1
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025. [PMID: 40249331 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
2
|
Zhi Y, Li T, Li Y, Zhang T, Du M, Zhang Q, Wang X, Hu G. Protective role of Cecropin AD against LPS-induced intestinal mucosal injury in chickens. Front Immunol 2023; 14:1290182. [PMID: 38162646 PMCID: PMC10757607 DOI: 10.3389/fimmu.2023.1290182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Cecropin AD (CAD), a renowned antimicrobial peptide, has shown promising potential in treating various bacterial infections. This study investigates the protective effects of CAD against lipopolysaccharide (LPS)-induced intestinal adversities in chickens. Methods Sixty SPF-grade chicks were divided into groups and exposed to different dosages of CAD, followed by LPS administration. The study assessed the impact of CAD on intestinal mucosal injury markers, oxidative stress, and inflammation. Results LPS significantly increased Diamine oxidase (DAO) and D-lactate (D-LA) levels, both indicators of intestinal mucosal injury. CAD treatment substantially attenuated these elevations, particularly at higher dosages. Additionally, CAD markedly reduced oxidative stress in intestinal tissues, as shown by normalized antioxidant levels and decreased reactive oxygen species. Histological analysis supported these findings, showing better-preserved villi structures in CAD-treated groups. Furthermore, CAD significantly reduced IL-6 and IL-8 expression post-LPS stimulation and effectively regulated the NLRP3 inflammasome pathway, decreasing associated factors like NLRP3, Caspase-1, IL-1b, and IL-18. Discussion The study demonstrates CAD's therapeutic potential in alleviating LPS-induced intestinal injuries. The protective effects are primarily attributed to its anti-inflammatory and antioxidative actions and modulation of the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Yan Zhi
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Tingyu Li
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yaxuan Li
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Tao Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Mengze Du
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Qian Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, the Key Laboratory of Otolaryngology-Head and Neck Surgery (Ministry of Education of China), Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Ge Hu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
3
|
Klinman DM, Goguet E, Tross D. TLR Agonist Therapy of Metastatic Breast Cancer in Mice. J Immunother 2023; 46:170-177. [PMID: 37103328 PMCID: PMC10168108 DOI: 10.1097/cji.0000000000000467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/20/2023] [Indexed: 04/28/2023]
Abstract
Toll-like receptor (TLR) 7/8 and 9 agonists stimulate an innate immune response that supports the development of tumor-specific immunity. Previous studies showed that either agonist individually could cure mice of small tumors and that when used in combination, they could prevent the progression of larger tumors (>300 mm 3 ). To examine whether these agents combined could control metastatic disease, syngeneic mice were challenged with the highly aggressive 66cl4 triple-negative breast tumor cell line. Treatment was not initiated until pulmonary metastases were established, as verified by bioluminescent imaging of luciferase-tagged tumor cells. Results show that combined therapy with TLR7/8 and TLR9 agonists delivered to both primary and metastatic tumor sites significantly reduced tumor burden and extended survival. The inclusion of cyclophosphamide and anti-PD-L1 resulted in optimal tumor control, characterized by a 5-fold increase in the average duration of survival.
Collapse
|
4
|
Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. Int J Mol Sci 2019; 20:ijms20236008. [PMID: 31795299 PMCID: PMC6929211 DOI: 10.3390/ijms20236008] [Citation(s) in RCA: 1072] [Impact Index Per Article: 178.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022] Open
Abstract
This review aims to briefly discuss a short list of a broad variety of inflammatory cytokines. Numerous studies have implicated that inflammatory cytokines exert important effects with regard to various inflammatory diseases, yet the reports on their specific roles are not always consistent. They can be used as biomarkers to indicate or monitor disease or its progress, and also may serve as clinically applicable parameters for therapies. Yet, their precise role is not always clearly defined. Thus, in this review, we focus on the existing literature dealing with the biology of cytokines interleukin (IL)-6, IL-1, IL-33, tumor necrosis factor-alpha (TNF-α), IL-10, and IL-8. We will briefly focus on the correlations and role of these inflammatory mediators in the genesis of inflammatory impacts (e.g., shock, trauma, immune dysregulation, osteoporosis, and/or critical illness).
Collapse
Affiliation(s)
- Shinwan Kany
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany;
- Department of Cardiology with Emphasis on Electrophysiology, University Heart Centre, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60590 Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6721395
| |
Collapse
|
5
|
Kany S, Janicova A, Relja B. Innate Immunity and Alcohol. J Clin Med 2019; 8:jcm8111981. [PMID: 31739600 PMCID: PMC6912266 DOI: 10.3390/jcm8111981] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
The innate immunity has evolved during millions of years, and thus, equivalent or comparable components are found in most vertebrates, invertebrates, and even plants. It constitutes the first line of defense against molecules, which are either pathogen-derived or a danger signal themselves, and not seldom both. These molecular patterns are comprised of highly conserved structures, a common trait in innate immunity, and constitute very potent triggers for inflammation mediated via extracellular or intracellular pattern recognition receptors. Human culture is often interweaved with the consumption of alcohol, in both drinking habits, its acute or chronical misuse. Apart from behavioral effects as often observed in intoxicated individuals, alcohol consumption also leads to immunological modulation on the humoral and cellular levels. In the last 20 years, major advances in this field of research have been made in clinical studies, as well as in vitro and in vivo research. As every physician will experience intoxicated patients, it is important to be aware of the changes that this cohort undergoes. This review will provide a summary of the current knowledge on the influence of alcohol consumption on certain factors of innate immunity after a hit, followed by the current studies that display the effect of alcohol with a description of the model, the mode of alcohol administration, as well as its dose. This will provide a way for the reader to evaluate the findings presented.
Collapse
|
6
|
Kim H, Griffith TS, Panyam J. Poly(d,l-lactide-co-glycolide) Nanoparticles as Delivery Platforms for TLR7/8 Agonist-Based Cancer Vaccine. J Pharmacol Exp Ther 2019; 370:715-724. [PMID: 30610006 DOI: 10.1124/jpet.118.254953] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/03/2019] [Indexed: 12/28/2022] Open
Abstract
Targeted drug delivery can significantly influence the efficacy of a drug. In the past decades, diverse drug-delivery technologies, including nano- and microparticles, co-crystals, and microneedles have been developed to maximize therapeutic efficacy and minimize undesired side effects of therapeutics. Nanoparticles-submicron-sized drug carriers-have been actively investigated for the delivery of antibiotics, nucleic acids, peptide/proteins, and chemotherapeutics. Recently, nanoparticles have gained attention as a vaccine delivery platform for tumor-associated antigens (TAAs) and/or vaccine adjuvants. Agonists of imidazoquinoline-based Toll-like receptor (TLR) 7/8 are potent cytokine inducers that are used as cancer vaccine adjuvants to elicit robust T-cell response by activating dendritic cells (DCs). Despite their in vitro potency, the translation of TLR7 agonists as cancer vaccine adjuvants in the clinic has been limited by their poor retention at the injection site. Therefore, a formulation that could improve the availability of TLR7/8 agonists to DCs via conventional vaccine administration routes (subcutaneous, intramuscular) can broaden the application of TLR7/8 agonists for cancer immunotherapy. Polymeric nanoparticles fabricated with poly(d,l-lactide-co-glycolide) (PLGA) can be an efficient TLR7/8 agonist delivery platform. PLGA is a biocompatible polymer, and nanoparticles prepared from this polymer are stable in saline and are small enough to be administered by subcutaneous or intramuscular injections. Furthermore, nanoparticulate TLR7/8 delivery can enhance DC uptake and facilitate lymphatic drainage, both of which can enhance the adjuvanticity of TLR7/8 agonists compared with soluble forms. In this review, we discuss the use of PLGA nanoparticles with TLR7/8 agonists for improving cancer immunotherapy.
Collapse
Affiliation(s)
- Hyunjoon Kim
- Departments of Pharmaceutics (H.K., J.P.) and Urology (T.S.G.), Center for Immunology (T.S.G.), Microbiology, Immunology, and Cancer Biology Graduate Program (T.S.G.), and Masonic Cancer Center (T.S.G., J.P.), University of Minnesota, Minneapolis, Minnesota
| | - Thomas S Griffith
- Departments of Pharmaceutics (H.K., J.P.) and Urology (T.S.G.), Center for Immunology (T.S.G.), Microbiology, Immunology, and Cancer Biology Graduate Program (T.S.G.), and Masonic Cancer Center (T.S.G., J.P.), University of Minnesota, Minneapolis, Minnesota
| | - Jayanth Panyam
- Departments of Pharmaceutics (H.K., J.P.) and Urology (T.S.G.), Center for Immunology (T.S.G.), Microbiology, Immunology, and Cancer Biology Graduate Program (T.S.G.), and Masonic Cancer Center (T.S.G., J.P.), University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
7
|
In vitro activation and maturation of human mononuclear phagocytes by stimulation with liposomes coated with a neoglycolipid containing α1–3, α1–6-mannotriose. Glycoconj J 2019; 36:185-197. [DOI: 10.1007/s10719-019-09870-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/28/2019] [Accepted: 04/04/2019] [Indexed: 01/22/2023]
|
8
|
Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials 2018; 164:38-53. [DOI: 10.1016/j.biomaterials.2018.02.034] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/05/2018] [Accepted: 02/17/2018] [Indexed: 12/21/2022]
|
9
|
Mego M, Gao H, Cohen EN, Anfossi S, Giordano A, Tin S, Fouad TM, De Giorgi U, Giuliano M, Woodward WA, Alvarez RH, Valero V, Ueno NT, Hortobagyi GN, Cristofanilli M, Reuben JM. Circulating tumor cells (CTCs) are associated with abnormalities in peripheral blood dendritic cells in patients with inflammatory breast cancer. Oncotarget 2018; 8:35656-35668. [PMID: 27374101 PMCID: PMC5482606 DOI: 10.18632/oncotarget.10290] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022] Open
Abstract
CTCs are involved in tumor dissemination and are an independent prognostic factor in primary and metastatic breast cancer patients. Dendritic cells (DCs) are the most efficient antigen presenting cells and are comprised of plasmacytoid-(pDC) and myeloid-(mDC) derived DC subsets. This study aimed to correlate CTC counts with the peripheral blood DC immunophenotypes and functions of inflammatory breast cancer (IBC) patients. This study included 65 IBC patients. Peripheral blood (PB) was obtained from patients prior to starting a new line of chemotherapy for CTCs enumeration by CellSearch® and DC phenotype and function by flow cytometry; the characteristics of DCs were then correlated with CTC counts and clinical outcome. Twenty-one (32.3%) patients with CTCs ≥5 had a significantly inferior overall survival (OS) compared to patients with <5 CTCs (p=0.045). In addition, patients with ≥5 CTCs had a lower percentage of mDCs capable of producing TNF-α before or after activation through the toll-like receptor (TLR), as well as a lower percentage of mDCs producing IL-12 after TLR-activation. There was a positive correlation between CTCs counts and expression of the activation (CCR7) and costimulatory (CD86) receptors on TLR-activated mDCs and pDCs, respectively. Moreover, presence of high percentage of mDC capable to produce increased levels of TNF-α was independently associated with inferior OS (p = 0.0006). An increase in the percentage of mDC producing TNF-α might induce a pro-inflammatory environment that could play a role in determining the poor clinical outcome in IBC patients and could add further prognostic value to CTCs.
Collapse
Affiliation(s)
- Michal Mego
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Department of Medical Oncology, Comenius University, School of Medicine, National Cancer Institute, Bratislava, Slovakia
| | - Hui Gao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Evan N Cohen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simone Anfossi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antonio Giordano
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Department of Medicine at Medical University of South Carolina, Charleston, SC, USA
| | - Sanda Tin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tamer M Fouad
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Department of Medical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ugo De Giorgi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) - IRCCS, Meldola (FC), Italy
| | - Mario Giuliano
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Wendy A Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ricardo H Alvarez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Massimo Cristofanilli
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Currently at Division of Hematology-Oncology at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of MD Anderson Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Patchett AL, Tovar C, Corcoran LM, Lyons AB, Woods GM. The toll-like receptor ligands Hiltonol ® (polyICLC) and imiquimod effectively activate antigen-specific immune responses in Tasmanian devils (Sarcophilus harrisii). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 76:352-360. [PMID: 28689773 DOI: 10.1016/j.dci.2017.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 06/07/2023]
Abstract
Devil facial tumour disease (DFTD) describes two genetically distinct transmissible tumours that pose a significant threat to the survival of the Tasmanian devil. A prophylactic vaccine could protect devils from DFTD transmission. For this vaccine to be effective, potent immune adjuvants will be required. Toll-like receptors (TLRs) promote robust immune responses in human cancer studies and are highly conserved across mammalian species. In this study, we investigated the proficiency of TLR ligands for immune activation in the Tasmanian devil using in vitro mononuclear cell stimulations and in vivo immunisation trials with a model antigen. We identified two such TLR ligands, polyICLC (Hiltonol®) (TLR3) and imiquimod (TLR7), that in combination induced significant IFNγ production from Tasmanian devil lymphocytes in vitro. Immunisation with these ligands and the model antigen keyhole limpet haemocyanin activated robust antigen-specific primary, secondary and long-term memory IgG responses. Our results support the conserved nature of TLR signaling across mammalian species. PolyICLC and imiquimod will be trialed as immune adjuvants in future DFTD vaccine formulations.
Collapse
Affiliation(s)
- Amanda L Patchett
- Menzies Institute for Medical Research, University of Tasmania, Hobart 7000, Tasmania, Australia.
| | - Cesar Tovar
- Menzies Institute for Medical Research, University of Tasmania, Hobart 7000, Tasmania, Australia
| | - Lynn M Corcoran
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3052, Victoria, Australia
| | - A Bruce Lyons
- School of Medicine, University of Tasmania, Hobart 7000, Tasmania, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart 7000, Tasmania, Australia; School of Medicine, University of Tasmania, Hobart 7000, Tasmania, Australia
| |
Collapse
|
11
|
Lövgren T, Sarhan D, Truxová I, Choudhary B, Maas R, Melief J, Nyström M, Edbäck U, Vermeij R, Scurti G, Nishimura M, Masucci G, Karlsson-Parra A, Lundqvist A, Adamson L, Kiessling R. Enhanced stimulation of human tumor-specific T cells by dendritic cells matured in the presence of interferon-γ and multiple toll-like receptor agonists. Cancer Immunol Immunother 2017; 66:1333-1344. [PMID: 28601925 PMCID: PMC5626805 DOI: 10.1007/s00262-017-2029-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/05/2017] [Indexed: 12/23/2022]
Abstract
Dendritic cell (DC) vaccines have been demonstrated to elicit immunological responses in numerous cancer immunotherapy trials. However, long-lasting clinical effects are infrequent. We therefore sought to establish a protocol to generate DC with greater immunostimulatory capacity. Immature DC were generated from healthy donor monocytes by culturing in the presence of IL-4 and GM-CSF and were further differentiated into mature DC by the addition of cocktails containing different cytokines and toll-like receptor (TLR) agonists. Overall, addition of IFNγ and the TLR7/8 agonist R848 during maturation was essential for the production of high levels of IL-12p70 which was further augmented by adding the TLR3 agonist poly I:C. In addition, the DC matured with IFNγ, R848, and poly I:C also induced upregulation of several other pro-inflammatory and Th1-skewing cytokines/chemokines, co-stimulatory receptors, and the chemokine receptor CCR7. For most cytokines and chemokines the production was even further potentiated by addition of the TLR4 agonist LPS. Concurrently, upregulation of the anti-inflammatory cytokine IL-10 was modest. Most importantly, DC matured with IFNγ, R848, and poly I:C had the ability to activate IFNγ production in allogeneic T cells and this was further enhanced by adding LPS to the cocktail. Furthermore, epitope-specific stimulation of TCR-transduced T cells by peptide- or whole tumor lysate-loaded DC was efficiently stimulated only by DC matured in the full maturation cocktail containing IFNγ and the three TLR ligands R848, poly I:C, and LPS. We suggest that this cocktail is used for future clinical trials of anti-cancer DC vaccines.
Collapse
Affiliation(s)
- Tanja Lövgren
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden. .,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden. .,Cancer Center Karolinska R8:01, Karolinska Universitetssjukhuset Solna, 171 76, Stockholm, Sweden.
| | - Dhifaf Sarhan
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Iva Truxová
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Bhavesh Choudhary
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Roeltje Maas
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Jeroen Melief
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Maria Nyström
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Edbäck
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Renee Vermeij
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Gina Scurti
- Department of Surgery, Loyola University Chicago, Maywood, IL, USA
| | | | - Giuseppe Masucci
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Alex Karlsson-Parra
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Lars Adamson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Proff J, Walterskirchen C, Brey C, Geyeregger R, Full F, Ensser A, Lehner M, Holter W. Cytomegalovirus-Infected Cells Resist T Cell Mediated Killing in an HLA-Recognition Independent Manner. Front Microbiol 2016; 7:844. [PMID: 27375569 PMCID: PMC4899442 DOI: 10.3389/fmicb.2016.00844] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023] Open
Abstract
In order to explore the potential of HLA-independent T cell therapy for human cytomegalovirus (HCMV) infections, we developed a chimeric antigen receptor (CAR) directed against the HCMV encoded glycoprotein B (gB), which is expressed at high levels on the surface of infected cells. T cells engineered with this anti-gB CAR recognized HCMV-infected cells and released cytokines and cytotoxic granules. Unexpectedly, and in contrast to analogous approaches for HIV, Hepatitis B or Hepatitis C virus, we found that HCMV-infected cells were resistant to killing by the CAR-modified T cells. In order to elucidate whether this phenomenon was restricted to the use of CARs, we extended our experiments to T cell receptor (TCR)-mediated recognition of infected cells. To this end we infected fibroblasts with HCMV-strains deficient in viral inhibitors of antigenic peptide presentation and targeted these HLA-class I expressing peptide-loaded infected cells with peptide-specific cytotoxic T cells (CTLs). Despite strong degranulation and cytokine production by the T cells, we again found significant inhibition of lysis of HCMV-infected cells. Impairment of cell lysis became detectable 1 day after HCMV infection and gradually increased during the following 3 days. We thus postulate that viral anti-apoptotic factors, known to inhibit suicide of infected host cells, have evolved additional functions to directly abrogate T cell cytotoxicity. In line with this hypothesis, CAR-T cell cytotoxicity was strongly inhibited in non-infected fibroblasts by expression of the HCMV-protein UL37x1, and even more so by additional expression of UL36. Our data extend the current knowledge on Betaherpesviral evasion from T cell immunity and show for the first time that, beyond impaired antigen presentation, infected cells are efficiently protected by direct blockade of cytotoxic effector functions through viral proteins.
Collapse
Affiliation(s)
- Julia Proff
- Children's Cancer Research Institute, St. Anna KinderkrebsforschungVienna, Austria; Children's University Hospital, Universitätsklinikum ErlangenErlangen, Germany
| | | | - Charlotte Brey
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung Vienna, Austria
| | - Rene Geyeregger
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung Vienna, Austria
| | - Florian Full
- Institute for Clinical and Molecular Virology, Universitätsklinikum ErlangenErlangen, Germany; Department of Microbiology, The University of ChicagoChicago, IL, USA
| | - Armin Ensser
- Institute for Clinical and Molecular Virology, Universitätsklinikum Erlangen Erlangen, Germany
| | - Manfred Lehner
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung Vienna, Austria
| | - Wolfgang Holter
- Children's Cancer Research Institute, St. Anna KinderkrebsforschungVienna, Austria; Department of Pediatrics, St. Anna Kinderspital, Medical University of ViennaVienna, Austria
| |
Collapse
|
13
|
Patchett AL, Latham R, Brettingham-Moore KH, Tovar C, Lyons AB, Woods GM. Toll-like receptor signaling is functional in immune cells of the endangered Tasmanian devil. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 53:123-133. [PMID: 26182986 DOI: 10.1016/j.dci.2015.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 06/04/2023]
Abstract
Devil facial tumour disease (DFTD) is a fatally transmissible cancer that threatens the Tasmanian devil population. As Tasmanian devils do not produce an immune response against DFTD cells, an effective vaccine will require a strong adjuvant. Activation of innate immune system cells through toll-like receptors (TLRs) could provide this stimulation. It is unknown whether marsupials, including Tasmanian devils, express functional TLRs. We isolated RNA from peripheral blood mononuclear cells and, with PCR, detected transcripts for TLRs 2, 3, 4, 5, 6, 7, 8, 9, 10 and 13. Stimulation of the mononuclear cells with agonists to these TLRs increased the expression of downstream TLR signaling products (IL1α, IL6, IL12A and IFNβ). Our data provide the first evidence that TLR signaling is functional in the mononuclear cells of the Tasmanian devil. Future DFTD vaccination trials will incorporate TLR agonists to enhance the immune response against DFTD.
Collapse
Affiliation(s)
- Amanda L Patchett
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Roger Latham
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | | | - Cesar Tovar
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - A Bruce Lyons
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia; School of Medicine, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
14
|
Huizinga R, van den Berg B, van Rijs W, Tio-Gillen AP, Fokkink WJR, Bakker-Jonges LE, Geleijns K, Samsom JN, van Doorn PA, Laman JD, Jacobs BC. Innate Immunity toCampylobacter jejuniin Guillain-Barré Syndrome. Ann Neurol 2015; 78:343-54. [DOI: 10.1002/ana.24442] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/18/2015] [Accepted: 05/25/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Ruth Huizinga
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Bianca van den Berg
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Wouter van Rijs
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Anne P. Tio-Gillen
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Willem Jan R. Fokkink
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | | | - Karin Geleijns
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Janneke N. Samsom
- Division Gastroenterology and Nutrition; Department of Pediatrics; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Pieter A. van Doorn
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Jon D. Laman
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Bart C. Jacobs
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
- Department of Neurology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| |
Collapse
|
15
|
Khan N, Vidyarthi A, Pahari S, Agrewala JN. Distinct Strategies Employed by Dendritic Cells and Macrophages in Restricting Mycobacterium tuberculosis Infection: Different Philosophies but Same Desire. Int Rev Immunol 2015; 35:386-398. [PMID: 25793750 DOI: 10.3109/08830185.2015.1015718] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dendritic cells (DCs) and macrophages (Mϕs) are professional antigen-presenting cells (APCs) that can efficiently phagocytose Mycobacterium tuberculosis (Mtb), the etiological agent of tuberculosis (TB). It is quite interesting to mention here that DCs and Mϕs use distinct strategies to combat and eliminate Mtb. Similarly, Mtb employs different mechanisms to counteract the action of DCs and Mϕs. Mϕs are evolved with specialized, innate, defensive machinery to restrict growth of Mtb at the initial phase of infection. However, DCs are more endowed toward initiating adaptive immunity by activating naïve T cells. During encounter with Mtb, DCs and Mϕs deliver discrete functions via triggering through different pattern recognition receptors (PRRs) expressed by these APCs. Mtb-infected DCs and Mϕs show differential expression of genes encoding cytokines, chemokines, costimulatory molecules, and adhesion molecules. Interestingly, Mtb impairs the immune defensive machinery by exploiting various PRRs. Remarkably, selective signaling through PRRs by Mtb abrogates the bactericidal activity of Mϕs, but subverts differentiation of monocytes to DCs. In this article, we highlight the role of PRRs in inducing distinct immune response by DCs and Mϕs against Mtb. Concurrently, we also discuss smart strategies exploited by Mtb to impair the function of host DCs and Mϕs.
Collapse
Affiliation(s)
- Nargis Khan
- a CSIR-Institute of Microbial Technology , Chandigarh , India
| | | | - Susanta Pahari
- a CSIR-Institute of Microbial Technology , Chandigarh , India
| | | |
Collapse
|
16
|
Zhao BG, Vasilakos JP, Tross D, Smirnov D, Klinman DM. Combination therapy targeting toll like receptors 7, 8 and 9 eliminates large established tumors. J Immunother Cancer 2014; 2:12. [PMID: 24982761 PMCID: PMC4075973 DOI: 10.1186/2051-1426-2-12] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 04/01/2014] [Indexed: 12/14/2022] Open
Abstract
Background The TLR7/8 agonist 3M-052 and the TLR9 agonist CpG ODN both trigger innate immune responses that support the induction of tumor-specific immunity. Previous studies showed that these agonists used individually could improve the survival of mice challenged with small tumors but were of limited therapeutic benefit against large/advanced tumors. Methods Normal mice were challenged with syngeneic tumors. Once these tumors reached clinically detectable size (500–800 mm3) they were treated by intra-tumoral injection with 3M-052 and/or CpG ODN. Anti-tumor immunity and tumor growth were evaluated. Results The co-delivery of agonists targeting TLRs 7, 8 and 9 increased the number and tumoricidal activity of tumor infiltrating CTL and NK cells while reducing the frequency of immunosuppressive MDSC. The combination of 3M-052 plus CpG ODN (but not each agent alone) eradicated large primary tumors and established long-term protective immunity. Conclusion The combination of agonists targeting TLRs 7/8 and 9 represents a significant improvement in cancer immunotherapy.
Collapse
Affiliation(s)
- By Gan Zhao
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, USA
| | | | - Debra Tross
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, USA
| | - Dmitri Smirnov
- 3M Drug Delivery Systems Division, St. Paul MN 55144, USA
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, USA
| |
Collapse
|
17
|
Redirecting T cells to Ewing's sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS One 2012; 7:e31210. [PMID: 22355347 PMCID: PMC3280271 DOI: 10.1371/journal.pone.0031210] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 01/04/2012] [Indexed: 11/19/2022] Open
Abstract
We explored the possibility to target Ewing's sarcoma family of tumors (ESFT) by redirecting T cells. To this aim, we considered NKG2D-ligands (NKG2D-Ls) as possible target antigens. Detailed analysis of the expression of MICA, MICB, ULBP-1, -2, and -3 in fourteen ESFT cell lines revealed consistent expression of at least one NKG2D-L. Thus, for redirecting T cells, we fused a CD3ζ/CD28-derived signaling domain to the ectodomain of NKG2D, however, opposite transmembrane orientation of this signaling domain and NKG2D required inverse orientation fusion of either of them. We hypothesized that the particularly located C-terminus of the NKG2D ectodomain should allow reengineering of the membrane anchoring from a native N-terminal to an artificial C-terminal linkage. Indeed, the resulting chimeric NKG2D receptor (chNKG2D) was functional and efficiently mediated ESFT cell death triggered by activated T cells. Notably, ESFT cells with even low NKG2D-L expression were killed by CD8(pos) and also CD4(pos) cells. Both, mRNA transfection and lentiviral transduction resulted in high level surface expression of chNKG2D. However, upon target-cell recognition receptor surface levels were maintained by tranfected RNA only during the first couple of hours after transfection. Later, target-cell contact resulted in strong and irreversible receptor down-modulation, whereas lentivirally mediated expression of chNKG2D remained constant under these conditions. Together, our study defines NKG2D-Ls as targets for a CAR-mediated T cell based immunotherapy of ESFT. A comparison of two different methods of gene transfer reveals strong differences in the susceptibility to ligand-induced receptor down-modulation with possible implications for the applicability of RNA transfection.
Collapse
|
18
|
Steinhagen F, Kinjo T, Bode C, Klinman DM. TLR-based immune adjuvants. Vaccine 2011; 29:3341-55. [PMID: 20713100 PMCID: PMC3000864 DOI: 10.1016/j.vaccine.2010.08.002] [Citation(s) in RCA: 378] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 07/27/2010] [Accepted: 08/01/2010] [Indexed: 12/29/2022]
Abstract
This work describes the nature and strength of the immune response induced by various Toll-like receptor ligands and their ability to act as vaccine adjuvants. It reviews the various ligands capable of triggering individual TLRs, and then focuses on the efficacy and safety of those agents for which clinical results are available.
Collapse
Affiliation(s)
- Folkert Steinhagen
- Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, United States
| | | | | | | |
Collapse
|
19
|
Steele JC, Rao A, Marsden JR, Armstrong CJ, Berhane S, Billingham LJ, Graham N, Roberts C, Ryan G, Uppal H, Walker C, Young LS, Steven NM. Phase I/II trial of a dendritic cell vaccine transfected with DNA encoding melan A and gp100 for patients with metastatic melanoma. Gene Ther 2011; 18:584-93. [PMID: 21307889 DOI: 10.1038/gt.2011.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This trial tested a dendritic cell (DC) therapeutic cancer vaccine in which antigen is loaded using a novel non-viral transfection method enabling the uptake of plasmid DNA condensed with a cationic peptide. Proof of principle required the demonstration of diverse T lymphocyte responses following vaccination, including multiple reactivities restricted through both major histocompatibility complex (MHC) class I and II. Patients with advanced melanoma were offered four cycles of vaccination with autologous DC expressing melan A and gp100. Disease response was measured using Response Evaluation Criteria in Solid Tumours. Circulating MHC class I- and II-restricted responses were measured against peptide and whole antigen targets using interferon-γ ELIspot and enzyme-linked immunosorbent assay assays, respectively. Responses were analyzed across the trial population and presented descriptively for some individuals. Twenty-five patients received at least one cycle. Vaccination was well tolerated. Three patients had reduction in disease volume. Across the trial population, vaccination resulted in an expansion of effector responses to both antigens, to the human leukocyte antigen A2-restricted modified epitope, melan A ELAGIGILTV, and to a panel of MHC class I- and II-restricted epitopes. Vaccination with mature DC non-virally transfected with DNA encoding antigen had biological effect causing tumour regression and inducing diverse T lymphocyte responses.
Collapse
Affiliation(s)
- J C Steele
- Cancer Research UK Clinical Trials Unit, School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Targeting Toll-like receptor 7/8 enhances uptake of apoptotic leukemic cells by monocyte-derived dendritic cells but interferes with subsequent cytokine-induced maturation. Cancer Immunol Immunother 2010; 60:37-47. [PMID: 20859626 PMCID: PMC3029806 DOI: 10.1007/s00262-010-0917-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 08/31/2010] [Indexed: 12/13/2022]
Abstract
Therapeutic vaccination with dendritic cells (DC) is an emerging investigational therapy for eradication of minimal residual disease in acute myeloid leukemia. Various strategies are being explored in manufacturing DC vaccines ex vivo, e.g., monocyte-derived DC (MoDC) loaded with leukemia-associated antigens (LAA). However, the optimal source of LAA and the choice of DC-activating stimuli are still not well defined. Here, loading with leukemic cell preparations (harboring both unknown and known LAA) was explored in combination with a DC maturation-inducing cytokine cocktail (CC; IL-1β, IL-6, TNF-α, and PGE2) and Toll-like receptor ligands (TLR-L) to optimize uptake. Since heat shock induced apoptotic blasts were more efficiently taken up than lysates, we focused on uptake of apoptotic leukemic cells. Uptake of apoptotic blast was further enhanced by the TLR7/8-L R848 (20–30%); in contrast, CC-induced maturation inhibited uptake. CC, and to a lesser extent R848, enhanced the ability of MoDC to migrate and stimulate T cells. Furthermore, class II-associated invariant chain peptide expression was down-modulated after R848- or CC-induced maturation, indicating enhanced processing and presentation of antigenic peptides. To improve both uptake and maturation, leukemic cells and MoDC were co-incubated with R848 for 24 h followed by addition of CC. However, this approach interfered with CC-mediated MoDC maturation as indicated by diminished migratory and T cell stimulatory capacity, and the absence of IL-12 production. Taken together, our data demonstrate that even though R848 improved uptake of apoptotic leukemic cells, the sequential use of R848 and CC is counter-indicated due to its adverse effects on MoDC maturation.
Collapse
|
21
|
Chi N, Maranchie JK, Appleman LJ, Storkus WJ. Update on vaccine development for renal cell cancer. Res Rep Urol 2010; 2:125-41. [PMID: 24198621 PMCID: PMC3703676 DOI: 10.2147/rru.s7242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) remains a significant health concern that frequently presents as metastatic disease at the time of initial diagnosis. Current first-line therapeutics for the advanced-stage RCC include antiangiogenic drugs that have yielded high rates of objective clinical response; however, these tend to be transient in nature, with many patients becoming refractory to chronic treatment with these agents. Adjuvant immunotherapies remain viable candidates to sustain disease-free and overall patient survival. In particular, vaccines designed to optimize the activation, maintenance, and recruitment of specific immunity within or into the tumor site continue to evolve. Based on the integration of increasingly refined immunomonitoring systems in both translational models and clinical trials, allowing for the improved understanding of treatment mechanism(s) of action, further refined (combinational) vaccine protocols are currently being developed and evaluated. This review provides a brief history of RCC vaccine development, discusses the successes and limitations in such approaches, and provides a rationale for developing combinational vaccine approaches that may provide improved clinical benefits to patients with RCC.
Collapse
Affiliation(s)
- Nina Chi
- Department of immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|
22
|
Buschow SI, Lasonder E, van Deutekom HWM, Oud MM, Beltrame L, Huynen MA, de Vries IJM, Figdor CG, Cavalieri D. Dominant processes during human dendritic cell maturation revealed by integration of proteome and transcriptome at the pathway level. J Proteome Res 2010; 9:1727-37. [PMID: 20131907 DOI: 10.1021/pr9008546] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Gene expression is commonly used to study the activation of dendritic cells (DCs) to identify proteins that determine whether these cells induce an immunostimulatory or tolerogenic immune response. RNA expression, however, does not necessarily predict protein abundance and often requires large numbers of experiments for statistical significance. Proteomics provides a direct view on protein expression but is costly and time consuming. Here, we combined a comprehensive quantitative proteome and transcriptome analysis on a single batch of immature and cytokine cocktail matured human DCs and integrated resulting data sets at the pathway level. Although overall correlation between differential mRNA and protein expression was low, correlation between components of DC relevant pathways was significantly higher. Differentially expressed proteins and genes partly mapped to identical but also to different pathway components demonstrating that RNA and protein data not only supported but also complemented each other. We identified 5 dominant pathways, which confirmed the importance of cytokines, cell adhesion, and migration in DC maturation and also indicated a fundamental role for lipid metabolism. From these pathways we extracted novel maturation markers that might improve DC vaccine design. For several of the candidate markers we confirmed widespread significance examining DCs from multiple individuals, underscoring the validity of our approach. We conclude that integration of different but related data sets at the pathway level can significantly increase the predictive power of multi "omics" analyses.
Collapse
Affiliation(s)
- Sonja I Buschow
- Department of Tumor Immunology and CMBI at the Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Alexandrescu DT, Ichim TE, Riordan NH, Marincola FM, Di Nardo A, Kabigting FD, Dasanu CA. Immunotherapy for melanoma: current status and perspectives. J Immunother 2010; 33:570-90. [PMID: 20551839 PMCID: PMC3517185 DOI: 10.1097/cji.0b013e3181e032e8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immunotherapy is an important modality in the therapy of patients with malignant melanoma. As our knowledge about this disease continues to expand, so does the immunotherapeutic armamentarium. Nevertheless, successful preclinical models do not always translate into clinically meaningful results. The authors give a comprehensive analysis of most recent advances in the immune anti-melanoma therapy, including interleukins, interferons, other cytokines, adoptive immunotherapy, biochemotherapy, as well as the use of different vaccines. We also present the fundamental concepts behind various immune enhancement strategies, passive immunotherapy, as well as the use of immune adjuvants. This review brings into discussion the results of newer and older clinical trials, as well as potential limitations and drawbacks seen with the utilization of various immune therapies in malignant melanoma. Development of novel therapeutic approaches, along with optimization of existing therapies, continues to hold a great promise in the field of melanoma therapy research. Use of anti-CTLA4 and anti-PD1 antibodies, realization of the importance of co-stimulatory signals, which translated into the use of agonist CD40 monoclonal antibodies, as well as activation of innate immunity through enhanced expression of co-stimulatory molecules on the surface of dendritic cells by TLR agonists are only a few items on the list of recent advances in the treatment of melanoma. The need to engineer better immune interactions and to boost positive feedback loops appear crucial for the future of melanoma therapy, which ultimately resides in our understanding of the complexity of immune responses in this disease.
Collapse
Affiliation(s)
- Doru T Alexandrescu
- Division of Dermatology, University of California at San Diego, San Diego, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Hartiala P, Hytönen J, Yrjänäinen H, Honkinen M, Terho P, Söderström M, Penttinen MA, Viljanen MK. TLR2 Utilization ofBorreliaDoes Not Induce p38- and IFN-β Autocrine Loop-Dependent Expression of CD38, Resulting in Poor Migration and Weak IL-12 Secretion of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:5732-42. [DOI: 10.4049/jimmunol.0803944] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Jurewicz M, Takakura A, Augello A, Movahedi Naini S, Ichimura T, Zandi-Nejad K, Abdi R. Ischemic injury enhances dendritic cell immunogenicity via TLR4 and NF-kappa B activation. THE JOURNAL OF IMMUNOLOGY 2010; 184:2939-48. [PMID: 20164431 DOI: 10.4049/jimmunol.0901889] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ischemic (isc) injury during the course of transplantation enhances the immunogenicity of allografts and thus results in poorer graft outcome. Given the central role of dendritic cells (DCs) in mounting alloimmune responses, activation of donor DCs by ischemia may have a primary function in the increased immunogenicity of isc allografts. In this study, we sought to investigate the effect of ischemia on DC activity in vitro. Following induction of ischemia, bone marrow-derived DCs were shown to augment allogeneic T cell proliferation as well as the IFN-gamma response. Isc DCs produced greater levels of IL-6, and isc insult was concurrent with NF-kappaB activation. TLR4 ligation was also shown to occur in isc DCs, most likely in response to the endogenous ligand heat shock protein 70, which was found to be elevated in DCs following isc injury, and lack of TLR4 abrogated the observed effects of isc DCs. As compared with control DCs, isc DCs injected into the footpads of mice demonstrated enhanced migration, which was concomitant with increased recipient T cell activity. Moreover, isc DCs underwent a greater degree of apoptosis in the lymph nodes of injected mice, which may further demonstrate enhanced immunogenicity of isc DCs. We thus show that isc injury of DCs enhances DC function, augments the allogeneic T cell response, and occurs via ligation of TLR4, followed by activation of NF-kappaB. These data may serve to identify novel therapeutic targets to attenuate graft immunogenicity following ischemia.
Collapse
Affiliation(s)
- Mollie Jurewicz
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Kim HJ, Kim HO, Lee K, Baek EJ, Kim HS. Two-step maturation of immature DCs with proinflammatory cytokine cocktail and poly(I:C) enhances migratory and T cell stimulatory capacity. Vaccine 2010; 28:2877-86. [PMID: 20156531 DOI: 10.1016/j.vaccine.2010.01.061] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/22/2010] [Accepted: 01/28/2010] [Indexed: 12/22/2022]
Abstract
Effective induction of cell-mediated immune responses strongly depends on the ability of dendritic cells (DCs) to produce Th1-polarizing cytokines, migrate to lymph nodes and stimulate T cells through antigen-presenting complex and costimulatory molecules. While various protocols for optimizing DC maturation with single or multiple stimuli mimicking infections or inflammatory milieu have been proposed for the generation of DCs with features desired for clinical application, stepwise maturation of DCs by these multiple stimuli has not been systemically assessed. Among the combinations of several immune-modulating factors with known effects on DC maturation, we found that stepwise DC maturation with cytokine cocktail (TNF-alpha+IL-6+IL-1 beta+PGE(2)) followed by poly(I:C) stimulation enhanced the production of IL-12 with strong allostimulatory capacity. While there were no significant differences between DC matured by simultaneous or sequential activation by cytokine cocktail and poly(I:C) in expression of markers and costimulatory molecules of mature DCs, the delivery of inflammatory signal prior to poly(I:C) results in sustained interleukin-12 expression with reduced IL-10 than DC matured by simultaneous stimulation. This sequential stimulation significantly increased migratory capacity in response to CCL21 and CXCL12 compared to DC matured with cytokine cocktail. Furthermore, these DCs retained their responsiveness to CD40L stimulation in secondary IL-12 production and efficiently generated autologous antigen-specific effector T cells as evidenced by ELISPOT assay. Thus, we propose a novel DC maturation protocol in which stimulation of DCs with cytokine cocktail and subsequently with poly(I:C) generates DCs with a high migratory capacity with a preferential Th1 inducing capacity.
Collapse
Affiliation(s)
- Hyung Jin Kim
- Division of Cardiovascular and Rare Disease, Korea Center for Disease Control and Prevention 194 Tongillo, Eunpyung-gu, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
27
|
Yu CF, Peng WM, Oldenburg J, Hoch J, Bieber T, Limmer A, Hartmann G, Barchet W, Eis-Hübinger AM, Novak N. Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation. THE JOURNAL OF IMMUNOLOGY 2009; 184:1159-67. [PMID: 20026744 DOI: 10.4049/jimmunol.0901706] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Signals involved in the commitment of Th17 differentiation are of substantial interest for our understanding of antimicrobial defense mechanisms and autoimmune disorders. Various ways in which myeloid dendritic cells modulate Th17 differentiation have been identified. However, although plasmacytoid dendritic cells (PDCs) are regarded as important players in antiviral/antimicrobial host defense and autoimmune diseases, a putative modulatory role of PDCs in Th17 differentiation has not yet been elucidated in detail. We demonstrated that PDCs are capable of promoting Th17 differentiation in response to TLR7 stimulation. Further, both the differentiation of Th17 cells from naive T cells and the amplification of Th17 effector functions of memory T cells are promoted by PDCs after TLR7 activation. Our data are of strong clinical relevance because TLR7 activation in PDCs might represent one of the missing links between innate and adaptive immune mechanisms and contribute to the amplification of Th17-driven autoimmune disorders as well as viral host defense.
Collapse
Affiliation(s)
- Chun-Feng Yu
- Department of Dermatology and Allergy, University Hospital, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Anguille S, Smits ELJM, Cools N, Goossens H, Berneman ZN, Van Tendeloo VFI. Short-term cultured, interleukin-15 differentiated dendritic cells have potent immunostimulatory properties. J Transl Med 2009; 7:109. [PMID: 20021667 PMCID: PMC2807857 DOI: 10.1186/1479-5876-7-109] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 12/18/2009] [Indexed: 12/24/2022] Open
Abstract
Background Optimization of the current dendritic cell (DC) culture protocol in order to promote the therapeutic efficacy of DC-based immunotherapy is warranted. Alternative differentiation of monocyte-derived DCs using granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-15 has been propagated as an attractive strategy in that regard. The applicability of these so-called IL-15 DCs has not yet been firmly established. We therefore developed a novel pre-clinical approach for the generation of IL-15 DCs with potent immunostimulatory properties. Methods Human CD14+ monocytes were differentiated with GM-CSF and IL-15 into immature DCs. Monocyte-derived DCs, conventionally differentiated in the presence of GM-CSF and IL-4, served as control. Subsequent maturation of IL-15 DCs was induced using two clinical grade maturation protocols: (i) a classic combination of pro-inflammatory cytokines (tumor necrosis factor-α, IL-1β, IL-6, prostaglandin E2) and (ii) a Toll-like receptor (TLR)7/8 agonist-based cocktail (R-848, interferon-γ, TNF-α and prostaglandin E2). In addition, both short-term (2-3 days) and long-term (6-7 days) DC culture protocols were compared. The different DC populations were characterized with respect to their phenotypic profile, migratory properties, cytokine production and T cell stimulation capacity. Results The use of a TLR7/8 agonist-based cocktail resulted in a more optimal maturation of IL-15 DCs, as reflected by the higher phenotypic expression of CD83 and costimulatory molecules (CD70, CD80, CD86). The functional superiority of TLR7/8-activated IL-15 DCs over conventionally matured IL-15 DCs was evidenced by their (i) higher migratory potential, (ii) advantageous cytokine secretion profile (interferon-γ, IL-12p70) and (iii) superior capacity to stimulate autologous, antigen-specific T cell responses after passive peptide pulsing. Aside from a less pronounced production of bioactive IL-12p70, short-term versus long-term culture of TLR7/8-activated IL-15 DCs resulted in a migratory profile and T cell stimulation capacity that was in favour of short-term DC culture. In addition, we demonstrate that mRNA electroporation serves as an efficient antigen loading strategy of IL-15 DCs. Conclusions Here we show that short-term cultured and TLR7/8-activated IL-15 DCs fulfill all pre-clinical prerequisites of immunostimulatory DCs. The results of the present study might pave the way for the implementation of IL-15 DCs in immunotherapy protocols.
Collapse
Affiliation(s)
- Sébastien Anguille
- University of Antwerp - Faculty of Medicine, Vaccine & Infectious Disease Institute (Vaxinfectio), Laboratory of Experimental Hematology, Universiteitsplein 1, B-2610 Wilrijk (Antwerp), Belgium.
| | | | | | | | | | | |
Collapse
|
29
|
Haenssle HA, Riedl P, Buhl T, Schardt A, Rosenberger A, Schön MP, Schirmbeck R. Intracellular delivery of major histocompatibility complex class I-binding epitopes: dendritic cells loaded and matured with cationic peptide/poly(I:C) complexes efficiently activate T cells. Exp Dermatol 2009; 19:19-28. [PMID: 19758325 DOI: 10.1111/j.1600-0625.2009.00954.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Based on their role for the induction of T-cell responses, dendritic cells (DCs) are popular candidates in cancer vaccine development. We established a novel single-step intracellular delivery of peptide/poly(I:C) complexes for antigen loading and Toll-like receptor-3 (TLR3)-mediated maturation of human DCs using a cell-penetrating peptide (tat(49-57): RKKRRQRRR) as delivery vector. Towards this end, a cationic tat-sequence was fused with an antigenic, major histocompatibility complex (MHC) class I-binding melanoma epitope (Melan-A/Mart-1 sequence: ELAGIGILTV) and then mixed with negatively charged poly(I:C) dsRNA to form peptide/nucleic acid complexes. Flow cytometry and confocal laser scanning microscopy confirmed intracellular localization of TLR3 in monocyte-derived immature DCs (iDCs). Peptide/poly(I:C) complexes were readily internalized by iDCs without negatively affecting cell viability. They induced DC maturation and secretion of bioactive interleukin (IL)-12p70. When peptide/poly(I:C) complex-loaded DCs were used for autologous T cell stimulation, epitope-specific interferon-gamma secretion was quantitatively superior in comparison to peptide-loaded DCs matured by a cytokine cocktail, as detected by enzyme-linked immunospot assays. Thus, complexes of cationic antigenic peptides and poly(I:C) might be of great utility for a TLR3-mediated DC maturation and intracellular peptide targeting in a single step. Resulting DCs induce a strong expansion/activation of antigen-specific T cells in the context of an IL-12p70 secretion.
Collapse
Affiliation(s)
- Holger A Haenssle
- Department of Dermatology, Georg-August-University Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
Lehner M, Grillhoesl C, Full F, Vogel B, Weller P, Müller-Fleckenstein I, Schmidt M, Fleckenstein B, Holter W, Ensser A. Transformation efficiency by Herpesvirus saimiri is not a limiting factor in clonal CD8pos T cell outgrowth. Virology 2009; 388:15-20. [PMID: 19375768 DOI: 10.1016/j.virol.2009.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 12/30/2008] [Accepted: 03/14/2009] [Indexed: 10/20/2022]
Abstract
The routine transformation of human CD8(pos) T cells by Herpesvirus saimiri has so far not been achieved in the case of pre-expanded antigen-specific CTLs. Here we transformed 73% of polyclonal EBV-specific CD8(pos) T cell cultures using an optimized culture medium supplemented with IL-2, IL-7, IL-12, and TGF-beta(1). Still, antigen-specific cytotoxicity was frequently lost and analysis of the TCR Vbeta-chain repertoire revealed a variable outgrowth of several initially subdominant populations. Limiting dilution cloning of cells in the presence of high titers of HVS did not result in clonal transformation but in the rapid loss of the viral genome in outgrowing clones. In summary, our data suggest that transformation of CD8(pos) T cells out of bulk cultures can be routinely achieved, while viral transformation itself remains an infrequent event on a per cell basis. The practical use of the improved immortalization of antigen-expanded CD8(pos) T cell lines, however, is limited by the arbitrary outgrowth of subdominant populations of unpredictable specificity.
Collapse
Affiliation(s)
- Manfred Lehner
- Labor für Zelltherapie, Abteilung Haematologie und Onkologie, Klinik für Kinder und Jugendliche, Universitätsklinikum, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Powell ND, Bailey MT, Mays JW, Stiner-Jones LM, Hanke ML, Padgett DA, Sheridan JF. Repeated social defeat activates dendritic cells and enhances Toll-like receptor dependent cytokine secretion. Brain Behav Immun 2009; 23:225-31. [PMID: 18848983 PMCID: PMC2711866 DOI: 10.1016/j.bbi.2008.09.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 09/08/2008] [Accepted: 09/18/2008] [Indexed: 02/02/2023] Open
Abstract
Stress hormones significantly impact dendritic cell (DC) activation and function, typically in a suppressive fashion. However, a social stressor termed social disruption (SDR) has been shown to induce an increase in inflammatory responses and a state of glucocorticoid resistance in splenic CD11b+ monocytes. These experiments were designed to determine the effects of SDR on DC activation, Toll-like receptor-induced cytokine secretion, and glucocorticoid sensitivity. Compared to cells obtained from control animals, splenic DCs from SDR mice displayed increased levels of MHC I, CD80, and CD44, indicative of an activated phenotype. In addition, DCs from SDR mice produced comparatively higher TNF-alpha, IL-6, and IL-10 in response to in vitro stimulation with LPS and CpG DNA. Increased amounts of TNF-alpha and IL-6 were also evident in SDR DC cultures stimulated with poly(I:C). Furthermore, as shown previously in CD11b+ monocytes, the CD11c+ DCs obtained from SDR mice were glucocorticoid resistant. Taken together, the data suggest that social stress, in the absence of any immune challenge, activates DCs, increases DC cytokine secretion in response to Toll-specific stimuli and renders DCs glucocorticoid resistant.
Collapse
Affiliation(s)
- ND Powell
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - MT Bailey
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - JW Mays
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - LM Stiner-Jones
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - ML Hanke
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - DA Padgett
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine. The Ohio State University, Columbus, OH, USA
| | - JF Sheridan
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine. The Ohio State University, Columbus, OH, USA
| |
Collapse
|
32
|
Benko S, Magyarics Z, Szabó A, Rajnavölgyi E. Dendritic cell subtypes as primary targets of vaccines: the emerging role and cross-talk of pattern recognition receptors. Biol Chem 2008; 389:469-85. [PMID: 18953714 DOI: 10.1515/bc.2008.054] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Preventive vaccination is the most successful approach against infectious diseases and has a great impact on world health. Vaccines operate through the activation of innate immunity that helps to stimulate antigen-specific T- and B-lymphocytes. These events are orchestrated by dendritic cells (DCs) that are able to sample foreign structures and concomitantly sense 'danger signals'. Thus, DCs provide a functional link between innate and acquired immunity, and due to their regulatory potential are referred to as natural adjuvants. Human conventional and plasmacytoid DCs express different sets of well-characterized Toll-like membrane receptors (TLRs) that recognize a broad range of conserved molecular patterns of pathogens. The recently discovered cytosolic Nod-like receptors (NLRs) and RIG-like helicases (RLHs) also turned out to participate in pathogen recognition and modulation of immune responses through interacting signaling pathways. As a result of their collaboration, the TLR, NLR and RLH recognition systems induce the secretion of different combinations of cytokines that play a fundamental role in T-cell activation and instruction. Ligands of the innate recognition systems emerge as new adjuvants for vaccine design, whereas manipulation of the signaling pathways mediated by these receptors offers new avenues for fine tuning immune responses and optimizing immunotherapies.
Collapse
Affiliation(s)
- Szilvia Benko
- Institute of Immunology, Medical and Health Science Centre, University of Debrecen, H-4032 Debrecen, Hungary
| | | | | | | |
Collapse
|
33
|
LPS and poly I:C induce chromatin modifications at a novel upstream region of the IL-23 p19 promoter. Inflammation 2008; 31:235-46. [PMID: 18587636 DOI: 10.1007/s10753-008-9070-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IL-23, a heterodimer of IL-12 p40 and IL-23 p19, is critical for an effective immune response to many infections and has been implicated in several autoimmune diseases, however, little is known about the regulation of IL-23 gene expression in monocytes. We found that poly I:C, LPS, flagellin, and zymogen activated significant IL-23 production in primary human monocytes. Using chromatin immunoprecipitation, we found that a distal upstream region of the IL-23 p19 promoter at -601 to -521 underwent extensive histone modifications in response to stimuli. This distal region of the promoter is not highly conserved between species and has not been previously implicated in the regulation of IL-23 expression. Knockdown of CBP markedly decreased IL-23 p19 responses to poly I:C but had a less dramatic effect on LPS responses, confirming different chromatin responses to these two stimuli. Our data suggest that one of the mechanisms regulating IL-23 expression is the regulation of histone modifications at this distal upstream region of the promoter.
Collapse
|
34
|
Verdijk P, Aarntzen EHJG, Punt CJA, de Vries IJM, Figdor CG. Maximizing dendritic cell migration in cancer immunotherapy. Expert Opin Biol Ther 2008; 8:865-74. [PMID: 18549318 DOI: 10.1517/14712598.8.7.865] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The success of dendritic cell (DC)-based immunotherapy in inducing cellular immunity against tumors is highly dependent on accurate delivery and trafficking of the DC to T-cell-rich areas of secondary lymphoid tissues. OBJECTIVE To provide an overview of DC migration in vivo and how migration to peripheral lymph nodes might be improved to optimize DC therapy. METHODS We focused on DC migration in preclinical models and human skin explants and on clinical vaccination trials studying migration of in vitro-generated DC. RESULTS/CONCLUSIONS DC migration requires an intricate interplay between the cell and its environment. To maximize migration for cellular therapy, it is important to optimize the generation of migratory DC as well as treatment strategies.
Collapse
Affiliation(s)
- Pauline Verdijk
- Radboud University Nijmegen Medical Centre, Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
35
|
|
36
|
Jasny E, Eisenblätter M, Mätz-Rensing K, Tenner-Racz K, Tenbusch M, Schrod A, Stahl-Hennig C, Moos V, Schneider T, Racz P, Uberla K, Kaup FJ, Ignatius R. IL-12-impaired and IL-12-secreting dendritic cells produce IL-23 upon CD154 restimulation. THE JOURNAL OF IMMUNOLOGY 2008; 180:6629-39. [PMID: 18453582 DOI: 10.4049/jimmunol.180.10.6629] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental studies in monkeys on the basis of ex vivo-generated, reinjected dendritic cells (DCs) allow investigations of primate DC biology in vivo. To study in vitro and in vivo properties of DCs with a reduced capacity to produce IL-12, we adapted findings obtained in vitro with human cells to the rhesus macaque model. Following exposure of immature monocyte-derived monkey DCs to the immunomodulating synthetic polypeptide glatiramer acetate (GA) and to dibutyryl-cAMP (d-cAMP; i.e., a cAMP enhancer that activates DCs but inhibits the induction of Th1 immune responses), the resulting DCs displayed a mature phenotype with enhanced Ag-specific T cell stimulatory function, notably also for memory Th1 cells. Phosphorylation of p38 MAPK was not induced in GA/d-cAMP-activated DCs. Accordingly, these cells secreted significantly less IL-12p40 (p < or = 0.001) than did cytokine-activated cells. However, upon restimulation with rhesus macaque CD154, GA/d-cAMP-activated DCs produced IL-12p40/IL-23. Additionally, DCs activated by proinflammatory cytokines following protocols for the generation of cells used in clinical studies secreted significantly more IL-23 upon CD154 restimulation than following prior activation. Two days after intradermal injection, GA/d-cAMP-activated fluorescence-labeled DCs were detected in the T cell areas of draining lymph nodes. When similarly injected, GA/d-cAMP as well as cytokine-activated protein-loaded DCs induced comparable Th immune responses characterized by secretion of IFN-gamma, TNF, and IL-17, and transiently expanded FOXP3(+) regulatory T cells. Reactivation of primate DCs through CD154 considerably influences their immmunostimulatory properties. This may have a substantial impact on the development of innovative vaccine approaches.
Collapse
Affiliation(s)
- Edith Jasny
- Institute of Microbiology and Hygiene, Department of Infection Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Dendritic cell vaccines in melanoma: From promise to proof? Crit Rev Oncol Hematol 2008; 66:118-34. [DOI: 10.1016/j.critrevonc.2007.12.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 12/03/2007] [Accepted: 12/13/2007] [Indexed: 02/02/2023] Open
|
38
|
Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother 2008; 57:1589-97. [PMID: 18322684 PMCID: PMC2522299 DOI: 10.1007/s00262-008-0489-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Accepted: 02/12/2008] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells of the immune system that play a key role in regulating T cell-based immunity. In vivo, the capacity of DC to activate T cells depends on their ability to migrate to the T cell areas of lymph nodes as well as on their maturation state. Depending on their cytokine-secreting profile, DC are able to skew the immune response in a specific direction. In particular, IL-12p70 producing DC drive T cells towards a T helper 1 type response. A serious disadvantage of current clinical grade ex vivo generated monocyte-derived DC is the poor IL-12p70 production. We have investigated the effects of Toll-like receptor (TLR)-mediated maturation on ex vivo generated human monocyte-derived DC. We demonstrate that in contrast to cytokine-matured DC, DC matured with poly(I:C) (TLR3 ligand) and/or R848 (TLR7/8 ligand) are able to produce vast amounts of IL-12p70, but exhibit a reduced migratory capacity. The addition of prostaglandin E2 (PGE2) improved the migratory capacity of TLR-ligand matured DC while maintaining their IL-12p70 production upon T cell encounter. We propose a novel clinical grade maturation protocol in which TLR ligands poly(I:C) and R848 are combined with PGE2 to generate DC with both high migratory capacity and IL-12p70 production upon T cell encounter.
Collapse
|