Linking Immunoevasion and Metabolic Reprogramming in B-Cell–Derived Lymphomas

Lymphomas represent a diverse group of malignancies that emerge from lymphocytes. Despite improvements in diagnosis and treatment of lymphomas of B-cell origin, relapsed and refractory disease represents an unmet clinical need. Therefore, it is of utmost importance to better understand the lymphomas’ intrinsic features as well as the interactions with their cellular microenvironment for developing novel therapeutic strategies. In fact, the role of immune-based approaches is steadily increasing and involves amongst others the use of monoclonal antibodies against tumor antigens, inhibitors of immunological checkpoints, and even genetically modified T-cells. Metabolic reprogramming and immune escape both represent well established cancer hallmarks. Tumor metabolism as introduced by Otto Warburg in the early 20th century promotes survival, proliferation, and therapeutic resistance. Simultaneously, malignant cells employ a plethora of mechanisms to evade immune surveillance. Increasing evidence suggests that metabolic reprogramming does not only confer cell intrinsic growth and survival advantages to tumor cells but also impacts local as well as systemic anti-tumor immunity. Tumor and immune cells compete over nutrients such as carbohydrates or amino acids that are critical for the immune cell function. Moreover, skewed metabolic pathways in malignant cells can result in abundant production and release of bioactive metabolites such as lactic acid, kynurenine or reactive oxygen species (ROS) that affect immune cell fitness and function. This “metabolic re-modeling” of the tumor microenvironment shifts anti-tumor immune reactivity toward tolerance. Here, we will review molecular events leading to metabolic alterations in B-cell lymphomas and their impact on anti-tumor immunity.

[1]  P. Strati,et al.  CAR T-Cell Therapy for B-Cell non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Clinical Trials and Real-World Experiences , 2020, Frontiers in Oncology.

[2]  S. Hamilton-Dutoit,et al.  Glycolytic biomarkers predict transformation in patients with follicular lymphoma , 2020, PloS one.

[3]  Jia Li,et al.  Diffuse large B-cell lymphoma with low 18F-fluorodeoxyglucose avidity features silent B-cell receptor signaling , 2020, Leukemia & lymphoma.

[4]  D. Vetrie,et al.  The leukaemia stem cell: similarities, differences and clinical prospects in CML and AML , 2020, Nature Reviews Cancer.

[5]  Xu Yixin,et al.  Adenosine Generated by Regulatory T Cells Induces CD8+ T Cell Exhaustion in Gastric Cancer through A2aR Pathway , 2019, BioMed research international.

[6]  G. Coukos,et al.  Adenosine mediates functional and metabolic suppression of peripheral and tumor-infiltrating CD8+ T cells , 2019, Journal of Immunotherapy for Cancer.

[7]  J. Tegnér,et al.  Exhaustion of CD4+ T-cells mediated by the Kynurenine Pathway in Melanoma , 2019, Scientific Reports.

[8]  A. Efeyan,et al.  Oncogenic Rag GTPase signalling enhances B cell activation and drives follicular lymphoma sensitive to pharmacological inhibition of mTOR , 2019, Nature Metabolism.

[9]  M. Horton,et al.  Targeting metabolism to regulate immune responses in autoimmunity and cancer , 2019, Nature Reviews Drug Discovery.

[10]  B. Nadel,et al.  GAPDH Expression Predicts the Response to R-CHOP, the Tumor Metabolic Status, and the Response of DLBCL Patients to Metabolic Inhibitors. , 2019, Cell metabolism.

[11]  D. Finlay,et al.  Competition for nutrients and its role in controlling immune responses , 2019, Nature Communications.

[12]  Michael L. Wang,et al.  Metabolic reprogramming toward oxidative phosphorylation identifies a therapeutic target for mantle cell lymphoma , 2019, Science Translational Medicine.

[13]  Meic H. Schmidt,et al.  Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. , 2019, Advances in biological regulation.

[14]  Ruoning Wang,et al.  A Metabolism Toolbox for CAR T Therapy , 2019, Front. Oncol..

[15]  J. Gribben,et al.  Aryl Hydrocarbon Receptor Interacting Protein Maintains Germinal Center B Cells through Suppression of BCL6 Degradation , 2019, Cell Reports.

[16]  M. Wasik,et al.  Metabolic Detection of Bruton's Tyrosine Kinase Inhibition in Mantle Cell Lymphoma Cells , 2019, Molecular Cancer Research.

[17]  W. Huber,et al.  Energy metabolism is co-determined by genetic variants in chronic lymphocytic leukemia and influences drug sensitivity , 2019, Haematologica.

[18]  Wei Zhou,et al.  Glutaminolysis Mediated by MALT1 Protease Activity Facilitates PD-L1 Expression on ABC-DLBCL Cells and Contributes to Their Immune Evasion , 2018, Front. Oncol..

[19]  R. Aloyz,et al.  Ibrutinib Resistance Is Reduced by an Inhibitor of Fatty Acid Oxidation in Primary CLL Lymphocytes , 2018, Front. Oncol..

[20]  E. Hanse,et al.  The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8+ T cells , 2018, Nature.

[21]  R. Davis,et al.  Increased Tumor Glycolysis Characterizes Immune Resistance to Adoptive T Cell Therapy. , 2018, Cell metabolism.

[22]  E. Pearce,et al.  Unraveling the Complex Interplay Between T Cell Metabolism and Function. , 2018, Annual review of immunology.

[23]  Z. Estrov,et al.  STAT3-activated CD36 facilitates fatty acid uptake in chronic lymphocytic leukemia cells , 2018, Oncotarget.

[24]  Yi Fang,et al.  Tumor-Repopulating Cells Induce PD-1 Expression in CD8+ T Cells by Transferring Kynurenine and AhR Activation. , 2018, Cancer cell.

[25]  Z. Estrov,et al.  Ibrutinib inhibits free fatty acid metabolism in chronic lymphocytic leukemia , 2018, Leukemia & lymphoma.

[26]  K. Young,et al.  PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. , 2018, Blood.

[27]  R. Davis,et al.  B-cell Receptor Signaling Regulates Metabolism in Chronic Lymphocytic Leukemia , 2017, Molecular Cancer Research.

[28]  D. Wallace,et al.  Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. , 2017, Cell metabolism.

[29]  B. Leiby,et al.  Mitochondrial and glycolytic metabolic compartmentalization in diffuse large B-cell lymphoma. , 2017, Seminars in oncology.

[30]  M. Konopleva,et al.  Targeting mantle cell lymphoma metabolism and survival through simultaneous blockade of mTOR and nuclear transporter exportin-1 , 2017, Oncotarget.

[31]  S. Loi,et al.  Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy , 2017, The Journal of clinical investigation.

[32]  D. Rossi,et al.  Adenosine signaling mediates hypoxic responses in the chronic lymphocytic leukemia microenvironment. , 2016, Blood advances.

[33]  S. Haferkamp,et al.  LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. , 2016, Cell metabolism.

[34]  B. Blanco,et al.  Targeting of PI3K/AKT/mTOR pathway to inhibit T cell activation and prevent graft-versus-host disease development , 2016, Journal of Hematology & Oncology.

[35]  Jun Yao,et al.  Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity , 2016, Nature Communications.

[36]  R. Gillies,et al.  Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. , 2016, Cancer research.

[37]  E. Pearce,et al.  Immunometabolism governs dendritic cell and macrophage function , 2016, The Journal of experimental medicine.

[38]  S. Barrans,et al.  Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma , 2015, Nature Genetics.

[39]  P. Hwu,et al.  Targeting the indoleamine 2,3-dioxygenase pathway in cancer , 2015, Journal of Immunotherapy for Cancer.

[40]  K. Akashi,et al.  Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. , 2015, Blood.

[41]  R. Moreno-Sánchez,et al.  Mitochondrial free fatty acid β-oxidation supports oxidative phosphorylation and proliferation in cancer cells. , 2015, The international journal of biochemistry & cell biology.

[42]  U. Günther,et al.  Metabolic plasticity in CLL: adaptation to the hypoxic niche , 2015, Leukemia.

[43]  H. Heslop,et al.  Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugs. , 2015, Blood.

[44]  D. Saul,et al.  Stromal cell-mediated glycolytic switch in CLL cells involves Notch-c-Myc signaling. , 2015, Blood.

[45]  Z. Estrov,et al.  Aberrant LPL Expression, Driven by STAT3, Mediates Free Fatty Acid Metabolism in CLL Cells , 2015, Molecular Cancer Research.

[46]  P. Kamper,et al.  Histologically transformed follicular lymphoma exhibits protein profiles different from both non-transformed follicular and de novo diffuse large B-cell lymphoma , 2015, Blood Cancer Journal.

[47]  T. Nakazato,et al.  Oxidative Stress Is Associated with Poor Prognosis in Patients with Follicular Lymphoma , 2014 .

[48]  N. Bec,et al.  Inhibition of CD39 Enzymatic Function at the Surface of Tumor Cells Alleviates Their Immunosuppressive Activity , 2014, Cancer Immunology Research.

[49]  D. Saul,et al.  Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia. , 2014, Blood.

[50]  Raul Rabadan,et al.  Genetics of follicular lymphoma transformation. , 2014, Cell reports.

[51]  Chih-Hao Chang,et al.  Fueling Immunity: Insights into Metabolism and Lymphocyte Function , 2013, Science.

[52]  Linda V. Sinclair,et al.  Antigen receptor control of amino acid transport coordinates the metabolic re-programming that is essential for T cell differentiation , 2013, Nature immunology.

[53]  G. Prendergast,et al.  IDO inhibits a tryptophan sufficiency signal that stimulates mTOR , 2012, Oncoimmunology.

[54]  Michael R. Green,et al.  Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. , 2012, Cancer cell.

[55]  A. Waickman,et al.  mTOR, metabolism, and the regulation of T‐cell differentiation and function , 2012, Immunological reviews.

[56]  N. Nagy,et al.  Activity and complexes of mTOR in diffuse large B-cell lymphomas—a tissue microarray study , 2012, Modern Pathology.

[57]  E. Jantunen,et al.  Oxidative stress and redox state-regulating enzymes have prognostic relevance in diffuse large B-cell lymphoma , 2012, Experimental Hematology & Oncology.

[58]  S. Olivares,et al.  Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. , 2011, Cancer research.

[59]  Hiroyasu Ito,et al.  Indoleamine 2,3-dioxygenase in tumor tissue indicates prognosis in patients with diffuse large B-cell lymphoma treated with R-CHOP , 2011, Annals of Hematology.

[60]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[61]  D. Mougiakakos,et al.  Increased thioredoxin-1 production in human naturally occurring regulatory T cells confers enhanced tolerance to oxidative stress. , 2011, Blood.

[62]  T. Habermann,et al.  A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma , 2010, Leukemia.

[63]  J. Fechner,et al.  An Interaction between Kynurenine and the Aryl Hydrocarbon Receptor Can Generate Regulatory T Cells , 2010, The Journal of Immunology.

[64]  Hiroyasu Ito,et al.  Serum concentration of L‐kynurenine predicts the clinical outcome of patients with diffuse large B‐cell lymphoma treated with R‐CHOP , 2010, European journal of haematology.

[65]  B. Coiffier,et al.  Phase III study to evaluate temsirolimus compared with investigator's choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[66]  B. Schraven,et al.  Adenosine regulates CD8 T‐cell priming by inhibition of membrane‐proximal T‐cell receptor signalling , 2009, Immunology.

[67]  Stefano Monti,et al.  SYK-dependent tonic B-cell receptor signaling is a rational treatment target in diffuse large B-cell lymphoma. , 2008, Blood.

[68]  G. Pawelec,et al.  Oxidative stress modulation and T cell activation , 2007, Experimental Gerontology.

[69]  J. Huss,et al.  Raising plasma fatty acid concentration induces increased biogenesis of mitochondria in skeletal muscle , 2007, Proceedings of the National Academy of Sciences.

[70]  V. Kuchroo,et al.  Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression , 2007, The Journal of experimental medicine.

[71]  Gregor Rothe,et al.  Inhibitory effect of tumor cell-derived lactic acid on human T cells. , 2007, Blood.

[72]  L. Moretta,et al.  The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. , 2006, Blood.

[73]  G. Laurent,et al.  Syk-dependent mTOR activation in follicular lymphoma cells. , 2006, Blood.

[74]  E. Jaffe,et al.  Constitutive activation of Akt contributes to the pathogenesis and survival of mantle cell lymphoma. , 2006, Blood.

[75]  W. Hofmann,et al.  Improved leukemia-free survival after postconsolidation immunotherapy with histamine dihydrochloride and interleukin-2 in acute myeloid leukemia: results of a randomized phase 3 trial. , 2006, Blood.

[76]  H. Döhner,et al.  High expression of lipoprotein lipase in poor risk B-cell chronic lymphocytic leukemia , 2005, Leukemia.

[77]  B. Baban,et al.  GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. , 2005, Immunity.

[78]  T. Golub,et al.  Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. , 2004, Blood.

[79]  Adrian Wiestner,et al.  A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[80]  B. Aggarwal,et al.  Adenosine suppresses activation of nuclear factor-κB selectively induced by tumor necrosis factor in different cell types , 2003, Oncogene.

[81]  E. Clementi,et al.  Mitochondrial Biogenesis in Mammals: The Role of Endogenous Nitric Oxide , 2003, Science.

[82]  G. Damonte,et al.  Tryptophan-derived Catabolites Are Responsible for Inhibition of T and Natural Killer Cell Proliferation Induced by Indoleamine 2,3-Dioxygenase , 2002, The Journal of experimental medicine.

[83]  L. Staudt,et al.  The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. , 2002, The New England journal of medicine.

[84]  F. Breedveld,et al.  Effect of Redox Balance Alterations on Cellular Localization of LAT and Downstream T-Cell Receptor Signaling Pathways , 2002, Molecular and Cellular Biology.

[85]  David Botstein,et al.  Relation of Gene Expression Phenotype to Immunoglobulin Mutation Genotype in B Cell Chronic Lymphocytic Leukemia , 2001, The Journal of experimental medicine.

[86]  R. Kiessling,et al.  Inhibition of Activated/Memory (CD45RO+) T Cells by Oxidative Stress Associated with Block of NF-κB Activation1 , 2001, The Journal of Immunology.

[87]  U. Mellqvist,et al.  Natural killer cell dysfunction and apoptosis induced by chronic myelogenous leukemia cells: role of reactive oxygen species and regulation by histamine , 2000 .

[88]  Ash A. Alizadeh,et al.  Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling , 2000, Nature.

[89]  Y. Lin,et al.  [The role of endogenous nitric oxide in airway hyperresponsiveness of asthmatic rats]. , 1999, Zhonghua jie he he hu xi za zhi = Zhonghua jiehe he huxi zazhi = Chinese journal of tuberculosis and respiratory diseases.

[90]  D. Munn,et al.  Inhibition of  T Cell Proliferation by Macrophage Tryptophan Catabolism , 1999, The Journal of experimental medicine.

[91]  B. Nathwani,et al.  A clinical evaluation of the International Lymphoma Study Group Classification of non-Hodgkin's lymphoma: a report of the Non-Hodgkin's Lymphoma Classification Project , 1997 .

[92]  T. Saito,et al.  Oxidative stress by tumor-derived macrophages suppresses the expression of CD3 zeta chain of T-cell receptor complex and antigen-specific T-cell responses. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[93]  N. Chiorazzi,et al.  mechanisms of disease Chronic Lymphocytic Leukemia , 2010 .

[94]  O. Warburg [Origin of cancer cells]. , 1956, Oncologia.

[95]  Deborah S. Barkauskas,et al.  A2AR Adenosine Signaling Suppresses Natural Killer Cell Maturation in the Tumor Microenvironment. , 2018, Cancer research.

[96]  G. Collins The next generation. , 2006, Scientific American.

[97]  D. Munn,et al.  IDO and tolerance to tumors. , 2004, Trends in molecular medicine.

[98]  D. McConkey,et al.  Increased mitochondrial biogenesis in primary leukemia cells: the role of endogenous nitric oxide and impact on sensitivity to fludarabine , 2004, Leukemia.

[99]  U. Mellqvist,et al.  Natural killer cell dysfunction and apoptosis induced by chronic myelogenous leukemia cells: role of reactive oxygen species and regulation by histamine. , 2000, Blood.

[100]  James Olen Armitage,et al.  A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma. The Non-Hodgkin's Lymphoma Classification Project. , 1997, Blood.