1
|
Jin C, Lu Z, Chen Y, Hu H, Zhou M, Zhang Y, Ouyang G, Li T, Sheng L. Identification of biomarkers for chronic lymphocytic leukemia risk: a proteome-wide Mendelian randomization study. Discov Oncol 2025; 16:2. [PMID: 39751938 PMCID: PMC11699013 DOI: 10.1007/s12672-024-01699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) is a common hematologic malignancy. Although previous research has explored associations between plasma proteins and CLL, the causal relationships remain unclear. This study used Mendelian randomization (MR) to investigate the causal relationship between 7156 plasma proteins and CLL risk. METHODS A two-sample MR analysis assessed the impact of specific plasma proteins on CLL risk, using data from the Finngen Proteomics project (analyzing 828 participants) and the UK Biobank. Additional analyses included colocalization, phenomenon-wide MR, and protein-protein interaction networks. RESULTS The study identified nine plasma proteins significantly associated with CLL risk. Increased levels of Peptidyl-prolyl cis-trans isomerase E (PPIE) (OR = 1.66, 95% CI 1.22-2.27, P = 0.001) were associated with an increased risk of developing CLL, whereas Protein O-Mannosyltransferase 2 (POMGNT2) (OR = 0.62, 95% CI 0.41-0.91, P = 0.017) and C-C Motif Chemokine Ligand 14(CCL14) (OR = 0.80, 95% CI 0.67-0.94, P = 0.010) were associated with a reduced risk of CLL. Colocalization analysis suggested that PPIE may share pathogenic variants with CLL (PP.H4 = 0.758). Phenomenon-wide MR analysis of PPIE also indicated associations with other clinical features, including rheumatic diseases and type 2 diabetes. Protein-protein interaction and drug-gene interaction analyses highlighted CDC5L and SNW1 as potential therapeutic targets. CONCLUSION This study identifies nine plasma proteins linked to CLL risk, with PPIE offering new insights into the disease's pathogenesis. Further research is needed to validate these findings and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Changyu Jin
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Zehong Lu
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Yuzhan Chen
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Huijie Hu
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Miao Zhou
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Yanli Zhang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China
| | - Guifang Ouyang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China.
| | - Tongyu Li
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China.
| | - Lixia Sheng
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No.59 Liu-Ting Road, Ningbo, 315000, People's Republic of China.
| |
Collapse
|
2
|
Vom Stein AF, Hallek M, Nguyen PH. Role of the tumor microenvironment in CLL pathogenesis. Semin Hematol 2024; 61:142-154. [PMID: 38220499 DOI: 10.1053/j.seminhematol.2023.12.004] [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: 11/07/2023] [Revised: 12/02/2023] [Accepted: 12/23/2023] [Indexed: 01/16/2024]
Abstract
Chronic lymphocytic leukemia (CLL) cells extensively interact with and depend on their surrounding tumor microenvironment (TME). The TME encompasses a heterogeneous array of cell types, soluble signals, and extracellular vesicles, which contribute significantly to CLL pathogenesis. CLL cells and the TME cooperatively generate a chronic inflammatory milieu, which reciprocally reprograms the TME and activates a signaling network within CLL cells, promoting their survival and proliferation. Additionally, the inflammatory milieu exerts chemotactic effects, attracting CLL cells and other immune cells to the lymphoid tissues. The intricate CLL-TME interactions also facilitate immune evasion and compromise leukemic cell surveillance. We also review recent advances that have shed light on additional aspects that are substantially influenced by the CLL-TME interplay.
Collapse
Affiliation(s)
- Alexander F Vom Stein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Michael Hallek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Phuong-Hien Nguyen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany.
| |
Collapse
|
3
|
Gorczynski R. Translation of Data from Animal Models of Cancer to Immunotherapy of Breast Cancer and Chronic Lymphocytic Leukemia. Genes (Basel) 2024; 15:292. [PMID: 38540350 PMCID: PMC10970502 DOI: 10.3390/genes15030292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 06/14/2024] Open
Abstract
The field of clinical oncology has been revolutionized over the past decade with the introduction of many new immunotherapies the existence of which have depended to a large extent on experimentation with both in vitro analysis and the use of various animal models, including gene-modified mice. The discussion below will review my own laboratory's studies, along with those of others in the field, on cancer immunotherapy. Our own studies have predominantly dwelt on two models of malignancy, namely a solid tumor model (breast cancer) and lymphoma. The data from our own laboratory, and that of other scientists, highlights the novel information so obtained, and the evidence that application of such information has already had an impact on immunotherapy of human oncologic diseases.
Collapse
Affiliation(s)
- Reginald Gorczynski
- Institute of Medical Science, Department of Immunology and Surgery, University of Toronto, C/O 429 Drewry Avenue, Toronto, ON M2R 2K6, Canada
| |
Collapse
|
4
|
Hu R, Ling X, Yang T, Zhang J, Gu X, Li F, Chen H, Wen Y, Li Z, Zou Y, Du Y. Cytokine levels in patients with non-M3 myeloid leukemia are key indicators of how well the disease responds to chemotherapy. Clin Exp Med 2023; 23:4623-4632. [PMID: 37925379 DOI: 10.1007/s10238-023-01242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant hematological neoplastic disease. Autocrine or paracrine cytokines released by leukemic cells regulate the proliferation of AML cells. It is uncertain whether cytokines can indicate whether patients with AML are in remission with chemotherapy. The goal of this study was to evaluate the levels of Th1/Th2/Th17 cytokines in AML patients before and after chemotherapy to determine whether the cytokine levels could predict disease remission after chemotherapy. It was found that the levels of IL-5, IL-6, IL-8, IL-10, TNF-α, TNF-β, IL-17F, and IL-22 were significantly increased at the time of AML diagnosis in patients who achieved remission after two chemotherapy treatments (P < 0.05). After chemotherapy, the cytokine levels were reduced in patients with remission, while the levels of IL-6 and IL-8 were raised in patients without remission (P < 0.05). A comparison of cytokine levels before and after chemotherapy in patients who achieved remission showed areas under the curve (AUCs) of 0.69 for both IL-6 and IL-8. In addition, a comparison of the remission and non-remission groups after chemotherapy showed an AUC of 0.77 for IL-6. We then calculated the cutoff value using receiver operating characteristic curves. Values of IL-6 < 9.99 and IL-8 < 8.46 at the time of diagnosis were predictive of chemotherapy success and remission, while IL-6 > 14.89 at diagnosis suggested that chemotherapy would not be successful and remission would not be achieved. Multifactorial analysis showed that age, Neu, IL-6, and IL-8 were independent risk factors for AML prognosis, and IL-6 (OR = 5.48, P = 0.0038) was superior to age (OR = 3.36, P = 0.0379), Neu (OR = 0.28, P = 0.0308), IL-8 (OR = 0.0421, P = 0.0421). In conclusion, IL-6 levels were found to be predictive of the likelihood of remission.
Collapse
Affiliation(s)
- Rui Hu
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xiaosui Ling
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Tonghua Yang
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jinping Zhang
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuezhong Gu
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Fan Li
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Heng Chen
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wen
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Zengzheng Li
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Yunlian Zou
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Yunyun Du
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.
- Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
| |
Collapse
|
5
|
Mesenchymal stromal cell senescence in haematological malignancies. Cancer Metastasis Rev 2023; 42:277-296. [PMID: 36622509 DOI: 10.1007/s10555-022-10069-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/17/2022] [Indexed: 01/10/2023]
Abstract
Acute myeloid leukaemia (AML), chronic lymphocytic leukaemia (CLL), and multiple myeloma (MM) are age-related haematological malignancies with defined precursor states termed myelodysplastic syndrome (MDS), monoclonal B-cell lymphocytosis (MBL), and monoclonal gammopathy of undetermined significance (MGUS), respectively. While the progression from asymptomatic precursor states to malignancy is widely considered to be mediated by the accumulation of genetic mutations in neoplastic haematopoietic cell clones, recent studies suggest that intrinsic genetic changes, alone, may be insufficient to drive the progression to overt malignancy. Notably, studies suggest that extrinsic, microenvironmental changes in the bone marrow (BM) may also promote the transition from these precursor states to active disease. There is now enhanced focus on extrinsic, age-related changes in the BM microenvironment that accompany the development of AML, CLL, and MM. One of the most prominent changes associated with ageing is the accumulation of senescent mesenchymal stromal cells within tissues and organs. In comparison with proliferating cells, senescent cells display an altered profile of secreted factors (secretome), termed the senescence-associated-secretory phenotype (SASP), comprising proteases, inflammatory cytokines, and growth factors that may render the local microenvironment favourable for cancer growth. It is well established that BM mesenchymal stromal cells (BM-MSCs) are key regulators of haematopoietic stem cell maintenance and fate determination. Moreover, there is emerging evidence that BM-MSC senescence may contribute to age-related haematopoietic decline and cancer development. This review explores the association between BM-MSC senescence and the development of haematological malignancies, and the functional role of senescent BM-MSCs in the development of these cancers.
Collapse
|
6
|
Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, Leivas A, Río P, Martínez-López J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol 2022; 13:953849. [PMID: 35990652 PMCID: PMC9381932 DOI: 10.3389/fimmu.2022.953849] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the impressive results of autologous CAR-T cell therapy in refractory B lymphoproliferative diseases, CAR-NK immunotherapy emerges as a safer, faster, and cost-effective approach with no signs of severe toxicities as described for CAR-T cells. Permanently scrutinized for its efficacy, recent promising data in CAR-NK clinical trials point out the achievement of deep, high-quality responses, thus confirming its potential clinical use. Although CAR-NK cell therapy is not significantly affected by the loss or downregulation of its CAR tumor target, as in the case of CAR-T cell, a plethora of common additional tumor intrinsic or extrinsic mechanisms that could also disable NK cell function have been described. Therefore, considering lessons learned from CAR-T cell therapy, the emergence of CAR-NK cell therapy resistance can also be envisioned. In this review we highlight the processes that could be involved in its development, focusing on cytokine addiction and potential fratricide during manufacturing, poor tumor trafficking, exhaustion within the tumor microenvironment (TME), and NK cell short in vivo persistence on account of the limited expansion, replicative senescence, and rejection by patient’s immune system after lymphodepletion recovery. Finally, we outline new actively explored alternatives to overcome these resistance mechanisms, with a special emphasis on CRISPR/Cas9 mediated genetic engineering approaches, a promising platform to optimize CAR-NK cell function to eradicate refractory cancers.
Collapse
Affiliation(s)
- Antonio Valeri
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Almudena García-Ortiz
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Eva Castellano
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Laura Córdoba
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Elena Maroto-Martín
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jessica Encinas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alejandra Leivas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Joaquín Martínez-López,
| |
Collapse
|
7
|
The Role of Neutrophils in the Pathogenesis of Chronic Lymphocytic Leukemia. Int J Mol Sci 2021; 23:ijms23010365. [PMID: 35008790 PMCID: PMC8745265 DOI: 10.3390/ijms23010365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 11/25/2022] Open
Abstract
Tumor-associated neutrophils appear to be a crucial element of the tumor microenvironment that actively participates in the development and progression of cancerous diseases. The increased lifespan, plasticity in changing of phenotype, and functions of neutrophils influence the course of the disease and may significantly affect survival. In patients with chronic lymphocytic leukemia (CLL), disturbances in neutrophils functions impede the effective immune defense against pathogens. Therefore, understanding the mechanism underlying such a phenomenon in CLL seems to be of great importance. Here we discuss the recent reports analyzing the phenotype and functions of neutrophils in CLL, the most common leukemia in adults. We summarize the data concerning both the phenotype and the mechanisms by which neutrophils directly support the proliferation and survival of malignant B cells.
Collapse
|
8
|
Alankus B, Ecker V, Vahl N, Braun M, Weichert W, Macher-Göppinger S, Gehring T, Neumayer T, Zenz T, Buchner M, Ruland J. Pathological RANK signaling in B cells drives autoimmunity and chronic lymphocytic leukemia. J Exp Med 2021; 218:211464. [PMID: 33075129 PMCID: PMC7868734 DOI: 10.1084/jem.20200517] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/30/2020] [Accepted: 09/03/2020] [Indexed: 12/28/2022] Open
Abstract
Clinical evidence suggests alterations in receptor activator of NF-κB (RANK) signaling are key contributors to B cell autoimmunity and malignancy, but the pathophysiological consequences of aberrant B cell–intrinsic RANK signaling remain unknown. We generated mice that express a human lymphoma–derived, hyperactive RANKK240E variant in B lymphocytes in vivo. Forced RANK signaling disrupted B cell tolerance and induced a fully penetrant systemic lupus erythematosus–like disease in addition to the development of chronic lymphocytic leukemia (CLL). Importantly, RANKK240E transgenic CLL cells as well as CLL cells of independent murine and of human origin depend on microenvironmental RANK ligand (RANKL) for tumor cell survival. Consequently, inhibition of the RANKL–RANK axis with anti-RANKL antibodies killed murine and human CLL cells in vitro and in vivo. These results establish pathological B cell–intrinsic RANK signaling as a potential driver of autoimmunity and B cell malignancy, and they suggest the exploitation of clinically available anti-RANKL compounds for CLL treatment.
Collapse
Affiliation(s)
- Begüm Alankus
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Veronika Ecker
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Nathalie Vahl
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martina Braun
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium, Heidelberg, Germany
| | | | - Torben Gehring
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tanja Neumayer
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Maike Buchner
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,German Cancer Consortium, Heidelberg, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,German Cancer Consortium, Heidelberg, Germany.,German Center for Infection Research, Munich, Germany
| |
Collapse
|
9
|
Impact of Immune Parameters and Immune Dysfunctions on the Prognosis of Patients with Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:cancers13153856. [PMID: 34359757 PMCID: PMC8345723 DOI: 10.3390/cancers13153856] [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: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary In chronic lymphocytic leukemia (CLL), immune alterations—affecting both the innate and adaptive immunity—are very common. As a clinical consequence, patients with CLL frequently present with autoimmune phenomena, increased risk of infections and second malignancies. The aim of this review article is to present available data on CLL-associated alterations of immune parameters that correlate with known prognostic markers and with clinical outcome. Also, data on the impact of immune-related clinical manifestations on the prognosis of patients with CLL will be discussed. Abstract Chronic lymphocytic leukemia (CLL) is characterized by a wide spectrum of immune alterations, affecting both the innate and adaptive immunity. These immune dysfunctions strongly impact the immune surveillance, facilitate tumor progression and eventually affect the disease course. Quantitative and functional alterations involving conventional T cells, γδ T cells, regulatory T cells, NK and NKT cells, and myeloid cells, together with hypogammaglobulinemia, aberrations in the complement pathways and altered cytokine signature have been reported in patients with CLL. Some of these immune parameters have been shown to associate with other CLL-related characteristics with a known prognostic relevance or to correlate with disease prognosis. Also, in CLL, the complex immune response dysfunctions eventually translate in clinical manifestations, including autoimmune phenomena, increased risk of infections and second malignancies. These clinical issues are overall the most common complications that affect the course and management of CLL, and they also may impact overall disease prognosis.
Collapse
|
10
|
Bozorgmehr N, Okoye I, Oyegbami O, Xu L, Fontaine A, Cox-Kennett N, Larratt LM, Hnatiuk M, Fagarasanu A, Brandwein J, Peters AC, Elahi S. Expanded antigen-experienced CD160 +CD8 +effector T cells exhibit impaired effector functions in chronic lymphocytic leukemia. J Immunother Cancer 2021; 9:jitc-2020-002189. [PMID: 33931471 PMCID: PMC8098955 DOI: 10.1136/jitc-2020-002189] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background T cell exhaustion compromises antitumor immunity, and a sustained elevation of co-inhibitory receptors is a hallmark of T cell exhaustion in solid tumors. Similarly, upregulation of co-inhibitory receptors has been reported in T cells in hematological cancers such as chronic lymphocytic leukemia (CLL). However, the role of CD160, a glycosylphosphatidylinositol-anchored protein, as one of these co-inhibitory receptors has been contradictory in T cell function. Therefore, we decided to elucidate how CD160 expression and/or co-expression with other co-inhibitory receptors influence T cell effector functions in patients with CLL. Methods We studied 56 patients with CLL and 25 age-matched and sex-matched healthy controls in this study. The expression of different co-inhibitory receptors was analyzed in T cells obtained from the peripheral blood or the bone marrow. Also, we quantified the properties of extracellular vesicles (EVs) in the plasma of patients with CLL versus healthy controls. Finally, we measured 29 different cytokines, chemokines or other biomarkers in the plasma specimens of patients with CLL and healthy controls. Results We found that CD160 was the most upregulated co-inhibitory receptor in patients with CLL. Its expression was associated with an exhausted T cell phenotype. CD160+CD8+ T cells were highly antigen-experienced/effector T cells, while CD160+CD4+ T cells were more heterogeneous. In particular, we identified EVs as a source of CD160 in the plasma of patients with CLL that can be taken up by T cells. Moreover, we observed a dominantly proinflammatory cytokine profile in the plasma of patients with CLL. In particular, interleukin-16 (IL-16) was highly elevated and correlated with the advanced clinical stage (Rai). Furthermore, we observed that the incubation of T cells with IL-16 results in the upregulation of CD160. Conclusions Our study provides a novel insight into the influence of CD160 expression/co-expression with other co-inhibitory receptors in T cell effector functions in patients with CLL. Besides, IL-16-mediated upregulation of CD160 expression in T cells highlights the importance of IL-16/CD160 as potential immunotherapy targets in patients with CLL. Therefore, our findings propose a significant role for CD160 in T cell exhaustion in patients with CLL.
Collapse
Affiliation(s)
- Najmeh Bozorgmehr
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Isobel Okoye
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Olaide Oyegbami
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lai Xu
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amelie Fontaine
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nanette Cox-Kennett
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Loree M Larratt
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Hnatiuk
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andrei Fagarasanu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph Brandwein
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anthea C Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shokrollah Elahi
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada .,Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Blanco G, Puiggros A, Sherry B, Nonell L, Calvo X, Puigdecanet E, Chiu PY, Kieso Y, Ferrer G, Palacios F, Arnal M, Rodríguez-Rivera M, Gimeno E, Abella E, Rai KR, Abrisqueta P, Bosch F, Calon A, Ferrer A, Chiorazzi N, Espinet B. Chronic lymphocytic leukemia-like monoclonal B-cell lymphocytosis exhibits an increased inflammatory signature that is reduced in early-stage chronic lymphocytic leukemia. Exp Hematol 2021; 95:68-80. [PMID: 33421548 DOI: 10.1016/j.exphem.2020.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 11/16/2022]
Abstract
Several studies in chronic lymphocytic leukemia (CLL) patients have reported impaired immune cell functions, which contribute to tumor evasion and disease progression. However, studies on CLL-like monoclonal B-cell lymphocytosis (MBL) are scarce. In the study described here, we characterized the immune environment in 62 individuals with clinical MBL, 56 patients with early-stage CLL, and 31 healthy controls. Gene expression arrays and quantitative reverse transcription polymerase chain reaction were performed on RNA from CD4+ peripheral blood cells; serum cytokines were measured with immunoassays; and HLA-DR expression on circulating monocytes, as well as the percentages of Th1, cytotoxic, exhausted, and effector CD4+ T cells, were evaluated by flow cytometry. In addition, cell cultures of clonal B cells and CD14-enriched or -depleted cell fractions were performed. Strikingly, MBL and early-stage CLL differed in pro-inflammatory signatures. An increased inflammatory drive orchestrated mainly by monocytes was identified in MBL, which exhibited enhanced phagocytosis, pattern recognition receptors, interleukin-8 (IL8), HMGB1, and acute response signaling pathways and increased pro-inflammatory cytokines (in particular IL8, interferon γ [IFNγ], and tumor necrosis factor α). This inflammatory signature was diminished in early-stage CLL (reduced IL8 and IFNγ levels, IL8 signaling pathway, and monocytic HLA-DR expression compared with MBL), especially in those patients with mutations in IGHV genes. Additionally, CD4+ T cells of MBL and early-stage CLL exhibited a similar upregulation of Th1 and cytotoxic genes and expanded CXCR3+ and perforin+ CD4+ T cells, as well as PD1+ CD4+ T cells, compared with controls. Cell culture assays disclosed tumor-supporting effects of monocytes similarly observed in MBL and early-stage CLL. These novel findings reveal differences in the inflammatory environment between MBL and CLL, highlighting an active role for antigen stimulation in the very early stages of the disease, potentially related to malignant B-cell transformation.
Collapse
Affiliation(s)
- Gonzalo Blanco
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Anna Puiggros
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Barbara Sherry
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY; Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, NY; Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY
| | | | - Xavier Calvo
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | | | - Pui Yan Chiu
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Yasmine Kieso
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Florencia Palacios
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | | | - María Rodríguez-Rivera
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Eva Gimeno
- Servei d'Hematologia, Hospital del Mar-IMIM, Barcelona, Spain; Grup de Recerca Clínica Aplicada en Neoplàsies Hematològiques, Cancer Research Program, IMIM-Hospital del Mar, Barcelona, Spain
| | - Eugènia Abella
- Servei d'Hematologia, Hospital del Mar-IMIM, Barcelona, Spain; Grup de Recerca Clínica Aplicada en Neoplàsies Hematològiques, Cancer Research Program, IMIM-Hospital del Mar, Barcelona, Spain
| | - Kanti R Rai
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, NY; Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Pau Abrisqueta
- Servei d'Hematologia, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Francesc Bosch
- Servei d'Hematologia, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Alexandre Calon
- Laboratori de Recerca Translacional en Microambient Tumoral, Cancer Research Program, IMIM, Barcelona, Spain
| | - Ana Ferrer
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Nicholas Chiorazzi
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, NY; Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY; Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Blanca Espinet
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Barcelona, Spain; Grup de Recerca Translacional en Neoplàsies Hematològiques, Cancer Research Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.
| |
Collapse
|
12
|
Olagoke O, Quigley BL, Timms P. Koalas vaccinated against Koala retrovirus respond by producing increased levels of interferon-gamma. Virol J 2020; 17:168. [PMID: 33129323 PMCID: PMC7602773 DOI: 10.1186/s12985-020-01442-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/27/2020] [Indexed: 01/01/2023] Open
Abstract
Koala retrovirus (KoRV) is believed to be in an active state of endogenization into the koala genome. KoRV is present as both an endogenous and exogenous infection in all koalas in northern Australia. KoRV has been linked to koala pathologies including neoplasia and increased susceptibility to Chlamydia. A KoRV vaccine recently trialled in 10 northern koalas improved antibody response and reduced viral load. This communication reports the expression of key immune genes underlining the innate and adaptive immune response to vaccination in these northern koalas. The results showed that prior to vaccination, IL-8 was expressed at the highest levels, with at least 200-fold greater expression compared to other cytokines, while CD8 mRNA expression was significantly higher than CD4 mRNA expression level. Interferon-γ was up-regulated at both 4- and 8-weeks post-vaccination while IL-8 was down-regulated at 8-weeks post-vaccination.
Collapse
Affiliation(s)
- Olusola Olagoke
- Genecology Research Centre, University of the Sunshine Coast, Sunshine Coast, QLD, Australia.
| | - Bonnie L Quigley
- Genecology Research Centre, University of the Sunshine Coast, Sunshine Coast, QLD, Australia
| | - Peter Timms
- Genecology Research Centre, University of the Sunshine Coast, Sunshine Coast, QLD, Australia
| |
Collapse
|
13
|
Dubois N, Crompot E, Meuleman N, Bron D, Lagneaux L, Stamatopoulos B. Importance of Crosstalk Between Chronic Lymphocytic Leukemia Cells and the Stromal Microenvironment: Direct Contact, Soluble Factors, and Extracellular Vesicles. Front Oncol 2020; 10:1422. [PMID: 32974152 PMCID: PMC7466743 DOI: 10.3389/fonc.2020.01422] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is caused by the accumulation of malignant B cells due to a defect in apoptosis and the presence of small population of proliferating cells principally in the lymph nodes. The abnormal survival of CLL B cells is explained by a plethora of supportive stimuli produced by the surrounding cells of the microenvironment, including follicular dendritic cells (FDCs), and mesenchymal stromal cells (MSCs). This crosstalk between malignant cells and normal cells can take place directly by cell-to-cell contact (assisted by adhesion molecules such as VLA-4 or CD100), indirectly by soluble factors (chemokines such as CXCL12, CXCL13, or CCL2) interacting with their receptors or by the exchange of material (protein, microRNAs or long non-coding RNAs) via extracellular vesicles. These different communication methods lead to different activation pathways (including BCR and NFκB pathways), gene expression modifications (chemokines, antiapoptotic protein increase, prognostic biomarkers), chemotaxis, homing in lymphoid tissues and survival of leukemic cells. In addition, these interactions are bidirectional, and CLL cells can manipulate the normal surrounding stromal cells in different ways to establish a supportive microenvironment. Here, we review this complex crosstalk between CLL cells and stromal cells, focusing on the different types of interactions, activated pathways, treatment strategies to disrupt this bidirectional communication, and the prognostic impact of these induced modifications.
Collapse
Affiliation(s)
- Nathan Dubois
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emerence Crompot
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Brussels, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
14
|
Kreuzberger N, Damen JA, Trivella M, Estcourt LJ, Aldin A, Umlauff L, Vazquez-Montes MD, Wolff R, Moons KG, Monsef I, Foroutan F, Kreuzer KA, Skoetz N. Prognostic models for newly-diagnosed chronic lymphocytic leukaemia in adults: a systematic review and meta-analysis. Cochrane Database Syst Rev 2020; 7:CD012022. [PMID: 32735048 PMCID: PMC8078230 DOI: 10.1002/14651858.cd012022.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic lymphocytic leukaemia (CLL) is the most common cancer of the lymphatic system in Western countries. Several clinical and biological factors for CLL have been identified. However, it remains unclear which of the available prognostic models combining those factors can be used in clinical practice to predict long-term outcome in people newly-diagnosed with CLL. OBJECTIVES To identify, describe and appraise all prognostic models developed to predict overall survival (OS), progression-free survival (PFS) or treatment-free survival (TFS) in newly-diagnosed (previously untreated) adults with CLL, and meta-analyse their predictive performances. SEARCH METHODS We searched MEDLINE (from January 1950 to June 2019 via Ovid), Embase (from 1974 to June 2019) and registries of ongoing trials (to 5 March 2020) for development and validation studies of prognostic models for untreated adults with CLL. In addition, we screened the reference lists and citation indices of included studies. SELECTION CRITERIA We included all prognostic models developed for CLL which predict OS, PFS, or TFS, provided they combined prognostic factors known before treatment initiation, and any studies that tested the performance of these models in individuals other than the ones included in model development (i.e. 'external model validation studies'). We included studies of adults with confirmed B-cell CLL who had not received treatment prior to the start of the study. We did not restrict the search based on study design. DATA COLLECTION AND ANALYSIS We developed a data extraction form to collect information based on the Checklist for Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modelling Studies (CHARMS). Independent pairs of review authors screened references, extracted data and assessed risk of bias according to the Prediction model Risk Of Bias ASsessment Tool (PROBAST). For models that were externally validated at least three times, we aimed to perform a quantitative meta-analysis of their predictive performance, notably their calibration (proportion of people predicted to experience the outcome who do so) and discrimination (ability to differentiate between people with and without the event) using a random-effects model. When a model categorised individuals into risk categories, we pooled outcome frequencies per risk group (low, intermediate, high and very high). We did not apply GRADE as guidance is not yet available for reviews of prognostic models. MAIN RESULTS From 52 eligible studies, we identified 12 externally validated models: six were developed for OS, one for PFS and five for TFS. In general, reporting of the studies was poor, especially predictive performance measures for calibration and discrimination; but also basic information, such as eligibility criteria and the recruitment period of participants was often missing. We rated almost all studies at high or unclear risk of bias according to PROBAST. Overall, the applicability of the models and their validation studies was low or unclear; the most common reasons were inappropriate handling of missing data and serious reporting deficiencies concerning eligibility criteria, recruitment period, observation time and prediction performance measures. We report the results for three models predicting OS, which had available data from more than three external validation studies: CLL International Prognostic Index (CLL-IPI) This score includes five prognostic factors: age, clinical stage, IgHV mutational status, B2-microglobulin and TP53 status. Calibration: for the low-, intermediate- and high-risk groups, the pooled five-year survival per risk group from validation studies corresponded to the frequencies observed in the model development study. In the very high-risk group, predicted survival from CLL-IPI was lower than observed from external validation studies. Discrimination: the pooled c-statistic of seven external validation studies (3307 participants, 917 events) was 0.72 (95% confidence interval (CI) 0.67 to 0.77). The 95% prediction interval (PI) of this model for the c-statistic, which describes the expected interval for the model's discriminative ability in a new external validation study, ranged from 0.59 to 0.83. Barcelona-Brno score Aimed at simplifying the CLL-IPI, this score includes three prognostic factors: IgHV mutational status, del(17p) and del(11q). Calibration: for the low- and intermediate-risk group, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high-risk group. Discrimination: the pooled c-statistic of four external validation studies (1755 participants, 416 events) was 0.64 (95% CI 0.60 to 0.67); 95% PI 0.59 to 0.68. MDACC 2007 index score The authors presented two versions of this model including six prognostic factors to predict OS: age, B2-microglobulin, absolute lymphocyte count, gender, clinical stage and number of nodal groups. Only one validation study was available for the more comprehensive version of the model, a formula with a nomogram, while seven studies (5127 participants, 994 events) validated the simplified version of the model, the index score. Calibration: for the low- and intermediate-risk groups, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high-risk group. Discrimination: the pooled c-statistic of the seven external validation studies for the index score was 0.65 (95% CI 0.60 to 0.70); 95% PI 0.51 to 0.77. AUTHORS' CONCLUSIONS Despite the large number of published studies of prognostic models for OS, PFS or TFS for newly-diagnosed, untreated adults with CLL, only a minority of these (N = 12) have been externally validated for their respective primary outcome. Three models have undergone sufficient external validation to enable meta-analysis of the model's ability to predict survival outcomes. Lack of reporting prevented us from summarising calibration as recommended. Of the three models, the CLL-IPI shows the best discrimination, despite overestimation. However, performance of the models may change for individuals with CLL who receive improved treatment options, as the models included in this review were tested mostly on retrospective cohorts receiving a traditional treatment regimen. In conclusion, this review shows a clear need to improve the conducting and reporting of both prognostic model development and external validation studies. For prognostic models to be used as tools in clinical practice, the development of the models (and their subsequent validation studies) should adapt to include the latest therapy options to accurately predict performance. Adaptations should be timely.
Collapse
Key Words
- adult
- female
- humans
- male
- age factors
- bias
- biomarkers, tumor
- calibration
- confidence intervals
- discriminant analysis
- disease-free survival
- genes, p53
- genes, p53/genetics
- immunoglobulin heavy chains
- immunoglobulin heavy chains/genetics
- immunoglobulin variable region
- immunoglobulin variable region/genetics
- leukemia, lymphocytic, chronic, b-cell
- leukemia, lymphocytic, chronic, b-cell/mortality
- leukemia, lymphocytic, chronic, b-cell/pathology
- models, theoretical
- neoplasm staging
- prognosis
- progression-free survival
- receptors, antigen, b-cell
- receptors, antigen, b-cell/genetics
- reproducibility of results
- tumor suppressor protein p53
- tumor suppressor protein p53/genetics
Collapse
MESH Headings
- Adult
- Age Factors
- Bias
- Biomarkers, Tumor
- Calibration
- Confidence Intervals
- Discriminant Analysis
- Disease-Free Survival
- Female
- Genes, p53/genetics
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Models, Theoretical
- Neoplasm Staging
- Prognosis
- Progression-Free Survival
- Receptors, Antigen, B-Cell/genetics
- Reproducibility of Results
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Nina Kreuzberger
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Johanna Aag Damen
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Angela Aldin
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Umlauff
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | - Karel Gm Moons
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Farid Foroutan
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Karl-Anton Kreuzer
- Center of Integrated Oncology Cologne-Bonn, Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
15
|
Allegra A, Musolino C, Tonacci A, Pioggia G, Casciaro M, Gangemi S. Clinico-Biological Implications of Modified Levels of Cytokines in Chronic Lymphocytic Leukemia: A Possible Therapeutic Role. Cancers (Basel) 2020; 12:cancers12020524. [PMID: 32102441 PMCID: PMC7072434 DOI: 10.3390/cancers12020524] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/08/2020] [Accepted: 02/22/2020] [Indexed: 12/19/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is the main cause of mortality among hematologic diseases in Western nations. B-CLL is correlated with an intense alteration of the immune system. The altered functions of innate immune elements and adaptive immune factors are interconnected in B-CLL and are decisive for its onset, evolution, and therapeutic response. Modifications in the cytokine balance could support the growth of the leukemic clone via a modulation of cellular proliferation and apoptosis, as some cytokines have been reported to be able to affect the life of B-CLL cells in vivo. In this review, we will examine the role played by cytokines in the cellular dynamics of B-CLL patients, interpret the contradictions sometimes present in the literature regarding their action, and evaluate the possibility of manipulating their production in order to intervene in the natural history of the disease.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Marco Casciaro
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
- Correspondence:
| |
Collapse
|
16
|
Abstract
Previous studies have shown that interleukin-24 (IL-24) has tumor-suppressing activity by multiple pathways. However, the immunogenicity moderation effect of IL-24 on malignant cells has not been explored extensively. In this study, we investigated the role of IL-24 in immunogenicity modulation of the myelogenous leukemia cells. Data show that myelogenous leukemia cells express low levels of immunogenicity molecules. Treatment with IL-24 could enhance leukemia cell immunogenicity, predominantly regulate leukemia cells to produce immune-associated cytokines, and improve the cytotoxic sensitivity of these cells to immune effector cells. IL-24 expression could retard transplanted leukemia cell tumor growth in vivo in athymic nude mice. Moreover, IL-24 had marked effects on downregulating the expression of angiogenesis-related proteins vascular endothelial growth factor, cluster of differentiation (CD) 31, CD34, collagen IV and metastasis-related factors CD147, membrane type-1 matrix metalloproteinase (MMP), and MMP-2 and MMP-9 in transplanted tumors. These findings indicated novel functions of this antitumor gene and characterized IL-24 as a promising agent for further clinical trial for hematologic malignancy immunotherapy.
Collapse
|
17
|
Redondo-Muñoz J, García-Pardo A, Teixidó J. Molecular Players in Hematologic Tumor Cell Trafficking. Front Immunol 2019; 10:156. [PMID: 30787933 PMCID: PMC6372527 DOI: 10.3389/fimmu.2019.00156] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4β1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4β1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.
Collapse
Affiliation(s)
- Javier Redondo-Muñoz
- Department of Immunology, Ophthalmology and ERL, Hospital 12 de Octubre Health Research Institute (imas12), School of Medicine, Complutense University, Madrid, Spain.,Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Angeles García-Pardo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
18
|
Gao C, Zhou C, Zhuang J, Liu L, Wei J, Liu C, Li H, Sun C. Identification of key candidate genes and miRNA‑mRNA target pairs in chronic lymphocytic leukemia by integrated bioinformatics analysis. Mol Med Rep 2018; 19:362-374. [PMID: 30431072 PMCID: PMC6297738 DOI: 10.3892/mmr.2018.9636] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignant clonal proliferative disorder of B cells. Inhibition of cell apoptosis and cell cycle arrest are the main pathological causes of this disease, but its molecular mechanism requires further investigation. The purpose of the present study was to identify biomarkers for the early diagnosis and treatment of CLL, and to explore the molecular mechanisms of CLL progression. A total of 488 differentially expressed genes (DEGs) and 32 differentially expressed microRNAs (miRNAs; DEMs) for CLL were identified by analyzing the gene chips GSE22529, GSE39411 and GSE62137. Functional and pathway enrichment analyses of DEGs demonstrated that DEGs were mainly involved in transcriptional dysregulation and multiple signaling pathways, such as the nuclear factor‑κB and mitogen‑activated protein kinase signaling pathways. In addition, Cytoscape software was used to visualize the protein‑protein interactions of these DEGs in order to identify hub genes, which could be used as biomarkers for the early diagnosis and treatment of CLL. Cytoscape software was also used to analyze the association between the predicted target mRNAs of DEMs and DEGs and increase knowledge about the miRNA‑mRNA regulatory network associated with the progression of CLL. Taken together, the present study provided a bioinformatics basis for advancing our understanding of the pathogenesis of CLL by identifying differentially expressed hub genes, miRNA‑mRNA target pairs and molecular pathways. In addition, hub genes may be used as novel biomarkers for the diagnosis of CLL and to guide the selection of CLL drug combinations.
Collapse
Affiliation(s)
- Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Chao Zhou
- Cancer Center, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Jing Zhuang
- Cancer Center, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Lijuan Liu
- Cancer Center, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Junyu Wei
- Cancer Center, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Changgang Sun
- Cancer Center, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
19
|
Nesseler JP, Schaue D, McBride WH, Nickers P. [Inflammatory and immune biomarkers of radiation response]. Cancer Radiother 2018; 22:180-192. [PMID: 29650389 DOI: 10.1016/j.canrad.2017.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
In radiotherapy, the treatment is adapted to each individual to protect healthy tissues but delivers most of time a standard dose according to the tumor histology and site. The only biomarkers studied to individualize the treatment are the HPV status with radiation dose de-escalation strategies, and tumor hypoxia with dose escalation to hypoxic subvolumes using FMISO- or FAZA-PET imaging. In the last decades, evidence has grown about the contribution of the immune system to radiation tumor response. Many preclinical studies have identified some of the mechanisms involved. In this context, we have realised a systematic review to highlight potential inflammatory and immune biomarkers of radiotherapy response. Some are inside the tumor microenvironment, as lymphocyte infiltration or PD-L1 expression, others are circulating biomarkers, including different types of hematological cells, cytokines and chemokines.
Collapse
Affiliation(s)
- J P Nesseler
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis.
| | - D Schaue
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - W H McBride
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - P Nickers
- Départment de radiothérapie, centre François-Baclesse, rue Émile-Mayrisch, 4240 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
20
|
Zhu F, McCaw L, Spaner DE, Gorczynski RM. Targeting the IL-17/IL-6 axis can alter growth of Chronic Lymphocytic Leukemia in vivo/in vitro. Leuk Res 2018; 66:28-38. [PMID: 29353760 DOI: 10.1016/j.leukres.2018.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/19/2017] [Accepted: 01/14/2018] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) is critical to the longevity of tumor B cells in chronic lymphocytic leukemia (CLL). Bone marrow mesenchymal stem cells (BMMSCs) and the cytokines they produce including IL-6 are important components of the TME in CLL. We found BMMSCs supported the survival of CLL cells in vitro through an IL-6 dependent mechanism. IL-17 which induces IL-6 generation in a variety of cells increased production of IL-6 both in CLL cells and BMMSCs in vitro. In a xenograft CLL mouse model, BMMSCs and the culture supernatant of BMMSCs increased engraftment of CLL cells through an IL-6 mediated mechanism with human recombinant IL-6 showing similar effects in vivo. Human recombinant IL-17 treatment also increased CLL engraftment in mice through an IL-6 mediated mechanism. Plasma of CLL patients showed elevated levels of both IL-6 and IL-17 by ELISA compared with healthy controls, with levels of IL-6 linearly correlated with IL-17 levels. CLL patients requiring fludarabine based chemotherapy expressed higher levels of IL-6 and IL-17, while CLL patients with the lowest levels of IgA/IgM had higher levels of IL-6, but not IL-17. These data imply an important role for the IL-17/IL-6 axis in CLL which could be therapeutic targets.
Collapse
Affiliation(s)
- Fang Zhu
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Transplant Research Division, Toronto General Hospital, Toronto, Canada
| | - Lindsay McCaw
- Biology Platform, Sunnybrook Research Institute, Toronto, Canada
| | - David E Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, Canada; Dept. of Medical Biophysics, University of Toronto, Toronto, Canada; Dept. of Immunology, University of Toronto, Toronto, Canada
| | - Reginald M Gorczynski
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Transplant Research Division, Toronto General Hospital, Toronto, Canada; Dept. of Immunology, University of Toronto, Toronto, Canada.
| |
Collapse
|
21
|
Kim W, Pyo J, Noh BJ, Jeong JW, Lee J, Kim JE. CCAR2 negatively regulates IL-8 production in cervical cancer cells. Oncotarget 2017; 9:1143-1155. [PMID: 29416683 PMCID: PMC5787426 DOI: 10.18632/oncotarget.23199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/27/2017] [Indexed: 01/10/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2) is a multifaceted protein that controls diverse cellular functions; however, its function in cancer is unclear. To better understand its potential role in cancer, we examined gene expression patterns regulated by CCAR2 in cervical cancer cells. Cytokine and chemokine production by CCAR2-deficient cells increased under oxidative conditions. In particular, H2O2-treated CCAR2-depleted cells showed a significant increase in interleukin-8 (IL-8) production, indicating a negative regulation of IL-8 by CCAR2. Upregulation of IL-8 expression in CCAR2-deficient cells occurred via activation of transcription factor AP-1. The negative correlation between CCAR2 and IL-8 expression was confirmed by examining mRNA and protein levels in tissues from cervical cancer patients. Furthermore, CCAR2-regulated IL-8 expression is associated with a shorter survival of cervical cancer patients. Overall, the data suggest that CCAR2 plays a critical role in controlling both the cancer secretome and cancer progression.
Collapse
Affiliation(s)
- Wootae Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehyuk Pyo
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Byeong-Joo Noh
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhie Lee
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
22
|
Revisiting the role of interleukin-8 in chronic lymphocytic leukemia. Sci Rep 2017; 7:15714. [PMID: 29146966 PMCID: PMC5691131 DOI: 10.1038/s41598-017-15953-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022] Open
Abstract
The proliferation and survival of malignant B cells in chronic lymphocytic leukemia (CLL) depend on signals from the microenvironment in lymphoid tissues. Among a plethora of soluble factors, IL-8 has been considered one of the most relevant to support CLL B cell progression in an autocrine fashion, even though the expression of IL-8 receptors, CXCR1 and CXCR2, on leukemic B cells has not been reported. Here we show that circulating CLL B cells neither express CXCR1 or CXCR2 nor they respond to exogenous IL-8 when cultured in vitro alone or in the presence of monocytes/nurse-like cells. By intracellular staining and ELISA we show that highly purified CLL B cells do not produce IL-8 spontaneously or upon activation through the B cell receptor. By contrast, we found that a minor proportion (<0.5%) of contaminating monocytes in enriched suspensions of leukemic cells might be the actual source of IL-8 due to their strong capacity to release this cytokine. Altogether our results indicate that CLL B cells are not able to secrete or respond to IL-8 and highlight the importance of methodological details in in vitro experiments.
Collapse
|
23
|
Marini C, Bruno S, Fiz F, Campi C, Piva R, Cutrona G, Matis S, Nieri A, Miglino M, Ibatici A, Maria Orengo A, Maria Massone A, Neumaier CE, Totero DD, Giannoni P, Bauckneht M, Pennone M, Tenca C, Gugiatti E, Bellini A, Borra A, Tedone E, Efetürk H, Rosa F, Emionite L, Cilli M, Bagnara D, Brucato V, Bruzzi P, Piana M, Fais F, Sambuceti G. Functional Activation of Osteoclast Commitment in Chronic Lymphocytic Leukaemia: a Possible Role for RANK/RANKL Pathway. Sci Rep 2017; 7:14159. [PMID: 29074954 PMCID: PMC5658396 DOI: 10.1038/s41598-017-12761-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/14/2017] [Indexed: 01/18/2023] Open
Abstract
Skeletal erosion has been found to represent an independent prognostic indicator in patients with advanced stages of chronic lymphocytic leukaemia (CLL). Whether this phenomenon also occurs in early CLL phases and its underlying mechanisms have yet to be fully elucidated. In this study, we prospectively enrolled 36 consecutive treatment-naïve patients to analyse skeletal structure and bone marrow distribution using a computational approach to PET/CT images. This evaluation was combined with the analysis of RANK/RANKL loop activation in the leukemic clone, given recent reports on its role in CLL progression. Bone erosion was particularly evident in long bone shafts, progressively increased from Binet stage A to Binet stage C, and was correlated with both local expansion of metabolically active bone marrow documented by FDG uptake and with the number of RANKL + cells present in the circulating blood. In immune-deficient NOD/Shi-scid, γcnull (NSG) mice, administration of CLL cells caused an appreciable compact bone erosion that was prevented by Denosumab. CLL cell proliferation in vitro correlated with RANK expression and was impaired by Denosumab-mediated disruption of the RANK/RANKL loop. This study suggests an interaction between CLL cells and stromal elements able to simultaneously impair bone structure and increase proliferating potential of leukemic clone.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Bioimages and Molecular Physiology, Milan, Italy.
- Nuclear Medicine IRCCS AOU San Martino-IST, Genoa, Italy.
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy.
| | - Silvia Bruno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Francesco Fiz
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
- Nuclear Medicine Unit, Department of Radiology, Uni-Klinikum Tuebingen, Germany
| | | | - Roberta Piva
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | | | - Serena Matis
- Molecular Pathology, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Alberto Nieri
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | - Maurizio Miglino
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | | | | | | | | | - Paolo Giannoni
- Stem Cell Laboratory, Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Matteo Bauckneht
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | | | - Claudya Tenca
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Elena Gugiatti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Alessandro Bellini
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | - Anna Borra
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | | | - Hülya Efetürk
- Nuclear Medicine IRCCS AOU San Martino-IST, Genoa, Italy
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| | | | - Laura Emionite
- Animal Facility, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Michele Cilli
- Animal Facility, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Davide Bagnara
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Karches Center for Oncology Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Valerio Brucato
- Department of Civil, Environmental, Aerospace, Materials Engineering, Aten Center, CHAB pole, University of Palermo, Viale delle Scienze, 6, Palermo, 90128, Italy
| | - Paolo Bruzzi
- Epidemiology and Clinical trial Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Michele Piana
- SPIN Institute, CNR, Genoa, Italy
- Department of Mathematics (DIMA), University of Genoa, Genoa, Italy
| | - Franco Fais
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Molecular Pathology, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine IRCCS AOU San Martino-IST, Genoa, Italy
- Nuclear Medicine, Department of Health Science, University of Genova, Genoa, Italy
| |
Collapse
|
24
|
Crassini K, Shen Y, Mulligan S, Giles Best O. Modeling the chronic lymphocytic leukemia microenvironment in vitro. Leuk Lymphoma 2016; 58:266-279. [PMID: 27756161 DOI: 10.1080/10428194.2016.1204654] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Microenvironments within the lymph node and bone marrow promote proliferation and drug resistance in chronic lymphocytic leukemia (CLL). Successful treatment of CLL must therefore target the leukemic cells within these compartments. A better understanding of the interaction between CLL cells and the tumor microenvironment has led to the development of in vitro models that mimic the mechanisms that support leukemic cell survival and proliferation in vivo. Employing these models as part of the pre-clinical evaluation of novel therapeutic agents enables a better approximation of their potential clinical efficacy. In this review we summarize the current literature describing how different aspects of the tumor microenvironment have been modeled in vitro and detail how these models have been employed to study the biology of the disease and potential efficacy of novel therapeutic agents.
Collapse
Affiliation(s)
- Kyle Crassini
- a Northern Blood Research Centre , Kolling Institute of Medical Research, Royal North Shore Hospital , Sydney , Australia
| | - Yandong Shen
- a Northern Blood Research Centre , Kolling Institute of Medical Research, Royal North Shore Hospital , Sydney , Australia
| | - Stephen Mulligan
- a Northern Blood Research Centre , Kolling Institute of Medical Research, Royal North Shore Hospital , Sydney , Australia.,b Chronic Lymphocytic Leukemia Research Consortium (CLLARC) , Australia
| | - O Giles Best
- a Northern Blood Research Centre , Kolling Institute of Medical Research, Royal North Shore Hospital , Sydney , Australia.,b Chronic Lymphocytic Leukemia Research Consortium (CLLARC) , Australia
| |
Collapse
|
25
|
Herman SEM, Wiestner A. Preclinical modeling of novel therapeutics in chronic lymphocytic leukemia: the tools of the trade. Semin Oncol 2016; 43:222-32. [PMID: 27040700 DOI: 10.1053/j.seminoncol.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the last decade our understanding of chronic lymphocytic leukemia (CLL) biology and pathogenesis has increased substantially. These insights have led to the development of several new agents with novel mechanisms of action prompting a change in therapeutic approaches from chemotherapy-based treatments to targeted therapies. Multiple preclinical models for drug development in CLL are available; however, with the advent of these targeted agents, it is becoming clear that not all models and surrogate readouts of efficacy are appropriate for all drugs. In this review we discuss in vitro and in vivo preclinical models, with a particular focus on the benefits and possible pitfalls of different model systems in the evaluation of novel therapeutics for the treatment of CLL.
Collapse
Affiliation(s)
- Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Awasthi A, Ayello J, Van de Ven C, Elmacken M, Sabulski A, Barth MJ, Czuczman MS, Islam H, Klein C, Cairo MS. Obinutuzumab (GA101) compared to rituximab significantly enhances cell death and antibody-dependent cytotoxicity and improves overall survival against CD20+ rituximab-sensitive/-resistant Burkitt lymphoma (BL) and precursor B-acute lymphoblastic leukaemia. Br J Haematol 2015; 171:763-75. [DOI: 10.1111/bjh.13764] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/22/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Aradhana Awasthi
- Department of Pediatrics; New York Medical College; Valhalla NY USA
| | - Janet Ayello
- Department of Pediatrics; New York Medical College; Valhalla NY USA
| | | | - Mona Elmacken
- Department of Pediatrics; New York Medical College; Valhalla NY USA
| | - Anthony Sabulski
- Department of Pediatrics; New York Medical College; Valhalla NY USA
| | - Matthew J. Barth
- Department of Medicine; Roswell Park Cancer Institute; Buffalo NY USA
| | - Myron S. Czuczman
- Department of Medicine; Roswell Park Cancer Institute; Buffalo NY USA
| | - Humayun Islam
- Department of Pathology; New York Medical College; Valhalla NY USA
| | - Christian Klein
- Roche Pharmaceutical Research & Early Development; Roche Innovation Centre; Zurich Switzerland
| | - Mitchell S. Cairo
- Department of Pediatrics; New York Medical College; Valhalla NY USA
- Department of Pathology; New York Medical College; Valhalla NY USA
- Department of Medicine; New York Medical College; Valhalla NY USA
- Department of Microbiology & Immunology; New York Medical College; Valhalla NY USA
- Department of Cell Biology & Anatomy; New York Medical College; Valhalla NY USA
| |
Collapse
|
27
|
Bernard S, Danglade D, Gardano L, Laguillier C, Lazarian G, Roger C, Thieblemont C, Marzec J, Gribben J, Cymbalista F, Varin-Blank N, Ledoux D, Baran-Marszak F. Inhibitors of BCR signalling interrupt the survival signal mediated by the micro-environment in mantle cell lymphoma. Int J Cancer 2014; 136:2761-74. [DOI: 10.1002/ijc.29326] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 10/27/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Sophie Bernard
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Damien Danglade
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Laura Gardano
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Christelle Laguillier
- Service de Biochimie, Hôpital Jean Verdier, Assistance Publique-Hôpitaux de Paris; Bondy France
| | - Gregory Lazarian
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Claudine Roger
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Catherine Thieblemont
- Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris; Paris France
| | - Jacek Marzec
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - John Gribben
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - Florence Cymbalista
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Nadine Varin-Blank
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Dominique Ledoux
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Fanny Baran-Marszak
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| |
Collapse
|
28
|
Perbellini O, Cioffi F, Malpeli G, Zanolin E, Lovato O, Scarpa A, Pizzolo G, Scupoli MT. Up-regulation of CXCL8/interleukin-8 production in response to CXCL12 in chronic lymphocytic leukemia. Leuk Lymphoma 2014; 56:1897-900. [DOI: 10.3109/10428194.2014.977889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
29
|
Visconti L, Nelissen K, Deckx L, van den Akker M, Adriaensen W, Daniels L, Matheï C, Linsen L, Hellings N, Stinissen P, Buntinx F. Prognostic value of circulating cytokines on overall survival and disease-free survival in cancer patients. Biomark Med 2014; 8:297-306. [PMID: 24521026 DOI: 10.2217/bmm.13.122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Through their tumor-promoting and/or tumor-suppressive properties, cytokines can influence progression of cancer. We systematically reviewed the current literature on the prognostic value of the circulating cytokines IL-1α/β, IL-6, IL-8, IL-10, IL-12, TNF-α, TGF-β and IFN-γ to predict overall and disease-free survival in any type of cancer patients. PubMed was systematically searched and based on eligibility assessment using our five criteria of the Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK) checklist, six unique studies were reviewed. Elevated IL-6 and IL-10 levels seem independently associated with worse prognosis in terms of overall and disease-free survival. The prognostic value of IL-1α/β, IL-8, IL-12, TNF-α, TGF-β and IFN-γ could not be demonstrated. The small number of selected studies underlines the need for large well-designed prospective studies, using the REMARK checklist as a guideline, to determine which cytokines have prognostic value on survival in cancer patients.
Collapse
Affiliation(s)
- Laura Visconti
- Faculty of Medicine & Life Sciences, Hasselt University, Campus Diepenbeek, Martelarenlaan 42, 3500 Hasselt, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pede V, Rombout A, Vermeire J, Naessens E, Vanderstraeten H, Philippé J, Verhasselt B. Expression of ZAP70 in chronic lymphocytic leukaemia activates NF-κB signalling. Br J Haematol 2013; 163:621-30. [DOI: 10.1111/bjh.12588] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 08/28/2013] [Indexed: 12/01/2022]
Affiliation(s)
- Valerie Pede
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Ans Rombout
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Jolien Vermeire
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Evelien Naessens
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Hanne Vanderstraeten
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Jan Philippé
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| | - Bruno Verhasselt
- Department of Clinical Chemistry, Microbiology and Immunology; Ghent University; Ghent Belgium
| |
Collapse
|
31
|
Ahn JS, Lee KH, Sun JM, Park K, Kang ES, Cho EK, Lee DH, Kim SW, Lee GW, Kang JH, Lee JS, Lee JW, Ahn MJ. A randomized, phase II study of vandetanib maintenance for advanced or metastatic non-small-cell lung cancer following first-line platinum-doublet chemotherapy. Lung Cancer 2013; 82:455-60. [PMID: 24075125 DOI: 10.1016/j.lungcan.2013.08.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 08/23/2013] [Accepted: 08/31/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND This randomized, phase II study investigated whether benefit could be obtained by giving vandetanib, an oral inhibitor of vascular endothelial and epithelial growth factor receptor, as a maintenance treatment in non-small cell lung cancer (NSCLC). METHODS Patients were randomly assigned to either vandetanib or placebo after completion of 4 cycles of first-line chemotherapy. A progression-free survival (PFS) rate at 3 months was selected as the primary endpoint. We set a maximum PFS rate at 3 months to 30% (null hypothesis), and a minimum PFS rate at 3 months to 50% (alternative hypothesis). RESULTS At the interim analysis, 9 of 24 patients in the vandetanib arm were progression-free at 3 months, whereas 7 of 24 in the placebo arm were progression-free. The placebo arm was closed at the first stage. The vandetanib arm proceeded to the second stage, and recruited a total of 75 patients. At the second stage, 28 out of 63 evaluable patients receiving vandetanib achieved PFS at 3 months. The alternative hypothesis that the PFS rate at 3 months is at least 50% was accepted. The median PFS was 2.7 months (95% CI, 1.9-4.4 months) in the vandetanib arm and 1.7 months (95% CI, 0.9-2.6 months) in the placebo arm. The most common adverse events in patients receiving vandetanib were rash (77.3%) and diarrhea (60.0%). CONCLUSIONS Maintenance therapy with vandetanib for patients with NSCLC after standard platinum doublet chemotherapy is well tolerated and may prolong PFS compared with placebo, and needs additional investigation.
Collapse
Affiliation(s)
- Jin Seok Ahn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University, 50 Irwon-dong, Gangnam-gu, Seoul 135-710, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lafarge ST, Johnston JB, Gibson SB, Marshall AJ. Adhesion of ZAP-70+ chronic lymphocytic leukemia cells to stromal cells is enhanced by cytokines and blocked by inhibitors of the PI3-kinase pathway. Leuk Res 2013; 38:109-15. [PMID: 23981382 DOI: 10.1016/j.leukres.2013.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/30/2013] [Indexed: 01/05/2023]
Abstract
CLL cell survival and proliferation is enhanced through direct contact with supporting cells present in lymphoid tissues. PI3Ks are critical signal transduction enzymes controlling B cell survival and activation. PI3K inhibitors have entered clinical trials and show promising therapeutic activity; however, it is unclear whether PI3K inhibitor drugs differentially affect ZAP-70 positive versus negative CLL cells or target specific microenvironmental interactions. Here we provide evidence that CD40L+IL-4, IL-8 or IL-6 enhance adhesion to stromal cells, with IL-6 showing a selective effect on ZAP-70 positive cells. Stimulatory effects of IL-8 or IL-6 are fully reversed by PI3K inhibition, while the effects of CD40L+IL-4 are partially reversed. While CD40L+IL-4 is the only stimulation increasing CLL cell survival for all patient groups, IL-6 protects ZAP-70 positive cells from cell death induced by PI3K inhibition. Altogether, our results indicate that targeting the PI3K pathway can reverse protective CLL-microenvironment interactions in both ZAP-70 positive and negative CLL despite their differences in cytokine responsiveness.
Collapse
Affiliation(s)
- Sandrine T Lafarge
- University of Manitoba, Department of Immunology, Winnipeg, MB, Canada; Cancercare Manitoba, Manitoba Institute of Cell Biology, Winnipeg, MB, Canada
| | - James B Johnston
- Cancercare Manitoba, Manitoba Institute of Cell Biology, Winnipeg, MB, Canada
| | - Spencer B Gibson
- University of Manitoba, Department of Immunology, Winnipeg, MB, Canada; Cancercare Manitoba, Manitoba Institute of Cell Biology, Winnipeg, MB, Canada; University of Manitoba, Department of Biochemistry and Medical Genetics, Winnipeg, MB, Canada
| | - Aaron J Marshall
- University of Manitoba, Department of Immunology, Winnipeg, MB, Canada; University of Manitoba, Department of Biochemistry and Medical Genetics, Winnipeg, MB, Canada.
| |
Collapse
|
33
|
Siddon AJ, Rinder HM. Pathology consultation on evaluating prognosis in incidental monoclonal lymphocytosis and chronic lymphocytic leukemia. Am J Clin Pathol 2013; 139:708-12. [PMID: 23690112 DOI: 10.1309/ajcplir4gzwx3xka] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a monoclonal B-cell lymphoproliferative disorder generally characterized by an indolent clinical course. However, some patients with CLL will have more aggressive disease progression, and identifying that subgroup may be important for early, or perhaps more aggressive, intervention. In addition, monoclonal B-cell lymphocytosis is often found on routine laboratory evaluation, and it is important to distinguish this entity from overt CLL. Moreover, since many patients with CLL are discovered incidentally and before significant disease progression, prognostic laboratory evaluation may become increasingly efficacious as therapeutic options replace the older strategy of expectant observation. Prognostication may be especially critical if it correctly identifies patients with early stage CLL who are at high risk of clonal evolution and/ or resistance to chemoimmunotherapy. Laboratory studies include surface CD38 and intracellular ZAP-70 expression by flow cytometry, serum β2-microglobulin, and immunoglobulin heavy-chain variable gene mutational status. Cytogenetics for targeted chromosome alterations may similarly aid in predicting outcome and guiding early intervention. This article concisely reviews the utility of commonly performed prognostic markers and addresses the laboratory evaluation in patients with incidentally discovered early stage CLL.
Collapse
|
34
|
Filip AA, Ciseł B, Koczkodaj D, Wąsik-Szczepanek E, Piersiak T, Dmoszyńska A. Circulating microenvironment of CLL: are nurse-like cells related to tumor-associated macrophages? Blood Cells Mol Dis 2013; 50:263-70. [PMID: 23313631 DOI: 10.1016/j.bcmd.2012.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/12/2012] [Indexed: 11/28/2022]
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is one of the most common hematologic malignancies in Western countries. Accumulation of leukemic lymphocytes in peripheral blood, bone marrow and secondary lymphatic organs of CLL patients is due to decreased apoptosis rather than to increased proliferation. The former is driven by signals from a specific microenvironment, created by stromal cells of mesenchymal origin, follicular dendritic cells, T lymphocytes and others. Nurse-like cells (NLCs) were first described to differentiate from peripheral blood mononuclear cells of CLL patients in vitro, then they have been also found in proliferation centers of their lymphatic tissues. Like tumor-associated macrophages (TAMs) in solid tumors, nurse-like cells promote survival of CLL lymphocytes. NLC gene expression patterns suggest their similarity to TAMs and differ between patients depending on ZAP70 protein expression status. NLC number in vitro corresponds with CD14 expressing cell count and beta-2-microglobulin serum level, and positively correlates with leukemic lymphocyte viability. As NLCs strongly express genes for adhesion molecules and secrete chemokines of antiapoptotic activity, they should be considered as a target for anti-microenvironment therapy of this incurable disease.
Collapse
Affiliation(s)
- Agata A Filip
- Department of Cancer Genetics, Medical University of Lublin, Radziwillowska 11, 20-080 Lublin, Poland.
| | | | | | | | | | | |
Collapse
|
35
|
Endogenous IL-8 acts as a CD16 co-activator for natural killer-mediated anti-CD20 B cell depletion in chronic lymphocytic leukemia. Leuk Res 2012; 37:440-6. [PMID: 23259986 DOI: 10.1016/j.leukres.2012.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 10/23/2012] [Accepted: 11/25/2012] [Indexed: 11/21/2022]
Abstract
Rituximab (RTX, anti-CD20 antibody) combined with chemotherapy is currently standard treatment for chronic lymphocytic leukemia (CLL). Serum level of IL-8 is a prognostic factor for CLL that correlates with disease stage. We investigated whether endogenous IL-8 affects RTX or Obinutuzumab (GA101) B-leukemic depletion mediated by natural killers (NK). Using whole peripheral blood lymphocytes from untreated CLL patients, RTX, but most significantly GA101, were effective in B-cell depletion and NK activation. IL-8 inhibition completely inhibited B-cell depletion by RTX and reduced GA101-induced B-cell depletion. Altogether results underline IL-8 as an endogenous NK co-activator and confirm GA101 therapeutic potential for CLL treatment.
Collapse
|
36
|
Schmiedel BJ, Scheible CA, Nuebling T, Kopp HG, Wirths S, Azuma M, Schneider P, Jung G, Grosse-Hovest L, Salih HR. RANKL expression, function, and therapeutic targeting in multiple myeloma and chronic lymphocytic leukemia. Cancer Res 2012; 73:683-94. [PMID: 23139212 DOI: 10.1158/0008-5472.can-12-2280] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bone destruction is a prominent feature of multiple myeloma, but conflicting data exist on the expression and pathophysiologic involvement of the bone remodeling ligand RANKL in this disease and the potential therapeutic benefits of its targeted inhibition. Here, we show that RANKL is expressed by primary multiple myeloma and chronic lymphocytic leukemia (CLL) cells, whereas release of soluble RANKL was observed exclusively with multiple myeloma cells and was strongly influenced by posttranscriptional/posttranslational regulation. Signaling via RANKL into multiple myeloma and CLL cells induced release of cytokines involved in disease pathophysiology. Both the effects of RANKL on osteoclastogenesis and cytokine production by malignant cells could be blocked by disruption of RANK-RANKL interaction with denosumab. As we aimed to combine neutralization of RANKL with induction of antibody-dependent cellular cytotoxicity of natural killer (NK) cells against RANKL-expressing malignant cells and as denosumab does not stimulate NK reactivity, we generated RANK-Fc fusion proteins with modified Fc moieties. The latter displayed similar capacity compared with denosumab to neutralize the effects of RANKL on osteoclastogenesis in vitro, but also potently stimulated NK cell reactivity against primary RANKL-expressing malignant B cells, which was dependent on their engineered affinity to CD16. Our findings introduce Fc-optimized RANK-Ig fusion proteins as attractive tools to neutralize the detrimental function of RANKL while at the same time potently stimulating NK cell antitumor immunity.
Collapse
|
37
|
Yoon JY, Lafarge S, Dawe D, Lakhi S, Kumar R, Morales C, Marshall A, Gibson SB, Johnston JB. Association of interleukin-6 and interleukin-8 with poor prognosis in elderly patients with chronic lymphocytic leukemia. Leuk Lymphoma 2012; 53:1735-42. [DOI: 10.3109/10428194.2012.666662] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Abstract
Chronic lymphocytic leukemia (CLL) is an indolent lymphoproliferative disorder characterized by both circulating peripheral disease as well as involvement of the lymph nodes and bone marrow. Increasing evidence suggests that the stromal microenvironment provides anti-apoptotic and pro-survival signals to CLL cells, and may contribute significantly to resistance to a wide variety of treatments. Our understanding of the complex interactions involved in CLL cell trafficking continues to grow. Chemokines and corresponding chemokine receptors are key factors for organizing CLL cell trafficking and homing and the complex cellular interactions between CLL and accessory cells. Important chemokines include CCL3, CCL4, and CCL22, which are released by CLL cells, and CXCL12, CXCL13, CXCL9, 10, 11, CCL 19, and CCL21, which are constitutively secreted by various stromal cells. Integrins such as VLA-4 (CD49d) as well as selectins and CD44 also likely play a role in directing CLL cell migration within the tissue microenvironments. Data are also emerging that other molecules such as MMP-9 and cytoskeletal proteins also contribute to CLL cell trafficking. Though this interplay is complex, it is critical that we improve our understanding of CLL cell trafficking to facilitate the development of novel therapies that target these pathways. Several drugs in clinical development, such as CXCR4 antagonists and PI3K, Btk, and Syk inhibitors appear to modulate CLL cell trafficking and CLL-stroma interactions. Here, we review the current understanding of the molecular interactions that underlie CLL cell trafficking and we highlight some of the promising approaches underway to target these pathways therapeutically in CLL.
Collapse
Affiliation(s)
- Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
39
|
Buechele C, Baessler T, Schmiedel BJ, Schumacher CE, Grosse-Hovest L, Rittig K, Salih HR. 4-1BB ligand modulates direct and Rituximab-induced NK-cell reactivity in chronic lymphocytic leukemia. Eur J Immunol 2011; 42:737-48. [PMID: 22144129 DOI: 10.1002/eji.201141920] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 11/04/2011] [Accepted: 11/21/2011] [Indexed: 12/17/2022]
Abstract
NK cells play an important role in tumor immunosurveillance and largely contribute to the therapeutic success of anti-tumor antibodies like Rituximab. Here, we studied the role of the TNF family member 4-1BB ligand (4-1BBL) during the interaction of NK cells with chronic lymphocytic leukemia (CLL) cells. 4-1BBL was highly expressed on patient B-CLL cells in all 56 investigated cases. Signaling via 4-1BBL following interaction with 4-1BB, which was detected on NK cells of CLL patients but not healthy individuals, led to the release of immunoregulatory cytokines including TNF by CLL cells. CLL patient sera contained elevated levels of TNF and induced 4-1BB upregulation on NK cells, which in turn impaired direct and Rituximab-induced NK-cell reactivity against 4-1BBL-expressing targets. NK-cell reactivity was not only enhanced by blocking the interaction of NK cell-expressed 4-1BB with 4-1BBL expressed by CLL cells, but also by preventing 4-1BB upregulation on NK cells via neutralization of TNF in patient serum with Infliximab. Our data indicate that 4-1BBL mediates NK-cell immunosubversion in CLL, and thus might contribute to the reportedly compromised efficacy of Rituximab to induce NK-cell reactivity in the disease, and that TNF neutralization may serve to enhance the efficacy of Rituximab treatment in CLL.
Collapse
Affiliation(s)
- Corina Buechele
- Department of Hematology, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Buechele C, Baessler T, Wirths S, Schmohl JU, Schmiedel BJ, Salih HR. Glucocorticoid-induced TNFR-related protein (GITR) ligand modulates cytokine release and NK cell reactivity in chronic lymphocytic leukemia (CLL). Leukemia 2011; 26:991-1000. [PMID: 22064350 DOI: 10.1038/leu.2011.313] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Natural killer (NK) cells play an important role in the immunosurveillance of hematopoietic malignancies. Their reactivity is influenced by activating and inhibitory signals mediated by tumor-expressed ligands for NK receptors. Many members of the tumor necrosis factor (TNF) family modulate differentiation, proliferation, activation and death of both tumor and immune effector cells. The TNF receptor family member glucocorticoid-induced TNFR-related protein (GITR) stimulates anti-tumor immunity in mice, but available data indicate that GITR may mediate different effects in mice and men and impairs the reactivity of human NK cells. Here, we comprehensively studied the expression and function of GITR ligand (GITRL) in leukemia. Among the different leukemia entities, pronounced expression of GITRL on leukemic cells was observed in chronic lymphocytic leukemia (CLL), and the GITR receptor was expressed at significantly higher levels on NK cells of CLL patients compared with healthy controls. Upon GITR-GITRL interaction, signaling via GITRL into the leukemia cells induced the release of interleukin (IL)-6, IL-8 and TNF, which act as growth and survival factors for CLL cells. In addition, GITRL impaired both direct and Rituximab-induced degranulation, cytotoxicity and interferon-γ production of NK cells, which could be restored by GITR blocking antibodies. Thus, GITRL may contribute to disease pathophysiology and resistance to direct and Rituximab-induced NK reactivity in CLL.
Collapse
Affiliation(s)
- C Buechele
- Department of Hematology and Oncology, Eberhard Karls-University, Tuebingen, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Chronic lymphocytic leukemia cells induce anti-apoptotic effects of bone marrow stroma. Ann Hematol 2011; 90:1381-90. [DOI: 10.1007/s00277-011-1218-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 03/14/2011] [Indexed: 11/25/2022]
|
42
|
de Vasconcellos JF, Laranjeira ABA, Zanchin NIT, Otubo R, Vaz TH, Cardoso AA, Brandalise SR, Yunes JA. Increased CCL2 and IL-8 in the bone marrow microenvironment in acute lymphoblastic leukemia. Pediatr Blood Cancer 2011; 56:568-77. [PMID: 21298741 DOI: 10.1002/pbc.22941] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 11/02/2010] [Indexed: 02/01/2023]
Abstract
BACKGROUND The interactions of acute lymphoblastic leukemia (ALL) blasts with bone marrow (BM) stromal cells have a positive impact on leukemia cell survival. In the present study, we proposed to identify and investigate the role of molecules critically involved in leukemia--microenvironment crosstalk. PROCEDURE Gene expression profiling analyses of BM mesenchymal stem cells (BMMSC) were performed following stimulation by ALL cells. CCL2 and IL-8 plasma levels were evaluated from ALL patients and controls. Expression of the CCL2 and IL-8 receptors in ALL was determined by RT-PCR. The biological effects of CCL2, IL-8 or its neutralizing antibodies in primary precursor-B ALL and BMMSC cells were evaluated using in vitro assays. RESULTS Leukemia stimulation of BMMSC upregulated the expression of several inflammatory chemokines, including CCL2 and IL-8. The BM plasma levels of CCL2 and IL-8 in children at diagnosis were significantly higher than in healthy controls (P < 0.001). Functional studies revealed that CCL2 and IL-8 enhanced the capacity of BMMSC to support adhesion of ALL cells. CCL2 and IL-8 were also found to enhance BMMSC survival and to increase their proliferation. ALL cells were not directly affected by CCL2 or IL-8. CONCLUSIONS The leukemic BM microenvironment had increased levels of CCL2 and IL-8. These chemokines are known to have suppressive effects in normal hematopoiesis. Our data indicate that CCL2 and IL-8 have a positive impact on BMMSC survival, proliferation, and adhesiveness to ALL cells. Leukemia-associated CCL2 and IL-8 upregulation may represent one possible mechanism of microenvironment perversion in favor of ALL cells.
Collapse
|
43
|
Shachar I, Haran M. The secret second life of an innocent chaperone: the story of CD74 and B cell/chronic lymphocytic leukemia cell survival. Leuk Lymphoma 2011; 52:1446-54. [PMID: 21417823 DOI: 10.3109/10428194.2011.565437] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This review deals with the cytokine macrophage migration inhibitory factor (MIF) and its receptor, CD74. MIF and CD74 have been shown to regulate peripheral B cell survival and were associated with tumor progression and metastasis. CD74 expression has been suggested to serve as a prognostic factor in many cancers, with higher relative expression of CD74 behaving as a marker of tumor progression. In chronic lymphocytic leukemia (CLL) cells, binding of MIF to CD74 induces nuclear factor-κB (NF-κB) activation and up-regulation of TAp63 expression, resulting in the secretion of interleukin 8 (IL-8), which in turn promotes cell survival. In addition, TAp63 expression elevates expression of the integrin VLA-4, particularly during the advanced stage of the disease. Blocking of CD74, TAp63, or VLA-4 inhibits the in vivo homing of CLL cells to the BM. Thus, CD74 and its target genes, TAp63 and VLA-4, facilitate migration of CLL cells back to the BM, where they interact with the supportive BM environment that helps rescue them from apoptosis. These results are expected to pave the way toward novel therapeutic strategies aimed at interrupting this survival pathway. One such agent, the monocolonal antibody milatuzumab directed at CD74, is already being studied in early clinical trials.
Collapse
Affiliation(s)
- Idit Shachar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
44
|
Tefferi A, Vaidya R, Caramazza D, Finke C, Lasho T, Pardanani A. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: a comprehensive cytokine profiling study. J Clin Oncol 2011; 29:1356-63. [PMID: 21300928 DOI: 10.1200/jco.2010.32.9490] [Citation(s) in RCA: 469] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Abnormal cytokine expression accompanies myelofibrosis and might be a therapeutic target for Janus-associated kinase (JAK) inhibitor drugs. This study describes the spectrum of plasma cytokine abnormalities in primary myelofibrosis (PMF) and examines their phenotypic correlates and prognostic significance. PATIENTS AND METHODS Patients included in this study were required to have archived plasma, bone marrow biopsy, and cytogenetic information available at the time of first referral to the Mayo Clinic. Multiplex biometric sandwich immunoassay was used to measure plasma levels of 30 cytokines. RESULTS In total, 127 PMF patients were studied; comparison with normal controls (n = 35) revealed significantly increased interleukin-1β (IL-1β), IL-1RA, IL-2R, IL-6, IL-8, IL-10, IL-12, IL-13, IL-15, tumor necrosis factor α (TNF-α), granulocyte colony-stimulating factor (G-CSF), interferon alfa (IFN-α), macrophage inflammatory protein 1α (MIP-1α), MIP-1β, hepatocyte growth factor (HGF), IFN-γ-inducible protein 10 (IP-10), monokine induced by IFN-γ (MIG), monocyte chemotactic protein 1 (MCP-1), and vascular endothelial growth factor (VEGF) levels and decreased IFN-γ levels. In treatment-naive patients (n = 90), increased levels of IL-8 (P < .001), IL-2R (P < .001), IL-12 (P < .001), IL-15 (P = .001), and IP-10 (P = .003) were independently predictive of inferior survival. A similar multivariable analysis that included all 127 study patients confirmed the prognostic value of these five cytokines, and IL-8, IL-2R, IL-12, and IL-15 remained significant when risk stratification, according to the recently revised Dynamic International Prognostic Scoring System (DIPSS plus), was added to the multivariable model. Leukemia-free survival was predicted by IL-8, which was also the only cytokine associated with ≥ 1% circulating blasts. Other cytokine-phenotype associations included increased IL-8 and constitutional symptoms; IL-2R, IL-12, and transfusion need; IL-2R, IL-8, and leukocytosis; IP-10 and thrombocytopenia; HGF, MIG, IL-1RA, and marked splenomegaly; and IL-1RA, IL-2R, IP-10, MIP-1β, and JAK2V617F. A two-cytokine (IL-8/IL-2R) -based risk categorization delineated prognostically different groups within specific DIPSS plus risk categories. CONCLUSION This study signifies the presence of specific cytokine-phenotype associations in PMF and a prognostically relevant plasma cytokine signature that might prove useful as a laboratory tool for predicting and monitoring treatment response.
Collapse
|
45
|
Burger JA. Chemokines and chemokine receptors in chronic lymphocytic leukemia (CLL): from understanding the basics towards therapeutic targeting. Semin Cancer Biol 2010; 20:424-30. [PMID: 20883788 DOI: 10.1016/j.semcancer.2010.09.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 09/16/2010] [Accepted: 09/21/2010] [Indexed: 02/03/2023]
Abstract
Chemokines and their receptors organize the recruitment and positioning of cells at each stage of the immune response, a system critically dependent upon coordination to get the right cells to the right place at the right time. Chemokine receptors expressed on CLL B cells are thought to function in a similar fashion, regulating the trafficking of the leukemia cells between blood, lymphoid organs, and the bone marrow, and within sub compartments within these tissues, in concert with adhesion molecules and other guidance cues. CLL cells not only respond to chemokines secreted in the microenvironment, the leukemia cells also secrete chemokines in response to external signals, such as B cell receptor engagement. These CLL cell-derived chemokines facilitate interactions between CLL cells, T cells, and other immune cells that shape the CLL microenvironment. CXCR4, the most prominent chemokine receptor in CLL, is now targeted in a first clinical trial, emphasizing that chemokines and their receptors have become a highly dynamic translational research field.
Collapse
Affiliation(s)
- Jan A Burger
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77230, USA.
| |
Collapse
|
46
|
Dichotomy in NF-kappaB signaling and chemoresistance in immunoglobulin variable heavy-chain-mutated versus unmutated CLL cells upon CD40/TLR9 triggering. Oncogene 2010; 29:5071-82. [PMID: 20581863 DOI: 10.1038/onc.2010.248] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic lymphocytic leukemia (CLL) cells circulating in peripheral blood (PB) differ from the leukemic fraction in lymph nodes (LNs) with respect to cell division and drug sensitivity. CD40 stimulation of PB CLL cells in vitro results in chemoresistance and provides a partial model for the LN microenvironment. The TLR9 ligand CpG induces proliferation in immunoglobulin variable heavy-chain-unmutated CLL, but apoptosis in immunoglobulin variable heavy-chain-mutated CLL. To juxtapose proliferative with antiapoptotic signals, we investigated the effects of CpG in the context of CD40 ligation in mutated versus unmutated CLL cells in this study. Prolonged CD40 ligation induced classical, followed by alternative nuclear factor-kappaB (NF-kappaB), activity in both subgroups, correlating with enhanced Bfl-1 and Bcl-X(L) levels, respectively. A dichotomy in NF-kappaB signaling occurred on combined CD40/TLR9 triggering. This induced declining p52 and Bcl-X(L) levels, and reversed chemoresistance only in mutated cells, whereas unmutated cells proliferated, maintained p52 and Bcl-X(L) and remained chemoresistant. The pivotal contribution of Bcl-X(L) to chemoresistance was shown by the BH3 mimetic ABT-737 and RNA interference. Finally, in ex vivo LN samples, p52, p65 and Bcl-X(L) levels were highly expressed, corroborating the in vitro findings. Thus, a distinction in NF-kappaB activation and drug susceptibility in mutated versus unmutated (LN-like) CLL cells was uncovered, which was causally linked to Bcl-X(L) levels.
Collapse
|
47
|
Cytokines in bronchoalveolar lavage fluid and serum of lung cancer patients during radiotherapy — Association of interleukin-8 and VEGF with survival. Cytokine 2010; 50:30-6. [DOI: 10.1016/j.cyto.2009.11.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 10/25/2009] [Accepted: 11/22/2009] [Indexed: 11/23/2022]
|
48
|
The role of chemokines in B cell chronic lymphocytic leukaemia: pathophysiological aspects and clinical impact. Ann Hematol 2009; 89:437-46. [PMID: 20020127 DOI: 10.1007/s00277-009-0876-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 11/26/2009] [Indexed: 12/17/2022]
Abstract
Chemokines are centrally involved in leukocyte migration, homing and haematopoiesis. Besides these physiological aspects, their role in pathological processes especially with respect to solid tumour and haematological neoplasias is well established. In this context, the focus was set here on disclosing their contribution in B cell chronic lymphocytic leukaemia (B-CLL), which is regarded as the most characteristic low-grade lymphoma. Up to now, it has been demonstrated that several chemokines are involved in migration of B-CLL cells to lymph nodes, secondary lymphoid organs and bone marrow. Moreover, some chemokines are known to have an anti-apoptotic effect and thus contribute to the survival of B-CLL cells. By interfering with both of these aspects, new therapeutic targets for this yet incurable disease may be developed. Furthermore, a correlation can be drawn between the concentration of some chemokines in patients' serum, the expression of their respective receptors on B-CLL cells and well-established predictive clinical parameters. Consequently, further systematic investigation of the chemokine network may lead to the identification of new diagnostic and prognostic markers. This review focuses on the impact of chemokines and their receptors on B-CLL pathophysiology and points out potential implications for both treatment and diagnosis.
Collapse
|
49
|
Mátrai Z, Németh J, Miklós K, Szabó Z, Masszi T. Serum beta2-microglobulin measured by immunonephelometry: expression patterns and reference intervals in healthy adults. Clin Chem Lab Med 2009; 47:585-9. [PMID: 19397485 DOI: 10.1515/cclm.2009.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Serum beta2-microglobulin (beta2m) has been established as a marker of disease activity in malignancies, autoimmune conditions and infections. Despite its important role in prognosis assessment and disease monitoring, relatively few studies are available on its expression in healthy individuals. Furthermore, interpretation of results is hampered by the variety in reference limits due to differences in methodology, sample population and statistics. METHODS Serum beta2m concentrations were measured using a microparticle-enhanced immunonephelometric method in 183 healthy blood donors aged 29-75 years. RESULTS The median beta2m concentration was 1.67 (0.88-2.75) mg/L with no difference between men and women (1.71 mg/L vs. 1.62 mg/L, p<0.07). A linear correlation was found between beta2m and age (p<0.0001), serum concentrations significantly higher in older subjects (1.55, 1.59, 1.70, and 1.87 mg/L in age groups of 29-40, 40-50, 50-60 and 60-75 years, respectively, p<0.0001). Reference intervals obtained by non-parametric estimation after partitioning by age were 1.02-2.46 mg/L vs. 1.29-2.70 mg/L in younger (29-49 years) vs. older (50-75 years) individuals. CONCLUSIONS These data can help standardise beta2m reference limits and support age-adjusted comparisons in clinical studies.
Collapse
Affiliation(s)
- Zoltán Mátrai
- Department of Haematology and Stem Cell Transplantation, St. László Hospital, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
50
|
Giannopoulos K, Dmoszynska A, Kowal M, Wasik-Szczepanek E, Bojarska-Junak A, Rolinski J, Döhner H, Stilgenbauer S, Bullinger L. Thalidomide exerts distinct molecular antileukemic effects and combined thalidomide/fludarabine therapy is clinically effective in high-risk chronic lymphocytic leukemia. Leukemia 2009; 23:1771-8. [PMID: 19440214 DOI: 10.1038/leu.2009.98] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thalidomide represents a promising immunomodulatory drug that targets both leukemia cells and the tumor microenvironment. We treated patients with chronic lymphocytic leukemia (CLL) with a combined thalidomide/fludarabine regimen and monitored cellular and molecular changes induced by thalidomide in vivo before fludarabine treatment. Thalidomide was given daily (100 mg p.o. per day) and fludarabine was administered on days 7-11 (25 mg/m(2) i.v. per day) within each 4-week cycle (maximum of 6 cycles). Twenty patients received thalidomide/fludarabine as first-line therapy and 20 patients were previously treated. Unmutated IgVH mutation status was found in 36 cases and 13 had high-risk cytogenetic aberrations (del17p, del11q). The overall response rate was 80 and 25% for untreated and previously treated patients, respectively. Although thalidomide reduced the number of CLL cells, the number of CD3 lymphocytes showed no significant change, but the number of CD4(+)CD25(hi)FOXP3(+) regulatory T cells (Tregs) was significantly decreased. Gene expression profiling revealed a thalidomide-induced signature containing both targets known to have a function in immunomodulatory drug action as well as novel candidate genes. Combined thalidomide/fludarabine therapy demonstrated efficacy in high-risk patients with CLL. Furthermore, our study provides novel biological insights into thalidomide effect, which might act by enhancing apoptosis of CLL cells and reducing Tregs, thereby enabling T-cell-dependent antitumor effect.
Collapse
Affiliation(s)
- K Giannopoulos
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|