, with some of them finely tuning this exquisite dependence of FL on its microenvironment. This precise characterization of the enemy (FL) and its allies (microenvironment) has paved the way for the development of novel therapies aimed at dismantling this contact network, weakening tumor cell support, and reactivating the host's immune response against the tumor. In this review, we will describe the main microenvironment actors, together with the current and future therapeutic approaches targeting them.
Collapse
|
26
|
Vlachonikola E, Stamatopoulos K, Chatzidimitriou A. T Cells in Chronic Lymphocytic Leukemia: A Two-Edged Sword. Front Immunol 2021; 11:612244. [PMID: 33552073 PMCID: PMC7857025 DOI: 10.3389/fimmu.2020.612244] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of mature, antigen-experienced B lymphocytes. Despite great progress recently achieved in the management of CLL, the disease remains incurable, underscoring the need for further investigation into the underlying pathophysiology. Microenvironmental crosstalk has an established role in CLL pathogenesis and progression. Indeed, the malignant CLL cells are strongly dependent on interactions with other immune and non-immune cell populations that shape a highly orchestrated network, the tumor microenvironment (TME). The composition of the TME, as well as the bidirectional interactions between the malignant clone and the microenvironmental elements have been linked to disease heterogeneity. Mounting evidence implicates T cells present in the TME in the natural history of the CLL as well as in the establishment of certain CLL hallmarks e.g. tumor evasion and immune suppression. CLL is characterized by restrictions in the T cell receptor gene repertoire, T cell oligoclonal expansions, as well as shared T cell receptor clonotypes amongst patients, strongly alluding to selection by restricted antigenic elements of as yet undisclosed identity. Further, the T cells in CLL exhibit a distinctive phenotype with features of “exhaustion” likely as a result of chronic antigenic stimulation. This might be relevant to the fact that, despite increased numbers of oligoclonal T cells in the periphery, these cells are incapable of mounting effective anti-tumor immune responses, a feature perhaps also linked with the elevated numbers of T regulatory subpopulations. Alterations of T cell gene expression profile are associated with defects in both the cytoskeleton and immune synapse formation, and are generally induced by direct contact with the malignant clone. That said, these abnormalities appear to be reversible, which is why therapies targeting the T cell compartment represent a reasonable therapeutic option in CLL. Indeed, novel strategies, including CAR T cell immunotherapy, immune checkpoint blockade and immunomodulation, have come to the spotlight in an attempt to restore the functionality of T cells and enhance targeted cytotoxic activity against the malignant clone.
Collapse
Affiliation(s)
- Elisavet Vlachonikola
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Genetics and Molecular Biology, Faculty of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Chatzidimitriou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Merdan S, Subramanian K, Ayer T, Van Weyenbergh J, Chang A, Koff JL, Flowers C. Gene expression profiling-based risk prediction and profiles of immune infiltration in diffuse large B-cell lymphoma. Blood Cancer J 2021; 11:2. [PMID: 33414466 PMCID: PMC7791044 DOI: 10.1038/s41408-020-00404-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
The clinical risk stratification of diffuse large B-cell lymphoma (DLBCL) relies on the International Prognostic Index (IPI) for the identification of high-risk disease. Recent studies suggest that the immune microenvironment plays a role in treatment response prediction and survival in DLBCL. This study developed a risk prediction model and evaluated the model’s biological implications in association with the estimated profiles of immune infiltration. Gene-expression profiling of 718 patients with DLBCL was done, for which RNA sequencing data and clinical covariates were obtained from Reddy et al. (2017). Using unsupervised and supervised machine learning methods to identify survival-associated gene signatures, a multivariable model of survival was constructed. Tumor-infiltrating immune cell compositions were enumerated using CIBERSORT deconvolution analysis. A four gene-signature-based score was developed that separated patients into high- and low-risk groups. The combination of the gene-expression-based score with the IPI improved the discrimination on the validation and complete sets. The gene signatures were successfully validated with the deconvolution output. Correlating the deconvolution findings with the gene signatures and risk score, CD8+ T-cells and naïve CD4+ T-cells were associated with favorable prognosis. By analyzing the gene-expression data with a systematic approach, a risk prediction model that outperforms the existing risk assessment methods was developed and validated.
Collapse
Affiliation(s)
- Selin Merdan
- Department of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Kritika Subramanian
- Department of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA. .,Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| | - Turgay Ayer
- Department of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Andres Chang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jean L Koff
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher Flowers
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
ROS Overproduction Sensitises Myeloma Cells to Bortezomib-Induced Apoptosis and Alleviates Tumour Microenvironment-Mediated Cell Resistance. Cells 2020; 9:cells9112357. [PMID: 33114738 PMCID: PMC7693395 DOI: 10.3390/cells9112357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm that remains incurable due to innate or acquired resistance. Although MM cells produce high intracellular levels of reactive oxygen species (ROS), we hypothesised that they could remain sensitive to ROS unbalance. We tested if the inhibition of ROS, on one hand, or the overproduction of ROS, on the other, could (re)sensitise cells to bortezomib (BTZ). Two drugs were used in a panel of MM cell lines with various responses to BTZ: VAS3947 (VAS), an inhibitor of NADPH oxidase and auranofin (AUR), an inhibitor of thioredoxin reductase (TXNRD1), an antioxidant enzyme overexpressed in MM cells. We used several culture models: in suspension, on a fibronectin layer, in coculture with HS-5 mesenchymal cells, and/or in 3-D culture (or spheroids) to study the response of MM primary cells and cell lines. Several MM cell lines were sensitive to VAS but the combination with BTZ showed antagonistic or additive effects at best. By contrast, in all culture systems studied, the combined AUR/BTZ treatment showed synergistic effects on cell lines, including those less sensitive to BTZ and primary cells. MM cell death is due to the activation of apoptosis and autophagy. Modulating the redox balance of MM cells could be an effective therapy for refractory or relapse post-BTZ patients.
Collapse
|
29
|
Younes S, Natkunam Y. FOXes at play in the lymphoma landscape. Leuk Lymphoma 2020; 62:5-7. [PMID: 33064049 DOI: 10.1080/10428194.2020.1834099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Sheren Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
30
|
Xu Y, Sun W, Li F. De Novo CD5+ Diffuse Large B-Cell Lymphoma: Biology, Mechanism, and Treatment Advances. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e782-e790. [DOI: 10.1016/j.clml.2020.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 12/27/2022]
|
31
|
Pollari M, Pellinen T, Karjalainen-Lindsberg ML, Kellokumpu-Lehtinen PL, Leivonen SK, Leppä S. Adverse prognostic impact of regulatory T-cells in testicular diffuse large B-cell lymphoma. Eur J Haematol 2020; 105:712-721. [PMID: 32632935 DOI: 10.1111/ejh.13484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Testicular diffuse large B-cell lymphoma (T-DLBCL) is a rare and aggressive extranodal lymphoma. We have previously shown that high content of tumor-infiltrating lymphocytes (TILs) and PD-1 expressing TILs associate with better survival in T-DLBCL. In this study, we have further characterized distinct TIL subtypes and their proportions in association with patient demographics and survival. METHODS We used multiplex immunohistochemistry to characterize TIL phenotypes, including cytotoxic T-cells (CTLs; CD8+ , OX40+ , Granzyme B+ , Ki-67+ , LAG-3+ , TIM-3+ , PD-1+ ), CD4+ T-cells (CD3+ , CD4+ , TIM-3+ , LAG-3+ ), regulatory T-cells (Tregs; CD3+ , CD4+ , FoxP3+ ), and T helper 1 cells (Th1; CD3+ , CD4+ , T-bet+ ) in 79 T-DLBCLs, and correlated the findings with patient demographics and outcome. RESULTS We observed a substantial variation in TIL phenotypes between the patients. The most prominent CD8+ TILs were Ki-67+ and TIM-3+ CTLs, whereas the most prominent CD4+ TILs were FoxP3+ Tregs. Despite the overall favorable prognostic impact of high TIL content, we found a subpopulation of T-bet+ FoxP3+ Tregs that had a significant adverse impact on survival. Lower content of CTLs with activated or exhausted phenotypes correlated with aggressive clinical features. CONCLUSIONS Our results demonstrate significant variation in TIL phenotypes and emphasize the adverse prognostic impact of Tregs in T-DLBCL.
Collapse
Affiliation(s)
- Marjukka Pollari
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | | | - Pirkko-Liisa Kellokumpu-Lehtinen
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Suvi-Katri Leivonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Sirpa Leppä
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.,iCAN, Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
32
|
McCulloch R, Eyre TA, Rule S. What Causes Bruton Tyrosine Kinase Inhibitor Resistance in Mantle Cell Lymphoma and How Should We Treat Such Patients? Hematol Oncol Clin North Am 2020; 34:923-939. [PMID: 32861287 DOI: 10.1016/j.hoc.2020.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this review, we explore insights into the pathophysiology of Bruton tyrosine kinase inhibitor (BTKi) resistance in mantle cell lymphoma, and consider potential therapeutic targets. We review the possible clinical benefits of giving BTKis alongside other novel therapies, and evaluate clinical data for treatment strategies post BTKi progression that may help guide current practice. We conclude by considering future approaches, including the potential role of chimeric antigen receptor T-cell therapy.
Collapse
Affiliation(s)
- Rory McCulloch
- Department of Haematology, Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Toby A Eyre
- Department of Haematology, Oxford University Hospitals, Oxford, UK
| | - Simon Rule
- Department of Haematology, Peninsula Medical School, University of Plymouth, John Bull Building, Plymouth PL6 8BU, UK.
| |
Collapse
|
33
|
Zhou H, Zheng C, Huang DS. A prognostic gene model of immune cell infiltration in diffuse large B-cell lymphoma. PeerJ 2020; 8:e9658. [PMID: 32844062 PMCID: PMC7414766 DOI: 10.7717/peerj.9658] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Background Immune cells in the tumor microenvironment are an important prognostic indicator in diffuse large B-cell lymphoma (DLBCL). However, information on the heterogeneity and risk stratification of these cells is limited. We sought to develop a novel immune model to evaluate the prognostic intra-tumoral immune landscape of patients with DLBCL. Methods The ESTIMATE and CIBERSORT algorithms were used to estimate the numbers of 22 infiltrating immune cells based on the gene expression profiles of 229 patients with DLBCL who were recruited from a public database. The least absolute shrinkage and selection operator (Lasso) penalized regression analyses and nomogram model were used to construct and evaluate the prognostic immunoscore (PIS) model for overall survival prediction. An immune gene prognostic score (IGPS) was generated by Gene Set Enrichment Analysis (GSEA) and Cox regression analysis was and validated in an independent NCBI GEO dataset (GSE10846). Results A higher proportion of activated natural killer cells was associated with a poor outcome. A total of five immune cells were selected in the Lasso model and DLBCL patients with high PIS showed a poor prognosis (hazard ratio (HR) 2.16; 95% CI [1.33-3.50]; P = 0.002). Differences in immunoscores and their related outcomes were attributed to eight specific immune genes involved in the cytokine-cytokine receptor interaction and chemokine signaling pathways. The IGPS based on a weighted formula of eight genes is an independent prognostic factor (HR: 2.14, 95% CI [1.40-3.28]), with high specificity and sensitivity in the validation dataset. Conclusions Our findings showed that a PIS model based on immune cells is associated with the prognosis of DLBCL. We developed a novel immune-related gene-signature model associated with the PIS model and enhanced the prognostic functionality for the prediction of overall survival in patients with DLBCL.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China.,Department of Impression Evidence Examination Technology, Criminal Investigation Police University of China, Shenyang, Liaoning, China
| | - Chang Zheng
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - De-Sheng Huang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China.,Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Department of Education, Shenyang, Liaoning, China
| |
Collapse
|
34
|
Inflammatory Cells in Diffuse Large B Cell Lymphoma. J Clin Med 2020; 9:jcm9082418. [PMID: 32731512 PMCID: PMC7463675 DOI: 10.3390/jcm9082418] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/17/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL), known as the most common non-Hodgkin lymphoma (NHL) subtype, is characterized by high clinical and biological heterogeneity. The tumor microenvironment (TME), in which the tumor cells reside, is crucial in the regulation of tumor initiation, progression, and metastasis, but it also has profound effects on therapeutic efficacy. The role of immune cells during DLBCL development is complex and involves reciprocal interactions between tumor cells, adaptive and innate immune cells, their soluble mediators and structural components present in the tumor microenvironment. Different immune cells are recruited into the tumor microenvironment and exert distinct effects on tumor progression and therapeutic outcomes. In this review, we focused on the role of macrophages, Neutrophils, T cells, natural killer cells and dendritic cells in the DLBCL microenvironment and their implication as target for DLBCL treatment. These new therapies, carried out by the induction of adaptive immunity through vaccination or passive of immunologic effectors delivery, enhance the ability of the immune system to react against the tumor antigens inducing the destruction of tumor cells.
Collapse
|
35
|
Solimando AG, Annese T, Tamma R, Ingravallo G, Maiorano E, Vacca A, Specchia G, Ribatti D. New Insights into Diffuse Large B-Cell Lymphoma Pathobiology. Cancers (Basel) 2020; 12:cancers12071869. [PMID: 32664527 PMCID: PMC7408689 DOI: 10.3390/cancers12071869] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma (NHL), accounting for about 40% of all cases of NHL. Analysis of the tumor microenvironment is an important aspect of the assessment of the progression of DLBCL. In this review article, we analyzed the role of different cellular components of the tumor microenvironment, including mast cells, macrophages, and lymphocytes, in the tumor progression of DLBCL. We examined several approaches to confront the available pieces of evidence, whereby three key points emerged. DLBCL is a disease of malignant B cells spreading and accumulating both at nodal and at extranodal sites. In patients with both nodal and extranodal lesions, the subsequent induction of a cancer-friendly environment appears pivotal. The DLBCL cell interaction with mature stromal cells and vessels confers tumor protection and inhibition of immune response while delivering nutrients and oxygen supply. Single cells may also reside and survive in protected niches in the nodal and extranodal sites as a source for residual disease and relapse. This review aims to molecularly and functionally recapitulate the DLBCL–milieu crosstalk, to relate niche and pathological angiogenic constitution and interaction factors to DLBCL progression.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico-IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080.5478326 (D.R.)
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy; (T.A.); (R.T.)
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy; (T.A.); (R.T.)
| | - Giuseppe Ingravallo
- Department of Emergency and Transplantation, Pathology Section, University of Bari Medical School, 70100 Bari, Italy; (G.I.); (E.M.)
| | - Eugenio Maiorano
- Department of Emergency and Transplantation, Pathology Section, University of Bari Medical School, 70100 Bari, Italy; (G.I.); (E.M.)
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
| | - Giorgina Specchia
- Department of Emergency and Transplantation, Hematology Section, University of Bari Medical School, 70100 Bari, Italy;
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy; (T.A.); (R.T.)
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080.5478326 (D.R.)
| |
Collapse
|
36
|
Li C, Romero-Masters JC, Huebner S, Ohashi M, Hayes M, Bristol JA, Nelson SE, Eichelberg MR, Van Sciver N, Ranheim EA, Scott RS, Johannsen EC, Kenney SC. EBNA2-deleted Epstein-Barr virus (EBV) isolate, P3HR1, causes Hodgkin-like lymphomas and diffuse large B cell lymphomas with type II and Wp-restricted latency types in humanized mice. PLoS Pathog 2020; 16:e1008590. [PMID: 32542010 PMCID: PMC7316346 DOI: 10.1371/journal.ppat.1008590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/25/2020] [Accepted: 05/01/2020] [Indexed: 12/15/2022] Open
Abstract
EBV transforms B cells in vitro and causes human B-cell lymphomas including classical Hodgkin lymphoma (CHL), Burkitt lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL). The EBV latency protein, EBNA2, transcriptionally activates the promoters of all latent viral protein-coding genes expressed in type III EBV latency and is essential for EBV's ability to transform B cells in vitro. However, EBNA2 is not expressed in EBV-infected CHLs and BLs in humans. EBV-positive CHLs have type II latency and are largely driven by the EBV LMP1/LMP2A proteins, while EBV-positive BLs, which usually have type I latency are largely driven by c-Myc translocations, and only express the EBNA1 protein and viral non-coding RNAs. Approximately 15% of human BLs contain naturally occurring EBNA2-deleted viruses that support a form of viral latency known as Wp-restricted (expressing the EBNA-LP, EBNA3A/3B/3C, EBNA1 and BHRF1 proteins), but whether Wp-restricted latency and/or EBNA2-deleted EBV can induce lymphomas in humanized mice, or in the absence of c-Myc translocations, is unknown. Here we show that a naturally occurring EBNA2-deleted EBV strain (P3HR1) isolated from a human BL induces EBV-positive B-cell lymphomas in a subset of infected cord blood-humanized (CBH) mice. Furthermore, we find that P3HR1-infected lymphoma cells support two different viral latency types and phenotypes that are mutually exclusive: 1) Large (often multinucleated), CD30-positive, CD45-negative cells reminiscent of the Reed-Sternberg (RS) cells in CHL that express high levels of LMP1 but not EBNA-LP (consistent with type II viral latency); and 2) smaller monomorphic CD30-negative DLBCL-like cells that express EBNA-LP and EBNA3A but not LMP1 (consistent with Wp-restricted latency). These results reveal that EBNA2 is not absolutely required for EBV to form tumors in CBH mice and suggest that P3HR1 virus can be used to model EBV positive lymphomas with both Wp-restricted and type II latency in vivo.
Collapse
MESH Headings
- Animals
- Cell Line
- Epstein-Barr Virus Infections/genetics
- Epstein-Barr Virus Infections/metabolism
- Epstein-Barr Virus Infections/pathology
- Epstein-Barr Virus Infections/virology
- Epstein-Barr Virus Nuclear Antigens/genetics
- Epstein-Barr Virus Nuclear Antigens/metabolism
- Gene Deletion
- Herpesvirus 4, Human/pathogenicity
- Herpesvirus 4, Human/physiology
- Hodgkin Disease/genetics
- Hodgkin Disease/metabolism
- Hodgkin Disease/pathology
- Hodgkin Disease/virology
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/virology
- Mice
- Viral Proteins/genetics
- Viral Proteins/metabolism
- Virus Latency
Collapse
Affiliation(s)
- Chunrong Li
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - James C. Romero-Masters
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shane Huebner
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Makoto Ohashi
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mitchell Hayes
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jillian A. Bristol
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Scott E. Nelson
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mark R. Eichelberg
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nicholas Van Sciver
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erik A. Ranheim
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rona S. Scott
- Center for Molecular and Tumor Virology, LSU Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Eric C. Johannsen
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shannon C. Kenney
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
37
|
The Interplay between MicroRNAs and the Components of the Tumor Microenvironment in B-Cell Malignancies. Int J Mol Sci 2020; 21:ijms21093387. [PMID: 32403283 PMCID: PMC7246984 DOI: 10.3390/ijms21093387] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/22/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
An increased focus is being placed on the tumorigenesis and contexture of tumor microenvironment in hematopoietic and solid tumors. Despite recent clinical revolutions in adoptive T-cell transfer approaches and immune checkpoint blockade, tumor microenvironment is a major obstacle to tumor regression in B-cell malignancies. A transcriptional alteration of coding and non-coding RNAs, such as microRNAs (miRNAs), has been widely demonstrated in the tumor microenvironment of B-cell malignancies. MiRNAs have been associated with different clinical-biological forms of B-cell malignancies and involved in the regulation of B lymphocyte development, maturation, and function, including B-cell activation and malignant transformation. Additionally, tumor-secreted extracellular vesicles regulate recipient cell functions in the tumor microenvironment to facilitate metastasis and progression by delivering miRNA contents to neighboring cells. Herein, we focus on the interplay between miRNAs and tumor microenvironment components in the different B-cell malignancies and its impact on diagnosis, proliferation, and involvement in treatment resistance.
Collapse
|
38
|
Granai M, Mundo L, Akarca AU, Siciliano MC, Rizvi H, Mancini V, Onyango N, Nyagol J, Abinya NO, Maha I, Margielewska S, Wi W, Bibas M, Piccaluga PP, Quintanilla-Martinez L, Fend F, Lazzi S, Leoncini L, Marafioti T. Immune landscape in Burkitt lymphoma reveals M2-macrophage polarization and correlation between PD-L1 expression and non-canonical EBV latency program. Infect Agent Cancer 2020; 15:28. [PMID: 32391073 PMCID: PMC7201729 DOI: 10.1186/s13027-020-00292-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The Tumor Microenviroment (TME) is a complex milieu that is increasingly recognized as a key factor in multiple stages of disease progression and responses to therapy as well as escape from immune surveillance. However, the precise contribution of specific immune effector and immune suppressor components of the TME in Burkitt lymphoma (BL) remains poorly understood. METHODS In this paper, we applied the computational algorithm CIBERSORT to Gene Expression Profiling (GEP) datasets of 40 BL samples to draw a map of immune and stromal components of TME. Furthermore, by multiple immunohistochemistry (IHC) and multispectral immunofluorescence (IF), we investigated the TME of additional series of 40 BL cases to evaluate the role of the Programmed Death-1 and Programmed Death Ligand-1 (PD-1/PD-L1) immune checkpoint axis. RESULTS Our results indicate that M2 polarized macrophages are the most prominent TME component in BL. In addition, we investigated the correlation between PD-L1 and latent membrane protein-2A (LMP2A) expression on tumour cells, highlighting a subgroup of BL cases characterized by a non-canonical latency program of EBV with an activated PD-L1 pathway. CONCLUSION In conclusion, our study analysed the TME in BL and identified a tolerogenic immune signature highlighting new potential therapeutic targets.
Collapse
Affiliation(s)
- Massimo Granai
- Department of Medical Biotechnology, University of Siena, Siena, Italy
- University Hospital of Tübingen, Institute of Pathology, Tübingen, Germany
| | - Lucia Mundo
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Ayse U. Akarca
- Department of Pathology, University College London, London, UK
| | | | - Hasan Rizvi
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Virginia Mancini
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Noel Onyango
- Department of Clinical Medicine and Therapeutics, University of Nairobi, Nairobi, Kenya
| | - Joshua Nyagol
- Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | | | - Ibrahim Maha
- South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Sandra Margielewska
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK and Durham University, Durham, UK
| | - Wenbin Wi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK and Durham University, Durham, UK
| | - Michele Bibas
- Clinical Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.C.S, Rome, Italy
| | - Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine Bologna University Medical School, S. Orsola Malpighi Hospital, Bologna and Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | | | - Falko Fend
- University Hospital of Tübingen, Institute of Pathology, Tübingen, Germany
| | - Stefano Lazzi
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Lorenzo Leoncini
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Teresa Marafioti
- Department of Pathology, University College London, London, UK
- Department of Cellular Pathology, University College Hospital, London, London UK
| |
Collapse
|
39
|
Sadeghi L, Arvidsson G, Merrien M, Wasik AM, Görgens A, Smith CE, Sander B, P. Wright A. Differential B-Cell Receptor Signaling Requirement for Adhesion of Mantle Cell Lymphoma Cells to Stromal Cells. Cancers (Basel) 2020; 12:cancers12051143. [PMID: 32370190 PMCID: PMC7281289 DOI: 10.3390/cancers12051143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
Interactions between lymphoma cells and stromal cells play a key role in promoting tumor survival and development of drug resistance. We identified differences in key signaling pathways between the JeKo-1 and REC-1 mantle cell lymphoma (MCL) cell lines, displaying different patterns of stromal cell adhesion and chemotaxis towards stroma-conditioned medium. The identified adhesion-regulated genes reciprocated important aspects of microenvironment-mediated gene modulation in MCL patients. Five-hundred and ninety genes were differently regulated between the cell lines upon adhesion to stromal cells, while 32 genes were similarly regulated in both cell lines. Regulation of B-cell Receptor (BCR) signature genes in adherent cells was specific for JeKo-1. Inhibition of BCR using siRNA or clinically approved inhibitors, Ibrutinib and Acalabrutinib, decreased adhesion of JeKo-1, but not REC-1 cells. Cell surface levels of chemokine receptor CXCR4 were higher in JeKo-1, facilitating migration and adhesion of JeKo-1 but not REC-1 cells. Surface levels of ICAM1 adhesion protein differ for REC-1 and JeKo-1. While ICAM1 played a positive role in adherence of both cell lines to stromal cells, S1PR1 had an inhibitory effect. Our results provide a model framework for further investigation of mechanistic differences in patient-response to new pathway-specific drugs.
Collapse
Affiliation(s)
- Laia Sadeghi
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Gustav Arvidsson
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Magali Merrien
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - Agata M. Wasik
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg, 45 147 Essen, Germany
| | - C.I. Edvard Smith
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Birgitta Sander
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - Anthony P. Wright
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
- Correspondence:
| |
Collapse
|
40
|
Abdi J, Rastgoo N, Chen Y, Chen GA, Chang H. Ectopic expression of BIRC5-targeting miR-101-3p overcomes bone marrow stroma-mediated drug resistance in multiple myeloma cells. BMC Cancer 2019; 19:975. [PMID: 31638931 PMCID: PMC6805455 DOI: 10.1186/s12885-019-6151-x] [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: 10/27/2018] [Accepted: 09/11/2019] [Indexed: 12/27/2022] Open
Abstract
Background Multiple myeloma (MM) cells gain protection against drugs through interaction with bone marrow stromal cells (BMSCs). This form of resistance largely accounts for resistance to therapy in MM patients which warrants further exploration to identify more potential therapeutic targets. Methods We performed miRNA/mRNA qPCR arrays and western blotting to analyze transcriptional and translational changes in MM cells co-cultured with BMSCs. Drug cytotoxicity and apoptosis in MMGFP-BMSC co-cultures were measured using fluorescence plate reader and flowcytometry, respectively. miRNA was overexpressed in MM cell lines using Lentiviral transduction, miRNA-3’UTR binding was examined using luciferase assay. Results We found that BMSCs downregulated miR-101-3p and upregulated survivin (BIRC5) in MM cells. Survivin was downregulated by miR-101-3p overexpression and found to be a direct target of miR-101-3p using 3’UTR luciferase assay. Overexpression of survivin increased viability of MM cells in the presence of anti-myeloma drugs, and miR-101-3p inhibition by anti-miR against miR-101-3p upregulated survivin. Furthermore, overexpression of miR-101-3p or silencing of survivin triggered apoptosis in MM cells and sensitized them to anti-myeloma drugs in the presence of BMSCs overcoming the stroma-induced drug resistance. Conclusions Our study demonstrates that BMSC-induced resistance to drugs is associated with survivin upregulation which is a direct target of miR-101-3p. This study also identifies miR-101-3p-survivin interaction as a druggable target involved in stroma-mediated drug resistance in MM and suggests it for developing more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Jahangir Abdi
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Nasrin Rastgoo
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Yan Chen
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada
| | - Guo An Chen
- Department of Hematology/Oncology, First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Hong Chang
- Dept. of Laboratory Hematology, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, Ontario, M5G 2C4, Canada. .,Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
41
|
Li J, Tian T, Zhou X. The role of exosomal shuttle RNA (esRNA) in lymphoma. Crit Rev Oncol Hematol 2019; 137:27-34. [DOI: 10.1016/j.critrevonc.2019.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/15/2019] [Accepted: 01/21/2019] [Indexed: 12/24/2022] Open
|
42
|
Huang Q, Liu F, Shen J. The significance of chemokines in diffuse large B-cell lymphoma: a systematic review and future insights. Future Oncol 2019; 15:1385-1395. [PMID: 30880459 DOI: 10.2217/fon-2018-0514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the progress made in molecular and clinical research, patients with diffuse large B-cell lymphoma (DLBCL) still have a bad prognosis. Recently, chemokines/chemokine receptors have become the subject of interest in relation to DLBCL. Studies have demonstrated the important role of chemokines/chemokine receptors in the communication between DLBCL cells and tumor microenvironment. Studies have also reported the ability of chemokines/chemokine receptors in promoting the proliferation and invasion of DLBCL cells. Here, we summarize the data on mechanisms of DLBCL supporting the involvement of chemokine/chemokine receptor changes. We focus on the available evidence regarding chemokines/chemokine receptors as biomarkers and therapeutic targets for DLBCL.
Collapse
Affiliation(s)
- Qian Huang
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Feifei Liu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Jianzhen Shen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
43
|
Lad D, Huang Q, Hoeppli R, Garcia R, Xu L, Levings M, Song K, Broady R. Evaluating the role of Tregs in the progression of multiple myeloma. Leuk Lymphoma 2019; 60:2134-2142. [PMID: 30773086 DOI: 10.1080/10428194.2019.1579324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of regulatory T-cells (Treg) and Th17 cells in the progression of multiple myeloma has been unclear. There are conflicting reports of the Treg and Th17 frequency being increased, decreased, and unchanged as compared with controls. In this study, we sought to characterize the T-cell subsets including Treg function in both blood and marrow compartments of monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma (MM). The Treg/Th17 ratio is skewed toward the suppressive phenotype in MGUS and MM. There are more activated and memory Tregs in the myeloma marrow. Although the myeloma Tregs are functional, they are less suppressive than Tregs in chronic lymphocytic leukemia where they drive disease progression. None of the T-cell subsets were found to have a clinical correlation with time to progression in MGUS or progression-free survival in myeloma. Tregs are important but unlikely major players in the progression of MGUS to MM.
Collapse
Affiliation(s)
- Deepesh Lad
- Clinical Hematology, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Qing Huang
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Romy Hoeppli
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Rosa Garcia
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Lixin Xu
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Megan Levings
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kevin Song
- Leukemia/BMT Program of BC, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - Raewyn Broady
- Leukemia/BMT Program of BC, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
44
|
Zhong W, Zhu Z, Xu X, Zhang H, Xiong H, Li Q, Wei Y. Human bone marrow-derived mesenchymal stem cells promote the growth and drug-resistance of diffuse large B-cell lymphoma by secreting IL-6 and elevating IL-17A levels. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:73. [PMID: 30755239 PMCID: PMC6373150 DOI: 10.1186/s13046-019-1081-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 02/06/2019] [Indexed: 02/08/2023]
Abstract
Background The drug-resistance and relapse of diffuse large B-cell lymphoma (DLBCL), which are related to mesenchymal stem cells (MSCs), have become increasingly common. However, the underlying mechanisms remain elusive. Methods CCK 8 assay, colony formation assay, and xenograft mouse model were used to investigate the effects of hBMSCs on DLBCL growth. Immunohistochemistry, qRT-PCR, and ELISA were used to study the expressions of IL-6 and IL-17A. Flow cytometry was used to analyze Th17 cells and Treg cells expressions. Western blot analysis, microarray analysis, and bioinformatics analysis were used to analyze the pathways of IL-6 or IL-17A mediated DLBCL growth. Results HBMSCs promoted DLBCL growth by secreting IL-6 in vitro and in vivo and simultaneously upregulating IL-17A in vitro. IL-6 and IL-17A synergistically promoted the growth and drug-resistance of DLBCL cells by protecting them from spontaneous or drug-induced apoptosis in vitro. IL-6 or IL-17A activated the JAK2/STAT3 pathway or upregulated cyclin D2 via activation of PI3K/Akt signaling in vitro, respectively. Conclusions The present results indicated that hBMSCs might have a “dual effect” on promoting DLBCL progression and drug-resistance by secreting IL-6 and upregulating IL-17A. IL-6, IL-17A, p-STAT3, p-Akt or cyclin D2 may be potential molecular targets for overcoming drug-resistance in patients with relapsed or refractory DLBCL.
Collapse
Affiliation(s)
- Weijie Zhong
- Department of Geriatrics, Hematology & Oncology ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Zhigang Zhu
- Department of Geriatrics, Hematology & Oncology ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Xin Xu
- Department of Geriatrics, Hematology & Oncology ward, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jinan, 272067, Shandong, China
| | - Huabao Xiong
- Immunology Institute, Mount Sinai School of Medicine, NY10029, New York, 5674, USA
| | - Qingshan Li
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Panfu Rd No.1, Yuexiu District, Guangzhou, 510180, Guangdong, China.
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Panfu Rd No.1, Yuexiu District, Guangzhou, 510180, Guangdong, China.
| |
Collapse
|
45
|
Nooka AK, Wang ML, Yee AJ, Kaufman JL, Bae J, Peterkin D, Richardson PG, Raje NS. Assessment of Safety and Immunogenicity of PVX-410 Vaccine With or Without Lenalidomide in Patients With Smoldering Multiple Myeloma: A Nonrandomized Clinical Trial. JAMA Oncol 2018; 4:e183267. [PMID: 30128502 DOI: 10.1001/jamaoncol.2018.3267] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Increasing evidence suggests the significance of the role of the immune system in the progression of smoldering multiple myeloma (SMM) to symptomatic multiple myeloma (MM). Boosting the immune system via vaccination in the earlier, asymptomatic SMM stage may provide a novel strategy to prevent or slow progression to active MM. Objective To determine the safety, tolerability, immunogenicity, and anti-MM activity of the PVX-410 multipeptide vaccine with or without lenalidomide. Design, Setting, and Participants This 3-cohort phase 1/2a multicenter dose-escalation study accrued 22 adults (≥18 years) with SMM with normal organ/marrow function who were human leukocyte antigen A2-positive and at moderate or high risk of progression to MM. Interventions Patients received 6 doses of PVX-410 emulsified in Montanide ISA 720 VG, 0.4 mg total (0.1 mg/peptide) (n = 3) or 0.8 mg total (0.2 mg/peptide) (n = 9), biweekly via subcutaneous injection. In the combination cohort (n = 10), patients also received three 21-day cycles of lenalidomide, 25 mg, orally daily every 28 days. All patients received 0.5 mL (1 mg) poly-ICLC (2 mg/mL) via intramuscular injection with each PVX-410 dose. Main Outcomes and Measures Adverse events (AEs) were evaluated using the Common Terminology Criteria for Adverse Events, version 4.03. PVX-410-specific T lymphocytes by flow cytometry to assess tetramer and interferon (IFN)-γ response. Disease response was assessed by investigators using the International Myeloma Working Group (IMWG) and modified European Group for Bone Marrow Transplantation (EBMT) criteria. Results Overall, 14 (64%) patients were men and the median age at enrollment was 56 years in the monotherapy and 57 years in the combination cohorts (overall range, 39-82 years). Six of 12 patients in the monotherapy and 9 of 10 in the combination cohorts were at moderate risk. The PVX-410 vaccine was well tolerated. The most common AEs were mild-to-moderate injection site reactions and constitutional symptoms. Of note, PVX-410 was immunogenic as monotherapy (10 of 11 patients) and in combination with lenalidomide (9 of 9 patients), as demonstrated by an increase in percentage of tetramer-positive cells and IFN-γ cells in the CD3+CD8+ cell population. The combination resulted in greater mean fold increases in proportions of CD3+CD8+ T cells that were tetramer-positive and IFN-γ-positive, statistically significant for IFN-γ-positive cells after 2 and 4 vaccinations. An increase and persistence of vaccine-specific effector memory cells was noted. In total, 7 of 12 patients in the PVX-410-alone cohort had stable disease with 2 of 3 (low-dose cohort) and 1 of 9 of the target-dose cohort progressing (median TTP, 36 weeks), whereas 5 of 12 patients in the combination cohort showed, clinical response, with 1 patient progressing (median TTP not reached). Conclusions and Relevance Overall, these results suggest that the vaccine is safe and immunogenic in this patient population and support continued study of PVX-410 in SMM. Trial Registration ClinicalTrials.gov identifier: NCT01718899.
Collapse
Affiliation(s)
- Ajay K Nooka
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | | | - Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Jonathan L Kaufman
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jooeun Bae
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Noopur S Raje
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
46
|
Gordiienko I, Shlapatska L, Kovalevska L, Sidorenko SP. SLAMF1/CD150 in hematologic malignancies: Silent marker or active player? Clin Immunol 2018; 204:14-22. [PMID: 30616923 DOI: 10.1016/j.clim.2018.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
SLAMF1/CD150 receptor is a founder of signaling lymphocyte activation molecule (SLAM) family of cell-surface receptors. It is widely expressed on cells within hematopoietic system. In hematologic malignancies CD150 cell surface expression is restricted to cutaneous T-cell lymphomas, few types of B-cell non-Hodgkin's lymphoma, near half of cases of chronic lymphocytic leukemia, Hodgkin's lymphoma, and multiple myeloma. Differential expression among various types of hematological malignancies allows considering CD150 as diagnostical and potential prognostic marker. Moreover, CD150 may be a target for antibody-based or measles virus oncolytic therapy. Due to CD150 signaling properties it is involved in regulation of malignant cell fate decision and tumor microenvironment in Hodgkin's lymphoma and chronic lymphocytic leukemia. This review summarizes evidence for the important role of CD150 in pathogenesis of hematologic malignancies.
Collapse
Affiliation(s)
- Inna Gordiienko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine.
| | - Larysa Shlapatska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Larysa Kovalevska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Svetlana P Sidorenko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
47
|
Leivonen SK, Pollari M, Brück O, Pellinen T, Autio M, Karjalainen-Lindsberg ML, Mannisto S, Kellokumpu-Lehtinen PL, Kallioniemi O, Mustjoki S, Leppä S. T-cell inflamed tumor microenvironment predicts favorable prognosis in primary testicular lymphoma. Haematologica 2018; 104:338-346. [PMID: 30237271 PMCID: PMC6355505 DOI: 10.3324/haematol.2018.200105] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/19/2018] [Indexed: 12/18/2022] Open
Abstract
Primary testicular lymphoma is a rare lymphoid malignancy, most often, histologically, representing diffuse large B-cell lymphoma. The tumor microenvironment and limited immune surveillance have a major impact on diffuse large B-cell lymphoma pathogenesis and survival, but the impact on primary testicular lymphoma is unknown. Here, the purpose of the study was to characterize the tumor microenvironment in primary testicular lymphoma, and associate the findings with outcome. We profiled the expression of 730 immune response genes in 60 primary testicular lymphomas utilizing the Nanostring platform, and used multiplex immunohistochemistry to characterize the immune cell phenotypes in the tumor tissue. We identified a gene signature enriched for T-lymphocyte markers differentially expressed between the patients. Low expression of the signature predicted poor outcome independently of the International Prognostic Index (progression-free survival: HR=2.810, 95%CI: 1.228-6.431, P=0.014; overall survival: HR=3.267, 95%CI: 1.406-7.590, P=0.006). The T-lymphocyte signature was associated with outcome also in an independent diffuse large B-cell lymphoma cohort (n=96). Multiplex immunohistochemistry revealed that poor survival of primary testicular lymphoma patients correlated with low percentage of CD3+CD4+ and CD3+CD8+ tumor-infiltrating lymphocytes (P<0.001). Importantly, patients with a high T-cell inflamed tumor microenvironment had a better response to rituximab-based immunochemotherapy, as compared to other patients. Furthermore, loss of membrane-associated human-leukocyte antigen complexes was frequent and correlated with low T-cell infiltration. Our results demonstrate that a T-cell inflamed tumor microenvironment associates with favorable survival in primary testicular lymphoma. This further highlights the importance of immune escape as a mechanism of treatment failure.
Collapse
Affiliation(s)
- Suvi-Katri Leivonen
- Research Program Unit, Medical Faculty, University of Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Finland
| | - Marjukka Pollari
- Research Program Unit, Medical Faculty, University of Helsinki, Finland.,Department of Oncology, Tampere University Hospital, Finland
| | - Oscar Brück
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Matias Autio
- Research Program Unit, Medical Faculty, University of Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Finland
| | | | - Susanna Mannisto
- Research Program Unit, Medical Faculty, University of Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Finland
| | - Pirkko-Liisa Kellokumpu-Lehtinen
- Department of Oncology, Tampere University Hospital, Finland.,University of Tampere, Faculty of Medicine and Life Sciences, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland.,Science for Life Laboratory, Karolinska Institutet, Department of Oncology and Pathology, Solna, Sweden
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki, Finland.,Department of Hematology, Comprehensive Cancer Center, Helsinki University Hospital, Finland
| | - Sirpa Leppä
- Research Program Unit, Medical Faculty, University of Helsinki, Finland .,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Finland
| |
Collapse
|
48
|
Recasens-Zorzo C, Cardesa-Salzmann T, Petazzi P, Ros-Blanco L, Esteve-Arenys A, Clot G, Guerrero-Hernández M, Rodríguez V, Soldini D, Valera A, Moros A, Climent F, González-Barca E, Mercadal S, Arenillas L, Calvo X, Mate JL, Gutiérrez-García G, Casanova I, Mangues R, Sanjuan-Pla A, Bueno C, Menéndez P, Martínez A, Colomer D, Tejedor RE, Teixidó J, Campo E, López-Guillermo A, Borrell JI, Colomo L, Pérez-Galán P, Roué G. Pharmacological modulation of CXCR4 cooperates with BET bromodomain inhibition in diffuse large B-cell lymphoma. Haematologica 2018; 104:778-788. [PMID: 29954928 PMCID: PMC6442946 DOI: 10.3324/haematol.2017.180505] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 06/25/2018] [Indexed: 01/15/2023] Open
Abstract
Constitutive activation of the chemokine receptor CXCR4 has been associated with tumor progression, invasion, and chemotherapy resistance in different cancer subtypes. Although the CXCR4 pathway has recently been suggested as an adverse prognostic marker in diffuse large B-cell lymphoma, its biological relevance in this disease remains underexplored. In a homogeneous set of 52 biopsies from patients, an antibody-based cytokine array showed that tissue levels of CXCL12 correlated with high microvessel density and bone marrow involvement at diagnosis, supporting a role for the CXCL12-CXCR4 axis in disease progression. We then identified the tetra-amine IQS-01.01RS as a potent inverse agonist of the receptor, preventing CXCL12-mediated chemotaxis and triggering apoptosis in a panel of 18 cell lines and primary cultures, with superior mobilizing properties in vivo than those of the standard agent. IQS-01.01RS activity was associated with downregulation of p-AKT, p-ERK1/2 and destabilization of MYC, allowing a synergistic interaction with the bromodomain and extra-terminal domain inhibitor, CPI203. In a xenotransplant model of diffuse large B-cell lymphoma, the combination of IQS-01.01RS and CPI203 decreased tumor burden through MYC and p-AKT downregulation, and enhanced the induction of apoptosis. Thus, our results point out an emerging role of CXCL12-CXCR4 in the pathogenesis of diffuse large B-cell lymphoma and support the simultaneous targeting of CXCR4 and bromodomain proteins as a promising, rationale-based strategy for the treatment of this disease.
Collapse
Affiliation(s)
- Clara Recasens-Zorzo
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | | | - Paolo Petazzi
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona
| | - Laia Ros-Blanco
- Grup d'Enginyeria Molecular, IQS School of Engineering, Universitat Ramon Llull, Barcelona
| | - Anna Esteve-Arenys
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Guillem Clot
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Martina Guerrero-Hernández
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Vanina Rodríguez
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Davide Soldini
- Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona
| | - Alexandra Valera
- Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona
| | - Alexandra Moros
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Fina Climent
- Pathology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat
| | - Eva González-Barca
- Institut Catalá d'Oncología, Hospital Duran I Reynals, L'Hospitalet de Llobregat
| | - Santiago Mercadal
- Institut Catalá d'Oncología, Hospital Duran I Reynals, L'Hospitalet de Llobregat
| | | | - Xavier Calvo
- Pathology Department, IMIM, Hospital del Mar, Barcelona
| | - José Luís Mate
- Pathology Department, Hospital Universitari Germans Trias i Pujol, Badalona
| | | | - Isolda Casanova
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona.,Grup d'Oncogènesi i Antitumorals, lnstitut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau) and Centro de Investigación Biomédica en Red CIBER-BBN, Barcelona
| | - Ramón Mangues
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona.,Grup d'Oncogènesi i Antitumorals, lnstitut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau) and Centro de Investigación Biomédica en Red CIBER-BBN, Barcelona
| | | | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona.,Institucio Catalana de Recerca I Estudis Avançats (ICREA), CIBERONC, Barcelona
| | - Antonio Martínez
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona.,Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona
| | - Dolors Colomer
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona.,Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona
| | - Roger Estrada Tejedor
- Grup d'Enginyeria Molecular, IQS School of Engineering, Universitat Ramon Llull, Barcelona
| | - Jordi Teixidó
- Grup d'Enginyeria Molecular, IQS School of Engineering, Universitat Ramon Llull, Barcelona
| | - Elias Campo
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona.,Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona
| | - Armando López-Guillermo
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona.,Department of Hematology, Hospital Clinic, Barcelona
| | - José Ignacio Borrell
- Grup d'Enginyeria Molecular, IQS School of Engineering, Universitat Ramon Llull, Barcelona
| | - Luis Colomo
- Hematopathology Unit, Department of Pathology, Hospital Clinic, Barcelona.,Pathology Department, IMIM, Hospital del Mar, Barcelona
| | - Patricia Pérez-Galán
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona
| | - Gaël Roué
- Division of Hemato-Oncology, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERONC, Barcelona .,Laboratory of Experimental Hematology, Department of Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
49
|
Trnĕný M, Verhoef G, Dyer MJ, Ben Yehuda D, Patti C, Canales M, Lopez A, Awan FT, Montgomery PG, Janikova A, Barbui AM, Sulek K, Terol MJ, Radford J, Guidetti A, Di Nicola M, Siraudin L, Hatteville L, Schwab S, Oprea C, Gianni AM. A phase II multicenter study of the anti-CD19 antibody drug conjugate coltuximab ravtansine (SAR3419) in patients with relapsed or refractory diffuse large B-cell lymphoma previously treated with rituximab-based immunotherapy. Haematologica 2018; 103:1351-1358. [PMID: 29748443 PMCID: PMC6068033 DOI: 10.3324/haematol.2017.168401] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/03/2018] [Indexed: 12/15/2022] Open
Abstract
This phase II, single-arm, multicenter study examined the efficacy and safety of coltuximab ravtansine (an anti-CD19 antibody drug conjugate) in 61 patients with histologically documented (de novo or transformed) relapsed or refractory diffuse large B-cell lymphoma who had previously received rituximab-containing immuno-chemotherapy. Patients had received a median of 2.0 (range 0-9) prior treatment regimens for diffuse large B-cell lymphoma and almost half (45.9%) had bulky disease (≥1 lesion >5 cm) at trial entry. Patients received coltuximab ravtansine (55 mg/m2) in 4 weekly and 4 biweekly administrations until disease progression or unacceptable toxicity. Forty-one patients were eligible for inclusion in the per protocol population. Overall response rate (International Working Group criteria) in the per protocol population, the primary end point, was 18/41 [43.9%; 90% confidence interval (CI:) 30.6-57.9%]. Median duration of response, progression-free survival, and overall survival (all treated patients) were 4.7 (range 0.0-8.8) months, 4.4 (90%CI: 3.02-5.78) months, and 9.2 (90%CI: 6.57-12.09) months, respectively. Common non-hematologic adverse events included asthenia/fatigue (30%), nausea (23%), and diarrhea (20%). Grade 3-4 adverse events were reported in 23 patients (38%), the most frequent being hepatotoxicity (3%) and abdominal pain (3%). Eye disorders occurred in 15 patients (25%); all were grade 1-2 and none required a dose modification. Coltuximab ravtansine monotherapy was well tolerated and resulted in moderate clinical responses in pre-treated patients with relapsed/refractory diffuse large B-cell lymphoma. (Registered at: clinicaltrials.gov identifier: 01472887)
Collapse
Affiliation(s)
- Marek Trnĕný
- Charles University, General Hospital, Prague, Czech Republic
| | - Gregor Verhoef
- Department of Hematology, University Hospital, Leuven, Belgium
| | - Martin Js Dyer
- Ernest and Helen Scott Haematological Research Institute, University of Leicester, UK
| | | | | | | | - Andrés Lopez
- Vall d'Hebron Research Institute, Barcelona, Spain
| | | | | | - Andrea Janikova
- Department of Hematology and Oncology, University Hospital Brno, Czech Republic
| | - Anna M Barbui
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Maria J Terol
- Hospital Clínico Universitario de Valencia, Health Research Institute INCLIVA, Spain
| | - John Radford
- University of Manchester and The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, UK
| | - Anna Guidetti
- Fondazione Istituto Nazionale Tumori, Milan, Italy.,University of Milan, Italy
| | | | | | | | | | | | - Alessandro M Gianni
- Fondazione Istituto Nazionale Tumori, Milan, Italy.,University of Milan, Italy
| |
Collapse
|
50
|
Pham LV, Pogue E, Ford RJ. The Role of Macrophage/B-Cell Interactions in the Pathophysiology of B-Cell Lymphomas. Front Oncol 2018; 8:147. [PMID: 29868471 PMCID: PMC5951963 DOI: 10.3389/fonc.2018.00147] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022] Open
Abstract
Macrophages (MPs) are heterogeneous, multifunctional, myeloid-derived leukocytes that are part of the innate immune system, playing wide-ranging critical roles in basic biological activities, including maintenance of tissue homeostasis involving clearance of microbial pathogens. Tumor-associated MPs (TAMs) are MPs with defined specific M2 phenotypes now known to play central roles in the pathophysiology of a wide spectrum of malignant neoplasms. Also, TAMs are often intrinsic cellular components of the essential tumor microenvironment (TME). In concert with lymphoid-lineage B and T cells at various developmental stages, TAMs can mediate enhanced tumor progression, often leading to poor clinical prognosis, at least partly through secretion of chemokines, cytokines, and various active proteases shown to stimulate tumor growth, angiogenesis, metastasis, and immunosuppression. Researchers recently showed that TAMs express certain key checkpoint-associated proteins [e.g., programmed cell death protein 1 (PD-1), programmed cell death-ligand 1 (PD-L1)] that appear to be involved in T-cell activation and that these proteins are targets of other specific checkpoint-blocking immunotherapies (anti-PD-1/PD-L1) currently part of new therapeutic paradigms for chemotherapy-resistant neoplasms. Although much is known about the wide spectrum and flexibility of MPs under many normal and neoplastic conditions, relatively little is known about the increasingly important interactions between MPs and B-lymphoid cells, particularly in the TME in patients with aggressive B-cell non-Hodgkin lymphoma (NHL-B). Normal and neoplastic lymphoid and myeloid cell/MP lineages appear to share many primitive cellular characteristics as well as transcriptional factor interactions in human and animal ontogenic studies. Such cells are capable of ectopic transcription factor-induced lineage reprogramming or transdifferentiation from early myeloid/monocytic lineages to later induce B-cell lymphomagenesis in experimental in vivo murine systems. Close cellular interactions between endogenous clonal neoplastic B cells and related aberrant myeloid precursor cells/MPs appear to be important interactive components of aggressive NHL-B that we discuss herein in the larger context of the putative role of B-cell/MP cellular lineage interactions involved in NHL-B pathophysiology during ensuing lymphoma development.
Collapse
Affiliation(s)
- Lan V Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth Pogue
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Richard J Ford
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|