1
|
Tiwari P, Yadav K, Shukla RP, Bakshi AK, Panwar D, Das S, Mishra PR. Extracellular vesicles-powered immunotherapy: Unleashing the potential for safer and more effective cancer treatment. Arch Biochem Biophys 2024; 756:110022. [PMID: 38697343 DOI: 10.1016/j.abb.2024.110022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Cancer treatment has seen significant advancements with the introduction of Onco-immunotherapies (OIMTs). Although some of these therapies have received approval for use, others are either undergoing testing or are still in the early stages of development. Challenges persist in making immunotherapy widely applicable to cancer treatment. To maximize the benefits of immunotherapy and minimize potential side effects, it's essential to improve response rates across different immunotherapy methods. A promising development in this area is the use of extracellular vesicles (EVs) as novel delivery systems. These small vesicles can effectively deliver immunotherapies, enhancing their effectiveness and reducing harmful side effects. This article discusses the importance of integrating nanomedicines into OIMTs, highlighting the challenges with current anti-OIMT methods. It also explores key considerations for designing nanomedicines tailored for OIMTs, aiming to improve upon existing immunotherapy techniques. Additionally, the article looks into innovative approaches like biomimicry and the use of natural biomaterial-based nanocarriers (NCs). These advancements have the potential to transform the delivery of immunotherapy. Lastly, the article addresses the challenges of moving OIMTs from theory to clinical practice, providing insights into the future of using advanced nanotechnology in cancer treatment.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Sweety Das
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
2
|
Kumari K, Singh A, Chaudhary A, Singh RK, Shanker A, Kumar V, Haque R. Neoantigen Identification and Dendritic Cell-Based Vaccines for Lung Cancer Immunotherapy. Vaccines (Basel) 2024; 12:498. [PMID: 38793749 PMCID: PMC11125796 DOI: 10.3390/vaccines12050498] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Immunotherapies can treat many cancers, including difficult-to-treat cases such as lung cancer. Due to its tolerability, long-lasting therapeutic responses, and efficacy in a wide spectrum of patients, immunotherapy can also help to treat lung cancer, which has few treatment choices. Tumor-specific antigens (TSAs) for cancer vaccinations and T-cell therapies are difficult to discover. Neoantigens (NeoAgs) from genetic mutations, irregular RNA splicing, protein changes, or viral genetic sequences in tumor cells provide a solution. NeoAgs, unlike TSAs, are non-self and can cause an immunological response. Next-generation sequencing (NGS) and bioinformatics can swiftly detect and forecast tumor-specific NeoAgs. Highly immunogenic NeoAgs provide personalized or generalized cancer immunotherapies. Dendritic cells (DCs), which originate and regulate T-cell responses, are widely studied potential immunotherapeutic therapies for lung cancer and other cancers. DC vaccines are stable, reliable, and safe in clinical trials. The purpose of this article is to evaluate the current status, limitations, and prospective clinical applications of DC vaccines, as well as the identification and selection of major histocompatibility complex (MHC) class I and II genes for NeoAgs. Our goal is to explain DC biology and activate DC manipulation to help researchers create extremely potent cancer vaccines for patients.
Collapse
Affiliation(s)
- Komal Kumari
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Amarnath Singh
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA;
| | - Archana Chaudhary
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Rakesh Kumar Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India;
| | - Asheesh Shanker
- Department of Bioinformatics, Central University of South Bihar, Gaya 824236, Bihar, India
| | - Vinay Kumar
- Heart and Vascular Institute, Pennsylvania State University, Hershey Medical Center, Hershey, PA 17033, USA;
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| |
Collapse
|
3
|
Jha A, Ahad A, Mishra GP, Sen K, Smita S, Minz AP, Biswas VK, Tripathy A, Senapati S, Gupta B, Acha-Orbea H, Raghav SK. SMRT and NCoR1 fine-tune inflammatory versus tolerogenic balance in dendritic cells by differentially regulating STAT3 signaling. Front Immunol 2022; 13:910705. [PMID: 36238311 PMCID: PMC9552960 DOI: 10.3389/fimmu.2022.910705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cell (DC) fine-tunes inflammatory versus tolerogenic responses to protect from immune-pathology. However, the role of co-regulators in maintaining this balance is unexplored. NCoR1-mediated repression of DC immune-tolerance has been recently reported. Here we found that depletion of NCoR1 paralog SMRT (NCoR2) enhanced cDC1 activation and expression of IL-6, IL-12 and IL-23 while concomitantly decreasing IL-10 expression/secretion. Consequently, co-cultured CD4+ and CD8+ T-cells depicted enhanced Th1/Th17 frequency and cytotoxicity, respectively. Comparative genomic and transcriptomic analysis demonstrated differential regulation of IL-10 by SMRT and NCoR1. SMRT depletion represses mTOR-STAT3-IL10 signaling in cDC1 by down-regulating NR4A1. Besides, Nfkbia and Socs3 were down-regulated in Ncor2 (Smrt) depleted cDC1, supporting increased production of inflammatory cytokines. Moreover, studies in mice showed, adoptive transfer of SMRT depleted cDC1 in OVA-DTH induced footpad inflammation led to increased Th1/Th17 and reduced tumor burden after B16 melanoma injection by enhancing oncolytic CD8+ T-cell frequency, respectively. We also depicted decreased Ncor2 expression in Rheumatoid Arthritis, a Th1/Th17 disease.
Collapse
Affiliation(s)
- Atimukta Jha
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Manipal Academy of Higher Education, Manipal, KA, India
| | - Abdul Ahad
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Manipal Academy of Higher Education, Manipal, KA, India
| | - Gyan Prakash Mishra
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Kaushik Sen
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Haryana, India
| | - Shuchi Smita
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Manipal Academy of Higher Education, Manipal, KA, India
| | - Aliva Prity Minz
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
| | - Viplov Kumar Biswas
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Archana Tripathy
- Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Shantibhushan Senapati
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Manipal Academy of Higher Education, Manipal, KA, India
| | - Bhawna Gupta
- Department of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Hans Acha-Orbea
- Department of Biochemistry Center of Immunity and Infection Lausanne (CIIL), University of Lausanne (UNIL), Epalinges, Switzerland
| | - Sunil Kumar Raghav
- Immuno-genomics & Systems Biology laboratory, Institute of Life Sciences (ILS), Bhubaneswar, OR, India
- Manipal Academy of Higher Education, Manipal, KA, India
| |
Collapse
|
4
|
GP96 and SMP30 Protein Priming of Dendritic Cell Vaccination Induces a More Potent CTL Response against Hepatoma. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2518847. [PMID: 35070229 PMCID: PMC8767371 DOI: 10.1155/2022/2518847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
Heat-shock protein (HSP) GP96 is a well-known adjuvant in immunotherapy. It belongs to the HSP90 family. Our previous study demonstrated that DC pulsed with recombinant senescence marker protein 30 (SMP30) could induce cytotoxic T lymphocytes (CTLs) against liver cancer cells in vitro. In this study, SMP30 and GP96 were subcloned into lentiviruses and transfected into DCs from healthy donors. We included six groups: the GP96-SMP30 group, GP96 group, SMP30 group, DC group, empty vector control group, and hepatoma extracted protein group. We used ELISA to detect cytokines and flow cytometry to assess CD80 and CD86 on DCs and the effect of CTLs. Our vector design was considered successful and further studied. In the SMP30 group, DC expresses more CCR7 and CD86 than the control group; in the SMP30+GP96 group, DC express more CCR7, CD86, and CD80 than the control group. Transfected DCs secreted more TNF-α and interferon-β and induced more CTLs than control DCs. SMP30 + GP96 effectively stimulated the proliferation of T cells compared with control treatment (P < 0.01). We detected the cytokines TNF-α, TNF-β, IL-12, and IFN (α, β, and γ) via ELISA (Figure 5) and verified the killing effect via FCM. Four E : T ratios (0 : 1, 10 : 1, 20 : 1, and 40 : 1) were tested. The higher the ratio was, the better the effects were. We successfully constructed a liver cancer model and tested the CTL effect in each group. The GP96 + SMP30 group showed a better effect than the other groups. GP96 and SMP30 can stimulate DCs together and produce more potent antitumor effects. Our research may provide a new efficient way to improve the therapeutic effect of DC vaccines in liver cancer.
Collapse
|
5
|
Stevens D, Ingels J, Van Lint S, Vandekerckhove B, Vermaelen K. Dendritic Cell-Based Immunotherapy in Lung Cancer. Front Immunol 2021; 11:620374. [PMID: 33679709 PMCID: PMC7928408 DOI: 10.3389/fimmu.2020.620374] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. The advent of immune checkpoint inhibitors has led to a paradigm shift in the treatment of metastatic non-small cell and small cell lung cancer. However, despite prolonged overall survival, only a minority of the patients derive clinical benefit from these treatments suggesting that the full anti-tumoral potential of the immune system is not being harnessed yet. One way to overcome this problem is to combine immune checkpoint blockade with different strategies aimed at inducing or restoring cellular immunity in a tumor-specific, robust, and durable way. Owing to their unique capacity to initiate and regulate T cell responses, dendritic cells have been extensively explored as tools for immunotherapy in many tumors, including lung cancer. In this review, we provide an update on the nearly twenty years of experience with dendritic cell-based immunotherapy in lung cancer. We summarize the main results from the early phase trials and give an overview of the future perspectives within this field.
Collapse
Affiliation(s)
- Dieter Stevens
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Sandra Van Lint
- Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium.,GMP Cell Therapy Unit, Department of Regenerative Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karim Vermaelen
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
6
|
Marino F, Semilietof A, Michaux J, Pak HS, Coukos G, Müller M, Bassani-Sternberg M. Biogenesis of HLA Ligand Presentation in Immune Cells Upon Activation Reveals Changes in Peptide Length Preference. Front Immunol 2020; 11:1981. [PMID: 32983136 PMCID: PMC7485268 DOI: 10.3389/fimmu.2020.01981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/22/2020] [Indexed: 02/05/2023] Open
Abstract
Induction of an effective tumor immunity is a complex process that includes the appropriate presentation of the tumor antigens, activation of specific T cells, and the elimination of malignant cells. Potent and efficient T cell activation is dependent on multiple factors, such as timely expression of co-stimulatory molecules, the differentiation state of professional antigen presenting cells (e.g., dendritic cells; DCs), the functionality of the antigen processing and presentation machinery (APPM), and the repertoire of HLA class I and II-bound peptides (termed immunopeptidome) presented to T cells. So far, how molecular perturbations underlying DCs maturation and differentiation affect the in vivo cross-presented HLA class I and II immunopeptidomes is largely unknown. Yet, this knowledge is crucial for further development of DC-based immunotherapy approaches. We applied a state-of-the-art sensitive MS-based immunopeptidomics approach to characterize the naturally presented HLA-I and -II immunopeptidomes eluted from autologous immune cells having distinct functional and biological states including CD14+ monocytes, immature DC (ImmDC) and mature DC (MaDC) monocyte-derived DCs and naive or activated T and B cells. We revealed a presentation of significantly longer HLA peptides upon activation that is HLA allotype specific. This was apparent in the self-peptidome upon cell activation and in the context of presentation of exogenously loaded antigens, suggesting that peptide length is an important feature with potential implications on the rational design of anti-cancer vaccines.
Collapse
Affiliation(s)
- Fabio Marino
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Aikaterini Semilietof
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Justine Michaux
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Hui-Song Pak
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - George Coukos
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Markus Müller
- Vital IT, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Agora Center, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
7
|
Morrissey ME, Byrne R, Nulty C, McCabe NH, Lynam-Lennon N, Butler CT, Kennedy S, O'Toole D, Larkin J, McCormick P, Mehigan B, Cathcart MC, Lysaght J, Reynolds JV, Ryan EJ, Dunne MR, O'Sullivan J. The tumour microenvironment of the upper and lower gastrointestinal tract differentially influences dendritic cell maturation. BMC Cancer 2020; 20:566. [PMID: 32552799 PMCID: PMC7302160 DOI: 10.1186/s12885-020-07012-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Only 10–30% of oesophageal and rectal adenocarcinoma patients treated with neoadjuvant chemoradiotherapy have a complete pathological response. Inflammatory and angiogenic mediators in the tumour microenvironment (TME) may enable evasion of anti-tumour immune responses. Methods The TME influence on infiltrating dendritic cells (DCs) was modelled by treating immature monocyte-derived DCs with Tumour Conditioned Media (TCM) from distinct gastrointestinal sites, prior to LPS-induced maturation. Results Cell line conditioned media from gastrointestinal cell lines inhibited LPS-induced DC markers and TNF-α secretion. TCM generated from human tumour biopsies from oesophageal, rectal and colonic adenocarcinoma induced different effects on LPS-induced DC markers - CD54, CD80, HLA-DR, CD86 and CD83 were enhanced by oesophageal cancer; CD80, CD86 and CD83 were enhanced by rectal cancer, whereas CD54, HLA-DR, CD86, CD83 and PD-L1 were inhibited by colonic cancer. Notably, TCM from all GI cancer types inhibited TNF-α secretion. Additionally, TCM from irradiated biopsies inhibited DC markers. Profiling the TCM showed that IL-2 levels positively correlated with maturation marker CD54, while Ang-2 and bFGF levels negatively correlated with CD54. Conclusion This study identifies that there are differences in DC maturational capacity induced by the TME of distinct gastrointestinal cancers. This could potentially have implications for anti-tumour immunity and response to radiotherapy.
Collapse
Affiliation(s)
- Maria E Morrissey
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Róisín Byrne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Celina Nulty
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Niamh H McCabe
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Clare T Butler
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Susan Kennedy
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Dermot O'Toole
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | | | | | | | - Mary-Clare Cathcart
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.,Oesophageal Unit, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, Education and Research Centre, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland.,Department of Biological Sciences, Health Research Institute, University of Limerick, Castletroy, Co., Limerick, Ireland
| | - Margaret R Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
8
|
Chang H, Zou Z, Wang Q, Li J, Jin H, Yin Q, Xing D. Targeting and Specific Activation of Antigen-Presenting Cells by Endogenous Antigen-Loaded Nanoparticles Elicits Tumor-Specific Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1900069. [PMID: 31921548 PMCID: PMC6947714 DOI: 10.1002/advs.201900069] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/03/2019] [Indexed: 05/19/2023]
Abstract
Immunotherapy has shown tremendous promise for improving cancer treatment. Unfortunately, antigen-presenting cells (APCs) in cancer patients cannot effectively recognize and process tumor antigens to activate host immune responses. In this study, an approach is developed to improve cancer immunotherapy that utilizes endogenous antigen-carrying nanoparticles (EAC-NPs), which encompasses a set of antigens isolated from solid tumors and adjuvants. The EAC-NPs specifically target APCs and subsequently result in enhanced T cell responses and improved antitumor efficacy. Mechanistic studies reveal that the EAC-NPs enhance and prolong the presence of immune compounds in APCs, which ensure persistent antigen loading and stimulation, induce a rapid proliferation of CD4+ and CD8+ T cells, and significantly increase the ratios of intratumoral CD4+ T/Treg and CD8+ T/Treg. The work using nanotechnology provides a promising strategy in improving antitumor immunity by enhancing the immunogenicity and presentation of tumor self-antigens for cancer immunotherapy.
Collapse
Affiliation(s)
- Hao‐Cai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Zheng‐Zhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qiu‐Hong Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Jie Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qian‐Xia Yin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| |
Collapse
|
9
|
Krekorian M, Fruhwirth GO, Srinivas M, Figdor CG, Heskamp S, Witney TH, Aarntzen EHJG. Imaging of T-cells and their responses during anti-cancer immunotherapy. Theranostics 2019; 9:7924-7947. [PMID: 31656546 PMCID: PMC6814447 DOI: 10.7150/thno.37924] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has proven to be an effective approach in a growing number of cancers. Despite durable clinical responses achieved with antibodies targeting immune checkpoint molecules, many patients do not respond. The common denominator for immunotherapies that have successfully been introduced in the clinic is their potential to induce or enhance infiltration of cytotoxic T-cells into the tumour. However, in clinical research the molecules, cells and processes involved in effective responses during immunotherapy remain largely obscure. Therefore, in vivo imaging technologies that interrogate T-cell responses in patients represent a powerful tool to boost further development of immunotherapy. This review comprises a comprehensive analysis of the in vivo imaging technologies that allow the characterisation of T-cell responses induced by anti-cancer immunotherapy, with emphasis on technologies that are clinically available or have high translational potential. Throughout we discuss their respective strengths and weaknesses, providing arguments for selecting the optimal imaging options for future research and patient management.
Collapse
Affiliation(s)
- Massis Krekorian
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Timothy H Witney
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Erik H J G Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Castiello L, Aricò E, D'Agostino G, Santodonato L, Belardelli F. In situ Vaccination by Direct Dendritic Cell Inoculation: The Coming of Age of an Old Idea? Front Immunol 2019; 10:2303. [PMID: 31611878 PMCID: PMC6773832 DOI: 10.3389/fimmu.2019.02303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
For more than 25 years, dendritic cell (DC) based vaccination has flashily held promises to represent a therapeutic approach for cancer treatment. While the vast majority of studies has focused on the use of antigen loaded DC, the intratumoral delivery of unloaded DC aiming at in situ vaccination has gained much less attention. Such approach grounds on the ability of inoculated DC to internalize and process antigens directly released by tumor (usually in combination with cell-death-inducing agents) to activate broad patient-specific antitumor T cell response. In this review, we highlight the recent studies in both solid and hematological tumors showing promising clinical results and discuss the main pitfalls and advantages of this approach for endogenous cancer vaccination. Lastly, we discuss how in situ vaccination by DC inoculation may fit with current immunotherapy approaches to expand and prolong patient response.
Collapse
Affiliation(s)
- Luciano Castiello
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Aricò
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Laura Santodonato
- FaBioCell, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Filippo Belardelli
- Consiglio Nazionale Delle Ricerche, Institute of Translational Pharmacology, Rome, Italy
| |
Collapse
|
11
|
Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol 2019; 9:3176. [PMID: 30719026 PMCID: PMC6348254 DOI: 10.3389/fimmu.2018.03176] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells, uniquely able to induce naïve T cell activation and effector differentiation. They are, likewise, involved in the induction and maintenance of immune tolerance in homeostatic conditions. Their phenotypic and functional heterogeneity points to their great plasticity and ability to modulate, according to their microenvironment, the acquired immune response and, at the same time, makes their precise classification complex and frequently subject to reviews and improvement. This review will present general aspects of the DC physiology and classification and will address their potential and actual uses in the management of human disease, more specifically cancer, as therapeutic and monitoring tools. New combination treatments with the participation of DC will be also discussed.
Collapse
Affiliation(s)
- Thiago A Patente
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana P Pinho
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline A Oliveira
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela C M Evangelista
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia C Bergami-Santos
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José A M Barbuto
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Discipline of Molecular Medicine, Department of Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Raaijmakers TK, Ansems M. Microenvironmental derived factors modulating dendritic cell function and vaccine efficacy: the effect of prostanoid receptor and nuclear receptor ligands. Cancer Immunol Immunother 2018; 67:1789-1796. [PMID: 29998375 PMCID: PMC6208817 DOI: 10.1007/s00262-018-2205-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/09/2018] [Indexed: 01/20/2023]
Abstract
Dendritic cells (DCs) are widely used in DC-based immunotherapies because of their capacity to steer immune responses. So far treatment success is limited and more functional knowledge on how DCs initiate and stably drive specific responses is needed. Many intrinsic and extrinsic factors contribute to how DCs skew the immune response towards immunity or tolerance. The origin and type of DC, its maturation status, but also factors they encounter in the in vitro or in vivo microenvironment they reside in during differentiation and maturation affect this balance. Treatment success of DC vaccines will, therefore, also depend on the presence of these factors during the process of vaccination. Identification and further knowledge of natural and pharmacological compounds that modulate DC differentiation and function towards a specific response may help to improve current DC-based immunotherapies. This review focuses on factors that could improve the efficacy of DC vaccines in (pre-)clinical studies to enhance DC-based immunotherapy, with a particular emphasis on compounds acting on prostanoid or nuclear receptor families.
Collapse
Affiliation(s)
- Tonke K Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
13
|
Nico D, Martins Almeida F, Maria Motta J, Soares dos Santos Cardoso F, Freire-de-Lima CG, Freire-de-Lima L, de Luca PM, Maria Blanco Martinez A, Morrot A, Palatnik-de-Sousa CB. NH36 and F3 Antigen-Primed Dendritic Cells Show Preserved Migrating Capabilities and CCR7 Expression and F3 Is Effective in Immunotherapy of Visceral Leishmaniasis. Front Immunol 2018; 9:967. [PMID: 29867949 PMCID: PMC5949526 DOI: 10.3389/fimmu.2018.00967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 04/18/2018] [Indexed: 01/23/2023] Open
Abstract
Physical contact between dendritic cells (DCs) and T cell lymphocytes is necessary to trigger the immune cell response. CCL19 and CCL21 chemokines bind to the CCR7 receptor of mature DCs, and of T cells and regulate DCs migration to the white pulp (wp) of the spleen, where they encounter lymphocytes. In visceral leishmaniasis (VL), cellular immunosuppression is mediated by impaired DC migration due to the decreased chemokine secretion by endothelium and to the reduced DCs CCR7 expression. The Leishmania (L.) donovani nucleoside hydrolase NH36 and its C-terminal domain, the F3 peptide are prominent antigens in the generation of preventive immunity to VL. We assessed whether these vaccines could prevent the migrating defect of DCs by restoring the expression of CCR7 receptors. C57Bl6 mice were vaccinated with NH36 and F3 and challenged with L. (L.) infantum chagasi. The F3 vaccine induced a 100% of survival and a long-lasting immune protection with an earlier CD4+Th1 response, with secretion of higher IFN-γ and TNF-α/IL-10 ratios, and higher frequencies of CD4+ T cells secreting IL-2+, TNF-α+, or IFN-γ+, or a combination of two or the three cytokines (IL-2+TNF-α+IFN-γ+). The CD8+ T cell response was promoted earlier by the NH36-vaccine, and later by the F3-vaccine. Maximal number of F3-primed DCs migrated in vitro in response to CCL19 and showed a high expression of CCR7 receptors (26.06%). Anti-CCR7 antibody treatment inhibited DCs migration in vitro (90%) and increased parasite load in vivo. When transferred into 28-day-infected mice, only 8% of DCs from infected, 59% of DCs from NH36-vaccinated, and 84% of DCs from F3-vaccinated mice migrated to the wp. Consequently, immunotherapy of infected mice with F3-primed DCs only, promoted increases in corporal weight and reductions of spleen and liver parasite loads and relative weights. Our findings indicate that vaccination with F3-vaccine preserves the maturation, migration properties and CCR7 expression of DCs, which are essential processes for the generation of cell-mediated immunity. The F3 vaccine is more potent in reversing the migration defect that occurs in VL and, therefore, more efficient in immunotherapy of VL.
Collapse
Affiliation(s)
- Dirlei Nico
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Martins Almeida
- Programa de Pós Graduação em Anatomia Patológica, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Graduação de Histologia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Maria Motta
- Programa de Glicobiologia, Instituto de Bioquímica Médica Leopoldo De Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Celio Geraldo Freire-de-Lima
- Programa de Imunobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Freire-de-Lima
- Programa de Medicina Regenerativa, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Melo de Luca
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Rio de Janeiro, Brazil
| | - Ana Maria Blanco Martinez
- Programa de Pós Graduação em Anatomia Patológica, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Rio de Janeiro, Brazil
- Centro de Pesquisas em Tuberculose, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarisa Beatriz Palatnik-de-Sousa
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia, São Paulo, Brazil
| |
Collapse
|
14
|
Malvicini M, Gutierrez-Moraga A, Rodriguez MM, Gomez-Bustillo S, Salazar L, Sunkel C, Nozal L, Salgado A, Hidalgo M, Lopez-Casas PP, Novella JL, Vaquero JJ, Alvarez-Builla J, Mora A, Gidekel M, Mazzolini G. A Tricin Derivative from Deschampsia antarctica Desv. Inhibits Colorectal Carcinoma Growth and Liver Metastasis through the Induction of a Specific Immune Response. Mol Cancer Ther 2018; 17:966-976. [DOI: 10.1158/1535-7163.mct-17-0193] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/31/2017] [Accepted: 02/09/2018] [Indexed: 11/16/2022]
|
15
|
Zhang X, Zhang Y, Xu J, Wang H, Zheng X, Lou Y, Han B. Antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B lymphocytes enhances the killing effect of cytotoxic T lymphocytes on tumor stem-like cells derived from cisplatin-resistant lung cancer cells. J Cancer 2018; 9:367-374. [PMID: 29344283 PMCID: PMC5771344 DOI: 10.7150/jca.20821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022] Open
Abstract
The present study investigated whether antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B lymphocytes can enhance the killing effect of CD8+ cytotoxic T lymphocytes (CTLs) on lung stem-like cancer cells (SLCCs). The CTLs were generated using an accelerated co-cultured dendritic cells (DC) (acDC) assay by incubating human peripheral blood mononuclear cells (PBMCs) from non-small-cell lung cancer patients with antigen peptides of Oct4 and Sox2 in the presence of several DC-activating agents. CD154+ NIH3T3 cells prepared by CD154 lentiviral transfection were used as feeder layer to activate primary B cells (CD19+) obtained from PBMCs. Activated B cells were co-cultured with CTLs to present antigen peptides of Oct4 and Sox2. CTLs co-cultured with activated B cells were evaluated for the levels of secreted inflammatory cytokines using ELISA. In addition, the killing effect of the CTLs on SLCCs derived from cisplatin-resistant strain of human lung cancer cell line PC9 was evaluated by flow cytometry using CFSE labeling of the target cells. After the acDC assay, the PBMCs exhibited a significant (p<0.01) increase in the population of CD8+/CD3+ cells, indicating successful preparation of CTLs. The primary B cells cultured on the CD154+ NIH3T3 feeder layer resulted in significant (p<0.01) increase in the proportions of population expressing CD80, CD86, or HLA-A, indicating successful activation of the B cells. The co-culture of CTLs with CD154-activated B cells presenting the Oct4 and Sox2 peptides caused significant increase in the levels of secretory inflammatory cytokines and exhibited enhanced killing of the SLCCs derived from cisplatin-resistant PC9 cells. Antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B cells can enhance the killing effect of CTLs towards lung SLCCs.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Yanwei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Jianlin Xu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Huimin Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Xiaoxuan Zheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Yuqing Lou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| |
Collapse
|
16
|
Yuan H, Jiang W, von Roemeling CA, Qie Y, Liu X, Chen Y, Wang Y, Wharen RE, Yun K, Bu G, Knutson KL, Kim BYS. Multivalent bi-specific nanobioconjugate engager for targeted cancer immunotherapy. NATURE NANOTECHNOLOGY 2017; 12:763-769. [PMID: 28459470 DOI: 10.1038/nnano.2017.69] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 03/15/2017] [Indexed: 05/17/2023]
Abstract
Tumour-targeted immunotherapy offers the unique advantage of specific tumouricidal effects with reduced immune-associated toxicity. However, existing platforms suffer from low potency, inability to generate long-term immune memory and decreased activities against tumour-cell subpopulations with low targeting receptor levels. Here we adopted a modular design approach that uses colloidal nanoparticles as substrates to create a multivalent bi-specific nanobioconjugate engager (mBiNE) to promote selective, immune-mediated eradication of cancer cells. By simultaneously targeting the human epidermal growth factor receptor 2 (HER2) expressed by cancer cells and pro-phagocytosis signalling mediated by calreticulin, the mBiNE stimulated HER2-targeted phagocytosis and produced durable antitumour immune responses against HER2-expressing tumours. Interestingly, although the initial immune activation mediated by the mBiNE was receptor dependent, the subsequent antitumour immunity also generated protective effects against tumour-cell populations that lacked the HER2 receptor. Thus, the mBiNE represents a new targeted, nanomaterial-immunotherapy platform to stimulate innate and adaptive immunity and promote a universal antitumour response.
Collapse
Affiliation(s)
- Hengfeng Yuan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Xuhui, Shanghai, China
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Christina A von Roemeling
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
- Mayo Graduate School, Mayo Clinic College of Medicine, 200 1st Street, Rochester, Minnesota 55902, USA
| | - Yaqing Qie
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Xiujie Liu
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Yuanxin Chen
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Yifan Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Robert E Wharen
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Kyuson Yun
- Department of Neurosurgery, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Keith L Knutson
- Department of Cancer Biology, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
- Department of Immunology, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Betty Y S Kim
- Department of Neurosurgery, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
- Department of Cancer Biology, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| |
Collapse
|
17
|
Huang E, Showalter L, Xu S, Czernliecki BJ, Koski GK. Calcium mobilizing treatment acts as a co-signal for TLR-mediated induction of Interleukin-12 (IL-12p70) secretion by murine bone marrow-derived dendritic cells. Cell Immunol 2017; 314:26-35. [PMID: 28190517 DOI: 10.1016/j.cellimm.2017.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/11/2017] [Accepted: 01/26/2017] [Indexed: 11/28/2022]
Abstract
We sought to determine whether pharmacological calcium-mobilizing agents could act in cooperation with Toll-like receptor (TLR) signals to induce high-level IL-12 production from murine bone marrow-derived dendritic cells. We found that calcium mobilization alone induced no IL-12, yet dramatically enhanced IL-12p70 secretion elicited by TLR ligands. Enhanced IL-12 production induced by calcium ionophore plus single TLR ligands, but not through dual TLR ligands, was inhibited by the calcineurin antagonist cyclosporine A, suggesting divergent mechanisms of IL-12 induction. Dendritic cells activated with calciumionophore plus the TLR9 ligand ODN1826 could induce Th1 polarization in naïve murine CD4pos T cells at levels equal or superior to dendritic cells activated with the most efficient TLR ligand pairing; ODN1826 plus bacterial lipopolysaccharide. Parallel analysis of 38 inflammation-associated soluble products showed calciumionophore enhancement was restricted to a small set of factors. These data demonstrate previously undocumented activation co-signals for IL-12 production by dendritic cells.
Collapse
Affiliation(s)
- Emily Huang
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States
| | - Loral Showalter
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States
| | - Shuwen Xu
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States
| | - Brian J Czernliecki
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States
| | - Gary K Koski
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States.
| |
Collapse
|
18
|
Castiello L, Sabatino M, Ren J, Terabe M, Khuu H, Wood LV, Berzofsky JA, Stroncek DF. Expression of CD14, IL10, and Tolerogenic Signature in Dendritic Cells Inversely Correlate with Clinical and Immunologic Response to TARP Vaccination in Prostate Cancer Patients. Clin Cancer Res 2017; 23:3352-3364. [PMID: 28073842 DOI: 10.1158/1078-0432.ccr-16-2199] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/05/2016] [Accepted: 12/20/2016] [Indexed: 12/25/2022]
Abstract
Purpose: Despite the vast number of clinical trials conducted so far, dendritic cell (DC)-based cancer vaccines have mostly shown unsatisfactory results. Factors and manufacturing procedures essential for these therapeutics to induce effective antitumor immune responses have yet to be fully characterized. We here aimed to identify DC markers correlating with clinical and immunologic response in a prostate carcinoma vaccination regimen.Experimental Design: We performed an extensive characterization of DCs used to vaccinate 18 patients with prostate carcinoma enrolled in a pilot trial of T-cell receptor gamma alternate reading frame protein (TARP) peptide vaccination (NCT00908258). Peptide-pulsed DC preparations (114) manufactured were analyzed by gene expression profiling, cell surface marker expression and cytokine release secretion, and correlated with clinical and immunologic responses.Results: DCs showing lower expression of tolerogenic gene signature induced strong antigen-specific immune response and slowing in PSA velocity, a surrogate for clinical response. These DCs were also characterized by lower surface expression of CD14, secretion of IL10 and MCP-1, and greater secretion of MDC. When combined, these four factors were able to remarkably discriminate DCs that were sufficiently potent to induce strong immunologic response.Conclusions: DC factors essential for the activation of immune responses associated with TARP vaccination in prostate cancer patients were identified. This study highlights the importance of in-depth characterization of DC vaccines and other cellular therapies, to understand the critical factors that hinder potency and potential efficacy in patients. Clin Cancer Res; 23(13); 3352-64. ©2017 AACR.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland.
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marianna Sabatino
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Jiaqiang Ren
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Hanh Khuu
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Lauren V Wood
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - David F Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| |
Collapse
|
19
|
Le PM, Tran TTB, Vu BT, Van Pham P. A preliminary comparison of dendritic cell maturation by total cellular RNA to total cellular lysate derived from breast cancer stem cells. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0028-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
20
|
Cabrales P, Scicinski J, Reid T, Kuypers F, Larkin S, Fens M, Oronsky A, Oronsky B. A look inside the mechanistic black box: Are red blood cells the critical effectors of RRx-001 cytotoxicity? Med Oncol 2016; 33:63. [PMID: 27229330 DOI: 10.1007/s12032-016-0775-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/13/2016] [Indexed: 12/18/2022]
Abstract
The therapeutic potential of epi-immunotherapeutic anticancer agent RRx-001 in cancer has been validated with preclinical and clinical studies, since RRx-001 has successfully completed a phase 1 trial and multiple single-agent and combination phase 2 trials with preliminary evidence of promising activity are underway. Previous experimental work has implicated diverse anticancer mechanisms such as oxidative stress, ATP and NADPH depletion, anti-angiogenesis and epigenetic modulation in the overall antitumor effect of RRx-001. The hypothesis of this study was that the RRx-001 red blood cells are the essential and de facto intermediaries responsible for the reprograming of tumor behavior via transfer of their intracellular and membrane contents. To test this hypothesis, and thereby resolve the "black box" incompleteness in the continuity of the mechanism, the fate of red blood cells incubated with RRx-001 was explored in vitro and in vivo both in healthy animals and in tumor-bearing mice. The collective results establish that RRx-001-derivatized red blood cells are the critical "missing links" to explain the specificity and anticancer activity of RRx-001, including its immunomodulatory effects on tumor-associated macrophages. These experimental results delineate a novel erythrocyte-based mechanism without precedent in the annals of oncology and open the door to rational combination strategies with RRx-001 both in cancer therapy and beyond, particularly in disease states that affect red blood cell and vascular function such as malaria, leishmaniasis, sickle-cell disease and hemorrhagic shock.
Collapse
Affiliation(s)
- Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jan Scicinski
- EpicentRx, Inc., 800 W El Camino Real, Suite 180, Mountain View, CA, 94040, USA
| | - Tony Reid
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Frans Kuypers
- Children's Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| | - Sandra Larkin
- Children's Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| | - Marcel Fens
- Children's Hospital Oakland Research Institute, Oakland, CA, 94609, USA
| | | | - Bryan Oronsky
- EpicentRx, Inc., 800 W El Camino Real, Suite 180, Mountain View, CA, 94040, USA.
| |
Collapse
|
21
|
Natarajan G, Oghumu S, Terrazas C, Varikuti S, Byrd JC, Satoskar AR. A Tec kinase BTK inhibitor ibrutinib promotes maturation and activation of dendritic cells. Oncoimmunology 2016; 5:e1151592. [PMID: 27471620 PMCID: PMC4938315 DOI: 10.1080/2162402x.2016.1151592] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/31/2016] [Accepted: 02/01/2016] [Indexed: 11/24/2022] Open
Abstract
Ibrutinib, a BTK inhibitor, is currently used to treat various hematological malignancies. We evaluated whether ibrutinib treatment during development of murine bone marrow-derived dendritic cells (DCs) modulates their maturation and activation. Ibrutinib treatment increased the proportion of CD11c+ DCs, upregulated the expression of MHC-II and CD80 and downregulated Ly6C expression by DCs. Additionally, ibrutinib treatment led to an increase in MHC-II+, CD80+ and CCR7+ DCs but a decrease in CD86+ DCs upon LPS stimulation. LPS/ibrutinib-treated DCs displayed increased IFNβ and IL-10 synthesis and decreased IL-6, IL-12 and NO production compared to DCs stimulated with LPS alone. Finally, LPS/ibrutinib-treated DCs promoted higher rates of CD4+ T cell proliferation and cytokine production compared to LPS only stimulated DCs. Taken together, our results indicate that ibrutinib enhances the maturation and activation of DCs to promote CD4+ T cell activation which could be exploited for the development of DC-based cancer therapies.
Collapse
Affiliation(s)
- Gayathri Natarajan
- Department of Microbiology, The Ohio State University , Columbus, OH, USA
| | - Steve Oghumu
- Department of Environmental Health Sciences, College of Public Health, The Ohio State University , Columbus, OH, USA
| | - Cesar Terrazas
- Department of Pathology, College of Medicine, The Ohio State University , Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, College of Medicine, The Ohio State University , Columbus, OH, USA
| | - John C Byrd
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA; Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Abhay R Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH, USA; Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
22
|
Behm B, Di Fazio P, Michl P, Neureiter D, Kemmerling R, Hahn EG, Strobel D, Gress T, Schuppan D, Wissniowski TT. Additive antitumour response to the rabbit VX2 hepatoma by combined radio frequency ablation and toll like receptor 9 stimulation. Gut 2016; 65:134-143. [PMID: 25524262 DOI: 10.1136/gutjnl-2014-308286] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Radiofrequency ablation (RFA), a palliative therapeutic option for solid hepatic tumours, stimulates localised and systemic antitumour cytotoxic T cells. We studied how far addition of CpG B oligonucleotides, toll like receptor (TLR) 9 agonists, would increase the antitumoural T cell response of RFA in the highly aggressive VX2 hepatoma. METHODS Rabbits were randomised to receive RFA, CpG B, their combination or no therapy. The antitumour efficacy of RFA alone or in combination with CpG B was further tested by rechallenging a separate group with intravenously injected VX2 tumour cells after 120 days. Animals were assessed for survival, tumour size and spread, and tumour and immune related histological markers after 120 days. Peripheral blood mononuclear cells were tested for tumour-specific T cell activation and cytotoxicity. Immune modulatory cytokines tumour necrosis factor α, interleukin (IL)-2/IL-8/IL-10/IL-12 and interferon γ, and vascular endothelial growth factor were measured in serum. RESULTS Mean survival of untreated animals was 36 days, as compared with 97, 78 and 114 days for RFA, CpG and combination therapy, respectively. Compared with untreated controls, antitumour T cell stimulation/cytotoxicity increased 26/16-fold, 32/17-fold and 50/38-fold 2 weeks after RFA, CpG and combination treatments, respectively. The combination inhibited tumour spread to lungs and peritoneum significantly and prohibited new tumour growth in animals receiving a secondary systemic tumour cell injection. RFA alone induced a Th1 cytokine pattern, while IL-8 and IL-10 were only upregulated in CpG treated animals and controls. CONCLUSIONS The combination of TLR9 stimulation with RFA resulted in a potentiated antitumour T cell response and cytotoxicity in the VX2 tumour model. Only this combination prevented subsequent tumour spread and resulted in a significantly improved survival, justifying the need for further exploration of the combination of ablative therapies and TLR9 agonists in liver cancer.
Collapse
Affiliation(s)
- Barbara Behm
- Department of Medicine 1, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Pietro Di Fazio
- Institute for Surgical Research, Philipps-University Marburg, Marburg, Germany
| | - Patrick Michl
- Division of Gastroenterology, University Hospital, Philipps-University Marburg, Marburg, Germany
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University, Salzburg, Austria
| | - Ralf Kemmerling
- Institute of Pathology, Paracelsus Medical University, Salzburg, Austria
| | - Eckhart Georg Hahn
- Department of Medicine 1, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Deike Strobel
- Department of Medicine 1, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Gress
- Division of Gastroenterology, University Hospital, Philipps-University Marburg, Marburg, Germany
| | - Detlef Schuppan
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA Institute of Translational Immunology, University Medical Center, Mainz, Germany
| | - Thaddaeus Till Wissniowski
- Department of Medicine 1, University Hospital Erlangen-Nuremberg, Erlangen, Germany Division of Gastroenterology, University Hospital, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
23
|
Wojas-Turek J, Szczygieł A, Kicielińska J, Rossowska J, Piasecki E, Pajtasz-Piasecka E. Treatment with cyclophosphamide supported by various dendritic cell-based vaccines induces diversification in CD4⁺ T cell response against MC38 colon carcinoma. Int J Oncol 2015; 48:493-505. [PMID: 26648160 PMCID: PMC4725454 DOI: 10.3892/ijo.2015.3278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/26/2015] [Indexed: 12/18/2022] Open
Abstract
The present study shows that an application of cyclophosphamide (CY) supported by dendritic cell (DC)-based vaccines affected differentiation of the activity of CD4+ T cell subpopulations accompanied by an alteration in CD8+ cell number. Vaccines were composed of bone marrow-derived DCs activated with tumor cell lysate (BM-DC/TAgTNF-α) and/or genetically modified DCs of JAWS II line (JAWS II/ Neo or JAWS II/IL-2 cells). Compared to untreated or CY-treated mice, the combined treatment of MC38 colon carcinoma-bearing mice resulted in significant tumor growth inhibition associated with an increase in influx of CD4+ and CD8+ T cells into tumor tissue. Whereas, the division of these cell population in spleen was not observed. Depending on the nature of DC-based vaccines and number of their applications, both tumor infiltrating cells and spleen cells were able to produce various amount of IFN-γ, IL-4 and IL-10 after mitogenic ex vivo stimulation. The administration of CY followed by BM-DC/TAgTNF-α and genetically modified JAWS II cells, increased the percentage of CD4+T-bet+ and CD4+GATA3+ cells and decreased the percentage of CD4+RORγt+ and CD4+FoxP3+ lymphocytes. However, the most intensive response against tumor was noted after the ternary treatment with CY + BM-DC/TAgTNF-α + JAWS II/IL-2 cells. Thus, the administration of various DC-based vaccines was responsible for generation of the diversified antitumor response. These findings demonstrate that the determination of the size of particular CD4+ T cell subpopulations may become a prognostic factor and be the basis for future development of anticancer therapy.
Collapse
Affiliation(s)
- Justyna Wojas-Turek
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Agnieszka Szczygieł
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Jagoda Kicielińska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Egbert Piasecki
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
24
|
Ahmed KK, Geary SM, Salem AK. Applying biodegradable particles to enhance cancer vaccine efficacy. Immunol Res 2015; 59:220-8. [PMID: 24838147 DOI: 10.1007/s12026-014-8537-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One of the primary goals of our group and our collaborators here at the University of Iowa is to develop therapeutic cancer vaccines using biodegradable and biocompatible polymer-based vectors. A major advantage of using discretely packaged immunogenic cargo over non-encapsulated vaccines is that they promote enhanced cellular immunity, a key requirement in achieving antitumor activity. We discuss the importance of co-encapsulation of tumor antigen and adjuvant, with specific focus on the synthetic oligonucleotide adjuvant, cytosine-phosphate-guanine oligodeoxynucleotides. We also discuss our research using a variety of polymers including poly(α-hydroxy acids) and polyanhydrides, with the aim of determining the effect that parameters, such as size and polymer type, can have on prophylactic and therapeutic tumor vaccine formulation efficacy. Aside from their role as vaccine vectors per se, we also address the research currently underway in our group that utilizes more novel applications of biodegradable polymer-based particles in facilitating other types of immune-based therapies.
Collapse
Affiliation(s)
- Kawther K Ahmed
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | | | | |
Collapse
|
25
|
InCVAX--a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett 2015; 359:169-77. [PMID: 25633839 DOI: 10.1016/j.canlet.2015.01.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
Abstract
A novel, promising potential cancer vaccine strategy was proposed to use a two-injection procedure for solid tumors to prompt the immune system to identify and systemically eliminate primary and metastatic cancers. The two-injection procedure consists of local photothermal application on a selected tumor intended to liberate whole cell tumor antigens, followed by a local injection of an immunoadjuvant that consists of a semi-synthetic functionalized glucosamine polymer, N-dihydro-galacto-chitosan (GC), which is intended to activate antigen presenting cells and facilitate an increased uptake of tumor antigens. This strategy is thus proposed as an in situ autologous cancer vaccine (inCVAX) that may activate antigen presenting cells and expose them to tumor antigens in situ, with the intention of inducing a systemic tumor specific T-cell response. Here, the development of inCVAX for the treatment of metastatic cancers in the past decades is systematically reviewed. The antitumor immune responses of local photothermal treatment and immunological stimulation with GC are also discussed. This treatment approach is also commonly referred to as laser immunotherapy (LIT).
Collapse
|
26
|
Li RL, Zhou S, Qin J, Liang CM, Luo GR. Effect of administration of BMDC vaccine sensitized by heat shocked hepal-6 cell proteins on intratumoral CD25 +Foxp3 + Tregs in mouse hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2014; 22:2081-2090. [DOI: 10.11569/wcjd.v22.i15.2081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine whether the bone marrow derived dendritic cell (BMDC) vaccine sensitized by heat shocked hepal-6 cell proteins affects the infiltration of intratumoral CD25+Foxp3+ Tregs in a mouse hepatocellular carcinoma (HCC) model.
METHODS: In the presence of GM-CSF and IL-4, BMDCs were induced in vitro. BMDCs were sensitized by heat shocked hepal-6 cell proteins to generate a vaccine for HCC. The expression of CD11c, CCR7, CD80 and CD86 on these sensitized BMDCs were analyzed by FACS. The anti-tumor effect of this vaccine was evaluated using a mouse HCC model established by subcutaneous injection of Hepal-6 cells. Eight days later, the tumor-bearing mice were divided into four groups, which underwent intratumoral injection of BMDCs sensitized by heat shocked hepal-6 cell proteins, serum-free culture medium, BMDCs without sensitization and BMDCs sensitized by unheated hepal-6 cell proteins (once every 7 d, 2 times altogether), respectively. Nine days after final administration, the mice were sacrificed and the tumor samples were taken for immunofluorescence staining for CD8+ cells and intratumoral CD25+Foxp3+ Tregs.
RESULTS: Light microscopy and scanning electron microscopy showed that BMDCs propagated in the presence of GM-CSF and IL-4 displayed the typical morphological characteristics of dendritic cells. Immunocytochemical staining showed that they expressed the dendritic cell marks including CD11c, CCR7, CD80 and CD86. Compared with the controls (BMDCs without sensitization or sensitized by unheated hepal-6 cells proteins), the BMDCs sensitized by heat shocked hepal-6 cells proteins showed increased expression of CD11c (67.2 ± 4.49 vs 52.4 ± 5.20, 58.4 ± 4.43), CCR7 (65.4 ± 5.34 vs 45.9 ± 5.04, 57.0 ± 3.46), CD80 (62.9 ± 4.69 vs 46.9 ± 4.75, 54.4 ± 3.47) and CD86 (73.3 ± 3.58 vs 60.1 ± 2.98, 63.7 ± 3.10) (P < 0.01 for all). Compared with the controls, the mice administrated with the BMDC vaccine sensitized by heat shocked Hepal-6 cell proteins showed increased CD8+ T cells (55.0 ± 4.11 vs 38.2 ± 3.34, 44.6 ± 4.29, 45.6 ± 4.92, P < 0.01 for all) and decreased intratumoral CD25+Foxp3+ Tregs (0.37 ± 0.028 vs 1.31 ± 0.020, 0.77 ± 0.057, 0.57 ± 0.062, P < 0.05 for all).
CONCLUSION: Heat shocked hepal-6 cell protein sensitization can upregulate the expression of CD11c, CCR7, CD80 and CD86 on BMDCs in vitro. Administration with this BMDC vaccine can increase CD8+ T cells and decrease intratumoral CD25+Foxp3+ Tregs in HCC mice.
Collapse
|
27
|
Tseng JY, Yang CY, Liang SC, Liu RS, Yang SH, Lin JK, Chen YM, Wu YC, Jiang JK, Lin CH. Interleukin-17A modulates circulating tumor cells in tumor draining vein of colorectal cancers and affects metastases. Clin Cancer Res 2014; 20:2885-97. [PMID: 24677375 DOI: 10.1158/1078-0432.ccr-13-2162] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Metastasis is the major cause of death in patients with colorectal cancer (CRC). Circulating tumor cells (CTC) are believed to cause metastasis and serve as a prognostic marker for mortality in clinical stage IV patients. However, most studies are conducted in late-stage cases when distant metastases have already occurred; thus, such results provide limited clinical use. This study focused on whether CTCs can predict the risk of metastasis after treatment of the primary tumor in early-stage patients with CRC. EXPERIMENTAL DESIGN CTCs were quantified using EpCAM-positive/CD45-negative immunoselection and flow cytometry in patients with CRC. A mouse model was used to investigate the mechanistic roles of CTCs and interleukin (IL)-17A in metastasis. RESULTS The number of mesenteric CTCs obtained from stage II patients was higher than that obtained from patients in stages I, III, and IV. In addition, following invasion of orthotopically implanted tumors in our mouse model, we found that CTCs exhibited an increase-then-decrease pattern, accompanied by corresponding changes in serum IL-17A levels and opposing changes in serum granulocyte macrophage colony-stimulating factor (GM-CSF) levels. Ablation of IL-17A and administration of rGM-CSF effectively suppressed the increase in CTCs and prevented metastasis in mice. Moreover, IL-17A promoted cancer cell motility, matrix digestion, and angiogenesis, whereas GM-CSF stimulated the elimination of CTCs by boosting host immunity. Notably, serum levels of IL-17A were also correlated with disease-free survival in patients with CRC. CONCLUSIONS Our results showed that CTCs and IL-17A could serve as prognostic markers and therapeutic targets for CRC metastasis. Clin Cancer Res; 20(11); 2885-97. ©2014 AACR.
Collapse
Affiliation(s)
- Ju-Yu Tseng
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Chih-Yung Yang
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Shu-Ching Liang
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Ren-Shyan Liu
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Shung-Haur Yang
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Jen-Kou Lin
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Yuh-Min Chen
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Yu-Chung Wu
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Jeng-Kai Jiang
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| | - Chi-Hung Lin
- Authors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, TaiwanAuthors' Affiliations: Institute of Microbiology and Immunology, School of Life Science; Department of Biomedical Imaging and Radiological Sciences; School of Medicine, National Yang-Ming University; Department of Education and Research, Taipei City Hospital; Department of Nuclear Medicine; Division of Colorectal Surgery, Department of Surgery; Section of Thoracic Oncology, Chest Department; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital; and VGH Yang-Ming Genome Research Center, Taipei, Taiwan
| |
Collapse
|
28
|
Cruz LJ, Rueda F, Simón L, Cordobilla B, Albericio F, Domingo JC. Liposomes containing NY-ESO-1/tetanus toxoid and adjuvant peptides targeted to human dendritic cells via the Fc receptor for cancer vaccines. Nanomedicine (Lond) 2014; 9:435-49. [DOI: 10.2217/nnm.13.66] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aim: To improve the immunological response against tumors, a vaccine based on nanoliposomes targeted to the Fcγ-receptor was developed to enhance the immunogenicity of tumor-associated antigens (TAAs). Materials & methods: Using human dendritic cells in vitro, a fragment of the TAA NY-ESO-1 combined with a T-helper peptide from the tetanus toxoid encapsulated in nanoliposomes was evaluated. In addition, peptides Palm-IL-1 and MAP-IFN-γwere coadministered as adjuvants to enhance the immunological response. Results: Coadministration of Palm-IL-1 or MAP-IFN-γpeptide adjuvants and the hybrid NY-ESO-1-tetanus toxoid (soluble or encapsulated in nanoliposomes without targeting) increased immunogenicity. However, the most potent immunological response was obtained when the peptide adjuvants were encapsulated in liposomes targeted to human dendritic cells via the Fc receptor. Conclusion: This targeted vaccine strategy is a promising tool to activate and deliver antigens to dendritic cells, thus improving immunotherapeutic response in situations in which the immune system is frequently compromised, as in advanced cancers.
Collapse
Affiliation(s)
- Luis J Cruz
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials & Nanomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Felix Rueda
- Department of Biochemistry & Molecular Biology, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Lorena Simón
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials & Nanomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Begoña Cordobilla
- Department of Biochemistry & Molecular Biology, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Fernando Albericio
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials & Nanomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona Science Park, Baldiri Reixac 10, 08028 Barcelona, Spain
- Department of Organic Chemistry, University of Barcelona, Marti i Franques 1, 08028-Barcelona, Spain
- School of Chemistry, University of KwaZulu Natal, Durban, Kwa-Zulu Natal, 4000, South Africa
| | - Joan C Domingo
- Department of Biochemistry & Molecular Biology, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
| |
Collapse
|
29
|
Spadaro M, Montone M, Arigoni M, Cantarella D, Forni G, Pericle F, Pascolo S, Calogero RA, Cavallo F. Recombinant human lactoferrin induces human and mouse dendritic cell maturation via Toll-like receptors 2 and 4. FASEB J 2013; 28:416-29. [PMID: 24088817 DOI: 10.1096/fj.13-229591] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lactoferrin, a key component of innate immunity, is a cationic monomeric 80-kDa glycoprotein of the transferrin superfamily. Recombinant human lactoferrin, known as talactoferrin (TLF), induces a distinct functional maturation program in human dendritic cells (DCs) derived from peripheral blood monocytes. However, the receptors and molecular mechanisms involved in this induction have not been fully determined. By exploiting genome-wide transcription profiling of immature DCs, TNF-α- and IL-1β-matured DCs (m-DCs), and TLF-matured DCs (TLF-DCs), we have detected a set of transcripts specific for m-DCs and one specific for TLF-DCs. Functional network reconstruction highlighted, as expected, the association of m-DC maturation with IL-1β, TNF-α, and NF-κB, whereas TLF-DC maturation was associated with ERK and NF-κB. This involvement of ERK and NF-κB transduction factors suggests direct involvement of Toll-like receptors (TLRs) in TLF-induced maturation. We have used MyD88 inhibition and siRNA silencing TLRs on human DCs and mouse TLR-2-knockout cells, to show that TLF triggers the maturation of both human and mouse DCs through TLR-2 and TLR-4.
Collapse
Affiliation(s)
- Michela Spadaro
- 1Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, Torino 10126, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Xu Z, Ramishetti S, Tseng YC, Guo S, Wang Y, Huang L. Multifunctional nanoparticles co-delivering Trp2 peptide and CpG adjuvant induce potent cytotoxic T-lymphocyte response against melanoma and its lung metastasis. J Control Release 2013; 172:259-265. [PMID: 24004885 DOI: 10.1016/j.jconrel.2013.08.021] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 08/14/2013] [Accepted: 08/19/2013] [Indexed: 11/19/2022]
Abstract
Immunotherapy has shown the potential to become an essential component of the successful treatment of various malignancies. In many cases, such as in melanoma, however, induction of a potent and specific T-cell response against the endogenous antigen or self-antigen still remains a major challenge. To induce a potent MHC I-restricted cytotoxic T-lymphocyte (CTL) response, cytosol delivery of an exogenous antigen into dendritic cells is preferred, if not required. Lipid-calcium-phosphate (LCP) nanoparticles represent a new class of intracellular delivery systems for impermeable drugs. We are interested in exploring the potential of LCP NPs for use as a peptide vaccine delivery system for cancer therapy. To increase the encapsulation of Trp2 peptide into the calcium phosphate precipitate core of LCP, two phosphor-serine residues were added to the N-terminal of the peptide (p-Trp2). CpG ODN was also co-encapsulated with p-Trp2 as an adjuvant. The NPs were further modified with mannose to enhance and prolong the cargo deposit into the lymph nodes (LNs), which ensured persistent antigen loading and stimulation. Compared with free Trp2 peptide/CpG, vaccination with LCP encapsulating p-Trp2 and CpG resulted in superior inhibition of tumor growth in both B16F10 subcutaneous and lung metastasis models. An IFN-γ production assay and in vivo CTL response study revealed that the improved efficacy was a result of a Trp2-specific immune response. Thus, encapsulation of phospho-peptide antigens into LCP may be a promising strategy for enhancing the immunogenicity of poorly immunogenic self-antigens for cancer therapy.
Collapse
Affiliation(s)
- Zhenghong Xu
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Srinivas Ramishetti
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yu-Cheng Tseng
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shutao Guo
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuhua Wang
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Gigi V, Stein J, Askenasy N, Yaniv I, Ash S. Early immunisation with dendritic cells after allogeneic bone marrow transplantation elicits graft vs tumour reactivity. Br J Cancer 2013; 108:1092-9. [PMID: 23511628 PMCID: PMC3619065 DOI: 10.1038/bjc.2013.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background: Perspectives of immunotherapy to cancer mediated by bone marrow transplantation (BMT) in conjunction with dendritic cell (DC)-mediated immune sensitisation have yielded modest success so far. In this study, we assessed the impact of DC on graft vs tumour (GvT) reactions triggered by allogeneic BMT. Methods: H2Ka mice implanted with congenic subcutaneous Neuro-2a neuroblastoma (NB, H2Ka) tumours were irradiated and grafted with allogeneic H2Kb bone marrow cells (BMC) followed by immunisation with tumour-inexperienced or tumour-pulsed DC. Results: Immunisation with tumour-pulsed donor DC after allogeneic BMT suppressed tumour growth through induction of T cell-mediated NB cell lysis. Early post-transplant administration of DC was more effective than delayed immunisation, with similar efficacy of DC inoculated into the tumour and intravenously. In addition, tumour inexperienced DC were equally effective as tumour-pulsed DC in suppression of tumour growth. Immunisation of DC did not impact quantitative immune reconstitution, however, it enhanced T-cell maturation as evident from interferon-γ (IFN-γ) secretion, proliferation in response to mitogenic stimulation and tumour cell lysis in vitro. Dendritic cells potentiate GvT reactivity both through activation of T cells and specific sensitisation against tumour antigens. We found that during pulsing with tumour lysate DC also elaborate a factor that selectively inhibits lymphocyte proliferation, which is however abolished by humoral and DC-mediated lymphocyte activation. Conclusion: These data reveal complex involvement of antigen-presenting cells in GvT reactions, suggesting that the limited success in clinical application is not a result of limited efficacy but suboptimal implementation. Although DC can amplify soluble signals from NB lysates that inhibit lymphocyte proliferation, early administration of DC is a dominant factor in suppression of tumour growth.
Collapse
Affiliation(s)
- V Gigi
- Zaizov Cancer Immunotherapy Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | | | | | | | | |
Collapse
|
32
|
Dendritic cells: cellular mediators for immunological tolerance. Clin Dev Immunol 2013; 2013:972865. [PMID: 23762100 PMCID: PMC3671285 DOI: 10.1155/2013/972865] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/07/2013] [Indexed: 01/07/2023]
Abstract
In general, immunological tolerance is acquired upon treatment with non-specific immunosuppressive drugs. This indiscriminate immunosuppression of the patient often causes serious side-effects, such as opportunistic infectious diseases. Therefore, the need for antigen-specific modulation of pathogenic immune responses is of crucial importance in the treatment of inflammatory diseases. In this perspective, dendritic cells (DCs) can have an important immune-regulatory function, besides their notorious antigen-presenting capacity. DCs appear to be essential for both central and peripheral tolerance. In the thymus, DCs are involved in clonal deletion of autoreactive immature T cells by presenting self-antigens. Additionally, tolerance is achieved by their interactions with T cells in the periphery and subsequent induction of T cell anergy, T cell deletion, and induction of regulatory T cells (Treg). Various studies have described, modulation of DC characteristics with the purpose to induce antigen-specific tolerance in autoimmune diseases, graft-versus-host-disease (GVHD), and transplantations. Promising results in animal models have prompted researchers to initiate first-in-men clinical trials. The purpose of current review is to provide an overview of the role of DCs in the immunopathogenesis of autoimmunity, as well as recent concepts of dendritic cell-based therapeutic opportunities in autoimmune diseases.
Collapse
|
33
|
Hobo W, Novobrantseva TI, Fredrix H, Wong J, Milstein S, Epstein-Barash H, Liu J, Schaap N, van der Voort R, Dolstra H. Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother 2013; 62:285-97. [PMID: 22903385 PMCID: PMC11028421 DOI: 10.1007/s00262-012-1334-1] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
Abstract
Dendritic cell (DC)-based vaccination boosting antigen-specific immunity is being explored for the treatment of cancer and chronic viral infections. Although DC-based immunotherapy can induce immunological responses, its clinical benefit has been limited, indicating that further improvement of DC vaccine potency is essential. In this study, we explored the generation of a clinical-grade applicable DC vaccine with improved immunogenic potential by combining PD-1 ligand siRNA and target antigen mRNA delivery. We demonstrated that PD-L1 and PD-L2 siRNA delivery using DLin-KC2-DMA-containing lipid nanoparticles (LNP) mediated efficient and specific knockdown of PD-L expression on human monocyte-derived DC. The established siRNA-LNP transfection method did not affect DC phenotype or migratory capacity and resulted in acceptable DC viability. Furthermore, we showed that siRNA-LNP transfection can be successfully combined with both target antigen peptide loading and mRNA electroporation. Finally, we demonstrated that these PD-L-silenced DC loaded with antigen mRNA superiorly boost ex vivo antigen-specific CD8(+) T cell responses from transplanted cancer patients. Together, these findings indicate that our PD-L siRNA-LNP-modified DC are attractive cells for clinical-grade production and in vivo application to induce and boost immune responses not only in transplanted cancer patients, but likely also in other settings.
Collapse
Affiliation(s)
- Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University, Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 8, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Hanny Fredrix
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University, Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 8, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Jamie Wong
- Alnylam Pharmaceuticals, Inc., Cambridge, MA USA
| | | | | | - Ju Liu
- Alnylam Pharmaceuticals, Inc., Cambridge, MA USA
| | - Nicolaas Schaap
- Department of Hematology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Robbert van der Voort
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University, Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 8, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University, Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 8, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
34
|
Induction of antitumor immunity ex vivo using dendritic cells transduced with fowl pox vector expressing MUC1, CEA, and a triad of costimulatory molecules (rF-PANVAC). J Immunother 2013; 35:555-69. [PMID: 22892452 DOI: 10.1097/cji.0b013e31826a73de] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The fowl pox vector expressing the tumor-associated antigens, mucin-1 and carcinoembryonic antigen in the context of costimulatory molecules (rF-PANVAC) has shown promise as a tumor vaccine. However, vaccine-mediated expansion of suppressor T-cell populations may blunt clinical efficacy. We characterized the cellular immune response induced by ex vivo dendritic cells (DCs) transduced with (rF)-PANVAC. Consistent with the functional characteristics of potent antigen-presenting cells, rF-PANVAC-DCs demonstrated strong expression of mucin-1 and carcinoembryonic antigen and costimulatory molecules, CD80, CD86, and CD83; decreased levels of phosphorylated STAT3, and increased levels of Tyk2, Janus kinase 2, and STAT1. rF-PANVAC-DCs stimulated expansion of tumor antigen-specific T cells with potent cytolytic capacity. However, rF-PANVAC-transduced DCs also induced the concurrent expansion of FOXP3 expressing CD4CD25 regulatory T cells (Tregs) that inhibited T-cell activation. Moreover, Tregs expressed high levels of Th2 cytokines [interleukin (IL)-10, IL-4, IL-5, and IL-13] together with phosphorylated STAT3 and STAT6. In contrast, the vaccine-expanded Treg population expressed high levels of Th1 cytokines IL-2 and interferon-γ and the proinflammatory receptor-related orphan receptor γt (RORγt) and IL-17A suggesting that these cells may share effector functions with conventional TH17 T cells. These data suggest that Tregs expanded by rF-PANVAC-DCs, exhibit immunosuppressive properties potentially mediated by Th2 cytokines, but simultaneous expression of Th1 and Th17-associated factors suggests a high degree of plasticity.
Collapse
|
35
|
Delirezh N, Shojaeefar E, Parvin P, Asadi B. Comparison the effects of two monocyte isolation methods, plastic adherence and magnetic activated cell sorting methods, on phagocytic activity of generated dendritic cells. CELL JOURNAL 2013; 15:218-23. [PMID: 24027662 PMCID: PMC3769603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 01/15/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVE It is believed that monocyte isolation methods and maturation factors affect the phenotypic and functional characteristics of resultant dendritic cells (DC). In the present study, we compared two monocyte isolation methods, including plastic adherence-dendritic cells (Adh-DC) and magnetic activated cell sorting- dendritic cells (MACS-DC), and their effects on phagocytic activity of differentiated immature DCs (immDCs). MATERIALS AND METHODS : In this experimental study, immDCs were generated from plastic adherence and MACS isolated monocytes in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4) in five days. The phagocytic activity of immDCs was analyzed by fluorescein isothiocyanate (FITC)-conjugated latex bead using flow cytometry. One way ANOVA test was used for statistical analysis of differences among experimental groups, including Adh-DC and MACS-DC groups. RESULTS We found that phagocytic activity of Adh-DC was higher than MACS-DC, whereas the mean fluorescence intensity (MFI) of phagocytic cells was higher in MACS-DC (p<0.05). CONCLUSION : We concluded that it would be important to consider phagocytosis parameters of generated DCs before making any decision about monocyte isolation methods to have fully functional DCs.
Collapse
Affiliation(s)
- Nowruz Delirezh
- * Corresponding Address: P.O.Box: 57153-1177Department of MicrobiologyFaculty of Veterinary MedicineUrmia UniversityUrmiaIran
| | | | | | | |
Collapse
|
36
|
Castiello L, Sabatino M, Zhao Y, Tumaini B, Ren J, Ping J, Wang E, Wood LV, Marincola FM, Puri RK, Stroncek DF. Quality controls in cellular immunotherapies: rapid assessment of clinical grade dendritic cells by gene expression profiling. Mol Ther 2012; 21:476-84. [PMID: 23147403 DOI: 10.1038/mt.2012.89] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell-based immunotherapies are among the most promising approaches for developing effective and targeted immune response. However, their clinical usefulness and the evaluation of their efficacy rely heavily on complex quality control assessment. Therefore, rapid systematic methods are urgently needed for the in-depth characterization of relevant factors affecting newly developed cell product consistency and the identification of reliable markers for quality control. Using dendritic cells (DCs) as a model, we present a strategy to comprehensively characterize manufactured cellular products in order to define factors affecting their variability, quality and function. After generating clinical grade human monocyte-derived mature DCs (mDCs), we tested by gene expression profiling the degrees of product consistency related to the manufacturing process and variability due to intra- and interdonor factors, and how each factor affects single gene variation. Then, by calculating for each gene an index of variation we selected candidate markers for identity testing, and defined a set of genes that may be useful comparability and potency markers. Subsequently, we confirmed the observed gene index of variation in a larger clinical data set. In conclusion, using high-throughput technology we developed a method for the characterization of cellular therapies and the discovery of novel candidate quality assurance markers.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lion E, Smits ELJM, Berneman ZN, Van Tendeloo VFI. NK cells: key to success of DC-based cancer vaccines? Oncologist 2012; 17:1256-70. [PMID: 22907975 DOI: 10.1634/theoncologist.2011-0122] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The cytotoxic and regulatory antitumor functions of natural killer (NK) cells have become attractive targets for immunotherapy. Manipulation of specific NK cell functions and their reciprocal interactions with dendritic cells (DCs) might hold therapeutic promise. In this review, we focus on the engagement of NK cells in DC-based cancer vaccination strategies, providing a comprehensive overview of current in vivo experimental and clinical DC vaccination studies encompassing the monitoring of NK cells. From these studies, it is clear that NK cells play a key regulatory role in the generation of DC-induced antitumor immunity, favoring the concept that targeting both innate and adaptive immune mechanisms may synergistically promote clinical outcome. However, to date, DC vaccination trials are only infrequently accompanied by NK cell monitoring. Here, we discuss different strategies to improve DC vaccine preparations via exploitation of NK cells and provide a summary of relevant NK cell parameters for immune monitoring. We underscore that the design of DC-based cancer vaccines should include the evaluation of their NK cell stimulating potency both in the preclinical phase and in clinical trials.
Collapse
Affiliation(s)
- Eva Lion
- Vaccine & Infectious Disease Institute (Vaxinfectio), Laboratory of Experimental Hematology, TIGR, University of Antwerp (UA), Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Antwerp, Belgium.
| | | | | | | |
Collapse
|
38
|
Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm 2012; 2012:690643. [PMID: 22851815 PMCID: PMC3407661 DOI: 10.1155/2012/690643] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/17/2012] [Indexed: 02/08/2023] Open
Abstract
Earlier investigations have revealed a surprising complexity and variety in the range of interaction between cells of the innate and adaptive immune system. Our understanding of the specialized roles of dendritic cell (DC) subsets in innate and adaptive immune responses has been significantly advanced over the years. Because of their immunoregulatory capacities and because very small numbers of activated DC are highly efficient at generating immune responses against antigens, DCs have been vigorously used in clinical trials in order to elicit or amplify immune responses against cancer and chronic infectious diseases. A better insight in DC immunobiology and function has stimulated many new ideas regarding the potential ways forward to improve DC therapy in a more fundamental way. Here, we discuss the continuous search for optimal in vitro conditions in order to generate clinical-grade DC with a potent immunogenic potential. For this, we explore the molecular and cellular mechanisms underlying adequate immune responses and focus on most favourable DC culture regimens and activation stimuli in humans. We envisage that by combining each of the features outlined in the current paper into a unified strategy, DC-based vaccines may advance to a higher level of effectiveness.
Collapse
|
39
|
Alyamkina EA, Leplina OY, Ostanin AA, Chernykh ER, Nikolin VP, Popova NA, Proskurina AS, Gvozdeva TS, Dolgova EV, Orishchenko KE, Rogachev VA, Sidorov SV, Varaksin NA, Ryabicheva TG, Bogachev SS, Shurdov MA. Effects of human exogenous DNA on production of perforin-containing CD8+ cytotoxic lymphocytes in laboratory setting and clinical practice. Cell Immunol 2012; 276:59-66. [PMID: 22578800 DOI: 10.1016/j.cellimm.2012.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/14/2012] [Accepted: 04/02/2012] [Indexed: 12/24/2022]
Abstract
We investigated the influence of Panagen DNA preparations on laboratory animals and IFN-induced human dendritic cells, as well as analyzed the data from a phase II clinical trial in the therapy of breast cancer. It was shown that this treatment resulted in increased number of CD8+/perforin+ T cells in peripheral lymphoid organs of experimental animals, in mixed lymphocyte culture population and in peripheral blood of breast cancer patients. Moreover, we demonstrated that when Panagen DNA preparations are used in combination with the standard FAC-based breast cancer therapies, non-specific immune response activity remains at the same levels as observed prior to therapy, whereas in FAC-placebo patients, non-specific immunity is greatly diminished.
Collapse
Affiliation(s)
- Ekaterina A Alyamkina
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Langerhans cells from human cutaneous squamous cell carcinoma induce strong type 1 immunity. J Invest Dermatol 2012; 132:1645-55. [PMID: 22402444 DOI: 10.1038/jid.2012.34] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Langerhans cells (LCs) are dendritic cells (DCs) localized to the epidermis. They should be the first antigen-presenting cells to encounter squamous cell carcinoma (SCC). The aim of this study was to investigate the ability of LCs isolated from human SCC to induce T-cell proliferation and polarization. We investigated the ability of LCs from SCC and peritumoral skin to induce T-cell proliferation and polarization. We also studied the effect of SCC supernatant on the ability of LCs from normal skin, in vitro-generated LCs, and DCs to activate and polarize T cells. LCs from SCC were stronger inducers of allogeneic CD4(+) and CD8(+) T-cell proliferation and IFN-γ production than LCs from peritumoral skin. We found that tumor supernatants (TSNs) were rich in immunosuppressive cytokines; despite this, allogeneic CD4(+) and CD8(+) T-cell proliferation and IFN-γ induction by LCs were augmented by TSN. Moreover, TSN facilitated IFN-γ induction by in vitro-generated LCs, but suppressed the ability of in vitro-generated DCs to expand allogeneic CD4(+) and CD8(+) T cells. We have demonstrated that LCs from SCC can induce type 1 immunity. TSN induces IFN-γ induction by in vitro-generated LCs. This contrasts greatly with prior studies showing that DCs from SCC cannot stimulate T cells. These data indicate that LCs may be superior to DCs for SCC immunotherapy and may provide a new rationale for harnessing LCs for the treatment of cancer patients.
Collapse
|
41
|
Weigand K, Voigt F, Encke J, Hoyler B, Stremmel W, Eisenbach C. Vaccination with dendritic cells pulsed with hepatitis C pseudo particles induces specific immune responses in mice. World J Gastroenterol 2012; 18:785-93. [PMID: 22371638 PMCID: PMC3286141 DOI: 10.3748/wjg.v18.i8.785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 03/26/2011] [Accepted: 06/13/2011] [Indexed: 02/06/2023] Open
Abstract
AIM To explore dendritic cells (DCs) multiple functions in immune modulation. METHODS We used bone-marrow derived dendritic cells from BALB/c mice pulsed with pseudo particles from the hepatitis C virus to vaccinate naive BALB/c mice. Hepatitis C virus (HCV) pseudo particles consist of the genotype 1b derived envelope proteins E1 and E2, covering a non-HCV core structure. Thus, not a single epitope, but the whole "viral surface" induces immunogenicity. For vaccination, mature and activated DC were injected subcutaneously twice. RESULTS Humoral and cellular immune responses measured by enzyme-linked immunosorbent assay and interferon-gamma enzyme-linked immunosorbent spot test showed antibody production as well as T-cells directed against HCV. Furthermore, T-cell responses confirmed two highly immunogenic regions in E1 and E2 outside the hypervariable region 1. CONCLUSION Our results indicate dendritic cells as a promising vaccination model for HCV infection that should be evaluated further.
Collapse
|
42
|
Le K, Li X, Figueroa D, Towner RA, Garteiser P, Saunders D, Smith N, Liu H, Hode T, Nordquist RE, Chen WR. Assessment of thermal effects of interstitial laser phototherapy on mammary tumors using proton resonance frequency method. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:128001. [PMID: 22191937 PMCID: PMC3245746 DOI: 10.1117/1.3659200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 10/18/2011] [Accepted: 10/19/2011] [Indexed: 05/31/2023]
Abstract
Laser immunotherapy (LIT) uses a synergistic approach to treat cancer systemically through local laser irradiation and immunological stimulation. Currently, LIT utilizes dye-assisted noninvasive laser irradiation to achieve selective photothermal interaction. However, LIT faces difficulties treating deeper tumors or tumors with heavily pigmented overlying skin. To circumvent these barriers, we use interstitial laser irradiation to induce the desired photothermal effects. The purpose of this study is to analyze the thermal effects of interstitial irradiation using proton resonance frequency (PRF). An 805-nm near-infrared laser with an interstitial cylindrical diffuser was used to treat rat mammary tumors. Different power settings (1.0, 1.25, and 1.5 W) were applied with an irradiation duration of 10 min. The temperature distributions of the treated tumors were measured by a 7 T magnetic resonance imager using PRF. We found that temperature distributions in tissue depended on both laser power and time settings, and that variance in tissue composition has a major influence in temperature elevation. The temperature elevations measured during interstitial laser irradiation by PRF and thermocouple were consistent, with some variations due to tissue composition and the positioning of the thermocouple's needle probes. Our results indicated that, for a tissue irradiation of 10 min, the elevation of rat tumor temperature ranged from 8 to 11°C for 1 W and 8 to 15°C for 1.5 W. This is the first time a 7 T magnetic resonance imager has been used to monitor interstitial laser irradiation via PRF. Our work provides a basic understanding of the photothermal interaction needed to control the thermal damage inside a tumor using interstitial laser treatment. Our work may lead to an optimal protocol for future cancer treatment using interstitial phototherapy in conjunction with immunotherapy.
Collapse
Affiliation(s)
- Kelvin Le
- University of Central Oklahoma, Department of Engineering and Physics, Edmond, Oklahoma 73034, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cools N, Petrizzo A, Smits E, Buonaguro FM, Tornesello ML, Berneman Z, Buonaguro L. Dendritic cells in the pathogenesis and treatment of human diseases: a Janus Bifrons? Immunotherapy 2011; 3:1203-1222. [PMID: 21995572 DOI: 10.2217/imt.11.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) represent the bridging cell compartment between a variety of nonself antigens (i.e., microbial, cancer and vaccine antigens) and adaptive immunity, orchestrating the quality and potency of downstream immune responses. Because of the central role of DCs in the generation and regulation of immunity, the modulation of DC function in order to shape immune responses is gaining momentum. In this respect, recent advances in understanding DC biology, as well as the required molecular signals for induction of T-cell immunity, have spurred many experimental strategies to use DCs for therapeutic immunological approaches for infections and cancer. However, when DCs lose control over such 'protective' responses - by alterations in their number, phenotype and/or function - undesired effects leading to allergy and autoimmune clinical manifestations may occur. Novel therapeutic approaches have been designed and currently evaluated in order to address DCs and silence these immunopathological processes. In this article we present recent concepts of DC biology and some medical implications in view of therapeutic opportunities.
Collapse
Affiliation(s)
- Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, B-2610 Wilrijk, Belgium
| | | | | | | | | | | | | |
Collapse
|
44
|
Selmeczi D, Hansen TS, Met O, Svane IM, Larsen NB. Efficient large volume electroporation of dendritic cells through micrometer scale manipulation of flow in a disposable polymer chip. Biomed Microdevices 2011; 13:383-92. [PMID: 21207149 DOI: 10.1007/s10544-010-9507-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We present a hybrid chip of polymer and stainless steel designed for high-throughput continuous electroporation of cells in suspension. The chip is constructed with two parallel stainless steel mesh electrodes oriented perpendicular to the liquid flow. The relatively high hydrodynamic resistance of the micrometer sized holes in the meshes compared to the main channel enforces an almost homogeneous flow velocity between the meshes. Thereby, very uniform electroporation of the cells can be accomplished. Successful electroporation of 20 million human dendritic cells with mRNA is demonstrated. The performance of the chip is similar to that of the traditional electroporation cuvette, but without an upper limit on the number of cells to be electroporated. The device is constructed with two female Luer parts and can easily be integrated with other microfluidic components. Furthermore it is fabricated from injection molded polymer parts and commercially available stainless steel mesh, making it suitable for inexpensive mass production.
Collapse
Affiliation(s)
- David Selmeczi
- Department of Micro- and Nanotechnology, Technical University of Denmark, DTU Nanotech, 2800 Kgs. Lyngby, Denmark
| | | | | | | | | |
Collapse
|
45
|
Castiello L, Sabatino M, Jin P, Clayberger C, Marincola FM, Krensky AM, Stroncek DF. Monocyte-derived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother 2011; 60:457-66. [PMID: 21258790 PMCID: PMC3086891 DOI: 10.1007/s00262-010-0954-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 11/30/2010] [Indexed: 12/17/2022]
Abstract
Ex vivo production of highly stimulator mature dendritic cells (DCs) for cellular therapy has been used to treat different pathological conditions with the aim of inducing a specific immune response. In the last decade, several protocols have been developed to mature monocyte-derived DCs: each one has led to the generation of DCs showing different phenotypes and stimulatory abilities, but it is not yet known which one is the best for inducing effective immune responses. We grouped several different maturation protocols according to the downstream pathways they activated and reviewed the shared features at a transcriptomic level to reveal the potential of DCs matured by each protocol to develop Th-polarized immune responses.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Marianna Sabatino
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ping Jin
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Carol Clayberger
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, and Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892 USA
| | - Alan M. Krensky
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - David F. Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
46
|
Current world literature. Curr Opin Oncol 2011; 23:227-34. [PMID: 21307677 DOI: 10.1097/cco.0b013e328344b687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Li X, Le H, Wolf RF, Chen VA, Sarkar A, Nordquist RE, Ferguson H, Liu H, Chen WR. Long-term effect on EMT6 tumors in mice induced by combination of laser immunotherapy and surgery. Integr Cancer Ther 2010; 10:368-73. [PMID: 21147818 DOI: 10.1177/1534735410387597] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Laser immunotherapy (LI) has been demonstrated to be a promising modality for cancer treatment. The present study was designed to further investigate the impact of LI combined with surgery. LI consists of a near-infrared laser, a light-absorbing dye (indocyanine green, ICG), and an immunostimulant (glycated chitosan, GC). ICG and GC were intratumorally injected, followed by laser irradiation. Female BALB/c mice bearing EMT6 tumor cells were divided into 4 groups: control, LI, LI followed by immediate surgery resection of residual tumor (LI + S(0wk)), and LI followed by surgical removal of residual tumor after 1 week (LI + S(1wk)). Successfully treated mice from all treatment groups were rechallenged twice with 10(5) and 5 × 10(5) EMT6 cells, respectively. The LI + S(1wk) group had the highest survival rate (72%) after 90 days, whereas the mice survival rates of the LI + S(0wk), LI, and control groups were 50%, 46%, 0%, respectively. The median survival times of control, LI, LI + S(0wk), and LI + S(1wk) groups were 32, 66, 74, and 90 days, respectively. Survival rates of the treated mice after the first and second tumor rechallenges, ranging from 73% to 95%, were not significantly different among the 4 groups (P > .05). The results show that LI is a useful tool for the treatment of tumor-bearing mice. Long-term antitumor effect can be induced by LI. They also indicate that combination of LI with surgery can further improve the therapeutic efficiency of LI.
Collapse
Affiliation(s)
- Xiaosong Li
- Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gujar SA, Marcato P, Pan D, Lee PWK. Reovirus virotherapy overrides tumor antigen presentation evasion and promotes protective antitumor immunity. Mol Cancer Ther 2010; 9:2924-33. [PMID: 20978162 DOI: 10.1158/1535-7163.mct-10-0590] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor-associated immunosuppressive strategies, such as lack of tumor antigen recognition and failure of lymphocyte activation and homing, resist the development of tumor-specific immunity and hamper the immune response-mediated elimination of cancerous cells. In this report, we show that reovirus virotherapy overrides such a tumor immune evasion and establishes clinically meaningful antitumor immunity capable of protecting against subsequent tumor challenge. Reovirus-mediated destruction of tumor cells facilitates the recognition of tumor antigens by promoting the display of otherwise inaccessible tumor-specific immunogenic peptides on the surface of dendritic cells (DC). Furthermore, on exposure to reovirus, DCs produce IL-1α, IL-1β, IL-6, IL-12p40/70, IL-17, CD30L, eotaxin, GM-CSF, KC, MCP-1, MCP-5, M-CSF, MIG, MIP-1α, RANTES, TNF-α, VCAM-1, VSGF, CXCL-16, AXL, and MCP-2; undergo maturation; and migrate into the tumor microenvironment along with CD8 T cells. These reovirus-activated DCs also acquire the capacity to prime tumor antigen-specific transgenic T cells in vitro and intrinsic antitumor T-cell response in vivo. Further, reovirus virotherapy augments the efficacy of DC- or T cell-based anticancer immunotherapies and synergistically enhances the survival in tumor-bearing mice. Most importantly, antitumor cellular immune responses initiated during reovirus oncotherapy protect the host against subsequent tumor challenge in a reovirus-independent but antigen-dependent manner. These reovirus oncotherapy-initiated antitumor immune responses represent an anticancer therapeutic entity that can maintain a long-term cancer-free health even after discontinuation of therapy.
Collapse
Affiliation(s)
- Shashi A Gujar
- Department of Pathology and Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Room 7-P, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | | | |
Collapse
|
49
|
Overview of cellular immunotherapy for patients with glioblastoma. Clin Dev Immunol 2010; 2010. [PMID: 20953324 PMCID: PMC2952949 DOI: 10.1155/2010/689171] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/13/2010] [Accepted: 08/27/2010] [Indexed: 12/22/2022]
Abstract
High grade gliomas (HGG) including glioblastomas (GBM) are the most common and devastating primary brain tumours. Despite important progresses in GBM treatment that currently includes surgery combined to radio- and chemotherapy, GBM patients' prognosis remains very poor. Immunotherapy is one of the new promising therapeutic approaches that can specifically target tumour cells. Such an approach could also maintain long term antitumour responses without inducing neurologic defects. Since the past 25 years, adoptive and active immunotherapies using lymphokine-activated killer cells, cytotoxic T cells, tumour-infiltrating lymphocytes, autologous tumour cells, and dendritic cells have been tested in phase I/II clinical trials with HGG patients. This paper inventories these cellular immunotherapeutic strategies and discusses their efficacy, limits, and future perspectives for optimizing the treatment to achieve clinical benefits for GBM patients.
Collapse
|
50
|
A liposome-based platform, VacciMax, and its modified water-free platform DepoVax enhance efficacy of in vivo nucleic acid delivery. Vaccine 2010; 28:6176-82. [PMID: 20656034 DOI: 10.1016/j.vaccine.2010.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 06/10/2010] [Accepted: 07/08/2010] [Indexed: 11/24/2022]
Abstract
Nucleic acid vaccines represent a promising alternative to killed bacterial antigen, recombinant protein or peptide vaccines for infectious diseases and cancer immunotherapy. Although significant advances are made with DNA vaccines in animal studies, there are severe limitations to deliver these vaccines effectively and considerable reservations exist about current methods used. In this study, a liposome-based vaccine platform, VacciMax (VM), and its modified water-free version, DepoVax (DPX), were tested for their ability to improve in vivo delivery of plasmid DNA (pDNA), mRNA and siRNA. Subcutaneously injected pDNA for IL12 and pDNA as well as mRNA for green fluorescent protein (GFP) in VM/DPX significantly enhanced their in vivo expression. Enhanced IL12 secretion and GFP expression was restricted to CD11b(+) and CD11c(+) antigen-presenting cells, but not B cells. Further, significant inhibition of plasmid/antigen-induced IL12 secretion was seen after injection of IL12-siRNA in VM. These findings suggest VM and DPX to be promising means of delivering nucleic acid vaccines in vivo, and warrant further studies on their role in inducing effective immune responses.
Collapse
|