101
|
Höchst B, Mikulec J, Baccega T, Metzger C, Welz M, Peusquens J, Tacke F, Knolle P, Kurts C, Diehl L, Ludwig-Portugall I. Differential induction of Ly6G and Ly6C positive myeloid derived suppressor cells in chronic kidney and liver inflammation and fibrosis. PLoS One 2015; 10:e0119662. [PMID: 25738302 PMCID: PMC4349817 DOI: 10.1371/journal.pone.0119662] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/28/2015] [Indexed: 12/20/2022] Open
Abstract
CD11b+Gr1+ myeloid derived suppressor cells (MDSC) are known to be very potent suppressors of T cell immunity and can be further stratified into granulocytic MDSC and monocytic MDSC in mice based on expression of Ly6G or Ly6C, respectively. Here, using these markers and functional assays, we aimed to identify whether MDSC are induced during chronic inflammation leading to fibrosis in both kidney and liver and whether additional markers could more specifically identify these MDSC subsets. In an adenine-induced model of kidney inflammation/fibrosis suppressive Ly6Gpos MDSC were induced. The suppressive function within the Ly6G+ MDSC population was exclusively present in IFNγRβ expressing cells. In contrast, in chronic inflammation in the liver induced by bile duct ligation, suppressive capacity was exclusively present in the Ly6Cpos MDSC subset. Gene expression analyses confirmed the differential origins and regulation of those MDSC subsets. Additionally, depletion of MDSC in either kidney or liver fibrosis enhanced fibrosis markers, indicating a protective role for MDSC in organ fibrosis. Thus, our data demonstrate that during liver inflammation and kidney fibrosis MDSC with similar function arise bearing a distinct marker profile and arising from different cell populations.
Collapse
Affiliation(s)
- Bastian Höchst
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Molecular Immunology, Technische Universität München, Munich, Germany
| | - Julita Mikulec
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
| | - Tania Baccega
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
| | | | - Meike Welz
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
| | - Julia Peusquens
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Percy Knolle
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Molecular Immunology, Technische Universität München, Munich, Germany
| | - Christian Kurts
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
| | - Linda Diehl
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (LD); (ILP)
| | - Isis Ludwig-Portugall
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
- * E-mail: (LD); (ILP)
| |
Collapse
|
102
|
Damuzzo V, Pinton L, Desantis G, Solito S, Marigo I, Bronte V, Mandruzzato S. Complexity and challenges in defining myeloid-derived suppressor cells. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2015; 88:77-91. [PMID: 25504825 PMCID: PMC4405078 DOI: 10.1002/cyto.b.21206] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 12/23/2022]
Abstract
Study of myeloid cells endowed with suppressive activity is an active field of research which has particular importance in cancer, in view of the negative regulatory capacity of these cells to the host's immune response. The expansion of these cells, called myeloid-derived suppressor cells (MDSCs), has been documented in many models of tumor-bearing mice and in patients with tumors of various origin, and their presence is associated with disease progression and reduced survival. For this reason, monitoring this type of cell expansion is of clinical importance, and flow cytometry is the technique of choice for their identification. Over the years, it has been demonstrated that MDSCs comprise a group of immature myeloid cells belonging both to monocytic and granulocytic lineages, with several stages of differentiation; their occurrence depends on tumor-derived soluble factors, which guide their expansion and determine their block of differentiation. Because of their heterogeneous composition, accurate phenotyping of these cells requires a multicolor approach, so that the expansion of all MDSC subsets can be appreciated. This review article focuses on identifying MDSCs and discusses problems associated with phenotyping circulating and tumor-associated MDSCs in humans and in mouse models.
Collapse
Affiliation(s)
- Vera Damuzzo
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | - Laura Pinton
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | | | - Samantha Solito
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | | | - Vincenzo Bronte
- Section of Immunology, Department of Pathology, Verona University HospitalVerona, Italy
| | - Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
- Veneto Institute of Oncology IOV—IRCCSPadova, Italy
| |
Collapse
|
103
|
Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 2015; 4:e954829. [PMID: 25949858 PMCID: PMC4368153 DOI: 10.4161/21624011.2014.954829] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/07/2014] [Indexed: 01/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) contribute to tumor-mediated immune escape and negatively correlate with overall survival of cancer patients. Nowadays, a variety of methods to target MDSCs are being investigated. Based on the intervention stage of MDSCs, namely development, expansion and activation, function and turnover, these methods can be divided into: (I) prevention or differentiation to mature cells, (II) blockade of MDSC expansion and activation, (III) inhibition of MDSC suppressive activity or (IV) depletion of intratumoral MDSCs. This review describes effective mono- or multimodal-therapies that target MDSCs for the benefit of cancer treatment.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- 5-Fluorouracil
- ADAM17, metalloproteinase domain-containing protein 17
- APCs, antigen presenting cells
- ARG1, arginase-1
- ATRA, all-trans retinoic acid
- CCL2, chemokine (C-C motif) ligand 2
- CD62L, L-selectin
- CDDO-Me, bardoxolone methyl
- COX2, cyclooxygenase 2
- CTLs, cytotoxic T lymphocytes
- CXCL12, chemokine (C-X-C motif) ligand 12
- CXCL15, chemokine (C-X-C motif) ligand 15
- DCs, dendritic cells
- ERK1/2, extracellular signal-regulated kinases
- Flt3, Fms-like tyrosine kinase 3
- FoxP3, forkhead box P3
- GITR, anti-glucocorticoid tumor necrosis factor receptor
- GM-CSF/CSF2, granulocyte monocyte colony stimulating factor
- GSH, glutathione
- HIF-1α, hypoxia inducible factor 1α
- HLA, human leukocyte antigen
- HNSCC, head and neck squamous cell carcinoma
- HPV-16, human papillomavirus 16
- HSCs, hematopoietic stem cells
- ICT, 3, 5, 7-trihydroxy-4′-emthoxy-8-(3-hydroxy-3-methylbutyl)-flavone
- IFNγ, interferon γ
- IL-10, interleukin 10
- IL-13, interleukin 13
- IL-1β, interleukin 1 β
- IL-4, interleukin 4
- IL-6, interleukin 6
- IMCs, immature myeloid cells
- JAK2, Janus kinase 2
- MDSCs, myeloid-derived suppressor cells
- MMPs, metalloproteinases (e.g., MMP9)
- Myd88, myeloid differentiation primary response protein 88
- NAC, N-acetyl cysteine
- NADPH, nicotinamide adenine dinucleotide phosphate-oxidase NK cells, natural killer cells
- NO, nitric oxide
- NOHA, N-hydroxy-L-Arginine
- NSAID, nonsteroidal anti-inflammatory drugs
- ODN, oligodeoxynucleotides
- PDE-5, phosphodiesterase type 5
- PGE2, prostaglandin E2
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SCF, stem cell factor
- STAT3, signal transducer and activator of transcription 3
- TAMs, tumor-associated macrophages
- TCR, T cell receptor
- TGFβ, transforming growth factor β
- TNFα, tumor necrosis factor α
- Tregs, regulatory T cells
- VEGFR, vascular endothelial growth factor receptor
- WA, withaferin A
- WRE, Withaferin somnifera
- all-trans retinoic acid
- bisphosphonates
- c-kit, Mast/stem cell growth factor receptor
- gemcitabine
- iNOS2, inducible nitric oxid synthase 2
- immune suppressive mechanisms
- mRCC, metastatic renal cell carcinoma
- myeloid-derived suppressor cells
- sunitinib therapeutic vaccination
Collapse
Affiliation(s)
- Oana Draghiciu
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Joyce Lubbers
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Hans W Nijman
- Department of Gynecology; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Toos Daemen
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| |
Collapse
|
104
|
Xiang D, Shigdar S, Qiao G, Wang T, Kouzani AZ, Zhou SF, Kong L, Li Y, Pu C, Duan W. Nucleic acid aptamer-guided cancer therapeutics and diagnostics: the next generation of cancer medicine. Am J Cancer Res 2015; 5:23-42. [PMID: 25553096 PMCID: PMC4265746 DOI: 10.7150/thno.10202] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/01/2014] [Indexed: 12/29/2022] Open
Abstract
Conventional anticancer therapies, such as chemo- and/or radio-therapy are often unable to completely eradicate cancers due to abnormal tumor microenvironment, as well as increased drug/radiation resistance. More effective therapeutic strategies for overcoming these obstacles are urgently in demand. Aptamers, as chemical antibodies that bind to targets with high affinity and specificity, are a promising new and novel agent for both cancer diagnostic and therapeutic applications. Aptamer-based cancer cell targeting facilitates the development of active targeting in which aptamer-mediated drug delivery could provide promising anticancer outcomes. This review is to update the current progress of aptamer-based cancer diagnosis and aptamer-mediated active targeting for cancer therapy in vivo, exploring the potential of this novel form of targeted cancer therapy.
Collapse
|
105
|
Botta C, Gullà A, Correale P, Tagliaferri P, Tassone P. Myeloid-derived suppressor cells in multiple myeloma: pre-clinical research and translational opportunities. Front Oncol 2014; 4:348. [PMID: 25538892 PMCID: PMC4258997 DOI: 10.3389/fonc.2014.00348] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/23/2014] [Indexed: 12/31/2022] Open
Abstract
Immunosuppressive cells have been reported to play an important role in tumor-progression mainly because of their capability to promote immune-escape, angiogenesis, and metastasis. Among them, myeloid-derived suppressor cells (MDSCs) have been recently identified as immature myeloid cells, induced by tumor-associated inflammation, able to impair both innate and adaptive immunity. While murine MDSCs are usually identified by the expression of CD11b and Gr1, human MDSCs represent a more heterogeneous population characterized by the expression of CD33 and CD11b, low or no HLA-DR, and variable CD14 and CD15. In particular, the last two may alternatively identify monocyte-like or granulocyte-like MDSC subsets with different immunosuppressive properties. Recently, a substantial increase of MDSCs has been found in peripheral blood and bone marrow (BM) of multiple myeloma (MM) patients with a role in disease progression and/or drug resistance. Pre-clinical models recapitulating the complexity of the MM-related BM microenvironment (BMM) are major tools for the study of the interactions between MM cells and cells of the BMM (including MDSCs) and for the development of new agents targeting MM-associated immune-suppressive cells. This review will focus on current strategies for human MDSCs generation and investigation of their immunosuppressive function in vitro and in vivo, taking into account the relevant relationship occurring within the MM–BMM. We will then provide trends in MDSC-associated research and suggest potential application for the treatment of MM.
Collapse
Affiliation(s)
- Cirino Botta
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | | | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy ; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University , Philadelphia, PA , USA
| |
Collapse
|
106
|
Chen S, Liu H, Su N, Zhang G, Wang L. Myeloid-derived suppressor cells promote age-related increase of lung cancer growth via B7-H1. Exp Gerontol 2014; 61:84-91. [PMID: 25479230 DOI: 10.1016/j.exger.2014.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/13/2014] [Accepted: 12/01/2014] [Indexed: 12/15/2022]
Abstract
Accumulation of myeloid-derived suppressor cells (MDSCs) in aged hosts contribute to the age-related increase of susceptibility to murine breast adenocarcinoma and co-stimulatory molecules expressed in MDSCs are essential for MDSCs-mediated immune suppression. However, the co-stimulatory molecules that exert a direct effect on MDSCs-mediated age-dependent tumor susceptibility and the regulatory mechanism of their expression remain unclear. In the present study, we found that accumulation of MDSCs in aged mice was closely correlated with age-dependent enhanced growth of lung cancer. Further analysis revealed that B7-H1 was highly expressed in the MDSCs of 18-month but not in 2-month old mice. Accordingly, inhibition of B7-H1 with B7-H1 specific antibody significantly reactivated T cells and reduced the tumor progression mediated by MDSCs. In addition, IL-10 released from 18-month old mice stimulated the expression of B7-H1 on MDSCs. These results suggest that B7-H1 expressed on MDSCs is a novel target for reducing lung cancer susceptibility as the age increases.
Collapse
Affiliation(s)
- Siwen Chen
- Special Procurement Ward, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Hongmei Liu
- Special Procurement Ward, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Nan Su
- Respiratory Intensive Care Unit, Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Guangbo Zhang
- Clinical Immunology Laboratory, First Affiliated Hospital, Suzhou 215006, Jiangsu Province, China
| | - Ling Wang
- Special Procurement Ward, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
107
|
Waldron TJ, Quatromoni JG, Karakasheva TA, Singhal S, Rustgi AK. Myeloid derived suppressor cells: Targets for therapy. Oncoimmunology 2014; 2:e24117. [PMID: 23734336 PMCID: PMC3654606 DOI: 10.4161/onci.24117] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 02/22/2013] [Accepted: 02/26/2013] [Indexed: 01/14/2023] Open
Abstract
The goal of achieving measurable response with cancer immunotherapy requires counteracting the immunosuppressive characteristics of tumors. One of the mechanisms that tumors utilize to escape immunosurveillance is the activation of myeloid derived suppressor cells (MDSCs). Upon activation by tumor-derived signals, MDSCs inhibit the ability of the host to mount an anti-tumor immune response via their capacity to suppress both the innate and adaptive immune systems. Despite their relatively recent discovery and characterization, anti-MDSC agents have been identified, which may improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Todd J Waldron
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA ; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | | | | | | | | |
Collapse
|
108
|
Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, Nazarian R, Califano J, Borrello I, Serafini P. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res 2014; 21:39-48. [PMID: 25320361 DOI: 10.1158/1078-0432.ccr-14-1711] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) play a key role in the progression of head and neck squamous cell carcinoma (HNSCC). On the basis of our preclinical data demonstrating that phosphodiesterase-5 (PDE5) inhibition can modulate these cell populations, we evaluated whether the PDE5 inhibitor tadalafil can revert tumor-induced immunosuppression and promote tumor immunity in patients with HNSCC. EXPERIMENTAL DESIGN First, we functionally and phenotypically characterized MDSCs in HNSCCs and determined, retrospectively, whether their presence at the tumor site correlates with recurrence. Then, we performed a prospective single-center, double-blinded, randomized, three-arm study in which patients with HNSCC undergoing definitive surgical resection of oral and oropharyngeal tumors were treated with tadalafil 10 mg/day, 20 mg/day, or placebo for at least 20 days preoperatively. Blood and tumor MDSC and Treg presence and CD8(+) T-cell reactivity to tumor antigens were evaluated before and after treatment. RESULTS MDSCs were characterized in HNSCC and their intratumoral presence significantly correlates with recurrence. Tadalafil treatment was well tolerated and significantly reduced both MDSCs and Treg concentrations in the blood and in the tumor (P < 0.05). In addition, the concentration of blood CD8(+) T cells reactive to autologous tumor antigens significantly increased after treatment (P < 0.05). Tadalafil immunomodulatory activity was maximized at an intermediate dose but not at higher doses. Mechanistic analysis suggests a possible off-target effect on PDE11 at high dosages that, by increasing intracellular cAMP, may negatively affect antitumor immunity. CONCLUSIONS Tadalafil seems to beneficially modulate the tumor micro- and macro-environment in patients with HNSCC by lowering MDSCs and Tregs and increasing tumor-specific CD8(+) T cells in a dose-dependent fashion.
Collapse
Affiliation(s)
- Donald T Weed
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Jennifer L Vella
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Isildinha M Reis
- Department of Public Health Sciences and Sylvester Biostatistics and Bioinformatics Core Resource, University of Miami, Miller School of Medicine, Miami, Florida
| | - Adriana C De la Fuente
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Carmen Gomez
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Zoukaa Sargi
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ronen Nazarian
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Joseph Califano
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland.,Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, Maryland
| | - Ivan Borrello
- Oncology Department, Johns Hopkins University, Baltimore, Maryland
| | - Paolo Serafini
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
109
|
Gervassi A, Lejarcegui N, Dross S, Jacobson A, Itaya G, Kidzeru E, Gantt S, Jaspan H, Horton H. Myeloid derived suppressor cells are present at high frequency in neonates and suppress in vitro T cell responses. PLoS One 2014; 9:e107816. [PMID: 25248150 PMCID: PMC4172591 DOI: 10.1371/journal.pone.0107816] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 08/04/2014] [Indexed: 12/26/2022] Open
Abstract
Over 4 million infants die each year from infections, many of which are vaccine-preventable. Young infants respond relatively poorly to many infections and vaccines, but the basis of reduced immunity in infants is ill defined. We sought to investigate whether myeloid-derived suppressor cells (MDSC) represent one potential impediment to protective immunity in early life, which may help inform strategies for effective vaccination prior to pathogen exposure. We enrolled healthy neonates and children in the first 2 years of life along with healthy adult controls to examine the frequency and function of MDSC, a cell population able to potently suppress T cell responses. We found that MDSC, which are rarely seen in healthy adults, are present in high numbers in neonates and their frequency rapidly decreases during the first months of life. We determined that these neonatal MDSC are of granulocytic origin (G-MDSC), and suppress both CD4+ and CD8+ T cell proliferative responses in a contact-dependent manner and gamma interferon production. Understanding the role G-MDSC play in infant immunity could improve vaccine responsiveness in newborns and reduce mortality due to early-life infections.
Collapse
Affiliation(s)
- Ana Gervassi
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Nicholas Lejarcegui
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Sandra Dross
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- University of Washington Department of Global Health, Seattle, Washington, United States of America
| | - Amanda Jacobson
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Grace Itaya
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Elvis Kidzeru
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Soren Gantt
- University of British Columbia Department of Pediatrics and Child and Family Research Institute, Vancouver, Canada
| | - Heather Jaspan
- University of Washington Seattle Children's Hospital, Seattle, Washington, United States of America
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Helen Horton
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- University of Washington Department of Global Health, Seattle, Washington, United States of America
- University of Washington Department of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
110
|
Sun H, Zhu X, Lu PY, Rosato RR, Tan W, Zu Y. Oligonucleotide aptamers: new tools for targeted cancer therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e182. [PMID: 25093706 PMCID: PMC4221593 DOI: 10.1038/mtna.2014.32] [Citation(s) in RCA: 362] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Aptamers are a class of small nucleic acid ligands that are composed of RNA or single-stranded DNA oligonucleotides and have high specificity and affinity for their targets. Similar to antibodies, aptamers interact with their targets by recognizing a specific three-dimensional structure and are thus termed “chemical antibodies.” In contrast to protein antibodies, aptamers offer unique chemical and biological characteristics based on their oligonucleotide properties. Hence, they are more suitable for the development of novel clinical applications. Aptamer technology has been widely investigated in various biomedical fields for biomarker discovery, in vitro diagnosis, in vivo imaging, and targeted therapy. This review will discuss the potential applications of aptamer technology as a new tool for targeted cancer therapy with emphasis on the development of aptamers that are able to specifically target cell surface biomarkers. Additionally, we will describe several approaches for the use of aptamers in targeted therapeutics, including aptamer-drug conjugation, aptamer-nanoparticle conjugation, aptamer-mediated targeted gene therapy, aptamer-mediated immunotherapy, and aptamer-mediated biotherapy.
Collapse
Affiliation(s)
- Hongguang Sun
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Xun Zhu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Jilin, China
| | | | - Roberto R Rosato
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Wen Tan
- School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
111
|
miR-34a expands myeloid-derived suppressor cells via apoptosis inhibition. Exp Cell Res 2014; 326:259-66. [DOI: 10.1016/j.yexcr.2014.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 03/21/2014] [Accepted: 04/16/2014] [Indexed: 01/08/2023]
|
112
|
The immune system and head and neck squamous cell carcinoma: from carcinogenesis to new therapeutic opportunities. Immunol Res 2014; 57:52-69. [PMID: 24218361 DOI: 10.1007/s12026-013-8462-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) exhibit complex interactions with the host immune system that may simultaneously explain resistance to various therapeutic modalities and that may also provide opportunities for therapeutic intervention. Discoveries in immunologic research over the last decade have led to an increased understanding of these interactions as well as the development of a multitude of investigational immunotherapies. Here, we describe the interaction between HNSCC and the immune system, including a discussion of immune cells involved with tumor carcinogenesis and the role of immune-modulating factors derived from tumors. We also describe the current immunotherapeutic approaches being investigated for HNSCC, including a discussion of the successes and limitations. With this review, we hope to present HNSCC as a model to guide future research in cancer immunology.
Collapse
|
113
|
Myeloid derived suppressor cells in physiological and pathological conditions: the good, the bad, and the ugly. Immunol Res 2014; 57:172-84. [PMID: 24203443 DOI: 10.1007/s12026-013-8455-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myeloid derived suppressor cells (MDSCs), a heterogeneous population of myeloid progenitors, are recognized as a key element in tumor escape and progression. The importance of MDSCs in human malignancies has been demonstrated in recent years, and new approaches targeting their suppressive/tolerogenic action are currently being tested in both preclinical model and clinical trials. However, emerging evidence suggests that MDSCs may play a prominent role as regulator of the physiologic, the chronic, and the pathologic immune responses. This review will focus on the biology of MDSC in light of these new findings and the possible role of this myeloid population not only in the progression of the tumor but also in its initiation.
Collapse
|
114
|
Shen J, Chen X, Wang Z, Zhang G, Chen W. Downregulation of CD40 expression contributes to the accumulation of myeloid-derived suppressor cells in gastric tumors. Oncol Lett 2014; 8:775-780. [PMID: 25009656 PMCID: PMC4081434 DOI: 10.3892/ol.2014.2174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 02/20/2014] [Indexed: 12/30/2022] Open
Abstract
An elevated number of myeloid-derived suppressor cells (MDSCs) in tumor-bearing hosts has been recognized as a crucial mediator of tumor progression due to the cells potent ability to suppress antitumor immunity. Cluster of differentiation (CD) 40, as a suppressive phenotype expressed in MDSCs, is essential for MDSC-mediated immune suppression and the expansion of T regulatory cells. However, whether CD40 exerts a direct effect on the accumulation of MDSCs remains unclear. In the present study, CD40 was observed to be highly expressed on the MDSCs obtained from mice bearing gastric tumors. Notably, a significant decrease in the level of CD40 expression was observed in addition to an increased number of MDSCs during tumor progression. Further analysis revealed that the MDSC levels were found to positively correlate with tumor progression and that CD40 expression levels inversely correlate with the accumulation of MDSCs. To confirm the potent correlation between CD40 expression and the accumulation of MDSCs, the apoptosis of the MDSCs was detected using agonistic anti-CD40 treatment. The results indicated that CD40 activation induces apoptosis in MDSCs and that the downregulation of CD40 expression may contribute to MDSC accumulation by facilitating MDSC resistance to apoptosis. Thus, these observations provide a novel mechanism to improve our understanding of the involvement of CD40 in MDSC accumulation during cancer development.
Collapse
Affiliation(s)
- Jin Shen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiaojuan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zhenxing Wang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Guangbo Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China ; Key Laboratory of Medicine and Clinical Immunology of Jiangsu Province, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China ; Key Laboratory of Medicine and Clinical Immunology of Jiangsu Province, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
115
|
Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N, El-Deiry WS, Winograd R, Vonderheide RH, English NR, Knight SC, Yagita H, McCaffrey JC, Antonia S, Hockstein N, Witt R, Masters G, Bauer T, Gabrilovich DI. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J Clin Invest 2014; 124:2626-39. [PMID: 24789911 DOI: 10.1172/jci74056] [Citation(s) in RCA: 303] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) dampen the immune response thorough inhibition of T cell activation and proliferation and often are expanded in pathological conditions. Here, we studied the fate of MDSCs in cancer. Unexpectedly, MDSCs had lower viability and a shorter half-life in tumor-bearing mice compared with neutrophils and monocytes. The reduction of MDSC viability was due to increased apoptosis, which was mediated by increased expression of TNF-related apoptosis-induced ligand receptors (TRAIL-Rs) in these cells. Targeting TRAIL-Rs in naive mice did not affect myeloid cell populations, but it dramatically reduced the presence of MDSCs and improved immune responses in tumor-bearing mice. Treatment of myeloid cells with proinflammatory cytokines did not affect TRAIL-R expression; however, induction of ER stress in myeloid cells recapitulated changes in TRAIL-R expression observed in tumor-bearing hosts. The ER stress response was detected in MDSCs isolated from cancer patients and tumor-bearing mice, but not in control neutrophils or monocytes, and blockade of ER stress abrogated tumor-associated changes in TRAIL-Rs. Together, these data indicate that MDSC pathophysiology is linked to ER stress, which shortens the lifespan of these cells in the periphery and promotes expansion in BM. Furthermore, TRAIL-Rs can be considered as potential targets for selectively inhibiting MDSCs.
Collapse
|
116
|
Shen XZ, Okwan-Duodu D, Blackwell WL, Ong FS, Janjulia T, Bernstein EA, Fuchs S, Alkan S, Bernstein KE. Myeloid expression of angiotensin-converting enzyme facilitates myeloid maturation and inhibits the development of myeloid-derived suppressor cells. J Transl Med 2014; 94:536-44. [PMID: 24614194 PMCID: PMC4221240 DOI: 10.1038/labinvest.2014.41] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 12/30/2013] [Accepted: 01/21/2014] [Indexed: 12/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells which accumulate in cancer, infection and chronic inflammation. These cells suppress T-cell function and the immune response. Angiotensin-converting enzyme (ACE) is a peptidase that is now known to regulate aspects of myelopoiesis. Here, we show that ACE expression correlates with myeloid maturation in vitro. Forced ACE overexpression in monocytic cells reduces the generation of MDSCs. In vivo, mice with a genetic change resulting in myeloid cell ACE overexpression have reduced numbers of blood and splenic MDSCs in a tumor model and in a model of chronic inflammation induced by complete Freund's adjuvant. In contrast, ACE-null mice produce large numbers of MDSCs during chronic inflammation. Macrophages from mice with myeloid ACE overexpressing are more pro-inflammatory and have more tumor-killing activity than cells from wild-type mice. Thus, manipulating myeloid ACE activity can interfere with MDSC development and the maturation of myeloid cells.
Collapse
Affiliation(s)
- Xiao Z. Shen
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US ,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Derick Okwan-Duodu
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US ,School of Medicine, Emory University, Atlanta, GA, US
| | - Wendell-Lamar Blackwell
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Frank S. Ong
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Tea Janjulia
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Ellen A. Bernstein
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Sebastien Fuchs
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US ,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Serhan Alkan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, US
| | - Kenneth E. Bernstein
- Division of Immunology, Department of Biomedical Science; Cedars-Sinai Medical Center, Los Angeles, CA, US ,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, US
| |
Collapse
|
117
|
Medina-Echeverz J, Aranda F, Berraondo P. Myeloid-derived cells are key targets of tumor immunotherapy. Oncoimmunology 2014; 3:e28398. [PMID: 25050208 PMCID: PMC4063142 DOI: 10.4161/onci.28398] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/14/2014] [Accepted: 03/01/2014] [Indexed: 12/22/2022] Open
Abstract
Tumors are composed of heterogeneous cell populations recruited by cancer cells to promote growth and metastasis. Among cells comprising the tumor stroma, myeloid-derived cells play pleiotropic roles in supporting tumorigenesis at distinct stages of tumor development. The tumor-infiltrating myeloid cell contingent is composed of mast cells, neutrophils, dendritic cells, macrophages, and myeloid-derived suppressor cells. Such cells are capable of evading the hostile tumor environment typically prone to immune cell destruction and can even promote angiogenesis, chronic inflammation, and invasion. This paper briefly summarizes the different myeloid-derived subsets that promote tumor development and the strategies that have been used to counteract the protumorigenic activity of these cells. These strategies include myeloid cell depletion, reduction of recruitment, and inactivation or remodeling of cell phenotype. Combining drugs designed to target tumor myeloid cells with immunotherapies that effectively trigger antitumor adaptive immune responses holds great promise in the development of novel cancer treatments.
Collapse
Affiliation(s)
- José Medina-Echeverz
- Division of Hepatology and Gene Therapy; Center for Applied Medical Research; University of Navarra; Pamplona, Spain
| | - Fernando Aranda
- Division of Hepatology and Gene Therapy; Center for Applied Medical Research; University of Navarra; Pamplona, Spain
| | - Pedro Berraondo
- Division of Hepatology and Gene Therapy; Center for Applied Medical Research; University of Navarra; Pamplona, Spain
| |
Collapse
|
118
|
Mirghorbani M, Van Gool S, Rezaei N. Myeloid-derived suppressor cells in glioma. Expert Rev Neurother 2014; 13:1395-406. [DOI: 10.1586/14737175.2013.857603] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
119
|
Abad C, Nobuta H, Li J, Kasai A, Yong WH, Waschek JA. Targeted STAT3 disruption in myeloid cells alters immunosuppressor cell abundance in a murine model of spontaneous medulloblastoma. J Leukoc Biol 2013; 95:357-67. [PMID: 24068730 DOI: 10.1189/jlb.1012531] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although the immune system may provide early protection against cancer, tumors may exploit the healing arm of the immune system to enhance their growth and metastasis. For example, myeloid derived suppressor cells (MDSCs) are thought to promote tumor growth by several mechanisms, including the suppression of T cell activity. It has been suggested that STAT3 activation in myeloid cells modulates multiple aspects of MDSC physiology, including their expansion and activity. Whereas most animal studies investigating tumor immunology have used tumor implants, we used transgenic mice (Smo*) that spontaneously develop medulloblastoma brain tumors to investigate the temporal accumulation of MDSCs within tumors and how myeloid STAT3 disruption affects MDSC and other immune cell types. We found distinct populations of MDSC in medulloblastoma tumors, with a high prevalence of CD11b(+)Ly6G(+)Ly6C(low/-) cells, described previously by others as G-MDSCs. These were found early in tumor development, in premalignant lesions located on the surface of the cerebellum of 28-day-old mice. In fully developed tumors, pSTAT3 was found in the majority of these cells. Conditional STAT3 gene disruption in myeloid cells resulted in an enhanced proinflammatory phenotype of macrophages in Smo* mice. Moreover, a significant reduction in the abundance of G-MDSCs and Tregs was observed within tumors along with an increased presence of CD4(+) and CD8(+) cells. Despite these alterations in immune cells induced by myeloid STAT3 disruption, we found no effect on tumor incidence in Smo* mice with this deletion.
Collapse
Affiliation(s)
- Catalina Abad
- 1.David Geffen School of Medicine, UCLA, 635 Charles E. Young Dr., South, NRB 345, Los Angeles, CA 90095-7332, USA.
| | | | | | | | | | | |
Collapse
|
120
|
Banerjee J, Nilsen-Hamilton M. Aptamers: multifunctional molecules for biomedical research. J Mol Med (Berl) 2013; 91:1333-42. [PMID: 24045702 DOI: 10.1007/s00109-013-1085-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 08/14/2013] [Accepted: 09/04/2013] [Indexed: 12/28/2022]
Abstract
Aptamers are single-stranded oligonucleotides that fold into well-defined three-dimensional shapes, allowing them to bind their targets with high affinity and specificity. They can be generated through an in vitro process called "Systemic Evolution of Ligands by Exponential Enrichment" and applied for specific detection, inhibition, and characterization of various targets like small organic and inorganic molecules, proteins, and whole cells. Aptamers have also been called chemical antibodies because of their synthetic origin and their similar modes of action to antibodies. They exhibit significant advantages over antibodies in terms of their small size, synthetic accessibility, and ability to be chemically modified and thus endowed with new properties. The first generation of aptamer drug "Macugen" was available for public use within 25 years of the discovery of aptamers. With others in the pipeline for clinical trials, this emerging field of medical biotechnology is raising significant interest. However, aptamers pose different problems for their development than for antibodies that need to be addressed to achieve practical applications. It is likely that current developments in aptamer engineering will be the basis for the evolution of improved future bioanalytical and biomedical applications. The present review discusses the development of aptamers for therapeutics, drug delivery, target validation and imaging, and reviews some of the challenges to fully realizing the promise of aptamers in biomedical applications.
Collapse
Affiliation(s)
- Jayeeta Banerjee
- Biology Department, Indian Institute of Science Education and Research (IISER), 900 NCL Innovation Park, Dr. Homi Bhabha Road, Pune, 411008, India,
| | | |
Collapse
|
121
|
Kohanbash G, McKaveney K, Sakaki M, Ueda R, Mintz AH, Amankulor N, Fujita M, Ohlfest JR, Okada H. GM-CSF promotes the immunosuppressive activity of glioma-infiltrating myeloid cells through interleukin-4 receptor-α. Cancer Res 2013; 73:6413-23. [PMID: 24030977 DOI: 10.1158/0008-5472.can-12-4124] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Malignant gliomas are lethal cancers in the brain and heavily infiltrated by myeloid cells. Interleukin-4 receptor-α (IL-4Rα) mediates the immunosuppressive functions of myeloid cells, and polymorphisms in the IL-4Rα gene are associated with altered glioma risk and prognosis. In this study, we sought to evaluate a hypothesized causal role for IL-4Rα and myeloid suppressor cells in glioma development. In both mouse de novo gliomas and human glioblastoma cases, IL-4Rα was upregulated on glioma-infiltrating myeloid cells but not in the periphery or in normal brain. Mice genetically deficient for IL-4Rα exhibited a slower growth of glioma associated with reduced production in the glioma microenvironment of arginase, a marker of myeloid suppressor cells, which is critical for their T-cell inhibitory function. Supporting this result, investigations using bone marrow-derived myeloid cells showed that IL-4Rα mediates IL-13-induced production of arginase. Furthermore, glioma-derived myeloid cells suppressed T-cell proliferation in an IL-4Rα-dependent manner, consistent with their identification as myeloid-derived suppressor cells (MDSC). Granulocyte macrophage colony-stimulating factor (GM-CSF) plays a central role for the induction of IL-4Rα expression on myeloid cells, and we found that GM-CSF is upregulated in both human and mouse glioma microenvironments compared with normal brain or peripheral blood samples. Together, our findings establish a GM-CSF-induced mechanism of immunosuppression in the glioma microenvironment via upregulation of IL-4Rα on MDSCs.
Collapse
Affiliation(s)
- Gary Kohanbash
- Authors' Affiliations: Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health; Brain Tumor Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center; Departments of Neurological Surgery, Surgery, and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Romero PJ, Withington T, Marincola F. Immune evasion in acute myeloid leukemia: current concepts and future directions. J Immunother Cancer 2013; 1:1/1/13. [PMID: 24353898 PMCID: PMC3864190 DOI: 10.1186/2051-1426-1-13] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 01/22/2023] Open
Abstract
Immune responses generated against malignant cells have the potential to inhibit tumor growth, or even eliminate transformed cells before a tumor forms. However, immune tolerance mechanisms that normally protect healthy tissues from autoimmune damage pose a formidable barrier to the development of effective anti-tumor immunity. Because malignant cells are derived from self-tissues, the majority of defined tumor antigens are either shared or aberrantly expressed self-proteins. Eliciting productive T cell responses against such proteins is challenging, as most high-affinity, self-reactive T cells are purged during thymic selection. Some T cells capable of tumor antigen recognition escape thymic deletion, but are functionally inhibited by peripheral tolerance mechanisms which limit their ability to attack a developing malignancy. Alternatively, some tumors express antigens derived from mutated self-proteins, viral proteins or self proteins expressed only during embryonic development. These antigens are recognized by the immune system as foreign and could be recognized by a relatively large number of peripheral T cells. Even in this scenario, tumors evade otherwise effective T cell responses by employing potent immunosuppressive mechanisms within their local environment. In the setting for solid malignancies, such as melanoma, a growing number of putative immune evasion mechanisms have been characterized. However, acute myeloid leukemia (AML) is a systemic disease, and the pathways it exploits to subvert the host immune response may be quite different than those of a solid tumor. Much remains unknown regarding the immune escape mechanisms promoted by AML, and whether efforts to thwart tolerance may influence the progression of this disease. Here, we review current concepts of immune evasion in AML, and speculate how potentially effective immunotherapeutic strategies might be developed to reverse immune tolerance in leukemia patients in the future.
Collapse
Affiliation(s)
- Pedro J Romero
- Ludwig Center for Cancer Research, Lausanne, Switzerland
| | - Tara Withington
- Society for Immunotherapy of Cancer (SITC), Milwaukee, WI, USA
| | | |
Collapse
|
123
|
Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol 2013; 91:493-502. [PMID: 23797066 DOI: 10.1038/icb.2013.29] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 05/26/2013] [Accepted: 05/27/2013] [Indexed: 02/07/2023]
Abstract
Immunosuppressive cells, mainly myeloid-derived suppressor cells (MDSCs) and T regulatory cells, downregulate antitumour immunity and cancer immunotherapy. MDSCs are a heterogeneous group of immature myeloid cells that negatively regulate the immune responses during tumour progression, inflammation and infection. Whilst there have been extensive laboratory investigations aimed at characterising the MDSC subsets in cancer, there remains a significant gap in our understanding of their phenotypical and functional heterogeneity. In this article, we review data concerning the phenotypical and functional role of MDSCs in cancers. Importantly, we analyse the value of MDSCs as a prognostic factor in various clinical settings and the possible therapeutic approaches towards elimination of their immunosuppressive activity and enhancement of beneficial antitumour immune responses. MDSCs promote tumour immune evasion by inhibiting T-cell responses, as well as by supporting tumour progression. Accumulation of MDSCs is associated with the progression of human cancers, and their elimination was shown to improve anti-tumour immune responses. Phenotypical characterisation of MDSCs has been poorly investigated in many human cancers and lacks comprehensive clinicopathological correlation data. Although the need for effective therapeutic agents to eliminate the MDSC suppressive effect is immense, their role has been examined only in a few clinical settings.
Collapse
Affiliation(s)
- Yazan S Khaled
- 1] Institutes of Cancer and Cardiovascular Sciences, University of Manchester, Manchester, UK [2] Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Manchester, UK [3] Department of Upper Gastrointestinal Surgery, Salford Royal Foundation Trust, Manchester, UK [4] Department of Hepatobiliary Surgery, North Manchester General Hospital, Manchester, UK [5] Section of Translational Anaesthetic and Surgical Sciences, Leeds Institute of Molecular Medicine, Leeds, UK
| | | | | |
Collapse
|
124
|
Aerts JG, Hegmans JP. Tumor-Specific Cytotoxic T Cells Are Crucial for Efficacy of Immunomodulatory Antibodies in Patients with Lung Cancer. Cancer Res 2013; 73:2381-8. [DOI: 10.1158/0008-5472.can-12-3932] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
125
|
Schilling B, Sucker A, Griewank K, Zhao F, Weide B, Görgens A, Giebel B, Schadendorf D, Paschen A. Vemurafenib reverses immunosuppression by myeloid derived suppressor cells. Int J Cancer 2013; 133:1653-63. [PMID: 23526263 DOI: 10.1002/ijc.28168] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/08/2013] [Indexed: 01/03/2023]
Abstract
Myeloid derived suppressor cells (MDSCs) suppress innate and adaptive immunity, thereby limiting anti-tumor immune responses in cancer patients. In patients with advanced melanoma, the phenotype and function of MDSCs remains controversial. In our study, we further explored two distinct subpopulations of MDSCs and investigated the impact of Vemurafenib on these cells. Flow cytometry analysis revealed that in comparison to healthy donors and patients with localized disease, PBMCs from patients with metastatic melanoma showed an increased frequency of CD14(+) HLA-DR(-/low) monocytic MDSCs (moMDSCs) and of a previously unrecognized population of CD14(-) CD66b(+) Arginase1(+) granulocytic MDSCs (grMDSCs). In vitro, both populations suppressed autologous T-cell proliferation, which was tested in CFSE-based proliferation assays. Vemurafenib treatment of melanoma patients reduced the frequency of both moMDSCs and grMDSCs. According to our in vivo finding, conditioned medium (CM) from Vemurafenib treated melanoma cells was less active in inducing moMDSCs in vitro than CM from untreated melanoma cells. In conclusion, patients with advanced melanoma show increased levels of moMDSCs, and of a population of CD14(-) CD66b(+) Arginase1(+) grMDSCs. Both MDSCs are distinct populations capable of suppressing autologous T-cell responses independently of each other. In vitro as well as in vivo, Vemurafenib inhibits the generation of human moMDSCs. Thus, Vemurafenib decreases immunosuppression in patients with advanced melanoma, indicating its potential as part of future immunotherapies.
Collapse
Affiliation(s)
- Bastian Schilling
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Bozeman EN, Cimino-Mathews A, Machiah DK, Patel JM, Krishnamoorthy A, Tien L, Shashidharamurthy R, Selvaraj P. Expression of membrane anchored cytokines and B7-1 alters tumor microenvironment and induces protective antitumor immunity in a murine breast cancer model. Vaccine 2013; 31:2449-56. [PMID: 23541884 DOI: 10.1016/j.vaccine.2013.03.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/25/2013] [Accepted: 03/15/2013] [Indexed: 01/08/2023]
Abstract
Many studies have shown that the systemic administration of cytokines or vaccination with cytokine-secreting tumors augments an antitumor immune response that can result in eradication of tumors. However, these approaches are hampered by the risk of systemic toxicity induced by soluble cytokines. In this study, we have evaluated the efficacy of 4TO7, a highly tumorigenic murine mammary tumor cell line, expressing glycosyl phosphatidylinositol (GPI)-anchored form of cytokine molecules alone or in combination with the costimulatory molecule B7-1 as a model for potential cell or membrane-based breast cancer vaccines. We observed that the GPI-anchored cytokines expressed on the surface of tumor cells greatly reduced the overall tumorigenicity of the 4TO7 tumor cells following direct live cell challenge as evidenced by transient tumor growth and complete regression within 30 days post challenge. Tumors co-expressing B7-1 and GPI-IL-12 grew the least and for the shortest duration, suggesting that this combination of immunostimulatory molecules is most potent. Protective immune responses were also observed following secondary tumor challenge. Further, the 4TO7-B7-1/GPI-IL-2 and 4TO7-B7-1/GPI-IL-12 transfectants were capable of inducing regression of a wild-type tumor growing at a distant site in a concomitant tumor challenge model, suggesting the tumor immunity elicited by the transfectants can act systemically and inhibit the tumor growth at a distant site. Additionally, when used as irradiated whole cell vaccines, 4TO7-B7-1/GPI-IL-12 led to a significant inhibition in tumor growth of day 7 established tumors. Lastly, we observed a significant decrease in the prevalence of myeloid-derived suppressor cells and regulatory T-cells in the tumor microenvironment on day 7 post challenge with 4TO7-B7-1/GPI-IL-12 cells, which provides mechanistic insight into antitumor efficacy of the tumor-cell membrane expressed IL-12. These studies have implications in designing membrane-based therapeutic vaccines with GPI-anchored cytokines for breast cancer.
Collapse
Affiliation(s)
- Erica N Bozeman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, United States.
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Kohanbash G, Okada H. Myeloid-derived suppressor cells (MDSCs) in gliomas and glioma-development. Immunol Invest 2013; 41:658-79. [PMID: 23017140 DOI: 10.3109/08820139.2012.689591] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells that inhibit anti-tumor immunity through a variety of mechanisms. Malignant gliomas are heavily infiltrated by myeloid cells, some of which appear to share biological functions of MDSCs. Our data with mouse de novo gliomas indicate critical roles of these cells in glioma development. This review summarizes the current understanding of MDSC biology in gliomas and discusses therapeutic interventions that can safely reverse the suppressive effects of MDSCs. The insight gained from these findings may lead to the development of novel immunotherapeutic strategies for gliomas.
Collapse
Affiliation(s)
- Gary Kohanbash
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
128
|
Hong EH, Chang SY, Lee BR, Pyun AR, Kim JW, Kweon MN, Ko HJ. Intratumoral injection of attenuated Salmonella vaccine can induce tumor microenvironmental shift from immune suppressive to immunogenic. Vaccine 2013; 31:1377-84. [PMID: 23318147 DOI: 10.1016/j.vaccine.2013.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 12/04/2012] [Accepted: 01/01/2013] [Indexed: 12/21/2022]
Abstract
Attenuated Salmonella vaccines show therapeutic anti-cancer effects, but the underlying mechanism has not been well investigated. In the current study, intratumoral (i.t.) injection of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine (RASV) significantly inhibited Her-2/neu-expressing tumor growth. Although depletion of CD8(+) cells in RASV-treated mice significantly restored tumor growth, the induction of Her-2/neu-specific cytotoxic T lymphocytes (CTLs) was not well correlated with the generation of the anti-tumor effect. Therefore, we hypothesized that RASV might induce a tumor microenvironmental shift, from immune suppressive to immunogenic, to reduce the suppressive force and finally elicit a successful anti-tumor response. We found that i.t. injection of RASV significantly increased the level of CD11b(+)Gr-1(+) myeloid cells identified as myeloid-derived suppressor cell (MDSC), but a significant portion of these cells were TNF-α-secreting Ly6-G(high) subsets, which can function as antitumor effector cells. We further investigated whether RASV can modulate immunosuppressive Treg cells, and CD4(+)CD25(+) Foxp3(+) Tregs was significantly reduced in RASV-treated mice. Thus, i.t. injection of RASV may offer a novel anti-cancer approach by eliciting transformation of immunosuppressive MDSCs into TNF-α-secreting neutrophils and reducing the generation of Treg cells, especially in the presence of tumor-specific CTLs. Collectively, these data will provide us an insight for the development of new anti-tumor approaches to overcome the immunosuppressive environment generated by tumors.
Collapse
Affiliation(s)
- Eun-Hye Hong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 200-701, South Korea
| | | | | | | | | | | | | |
Collapse
|
129
|
Heuvers ME, Aerts JG, Cornelissen R, Groen H, Hoogsteden HC, Hegmans JP. Patient-tailored modulation of the immune system may revolutionize future lung cancer treatment. BMC Cancer 2012; 12:580. [PMID: 23217146 PMCID: PMC3533940 DOI: 10.1186/1471-2407-12-580] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/15/2012] [Indexed: 12/16/2022] Open
Abstract
Cancer research has devoted most of its energy over the past decades on unraveling the control mechanisms within tumor cells that govern its behavior. From this we know that the onset of cancer is the result of cumulative genetic mutations and epigenetic alterations in tumor cells leading to an unregulated cell cycle, unlimited replicative potential and the possibility for tissue invasion and metastasis. Until recently it was often thought that tumors are more or less undetected or tolerated by the patient’s immune system causing the neoplastic cells to divide and spread without resistance. However, it is without any doubt that the tumor environment contains a wide variety of recruited host immune cells. These tumor infiltrating immune cells influence anti-tumor responses in opposing ways and emerges as a critical regulator of tumor growth. Here we provide a summary of the relevant immunological cell types and their complex and dynamic roles within an established tumor microenvironment. For this, we focus on both the systemic compartment as well as the local presence within the tumor microenvironment of late-stage non-small cell lung cancer (NSCLC), admitting that this multifaceted cellular composition will be different from earlier stages of the disease, between NSCLC patients. Understanding the paradoxical role that the immune system plays in cancer and increasing options for their modulation may alter the odds in favor of a more effective anti-tumor immune response. We predict that the future standard of care of lung cancer will involve patient-tailor-made combination therapies that associate (traditional) chemotherapeutic drugs and biologicals with immune modulating agents and in this way complement the therapeutic armamentarium for this disease.
Collapse
Affiliation(s)
- Marlies E Heuvers
- Department of Pulmonary Medicine, Erasmus Medical Center, Postbox 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
130
|
Zhou J, Bobbin ML, Burnett JC, Rossi JJ. Current progress of RNA aptamer-based therapeutics. Front Genet 2012; 3:234. [PMID: 23130020 PMCID: PMC3486975 DOI: 10.3389/fgene.2012.00234] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/15/2012] [Indexed: 01/07/2023] Open
Abstract
Aptamers are single-stranded nucleic acids that specifically recognize and bind tightly to their cognate targets due to their stable three-dimensional structure. Nucleic acid aptamers have been developed for various applications, including diagnostics, molecular imaging, biomarker discovery, target validation, therapeutics, and drug delivery. Due to their high specificity and binding affinity, aptamers directly block or interrupt the functions of target proteins making them promising therapeutic agents for the treatment of human maladies. Additionally, aptamers that bind to cell surface proteins are well suited for the targeted delivery of other therapeutics, such as conjugated small interfering RNAs (siRNA) that induce RNA interference (RNAi). Thus, aptamer-siRNA chimeras may offer dual-functions, in which the aptamer inhibits a receptor function, while the siRNA internalizes into the cell to target a specific mRNA. This review focuses on the current progress and therapeutic potential of RNA aptamers, including the use of cell-internalizing aptamers as cell-type specific delivery vehicles for targeted RNAi. In particular, we discuss emerging aptamer-based therapeutics that provide unique clinical opportunities for the treatment various cancers and neurological diseases.
Collapse
Affiliation(s)
- Jiehua Zhou
- Division of Molecular and Cellular Biology, Beckman Research Institute of City of Hope Duarte, CA, USA
| | | | | | | |
Collapse
|
131
|
Pratico ED, Sullenger BA, Nair SK. Identification and characterization of an agonistic aptamer against the T cell costimulatory receptor, OX40. Nucleic Acid Ther 2012; 23:35-43. [PMID: 23113766 DOI: 10.1089/nat.2012.0388] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Induction of an effective immune response that can target and eliminate malignant cells or virus-infected cells requires the stimulation of antigen-specific effector T cells. A productive and long-lasting memory response requires 2 signals: a specific signal provided by antigen recognition through engagement of the T cell receptor and a secondary signal via engagement of costimulatory molecules (such as OX40) on these newly activated T cells. The OX40-OX40-ligand interaction is critical for the generation of productive effector and memory T cell functions. Thus agonistic antibodies that stimulate OX40 on activated T cells have been used as adjuvants to augment immune responses. We previously demonstrated that an aptamer modified to stimulate murine OX40 enhanced vaccine-mediated immune responses in a murine melanoma model. In this study, we describe the development of an agonistic aptamer that targets human OX40 (hOX40). This hOX40 aptamer was isolated using systematic evolution of ligands by exponential enrichment and binds the target purified protein with high affinity [dissociation constants (K(d))<10 nM]. Moreover, the hOX40 aptamer-streptavidin complex has an apparent binding affinity of ~50 nM for hOX40 on activated T cells as determined by flow cytometry and specifically binds activated human T cells. A multivalent version of the aptamer, but not a mutant version of the aptamer, was able to stimulate OX40 on T cells and enhance cell proliferation and interferon-gamma production. Future studies will assess the therapeutic potential of hOX40 aptamers for ex vivo stimulation of antigen specific T cells in conjunction with dendritic cell-based vaccines for adoptive cellular therapy.
Collapse
Affiliation(s)
- Elizabeth D Pratico
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.
| | | | | |
Collapse
|
132
|
Zhao X, Rong L, Zhao X, Li X, Liu X, Deng J, Wu H, Xu X, Erben U, Wu P, Syrbe U, Sieper J, Qin Z. TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest 2012; 122:4094-104. [PMID: 23064360 DOI: 10.1172/jci64115] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 08/30/2012] [Indexed: 12/22/2022] Open
Abstract
TNF, an inflammatory cytokine that is enriched in the tumor microenvironment, promotes tumor growth and subverts innate immune responses to cancer cells. We previously reported that tumors implanted in TNF receptor-deficient (Tnfr-/-) mice are spontaneously rejected; however, the molecular mechanisms underlying this rejection are unclear. Here we report that TNF signaling drives the peripheral accumulation of myeloid-derived suppressor cells (MDSCs). MDSCs expand extensively during inflammation and tumor progression in mice and humans and can enhance tumor growth by repressing T cell-mediated antitumor responses. Peripheral accumulation of MDSCs was drastically impaired in Tnfr-/- mice. Signaling of TNFR-2, but not TNFR-1, promoted MDSC survival through upregulation of cellular FLICE-inhibitory protein (c-FLIP) and inhibition of caspase-8 activity. Loss of TNFRs impaired the induction of MDSCs from bone marrow cells, but this could be reversed by treatment with caspase inhibitors. These results demonstrate that TNFR-2 signaling promotes MDSC survival and accumulation and helps tumor cells evade the immune system.
Collapse
Affiliation(s)
- Xueqiang Zhao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Melanoma-induced immunosuppression and its neutralization. Semin Cancer Biol 2012; 22:319-26. [PMID: 22349515 DOI: 10.1016/j.semcancer.2012.02.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/06/2012] [Indexed: 02/08/2023]
Abstract
Malignant melanoma is characterized by a rapid progression, metastasis to distant organs, and resistance to chemo- and radiotherapy. Well-defined immunogenic capacities of melanoma cells should allow a successful application of different immunotherapeutic strategies. However, the overall results of immunotherapeutic clinical studies are not satisfactory. These paradoxical observations are supposed to be due to the profound immunosuppression mediated by different mechanisms dealing with alterations in tumor and surrounding stroma cells. Melanoma microenvironment has been characterized by a remarkable accumulation of highly immunosuppressive regulatory leucocytes, in particular, myeloid-derived suppressor cells (MDSCs). Their migration, retention and high activity in the tumor lesions have been demonstrated to be induced by chronic inflammatory conditions developing in the tumor microenvironment and characterized by the long-term secretion of various inflammatory mediators (cytokines, chemokines, growth factors, reactive oxygen and nitrogen species, prostaglandins etc.) leading to further cancer progression. Here, we discuss the role of chronic inflammation in the recruitment and activation of MDSCs in melanoma lesions as well as therapeutic approaches of MDSC targeting to overcome tumor immunosuppressive microenvironment induced by chronic inflammation and enhance the efficiency of melanoma immunotherapies.
Collapse
|