1
|
Immunomodulatory Actions of Mesenchymal Stromal Cells (MSCs) in Osteoarthritis of the Knee. OSTEOLOGY 2021. [DOI: 10.3390/osteology1040020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cellular therapy offers regeneration which curbs osteoarthritis of the knee. Among cellular therapies, mesenchymal stromal cells (MSCs) are readily isolated from various sources as culture expanded and unexpanded cellular population which are used as therapeutic products. Though MSCs possess a unique immunological and regulatory profile through cross-talk between MSCs and immunoregulatory cells (T cells, NK cells, dendritic cells, B cells, neutrophils, monocytes, and macrophages), they provide an immunotolerant environment when transplanted to the site of action. Immunophenotypic profile allows MSCs to escape immune surveillance and promotes their hypoimmunogenic or immune-privileged status. MSCs do not elicit a proliferative response when co-cultured with allogeneic T cells in vitro. MSCs secrete a wide range of anti-inflammatory mediators such as PGE-2, IDO, IL-1Ra, and IL-10. They also stimulate the resilient chondrogenic progenitors and enhance the chondrocyte differentiation by secretion of BMPs and TGFβ1. We highlight the various mechanisms of MSCs during tissue healing signals, their interaction with the immune system, and the impact of their lifespan in the management of osteoarthritis of the knee. A better understanding of the immunobiology of MSC renders them as an efficient therapeutic product for the management of osteoarthritis of the knee.
Collapse
|
2
|
Nakasone H, Kikuchi M, Kawamura K, Akahoshi Y, Sato M, Kawamura S, Yoshino N, Takeshita J, Yoshimura K, Misaki Y, Gomyo A, Tanihara A, Kusuda M, Tamaki M, Kimura SI, Kako S, Kanda Y. Increased CD83 expression of CD34-positive monocytes in donors during peripheral blood stem cell mobilization in humans. Sci Rep 2019; 9:16499. [PMID: 31712609 PMCID: PMC6848192 DOI: 10.1038/s41598-019-53020-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/26/2019] [Indexed: 12/02/2022] Open
Abstract
CD34-positive monocytes (CD34+mono) have recently been identified in grafts mobilized by granulocyte-colony stimulating factor. We analyzed transplant outcomes of 73 patients whose donor's peripheral blood cells were cryopreserved during mobilization. CD34+mono was detected more frequently in male donors (67% vs. 40%, P = 0.03), while the detection of CD34+mono in donors was not associated with the patient background. Although there was no significant difference in overall survival in the whole cohort, the detection of CD34+mono in donors were significantly associated with a decreased risk of non-relapse mortality (HR 0.23, P = 0.035). Fatal infectious events tended to be less frequent in donors with CD34+mono. Gene expression profile analyses of CD34+mono in humans revealed that the expressions of pro-inflammatory cytokines like IL6, CCL3, IL8, VEGFA, and IL1A were elevated in CD34+mono, and those cytokines were enriched in the immune response, especially against infectious pathogens in the gene ontology analyses. In addition, the expression of CD83 was specifically increased in CD34+mono. It might play a role of antigen presentation in the immune network, leading in a clinical benefit against infections. Further investigations will be required to confirm the biological functions and clinical roles of CD34+mono in transplantation.
Collapse
Affiliation(s)
- Hideki Nakasone
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Misato Kikuchi
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Koji Kawamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yu Akahoshi
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Miki Sato
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shunto Kawamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Nozomu Yoshino
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Junko Takeshita
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Kazuki Yoshimura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yukiko Misaki
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Ayumi Gomyo
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Aki Tanihara
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Machiko Kusuda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Masaharu Tamaki
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shun-Ichi Kimura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shinichi Kako
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan.
| |
Collapse
|
3
|
Large-scale production and directed induction of functional dendritic cells ex vivo from serum-free expanded human hematopoietic stem cells. Cytotherapy 2019; 21:755-768. [PMID: 31105040 DOI: 10.1016/j.jcyt.2019.04.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/01/2019] [Accepted: 04/28/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dendritic cells (DCs) that are derived from hematopoietic stem cells (HSCs) are the most potent antigen-presenting cells and play a pivotal role in initiating the immune response. Hence, large-scale production and direct induction of functional DCs ex vivo from HSCs are crucial to HSC research and clinical potential, such as vaccines for cancer and immune therapy. METHODS In a previous study, we developed a serum-free HSC expansion system (SF-HSC medium) to expand large numbers of primitive HSCs ex vivo. Herein, a DC induction and expansion medium (DC medium) was proposed to further generate large numbers of functional DCs from serum-free expanded HSCs, which were developed and optimized by factorial design and the steepest ascent method. RESULTS The DC medium is composed of effective basal medium (Iscove's modified Dulbecco's medium [IMDM]) and cytokines (2.9 ng/mL stem cell factor [SCF], 2.1 ng/mL Flt-3 ligand, 3.6 ng/mL interleukin [IL]-1β, 19.3 ng/mL granulocyte-macrophage colony-stimulating factor [GM-CSF] and 20.0 ng/mL tumor necrosis factor-α [TNF-α]). After 10-day culture in DC medium, the maximum fold expansion for accumulated CD1a+CD11c+ DCs was more than 4000-fold, and the induced DCs were characterized and confirmed by analysis of growth kinetics, surface antigen expression, endocytosis ability, mixed lymphocyte reaction, specific cytokine secretion and lipopolysaccharide stimulation. DISCUSSION In conclusion, the combination of DC medium and SF-HSC medium can efficiently induce and expand a large amount of functional DCs from a small scale of HSCs and might be a promising source of DCs for vaccine and immune therapy in the near future.
Collapse
|
4
|
Cornel AM, van Til NP, Boelens JJ, Nierkens S. Strategies to Genetically Modulate Dendritic Cells to Potentiate Anti-Tumor Responses in Hematologic Malignancies. Front Immunol 2018; 9:982. [PMID: 29867960 PMCID: PMC5968097 DOI: 10.3389/fimmu.2018.00982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) vaccination has been investigated as a potential strategy to target hematologic malignancies, while generating sustained immunological responses to control potential future relapse. Nonetheless, few clinical trials have shown robust long-term efficacy. It has been suggested that a combination of surmountable shortcomings, such as selection of utilized DC subsets, DC loading and maturation strategies, as well as tumor-induced immunosuppression may be targeted to maximize anti-tumor responses of DC vaccines. Generation of DC from CD34+ hematopoietic stem and progenitor cells (HSPCs) may provide potential in patients undergoing allogeneic HSPC transplantations for hematologic malignancies. CD34+ HSPC from the graft can be genetically modified to optimize antigen presentation and to provide sufficient T cell stimulatory signals. We here describe beneficial (gene)-modifications that can be implemented in various processes in T cell activation by DC, among which major histocompatibility complex (MHC) class I and MHC class II presentation, DC maturation and migration, cross-presentation, co-stimulation, and immunosuppression to improve anti-tumor responses.
Collapse
Affiliation(s)
- Annelisa M Cornel
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niek P van Til
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, Netherlands.,Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
5
|
Tremble LF, Forde PF, Soden DM. Clinical evaluation of macrophages in cancer: role in treatment, modulation and challenges. Cancer Immunol Immunother 2017; 66:1509-1527. [PMID: 28948324 PMCID: PMC11028704 DOI: 10.1007/s00262-017-2065-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022]
Abstract
The focus of immunotherapeutics has been placed firmly on anti-tumour T cell responses. Significant progress has been made in the treatment of both local and systemic malignancies, but low response rates and rising toxicities are limiting this approach. Advancements in the understanding of tumour immunology are opening up a new range of therapeutic targets, including immunosuppressive factors in the tumour microenvironment. Macrophages are a heterogeneous group of cells that have roles in innate and adaptive immunity and tissue repair, but become co-opted by tumours to support tumour growth, survival, metastasis and immunosuppression. Macrophages also support tumour resistance to conventional therapy. In preclinical models, interference with macrophage migration, macrophage depletion and macrophage re-education have all been shown to reduce tumour growth and support anti-tumour immune responses. Here we discuss the role of macrophages in prognosis and sensitivity to therapy, while examining the significant progress which has been made in modulating the behaviour of these cells in cancer patients.
Collapse
Affiliation(s)
- Liam Friel Tremble
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland.
| | - Patrick F Forde
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| | - Declan M Soden
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| |
Collapse
|
6
|
García-Mendoza MG, Inman DR, Ponik SM, Jeffery JJ, Sheerar DS, Van Doorn RR, Keely PJ. Neutrophils drive accelerated tumor progression in the collagen-dense mammary tumor microenvironment. Breast Cancer Res 2016; 18:49. [PMID: 27169366 PMCID: PMC4864897 DOI: 10.1186/s13058-016-0703-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/12/2016] [Indexed: 12/27/2022] Open
Abstract
Background High mammographic density has been correlated with a 4-fold to 6-fold increased risk of developing breast cancer, and is associated with increased stromal deposition of extracellular matrix proteins, including collagen I. The molecular and cellular mechanisms responsible for high breast tissue density are not completely understood. Methods We previously described accelerated tumor formation and metastases in a transgenic mouse model of collagen-dense mammary tumors (type I collagen-α1 (Col1α1)tm1Jae and mouse mammary tumor virus - polyoma virus middle T antigen (MMTV-PyVT)) compared to wild-type mice. Using ELISA cytokine arrays and multi-color flow cytometry analysis, we studied cytokine signals and the non-malignant, immune cells in the collagen-dense tumor microenvironment that may promote accelerated tumor progression and metastasis. Results Collagen-dense tumors did not show any alteration in immune cell populations at late stages. The cytokine signals in the mammary tumor microenvironment were clearly different between wild-type and collagen-dense tumors. Cytokines associated with neutrophil signaling, such as granulocyte monocyte-colony stimulated factor (GM-CSF), were increased in collagen-dense tumors. Depleting neutrophils with anti-Ly6G (1A8) significantly reduced the number of tumors, and blocked metastasis in over 80 % of mice with collagen-dense tumors, but did not impact tumor growth or metastasis in wild-type mice. Conclusion Our study suggests that tumor progression in a collagen-dense microenvironment is mechanistically different, with pro-tumor neutrophils, compared to a non-dense microenvironment. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0703-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- María G García-Mendoza
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, Madison, WI, USA.,UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA.,Present Address: Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David R Inman
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, Madison, WI, USA.,UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, Madison, WI, USA.,UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Justin J Jeffery
- UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Dagna S Sheerar
- UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Rachel R Van Doorn
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, Madison, WI, USA
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, Madison, WI, USA. .,UW Carbone Cancer Center, University of Wisconsin - Madison, Madison, WI, USA. .,Wisconsin Institutes of Medical Research, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
7
|
Bie Y, Xu Q, Zhang Z. Isolation of dendritic cells from umbilical cord blood using magnetic activated cell sorting or adherence. Oncol Lett 2015; 10:67-70. [PMID: 26170978 DOI: 10.3892/ol.2015.3198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 03/17/2015] [Indexed: 02/04/2023] Open
Abstract
Dendritic cells (DCs) are a highly specialized type of antigen-presenting cell. The present study describes and compares two methods for preparing DCs from umbilical cord blood. The first method involves the isolation of DCs by magnetic activated cell sorting (MACS). This technique isolates CD34+ cells from cord blood and induces the formation of DCs by the addition of cytokines, granulocyte macrophage colony-stimulating factor and interleukin-4. The second method involves the generation of large numbers of DCs from cord blood using an adherent method, which isolates umbilical cord blood mononuclear cells and induces DCs in the same conditions as those used in MACS. The DCs were harvested following 7 days of incubation and observed with an inverted microscope. The phenotype of the cells was then analyzed by flow cytometry. The results revealed that, subsequent to 7 days of incubation, the differentiated DCs obtained using the adherent method were more mature than those isolated using MACS. However, these cells were unable to be maintained in culture for more than 9-10 days. By contrast, the DCs derived from CD34+ cells by MACS were phenotypically stable and could be maintained for up to 3 weeks in culture. Either method produced DCs from cord blood. However, the DCs isolated using the MACS method demonstrated higher homogeneity, yield and viability than those obtained using the adherent method. Due to the various compositions of the monocyte subsets isolated, isolation methods affect the phenotypes and functions of the resultant DCs.
Collapse
Affiliation(s)
- Yachun Bie
- Department of Obstetrics and Gynecology, Beijng Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Qiuxiang Xu
- Department of Obstetrics and Gynecology, Beijng Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijng Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
8
|
Abstract
The immunotherapy for malignant glioma faces unique difficult, due to some anatomical and immunological characteristics including the existence of blood brain barrier, the absence of lymphatic tissues and dendritic cells (DCs) in the central nervous system (CNS) parenchyma, and the presence of an immunosuppressive microenvironment. Therefore, immunotherapeutic approaches will not be beneficial unless the compromised immune status in malignant glioma patients is overcome. DC-based immunotherapy, vaccinating cancer patients with DCs pulsed with various tumor antigens, is one of the most promising immunotherapeutic approaches for treatment of malignant glioma because it seems able to overcome, at least partially, the immunosuppressive state associated with primary malignancies. The preparation of DCs, choice of antigen, and route and schedule of administration are improving and optimizing with rapid development of molecular biology and gene engineering technology. DC vaccination in humans, after a number of pre-clinical models and clinical trials, would increase the clinical benefits for malignant glioma immunotherapy.
Collapse
Affiliation(s)
- Jin-Hai Gu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | | |
Collapse
|
9
|
Tahoori MT, Pourfathollah AA, Soleimani M, Vasheghani-Farahani E, Mohammadzadeh A, Amari A, Hashemi SM, Mossahebi-Mohammadi M. Fibroblasts feeder niche and Flt3 Ligand as a novel inducer of plasmacytoid dendritic cells development in vitro. Int Immunopharmacol 2014; 24:474-480. [PMID: 25445955 DOI: 10.1016/j.intimp.2014.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
Plasmacytoid dendritic cell (pDC), plays central role in antiviral immunity. The aim of this study was to assess the effect of Flt3 ligand (FL) alone or with L929 fibroblast feeder or L929 conditioned media on differentiation of mouse bone marrow (BM) cells into pDC in vitro. Murine BM cells were cultured with FL or with L929 or conditioned media for 9days. The differentiated cells were analyzed using flow cytometry for PDCA-1, B220 and CXCR4. The relative expression of Stat3, CXCR4, CXCR7, IFN-β, TGF-β and Runx2 in differentiated cells determined by real time PCR. The development of pDC showed up to 19% increase after co-culture of BM cells with fibroblast feeder. Upregulation of Stat3, Runx2 and CXCR4 due to the presence of fibroblast feeder with FL in culture results in improved pDC development. Furthermore, 30% L929 supernatant along with Flt3 ligand was able to derive pDC up to 8.9% in comparison with FL alone, which was 6.6% in vitro. Thus, for the first time we introduced L929 fibroblast feeder as a niche producer of M-CSF and probably other growth factors and chemokines, which promotes the development of pDC in vitro along with FL, similar to in vivo niche.
Collapse
Affiliation(s)
- Mohammad Taher Tahoori
- Department of Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| | - Masoud Soleimani
- Department of Stem Cell Biology, Stem Cell Technology Research Center, Tehran Iran; Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | | | - Adel Mohammadzadeh
- Department of Microbiology, Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Afshin Amari
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
10
|
Pham VQ, Nguyen ST, Van Pham P. Production of functional dendritic cells from mouse bone marrow. BIOMEDICAL RESEARCH AND THERAPY 2014. [DOI: 10.7603/s40730-014-0020-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
11
|
Harrison SJ, Cook G, Nibbs RJB, Prince HM. Immunotherapy of multiple myeloma: the start of a long and tortuous journey. Expert Rev Anticancer Ther 2014; 6:1769-85. [PMID: 17181491 DOI: 10.1586/14737140.6.12.1769] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The field of tumor immunotherapy is still in its infancy. It is becoming clear that the human immune response is the result of highly complex, continuously evolving interactions between cells of the adaptive and innate arms of the immune system, the internal and external environments, and normal and abnormal cells (e.g., myeloma plasma cells). Despite the considerable advances in our knowledge over the past 30 years, we have still only scratched the surface of the immune system's interaction with malignant diseases such as myeloma and to date, this has not translated into significantly better outcomes for patients with this disease. This review will summarize our current knowledge of the fundamental immunology of myeloma, review immunotherapy trials reported to date and discuss whether, in light of the current information, immunotherapy of multiple myeloma is an achievable goal.
Collapse
Affiliation(s)
- Simon J Harrison
- DHMO, Peter MacCallum Cancer Centre and Locked Bag 1, A'becket Street, Melbourne, Victoria 8006, Australia.
| | | | | | | |
Collapse
|
12
|
Klammer M, Roddie PH. Current progress in the development of a cell-based vaccine for the immunotherapy of acute myeloid leukemia. Expert Rev Vaccines 2014; 5:211-22. [PMID: 16608421 DOI: 10.1586/14760584.5.2.211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Evidence that immunological control contributes to the elimination of residual leukemia has emerged from allogeneic hematopoietic stem cell transplantation. This review assesses the current understanding of immunobiology of acute myeloid leukemia and how dendritic cells and T cells may be harnessed using in vitro and in vivo priming techniques. Preclinical and clinical dendritic cell vaccine trials reported to date are considered and the prospects for immunotherapy with dendritic cell-based vaccine constructs evaluated.
Collapse
Affiliation(s)
- Matthias Klammer
- Western General Hospital, University of Edinburgh-Leukaemia Research Fund, John Hughes Bennett Laboratory and Department of Haematology, Western General Hospital, Edinburgh, UK.
| | | |
Collapse
|
13
|
Abstract
The role of granulocyte-macrophage-colony-stimulating factor (GM-CSF) in the supportive care of cancer patients has been evaluated with promising results. More recently, GM-CSF has been added to regimens for the mobilization of hematopoietic progenitor cells. An expanding role for GM-CSF in regulating immune responses has been recognized based upon its activity on the development and maturation of antigen presenting cells and its capability for skewing the immune system toward Th1-type responses. GM-CSF has been shown to preferentially enhance both the numbers and activity of type 1 dendritic cells (DC1), the subsets of dendritic cells responsible for initiating cytotoxic immune responses. The increase in DC1 content and activity following local and systemic GM-CSF administration support a role for GM-CSF as an immune stimulant and vaccine adjuvant in cancer patients. GM-CSF has shown clinical activity as an immune stimulant in tumor cell and dendritic cell vaccines, and may increase antibody-dependent cellular cytotoxicity. The successful use of myeloid acting cytokines to enhance anti-tumor responses will likely require the utilization of GM-CSF in combination with cytotoxic or other targeted therapies.
Collapse
Affiliation(s)
- Martha Arellano
- Emory University, Winship Cancer Institute, Atlanta, GA, USA
| | | |
Collapse
|
14
|
Phuc PV, Lam DH, Ngoc VB, Thu DT, Nguyet NTM, Ngoc PK. Production of functional dendritic cells from menstrual blood--a new dendritic cell source for immune therapy. In Vitro Cell Dev Biol Anim 2011; 47:368-75. [PMID: 21424240 DOI: 10.1007/s11626-011-9399-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 02/22/2011] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are the most professional antigen-presenting cells of the mammalian immune system. They are able to phagocytize, process antigen materials, and then present them to the surface of other cells including T lymphocytes in the immune system. These capabilities make DC therapy become a novel and promising immune-therapeutic approach for cancer treatment as well as for cancer vaccination. Many trials of DC therapy to treat cancers have been performed and have shown their application value. They involve harvesting monocytes or hematopoietic stem cells from a patient and processing them in the laboratory to produce DCs and then reintroduced into a patient in order to activate the immune system. DCs were successfully produced from peripheral, umbilical cord blood-derived monocytes or hematopoietic stem cells. In this research, we produced DCs from human menstrual blood-derived monocytes. Briefly, monocytes were isolated by FACS based on FSC vs. SSC plot from lysed menstrual blood. Obtained monocytes were induced into DCs by a two-step protocol. In the first step, monocytes were incubated in RPMI medium supplemented with 2% FBS, GM-CSF, and IL-4, followed by incubation in RPMI medium supplemented with α-TNF in the second step. Our data showed that induced monocytes had typical morphology of DCs, expressed HLA-DR, HLA-ABC, CD80 and CD86 markers, exhibited uptake of dextran-FITC, stimulated allogenic T cell proliferation, and released IL-12. These results demonstrated that menstrual blood can not only be a source of stromal stem cell but also DCs, which are a potential candidate for immune therapy.
Collapse
Affiliation(s)
- Pham Van Phuc
- Laboratory of Stem cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh, Vietnam.
| | | | | | | | | | | |
Collapse
|
15
|
Ex vivo development, expansion and in vivo analysis of a novel lineage of dendritic cells from hematopoietic stem cells. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2010; 8:8. [PMID: 21106069 PMCID: PMC3004889 DOI: 10.1186/1476-8518-8-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 11/24/2010] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play a key role in innate and adaptive immunity but the access to sufficient amount of DCs for basic and translational research has been limited. We established a novel ex vivo system to develop and expand DCs from hematopoietic stem/progenitor cells (HPCs). Both human and mouse HPCs were expanded first in feeder culture supplemented with c-Kit ligand (KL, stem cell factor, steel factor or CD117 ligand), Flt3 ligand (fms-like tyrosine kinase 3, Flt3L, FL), thrombopoietin (TPO), IL-3, IL-6, and basic fibroblast growth factor (bFGF), and then in a second feeder culture ectopically expressing all above growth factors plus GM-CSF and IL-15. In the dual culture system, CD34+ HPCs differentiated toward DC progenitors (DCPs), which expanded more than five orders of magnitude. The DCPs showed myeloid DC surface phenotype with up-regulation of transcription factors PU.1 and Id2, and DC-related factors homeostatic chemokine ligand 17 (CCL17) and beta-chemokine receptor 6 (CCR6). Multiplex ELISA array and cDNA microarray analyses revealed that the DCPs shared some features of IL-4 and IL-15 DCs but displayed a pronounced proinflammatory phenotype. DCP-derived DCs showed antigen-uptake and immune activation functions analogous to that of the peripheral blood-derived DCs. Furthermore, bone marrow HPC-derived DCP vaccines of tumor-bearing mice suppressed tumor growth in vivo. This novel approach of generating DCP-DCs, which are different from known IL-4 and IL-15 DCs, overcomes both quantitative and qualitative limitations in obtaining functional autologous DCs from a small number of HPCs with great translational potential.
Collapse
|
16
|
Abstract
IMPORTANCE OF THE FIELD Pulmonary alveolar proteinosis (PAP) is a rare disease in which the abnormalities in surfactant metabolism are caused most often by impairments of GM-CSF pathway at different levels in different disease subsets (congenital, secondary, acquired/idiopathic) and for which there are only few, costly invasive therapeutic methods. AREAS COVERED IN THIS REVIEW This review discusses these impairments, and their pathogenic and clinical consequences along with potential corrective therapies such as exogenous inhaled GM-CSF. WHAT THE READER WILL GAIN Among the PAP disease subsets, in autoimmune PAP the GM-CSF autoantibodies play a major role in disease pathogenesis and their deleterious pulmonary effects can be blocked efficaciously with inhaled GM-CSF. TAKE HOME MESSAGE In PAP correction of the abnormalities of the GM-CSF pathway represent a plausible approach demonstrated to be efficacious also in the case of inhaled GM-CSF used for autoimmune PAP.
Collapse
Affiliation(s)
- Sabina A Antoniu
- University of Medicine and Pharmacy Gr T Popa Iasi, Pulmonary Disease University Hospital, Department of Internal Medicine II- Pulmonary Disease, 30 Dr I Cihac Str, 700115 Iasi, Romania.
| |
Collapse
|
17
|
Nie YJ, Mok MY, Chan GCF, Chan AW, Jin OU, Kavikondala S, Lie AKW, Lau CS. Phenotypic and functional abnormalities of bone marrow-derived dendritic cells in systemic lupus erythematosus. Arthritis Res Ther 2010; 12:R91. [PMID: 20478074 PMCID: PMC2911875 DOI: 10.1186/ar3018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 08/30/2009] [Accepted: 05/18/2010] [Indexed: 12/29/2022] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoreactive T and B cells, which are believed to be secondary to deficient dendritic cells (DCs). However, whether DC abnormalities occur during their development in the bone marrow (BM) or in the periphery is not known. Methods Thirteen patients with SLE and 16 normal controls were recruited. We studied the morphology, phenotype, and functional abilities of bone marrow-derived dendritic cells (BMDCs) generated by using two culture methods: FMS-like tyrosine kinase 3 (Flt3)-ligand (FL) and granulocyte-macrophage colony-stimulating factor (GM-CSF) plus interleukin-4 (IL-4), respectively. Results BMDCs induced by FL exhibited both myeloid (mDC) and plasmacytoid DC (pDC) features, whereas GM-CSF/IL-4 induced mDC generation. Substantial phenotypic and functional defects of BMDCs were found from patients with SLE at different stages of cell maturation. When compared with healthy controls, SLE immature BM FLDCs expressed higher levels of CCR7. Both immature and mature SLE BM FLDCs expressed higher levels of CD40 and CD86 and induced stronger T-cell proliferation. SLE BM mDCs expressed higher levels of CD40 and CD86 but lower levels of HLA-DR and a lower ability to stimulate T-cell proliferation when compared with control BM mDCs. Conclusions Our data are in accordance with previous reports that suggest that DCs have a potential pathogenic role in SLE. Defects of these cells are evident during their development in BM. BM mDCs are deficient, whereas BM pDCs, which are part of BM FLDCs, are the likely culprit in inducing autoimmunity in SLE.
Collapse
Affiliation(s)
- Ying J Nie
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Everts B, Adegnika AA, Kruize YCM, Smits HH, Kremsner PG, Yazdanbakhsh M. Functional impairment of human myeloid dendritic cells during Schistosoma haematobium infection. PLoS Negl Trop Dis 2010; 4:e667. [PMID: 20422029 PMCID: PMC2857749 DOI: 10.1371/journal.pntd.0000667] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 03/11/2010] [Indexed: 01/27/2023] Open
Abstract
Chronic Schistosoma infection is often characterized by a state of T cell hyporesponsiveness of the host. Suppression of dendritic cell (DC) function could be one of the mechanisms underlying this phenomenon, since Schistosoma antigens are potent modulators of dendritic cell function in vitro. Yet, it remains to be established whether DC function is modulated during chronic human Schistosoma infection in vivo. To address this question, the effect of Schistosoma haematobium infection on the function of human blood DC was evaluated. We found that plasmacytoid (pDC) and myeloid DC (mDC) from infected subjects were present at lower frequencies in peripheral blood and that mDC displayed lower expression levels of HLA-DR compared to those from uninfected individuals. Furthermore, mDC from infected subjects, but not pDC, were found to have a reduced capacity to respond to TLR ligands, as determined by MAPK signaling, cytokine production and expression of maturation markers. Moreover, the T cell activating capacity of TLR-matured mDC from infected subjects was lower, likely as a result of reduced HLA-DR expression. Collectively these data show that S. haematobium infection is associated with functional impairment of human DC function in vivo and provide new insights into the underlying mechanisms of T cell hyporesponsiveness during chronic schistosomiasis.
Collapse
Affiliation(s)
- Bart Everts
- Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
19
|
Shao L, Li T, Mo X, Majdic O, Zhang Y, Seyerl M, Schrauf C, Ma D, Stöckl J, Han W. Expressional and functional studies of CKLF1 during dendritic cell maturation. Cell Immunol 2010; 263:188-95. [DOI: 10.1016/j.cellimm.2010.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 03/09/2010] [Accepted: 03/18/2010] [Indexed: 10/19/2022]
|
20
|
Balan S, Kale VP, Limaye LS. A simple two-step culture system for the large-scale generation of mature and functional dendritic cells from umbilical cord blood CD34+ cells. Transfusion 2009; 49:2109-21. [DOI: 10.1111/j.1537-2995.2009.02231.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
21
|
Kalinski P, Urban J, Narang R, Berk E, Wieckowski E, Muthuswamy R. Dendritic cell-based therapeutic cancer vaccines: what we have and what we need. Future Oncol 2009; 5:379-90. [PMID: 19374544 DOI: 10.2217/fon.09.6] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Therapeutic cancer vaccines rely on the immune system to eliminate tumor cells. In contrast to chemotherapy or passive (adoptive) immunotherapies with antibodies or ex vivo-expanded T cells, therapeutic vaccines do not have a direct anti-tumor activity, but aim to reset patients' immune systems to achieve this goal. Recent identification of effective ways of enhancing immunogenicity of tumor-associated antigens, including the use of dendritic cells and other potent vectors of cancer vaccines, provide effective tools to induce high numbers of circulating tumor-specific T cells. However, despite indications that some of the new cancer vaccines may be able to delay tumor recurrence or prolong the survival of cancer patients, their ability to induce cancer regression remains low. Recent reports help to identify and prospectively remove the remaining obstacles towards effective therapeutic vaccination of cancer patients. They indicate that the successful induction of tumor-specific T cells by cancer vaccines is not necessarily associated with the induction of functional cytotoxic T lymphocytes, and that current cancer vaccines may promote undesirable expansion of Treg cells. Furthermore, recent studies also identify the tools to counteract such phenomena, in order to assure the desirable induction of Th1-cytotoxic T lymphocytes, NK-mediated type-1 immunity and appropriate homing of effector cells to tumors.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Surgery, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Res. Pavilion, Suite 1.46, 5117 Center Avenue, PA 15213-1863, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Martino S, di Girolamo I, Tiribuzi R, D'Angelo F, Datti A, Orlacchio A. Efficient siRNA delivery by the cationic liposome DOTAP in human hematopoietic stem cells differentiating into dendritic cells. J Biomed Biotechnol 2009; 2009:410260. [PMID: 19503805 PMCID: PMC2688684 DOI: 10.1155/2009/410260] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 03/20/2009] [Indexed: 01/12/2023] Open
Abstract
RNA interference technology is an ideal strategy to elucidate the mechanisms associated with human CD34(+) hematopoietic stem cell differentiation into dendritic cells. Simple manipulations in vitro can unequivocally yield alloreactive or tolerogenic populations, suggesting key implications of biochemical players that might emerge as therapeutic targets for cancer or graft-versus-host disease. To knockdown proteins typically involved in the biology of dendritic cells, we employed an siRNA delivery system based on the cationic liposome DOTAP as the carrier. Freshly-isolated CD34(+) cells were transfected with siRNA for cathepsin S with negligible cytotoxicity and transfection rates (>60%) comparable to the efficiency shown by lentiviral vectors. Further, cathepsin S knockdown was performed during both cell commitment and through the entire 14-day differentiation process with repeated transfection rounds that had no effect per se on cell development. Tested in parallel, other commercially-available chemical reagents failed to meet acceptable standards. In addition to safe and practical handling, a direct advantage of DOTAP over viral-mediated techniques is that transient silencing effects can be dynamically appraised through the recovery of targeted proteins. Thus, our findings identify DOTAP as an excellent reagent for gene silencing in resting and differentiating CD34(+) cells, suggesting a potential for applications in related preclinical models.
Collapse
Affiliation(s)
- Sabata Martino
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| | - Ilaria di Girolamo
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| | - Roberto Tiribuzi
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| | - Francesco D'Angelo
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| | - Alessandro Datti
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| | - Aldo Orlacchio
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Sezione di Biochimica e Biologia Molecolare, Università di Perugia, Via del Giochetto, 06100 Perugia, Italy
| |
Collapse
|
23
|
Abstract
BACKGROUND Cancer vaccines are a unique approach to cancer therapy. They exert an antitumor effect by engaging the host immune response, and have great potential for circumventing the intrinsic drug resistance that limits standard cancer management. Additional advantages of cancer vaccines are exquisite specificity, low toxicity, and the potential for a durable treatment effect due to immunologic memory. OBJECTIVES This review aims to consider the promise of cancer vaccines, review the current state of cancer vaccine development, and suggest directions for future research. METHODS The scope of this review was defined peer-reviewed information found on Medline, and information found on the Internet about Phase III clinical trials that are ongoing and not yet published. RESULTS/CONCLUSIONS Multiple Phase III clinical trials have demonstrated the promise and challenges posed by therapeutic vaccines, and defined the next steps in their clinical development. Determining the optimal integration of cancer vaccines with chemotherapy, radiation, surgery, and biologically targeted therapies, defining predictive biomarkers of immunologic and clinical response, and combining tumor vaccines with new drugs that effectively modulate the antitumor immune response, will ensure that cancer vaccines become part of standard cancer therapy and prevention.
Collapse
Affiliation(s)
- Leisha A Emens
- Johns Hopkins University, Tumor Immunology and Breast Cancer Research Programs, Department of Oncology, 1650 Orleans Street, Room 409, Bunting Blaustein Cancer Research Building, Baltimore, MD 21231-1000, USA.
| |
Collapse
|
24
|
Abstract
Dendritic cells (DCs) play a crucial role in the induction of antigen-specific T-cell responses, and therefore their use for the active immunotherapy of malignancies has been studied with considerable interest. More than a decade has passed since the publication of the first clinical data of DC-based vaccines, and through this and subsequent studies, a number of important developmental insights have been gleaned. These include the ideal source and type of DCs, the discovery of novel antigens and methods of loading DCs, the role of DC maturation, and the most efficient route of immunization. The generation of immune responses against tumor antigens after DC immunization has been demonstrated, and favorable clinical responses have been reported in some patients; however, it is difficult to pool the results as a whole, and thus the body of data remains inconclusive, in part because of varying DC preparation and vaccination protocols, the use of different forms of antigens, and, most importantly, a lack of rigorous criteria for defining clinical responses. As such, the standardization of clinical and immunologic criteria utilized, as well as DC preparations employed, will allow for the comparison of results across multiple clinical studies and is required in order for future trials to measure the true value and role of this treatment modality. In addition, issues regarding the optimal dose and clinical setting for the application of DC vaccines remain to be resolved, and recent clinical studies have been designed to begin to address these questions.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Program in Molecular Therapeutics, Comprehensive Cancer Center, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
25
|
Morandi B, Costa R, Falco M, Parolini S, De Maria A, Ratto G, Mingari MC, Melioli G, Moretta A, Ferlazzo G. Distinctive Lack of CD48 Expression in Subsets of Human Dendritic Cells Tunes NK Cell Activation. THE JOURNAL OF IMMUNOLOGY 2005; 175:3690-7. [PMID: 16148114 DOI: 10.4049/jimmunol.175.6.3690] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD48 is a glycosyl phosphatidylinositol anchor protein known to be virtually expressed by all human leukocytes. Its ligand, 2B4, is a signaling lymphocyte activation molecule-related receptor involved in NK cell activation. Because dendritic cells (DCs) are strong inducers of NK cell functions, we analyzed the expression of CD48 in different human DC subsets. We observed that monocytes differentiating in DCs promptly down-regulate CD48. Similarly, DCs isolated from inflamed lymph nodes generally do not express CD48. Plasmocytoid DCs do not express CD48 either, whereas myeloid DCs harbored in blood, bone marrow, and thymus express it. In addition, we showed that CD48 expression in DCs affects NK cell functions during NK/DC cross-talk, because NK cells obtained from normal donors and from X-linked lymphoproliferative disease patients are, respectively, triggered or inhibited by DCs expressing surface CD48. Remarkably, IFN-gamma production by lymph node NK cells, in contrast to blood NK cells, can be negatively modulated by 2B4/CD48 interactions, indicating a 2B4 inhibitory pathway in lymph node NK cells. Therefore, the CD48 deficiency of DCs harbored in inflamed lymph nodes that we report in this study might be relevant to successfully activate lymph node NK cells in the early phase of the immune response. Our results show that distinct subsets of human DCs, differently from all other mononuclear hemopoietic cells, specifically do not express CD48. Moreover, the expression of CD48 depends on the anatomic location of DCs and might be related to the tissue-specific 2B4 function (activating or inhibitory) of the NK cells with which they interact.
Collapse
|
26
|
Abstract
In a small number of patients with multiple myeloma (MM), long-term disease-free survival has been achieved by harnessing the immune phenomenon, 'graft-versus-tumour' effect, induced by allogeneic haemopoietic stem cell transplantation. This has prompted many investigators to examine ways in which a patient's own immune system can be more effectively directed against their disease, with the ultimate aim of tumour eradication. In this review we assess the current understanding of immunobiology in MM, and how the different components of the immune system, such as dendritic cells, T cells and natural killer cells, may be harnessed using in-vitro and in-vivo priming techniques. We look at the clinical immunotherapy trials reported to date and whether, in light of the current information, immunotherapy for MM is an achievable goal.
Collapse
Affiliation(s)
- S J Harrison
- ATMU and Cancer Division, Section of Experimental Haematology, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
27
|
Syme R, Bajwa R, Robertson L, Stewart D, Glück S. Comparison of CD34 and monocyte-derived dendritic cells from mobilized peripheral blood from cancer patients. Stem Cells 2005; 23:74-81. [PMID: 15625124 DOI: 10.1634/stemcells.2004-0070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that are integral to the initiation of T-cell immunity. Two cell types can be used as a source for generating DCs: monocytes and CD34(+) stem cells. Despite many investigations characterizing DCs, none have performed a direct paired comparison of monocyte and stem cell-derived DCs. Therefore, it is unclear whether one cell source has particular advantages over the other, or whether inherent differences exist between the two populations. We undertook the following study to determine if there were any differences in DCs generated from monocytes or CD34(+) cells from mobilized peripheral blood. DCs were generated by culturing the adherent cells (monocytes) in interleukin-4 and GM-CSF for 7 days, or by culturing nonadherent cells (CD34(+)) in the presence of GM-CSF and tumor necrosis factor alpha for 14 days. The resulting DCs were compared morphologically, phenotypically, functionally, and by yield. We could generate morphologically and phenotypically similar DCs. Differences were encountered when expression levels of some cell surface markers were examined (CD86, HLA-DR). There was no difference in how the DCs performed in a mixed lymphocyte reaction (p = .3). Further, no statistical difference was discovered when we examined cellular (DC) yield (p = .1); however, there was a significant difference when yield was normalized to the starting number of monocytes or CD34(+) cells (p = .016). Together, these data demonstrate that differences do exist between monocyte-derived DCs and CD34-derived DCs from the same cellular product (apheresis) from the same individual.
Collapse
Affiliation(s)
- Rachel Syme
- Clinical Research Program, Department of Oncology, Tom Baker Cancer Centre, 1331 29th Street NW, Calgary, AB, T2N 4N2, Canada.
| | | | | | | | | |
Collapse
|
28
|
da Costa CET, Annels NE, Faaij CMJM, Forsyth RG, Hogendoorn PCW, Egeler RM. Presence of osteoclast-like multinucleated giant cells in the bone and nonostotic lesions of Langerhans cell histiocytosis. J Exp Med 2005; 201:687-93. [PMID: 15753204 PMCID: PMC2212837 DOI: 10.1084/jem.20041785] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 11/15/2004] [Indexed: 11/23/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is a disease that can involve one or multiple organ systems characterized by an accumulation of CD1a(+) Langerhans-like cells as well as several other myeloid cell types. The precise origin and role of one of these populations, the multinucleated giant cell (MGC), in this disease remains unknown. This work shows that in three different lesional tissues, bone, skin, and lymph node, the MGCs expressed the characteristic osteoclast markers, tartrate-resistant acid phosphatase and vitronectin receptor, as well as the enzymes cathepsin K and matrix metalloproteinase-9. Although, in bone lesions, the osteoclast-like MGCs were only CD68(+), in the nonostotic sites, they coexpressed CD1a. The presence of osteoclast-like MGCs may be explained by the production of osteoclast-inducing cytokines such as receptor activator of nuclear factor kappaB ligand and macrophage colony-stimulating factor by both the CD1a(+) LCH cells and T cells in these lesions. As osteoclast-derived enzymes play a major role in tissue destruction, the osteoclast-like nature of MGCs in all LCH lesions makes them a potential target for the treatment of this disease.
Collapse
Affiliation(s)
- Cristiana E T da Costa
- Department of Pediatric Immunology, Leiden University Medical Centre, 2300 RC Leiden, Netherlands
| | | | | | | | | | | |
Collapse
|
29
|
Della Bella S, Nicola S, Timofeeva I, Villa ML, Santoro A, Berardi AC. Are interleukin-16 and thrombopoietin new tools for the in vitro generation of dendritic cells? Blood 2004; 104:4020-8. [PMID: 15304384 DOI: 10.1182/blood-2004-03-0885] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The effects of interleukin 16 (IL-16) on dendritic cell (DC) generation from human CD34+ progenitor cells are not known. Here, we show that IL-16 added to a basal cocktail comprised of granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-4, Flt-3 ligand (Flt3L), and tumor necrosis factor α (TNF-α) does induce the CD34+ hematopoietic cells to proliferate in vitro and to differentiate into phenotypically and functionally mature DCs. IL-16 exerts this function more efficiently than stem cell factor (SCF) as a control, thrombopoietin (TPO), or IL-16 plus TPO. Moreover, we show that the combination of IL-16 plus TPO induces the generation of tolerogenic DCs, able to induce an anergic state in T cells that persists when T cells are rechallenged with immunogenic DCs. An altered pattern of cytokine production, a reduced expression of the C-type lectin DC-SIGN, and an increased surface expression of the inhibitory molecules immunoglobulin-like transcript 2 (ILT-2), ILT-3, and ILT-4 may all contribute to confer the tolerogenic properties of these DCs. Generation of tolerogenic DCs may aid the exploration of new therapeutic strategies to promote tolerance to autoantigens and prevent disease development. (Blood. 2004;104:4020-4028)
Collapse
Affiliation(s)
- Silvia Della Bella
- Dipartimento di Scienze e Tecnologie Biomediche, Laboratorio di Immunologia, Università di Milano, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Markiewicz MA, Kast WM. Progress in the Development of Immunotherapy of Cancer Using Ex Vivo-Generated Dendritic Cells Expressing Multiple Tumor Antigen Epitopes. Cancer Invest 2004; 22:417-34. [PMID: 15493363 DOI: 10.1081/cnv-200029072] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy with tumor-associated antigen-pulsed, ex vivo-generated dendritic cells (DCs) is a promising approach for the treatment of cancer that has shown efficacy in animal models and is now being tested in the clinic. The majority of studies performed to date make use of a single tumor-associated epitope. However, because of the high rate of mutation in tumor cells allowing for loss of expression of a single antigen, it is likely that use of multiple antigenic epitopes will induce a broader, longer-lasting, and effective tumor-specific immune response. Multiple vehicles for loading DCs with multiple antigenic epitopes are under investigation to determine the most effective method for vaccination, with many of these methods showing promise. These loading methods, as well as other critical considerations for making DC vaccination as efficacious as possible, are discussed in this article.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Cell Fusion
- Cells, Cultured/immunology
- Cells, Cultured/transplantation
- Clinical Trials as Topic
- DNA, Neoplasm/administration & dosage
- DNA, Neoplasm/genetics
- DNA, Neoplasm/immunology
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Epitopes/immunology
- Gene Transfer Techniques
- Humans
- Hybrid Cells/transplantation
- Immunotherapy, Adoptive
- Neoplasm Proteins/immunology
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Peptide Fragments/immunology
- RNA, Neoplasm/administration & dosage
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Mary A Markiewicz
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089-2821, USA
| | | |
Collapse
|
31
|
Cranmer LD, Trevor KT, Hersh EM. Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother 2004; 53:275-306. [PMID: 14648069 PMCID: PMC11032969 DOI: 10.1007/s00262-003-0432-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2003] [Accepted: 07/30/2003] [Indexed: 02/06/2023]
Abstract
Dendritic cell (DC) immunotherapy has shown significant promise in animal studies as a potential treatment for cancer. Its application in the clinic depends on the results of human trials. Here, we review the published clinical trials of cancer immunotherapy using exogenously antigen-exposed DCs. We begin with a short review of general properties and considerations in the design of such vaccines. We then review trials by disease type. Despite great efforts on the part of individual investigative groups, most trials to date have not yielded data from which firm conclusions can be drawn. The reasons for this include nonstandard DC preparation and vaccination protocols, use of different antigen preparations, variable means of immune assessment, and nonrigorous criteria for defining clinical response. While extensive animal studies have been conducted using DCs, optimal parameters in humans remain to be established. Unanswered questions include optimal cell dose, use of mature versus immature DCs for vaccination, optimal antigen preparation, optimal route, and optimal means of assessing immune response. It is critical that these questions be answered, as DC therapy is labor- and resource-intensive. Cooperation is needed on the part of the many investigators in the field to address these issues. If such cooperation is not forthcoming, the critical studies that will be required to make DC therapy a clinically and commercially viable enterprise will not take place, and this therapy, so promising in preclinical studies, will not be able to compete with the many other new approaches to cancer therapy presently in development. Trials published in print through June 2003 are included. We exclude single case reports, except where relevant, and trials with so many variables as to prevent interpretation about DC therapy effects.
Collapse
Affiliation(s)
- Lee D. Cranmer
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Katrina T. Trevor
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Evan M. Hersh
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| |
Collapse
|
32
|
Moldenhauer A, Nociari M, Lam G, Salama A, Rafii S, Moore MAS. Tumor necrosis factor alpha-stimulated endothelium: an inducer of dendritic cell development from hematopoietic progenitors and myeloid leukemic cells. Stem Cells 2004; 22:144-57. [PMID: 14990854 DOI: 10.1634/stemcells.22-2-144] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Especially when exposed to inflammatory stimuli, endothelial cells (EC) have been shown to promote the maturation of monocytes into dendritic cells (DC) and the long-term proliferation of CD34+ cells by constitutive cytokine production and direct cellular contact. We therefore hypothesized that cytokine-stimulated EC would induce hematopoietic progenitor cells to develop into mature dendritic cells. To test this theory, human CD34+ cells derived from cord blood or leukapheresis products were cultured with a monolayer of either interleukin (IL)-1beta, IL-4, or tumor necrosis factor (TNF)-alpha-stimulated human umbilical cord EC. The cells in suspension were analyzed weekly over a period of 6 weeks. IL-1beta supported cell expansion, whereas IL-4 had no effect on cell expansion or DC differentiation. Only TNF-alpha-stimulated EC induced the development of mature, allostimulatory DC with a high expression of CD83, HLA-DR, CD1a, and costimulatory molecules like CD80 and CD86. Acute myeloid leukemia cells from the cell line Kasumi-1 also developed DC-like features when cocultured with TNF-alpha-stimulated EC. Direct contact between endothelial and progenitor cells increased the number of developing DC. Cell cycle analysis and apoptosis studies demonstrated a reduced G2M fraction, an increased S fraction, and a decrease in TNF-alpha-dependent apoptosis of DC developing in the presence of endothelial cells. As shown by electron and confocal microscopic studies, intimate interactions between EC and DC occurred, resulting in the internalization of the developing DC within the EC monolayer and a bidirectional exchange of proteins. We conclude that, via the action of TNF-alpha, inflamed human endothelium can induce CD34+ and leukemic cells to differentiate into dendritic cells.
Collapse
Affiliation(s)
- Anja Moldenhauer
- Institute for Transfusion Medicine, Charité, Universitätsmedizen Berlin, Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
33
|
Laderach D, Wesa A, Galy A. 4-1BB-ligand is regulated on human dendritic cells and induces the production of IL-12. Cell Immunol 2003; 226:37-44. [PMID: 14746806 DOI: 10.1016/j.cellimm.2003.11.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Co-stimulation via 4-1BB and its ligand 4-1BB ligand (4-1BB-L) plays an important role in cytotoxic and pro-inflammatory immune responses. 4-1BB-L is generally described on activated antigen-presenting cells but there is limited information on its expression and function in human dendritic cells (DC). We herein compared purified CD1a+CD14- DC issued from monocytes or from hematopoietic progenitor cells (HPC). These DC expressed 4-1BB-L mRNA transcripts with highest cell surface levels on HPC-derived DC cultured with IL-1. Pro-inflammatory activation, particularly CD40 ligand+IL-1, up-regulated 4-1BB-L on DC. We confirmed reverse signaling via 4-1BB-L as immobilized 4-1BB in conjunction with CD40-L, enhanced IL-12beta mRNA and the secretion of IL-12 p70 in various APC, including monocytes. Altogether, DC may differ in T cell co-stimulation properties due to variable and regulated levels of 4-1BB-L. Data illustrate reciprocal stimulations between T cells and APC via up-regulated receptor/ligands and production of key cytokines that consolidate cellular immune responses.
Collapse
Affiliation(s)
- Diego Laderach
- Department of Immunology and Microbiology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | | | | |
Collapse
|
34
|
Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells that initiate T cell-mediated immune responses. The development of methods to generate a large number of DC in vitro has facilitated their application to immunotherapy. Adding tumor antigens to DC in vitro and administering them to cancer patients is expected to induce effective immune responses to cancers, which are poorly immunogenic in most cases. There are several parameters that need to be optimized to improve the efficacy of DC-based immunotherapy. Because the immune system has developed in order to eliminate microbial pathogens and is thus well equipped with machinery for that purpose, reproducing events occurring during anti-infection immune responses for antitumor immunity may lead to the development of effective tumor immunotherapy.
Collapse
Affiliation(s)
- Norimitsu Kadowaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
35
|
Mosca PJ, Clay TM, Kim Lyerly H, Morse MA. Current status of dendritic cell immunotherapy of malignancies. Int Rev Immunol 2003; 22:255-81. [PMID: 12745642 DOI: 10.1080/08830180305223] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Because dendritic cells (DC) are central to the induction of antigen-specific T cell responses, their use for the active immunotherapy of malignancies has been of considerable interest. Since clinical trials with DC-based vaccines have been initiated, a number of important developmental issues have become apparent. These include the ideal source and type of DC, the form of antigen and method of loading DC, whether to induce maturation, the route and timing of immunization, and the optimal clinical scenario. Clinical responses such as stability of disease and tumor regressions have been reported in some patients, particularly with melanoma, myeloma, and prostate cancer.
Collapse
Affiliation(s)
- Paul J Mosca
- Departments of Surgery and Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | |
Collapse
|
36
|
Banchereau J, Paczesny S, Blanco P, Bennett L, Pascual V, Fay J, Palucka AK. Dendritic cells: controllers of the immune system and a new promise for immunotherapy. Ann N Y Acad Sci 2003; 987:180-7. [PMID: 12727638 DOI: 10.1111/j.1749-6632.2003.tb06047.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The immune system is controlled by dendritic cells (DCs). Just as lymphocytes comprise different subsets, DCs comprise several subsets that differentially control lymphocyte function. In humans, the myeloid pathway includes Langerhans cells (LCs) and interstitial DCs (intDCs). While both subsets produce IL-12, only intDCs make IL-10 and induce B cell differentiation. Another pathway includes plasmacytoid DCs, which promptly secrete large amounts of IFN-alpha/beta viral encounter. Thus, insights into in vivo DC functions are important to understand the launching and modulation of immunity.
Collapse
|
37
|
Prósper F, Solá C, Hornedo J, Arbona C, Menéndez P, Orfao A, Lluch A, Cortés-Funes H, López JJ, García-Conde J. Mobilization of peripheral blood progenitor cells with a combination of cyclophosphamide, r-metHuSCF and filgrastim in patients with breast cancer previously treated with chemotherapy. Leukemia 2003; 17:437-41. [PMID: 12592344 DOI: 10.1038/sj.leu.2402750] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2002] [Accepted: 07/22/2002] [Indexed: 11/09/2022]
Abstract
The objective of our study was to determine the effect of adding r-metHuSCF to Filgrastim and cyclophosphamide for mobilization of peripheral blood progenitor cells (PBPC), on collection of CD34(+) cells and engraftment after autologous stem cell transplant. Twenty-three patients with previously treated stage II-IV breast cancer received cyclophosphamide (3 g/m(2)), Filgrastim 5 microg/kg daily and r-metHuSCF 20 microg/kg daily. Two PBPC collections were performed on consecutive days starting the day the WBC count was above 7.5 x 10(3)/microl. Collection was performed between days +9 and +12 and the median number of CD34(+) cells collected was 9.9 x 10(6)/kg (1.1-53.1) and 6.6 x 10(6)/kg (1.4-33.8) for the first and second apheresis, respectively. Despite being previously treated patients, the target CD34(+) cell dose required for SCT was obtained in all patients. SCT was associated with rapid neutrophil and platelet engraftment and a highly significant correlation was observed between the number of CD34(+) cells infused and engraftment. Treatment with SCF plus filgrastim was well tolerated, with mild to moderate local skin rash being the most frequently reported adverse event. In conclusion, addition of r-metHuSCF induces mobilization of a large number of CD34(+) cells which results in shortening of time to engraftment and hospitalization.
Collapse
Affiliation(s)
- F Prósper
- Servicio de Hematología y Oncología, Hospital Clínico, Universidad de Valencia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim EM, Sivanandham M, Stavropoulos CI, Wallack MK. Adjuvant effect of a Flt3 ligand (FL) gene-transduced xenogeneic cell line in a murine colon cancer model. J Surg Res 2002; 108:148-56. [PMID: 12443727 DOI: 10.1006/jsre.2002.6540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Flt3 Ligand (FL) has been shown to elicit antitumor responses induced by tumor antigen stimulation. Allogeneic and xenogeneic cell lines transduced with cytokine genes may be used to augment the antitumor efficacy of tumor antigens. OBJECTIVES The objective was to evaluate the augmentation of tumor lysate-induced immunity by a more clinically applicable FL gene-transduced xenogeneic cell line in combination with interleukin-2 (IL-2) in a CC-36 murine colon cancer model. METHODS Human 143B osteosarcoma tumor cells were transduced with full-length FL cDNA (143B-FL). Secretion of FL from 143B-FL was analyzed in vivo in normal BALB/c mice transplanted with 143B-FL, and expansion of dendritic cells (DC) was also analyzed in the same mice by flow cytometry. Eight-week-old, male BALB/c mice were used in a prophylactic vaccination protocol utilizing tumor lysate (CLy), 143B-FL, and soluble IL-2. Prophylactic group designations (n = 10/group) were as follows: ten million 143B-FL cells (alone, with tumor lysate, or with tumor lysate and IL-2), IL-2 with tumor lysate, IL-2 alone, or a no treatment control. The tumor lysate (200 microg of protein) and IL-2 (100,000 IU) injections were administered intraperitoneally. Mice were challenged subcutaneously with 10(3) CC-36 tumor cells. Tumor protection and tumor burden (TB), as mean tumor diameter, were determined. Peripheral blood lymphocytes (PBLs) from the 143B-FL + IL-2 + tumor lysate vaccinated group were analyzed for cytolytic activity in 4-h chromium release assays. In addition, plasma cytokine concentrations of interleukin-12 (IL-12) and interferon gamma (IFN-gamma) were assessed by ELISA. Student's t tests were used for all statistical comparisons. RESULTS In vivo expression of FL was observed 24 h following the inoculation of 143B-FL, and a four fold increase in DCs was observed in the peripheral blood of these mice. Mice immunized with a combination of 143B-FL, tumor lysate and IL-2 showed statistically significant protection against tumor development (10%) for 100 days after tumor challenge; incidences in other groups ranged from 40 to 100% (P < 0.05). Moreover, this immunization protocol produced the lowest TB at 3- and 6-week time points (0, 1.6 mm) when compared to all other groups (TB between 7.2 and 15.9 mm) (P < 0.05). In addition, PBLs from vaccinated mice showed increased cytolytic activity against CC-36 target cells. This corresponded to increased levels of IL-12 and IFN-g in the plasma of mice following vaccination. CONCLUSIONS These data suggest that FL gene-transduced xenogeneic tumor cells may augment the immunity induced by tumor antigens and systemic IL-2 through the activation of dendritic cells and T-cell-mediated mechanisms.
Collapse
Affiliation(s)
- Eugene M Kim
- Department of Surgery, Saint Vincents Hospital & Medical Center of New York, New York Medical College, 10011, USA
| | | | | | | |
Collapse
|
39
|
Wesa A, Galy A. Increased production of pro-inflammatory cytokines and enhanced T cell responses after activation of human dendritic cells with IL-1 and CD40 ligand. BMC Immunol 2002; 3:14. [PMID: 12385649 PMCID: PMC134468 DOI: 10.1186/1471-2172-3-14] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2002] [Accepted: 10/18/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Various microbial, inflammatory and immune signals regulate the activation of dendritic cells (DC), determining their ability to interact with naïve T cells and to produce cytokines that direct T cell development. In particular, CD40L and IL-1 cooperatively activate DC to secrete high levels of IL-12. The immuno-stimulatory capacity of such DC is otherwise not well-defined prompting further characterization of the effects of IL-1 and family members on DC activation in comparison with other pro-inflammatory stimuli. RESULTS Human DC co-activated in vitro by CD40L and IL-1beta expressed numerous cytokine genes including IL-12beta, IL-23 p19, IL-1beta, IL-1alpha, IL-1Ra, IL-10, IL-6, IL-18 and IFN-gamma. These DC produced high levels of IL-12 protein and appeared capable of producing IFN-gamma. Potent CD4+ and CD8+ T cell-stimulatory properties were acquired by DC under conditions that also induced IL-12. Notably, these DC induced rapid differentiation of fluMP-specific CD8+ T cells. Molecules related to IL-1beta, like IL-1alpha, co-induced IL-12 secretion whereas IL-18 did not. Conversely, the inhibitor IL-1Ra, produced endogenously by DC curtailed IL-12 production in response to CD40L. CONCLUSIONS IL-1 and IL-1Ra play a biologically-relevant role in the positive and negative regulation of DC activation. In conjunction with CD40L, IL-1 sends a powerful activation signal to DC that could be distinguished from other modes of activation. This signal enables the production of pro-inflammatory cytokines by DC, and enhances the differentiation of naïve T cells into effectors of type-1 cellular immune responses.
Collapse
Affiliation(s)
- Amy Wesa
- Cancer Biology Program and Department of Immunology and Microbiology, Barbara Ann Karmanos Cancer Institute of Wayne State University, Detroit, USA
| | - Anne Galy
- Cancer Biology Program and Department of Immunology and Microbiology, Barbara Ann Karmanos Cancer Institute of Wayne State University, Detroit, USA
- INSERM U 362, Institut Gustave Roussy, Villejuif and Genethon, Evry, France
| |
Collapse
|
40
|
Laderach D, Movassagh M, Johnson A, Mittler RS, Galy A. 4-1BB co-stimulation enhances human CD8(+) T cell priming by augmenting the proliferation and survival of effector CD8(+) T cells. Int Immunol 2002; 14:1155-67. [PMID: 12356681 DOI: 10.1093/intimm/dxf080] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interactions between 4-1BB and its ligand, 4-1BBL, enhance CD8(+) T cell-mediated antiviral and antitumor immunity in vivo. However, mechanisms regulating the priming of CD8(+) T cell responses by 4-1BB remain unclear, particularly in humans. The 4-1BB receptor was undetectable on naive or resting human CD8(+) T cells and induced in vitro by TCR triggering. Naive cord blood cells were therefore primed in vitro against peptides or cellular antigens and then co-stimulated with 4-1BBL or agonistic antibodies. Co-stimulation enhanced effector function such as IFN-gamma production and cytotoxicity by augmenting numbers of antigen-specific and effector CD8(+) T cells. OKT3 responses also showed reduced cell death and revealed that the proliferation of CD8(+) T cells required two independently regulated events. One, the induction of IL-2 production, could be directly triggered by 4-1BB engagement on CD8(+) T cells in the absence of accessory cells. The other, expression of CD25, was induced with variable efficacy by accessory cells. Thus, suboptimal accessory cells and 4-1BB co-stimulation combined their effects to enhance IL-2 production and proliferation. Reduced apoptosis observed after co-stimulation in the presence of accessory cells correlated with increased levels of Bcl-X(L) in CD8(+) T cells, while Bcl-2 expression remained unchanged. Altogether, 4-1BB enhanced expansion, survival and effector functions of newly primed CD8(+) T cells, acting in part directly on these cells. As 4-1BB triggering could be protracted from the TCR signal, 4-1BB agonists may function through these mechanisms to enhance or rescue suboptimal immune responses.
Collapse
Affiliation(s)
- Diego Laderach
- Barbara Ann Karmanos Cancer Institute, Department of Immunology Microbiology, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
41
|
Muench MO, Suskind DL, Bárcena A. Isolation, growth and identification of colony-forming cells with erythroid, myeloid, dendritic cell and NK-cell potential from human fetal liver. Biol Proced Online 2002; 4:10-23. [PMID: 12734573 PMCID: PMC145552 DOI: 10.1251/bpo29] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2002] [Revised: 05/16/2002] [Accepted: 05/27/2002] [Indexed: 12/04/2022] Open
Abstract
The study of hematopoietic stem cells (HSCs) and the process by which they differentiate into committed progenitors has been hampered by the lack of in vitro clonal assays that can support erythroid, myeloid and lymphoid differentiation. We describe a method for the isolation from human fetal liver of highly purified candidate HSCs and progenitors based on the phenotypes CD38(-)CD34(++) and CD38(+)CD34(++), respectively. We also describe a method for the growth of colony-forming cells (CFCs) from these cell populations, under defined culture conditions, that supports the differentiation of erythroid, CD14/CD15(+) myeloid, CD1a(+) dendritic cell and CD56(+) NK cell lineages. Flow cytometric analyses of individual colonies demonstrate that CFCs with erythroid, myeloid and lymphoid potential are distributed among both the CD38(-) and CD38(+) populations of CD34(++) progenitors.
Collapse
Affiliation(s)
- Marcus O Muench
- Department of Laboratory Medicine, University of California at San Francisco. 3rd & Parnassus Ave., Room U-440; San Francisco, CA 94143-0793.
| | | | | |
Collapse
|
42
|
Schui DK, Singh L, Schneider B, Knau A, Hoelzer D, Weidmann E. Inhibiting effects on the induction of cytotoxic T lymphocytes by dendritic cells pulsed with lysates from acute myeloid leukemia blasts. Leuk Res 2002; 26:383-9. [PMID: 11839382 DOI: 10.1016/s0145-2126(01)00141-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dendritic cells (DCs) were established from 25 patients in complete remission of acute myeloid leukemia (AML). In patients during hematopoietic regeneration following chemotherapy the yield of DC was comparable to that of healthy donors. In patients, more than 2 months after chemotherapy, significantly less DC were generated. Comparison of the antigen-presenting capacity using tetanus toxoid of six AML patients and six healthy volunteers did not show significant differences. In six AML patients, lymphocytes stimulated with blast cell lysate pulsed DC were analyzed for cytotoxic activity against autologous blast cells. 8.4-35.6% of autologous blast cells were lysed by DC stimulated lymphocytes. In three of the six patients maximum lysis of target cells was achieved by unpulsed DC. Thus, it seems that in some patients blast cell lysates mediate inhibitory effects, which may explain to some extend immune escape mechanisms in AML.
Collapse
Affiliation(s)
- Daniela K Schui
- Department of Medicine III, Haematology/Oncology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Muench MO, Bárcena A. Broad distribution of colony-forming cells with erythroid, myeloid, dendritic cell, and NK cell potential among CD34(++) fetal liver cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4902-9. [PMID: 11673495 DOI: 10.4049/jimmunol.167.9.4902] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The generation of erythroid, myeloid, and lymphoid cells from human fetal liver progenitors was studied in colony-forming cell (CFC) assays. CD38(-) and CD38(+) progenitors that expressed high levels of CD34 were grown in serum-deprived medium supplemented with kit ligand, flk2/flt3 ligand, GM-CSF, c-mpl ligand, erythropoietin, and IL-15. The resulting colonies were individually analyzed by flow cytometry. CD56(+) NK cells were detected in 21.9 and 9.9% of colonies grown from CD38(-) and CD38(+) progenitors, respectively. NK cells were detected in mostly large CD14(+)/CD15(+) myeloid colonies that also, in some cases, contained red cells. NK cells were rarely detected in erythroid colonies, suggesting an early split between the erythroid and the NK cell lineages. CD1a(+) dendritic cells were also present in three-quarters of the colonies grown from CD38(-) and CD38(+) progenitors. Multilineage colonies containing erythrocytes, myeloid cells, and NK cells were present in 13.7 and 2.7% of colonies grown from CD38(-) and CD38(+) progenitors, respectively. High proliferative-potential CFCs that generated multilineage colonies were also detected among both populations of progenitors. The total number of high proliferative-potential CFCs with erythroid, myeloid, and NK cell potential was estimated to be 2-fold higher in the CD38(+) fraction compared with the CD38(-) fraction because of the higher frequency of CD38(+) cells among CD34(++) cells. The broad distribution of multipotent CFCs among CD38(-) and CD38(+) progenitors suggests that the segregation of the erythroid, myeloid, and lymphoid lineages may not always be an early event in hemopoiesis. Alternatively, some stem cells may be present among CD38(+) cells.
Collapse
Affiliation(s)
- M O Muench
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA.
| | | |
Collapse
|
44
|
|
45
|
Abstract
The cytokine IL-12, a product of dendritic cells (DC), plays a major role in cellular immunity, notably by inducing lymphocytes to produce IFN-gamma. Microbial products, T cell signals and cytokines induce the production of IL-12. Here, IL-1 beta is identified as a new IL-12-inducing agent, acting conjointly with CD40 ligand (CD40L) on human monocyte-derived DC in vitro. The effects of IL-1 beta were dose dependent, specifically blocked by neutralizing antibodies, and were observed both in immature and mature DC. Immature DC secreted more IL-12 than mature DC, but the effects of IL-1 beta were not due to a block of DC maturation as determined by analysis of DC surface markers. The mechanisms of action of IL-1 beta could be contrasted to that of other inducers of IL-12 such as IFN-gamma and lipopolysaccharide (LPS). Either IL-1 beta or IFN-gamma co-induced IL-12 with CD40L but conjointly, IL-1 beta, CD40L and IFN-gamma synergized, inducing very high levels of IL-12. The effects of IL-1 beta differed from those of LPS in that IL-1 beta, unlike LPS, could not induce IL-12 solely after IFN-gamma priming; and when combined with CD40L, IL-1 beta, unlike LPS, induced little IL-10. The mechanism of action of IL-1 beta involves IL-12 alpha mRNA up-regulation, and we show that the combination of CD40L and IL-1 beta induces high levels of IL-12 alpha and IL-12 beta mRNA in DC. Altogether, these results delineate a new mechanism linking adaptive and innate immune responses for the regulation of IL-12 production in DC and for the role of IL-1 beta in the development of cellular immunity.
Collapse
Affiliation(s)
- A K Wesa
- Barbara Ann Karmanos Cancer Institute of Wayne State University, Detroit, MI 48201, USA
| | | |
Collapse
|
46
|
Syme R, Glück S. Generation of dendritic cells: role of cytokines and potential clinical applications. Transfus Apher Sci 2001; 24:117-24. [PMID: 11501570 DOI: 10.1016/s1473-0502(01)00005-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the last decade there has been great interest in generating populations of antigen presenting cells (APC), which can be exploited to improve immune responses to a variety of diseases including malignancies. Dendritic cells (DC) are an APC population that are easily generated ex vivo using a variety of cytokines. Cytokines can also be used to further manipulate these cells in maturation and function. The following discussion will provide an overview of dendritic cell isolation and generation with a focus on the role cytokines play in this process.
Collapse
Affiliation(s)
- R Syme
- Department of Oncology, Faculty of Medicine, University of Calgary, Tom Baker Cancer Centre, AB, Canada
| | | |
Collapse
|
47
|
Abstract
The characterization of tumor-associated antigens recognized by human T lymphocytes in a major histocompatibility complex (MHC)-restricted fashion has opened new possibilities for immunotherapeutic approaches to the treatment of human cancers. Dendritic cells (DC) are professional antigen presenting cells that are well suited to activate T cells toward various antigens, such as tumor-associated antigens, due to their potent costimulatory activity. The availability of large numbers of DC, generated either from hematopoietic progenitor cells or monocytes in vitro or isolated from peripheral blood, has profoundly changed pre-clinical research as well as the clinical evaluation of these cells. Accordingly, appropriately pulsed or transfected DC may be used for vaccination in the field of infectious diseases or tumor immunotherapy to induce antigen-specific T cell responses. These observations led to pilot clinical trials of DC vaccination for patients with cancer in order to investigate the feasibility, safety, as well as the immunologic and clinical effects of this approach. Initial clinical studies of human DC vaccines are generating encouraging preliminary results demonstrating induction of tumor-specific immune responses and tumor regression. Nevertheless, much work is still needed to address several variables that are critical for optimizing this approach and to determine the role of DC-based vaccines in tumor immunotherapy.
Collapse
Affiliation(s)
- N Meidenbauer
- Department of Hematology/Oncology, University of Regensburg, Germany
| | | | | |
Collapse
|
48
|
Wesa AK, Galy A. Regulation of T cell cytokine production by dendritic cells generated in vitro from hematopoietic progenitor cells. Cell Immunol 2001; 208:115-24. [PMID: 11333144 DOI: 10.1006/cimm.2001.1773] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
When dendritic cells (DC) present antigens to T cells, reciprocal cellular interactions occur that lead to cytokine production. This cytokine response is regulated by specific properties of DC, notably their maturation/activation status and perhaps their origin. The latter possibility prompted us to determine if DC produced along distinct developmental pathways induced distinct T cell responses. Hematopoietic progenitor cells with the potential to differentiate into multiple lineages of cells were induced to differentiate into DC along two pathways. One leads to the formation of lymphoid-related DC but not of monocyte-derived DC and is induced by culture of CD34(+) cells with flt-3 ligand (F), c-kit ligand (K), GM-CSF (Gm), IL-1beta ("1"), and IL-7 ("7") (FKGm17). Another pathway with distinct molecular requirements supports in part monocyte-derived DC and is induced by the cytokines F, K, Gm, TNF-alpha (T), and IL-4 ("4") (FKGmT4). DC produced along these two pathways were isolated by flow cytometry and compared. They differed only slightly in phenotype and morphology and both induced Th1-type cytokine production in MLR (mixed lymphocyte reactions). However, on a cell-per-cell basis, FKGm17-DC produced more IL-18 or IL-12 and induced more IFN-gamma by T cells in MLR. Such superior properties were not intrinsically determined by the origin of the DC but were induced by FKGm17 cytokines. We conclude that lymphoid-related DC have the potential to induce Th1 T cell responses but that environmental signals strongly influence T-cell-stimulating properties of DC.
Collapse
Affiliation(s)
- A K Wesa
- Barbara Ann Karmanos Cancer Institute of Wayne State University, Detroit, Michigan, 48201, USA
| | | |
Collapse
|
49
|
Syme R, Glück S. Effects of cytokines on the culture and differentiation of dendritic cells in vitro. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:43-51. [PMID: 11276358 DOI: 10.1089/152581601750098219] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ability to culture dendritic cells (DC) in vitro has been integral to the dramatic increase in research in the area of immunotherapy. Over time, a number of methods for generating these cells have been developed. This article will provide an overview of the isolation and generation of DC and will give a detailed description of the role specific cytokines play in this process from the mobilization of precursors to the final maturation of DC.
Collapse
Affiliation(s)
- R Syme
- Departments of Oncology, Medicine and Pharmacology & Therapeutics, Faculty of Medicine, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta T2N 4N2, Canada.
| | | |
Collapse
|
50
|
|