51
|
Domka K, Goral A, Firczuk M. cROSsing the Line: Between Beneficial and Harmful Effects of Reactive Oxygen Species in B-Cell Malignancies. Front Immunol 2020; 11:1538. [PMID: 32793211 PMCID: PMC7385186 DOI: 10.3389/fimmu.2020.01538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/11/2020] [Indexed: 01/06/2023] Open
Abstract
B-cell malignancies are a heterogeneous group of hematological neoplasms derived from cells at different stages of B-cell development. Recent studies revealed that dysregulated redox metabolism is one of the factors contributing to the pathogenesis and progression of B-cell malignancies. Elevated levels of oxidative stress markers usually correlate with the advanced stage of various B-cell malignancies. In the complex tumor microenvironment, reactive oxygen species affect not only malignant cells but also bystander cells, including immune cells. Importantly, malignant cells, due to genetic dysregulation, are able to adapt to the increased demands for energy and reducing equivalents via metabolic reprogramming and upregulation of antioxidants. The immune cells, however, are more sensitive to oxidative imbalance. This may cause their dysfunction, leading to immune evasion and tumor progression. On the other hand, the already imbalanced redox homeostasis renders malignant B-cells particularly sensitive to further elevation of reactive oxygen species. Indeed, targeting antioxidant systems has already presented anti-leukemic efficacy in preclinical models. Moreover, the prooxidant treatment that triggers immunogenic cell death has been utilized to generate autologous anti-leukemic vaccines. In this article, we review novel research on the dual role of the reactive oxygen species in B-cell malignancies. We highlight the mechanisms of maintaining redox homeostasis by malignant B-cells along with the antioxidant shield provided by the microenvironment. We summarize current findings regarding therapeutic targeting of redox metabolism in B-cell malignancies. We also discuss how the oxidative stress affects antitumor immune response and how excessive reactive oxygens species influence anticancer prooxidant treatments and immunotherapies.
Collapse
Affiliation(s)
- Krzysztof Domka
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Goral
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
52
|
Immune Response Dysfunction in Chronic Lymphocytic Leukemia: Dissecting Molecular Mechanisms and Microenvironmental Conditions. Int J Mol Sci 2020; 21:ijms21051825. [PMID: 32155826 PMCID: PMC7084946 DOI: 10.3390/ijms21051825] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 01/05/2023] Open
Abstract
Representing the major cause of morbidity and mortality for chronic lymphocytic leukemia (CLL) patients, immunosuppression is a common feature of the disease. Effectors of the innate and the adaptive immune response show marked dysfunction and skewing towards the generation of a tolerant environment that favors disease expansion. Major deregulations are found in the T lymphocyte compartment, with inhibition of CD8+ cytotoxic and CD4+ activated effector T cells, replaced by exhausted and more tolerogenic subsets. Likewise, differentiation of monocytes towards a suppressive M2-like phenotype is induced at the expense of pro-inflammatory sub-populations. Thanks to their B-regulatory phenotype, leukemic cells play a central role in driving immunosuppression, progressively inhibiting immune responses. A number of signaling cascades triggered by soluble mediators and cell–cell contacts contribute to immunomodulation in CLL, fostered also by local environmental conditions, such as hypoxia and derived metabolic acidosis. Specifically, molecular pathways modulating T-cell activity in CLL, spanning from the best known cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death 1 (PD-1) to the emerging T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domains (TIGIT)/CD155 axes, are attracting increasing research interest and therapeutic relevance also in the CLL field. On the other hand, in the microenvironment, the B cell receptor (BCR), which is undoubtedly the master regulator of leukemic cell behavior, plays an important role in orchestrating immune responses, as well. Lastly, local conditions of hypoxia, typical of the lymphoid niche, have major effects both on CLL cells and on non-leukemic immune cells, partly mediated through adenosine signaling, for which novel specific inhibitors are currently under development. In summary, this review will provide an overview of the molecular and microenvironmental mechanisms that modify innate and adaptive immune responses of CLL patients, focusing attention on those that may have therapeutic implications.
Collapse
|
53
|
Torres-Aguila NP, Carrera C, Giese AK, Cullell N, Muiño E, Cárcel-Márquez J, Gallego-Fabrega C, González-Sánchez J, Del Mar Freijo M, Álvarez-Sabín J, Molina C, Ribó M, Jimenez-Conde J, Roquer J, Sobrino T, Campos F, Castillo J, Muñoz-Narbona L, Lopez-Cancio E, Dàvalos A, Diaz-Navarro R, Tur S, Vives-Bauza C, Serrano-Heras G, Segura T, Krupinski J, Delgado-Mederos R, Martí-Fàbregas J, Heitsch L, Ibañez L, Cruchaga C, Rost NS, Montaner J, Lee JM, Fernandez-Cadenas I. Genome-Wide Association Study of White Blood Cell Counts in Patients With Ischemic Stroke. Stroke 2019; 50:3618-3621. [PMID: 31587654 DOI: 10.1161/strokeaha.119.026593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background and Purpose- Immune cells play a key role in the first 24h poststroke (acute phase), being associated with stroke outcome. We aimed to find genetic risk factors associated with leukocyte counts during the acute phase of stroke. Methods- Ischemic stroke patients with leukocyte counts data during the first 24h were included. Genome-wide association study and gene expression studies were performed. Results- Our genome-wide association study, which included 2064 (Discovery) and 407 (Replication) patients, revealed a new locus (14q24.3) associated with leukocyte counts. After Joint analysis (n=2471) 5 more polymorphisms reached genome-wide significance (P<5×10-8). The 14q24.3 locus was associated with acute stroke outcome (rs112809786, P=0.036) and with ACOT1 and PTGR2 gene expression. Previous polymorphisms associated with leukocyte counts in general-population did not show any significance in our study. Conclusions- We have found the first locus associated with leukocyte counts in ischemic stroke, also associated with acute outcome. Genetic analysis of acute endophenotypes could be useful to find the genetic factors associated with stroke outcome. Our findings suggested a different modulation of immune cells in stroke compared with healthy conditions.
Collapse
Affiliation(s)
- Nuria P Torres-Aguila
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.).,Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Barcelona, Spain (N.P.T.-A., C.C., J.M.)
| | - Caty Carrera
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Barcelona, Spain (N.P.T.-A., C.C., J.M.)
| | - Anne-Katrine Giese
- Neurology Department, Massachusetts General Hospital, Boston (A.-K.G., N.S.R.)
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Laboratory, Fundació Mútua Terrasa, Hospital Universitari Mútua Terrassa, Spain (N.C., C.G.-F., J.G.-S.)
| | - Elena Muiño
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.)
| | - Jara Cárcel-Márquez
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.)
| | - Cristina Gallego-Fabrega
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.).,Stroke Pharmacogenomics and Genetics Laboratory, Fundació Mútua Terrasa, Hospital Universitari Mútua Terrassa, Spain (N.C., C.G.-F., J.G.-S.)
| | - Jonathan González-Sánchez
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.).,Stroke Pharmacogenomics and Genetics Laboratory, Fundació Mútua Terrasa, Hospital Universitari Mútua Terrassa, Spain (N.C., C.G.-F., J.G.-S.).,School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom (J.G.-S., J.K.)
| | | | - José Álvarez-Sabín
- Stroke Unit, Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.Á.-S., C.M., M.R.)
| | - Carlos Molina
- Stroke Unit, Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.Á.-S., C.M., M.R.)
| | - Marc Ribó
- Stroke Unit, Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.Á.-S., C.M., M.R.)
| | - Jordi Jimenez-Conde
- Neurology Department, Institut Hospital del Mar d'Investigacions Mèdiques-Hospital del Mar Barcelona, Spain (J.J.-C., J.R.).,Neurovascular Research Group, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain (J.J.-C., J.R.).,Universitat Autónoma de Barcelona/Departamento de Ciencias Experimentales y de la Salud, Universitat Pompeu Fabra, Barcelona, Spain (J.J.-C., J.R.)
| | - Jaume Roquer
- Neurology Department, Institut Hospital del Mar d'Investigacions Mèdiques-Hospital del Mar Barcelona, Spain (J.J.-C., J.R.).,Neurovascular Research Group, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain (J.J.-C., J.R.).,Universitat Autónoma de Barcelona/Departamento de Ciencias Experimentales y de la Salud, Universitat Pompeu Fabra, Barcelona, Spain (J.J.-C., J.R.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain (T.S., F.C., J.C.)
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain (T.S., F.C., J.C.)
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain (T.S., F.C., J.C.)
| | - Lucia Muñoz-Narbona
- Neurosciences Department, Hospital Germans Trias I Pujol-Universitat Autónoma de Barcelona, Barcelona, Spain (L.M.-N., A.D.)
| | - Elena Lopez-Cancio
- Stroke Unit, Hospital Universitario Central de Asturias, Oviedo, Spain (E.L.-C.)
| | - Antoni Dàvalos
- Neurosciences Department, Hospital Germans Trias I Pujol-Universitat Autónoma de Barcelona, Barcelona, Spain (L.M.-N., A.D.)
| | - Rosa Diaz-Navarro
- Neuroscience Laboratory, Fundació Institut d'Investigació Sanitària Illes Balears, Mallorca, Spain (R.D.-N., S.T., C.V.-B.)
| | - Silvia Tur
- Neuroscience Laboratory, Fundació Institut d'Investigació Sanitària Illes Balears, Mallorca, Spain (R.D.-N., S.T., C.V.-B.)
| | - Cristòfol Vives-Bauza
- Neuroscience Laboratory, Fundació Institut d'Investigació Sanitària Illes Balears, Mallorca, Spain (R.D.-N., S.T., C.V.-B.)
| | - Gemma Serrano-Heras
- Neurology Department, University Hospital of Albacete, Spain (G.S.-H., T.S.)
| | - Tomás Segura
- Neurology Department, University Hospital of Albacete, Spain (G.S.-H., T.S.)
| | - Jerzy Krupinski
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom (J.G.-S., J.K.).,Neurology Service, Hospital Universitari Mútua Terrassa, Spain (J.K.)
| | - Raquel Delgado-Mederos
- Stroke Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (R.D.-M., J.M.-F.)
| | - Joan Martí-Fàbregas
- Stroke Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (R.D.-M., J.M.-F.)
| | - Laura Heitsch
- Emergency Medicine (L.H.), Washington University School of Medicine, Saint Louis, MO.,Neurology Department (L.H., J.-M.L.), Washington University School of Medicine, Saint Louis, MO
| | - Laura Ibañez
- Psychiatry Department (L.I., C.C.), Washington University School of Medicine, Saint Louis, MO
| | - Carlos Cruchaga
- Psychiatry Department (L.I., C.C.), Washington University School of Medicine, Saint Louis, MO
| | - Natalia S Rost
- Neurology Department, Massachusetts General Hospital, Boston (A.-K.G., N.S.R.)
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Barcelona, Spain (N.P.T.-A., C.C., J.M.).,Stroke Research Program, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/University of Seville (J.M.).,Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain (J.M.)
| | - Jin-Moo Lee
- Neurology Department (L.H., J.-M.L.), Washington University School of Medicine, Saint Louis, MO
| | - Israel Fernandez-Cadenas
- From the Stroke Pharmacogenomics and Genetics Laboratory, Hospital de la Santa Creu i Sant Pau Research Institute, Barcelona Spain (N.P.T.-A., E.M., J.C.-M., C.G.-F., J.G.-S., I.F.-C.)
| |
Collapse
|
54
|
Schulze-Edinghausen L, Dürr C, Öztürk S, Zucknick M, Benner A, Kalter V, Ohl S, Close V, Wuchter P, Stilgenbauer S, Lichter P, Seiffert M. Dissecting the Prognostic Significance and Functional Role of Progranulin in Chronic Lymphocytic Leukemia. Cancers (Basel) 2019; 11:E822. [PMID: 31200555 PMCID: PMC6627891 DOI: 10.3390/cancers11060822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is known for its strong dependency on the tumor microenvironment. We found progranulin (GRN), a protein that has been linked to inflammation and cancer, to be upregulated in the serum of CLL patients compared to healthy controls, and increased GRN levels to be associated with an increased hazard for disease progression and death. This raised the question of whether GRN is a functional driver of CLL. We observed that recombinant GRN did not directly affect viability, activation, or proliferation of primary CLL cells in vitro. However, GRN secretion was induced in co-cultures of CLL cells with stromal cells that enhanced CLL cell survival. Gene expression profiling and protein analyses revealed that primary mesenchymal stromal cells (MSCs) in co-culture with CLL cells acquire a cancer-associated fibroblast-like phenotype. Despite its upregulation in the co-cultures, GRN treatment of MSCs did not mimic this effect. To test the relevance of GRN for CLL in vivo, we made use of the Eμ-TCL1 CLL mouse model. As we detected strong GRN expression in myeloid cells, we performed adoptive transfer of Eμ-TCL1 leukemia cells to bone marrow chimeric Grn-/- mice that lack GRN in hematopoietic cells. Thereby, we observed that CLL-like disease developed comparable in Grn-/- chimeras and respective control mice. In conclusion, serum GRN is found to be strongly upregulated in CLL, which indicates potential use as a prognostic marker, but there is no evidence that elevated GRN functionally drives the disease.
Collapse
Affiliation(s)
- Lena Schulze-Edinghausen
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Claudia Dürr
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Selcen Öztürk
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway.
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Verena Kalter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sibylle Ohl
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Viola Close
- Internal Medicine III, University of Ulm, 89081 Ulm, Germany, and Cooperation Unit Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany.
| | - Stephan Stilgenbauer
- Internal Medicine III, University of Ulm, 89081 Ulm, Germany, and Department of Internal Medicine I, Saarland University, 66421 Homburg, Germany.
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- German Cancer Research Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
55
|
Hanna BS, Öztürk S, Seiffert M. Beyond bystanders: Myeloid cells in chronic lymphocytic leukemia. Mol Immunol 2019; 110:77-87. [DOI: 10.1016/j.molimm.2017.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 12/31/2022]
|
56
|
Gohil SH, Wu CJ. Dissecting CLL through high-dimensional single-cell technologies. Blood 2019; 133:1446-1456. [PMID: 30728142 PMCID: PMC6440295 DOI: 10.1182/blood-2018-09-835389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
We now have the potential to undertake detailed analysis of the inner workings of thousands of cancer cells, one cell at a time, through the emergence of a range of techniques that probe the genome, transcriptome, and proteome combined with the development of bioinformatics pipelines that enable their interpretation. This provides an unprecedented opportunity to better understand the heterogeneity of chronic lymphocytic leukemia and how mutations, activation states, and protein expression at the single-cell level have an impact on disease course, response to treatment, and outcomes. Herein, we review the emerging application of these new techniques to chronic lymphocytic leukemia and examine the insights already attained through this transformative technology.
Collapse
Affiliation(s)
- Satyen H Gohil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA; and
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
57
|
Giannoni P, Fais F, Cutrona G, Totero DD. Hepatocyte Growth Factor: A Microenvironmental Resource for Leukemic Cell Growth. Int J Mol Sci 2019; 20:ijms20020292. [PMID: 30642077 PMCID: PMC6359660 DOI: 10.3390/ijms20020292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the progressive expansion of B lymphocytes CD5+/CD23+ in peripheral blood, lymph-nodes, and bone marrow. The pivotal role played by the microenvironment in disease pathogenesis has become increasingly clear. We demonstrated that bone marrow stromal cells and trabecular bone cells sustain survival of leukemic B cells through the production of hepatocyte growth factor (HGF). Indeed the trans-membrane kinase receptor for HGF, c-MET, is expressed on CLL cells and STAT3 TYR705 or AKT phosphorylation is induced after HGF/c-MET interaction. We have further observed that c-MET is also highly expressed in a peculiar type of cells of the CLL-microenvironment showing nurturing features for the leukemic clone (nurse-like cells: NLCs). Since HGF treatment drives monocytes toward the M2 phenotype and NLCs exhibit features of tumor associated macrophages of type 2 we suggested that HGF, released either by cells of the microenvironment or leukemic cells, exerts a double effect: (i) enhances CLL cells survival and (ii) drives differentiation of monocytes-macrophages to an oriented immune suppressive phenotype. We here discuss how paracrine, but also autocrine production of HGF by malignant cells, may favor leukemic clone expansion and resistance to conventional drug treatments in CLL, as well as in other hematological malignancies. Novel therapeutic approaches aimed to block HGF/c-MET interactions are further proposed.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Department of Experimental Medicine, University of Genoa, V. Pastore 3, 16132 Genova, Italy.
| | - Franco Fais
- Molecular Pathology Unit, IRCCS Polyclinic Hospital San Martino, L.go R. Benzi n.10, 16132 Genova, Italy.
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Polyclinic Hospital San Martino, L.go R. Benzi n.10, 16132 Genova, Italy.
| | - Daniela de Totero
- Molecular Pathology Unit, IRCCS Polyclinic Hospital San Martino, L.go R. Benzi n.10, 16132 Genova, Italy.
| |
Collapse
|
58
|
Dong S, Harrington BK, Hu EY, Greene JT, Lehman AM, Tran M, Wasmuth RL, Long M, Muthusamy N, Brown JR, Johnson AJ, Byrd JC. PI3K p110δ inactivation antagonizes chronic lymphocytic leukemia and reverses T cell immune suppression. J Clin Invest 2018; 129:122-136. [PMID: 30457982 DOI: 10.1172/jci99386] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/02/2018] [Indexed: 12/20/2022] Open
Abstract
Targeted therapy with small molecules directed at essential survival pathways in leukemia represents a major advance, including the phosphatidylinositol-3'-kinase (PI3K) p110δ inhibitor idelalisib. Here, we found that genetic inactivation of p110δ (p110δD910A/D910A) in the Eμ-TCL1 murine chronic lymphocytic leukemia (CLL) model impaired B cell receptor signaling and B cell migration, and significantly delayed leukemia pathogenesis. Regardless of TCL1 expression, p110δ inactivation led to rectal prolapse in mice resembling autoimmune colitis in patients receiving idelalisib. Moreover, we showed that p110δ inactivation in the microenvironment protected against CLL and acute myeloid leukemia. After receiving higher numbers of TCL1 leukemia cells, half of p110δD910A/D910A mice spontaneously recovered from high disease burden and resisted leukemia rechallenge. Despite disease resistance, p110δD910A/D910A mice exhibited compromised CD4+ and CD8+ T cell response, and depletion of CD4+ or CD8+ T cells restored leukemia. Interestingly, p110δD910A/D910A mice showed significantly impaired Treg expansion that associated with disease clearance. Reconstitution of p110δD910A/D910A mice with p110δWT/WT Tregs reversed leukemia resistance. Our findings suggest that p110δ inhibitors may have direct antileukemic and indirect immune-activating effects, further supporting that p110δ blockade may have a broader immune-modulatory role in types of leukemia that are not sensitive to p110δ inhibition.
Collapse
Affiliation(s)
- Shuai Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy.,Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| | - Bonnie K Harrington
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center.,College of Veterinary Medicine
| | - Eileen Y Hu
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center.,Medical Scientist Training Program
| | - Joseph T Greene
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center.,Molecular, Cellular, and Developmental Biology Program, and
| | - Amy M Lehman
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, USA
| | - Minh Tran
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| | - Ronni L Wasmuth
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| | - Meixiao Long
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| | - Jennifer R Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Amy J Johnson
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center.,Janssen Research and Development LLC, Spring House, Pennsylvania, USA
| | - John C Byrd
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy.,Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center
| |
Collapse
|
59
|
Methylation level of Rap1GAP and the clinical significance in MDS. Oncol Lett 2018; 16:7287-7294. [PMID: 30546468 DOI: 10.3892/ol.2018.9503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
Previous studies on the pathogenesis of myelodysplastic syndrome (MDS) have identified multiple associated gene mutations, including mutations of tetmethylcytosinedioxygenase 2, isocitrate dehydrogenase [NADP(+)] 1 cytosolic, isocitrate dehydrogenase [NADP(+)] 2 mitochondrial and additional sex combs like 1 transcriptional regulator, all of which may be considered epigenetic regulators. Furthermore, mutations of RAS type GTPase family genes have been identified in 10-15% patients with MDS. The authors' previous study on the gene expression profile of cluster of differentiation 34+ cells using microarray analysis identified elevated expression of RAP1GTPase activating protein 1 (Rap1GAP) in patients with MDS compared with that in non-malignant blood diseases (NM) control group. To further investigate the mechanism of increased Rap1GAP expression, the methylation pattern of the promoter of this gene was determined in 86 patients with MDS (n=29), acute myeloid leukemia (AML) (n=31) or NM (n=26) using bisulfite-specific polymerase chain reaction and DNA sequencing. The results demonstrated that the methylation of Rap1GAP occurred in all 29 patients with MDS at multiple CpG sites. The methylation level of Rap1GAP in patients with MDS was decreased compared with that in patients with NM. Significant differences at 4CpG sites (5,7,8 and 12) of Rap1GAP promoter were identified between MDS and NM. Furthermore, based on the present clinical records of the patient cohort, the methylation status of Rap1GAP promoter did not appear to be associated with the clinicopathological characteristics of patients with MDS, including age, gender and International Prognosis Score System. The difference in methylation level at CpG site 8 of Rap1GAP promoter was identified to be significantly increased in patients with MDS-refractory anemia with ring sideroblasts compared with that in the MDS-refractory cytopenia with multilineage dysplasia or MDS-unclassified groups. The results of the present study suggest that patients with MDS exhibit a lower overall methylation level within Rap1GAP promoter compared with patients with NM or AML. In addition, the methylation level at the four identified CpG sites can distinguish between MDS and NM.
Collapse
|
60
|
Palma M, Krstic A, Peña Perez L, Berglöf A, Meinke S, Wang Q, Blomberg KEM, Kamali-Moghaddam M, Shen Q, Jaremko G, Lundin J, De Paepe A, Höglund P, Kimby E, Österborg A, Månsson R, Smith CIE. Ibrutinib induces rapid down-regulation of inflammatory markers and altered transcription of chronic lymphocytic leukaemia-related genes in blood and lymph nodes. Br J Haematol 2018; 183:212-224. [PMID: 30125946 DOI: 10.1111/bjh.15516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/11/2018] [Indexed: 01/05/2023]
Abstract
In chronic lymphocytic leukaemia (CLL) patients, treatment with the Bruton tyrosine kinase inhibitor ibrutinib induces a rapid shift of tumour cells from lymph nodes (LN) to peripheral blood (PB). Here, we characterized in depth the dynamics of ibrutinib-induced inflammatory, transcriptional and cellular changes in different compartments immediately after treatment initiation in seven relapsed/refractory CLL patients. Serial PB and LN samples were taken before start and during the first 29 days of treatment. Changes in plasma inflammation-related biomarkers, CLL cell RNA expression, B-cell activation and migration markers expression, and PB mononuclear cell populations were assessed. A significant reduction of 10 plasma inflammation markers, the majority of which were chemokines and not CLL-derived, was observed within hours, and was paralleled by very early increase of CD19+ circulating cells. At the RNA level, significant and continuous changes in transcription factors and signalling molecules linked to B-cell receptor signalling and CLL biology was observed in both PB and LN CLL cells already after 2 days of treatment. In conclusion, ibrutinib seems to instantly shut off an ongoing inflammatory response and interfere with diverse sensitive pathways in the LN.
Collapse
Affiliation(s)
- Marzia Palma
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Haematology, Karolinska University Hospital, Stockholm, Sweden
| | - Aleksandra Krstic
- Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lucia Peña Perez
- Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Berglöf
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Meinke
- Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Qing Wang
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Qiujin Shen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Georg Jaremko
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Jeanette Lundin
- Department of Haematology, Karolinska University Hospital, Stockholm, Sweden
| | - Ayla De Paepe
- Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Petter Höglund
- Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Kimby
- Department of Haematology, Karolinska University Hospital, Stockholm, Sweden.,Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Österborg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Haematology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert Månsson
- Department of Haematology, Karolinska University Hospital, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
61
|
Assis-Mendonça GR, Crepaldi AH, Delamain MT, Moreira AH, Costa FD, Lima VCCD, Souza CAD, Soares FA, Vassallo J. Characteristics of follicular and mantle cell lymphoma in Brazil: prognostic impact of clinical parameters and treatment conditions in two hospitals. Hematol Transfus Cell Ther 2018; 40:343-353. [PMID: 30370412 PMCID: PMC6200674 DOI: 10.1016/j.htct.2018.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 02/07/2018] [Indexed: 12/31/2022] Open
Abstract
Objective Follicular and mantle cell lymphoma are low-grade B-cell malignancies that lack good responses to chemoimmunotherapy. This study aimed to assess retrospectively clinicopathological features and to determine independent prognostic factors for follicular and mantle cell lymphoma patients treated at two Brazilian medical centers: the Hematology and Hemotherapy Center of the Universidade Estadual de Campinas (Unicamp), a public university hospital, and AC. Camargo Cancer Center, a specialized cancer center. Methods Two hundred and twenty-seven follicular and 112 mantle cell lymphoma cases were diagnosed between 1999 and 2016. Archived paraffin blocks were retrieved and reviewed. Corresponding demographics and clinical data were recovered from medical charts. Outcome analyses considered both overall and event-free survival. Results For follicular lymphoma treated with the R-CHOP (rituximab, cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, prednisone) and R-CVP (rituximab, cyclophosphamide, vincristine sulfate, prednisone) regimens, both B-symptoms (p-value < 0.01 for overall and event-free survival) and high-risk Follicular Lymphoma International Prognostic Index (p-value < 0.01 for overall survival) were independently associated to worse prognosis. Maintenance with rituximab improved the prognosis (p-value < 0.01 for overall survival). For mantle cell lymphoma, B-symptoms (p-value = 0.03 for overall survival and event-free survival) and bone marrow infiltration (p-value = 0.01 for overall survival) independently predicted reduced survival, and rituximab at induction increased both event-free and overall survival (p-value < 0.01 in both analyses). Combinations of these deleterious features could identify extremely poor prognostic subgroups. The administration of rituximab was more frequent in the AC. Camargo Cancer Center, which was the institution associated with better overall survival for both neoplasias. Conclusion This study represents the largest cohort of follicular and mantle cell lymphoma in South America thus far. Some easily assessable clinical variables were able to predict prognosis and should be considered in low-income centers. In addition, the underuse of rituximab in the Brazilian public health system should be reconsidered in future health policies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - José Vassallo
- Universidade Estadual de Campinas (Unicamp), Campinas, SP, Brazil.,A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| |
Collapse
|
62
|
Pollyea DA, Hedin BR, O'Connor BP, Alper S. Monocyte function in patients with myelodysplastic syndrome. J Leukoc Biol 2018; 104:641-647. [PMID: 29656609 DOI: 10.1002/jlb.5ab1017-419rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/14/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a malignant hematopoietic stem cell disorder that frequently evolves into acute myeloid leukemia (AML). Patients with MDS are prone to infectious complications, in part due to the presence of severe neutropenia and/or neutrophil dysfunction. However, not all patients with neutropenia become infected, suggesting that other immune cells may compensate in these patients. Monocytes are also integral to immunologic defense; however, much less is known about monocyte function in patients with MDS. In the current study, we monitor the composition of peripheral blood monocytes and several aspects of monocyte function in MDS patients, including HLA-DR expression, LPS-induced inflammatory cytokine production, and phagocytosis. We find that monocytes from MDS patients exhibit relatively normal innate immune functions compared to monocytes from healthy control subjects. We also find that HLA-DR expression is moderately increased in monocytes from MDS patients. These results suggest that monocytes could compensate for other immune deficits in MDS patients to help fight infection. We also find that the range of immune functions in monocytes from MDS patients correlates with several key clinical parameters, including blast cell count, monocyte count, and revised International Prognostic Scoring System score, suggesting that disease severity impacts monocyte function in MDS patients.
Collapse
Affiliation(s)
- Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Brenna R Hedin
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA.,Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Brian P O'Connor
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA.,Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA.,Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Scott Alper
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA.,Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
63
|
van Attekum MH, Eldering E, Kater AP. Chronic lymphocytic leukemia cells are active participants in microenvironmental cross-talk. Haematologica 2017; 102:1469-1476. [PMID: 28775118 PMCID: PMC5685246 DOI: 10.3324/haematol.2016.142679] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
The importance of the tumor microenvironment in chronic lymphocytic leukemia is
widely accepted. Nevertheless, the understanding of the complex interplay
between the various types of bystander cells and chronic lymphocytic leukemia
cells is incomplete. Numerous studies have indicated that bystander cells
provide chronic lymphocytic leukemia-supportive functions, but it has also
become clear that chronic lymphocytic leukemia cells actively engage in the
formation of a supportive tumor microenvironment through several cross-talk
mechanisms. In this review, we describe how chronic lymphocytic leukemia cells
participate in this interplay by inducing migration and tumor-supportive
differentiation of bystander cells. Furthermore, chronic lymphocytic
leukemia-mediated alterations in the interactions between bystander cells are
discussed. Upon bystander cell interaction, chronic lymphocytic leukemia cells
secrete cytokines and chemokines such as migratory factors [chemokine
(C-C motif) ligand 22 and chemokine (CC motif) ligand 2], which result
in further recruitment of T cells but also of monocyte-derived cells. Within the
tumor microenvironment, chronic lymphocytic leukemia cells induce
differentiation towards a tumor-supportive M2 phenotype of monocyte-derived
cells and suppress phagocytosis, but also induce increased numbers of supportive
regulatory T cells. Like other tumor types, the differentiation of stromal cells
towards supportive cancer-associated fibroblasts is critically dependent on
chronic lymphocytic leukemia-derived factors such as exosomes and
platelet-derived growth factor. Lastly, both chronic lymphocytic leukemia and
bystander cells induce a tolerogenic tumor microenvironment; chronic lymphocytic
leukemia-secreted cytokines, such as interleukin-10, suppress cytotoxic T-cell
functions, while chronic lymphocytic leukemia-associated monocyte-derived cells
contribute to suppression of T-cell function by producing the immune checkpoint
factor, programmed cell death-ligand 1. Deeper understanding of the active
involvement and cross-talk of chronic lymphocytic leukemia cells in shaping the
tumor microenvironment may offer novel clues for designing therapeutic
strategies.
Collapse
Affiliation(s)
- Martijn Ha van Attekum
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands .,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
64
|
Haderk F, Schulz R, Iskar M, Cid LL, Worst T, Willmund KV, Schulz A, Warnken U, Seiler J, Benner A, Nessling M, Zenz T, Göbel M, Dürig J, Diederichs S, Paggetti J, Moussay E, Stilgenbauer S, Zapatka M, Lichter P, Seiffert M. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol 2017; 2:2/13/eaah5509. [PMID: 28754746 DOI: 10.1126/sciimmunol.aah5509] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 03/23/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
Abstract
In chronic lymphocytic leukemia (CLL), monocytes and macrophages are skewed toward protumorigenic phenotypes, including the release of tumor-supportive cytokines and the expression of immunosuppressive molecules such as programmed cell death 1 ligand 1 (PD-L1). To understand the mechanism driving protumorigenic skewing in CLL, we evaluated the role of tumor cell-derived exosomes in the cross-talk with monocytes. We carried out RNA sequencing and proteome analyses of CLL-derived exosomes and identified noncoding Y RNA hY4 as a highly abundant RNA species that is enriched in exosomes from plasma of CLL patients compared with healthy donor samples. Transfer of CLL-derived exosomes or hY4 alone to monocytes resulted in key CLL-associated phenotypes, including the release of cytokines, such as C-C motif chemokine ligand 2 (CCL2), CCL4, and interleukin-6, and the expression of PD-L1. These responses were abolished in Toll-like receptor 7 (TLR7)-deficient monocytes, suggesting exosomal hY4 as a driver of TLR7 signaling. Pharmacologic inhibition of endosomal TLR signaling resulted in a substantially reduced activation of monocytes in vitro and attenuated CLL development in vivo. Our results indicate that exosome-mediated transfer of noncoding RNAs to monocytes contributes to cancer-related inflammation and concurrent immune escape via PD-L1 expression.
Collapse
Affiliation(s)
- Franziska Haderk
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralph Schulz
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Murat Iskar
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Llaó Cid
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Worst
- Division of Signaling and Functional Genomics, DKFZ, Heidelberg, Germany
| | - Karolin V Willmund
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angela Schulz
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Uwe Warnken
- Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Jana Seiler
- Division of RNA Biology and Cancer (B150), DKFZ, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, DKFZ, Heidelberg, Germany
| | | | - Thorsten Zenz
- Department of Molecular Therapy in Hematology and Oncology and Department of Translational Oncology, National Center for Tumor Diseases (NCT); DKFZ; and Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Maria Göbel
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Jan Dürig
- Department of Hematology, Essen University Hospital, Essen, Germany
| | - Sven Diederichs
- Division of RNA Biology and Cancer (B150), DKFZ, Heidelberg, Germany.,Division of Cancer Research, Department of Thoracic Surgery, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany
| | - Jérôme Paggetti
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Etienne Moussay
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | | | - Marc Zapatka
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Seiffert
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
65
|
Purroy N, Wu CJ. Coevolution of Leukemia and Host Immune Cells in Chronic Lymphocytic Leukemia. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026740. [PMID: 28096240 DOI: 10.1101/cshperspect.a026740] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cumulative studies on the dissection of changes in driver genetic lesions in cancer across the course of the disease have provided powerful insights into the adaptive mechanisms of tumors in response to the selective pressures of therapy and environmental changes. In particular, the advent of next-generation-sequencing (NGS)-based technologies and its implementation for the large-scale comprehensive analyses of cancers have greatly advanced our understanding of cancer as a complex dynamic system wherein genetically distinct subclones interact and compete during tumor evolution. Aside from genetic evolution arising from interactions intrinsic to the cell subpopulations within tumors, it is increasingly appreciated that reciprocal interactions between the tumor cell and cellular constituents of the microenvironment further exert selective pressures on specific clones that can impact the balance between tumor immunity and immunologic evasion and escape. Herein, we review the evidence supporting these concepts, with a particular focus on chronic lymphocytic leukemia (CLL), a disease that has been highly amenable to genomic interrogation and studies of clonal heterogeneity and evolution. Better knowledge of the basis for immune escape has an important clinical impact on prognostic stratification and on the pursuit of new therapeutic opportunities.
Collapse
Affiliation(s)
- Noelia Purroy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
66
|
Turrini R, Pabois A, Xenarios I, Coukos G, Delaloye JF, Doucey MA. TIE-2 expressing monocytes in human cancers. Oncoimmunology 2017; 6:e1303585. [PMID: 28507810 PMCID: PMC5414874 DOI: 10.1080/2162402x.2017.1303585] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAM) are well known as a key player in the tumor microenvironment, which support cancer progression. More recently, a lineage of monocytes characterized by the expression of the TIE-2/Tek angiopoietin receptor identified a subset of circulating and tumor-associated monocytes endowed with proangiogenic activity. TIE-2 expressing monocytes (TEM) were found both in humans and mice. Here, we review the phenotypes and functions of TEM reported so far in human cancer and their potential use as markers of cancer progression and metastasis. Finally, we discuss the therapeutic approaches currently used or proposed to target TEM.
Collapse
Affiliation(s)
- Riccardo Turrini
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Angélique Pabois
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Xenarios
- Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Marie-Agnès Doucey
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
67
|
Yao Y, Strauss-Albee DM, Zhou JQ, Malawista A, Garcia MN, Murray KO, Blish CA, Montgomery RR. The natural killer cell response to West Nile virus in young and old individuals with or without a prior history of infection. PLoS One 2017; 12:e0172625. [PMID: 28235099 PMCID: PMC5325267 DOI: 10.1371/journal.pone.0172625] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
West Nile virus (WNV) typically leads to asymptomatic infection but can cause severe neuroinvasive disease or death, particularly in the elderly. Innate NK cells play a critical role in antiviral defenses, yet their role in human WNV infection is poorly defined. Here we demonstrate that NK cells mount a robust, polyfunctional response to WNV characterized by cytolytic activity, cytokine and chemokine secretion. This is associated with downregulation of activating NK cell receptors and upregulation of NK cell activating ligands for NKG2D. The NK cell response did not differ between young and old WNV-naïve subjects, but a history of symptomatic infection is associated with more IFN-γ producing NK cell subsets and a significant decline in a specific NK cell subset. This NK repertoire skewing could either contribute to or follow heightened immune pathogenesis from WNV infection, and suggests that NK cells could play an important role in WNV infection in humans.
Collapse
Affiliation(s)
- Yi Yao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Dara M. Strauss-Albee
- Stanford Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Julian Q. Zhou
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Anna Malawista
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Melissa N. Garcia
- Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, United States of America
| | - Kristy O. Murray
- Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, United States of America
| | - Catherine A. Blish
- Stanford Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ruth R. Montgomery
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Program on Human Translational Immunology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
68
|
Th1 and Th17 proinflammatory profile characterizes invariant natural killer T cells in virologically suppressed HIV+ patients with low CD4+/CD8+ ratio. AIDS 2016; 30:2599-2610. [PMID: 27782963 DOI: 10.1097/qad.0000000000001247] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Scanty data exist on the phenotype and functionality of invariant natural killer T (iNKT) cells in HIV-infected (HIV+) patients. METHODS By flow cytometry, we studied iNKT cells from 54 HIV+ patients who started combined antiretroviral therapy and had undetectable viral load for more than 1 year. Twenty-five maintained a CD4/CD8 ratio less than 0.4, whereas 29 reached a ratio more than 1.1; 32 age-matched and sex-matched patients were healthy controls (CTR). RESULTS Patients with low ratio had lower percentage of CD4 iNKT cells compared with patients with high ratio and higher CD8 iNKT cell percentage; double-negative iNKT cells were lower in HIV+ patients compared with CTR. Patients with low ratio had higher percentage of CD4 and double-negative iNKT cells expressing CD38 and HLA-DR compared with patients with high ratio. CD4 iNKT cells expressing PD-1 were higher in patients with CD4/CD8 ratio less than 0.4, whereas double-negative iNKT cells expressing PD-1 were lower compared with patients with ratio more than 1.1. Patients with low ratio had higher CD4 iNKT cells producing IL-17, CD8 iNKT cells producing IFN-γ, TNF-α or IFN-γ and TNF-α, and double-negative iNKT cells producing IL-17 or IL-17 and IFN-γ compared with CTR. Activated CD4 (or CD8) T cells correlated with activated CD4 (or CD8) iNKT cells, as well as the percentages of CD4 (or CD8) T cells expressing PD-1 was correlated to that of CD4 (or CD8) iNKT cells expressing PD-1. CONCLUSION Low CD4/CD8 ratio despite effective combined antiretroviral therapy is associated with altered iNKT cell subsets, enhanced activation, and prominent Th1/Th17 proinflammatory profile.
Collapse
|
69
|
Stevens WBC, Netea MG, Kater AP, van der Velden WJFM. 'Trained immunity': consequences for lymphoid malignancies. Haematologica 2016; 101:1460-1468. [PMID: 27903713 DOI: 10.3324/haematol.2016.149252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022] Open
Abstract
In hematological malignancies complex interactions exist between the immune system, microorganisms and malignant cells. On one hand, microorganisms can induce cancer, as illustrated by specific infection-induced lymphoproliferative diseases such as Helicobacter pylori-associated gastric mucosa-associated lymphoid tissue lymphoma. On the other hand, malignant cells create an immunosuppressive environment for their own benefit, but this also results in an increased risk of infections. Disrupted innate immunity contributes to the neoplastic transformation of blood cells by several mechanisms, including the uncontrolled clearance of microbial and autoantigens resulting in chronic immune stimulation and proliferation, chronic inflammation, and defective immune surveillance and anti-cancer immunity. Restoring dysfunction or enhancing responsiveness of the innate immune system might therefore represent a new angle for the prevention and treatment of hematological malignancies, in particular lymphoid malignancies and associated infections. Recently, it has been shown that cells of the innate immune system, such as monocytes/macrophages and natural killer cells, harbor features of immunological memory and display enhanced functionality long-term after stimulation with certain microorganisms and vaccines. These functional changes rely on epigenetic reprogramming and have been termed 'trained immunity'. In this review the concept of 'trained immunity' is discussed in the setting of lymphoid malignancies. Amelioration of infectious complications and hematological disease progression can be envisioned to result from the induction of trained immunity, but future studies are required to prove this exciting new hypothesis.
Collapse
Affiliation(s)
- Wendy B C Stevens
- Department of Hematology, Radboud University Medical Centre, Nijmegen
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Centre, and Radboud Center for Infectious Diseases, Nijmegen.,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| | - Arnon P Kater
- Department of Hematology, Lymphoma and Myeloma Center Amsterdam (LYMMCARE) Academic Medical Center, University of Amsterdam, The Netherlands
| | - Walter J F M van der Velden
- Department of Hematology, Radboud University Medical Centre, Nijmegen .,Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen
| |
Collapse
|
70
|
Nguyen PH, Fedorchenko O, Rosen N, Koch M, Barthel R, Winarski T, Florin A, Wunderlich FT, Reinart N, Hallek M. LYN Kinase in the Tumor Microenvironment Is Essential for the Progression of Chronic Lymphocytic Leukemia. Cancer Cell 2016; 30:610-622. [PMID: 27728807 DOI: 10.1016/j.ccell.2016.09.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/16/2016] [Accepted: 09/15/2016] [Indexed: 01/09/2023]
Abstract
Survival of chronic lymphocytic leukemia (CLL) cells strictly depends on the support of an appropriate tumor microenvironment. Here, we demonstrate that LYN kinase is essential for CLL progression. Lyn deficiency results in a significantly reduced CLL burden in vivo. Loss of Lyn within leukemic cells reduces B cell receptor (BCR) signaling including BTK phosphorylation, but surprisingly does not affect leukemic cell expansion. Instead, syngeneic CLL transplantation of CLL cells into Lyn- or Btk-deficient recipients results in a strongly delayed leukemic progression and prolonged survival. Moreover, Lyn deficiency in macrophages hinders nursing functions for CLL cells, which is mediated by direct contact rather than secretion of soluble factors. Taken together, LYN and BTK seem essential for the formation of a microenvironment supporting leukemic growth.
Collapse
MESH Headings
- Animals
- Cell Proliferation/physiology
- Disease Progression
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Signal Transduction
- Tumor Microenvironment
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Phuong-Hien Nguyen
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Oleg Fedorchenko
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Natascha Rosen
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Maximilian Koch
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Romy Barthel
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Tomasz Winarski
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Alexandra Florin
- Institute of Pathology, University Hospital of Cologne, 50931 Cologne, Germany
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research; Institute for Genetics, University of Cologne, 50931 Cologne, Germany
| | - Nina Reinart
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
71
|
Takeda Y, Kato T, Ito H, Kurota Y, Yamagishi A, Sakurai T, Araki A, Nara H, Tsuchiya N, Asao H. The pattern of GPI-80 expression is a useful marker for unusual myeloid maturation in peripheral blood. Clin Exp Immunol 2016; 186:373-386. [PMID: 27569996 DOI: 10.1111/cei.12859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2016] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have a wide spectrum of immunosuppressive activity; control of these cells is a new target for improving clinical outcomes in cancer patients. MDSCs originate from unusual differentiation of neutrophils or monocytes induced by inflammatory cytokines, including granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophage (GM)-CSF. However, MDSCs are difficult to detect in neutrophil or monocyte populations because they are not uniform cells, resembling both neutrophils and monocytes; thus, they exist in a heterogeneous population. In this study, we investigated GPI-80, a known regulator of Mac-1 (CD11b/CD18) and associated closely with neutrophil maturation, to clarify this unusual differentiation. First, we demonstrated that the mean fluorescence intensity (MFI) of GPI-80 and coefficient of variation (CV) of GPI-80 were increased by treatment with G-CSF and GM-CSF, respectively, using a human promyelocytic leukaemia (HL60) cell differentiation model. To confirm the value of GPI-80 as a marker of unusual differentiation, we measured GPI-80 expression and MDSC functions using peripheral blood cells from metastatic renal cell carcinoma patients. The GPI-80 CV was augmented significantly in the CD16hi neutrophil cell population, and GPI-80 MFI was increased significantly in the CD33hi monocyte cell population. Furthermore, the GPI-80 CV in the CD16hi population was correlated inversely with the proliferative ability of T cells and the GPI-80 MFI of the CD33hi population was correlated with reactive oxygen species production. These results led us to propose that the pattern of GPI-80 expression in these populations is a simple and useful marker for unusual differentiation, which is related to MDSC functions.
Collapse
Affiliation(s)
- Y Takeda
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - T Kato
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Ito
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - Y Kurota
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - A Yamagishi
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - T Sakurai
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - A Araki
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Nara
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - N Tsuchiya
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Asao
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
72
|
The PD-1/PD-L1 axis contributes to immune metabolic dysfunctions of monocytes in chronic lymphocytic leukemia. Leukemia 2016; 31:470-478. [DOI: 10.1038/leu.2016.214] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/03/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
|
73
|
Kocher T, Asslaber D, Zaborsky N, Flenady S, Denk U, Reinthaler P, Ablinger M, Geisberger R, Bauer JW, Seiffert M, Hartmann TN, Greil R, Egle A, Piñón Hofbauer J. CD4+ T cells, but not non-classical monocytes, are dispensable for the development of chronic lymphocytic leukemia in the TCL1-tg murine model. Leukemia 2016; 30:1409-13. [PMID: 26522084 PMCID: PMC4895160 DOI: 10.1038/leu.2015.307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- T Kocher
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- EB House Austria, Research Program for the Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - D Asslaber
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - N Zaborsky
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - S Flenady
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - U Denk
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - P Reinthaler
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - M Ablinger
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - R Geisberger
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - J W Bauer
- EB House Austria, Research Program for the Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - M Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T N Hartmann
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - R Greil
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - A Egle
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute, Salzburg, Austria
| | - J Piñón Hofbauer
- Laboratory for Immunological and Molecular Cancer Research, Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- EB House Austria, Research Program for the Molecular Therapy of Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
74
|
Aguirre Palma LM, Flamme H, Gerke I, Kreuzer KA. Angiopoietins Modulate Survival, Migration, and the Components of the Ang-Tie2 Pathway of Chronic Lymphocytic Leukaemia (CLL) Cells In Vitro. CANCER MICROENVIRONMENT 2016; 9:13-26. [PMID: 26846110 DOI: 10.1007/s12307-016-0180-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 01/24/2016] [Indexed: 12/15/2022]
Abstract
In actuality, chronic lymphocytic leukaemia (CLL) remains an incurable haematopoietic malignancy of high prevalence amongst elderly populations in the West. Malignant CLL cells characteristically accumulate in the peripheral blood, bone marrow, lymph nodes, and spleen of CLL patients. There is evidence that CLL cells express Ang2 and Tie1, two central components of the Ang-Tie2 pro-angiogenic pathway. Central to blood vessel development and maintenance, at present it remains unclear how the Ang-Tie2 pathway modulates CLL pathophysiology. Here we evaluate the status of the Ang-Tie2 pathway in CLL cells and assess Ang1 levels in plasma/cell medium from CLL samples. To understand how angiopoietins in the microenvironment regulate the components of Ang-Tie2 pathway, survival, migration, and metabolic fitness of CLL cells, we exposed CLL cells to recombinant angiopoietins. CLL plasma and CLL cells in culture present significant lower levels of Ang1. CLL cells simultaneously express Ang1, Ang2, and Tie1 mRNA, but lack that of Tie2 and its regulator, VE-PTP. Exposure to Ang1 confers survival advantage in the long-term, whereas Ang2 and trebananib, an angiopoietin blocker, proved detrimental. Angiopoietins differentially modulate expression of Ang1, Ang2, and Tie1 transcripts. Ang2, but not Ang1, induces the concomitant and transient expression of Tie2 and VE-PTP mRNA. Both angiopoietins, particularly Ang2, increase CLL-Tie1 expression and Ang1 clearly induces chemotaxis and transendothelial-like migration of CLL cells. Besides, changes in caspase and ATP content corroborate the sensitivity of CLL cells to angiopoietin exposure. Altogether, this work shows that angiopoietins regulate the fate of CLL cells in a Tie2-independent manner and highlights the potential of the Ang-Tie2 pathway as a therapeutic target in CLL research.
Collapse
Affiliation(s)
| | - Hanna Flamme
- Department I of Internal Medicine, University at Cologne, Kerpener Strasse 62, Cologne, Germany
| | - Iris Gerke
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, MB, Canada
| | - Karl-Anton Kreuzer
- Department I of Internal Medicine, University at Cologne, Kerpener Strasse 62, Cologne, Germany.
| |
Collapse
|
75
|
Niemann CU, Herman SEM, Maric I, Gomez-Rodriguez J, Biancotto A, Chang BY, Martyr S, Stetler-Stevenson M, Yuan CM, Calvo KR, Braylan RC, Valdez J, Lee YS, Wong DH, Jones J, Sun C, Marti GE, Farooqui MZH, Wiestner A. Disruption of in vivo Chronic Lymphocytic Leukemia Tumor-Microenvironment Interactions by Ibrutinib--Findings from an Investigator-Initiated Phase II Study. Clin Cancer Res 2015; 22:1572-82. [PMID: 26660519 DOI: 10.1158/1078-0432.ccr-15-1965] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental interactions for proliferation and survival that are at least partially mediated through B-cell receptor (BCR) signaling. Ibrutinib, a Bruton tyrosine kinase inhibitor, disrupts BCR signaling and leads to the egress of tumor cells from the microenvironment. Although the on-target effects on CLL cells are well defined, the impact on the microenvironment is less well studied. We therefore sought to characterize the in vivo effects of ibrutinib on the tumor microenvironment. EXPERIMENTAL DESIGN Patients received single-agent ibrutinib on an investigator-initiated phase II trial. Serial blood and tissue samples were collected pretreatment and during treatment. Changes in cytokine levels, cellular subsets, and microenvironmental interactions were assessed. RESULTS Serum levels of key chemokines and inflammatory cytokines decreased significantly in patients on ibrutinib. Furthermore, ibrutinib treatment decreased circulating tumor cells and overall T-cell numbers. Most notably, a reduced frequency of the Th17 subset of CD4(+)T cells was observed concurrent with reduced expression of activation markers and PD-1 on T cells. Consistent with direct inhibition of T cells, ibrutinib inhibited Th17 differentiation of murine CD4(+)T cells in vitro Finally, in the bone marrow microenvironment, we found that ibrutinib disaggregated the interactions of macrophages and CLL cells, inhibited secretion of CXCL13, and decreased the chemoattraction of CLL cells. CONCLUSIONS In conjunction with inhibition of BCR signaling, these changes in the tumor microenvironment likely contribute to the antitumor activity of ibrutinib and may impact the efficacy of immunotherapeutic strategies in patients with CLL. See related commentary by Bachireddy and Wu, p. 1547.
Collapse
Affiliation(s)
- Carsten U Niemann
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland. Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Irina Maric
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | | | - Angelique Biancotto
- Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, Maryland
| | | | - Sabrina Martyr
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | | | - Constance M Yuan
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Katherine R Calvo
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | - Raul C Braylan
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | - Janet Valdez
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Yuh Shan Lee
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Deanna H Wong
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Jade Jones
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland. Medical Research Scholars Program, NIH, Bethesda, Maryland
| | - Clare Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Gerald E Marti
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Mohammed Z H Farooqui
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
76
|
Hounkpe BW, Fiusa MML, Colella MP, da Costa LNG, Benatti RDO, Saad STO, Costa FF, dos Santos MNN, De Paula EV. Role of innate immunity-triggered pathways in the pathogenesis of Sickle Cell Disease: a meta-analysis of gene expression studies. Sci Rep 2015; 5:17822. [PMID: 26648000 PMCID: PMC4673434 DOI: 10.1038/srep17822] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/06/2015] [Indexed: 12/19/2022] Open
Abstract
Despite the detailed characterization of the inflammatory and endothelial changes observed in Sickle Cell Disease (SCD), the hierarchical relationship between elements involved in the pathogenesis of this complex disease is yet to be described. Meta-analyses of gene expression studies from public repositories represent a novel strategy, capable to identify key mediators in complex diseases. We performed several meta-analyses of gene expression studies involving SCD, including studies with patient samples, as well as in-vitro models of the disease. Meta-analyses were performed with the Inmex bioinformatics tool, based on the RankProd package, using raw gene expression data. Functional gene set analysis was performed using more than 60 gene-set libraries. Our results demonstrate that the well-characterized association between innate immunity, hemostasis, angiogenesis and heme metabolism with SCD is also consistently observed at the transcriptomic level, across independent studies. The enrichment of genes and pathways associated with innate immunity and damage repair-associated pathways supports the model of erythroid danger-associated molecular patterns (DAMPs) as key mediators of the pathogenesis of SCD. Our study also generated a novel database of candidate genes, pathways and transcription factors not previously associated with the pathogenesis of SCD that warrant further investigation in models and patients of SCD.
Collapse
Affiliation(s)
| | - Maiara Marx Luz Fiusa
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | - Marina Pereira Colella
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | | | | | - Sara T Olalla Saad
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| | | | - Erich Vinicius De Paula
- Faculty of Medical Sciences, University of Campinas/Hematology and Hemotherapy Center, Campinas, SP, Brazil
| |
Collapse
|
77
|
Hanna BS, McClanahan F, Yazdanparast H, Zaborsky N, Kalter V, Rößner PM, Benner A, Dürr C, Egle A, Gribben JG, Lichter P, Seiffert M. Depletion of CLL-associated patrolling monocytes and macrophages controls disease development and repairs immune dysfunction in vivo. Leukemia 2015; 30:570-9. [PMID: 26522085 DOI: 10.1038/leu.2015.305] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/22/2015] [Accepted: 10/22/2015] [Indexed: 02/08/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by apoptosis resistance and a dysfunctional immune system. Previous reports suggested a potential role of myeloid cells in mediating these defects. However, the composition and function of CLL-associated myeloid cells have not been thoroughly investigated in vivo. Using the Eμ-TCL1 mouse model, we observed severe skewing of myeloid cell populations with CLL development. Monocytes and M2-like macrophages infiltrated the peritoneal cavity of leukemic mice. Monocytes also accumulated in the spleen in a CCR2-dependent manner, and were severely skewed toward Ly6C(low) patrolling or nonclassical phenotype. In addition, the percentage of MHC-II(hi) dendritic cells and macrophages significantly dropped in the spleen. Gene expression profiling of CLL-associated monocytes revealed aberrantly high PD-L1 expression and secretion of multiple inflammatory and immunosuppressive cytokines like interleukin-10, tumor necrosis factor-α and CXCL9. In vivo myeloid cell depletion using liposomal Clodronate resulted in a significant control of CLL development accompanied by a pronounced repair of innate immune cell phenotypes and a partial resolution of systemic inflammation. In addition, CLL-associated skewing of T cells toward antigen-experienced phenotypes was repaired. The presented data suggest that targeting nonmalignant myeloid cells might serve as a novel immunotherapeutical strategy for CLL.
Collapse
Affiliation(s)
- B S Hanna
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - F McClanahan
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Centre for Haemato-Oncology, Barts Cancer Institute, London, UK
| | - H Yazdanparast
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - N Zaborsky
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department for Hematology, Paracelsus Private Medical University Hospital, Salzburg, Austria
| | - V Kalter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P M Rößner
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Dürr
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Egle
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department for Hematology, Paracelsus Private Medical University Hospital, Salzburg, Austria
| | - J G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, London, UK
| | - P Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Seiffert
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
78
|
Church AK, VanDerMeid KR, Baig NA, Baran AM, Witzig TE, Nowakowski GS, Zent CS. Anti-CD20 monoclonal antibody-dependent phagocytosis of chronic lymphocytic leukaemia cells by autologous macrophages. Clin Exp Immunol 2015; 183:90-101. [PMID: 26307241 DOI: 10.1111/cei.12697] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 01/01/2023] Open
Abstract
Unconjugated monoclonal antibodies (mAbs) are an important component of effective combination therapies for chronic lymphocytic leukaemia (CLL). Antibody-dependent phagocytosis (ADP) is a major mediator of mAb cytotoxicity, but there is limited knowledge of the determinants of ADP efficacy. We used macrophages derived in vitro from autologous circulating monocytes to test the effects of mAb structure and concentration, target : effector cell ratio, duration of co-incubation and CLL cell CD20 expression on ADP. Next-generation anti-CD20 mAbs (ofatumumab, ublituximab, obinutuzumab, ocaratuzumab) were significantly more effective at inducing ADP compared to rituximab, but none were as effective as the anti-CD52 mAb alemtuzumab. Ofatumumab (10 μg/ml) used as a representative next-generation anti-CD20 mAb achieved an ADP plateau at 3 h co-incubation with a target : effector ratio of 10 : 1 (mean = 2.1 CLL cells/macrophage, range = 1.5-3.5). At 0.156 μg/ml (the lowest concentration tested) ofatumumab ADP was significantly higher than alemtuzumab. However, ofatumumab-induced ADP did not increase significantly at higher mAb concentrations. We show that anti-CD20 mAb ADP efficacy is determined by the mAb characteristics, target : effector ratio and incubation time. We suggest that preclinical evaluation of anti-CD20 mAbs to understand the determinants of ADP could be useful in designing future combination therapies for CLL.
Collapse
Affiliation(s)
- A K Church
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - K R VanDerMeid
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - N A Baig
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - A M Baran
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - T E Witzig
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - C S Zent
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
79
|
LAPUC IZABELA, BOLKUN LUKASZ, ELJASZEWICZ ANDRZEJ, RUSAK MALGORZATA, LUKSZA EWA, SINGH PAULINA, MIKLASZ PAULA, PISZCZ JAROSLAW, PTASZYNSKA-KOPCZYNSKA KATARZYNA, JASIEWICZ MALGORZATA, KAMINSKI KAROL, DABROWSKA MILENA, BODZENTA-LUKASZYK ANNA, KLOCZKO JANUSZ, MONIUSZKO MARCIN. Circulating classical CD14++CD16− monocytes predict shorter time to initial treatment in chronic lymphocytic leukemia patients: Differential effects of immune chemotherapy on monocyte-related membrane and soluble forms of CD163. Oncol Rep 2015; 34:1269-78. [PMID: 26135617 DOI: 10.3892/or.2015.4088] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/29/2015] [Indexed: 11/06/2022] Open
|
80
|
Perturbation of the normal immune system in patients with CLL. Blood 2015; 126:573-81. [PMID: 26084672 DOI: 10.1182/blood-2015-03-567388] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/08/2015] [Indexed: 12/22/2022] Open
Abstract
Immune dysregulation is a cardinal feature of chronic lymphocytic leukemia (CLL) from its early stage and worsens during clinical observation, even in absence of disease progression. Although the mechanisms remain unclear, new insights are emerging into the complex relationship between the CLL clone and its immune environment. T cells are increased in early-stage disease and show progressive accumulation and exhaustion. The mechanisms that drive this expansion may include auto-antigens involved in the original clonal expansion. In addition, chronic viral infections such as cytomegalovirus generate huge virus-specific immune responses, which are further expanded in CLL. Attention is now focused largely on the direct immunosuppressive properties of the tumor. Remarkably, CLL clones often have features of the recently described regulatory B cells producing immunosuppressive IL-10. Better knowledge of the regulatory properties intrinsic to CLL cells may soon become more important with the switch from chemotherapy-based treatments, which trade control of CLL with further impairment of immune function, to the new agents targeting CLL B-cell receptor-associated signaling. Treatment with these new agents is associated with evidence of immune recovery and reduced infectious complications. As such, they offer the prospect of immunologic rehabilitation and a platform from which to ultimately replace chemotherapy.
Collapse
|
81
|
Szerafin L, Jakó J, Riskó F. [Prognostic value of absolute monocyte count in chronic lymphocytic leukaemia]. Orv Hetil 2015; 156:592-7. [PMID: 25845318 DOI: 10.1556/oh.2015.30126] [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] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The low peripheral absolute lymphocyte and high monocyte count have been reported to correlate with poor clinical outcome in various lymphomas and other cancers. However, a few data known about the prognostic value of absolute monocyte count in chronic lymphocytic leukaemia. AIM The aim of the authors was to investigate the impact of absolute monocyte count measured at the time of diagnosis in patients with chronic lymphocytic leukaemia on the time to treatment and overal survival. METHOD Between January 1, 2005 and December 31, 2012, 223 patients with newly-diagnosed chronic lymphocytic leukaemia were included. The rate of patients needing treatment, time to treatment, overal survival and causes of mortality based on Rai stages, CD38, ZAP-70 positivity and absolute monocyte count were analyzed. RESULTS Therapy was necessary in 21.1%, 57.4%, 88.9%, 88.9% and 100% of patients in Rai stage 0, I, II, III an IV, respectively; in 61.9% and 60.8% of patients exhibiting CD38 and ZAP-70 positivity, respectively; and in 76.9%, 21.2% and 66.2% of patients if the absolute monocyte count was <0.25 G/l, between 0.25-0.75 G/l and >0.75 G/l, respectively. The median time to treatment and the median overal survival were 19.5, 65, and 35.5 months; and 41.5, 65, and 49.5 months according to the three groups of monocyte counts. The relative risk of beginning the therapy was 1.62 (p<0.01) in patients with absolute monocyte count <0.25 G/l or >0.75 G/l, as compared to those with 0.25-0.75 G/l, and the risk of overal survival was 2.41 (p<0.01) in patients with absolute monocyte count lower than 0.25 G/l as compared to those with higher than 0.25 G/l. The relative risks remained significant in Rai 0 patients, too. The leading causes of mortality were infections (41.7%) and the chronic lymphocytic leukaemia (58.3%) in patients with low monocyte count, while tumours (25.9-35.3%) and other events (48.1 and 11.8%) occurred in patients with medium or high monocyte counts. CONCLUSIONS Patients with low and high monocyte counts had a shorter time to treatment compared to patients who belonged to the intermediate monocyte count group. The low absolute monocyte count was associated with increased mortality caused by infectious complications and chronic lymphocytic leukaemia. The absolute monocyte count may give additional prognostic information in Rai stage 0, too.
Collapse
Affiliation(s)
- László Szerafin
- Szabolcs-Szatmár-Bereg Megyei Jósa András Oktatókórház Hematológiai Osztály Nyíregyháza Szent István u. 68. 4400
| | - János Jakó
- Szabolcs-Szatmár-Bereg Megyei Jósa András Oktatókórház Hematológiai Osztály Nyíregyháza Szent István u. 68. 4400
| | - Ferenc Riskó
- Szabolcs-Szatmár-Bereg Megyei Jósa András Oktatókórház Sürgősségi Betegellátó Centrum Nyíregyháza
| |
Collapse
|
82
|
PD-L1 checkpoint blockade prevents immune dysfunction and leukemia development in a mouse model of chronic lymphocytic leukemia. Blood 2015; 126:203-11. [PMID: 25800048 DOI: 10.1182/blood-2015-01-622936] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/14/2015] [Indexed: 01/12/2023] Open
Abstract
Blockade of the programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint augments antitumor immunity and induces durable responses in patients with solid cancers, but data on clinical efficacy in leukemias are sparse. Chronic lymphocytic leukemia (CLL) is associated with a tumor-supportive microenvironment and a dysfunctional immune system, as shown by "exhausted" T cells, defective immunologic synapse formation, and immunosuppressive myeloid cells. These defects involve aberrant expression of PD-L1 and are closely mirrored in the Eµ-TCL1 mouse model for CLL. In this study, we treated mice after adoptive transfer of Eµ-TCL1 CLL with PD-L1-blocking antibodies, which prevented CLL development and was accompanied by a reactivation of immune effector functions. This included restoration of mature macrophages and major histocompatibility complex class II-expressing dendritic cells and prevention of aberrant and exhaustion-like T-cell phenotypes. In addition, PD-L1 blockade restored CD8 T-cell cytotoxicity and immune synapse formation and normalized T-cell cytokines and proliferation ex vivo and in vivo. Our data demonstrate that early PD-L1 blockade effectively corrects leukemia-induced immune dysfunction and thus prevents CLL development in mice. Targeting PD-L1/PD-1 interactions should therefore be further explored in clinical studies with CLL patients, ideally in combination with novel compounds to help eliminate CLL.
Collapse
|
83
|
Boissard F, Fournié JJ, Laurent C, Poupot M, Ysebaert L. Nurse like cells: chronic lymphocytic leukemia associated macrophages. Leuk Lymphoma 2015; 56:1570-2. [PMID: 25586606 DOI: 10.3109/10428194.2014.991731] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD14 + cells are able to differentiate into large and adherent cells if in contact with chronic lymphocytic leukemia (CLL) cells or healthy B lymphocytes. In CLL these cells, called CLL-nurse like cells (NLCs), express a very high amount of CD163 and CD68 and are able to rescue CLL cells through CCL4 production. Adherent cells derived from healthy donors, called HD-NLCs, express very little CD163 and CD68, do not produce CCL4 and are unable to rescue CCL cells. This study reveals that CLL-NLCs are the specific nurse cells in CLL, protecting CLL cells from death.
Collapse
Affiliation(s)
- Frédéric Boissard
- INSERM UMR1037 - Cancer Research Center of Toulouse , Toulouse , France
| | | | | | | | | |
Collapse
|
84
|
Extracellular nicotinamide phosphoribosyltransferase (NAMPT) promotes M2 macrophage polarization in chronic lymphocytic leukemia. Blood 2015; 125:111-23. [DOI: 10.1182/blood-2014-07-589069] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Key Points
CLL lymphocytes show high intracellular and extracellular NAMPT levels, further increased upon activation. eNAMPT prompts differentiation of CLL monocytes into M2 macrophages that sustain CLL survival and reduce T-cell proliferation.
Collapse
|
85
|
Park HR, Lee SE, Yang H, Son GW, Park YS. Functional screening of altered microRNA expression in 3-methylcholanthrene-treated human umbilical vein endothelial cells. BIOCHIP JOURNAL 2014. [DOI: 10.1007/s13206-014-8403-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
86
|
ten Hacken E, Burger JA. Microenvironment dependency in Chronic Lymphocytic Leukemia: The basis for new targeted therapies. Pharmacol Ther 2014; 144:338-48. [PMID: 25050922 DOI: 10.1016/j.pharmthera.2014.07.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/11/2014] [Indexed: 02/03/2023]
Abstract
Chronic Lymphocytic Leukemia (CLL) is a prototype microenvironment-dependent B-cell malignancy, in which the neoplastic B cells co-evolve together with a supportive tissue microenvironment, which promotes leukemia cell survival, growth, and drug-resistance. Chemo-immunotherapy is an established treatment modality for CLL patients, resulting in high rates of responses and improved survival, especially in low-risk CLL. New, alternative treatments target B-cell receptor (BCR) signaling and the Chemokine (C-X-C motif) Receptor 4 (CXCR4)-Chemokine (C-X-C motif) Ligand 12 (CXCL12) axis, which are key pathways of CLL-microenvironment cross talk. The remarkable clinical efficacy of inhibitors targeting the BCR-associated kinases Bruton's tyrosine kinase (BTK) and phosphoinositide 3-kinase delta (PI3Kδ) challenges established therapeutic paradigms and corroborates the central role of BCR signaling in CLL pathogenesis. In this review, we discuss the cellular and molecular components of the CLL microenvironment. We also describe the emerging therapeutic options for CLL patients, with a focus on inhibitors of CXCR4-CXCL12 and BCR signaling.
Collapse
Affiliation(s)
- Elisa ten Hacken
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Jan A Burger
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
87
|
Fiorcari S, Martinelli S, Bulgarelli J, Audrito V, Zucchini P, Colaci E, Potenza L, Narni F, Luppi M, Deaglio S, Marasca R, Maffei R. Lenalidomide interferes with tumor-promoting properties of nurse-like cells in chronic lymphocytic leukemia. Haematologica 2014; 100:253-62. [PMID: 25398834 DOI: 10.3324/haematol.2014.113217] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lenalidomide is an immunomodulatory agent clinically active in chronic lymphocytic leukemia patients. The specific mechanism of action is still undefined, but includes modulation of the microenvironment. In chronic lymphocytic leukemia patients, nurse-like cells differentiate from CD14(+) mononuclear cells and protect chronic lymphocytic leukemia cells from apoptosis. Nurse-like cells resemble M2 macrophages with potent immunosuppressive functions. Here, we examined the effect of lenalidomide on the monocyte/macrophage population in chronic lymphocytic leukemia patients. We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. We observed that lenalidomide increases the number of nurse-like cells that lost the ability to nurture chronic lymphocytic leukemia cells, acquired properties of phagocytosis and promoted T-cell proliferation. Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Our data provide new insights into the mechanism of action of lenalidomide that mediates a pro-inflammatory switch of nurse-like cells affecting the protective microenvironment generated by chronic lymphocytic leukemia into tissues.
Collapse
Affiliation(s)
- Stefania Fiorcari
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jenny Bulgarelli
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Audrito
- Department of Medical Sciences, University of Turin and Human Genetics Foundation, Italy
| | - Patrizia Zucchini
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Colaci
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Narni
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin and Human Genetics Foundation, Italy
| | - Roberto Marasca
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossana Maffei
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
88
|
Aguirre Palma LM, Gehrke I, Kreuzer KA. Angiogenic factors in chronic lymphocytic leukaemia (CLL): Where do we stand? Crit Rev Oncol Hematol 2014; 93:225-36. [PMID: 25459668 DOI: 10.1016/j.critrevonc.2014.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/23/2014] [Accepted: 10/01/2014] [Indexed: 01/09/2023] Open
Abstract
The role of angiogenesis in haematological malignancies such as chronic lymphocytic leukaemia (CLL) is difficult to envision, because leukaemia cells are not dependent on a network of blood vessels to support basic physiological requirements. Regardless, CLL cells secrete high levels of major angiogenic factors, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and platelet derived growth factor (PDGF). Nonetheless, it remains unclear how most angiogenic factors regulate accumulation and delayed apoptosis of CLL cells. Angiogenic factors such as leptin, granulocyte colony-stimulating factor (G-CSF), follistatin, angiopoietin-1 (Ang1), angiogenin (ANG), midkine (MK), pleiotrophin (PTN), progranulin (PGRN), proliferin (PLF), placental growth factor (PIGF), and endothelial locus-1 (Del-1), represent novel therapeutic targets of future CLL research but have remained widely overlooked. This review aims to outline our current understanding of angiogenic growth factors and their relationship with CLL, a still uncured haematopoietic malignancy.
Collapse
Affiliation(s)
| | - Iris Gehrke
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, MB, Canada.
| | - Karl-Anton Kreuzer
- Department I of Internal Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
89
|
Radom-Aizik S, Zaldivar FP, Haddad F, Cooper DM. Impact of brief exercise on circulating monocyte gene and microRNA expression: implications for atherosclerotic vascular disease. Brain Behav Immun 2014; 39:121-9. [PMID: 24423463 PMCID: PMC4101903 DOI: 10.1016/j.bbi.2014.01.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 12/24/2013] [Accepted: 01/04/2014] [Indexed: 12/14/2022] Open
Abstract
Physical activity can prevent and/or attenuate atherosclerosis, a disease clearly linked to inflammation. Paradoxically, even brief exercise induces a stress response and increases inflammatory cells like monocytes in the circulation. We hypothesized that exercise would regulate the expression of genes, gene pathways, and microRNAs in monocytes in a way that could limit pro-inflammatory function and drive monocytes to prevent, rather than contribute to, atherosclerosis. Twelve healthy men (22-30year old) performed ten 2-min bouts of cycle ergometer exercise at a constant work equivalent to an average of 82% of maximum O2 consumption interspersed with 1-min rest. Blood was drawn before and immediately after the exercise. Monocytes were isolated from peripheral blood mononuclear cells. Flow cytometry was used to identify monocyte subtypes. We used Affymetrix U133 + 2.0 arrays for gene expression and Agilent Human miRNA V2 Microarray for miRNAs. A stringent statistical approach (FDR <0.05) was used to determine that exercise significantly altered the expression of 894 annotated genes and 19 miRNAs. We found distinct gene alterations that were likely to direct monocytes in an anti-inflammatory, anti-atherogenic pathway, including the downregulation of monocyte TNF, TLR4, and CD36 genes and the upregulation of EREG and CXCR4. Exercise significantly altered a number of microRNAs that likely influence monocytes involvement in vascular health. Exercise leads to a novel genomic profile of circulating monocytes, which appears to promote cardiovascular health despite the overall stress response.
Collapse
Affiliation(s)
- Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine, United States.
| | - Frank P. Zaldivar
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| | - Fadia Haddad
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| | - Dan M. Cooper
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| |
Collapse
|
90
|
Maffei R, Bulgarelli J, Fiorcari S, Martinelli S, Castelli I, Valenti V, Rossi D, Bonacorsi G, Zucchini P, Potenza L, Vallisa D, Gattei V, Poeta GD, Forconi F, Gaidano G, Narni F, Luppi M, Marasca R. Endothelin-1 promotes survival and chemoresistance in chronic lymphocytic leukemia B cells through ETA receptor. PLoS One 2014; 9:e98818. [PMID: 24901342 PMCID: PMC4046988 DOI: 10.1371/journal.pone.0098818] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/07/2014] [Indexed: 11/26/2022] Open
Abstract
The endothelin axis, comprising endothelins (ET-1, ET-2 and ET-3) and their receptors (ET(A)R and ETBR), has emerged as relevant player in tumor growth and metastasis. Here, we investigated the involvement of ET-1/ET(A)R axis in chronic lymphocytic leukemia (CLL). CLL cells expressed higher levels of ET-1 and ETA receptor as compared to normal B cells. ET-1 peptide stimulated phosphoinositide-3-kinase and mitogen-activated protein kinase signaling pathways, improved survival and promoted proliferation of leukemic cells throughout ET(A)R triggering. Moreover, the blockade of ET(A)R by the selective antagonist BQ-123 inhibited the survival advantage acquired by CLL cells in contact with endothelial layers. We also found that blocking ET(A)R via BQ-123 interferes with ERK phosphorylation and CLL pro-survival effect mediated by B-cell receptor (BCR) activation. The pro-apoptotic effect of phosphoinositide-3-kinase δ inhibitor idelalisib and mitogen-activated protein kinase inhibitor PD98059 was decreased by the addition of ET-1 peptide. Then, ET-1 also reduced the cytotoxic effect of fludarabine on CLL cells cultured alone or co-cultured on endothelial layers. ET(A)R blockade by BQ-123 inhibited the ET-1-mediated protection against drug-induced apoptosis. Lastly, higher plasma levels of big ET-1 were detected in patients (n = 151) with unfavourable prognostic factors and shorter time to first treatment. In conclusion, our data describe for the first time a role of ET-1/ET(A)R signaling in CLL pathobiology. ET-1 mediates survival, drug-resistance, and growth signals in CLL cells that can be blocked by ET(A)R inhibition.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Proliferation
- Cell Survival/drug effects
- Cell Survival/genetics
- Drug Resistance, Neoplasm/genetics
- Endothelin A Receptor Antagonists/pharmacology
- Endothelin-1/blood
- Endothelin-1/genetics
- Endothelin-1/metabolism
- Gene Expression
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Prognosis
- Receptor, Endothelin A/genetics
- Receptor, Endothelin A/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/drug effects
- Treatment Outcome
Collapse
Affiliation(s)
- Rossana Maffei
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jenny Bulgarelli
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Fiorcari
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ilaria Castelli
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Davide Rossi
- Hematology Division, Department of Clinical and Experimental Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Goretta Bonacorsi
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Patrizia Zucchini
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico, I.R.C.C.S., Aviano (PN), Italy
| | - Giovanni Del Poeta
- Hematology Division, S.Eugenio Hospital and University of Tor Vergata, Rome, Italy
| | - Francesco Forconi
- Cancer Sciences Unit, CRUK Clinical Centre, University of Southampton, Southampton, United Kingdom
- Hematology Division, Department of Clinical Medicine and Immunological Sciences, University of Siena, Siena, Italy
| | - Gianluca Gaidano
- Hematology Division, Department of Clinical and Experimental Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Franco Narni
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Marasca
- Hematology Division, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
91
|
Chen JC, Johnson BA, Erikson DW, Piltonen TT, Barragan F, Chu S, Kohgadai N, Irwin JC, Greene WC, Giudice LC, Roan NR. Seminal plasma induces global transcriptomic changes associated with cell migration, proliferation and viability in endometrial epithelial cells and stromal fibroblasts. Hum Reprod 2014; 29:1255-70. [PMID: 24626806 PMCID: PMC4017943 DOI: 10.1093/humrep/deu047] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/30/2014] [Accepted: 02/12/2014] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION How does seminal plasma (SP) affect the transcriptome of human primary endometrial epithelial cells (eEC) and stromal fibroblasts (eSF)? SUMMARY ANSWER Exposure of eEC and eSF to SP in vitro increases expression of genes and secreted proteins associated with cellular migration, proliferation, viability and inhibition of cell death. WHAT IS KNOWN ALREADY Studies in both humans and animals suggest that SP can access and induce physiological changes in the upper female reproductive tract (FRT), which may participate in promoting reproductive success. STUDY DESIGN, SIZE, DURATION This is a cross sectional study involving control samples versus treatment. SP (pooled from twenty donors) was first tested for dose- and time-dependent cytotoxic effects on eEC and eSF (n = 4). As exposure of eEC or eSF to 1% SP for 6 h proved to be non-toxic, a second set of eEC/eSF samples (n = 4) was treated under these conditions for transcriptome, protein and functional analysis. With a third set of samples (n = 3), we further compared the transcriptional response of the cells to SP versus fresh semen. PARTICIPANTS/MATERIALS, SETTING, METHODS eEC and eSF were isolated from endometrial biopsies from women of reproductive age undergoing benign gynecologic procedures and maintained in vitro. RNA was isolated and processed for microarray studies to analyze global transcriptomic changes. Secreted factors in conditioned media from SP-treated cells were analyzed by Luminex and for the ability to stimulate migration of CD14+ monocytes and CD4+ T cells. MAIN RESULTS AND THE ROLE OF CHANCE Pathway identifications were determined using the Z-scoring system in Ingenuity Pathways Analysis (Z scores ≥|1.5|). SP induced transcriptomic changes (P < 0.05) associated with promoting leukocyte and endothelial cell recruitment, and proliferation of eEC and eSF. Cell viability pathways were induced, while those associated with cell death were suppressed (P < 0.05). SP and fresh semen induced similar sets of pathways, suggesting that SP can model the signaling effects of semen in the endometrium. SP also induced secretion of pro-inflammatory and pro-chemotactic cytokines, as well as pro-angiogenic and proliferative growth factors (P < 0.05) in both eEC and eSF. Finally, functional assays revealed that conditioned media from SP-treated eEC and eSF significantly increased (P < 0.05) chemotaxis of CD14+ monocytes and CD4+ T cells. LIMITATIONS, REASONS FOR CAUTION This study is limited to in vitro analyses of the effects of SP on endometrial cells. In addition, the measured response to SP was conducted in the absence of the ovarian hormones estradiol and progesterone, as well as epithelial-stromal paracrine signaling. While this study focused on establishing the baseline cellular response of endometrial cells to SP, future work should assess how hormone signaling in the presence of appropriate paracrine interactions affects SP-induced genes in these cells. WIDER IMPLICATIONS OF THE FINDINGS The results of this study support previous findings that SP and semen contain bioactive factors capable of eliciting chemotactic responses in the uterus, which can lead to recruitment of leukocytes to the endometrium. Future directions will explore if similar changes in gene expression do indeed occur after coitus in vivo, and how the signaling cascades initiated by SP in the endometrium can affect reproductive success, female reproductive health and susceptibility to sexually transmitted diseases. The gene list provided by the transcriptome analysis reported here should prove a valuable resource for understanding the response of the upper FRT to SP exposure. STUDY FUNDING/COMPETING INTEREST(S) This project was supported by NIH AI083050-04 (W.C.G./L.C.G.); NIH U54HD 055764 (L.C.G.); NIH 1F32HD074423-02 (J.C.C.); DOD W81XWH-11-1-0562 (W.C.G.); NIH 5K12-DK083021-04, NIH 1K99AI104262-01A1, The UCSF Hellman Award (N.R.R.). The authors have nothing to disclose.
Collapse
Affiliation(s)
- Joseph C. Chen
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Brittni A. Johnson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - David W. Erikson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Terhi T. Piltonen
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
- Department of Obstetrics and Gynecology and Center of Clinical Research, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Fatima Barragan
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Simon Chu
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA
| | - Nargis Kohgadai
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Juan C. Irwin
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Warner C. Greene
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA
- Department of Medicine, and Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Linda C. Giudice
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Nadia R. Roan
- Gladstone Institute of Virology and Immunology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| |
Collapse
|
92
|
Franco G, Guarnotta C, Frossi B, Piccaluga PP, Boveri E, Gulino A, Fuligni F, Rigoni A, Porcasi R, Buffa S, Betto E, Florena AM, Franco V, Iannitto E, Arcaini L, Pileri SA, Pucillo C, Colombo MP, Sangaletti S, Tripodo C. Bone marrow stroma CD40 expression correlates with inflammatory mast cell infiltration and disease progression in splenic marginal zone lymphoma. Blood 2014; 123:1836-1849. [PMID: 24452203 DOI: 10.1182/blood-2013-04-497271] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Splenic marginal zone lymphoma (SMZL) is a mature B-cell neoplasm characterized by rather indolent clinical course. However, nearly one third of patients experience a rapidly progressive disease with a dismal outcome. Despite the characterization of clone genetics and the recognition of deregulated immunologic stimulation in the pathogenesis of SMZL, little is known about microenvironment dynamics and their potential biological influence on disease outcome. Here we investigate the effect of stroma-intrinsic features on SMZL disease progression by focusing on the microenvironment of the bone marrow (BM), which represents an elective disease localization endorsing diagnostic and prognostic relevance. We show that the quality of the BM stromal meshwork of SMZL infiltrates correlates with time to progression. In particular, we describe the unfavorable prognostic influence of dense CD40 expression by BM stromal cells, which involves the contribution of CD40 ligand (CD40L)-expressing bystander mast cells infiltrating SMZL BM aggregates. The CD40/CD40L-assisted crosstalk between mesenchymal stromal cells and mast cells populating the SMZL microenvironment finds correlation in p53(-/-) mice developing SMZL and contributes to the engendering of detrimental proinflammatory conditions. Our study highlights a dynamic interaction, playing between nonneoplastic elements within the SMZL niche, toward disease progression.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CD40 Antigens/metabolism
- CD40 Ligand/metabolism
- Cell Differentiation
- Cell Proliferation
- Cytokines/biosynthesis
- Disease Progression
- Disease-Free Survival
- Female
- Genes, p53
- Humans
- Inflammation Mediators/metabolism
- Lymphoma, B-Cell, Marginal Zone/etiology
- Lymphoma, B-Cell, Marginal Zone/immunology
- Lymphoma, B-Cell, Marginal Zone/pathology
- Male
- Mast Cells/immunology
- Mast Cells/pathology
- Mesenchymal Stem Cells/immunology
- Mesenchymal Stem Cells/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Middle Aged
- Prognosis
- Tumor Microenvironment/immunology
Collapse
|
93
|
Giannoni P, Pietra G, Travaini G, Quarto R, Shyti G, Benelli R, Ottaggio L, Mingari MC, Zupo S, Cutrona G, Pierri I, Balleari E, Pattarozzi A, Calvaruso M, Tripodo C, Ferrarini M, de Totero D. Chronic lymphocytic leukemia nurse-like cells express hepatocyte growth factor receptor (c-MET) and indoleamine 2,3-dioxygenase and display features of immunosuppressive type 2 skewed macrophages. Haematologica 2014; 99:1078-87. [PMID: 24561793 DOI: 10.3324/haematol.2013.091405] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatocyte growth factor, produced by stromal and follicular dendritic cells, and present at high concentrations in the sera of patients with chronic lymphocytic leukemia, prolongs the survival of leukemic B cells by interacting with their receptor, c-MET. It is, however, unknown whether hepatocyte growth factor influences microenvironmental cells, such as nurse-like cells, which deliver survival signals to the leukemic clone. We evaluated the expression of c-MET on nurse-like cells and monocytes from patients with chronic lymphocytic leukemia and searched for phenotypic/functional features supposed to be influenced by the hepatocyte growth factor/c-MET interaction. c-MET is expressed at high levels on nurse-like cells and at significantly higher levels than normal on monocytes from patients. Moreover, the hepatocyte growth factor/c-MET interaction activates STAT3(TYR705) phosphorylation in nurse-like cells. Indoleamine 2,3-dioxygenase, an enzyme modulating T-cell proliferation and induced on normal monocytes after hepatocyte growth factor treatment, was detected together with interleukin-10 on nurse-like cells, and on freshly-prepared patients' monocytes. Immunohistochemical/immunostaining analyses demonstrated the presence of c-MET(+) and indoleamine 2,3-dioxygenase(+) cells in lymph node biopsies, co-expressed with CD68 and vimentin. Furthermore nurse-like cells and chronic lymphocytic monocytes significantly inhibited T-cell proliferation, prevented by anti-transforming growth factor beta and interleukin-10 antibodies and indoleamine 2,3-dioxygenase inhibitors, and supported CD4(+)CD25(high+)/FOXP3(+) T regulatory cell expansion. We suggest that nurse-like cells display features of immunosuppressive type 2 macrophages: higher hepatocyte growth factor levels, produced by leukemic or other microenvironmental surrounding cells, may cooperate to induce M2 polarization. Hepatocyte growth factor may thus have a dual pathophysiological role: directly through enhancement of survival of the leukemic clone and indirectly by favoring T-cell immunosuppression.
Collapse
Affiliation(s)
- Paolo Giannoni
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Gabriella Pietra
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Giorgia Travaini
- Transfer Gene Laboratory, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Rodolfo Quarto
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Genti Shyti
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Roberto Benelli
- Immunology, Dept. of Integrated Oncology Therapies, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Laura Ottaggio
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Maria Cristina Mingari
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy Excellence Center for Biomedical Research, University of Genoa, Genoa, Italy Immunology, Dept. of Integrated Oncology Therapies, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Simona Zupo
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Giovanna Cutrona
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Ivana Pierri
- Clinical Oncohematology, University of Genoa, Genoa, Italy
| | - Enrico Balleari
- Clinic of Internal Medicine 3, Dept. of Hematology, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Marco Calvaruso
- Pathological Anatomy, Tumor Immunology Unit, Dept. of Science for Health Promotion and Maternal Infantile "G. D'Alessandro", Palermo, Italy
| | - Claudio Tripodo
- Pathological Anatomy, Tumor Immunology Unit, Dept. of Science for Health Promotion and Maternal Infantile "G. D'Alessandro", Palermo, Italy
| | | | | |
Collapse
|
94
|
Peripheral blood lymphocyte/monocyte ratio predicts outcome for patients with diffuse large B cell lymphoma after standard first-line regimens. Ann Hematol 2013; 93:617-26. [PMID: 24141331 DOI: 10.1007/s00277-013-1916-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/26/2013] [Indexed: 02/06/2023]
Abstract
To determine whether peripheral blood absolute lymphocyte/absolute monocyte counts ratio (ALC/AMC ratio) at diagnosis predicts survival of diffuse large B cell lymphoma (DLBCL) patients treated with standard first-line regimens, we retrospectively analyzed 244 patients with DLBCL who were treated with standard cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone, or rituximab-cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone. Progression-free survival and overall survival (PFS and OS) were estimated using the Kaplan-Meier method and two-tailed log-rank; The Cox proportional hazards model was used to evaluate ALC/AMC ratio as prognostic factors when adjusting for the International Prognostic Index (IPI). On univariate and multivariate analyses performed with factors included in the IPI, the ALC/AMC ratio at diagnosis remained an independent predictor of OS and PFS (OS: P < 0.001; PFS: P < 0.001). Patients with lower ALC/AMC ratio (<3.8) seemed to have lower complete remission rate, 2-year PFS and 3-year OS when compared to patients with ALC/AMC ratio ≥3.8, respectively (26 versus 90 %, P < 0.001; 18 versus 82 %, P < 0.001; 24 versus 86 %; P < 0.001, respectively). Moreover, the ALC/AMC ratio was able to further risk-stratify IPI 0-2 and three-five risk patient groups, respectively. The ALC/AMC ratio at the time of diagnosis may provide additional prognostic information beyond that of the IPI for patients with DLBCL who receive standard first-line regimens.
Collapse
|
95
|
Herishanu Y, Kay S, Sarid N, Kohan P, Braunstein R, Rotman R, Deutsch V, Ben-Ezra J, Naparstek E, Perry C, Katz BZ. Absolute monocyte count trichotomizes chronic lymphocytic leukemia into high risk patients with immune dysregulation, disease progression and poor survival. Leuk Res 2013; 37:1222-8. [DOI: 10.1016/j.leukres.2013.07.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/18/2013] [Indexed: 10/26/2022]
|