1
|
Shi H, Sun X, Wu Y, Cui Q, Sun S, Ji N, Liu Y. Targeting the tumor microenvironment in primary central nervous system lymphoma: Implications for prognosis. J Clin Neurosci 2024; 124:36-46. [PMID: 38642434 DOI: 10.1016/j.jocn.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/06/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma, and there is limited research on its tumor microenvironment (TME). Nevertheless, more and more studies have evidence that TME has essential effects on tumor cell proliferation, immune escape, and drug resistance. Thus, it is critical to elucidate the role of TME in PCNSL. The understanding of the PCNSL TME is gradually unfolding, including factors that distinguish it from systemic diffuse large B-cell lymphoma (DLBCL). The TME in PCNSL exhibits both transcriptional and spatial intratumor heterogeneity. Cellular interactions between tumor cells and stroma cells reveal immune evasion signaling. The comparative analysis between PCNSL and DLBCL suggests that PCNSL is more likely to be an immunologically deficient tumor. In PCNSL, T cell exhaustion and downregulation of macrophage immune function are accompanied by suppressive microenvironmental factors such as M2 polarized macrophages, endothelin B receptor, HLA depletion, PD-L1, and TIM-3. MMP-9, Integrin-β1, and ICAM-1/LFA-1 play crucial roles in transendothelial migration towards the CNS, while CXCL13/CXCR5, CD44, MAG, and IL-8 are essential for brain parenchymal invasion. Further, macrophages, YKL-40, CD31, CD105, PD-1/PD-L1 axis, osteopontin, galectin-3, aggregative perivascular tumor cells, and HLA deletion may contribute to poor outcomes in patients with PCNSL. This article reviews the effect of various components of TME on the progression and prognosis of PCNSL patients to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Han Shi
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Xuefei Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuchen Wu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Qu Cui
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Shengjun Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Nan Ji
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuanbo Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China.
| |
Collapse
|
2
|
Zheng X, Wang C, Chen F, Li S, Zhang H, Dong G, Yang S, Kang X, Kang Z, Han C, Yin S, Li W. Zanubrutinib delays selinexor resistance evolution in biopsy sample-derived primary central nervous system lymphoma models. iScience 2024; 27:109799. [PMID: 38726367 PMCID: PMC11079464 DOI: 10.1016/j.isci.2024.109799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/18/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and aggressive lymphoma of the brain with poor prognosis. The scarcity of cell lines established using PCNSL makes it difficult to conduct preclinical studies on new drugs. We aimed to explore the effect of selinexor combined with zanubrutinib in PCNSL using established PCNSL cells and an orthotopic PCNSL model. Primary PCNSL cells were successfully cultured. Selinexor inhibited proliferation, induced G1 phase arrest, and promoted apoptosis, however, induced drug resistance in PCNSL. Selinexor combined with zanubrutinib had a synergistic effect on PCNSL and prevented the onset of selinexor resistance in PCNSL by inhibiting AKT signaling. Moreover, selinexor combined with zanubrutinib notably slowed tumor growth and prolonged survival compared to that of the control. Overall, the addition of zanubrutinib to selinexor monotreatment had a synergistic effect in vitro and prolonged survival in vivo.
Collapse
Affiliation(s)
- Xiaohong Zheng
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenglan Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hua Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shoubo Yang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xun Kang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunlei Han
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuo Yin
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Wu D, Liu D, Tateishi K, Qi F, Yang F, Ke C, You H. Understanding the molecular pathogenesis of primary central nervous system lymphoma by experimental animal models. J Cell Physiol 2023; 238:2191-2205. [PMID: 37642377 DOI: 10.1002/jcp.31107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and invasive diffuse large B cell lymphoma confined in central nervous system (CNS). The effort to press forward the translational progress has been frustrated by the insufficient understanding of immunophenotype of CNS and tumor genetic alterations of PCNSL, and the lack of validated diagnostic biomarkers. Researchers now have a variety of PCNSL animal models at their disposal that resemble the morphology and immunophenotype of PCNSL, however, a careful and detailed re-examination of these animal models is needed to clarify the differences in genetic alterations, migration capability, and immune status. In this review, we present the knowledge about the phenotypic and genotypic features of PCNSL tumor cells, and compile the preclinical animal models of PCNSL with regard to various injection sites, cell origins, recipient animals, and immune status, and elaborate on the tropism and migration of tumor cells and novel therapeutic strategies for PCNSL. We envisage that the selection of suitable animal models will serve as a well-defined preclinical system to understand the molecular pathogenesis of PCNSL, thereby galvanizing the development of novel and potent therapeutic approaches.
Collapse
Affiliation(s)
- Di Wu
- Department of Basic Medicine, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Dahai Liu
- Department of Basic Medicine, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Kensuke Tateishi
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Fei Qi
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Fang Yang
- Department of Basic Medicine, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Chao Ke
- State Key Laboratory of Oncology in South China, Department of Neurosurgery and Neuro-oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hua You
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Montesinos-Rongen M, Sanchez-Ruiz M, Siebert S, Winter C, Siebert R, Brunn A, Deckert M. AMD3100-mediated CXCR4 inhibition impairs development of primary lymphoma of the central nervous system. THE AMERICAN JOURNAL OF PATHOLOGY 2023:S0002-9440(23)00163-3. [PMID: 37196929 DOI: 10.1016/j.ajpath.2023.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/23/2023] [Accepted: 04/26/2023] [Indexed: 05/19/2023]
Abstract
A hallmark of primary lymphoma of the central nervous system (PCNSL, CNS) is the strong CXCR4 expression of the tumor cells, the function of which is still unknown. In vitro treatment of BAL17CNS lymphoma cells by AMD3100 which inhibits CXCR4-CXCL12 interactions resulted in the significantly differential expression of 273 genes encoding proteins involved in cell motility, cell-cell signaling and interaction, hematological system development and function, and immunological disease. Among the genes downregulated was the one encoding CD200, a regulator of CNS immunological activity. These data directly translated into the in vivo situation; BAL17CNS CD200 expression was downregulated by 89% (3% vs. 28% CD200+ lymphoma cells) in AMD3100-treated vs. untreated mice with BAL17CNS-induced PCNSL. Reduced lymphoma cell CD200 expression may contribute to the markedly increased microglial activation in AMD3100-treated mice. AMD3100 also maintained the structural integrity of blood-brain barrier tight junctions and the outer basal lamina of cerebral blood vessels. Subsequently, lymphoma cell invasion of the brain parenchyma was impaired and maximal parenchymal tumor size was significantly reduced by 82% in the induction phase. Thus, AMD3100 qualified as potentially attractive candidate to be included into the therapeutic concept of PCNSL. Beyond therapy, CXCR4-induced suppression of microglial activity is of general neuroimmunological interest and identifies CD200 expressed by the lymphoma cells as a novel mechanism of immune escape in PCNSL.
Collapse
Affiliation(s)
- Manuel Montesinos-Rongen
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Monica Sanchez-Ruiz
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Susann Siebert
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Claudia Winter
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Anna Brunn
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; present address: Institute of Neuropathology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martina Deckert
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; present address: Institute of Neuropathology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
5
|
Montesinos-Rongen M, Sanchez-Ruiz M, Siebert S, Winter C, Siebert R, Brunn A, Deckert M. Expression of Cas9 in a Syngeneic Model of Primary Central Nervous System Lymphoma Induces Intracerebral NK and CD8 T Cell-Mediated Lymphoma Cell Lysis Via Perforin. CRISPR J 2022; 5:726-739. [PMID: 36260299 DOI: 10.1089/crispr.2022.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The development of clustered regulatory interspaced short palindromic repeats/CRISPR associated protein 9 (CRISPR-Cas9)-mediated gene modification has opened an exciting avenue of targeting genes to study the pathogenesis of diseases and to develop novel therapeutic concepts. However, as the effector protein Cas9 is of bacterial origin, unwanted side effects due to a host immune response against Cas9 need to be considered. Here, we used the syngeneic model of BAL17CNS-induced primary lymphoma of the central nervous system (PCNSL, CNS) in BALB/c mice to address this issue. Surprisingly, stable expression of Cas9 in BAL17CNS (BAL17CNS/Cas9) cells rendered them unable to establish PCNSL on intracerebral transplantation. Instead, they induced a prominent intracerebral immune response mediated by CD8 T cells, which lysed BAL17CNS/Cas9 cells via perforin. In addition, B cells contributed to the immune response as evidenced by serum anti-Cas9 antibodies in BALB/c mice as early as day 8 after transplantation of BAL17CNS/Cas9 cells. In athymic BALB/cnu/nu mice, NK cells mounted a vigorous intracerebral immune response with perforin-mediated destruction of BAL17CNS/Cas9 cells. Thus, in the CNS, perforin produced by NK and CD8 T cells was identified as a mediator of cytotoxicity against BAL17CNS/Cas9 cells. These observations should be taken into account when considering therapeutic CRISPR-Cas9-mediated tumor cell manipulation for PCNSL.
Collapse
Affiliation(s)
- Manuel Montesinos-Rongen
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Monica Sanchez-Ruiz
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Susann Siebert
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Claudia Winter
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Anna Brunn
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Martina Deckert
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany, and Ulm University and Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
6
|
Montesinos-Rongen M, Brunn A, Sanchez-Ruiz M, Küppers R, Siebert R, Deckert M. Impact of a Faulty Germinal Center Reaction on the Pathogenesis of Primary Diffuse Large B Cell Lymphoma of the Central Nervous System. Cancers (Basel) 2021; 13:cancers13246334. [PMID: 34944954 PMCID: PMC8699297 DOI: 10.3390/cancers13246334] [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: 11/30/2021] [Accepted: 12/13/2021] [Indexed: 01/12/2023] Open
Abstract
Simple Summary The pathogenetic mechanisms and peculiar tropism of primary CNS lymphoma (PCNSL) of the central nervous system (CNS) have been the subject of debate for decades. Hypothesis-driven targeted molecular studies have revealed that PCNSLs derived from self-/polyreactive B cells that have escaped developmental control mechanisms. The early acquisition of activating mutations targeting the B cell receptor pathway provides a survival advantage. The failure of the germinal center (GC) reaction and its checkpoints increases tumor B cell affinity for the CNS. During this faulty GC reaction, PCNSL tumor cells acquire further oncogenic alterations converging on the Toll-like receptor, B cell receptor, and NF-κB pathway. These activated pathways sustain proliferation. Concomitantly, cells become unable to complete terminal B cell differentiation, becoming trapped within the vicious cycle of the GC reaction as low-affinity IgM+ B cells related to memory cells. Abstract Primary lymphoma of the central nervous system (PCNSL, CNS) is a specific diffuse large B cell lymphoma (DLBCL) entity confined to the CNS. Key to its pathogenesis is a failure of B cell differentiation and a lack of appropriate control at differentiation stages before entrance and within the germinal center (GC). Self-/polyreactive B cells rescued from apoptosis by MYD88 and/or CD79B mutations accumulate a high load of somatic mutations in their rearranged immunoglobulin (IG) genes, with ongoing somatic hypermutation (SHM). Furthermore, the targeting of oncogenes by aberrant SHM (e.g., PIM1, PAX5, RHOH, MYC, BTG2, KLHL14, SUSD2), translocations of the IG and BCL6 genes, and genomic instability (e.g., gains of 18q21; losses of 9p21, 8q12, 6q21) occur in these cells in the course of their malignant transformation. Activated Toll-like receptor, B cell receptor (BCR), and NF-κB signaling pathways foster lymphoma cell proliferation. Hence, tumor cells are arrested in a late B cell differentiation stage, corresponding to late GC exit B cells, which are genetically related to IgM+ memory cells. Paradoxically, the GC reaction increases self-/polyreactivity, yielding increased tumor BCR reactivity for multiple CNS proteins, which likely contributes to CNS tropism of the lymphoma. The loss of MHC class I antigen expression supports tumor cell immune escape. Thus, specific and unique interactions of the tumor cells with resident CNS cells determine the hallmarks of PCNSL.
Collapse
Affiliation(s)
- Manuel Montesinos-Rongen
- Institute of Neuropathology, Faculty of Medicine, University Hospital Cologne, 50937 Cologne, Germany; (M.M.-R.); (A.B.); (M.S.-R.)
| | - Anna Brunn
- Institute of Neuropathology, Faculty of Medicine, University Hospital Cologne, 50937 Cologne, Germany; (M.M.-R.); (A.B.); (M.S.-R.)
| | - Monica Sanchez-Ruiz
- Institute of Neuropathology, Faculty of Medicine, University Hospital Cologne, 50937 Cologne, Germany; (M.M.-R.); (A.B.); (M.S.-R.)
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), Medical School, University of Duisburg-Essen, 45122 Essen, Germany;
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89081 Ulm, Germany;
| | - Martina Deckert
- Institute of Neuropathology, Faculty of Medicine, University Hospital Cologne, 50937 Cologne, Germany; (M.M.-R.); (A.B.); (M.S.-R.)
- Correspondence: ; Tel.: +49-221-478-5265; Fax: +49-221-478-3712
| |
Collapse
|
7
|
You H, Wei L, Kaminska B. Emerging insights into origin and pathobiology of primary central nervous system lymphoma. Cancer Lett 2021; 509:121-129. [PMID: 33766752 DOI: 10.1016/j.canlet.2021.02.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 01/03/2023]
Abstract
Primary central nervous system lymphoma (PCNSL) is an aggressive cancer typically confined to the brain, eyes, leptomeninges and spinal cord, without evidence of systemic involvement. PCNSL remains a challenge for scientists and clinicians due to insufficient biological knowledge, a lack of appropriate animal models and validated diagnostic biomarkers. We summarize recent findings on genomic, transcriptomic and epigenetic alterations identified in PCNSL. These findings help to define pathobiology of the disease and delineate defects in B cell differentiation. Evidence from genomic and transcriptomic studies helps to separate PCNSL from other hematological malignancies, improves diagnostics and reveals new therapeutic targets for treatment. Discovery of the CNS lymphatic system may be instrumental in better understanding the origin of the disease. We critically assess the attempts to model PCNSL in rodents, and conclude that there is a lack of a genetic/transgenic model that adequately mimics pathogenesis of the disease. Contribution of the tumor microenvironment in tumorigenesis and aggressiveness of PCNSL remains understudied. Assessing heterogeneity of immune infiltrates, cytokine profiling and molecular markers, may improve diagnostics and put forward new therapeutic strategies.
Collapse
Affiliation(s)
- Hua You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Li Wei
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Bozena Kaminska
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
8
|
You H, Baluszek S, Kaminska B. Immune Microenvironment of Brain Metastases-Are Microglia and Other Brain Macrophages Little Helpers? Front Immunol 2019; 10:1941. [PMID: 31481958 PMCID: PMC6710386 DOI: 10.3389/fimmu.2019.01941] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Abstract
Brain metastases are common intracranial neoplasms and their frequency increases with prolonged survival of cancer patients. New pharmaceuticals targeting oncogenic kinases and immune checkpoint inhibitors augment both overall and progression-free survival in patients with brain metastases, but are not fully successful in reducing metastatic burden and still a majority of oncologic patients die due to dissemination of the disease. Despite therapy advancements, median survival of patients with brain metastases is several months, although it may vary in different types or subtypes of cancer. Contribution of the innate immune system to cancer progression is well established. Tumor-associated macrophages (TAMs), instead of launching antitumor responses, promote extracellular matrix degradation, secrete immunosuppressive cytokines, promote neoangiogenesis and tumor growth. While their roles as pro-tumorigenic cells facilitating tissue remodeling, invasion and metastasis is well documented, much less is known about the immune microenvironment of brain metastases and roles of specific immune cells in those processes. The central nervous system (CNS) is armed in resident myeloid cells: microglia and perivascular macrophages which colonize CNS in early development and maintain homeostasis in brain parenchyma and at brain-blood vessels interfaces. In this study we discuss available data on the immune composition of most common brain metastases, focusing on interactions between metastatic cancer cells and microglia, perivascular and meningeal macrophages. Cancer cells ‘highjack’ several CNS protective mechanisms and may employ microglia and CNS-border associated macrophages into helping cancer cells to colonize a pre-metastatic niche. We describe emerging molecular insights into mechanisms governing communication between microglia and metastatic cancer cells that culminate in activation of CNS resident microglia and trafficking of monocytic cells from the periphery. We present mechanisms controlling those processes in brain metastases and hypothesize on potential therapeutic approaches. In summary, microglia and non-parenchymal brain macrophages are involved in multiple stages of a metastatic disease and, unlike tumor cells, are genetically stable and predictable, which makes them an attractive target for anticancer therapies.
Collapse
Affiliation(s)
- Hua You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,School of Laboratory Medicine, YouJiang Medical University for Nationalities, Baise, China.,Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Szymon Baluszek
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
9
|
Knittel G, Rehkämper T, Korovkina D, Liedgens P, Fritz C, Torgovnick A, Al-Baldawi Y, Al-Maarri M, Cun Y, Fedorchenko O, Riabinska A, Beleggia F, Nguyen PH, Wunderlich FT, Ortmann M, Montesinos-Rongen M, Tausch E, Stilgenbauer S, P Frenzel L, Herling M, Herling C, Bahlo J, Hallek M, Peifer M, Buettner R, Persigehl T, Reinhardt HC. Two mouse models reveal an actionable PARP1 dependence in aggressive chronic lymphocytic leukemia. Nat Commun 2017; 8:153. [PMID: 28751718 PMCID: PMC5532225 DOI: 10.1038/s41467-017-00210-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) remains an incurable disease. Two recurrent cytogenetic aberrations, namely del(17p), affecting TP53, and del(11q), affecting ATM, are associated with resistance against genotoxic chemotherapy (del17p) and poor outcome (del11q and del17p). Both del(17p) and del(11q) are also associated with inferior outcome to the novel targeted agents, such as the BTK inhibitor ibrutinib. Thus, even in the era of targeted therapies, CLL with alterations in the ATM/p53 pathway remains a clinical challenge. Here we generated two mouse models of Atm- and Trp53-deficient CLL. These animals display a significantly earlier disease onset and reduced overall survival, compared to controls. We employed these models in conjunction with transcriptome analyses following cyclophosphamide treatment to reveal that Atm deficiency is associated with an exquisite and genotype-specific sensitivity against PARP inhibition. Thus, we generate two aggressive CLL models and provide a preclinical rational for the use of PARP inhibitors in ATM-affected human CLL. ATM and TP53 mutations are associated with poor prognosis in chronic lymphocytic leukaemia (CLL). Here the authors generate mouse models of Tp53- and Atm-defective CLL mimicking the high-risk form of human disease and show that Atm-deficient CLL is sensitive to PARP1 inhibition.
Collapse
Affiliation(s)
- Gero Knittel
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany. .,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany. .,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.
| | - Tim Rehkämper
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Darya Korovkina
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Paul Liedgens
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Christian Fritz
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Alessandro Torgovnick
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Yussor Al-Baldawi
- Department of Radiology, Medical Faculty, University Hospital of Cologne, Cologne, 50931, Germany
| | - Mona Al-Maarri
- Max-Planck-Institute for Metabolism Research, Cologne, 50931, Germany
| | - Yupeng Cun
- Department of Translational Genomics, University of Cologne, Cologne, 50931, Germany
| | - Oleg Fedorchenko
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Arina Riabinska
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Filippo Beleggia
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Phuong-Hien Nguyen
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | | | - Monika Ortmann
- Institute of Pathology, University Hospital of Cologne, Cologne, 50931, Germany
| | | | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Ulm, 89070, Germany
| | | | - Lukas P Frenzel
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Marco Herling
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, 50931, Germany
| | - Carmen Herling
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Jasmin Bahlo
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany
| | - Michael Hallek
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany.,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany
| | - Martin Peifer
- Department of Translational Genomics, University of Cologne, Cologne, 50931, Germany
| | - Reinhard Buettner
- Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany.,Institute of Pathology, University Hospital of Cologne, Cologne, 50931, Germany
| | - Thorsten Persigehl
- Department of Radiology, Medical Faculty, University Hospital of Cologne, Cologne, 50931, Germany
| | - H Christian Reinhardt
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, 50931, Germany. .,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 50931, Germany. .,Center of Integrated Oncology (CIO), University Hospital of Cologne, Cologne, 50931, Germany. .,Center of Molecular Medicine, University of Cologne, Cologne, 50931, Germany.
| |
Collapse
|
10
|
Endogenous Il10 alleviates the systemic antiviral cellular immune response and T cell-mediated immunopathology in select organs of acutely LCMV-infected mice. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 185:3025-38. [PMID: 26506472 DOI: 10.1016/j.ajpath.2015.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/10/2015] [Accepted: 07/14/2015] [Indexed: 12/22/2022]
Abstract
The immunoregulatory cytokine IL-10 suppresses T-cell immunity. The complementary question, whether IL-10 is also involved in limiting the collateral damage of vigorous T cell responses, has not been addressed in detail. Here, we report that the particularly strong virus-specific immune response during acute primary infection with the lymphocytic choriomeningitis virus (LCMV) in mice is significantly further increased in Il10-deficient mice, particularly regarding frequencies and cytotoxic activity of CD8(+) T cells. This increase results in exacerbating immunopathology in select organs, ranging from transient local swelling to an increased risk for mortality. Remarkably, LCMV-induced, T cell-mediated hepatitis is not affected by endogenous Il10. The alleviating effect of Il10 on LCMV-induced immunopathology was found to be operative in delayed-type hypersensitivity footpad-swelling reaction and in debilitating meningitis in mice of both the C57BL/6 and BALB/c strains. These strains are prototypic counterpoles for genetically imprinted type 1-biased versus type 2-biased T cell-mediated immune responses against various infectious pathogens. However, during acute LCMV infection, neither systemic cytokine patterns nor the impact of Il10 on LCMV-induced immunopathology differed conspicuously between these two strains of mice. This study documents a physiological role of Il10 in the regulation of a balanced T-cell response limiting immunopathological damage.
Collapse
|
11
|
B-cell-specific conditional expression of Myd88p.L252P leads to the development of diffuse large B-cell lymphoma in mice. Blood 2016; 127:2732-41. [PMID: 27048211 DOI: 10.1182/blood-2015-11-684183] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/30/2016] [Indexed: 12/20/2022] Open
Abstract
The adaptor protein MYD88 is critical for relaying activation of Toll-like receptor signaling to NF-κB activation. MYD88 mutations, particularly the p.L265P mutation, have been described in numerous distinct B-cell malignancies, including diffuse large B-cell lymphoma (DLBCL). Twenty-nine percent of activated B-cell-type DLBCL (ABC-DLBCL), which is characterized by constitutive activation of the NF-κB pathway, carry the p.L265P mutation. In addition, ABC-DLBCL frequently displays focal copy number gains affecting BCL2 Here, we generated a novel mouse model in which Cre-mediated recombination, specifically in B cells, leads to the conditional expression of Myd88(p.L252P) (the orthologous position of the human MYD88(p.L265P) mutation) from the endogenous locus. These mice develop a lymphoproliferative disease and occasional transformation into clonal lymphomas. The clonal disease displays the morphologic and immunophenotypical characteristics of ABC-DLBCL. Lymphomagenesis can be accelerated by crossing in a further novel allele, which mediates conditional overexpression of BCL2 Cross-validation experiments in human DLBCL samples revealed that both MYD88 and CD79B mutations are substantially enriched in ABC-DLBCL compared with germinal center B-cell DLBCL. Furthermore, analyses of human DLBCL genome sequencing data confirmed that BCL2 amplifications frequently co-occurred with MYD88 mutations, further validating our approach. Finally, in silico experiments revealed that MYD88-mutant ABC-DLBCL cells in particular display an actionable addiction to BCL2. Altogether, we generated a novel autochthonous mouse model of ABC-DLBCL that could be used as a preclinical platform for the development and validation of novel therapeutic approaches for the treatment of ABC-DLBCL.
Collapse
|
12
|
Deckert M, Montesinos-Rongen M, Brunn A, Siebert R. Systems biology of primary CNS lymphoma: from genetic aberrations to modeling in mice. Acta Neuropathol 2014; 127:175-88. [PMID: 24240734 DOI: 10.1007/s00401-013-1202-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/21/2013] [Indexed: 12/15/2022]
Abstract
Primary lymphoma of the central nervous system (CNS, PCNSL) is a specific diffuse large B cell lymphoma entity arising in and confined to the CNS. Despite extensive research since many decades, the pathogenetic mechanisms underlying the remarkable tropism of this peculiar malignant hematopoietic tumor remain still to be elucidated. In the present review, we summarize the present knowledge on the genotypic and phenotypic characteristics of the tumor cells of PCNSL, give an overview over deregulated molecular pathways in PCNSL and present recent progress in the field of preclinical modeling of PCNSL in mice. With regard to the phenotype, PCNSL cells resemble late germinal center exit IgM+IgD+ B cells with blocked terminal B cell differentiation. They show continued BCL6 activity in line with ongoing activity of the germinal center program. This together with the pathways deregulated by genetic alterations may foster B cell activation and brisk proliferation, which correlated with the simultaneous MYC and BCL2 overexpression characteristic for PCNSL. On the genetic level, PCNSL are characterized by ongoing aberrant somatic hypermutation that, besides the IG locus, targets the PAX5, TTF, MYC, and PIM1 genes. Moreover, PCNSL cells show impaired IG class switch due to sμ region deletions, and PRDM1 mutations. Several important pathways, i.e., the B cell receptor (BCR), the toll-like receptor, and the nuclear factor-κB pathway, are activated frequently due to genetic changes affecting genes like CD79B, SHIP, CBL, BLNK, CARD11, MALT1, BCL2, and MYD88. These changes likely foster tumor cell survival. Nevertheless, many of these features are also present in subsets of systemic DLBLC and might not be the only reasons for the peculiar tropism of PCNSL. Here, preclinical animal models that closely mimic the clinical course and neuropathology of human PCNSL may provide further insight and we discuss recent advances in this field. Such models enable us to understand the pathogenetic interaction between the malignant B cells, resident cell populations of the CNS, and the associated inflammatory infiltrate. Indeed, the immunophenotype of the CNS as well as tumor cell characteristics and intracerebral interactions may create a micromilieu particularly conducive to PCNSL that may foster aggressiveness of tumor cells and accelerate the fatal course of disease. Suitable animal models may also serve as a well-defined preclinical system and may provide a useful tool for developing new specific therapeutic strategies.
Collapse
|