1
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
2
|
Seshadri G, Vivek S, Prizment A, Crimmins EM, Klopack ET, Faul J, Guan W, Meier HCS, Thyagarajan B. Immune cells are associated with mortality: the Health and Retirement Study. Front Immunol 2023; 14:1280144. [PMID: 37928548 PMCID: PMC10623116 DOI: 10.3389/fimmu.2023.1280144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Age-related immunosenescence is characterized by changes in immune cell subsets and is associated with mortality. However, since immunosenescence is associated with other concurrent age-related changes such as inflammation and multi-organ dysfunction, it is unclear whether the association between age-related immunosenescence and mortality is independent of other concurrent age-related changes. To address these limitations, we evaluated the independent association between immune cell subsets and mortality after adjustment for age-related inflammation and biologic age. Methods Data for this study was obtained from the 2016 interview of the Health and Retirement Study (N=6802). Cox proportional hazards regression models were used to estimate the association between 25 immune cell subsets (11 T-cell subsets, 4 B-cell subsets, 3 monocyte subsets, 3 natural killer cell subsets, 3 dendritic cell subsets, and neutrophils) and 4-year mortality adjusting for covariates such as the Klemera-Doubal algorithm biological age, chronological age, gender, race/ethnicity, BMI, smoking status, comorbidity index, CMV seropositivity, and inflammatory latent variable comprising C-reactive protein, and 4 cytokines (interleukin-10, interleukin-1 receptor antagonist, interleukin-6, and soluble tumor necrosis factor). Results Four hundred and seventy-six participants died during the study period with an overall median follow up time of 2.5 years. After controlling for covariates and adjustment for sample-weights, total T cells [HR: 0.86, p=0.004], NK CD56LO cells [HR: 0.88, p=0.005], and neutrophils [HR: 1.22, p=0.004] were significantly associated with mortality. Conclusions These findings support the idea that an aging immune system is associated with short-term mortality independent of age-related inflammation or other age-related measures of physiological dysfunction. If replicated in other external cohorts, these findings could identify novel targets for both monitoring and intervention to reduce the age-related mortality.
Collapse
Affiliation(s)
- Gokul Seshadri
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Sithara Vivek
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Eileen M. Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Eric T. Klopack
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Jessica Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Helen C. S. Meier
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
3
|
Trelford CB, Dagnino L, Di Guglielmo GM. Transforming growth factor-β in tumour development. Front Mol Biosci 2022; 9:991612. [PMID: 36267157 PMCID: PMC9577372 DOI: 10.3389/fmolb.2022.991612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/15/2022] [Indexed: 11/14/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a ubiquitous cytokine essential for embryonic development and postnatal tissue homeostasis. TGFβ signalling regulates several biological processes including cell growth, proliferation, apoptosis, immune function, and tissue repair following injury. Aberrant TGFβ signalling has been implicated in tumour progression and metastasis. Tumour cells, in conjunction with their microenvironment, may augment tumourigenesis using TGFβ to induce epithelial-mesenchymal transition, angiogenesis, lymphangiogenesis, immune suppression, and autophagy. Therapies that target TGFβ synthesis, TGFβ-TGFβ receptor complexes or TGFβ receptor kinase activity have proven successful in tissue culture and in animal models, yet, due to limited understanding of TGFβ biology, the outcomes of clinical trials are poor. Here, we review TGFβ signalling pathways, the biology of TGFβ during tumourigenesis, and how protein quality control pathways contribute to the tumour-promoting outcomes of TGFβ signalling.
Collapse
Affiliation(s)
- Charles B. Trelford
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Children’s Health Research Institute and Lawson Health Research Institute, London, ON, Canada
| | - Gianni M. Di Guglielmo
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
4
|
Zhao L, Zhang S, Kepp O, Kroemer G, Liu P. Dendritic cell transfer for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:33-64. [PMID: 35798506 DOI: 10.1016/bs.ircmb.2022.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Dendritic cells (DCs) play a major role in cancer immunosurveillance as they bridge innate and adaptive immunity by detecting tumor-associated antigens and presenting them to T lymphocytes. The adoptive transfer of antigen loaded DCs has been proposed as an immunotherapeutic approach for the treatment of various types of cancer. Nevertheless, despite promising preclinical data, the therapeutic efficacy of DC transfer is still deceptive in cancer patients. Here we summarize recent findings in DC biology with a special focus on the development of actionable therapeutic strategies and discuss experimental and clinical approaches that aim at improving the efficacy of DC-based immunotherapies, including, but not limited to, optimized DC production and antigen loading, stimulated maturation, the co-treatment with additional immunotherapies, as well as the inhibition of DC checkpoints.
Collapse
Affiliation(s)
- Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Shuai Zhang
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Institut du Cancer Paris Carpem, Department of Biology, Hôpital Européen Georges Pompidou, APHP, Paris, France.
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| |
Collapse
|
5
|
Tan L, Shen X, He Z, Lu Y. The Role of Photodynamic Therapy in Triggering Cell Death and Facilitating Antitumor Immunology. Front Oncol 2022; 12:863107. [PMID: 35692783 PMCID: PMC9184441 DOI: 10.3389/fonc.2022.863107] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is a major threat to human health because of its high mortality, easy recurrence, strong invasion, and metastasis. Photodynamic therapy (PDT) is a promising minimally invasive treatment for tumor. Compared with traditional treatment methods, PDT is less invasive and does not easily damage normal tissues. Most of the effects of this treatment are due to the direct effects of singlet oxygen together with reactive oxygen species. PDT can provide the source of active oxygen for the Fenton reaction, which enhances ferroptosis and also improves the efficacy of PDT in antitumor therapy. Additionally, in contrast to chemotherapy and radiotherapy, PDT has the effect of stimulating the immune response, which can effectively induce immunogenic cell death (ICD) and stimulate immunity. PDT is an ideal minimally invasive treatment method for tumors. In this paper, according to the characteristics of anti-tumor immunity of PDT, some tumor treatment strategies of PDT combined with anti-tumor immunotherapy are reviewed.
Collapse
|
6
|
Ramírez-Rico G, Drago-Serrano ME, León-Sicairos N, de la Garza M. Lactoferrin: A Nutraceutical with Activity against Colorectal Cancer. Front Pharmacol 2022; 13:855852. [PMID: 35264972 PMCID: PMC8899398 DOI: 10.3389/fphar.2022.855852] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
Homeostasis in the human body results from the tight regulation of several events, since too little inflammation disrupts the process of tissue repair and remodeling, whereas too much exerts a collateral effect by causing tissue damage with life-threatening consequences. In some clinical conditions, such as inflammatory bowel disease (IBD), inflammation functions as a double-edged sword by either enabling or inhibiting cancer development and progression. Generally, cancer develops through evasion mechanisms that regulate cell growth, causing a high rate of uncontrolled proliferation, and mechanisms for evading cell death, such as apoptosis. Moreover, chronic inflammation is a factor that contributes to colorectal cancer (CRC), as observed in individuals with IBD; all these conditions favor an increased rate of angiogenesis and eventual metastasis. Lactoferrin (Lf) is a mammalian iron-binding multifunctional glycoprotein regarded as a natural compound that up- and downregulates both humoral and cellular components of immunity involved in regulating the inflammatory response and maintaining gut homeostasis. Human and bovine Lf share high sequence homology and have very similar antimicrobial, anti-inflammatory, and immunomodulatory activities. Bovine Lf from milk is considered a safe molecule and is commercially available in large quantities. This review mainly focuses on the regulatory effects of orally administered bovine Lf on the inflammatory response associated with CRC; this approach indicates that CRC is one of the most frequently diagnosed cancers and affects the intestinal tract with high clinical and epidemiologic relevance. Thus, this review may provide foundations for the potential use of bovine Lf alone or as a natural adjunct agent to increase the effectiveness and reduce the side effects of anticancer chemotherapy.
Collapse
Affiliation(s)
- Gerardo Ramírez-Rico
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), México City, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City, Mexico
| | - Nidia León-Sicairos
- Centro de Investigación Aplicada a La Salud Pública (CIASaP), Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán, Mexico
- Hospital Pediátrico de Sinaloa, Culiacán, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- *Correspondence: Mireya de la Garza,
| |
Collapse
|
7
|
Lu Y, Shi Y, You J. Strategy and clinical application of up-regulating cross presentation by DCs in anti-tumor therapy. J Control Release 2021; 341:184-205. [PMID: 34774890 DOI: 10.1016/j.jconrel.2021.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/20/2022]
Abstract
The cross presentation of exogenous antigen (Ag) by dendritic cells (DCs) facilitates a diversified mode of T-cell activation, orchestrates specific humoral and cellular immunity, and contributes to an efficient anti-tumor immune response. DCs-mediated cross presentation is subject to both intrinsic and extrinsic factors, including the homing and phenotype of DCs, the spatiotemporal trafficking and degradation kinetics of Ag, and multiple microenvironmental clues, with many details largely unexplored. Here, we systemically review the current mechanistic understanding and regulation strategies of cross presentation by heterogeneous DC populations. We also provide insights into the future exploitation of DCs cross presentation for a better clinical efficacy in anti-tumor therapy.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
8
|
Li Y, Wang X, Ma X, Liu C, Wu J, Sun C. Natural Polysaccharides and Their Derivates: A Promising Natural Adjuvant for Tumor Immunotherapy. Front Pharmacol 2021; 12:621813. [PMID: 33935714 PMCID: PMC8080043 DOI: 10.3389/fphar.2021.621813] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/15/2021] [Indexed: 12/30/2022] Open
Abstract
The treatment process of tumor is advanced with the development of immunotherapy. In clinical experience, immunotherapy has achieved very significant results. However, the application of immunotherapy is limited by a variety of immune microenvironment. For a long time in the past, polysaccharides such as lentinan and Ganoderma lucidum glycopeptide have been used in clinic as adjuvant drugs to widely improve the immunity of the body. However, their mechanism in tumor immunotherapy has not been deeply discussed. Studies have shown that natural polysaccharides can stimulate innate immunity by activating upstream immune cells so as to regulate adaptive immune pathways such as T cells and improve the effect of immunotherapy, suggesting that polysaccharides also have a promising future in cancer therapy. This review systematically discusses that polysaccharides can directly or indirectly activate macrophages, dendritic cells, natural killer cells etc., binding to their surface receptors, inducing PI3K/Akt, mitogen-activated protein kinase, Notch and other pathways, promote their proliferation and differentiation, increasing the secretion of cytokines, and improve the state of immune suppression. These results provide relevant basis for guiding polysaccharide to be used as adjuvants of cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaomin Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.,Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
9
|
Larmonier N, Bonnotte B, Katsanis E. Cytotoxic and antigen presenting functions of T helper-1-activated dendritic cells. Oncoimmunology 2021; 1:566-568. [PMID: 22754789 PMCID: PMC3382885 DOI: 10.4161/onci.19370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although primarily defined by their cardinal antigen-presenting function, dendritic cells (DCs) are also equipped with cytotoxic properties. We have recently reported that DCs activated by IFNγ-secreting Th-1 lymphocytes can kill cancer cells and subsequently present the acquired tumor-derived antigens to T lymphocytes both in vitro and in vivo.
Collapse
Affiliation(s)
- Nicolas Larmonier
- Department of Pediatrics; Steele Children's Research Center; Department of immunobiology; BIO5 Institute and Arizona Cancer Center; University of Arizona; Tucson, AZ USA
| | | | | |
Collapse
|
10
|
Batchu RB, Gruzdyn OV, Kolli BK, Dachepalli R, Umar PS, Rai SK, Singh N, Tavva PS, Weaver DW, Gruber SA. IL-10 Signaling in the Tumor Microenvironment of Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:51-65. [PMID: 33559854 DOI: 10.1007/978-3-030-55617-4_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Unlike other malignancies, ovarian cancer (OC) creates a complex tumor microenvironment with distinctive peritoneal ascites consisting of a mixture of several immunosuppressive cells which impair the ability of the patient's immune system to fight the disease. The poor survival rates observed in advanced stage OC patients and the lack of effective conventional therapeutic options have been attributed in large part to the immature dendritic cells (DCs), IL-10 secreting regulatory T cells, tumor-associated macrophages, myeloid-derived suppressor cells, and cancer stem cells that secrete inhibitory cytokines. This review highlights the critical role played by the intraperitoneal presence of IL-10 in the generation of an immunosuppressive tumor microenvironment. Further, the effect of antibody neutralization of IL-10 on the efficacy of DC and chimeric antigen receptor T-cell vaccines will be discussed. Moreover, we will review the influence of IL-10 in the promotion of cancer stemness in concert with the NF-κB signaling pathway with regard to OC progression. Finally, understanding the role of IL-10 and its crosstalk with various cells in the ascitic fluid may contribute to the development of novel immunotherapeutic approaches with the potential to kill drug-resistant OC cells while minimizing toxic side effects.
Collapse
Affiliation(s)
- Ramesh B Batchu
- Wayne State University School of Medicine, Detroit, MI, USA. .,John D. Dingell VA Medical Center, Detroit, MI, USA.
| | - Oksana V Gruzdyn
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Bala K Kolli
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA.,Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | - Prem S Umar
- Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | | | | | | | - Scott A Gruber
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
11
|
Varga Z, Rácz E, Mázló A, Korodi M, Szabó A, Molnár T, Szöőr Á, Veréb Z, Bácsi A, Koncz G. Cytotoxic activity of human dendritic cells induces RIPK1-dependent cell death. Immunobiology 2020; 226:152032. [PMID: 33316542 DOI: 10.1016/j.imbio.2020.152032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/05/2020] [Accepted: 11/18/2020] [Indexed: 02/05/2023]
Abstract
Dendritic cells (DCs), as potent phagocytes engulf dead cells and present peptide fragments of tumor antigens or pathogens derived from infected cells to naïve CD8+ T-lymphocytes. Dendritic cells can also induce apoptosis in target cells, thus getting an opportunity to sample their microenvironment. Here, we present that the supernatants of LPS- or CL075-activated DCs induced cell death in different cell lines, but during the differentiation to mature DCs, they lost their cytotoxic potential. Dexamethasone-pre-treated tolerogenic DCs induced less intensive death indicating that the tissue microenvironment can downregulate DC-mediated killing. Exploring the signaling of DC-induced cell death, we observed that the supernatant of activated DCs induced TNF-dependent cell death, since TNF antagonist blocked the cytotoxic activity of DCs, contrary to inhibitors of Fas and TRAIL receptors. We identified that the DC-induced killing is at least partially a RIPK1-dependent process, as RIPK1 positive target cells were more susceptible to DC-induced cell death than their RIPK1 deficient counterparts. Moreover, both the elevated phosphorylation of RIPK1 and the increase in RIPK1-caspase-8 interaction in target cells suggest that RIPK1-mediated signals contribute to DC supernatant-induced cell death. We also proved that the cytotoxic activity of DC-derived supernatant induced apoptosis in the target cells and not necroptosis, as it was completely abrogated with the pan caspase inhibitor (Z-VAD), while the necroptosis inhibitor (Nec-1) had no effect. Our work revealed that the supernatant of activated DCs induces the apoptosis of target cells in a RIPK1-dependent manner. This phenomenon could be relevant for the initiation of cross-presentation and may broaden the plethora of cytotoxic mechanisms acting against tumor cells.
Collapse
Affiliation(s)
- Zsófia Varga
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Cellular and Immune Biology, Debrecen, Hungary
| | - Evelin Rácz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Cellular and Immune Biology, Debrecen, Hungary
| | - Mónika Korodi
- University of Pécs, Doctoral School of Chemistry, Department of Chemistry, Faculty of Sciences, Pécs, Hungary
| | - Anikó Szabó
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Molnár
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Cellular and Immune Biology, Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory (HECRIN), Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; Institute for Translational Medicine, University of Szeged, Szeged, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
12
|
Korbecki J, Olbromski M, Dzięgiel P. CCL18 in the Progression of Cancer. Int J Mol Sci 2020; 21:ijms21217955. [PMID: 33114763 PMCID: PMC7663205 DOI: 10.3390/ijms21217955] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
A neoplastic tumor consists of cancer cells that interact with each other and non-cancerous cells that support the development of the cancer. One such cell are tumor-associated macrophages (TAMs). These cells secrete many chemokines into the tumor microenvironment, including especially a large amount of CCL18. This chemokine is a marker of the M2 macrophage subset; this is the reason why an increase in the production of CCL18 is associated with the immunosuppressive nature of the tumor microenvironment and an important element of cancer immune evasion. Consequently, elevated levels of CCL18 in the serum and the tumor are connected with a worse prognosis for the patient. This paper shows the importance of CCL18 in neoplastic processes. It includes a description of the signal transduction from PITPNM3 in CCL18-dependent migration, invasion, and epithelial-to-mesenchymal transition (EMT) cancer cells. The importance of CCL18 in angiogenesis has also been described. The paper also describes the effect of CCL18 on the recruitment to the cancer niche and the functioning of cells such as TAMs, regulatory T cells (Treg), cancer-associated fibroblasts (CAFs) and tumor-associated dendritic cells (TADCs). The last part of the paper describes the possibility of using CCL18 as a therapeutic target during anti-cancer therapy.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
- Correspondence: ; Tel.: +48-717-841-354
| | - Mateusz Olbromski
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
- Department of Physiotherapy, Wroclaw University School of Physical Education, Ignacego Jana Paderewskiego 35 Av., 51-612 Wroclaw, Poland
| |
Collapse
|
13
|
Patidar A, Selvaraj S, Chauhan P, Guzman CA, Ebensen T, Sarkar A, Chattopadhyay D, Saha B. Peptidoglycan-treated tumor antigen-pulsed dendritic cells impart complete resistance against tumor rechallenge. Clin Exp Immunol 2020; 201:279-288. [PMID: 32443171 DOI: 10.1111/cei.13468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/28/2022] Open
Abstract
Solid tumors elicit suppressive T cell responses which impair antigen-presenting cell (APC) functions. Such immune suppression results in uncontrolled tumor growth and mortality. Addressing APC dysfunction, dendritic cell (DC)-mediated anti-tumor vaccination was extensively investigated in both mice and humans. These studies never achieved full resistance to tumor relapse. Herein, we describe a repetitive RM-1 murine tumor rechallenge model for recurrence in humans. Using this newly developed model, we show that priming with tumor antigen-pulsed, Toll-like receptor (TLR)2 ligand-activated DCs elicits a host-protective anti-tumor immune response in C57BL/6 mice. Upon stimulation with the TLR2 ligand peptidoglycan (PGN), the tumor antigen-pulsed DCs induce complete resistance to repetitive tumor challenges. Intra-tumoral injection of PGN reduces tumor growth. The tumor resistance is accompanied by increased expression of interleukin (IL)-27, T-box transcription factor TBX21 (T-bet), IL-12, tumor necrosis factor (TNF)-α and interferon (IFN)-γ, along with heightened cytotoxic T lymphocyte (CTL) functions. Mice primed four times with PGN-stimulated tumor antigen-pulsed DCs remain entirely resistant to repeat challenges with RM-1 tumor cells, suggesting complete prevention of relapse and recurrence of tumor. Adoptive transfer of T cells from these mice, which were fully protected from RM-1 rechallenge, confers anti-tumor immunity to syngeneic naive recipient mice upon RM-1 challenge. These observations indicate that PGN-activated DCs induce robust host-protective anti-tumor T cells that completely resist tumor growth and recurrence.
Collapse
Affiliation(s)
- A Patidar
- National Centre for Cell Science, Pune, India
| | - S Selvaraj
- National Centre for Cell Science, Pune, India
| | - P Chauhan
- National Centre for Cell Science, Pune, India
| | - C A Guzman
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - T Ebensen
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - A Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | | | - B Saha
- National Centre for Cell Science, Pune, India.,Trident Academy of Creative Technology, Bhubaneswar, India.,National Institute of Traditional Medicine, Belagavi, India
| |
Collapse
|
14
|
Kounis NG, Ren HL, Kavalioti M, Koniari I, Theoharides TC. Intimate Contact Could Be Dangerous for Your Health. Clin Ther 2019; 41:1222-1226. [PMID: 31076202 DOI: 10.1016/j.clinthera.2019.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/28/2019] [Accepted: 04/09/2019] [Indexed: 11/25/2022]
Abstract
PURPOSE To review evidence of hypersensitivity reactions to allergens and/or pathogens transmitted via intimate contact. METHODS We reviewed PubMed for publications in English between 1980 and 2018 using the terms allergy, drugs, foods, hypersensitivity, intercourse, kissing, Kounis syndrome, mast cells, and semen. FINDINGS In human RELATIONSHIPS, intimate contact can occasionally have disastrous or even fatal consequences because antigens and pathogens can be transmitted via the oral and vaginal mucosa. Hypersensitivity to semen is an underrecognized problem. Some individuals also developed acute coronary hypersensitivity, which mimics myocardial infarction, known as Kounis syndrome. IMPLICATIONS Hypersensitivity reactions to allergens and/or pathogens via intimate contact are common and should be recognized. Sensitive patients should be evaluated for atopic diathesis because such patients may be more susceptible and could also develop Kounis syndrome.
Collapse
Affiliation(s)
- Nicholas G Kounis
- Patras Highest Institute of Education and Technology, Patras, Greece
| | - Huali L Ren
- Department of Otolaryngology, Beijing Electric Power Hospital, Beijing, China
| | - Maria Kavalioti
- Department of Biomedical Sciences, University of Greenwich, United Kingdom
| | - Ioanna Koniari
- Patras Highest Institute of Education and Technology, Patras, Greece
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA; Department of Internal Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
15
|
Galati D, Zanotta S, Corazzelli G, Bruzzese D, Capobianco G, Morelli E, Arcamone M, De Filippi R, Pinto A. Circulating dendritic cells deficiencies as a new biomarker in classical Hodgkin lymphoma. Br J Haematol 2018; 184:594-604. [DOI: 10.1111/bjh.15676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/08/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Domenico Galati
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Serena Zanotta
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Gaetano Corazzelli
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Dario Bruzzese
- Department of Public Health Università degli Studi di Napoli Federico II Napoli Italia
| | - Gaetana Capobianco
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Emanuela Morelli
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Manuela Arcamone
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery Università degli Studi di Napoli Federico II Napoli Italia
| | - Antonio Pinto
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| |
Collapse
|
16
|
Haddad D. Genetically Engineered Vaccinia Viruses As Agents for Cancer Treatment, Imaging, and Transgene Delivery. Front Oncol 2017; 7:96. [PMID: 28589082 PMCID: PMC5440573 DOI: 10.3389/fonc.2017.00096] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/27/2017] [Indexed: 01/08/2023] Open
Abstract
Despite advances in technology, the formidable challenge of treating cancer, especially if advanced, still remains with no significant improvement in survival rates, even with the most common forms of cancer. Oncolytic viral therapies have shown great promise for the treatment of various cancers, with the possible advantages of stronger treatment efficacy compared to conventional therapy due to higher tumor selectivity, and less toxicity. They are able to preferentially and selectively propagate in cancer cells, consequently destroying tumor tissue mainly via cell lysis, while leaving non-cancerous tissues unharmed. Several wild-type and genetically engineered vaccinia virus (VACV) strains have been tested in both preclinical and clinical trials with promising results. Greater understanding and advancements in molecular biology have enabled the generation of genetically engineered oncolytic viruses for safer and more efficacious treatment, including arming VACVs with cytokines and immunostimulatory molecules, anti-angiogenic agents, and enzyme prodrug therapy, in addition to combining VACVs with conventional external and systemic radiotherapy, chemotherapy, immunotherapy, and other virus strains. Furthermore, novel oncolytic vaccinia virus strains have been generated that express reporter genes for the tracking and imaging of viral therapy and monitoring of therapeutic response. Further study is needed to unlock VACVs’ full potential as part of the future of cancer therapy.
Collapse
Affiliation(s)
- Dana Haddad
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Heinrich B, Klein J, Delic M, Goepfert K, Engel V, Geberzahn L, Lusky M, Erbs P, Preville X, Moehler M. Immunogenicity of oncolytic vaccinia viruses JX-GFP and TG6002 in a human melanoma in vitro model: studying immunogenic cell death, dendritic cell maturation and interaction with cytotoxic T lymphocytes. Onco Targets Ther 2017; 10:2389-2401. [PMID: 28496337 PMCID: PMC5422459 DOI: 10.2147/ott.s126320] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oncolytic virotherapy is an emerging immunotherapeutic modality for cancer treatment. Oncolytic viruses with genetic modifications can further enhance the oncolytic effects on tumor cells and stimulate antitumor immunity. The oncolytic vaccinia viruses JX-594-GFP+/hGM-CSF (JX-GFP) and TG6002 are genetically modified by secreting granulocyte-macrophage colony-stimulating factor (GM-CSF) or transforming 5-fluorocytosine (5-FC) into 5-fluorouracil (5-FU). We compared their properties to kill tumor cells and induce an immunogenic type of cell death in a human melanoma cell model using SK29-MEL melanoma cells. Their influence on human immune cells, specifically regarding the activation of dendritic cells (DCs) and the interaction with the autologous cytotoxic T lymphocyte (CTL) clone, was investigated. Melanoma cells were infected with either JX-GFP or TG6002 alone or in combination with 5-FC and 5-FU. The influence of viral infection on cell viability followed a time- and multiplicity of infection dependent manner. Combination of virus treatment with 5-FU resulted in stronger reduction of cell viability. TG6002 in combination with 5-FC did not significantly strengthen the reduction of cell viability in this setting. Expression of calreticulin and high mobility group 1 protein (HMGB1), markers of immunogenic cell death (ICD), could be detected after viral infection. Accordingly, DC maturation was noted after viral oncolysis. DCs presented stronger expression of activation and maturation markers. The autologous CTL clone IVSB expressed the activation marker CD69, but viral treatment failed to enhance cytotoxicity marker. In summary, vaccinia viruses JX-GFP and TG6002 lyse melanoma cells and induce additional immunostimulatory effects to promote antitumor immune response. Further investigation in vivo is needed to consolidate the data.
Collapse
Affiliation(s)
- B Heinrich
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - J Klein
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - M Delic
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - K Goepfert
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - V Engel
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - L Geberzahn
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| | - M Lusky
- Transgene SA, Illkirch-Graffenstaden
| | - P Erbs
- Transgene SA, Illkirch-Graffenstaden
| | | | - M Moehler
- First Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
18
|
Dendritic cells in hematological malignancies. Crit Rev Oncol Hematol 2016; 108:86-96. [DOI: 10.1016/j.critrevonc.2016.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
|
19
|
Yang Y, Hu Y, Wang H. Targeting Antitumor Immune Response for Enhancing the Efficacy of Photodynamic Therapy of Cancer: Recent Advances and Future Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5274084. [PMID: 27672421 PMCID: PMC5031843 DOI: 10.1155/2016/5274084] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/04/2016] [Indexed: 02/06/2023]
Abstract
Photodynamic therapy (PDT) is a minimally invasive therapeutic strategy for cancer treatment, which can destroy local tumor cells and induce systemic antitumor immune response, whereas, focusing on improving direct cytotoxicity to tumor cells treated by PDT, there is growing interest in developing approaches to further explore the immune stimulatory properties of PDT. In this review we summarize the current knowledge of the innate and adaptive immune responses induced by PDT against tumors, providing evidence showing PDT facilitated-antitumor immunity. Various immunotherapeutic approaches on different cells are reviewed for their effectiveness in improving the treatment efficiency in concert with PDT. Future perspectives are discussed for further enhancing PDT efficiency via intracellular targetable drug delivery as well as optimized experimental model development associated with the study of antitumor immune response.
Collapse
Affiliation(s)
- Yamin Yang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, 169 Sheng Tai West Road, Nanjing, Jiangsu 211106, China
| | - Yue Hu
- Department of Biological and Environmental Engineering, Cornell University, 120 Riley Robb, Ithaca, NY 14853, USA
| | - Hongjun Wang
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Castle Point on Hudson, Hoboken, NJ 07030, USA
| |
Collapse
|
20
|
da Costa Souza P, Parra ER, Atanazio MJ, da Silva OB, Noleto GS, Ab'Saber AM, de Morais Fernezlian S, Takagaki T, Capelozzi VL. Different morphology, stage and treatment affect immune cell infiltration and long-term outcome in patients with non-small-cell lung carcinoma. Histopathology 2016; 61:587-96. [PMID: 22716510 DOI: 10.1111/j.1365-2559.2012.04318.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Development of effective immune-based therapies for patients with non-small-cell lung carcinoma (NSCLC) depends on an accurate characterization of complex interactions that occur between immune cells and the tumour environment. METHODS AND RESULTS Innate and adaptive immune responses were evaluated in relation to prognosis in 65 patients with surgically excised NSCLC. Immunohistochemistry and morphometry were used to determine the abundance and distribution of immune cells. We found low numbers of immune cells and levels of cytokines in the tumour environment when compared with surrounding parenchyma. Smoking was associated inversely with the adaptive immune response and directly with innate immunity. We observed a prominent adaptive immune response in squamous cell carcinomas (SCC) but greater innate immune responses in adenocarcinomas and large cell carcinomas. Cox model analysis showed a low risk of death for smoking <41 packs/year, N0 tambour stage, squamous carcinoma, CD4(+) > 16.81% and macrophages/monocytes >4.5%. Collectively, the data indicate that in NSCLC there is not a substantive local immune cell infiltrate within the tumour. CONCLUSION Although immune cell infiltration is limited in NSCLC it appears to have an impact on prognosis and this may be of relevance for new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Paola da Costa Souza
- Department of PathologyDiscipline of Oncology, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Boyle ST, Faulkner JW, McColl SR, Kochetkova M. The chemokine receptor CCR6 facilitates the onset of mammary neoplasia in the MMTV-PyMT mouse model via recruitment of tumor-promoting macrophages. Mol Cancer 2015; 14:115. [PMID: 26047945 PMCID: PMC4464622 DOI: 10.1186/s12943-015-0394-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/29/2015] [Indexed: 01/02/2023] Open
Abstract
Background The expression of the chemokine receptor CCR6 has been previously correlated with higher grades and stages of breast cancer and decreased relapse-free survival. Also, its cognate chemokine ligand CCL20 has been reported to induce proliferation of cultured human breast epithelial cells. Methods To establish if CCR6 plays a functional role in mammary tumorigenesis, a bigenic MMTV-PyMT CCR6-null mouse was generated and mammary tumor development was assessed. Levels of tumor-infiltrating immune cells within tumor-bearing mammary glands from MMTV-PyMT Ccr6WT and Ccr6−/− mice were also analyzed. Results Deletion of CCR6 delayed tumor onset, significantly reduced the extent of initial hyperplastic outgrowth, and decreased tumor incidence in PyMT transgenic mice. CCR6 was then shown to promote the recruitment of pro-tumorigenic macrophages to the tumor site, facilitating the onset of neoplasia. Conclusions This study delineated for the first time a role for CCR6 in the development of breast cancer, and demonstrated a critical function for this receptor in maintaining the pro-tumorigenic cancer microenvironment. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0394-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarah T Boyle
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica W Faulkner
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Shaun R McColl
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Centre for Molecular Pathology, University of Adelaide, Adelaide, South Australia, Australia
| | - Marina Kochetkova
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
22
|
Chang J, Liu Y, Han B, Zhou C, Bai C, Li J. Pseudomonas aeruginosa preparation plus chemotherapy for advanced non-small-cell lung cancer: a randomized, multicenter, double-blind phase III study. Med Oncol 2015; 32:139. [DOI: 10.1007/s12032-015-0583-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/14/2015] [Indexed: 10/23/2022]
|
23
|
Gasparoto TH, de Oliveira CE, de Freitas LT, Pinheiro CR, Hori JI, Garlet GP, Cavassani KA, Schillaci R, da Silva JS, Zamboni DS, Campanelli AP. Inflammasome activation is critical to the protective immune response during chemically induced squamous cell carcinoma. PLoS One 2014; 9:e107170. [PMID: 25268644 PMCID: PMC4182037 DOI: 10.1371/journal.pone.0107170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/07/2014] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation affects most stages of tumorigenesis, including initiation, promotion, malignant differentiation, invasion and metastasis. Inflammasomes have been described as involved with persistent inflammation and are known to exert both pro and antitumour effects. We evaluated the influence of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and caspase (CASP)-1 in the antitumor immune response using a multistage model of squamous cell carcinoma (SCC) development. Absence of ASC and CASP-1 resulted in an earlier incidence and increased number of papilloma. Loss of inflammassome function in mice resulted in decreased presence of natural killer (NK), dendritic (DC), CD4+, CD8+ and CD45RB+ T cells in the tumor lesions as well as in lymph nodes (LN) compared with WT mice. Increased percentage of CD4+CD25+Foxp3+ T cells was associated with association with inflammasome loss of function. Moreover, significant differences were also found with neutrophils and macrophage infiltrating the lesions. Myeloperoxidase (MPO), but not elastase (ELA), activity oscillated among the groups during the SCC development. Levels of proinflammatory cytokines IL-1β, IL-18, Tumor Necrosis Factor (TNF)-α and Interferon (IFN)-γ were decreased in the tumor microenvironment in the absence of inflammasome proteins. These observations suggest a link between inflammasome function and SCC tumorigenesis, indicating an important role for inflammasome activation in the control of SCC development.
Collapse
Affiliation(s)
- Thais Helena Gasparoto
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Carine Ervolino de Oliveira
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- Department of Stomatology - Oral Pathology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Luisa Thomazini de Freitas
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Claudia Ramos Pinheiro
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Juliana Issa Hori
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Karen Angélica Cavassani
- Departament of Pathology, Medical School, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Roxana Schillaci
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Buenos Aires, Argentina
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Dario Simões Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences - Microbiology and Immunology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- * E-mail:
| |
Collapse
|
24
|
Hanke NT, LaCasse CJ, Larmonier CB, Alizadeh D, Trad M, Janikashvili N, Bonnotte B, Katsanis E, Larmonier N. PIAS1 and STAT-3 impair the tumoricidal potential of IFN-γ-stimulated mouse dendritic cells generated with IL-15. Eur J Immunol 2014; 44:2489-2499. [PMID: 24777831 DOI: 10.1002/eji.201343803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 04/04/2014] [Accepted: 04/17/2014] [Indexed: 01/22/2023]
Abstract
Primarily defined by their antigen-presenting property, dendritic cells (DCs) are being implemented as cancer vaccines in immunotherapeutic interventions. DCs can also function as direct tumor cell killers. How DC cytotoxic activity can be efficiently harnessed and the mechanisms controlling this nonconventional property are not fully understood. We report here that the tumoricidal potential of mouse DCs generated from myeloid precursors with GM-CSF and IL-15 (IL-15 DCs) can be triggered with the Toll-like receptor (TLR) 4 ligand lipopolysaccharide to a similar extent compared with that of their counterparts, conventionally generated with IL-4 (IL-4 DCs). The mechanism of tumor cell killing depends on the induction of iNOS expression by DCs. In contrast, interferon (IFN)-γ induces the cytotoxic activity of IL-4 but not IL-15 DCs. Although the IFN-γ-STAT-1 signaling pathway is overall functional in IL-15 DCs, IFN-γ fails to induce iNOS expression in these cells. iNOS expression is negatively controlled in IFN-γ-stimulated IL-15 DCs by the cooperation between the E3 SUMO ligase PIAS1 and STAT-3, and can be partially restored with PIAS1 siRNA and STAT-3 inhibitors.
Collapse
Affiliation(s)
- Neale T Hanke
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Collin J LaCasse
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Claire B Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America
| | - Darya Alizadeh
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America
| | - Malika Trad
- INSERM UMR 1098, Faculty of Medicine, Dijon, France
| | | | | | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America.,Department of Immunobiology, BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
| | - Nicolas Larmonier
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States of America.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States of America.,Department of Immunobiology, BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
25
|
Heinrich B, Goepfert K, Delic M, Galle PR, Moehler M. Influence of the oncolytic parvovirus H-1, CTLA-4 antibody tremelimumab and cytostatic drugs on the human immune system in a human in vitro model of colorectal cancer cells. Onco Targets Ther 2013; 6:1119-27. [PMID: 23986643 PMCID: PMC3754820 DOI: 10.2147/ott.s49371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor-directed and immune-system-stimulating therapies are of special interest in cancer treatment. Here, we demonstrate the potential of parvovirus H-1 (H-1PV) to efficiently kill colorectal cancer cells and induce immunogenicity of colorectal tumors by inducing maturation of dendritic cells (DCs) alone and also in combination with cytostatic drugs in vitro. Using our cell culture model, we have additionally investigated the effects of anti-CTLA-4 (cytotoxic T-lymphocyte-associated antigen 4) receptor antibody tremelimumab on this process. Materials and methods Colon carcinoma cell lines were treated with different concentrations of cytostatic drugs or tremelimumab or were infected with H-1PV in different multiplicities of infection (MOIs), and viability was determined using MTT assays. Expression of CTLA-4 in colon carcinoma cell lines was measured by FACScan™. For the coculture model, we isolated monocytes using adherence, and differentiation into immature DCs (iDCs) was stimulated using interleukin-4 and granulocyte-macrophage colony-stimulating factor. Maturation of iDCs into mature DCs (mDCs) was induced by a cytokine cocktail. SW480 colon carcinoma cells were infected with H-1PV or treated with cytostatic drugs. Drug treated and H-1PV-infected SW480 colon carcinoma cells were cocultured with iDCs and expression of maturation markers was measured using FACScan™. Cytokine measurements were performed using enzyme-linked immunosorbent assay. Results Colon carcinoma cells SW480 were potently infected and killed by H-1PV. CTLA-4 expression in SW480 cells increased after infection with H-1PV and also after treatment with cytostatic drugs. Tremelimumab had no influence on viability of the colon carcinoma cell line. There was no maturation of iDCs after coculture with SW480; instead, H-1PV-infected or drug pretreated SW480 induced maturation. Cytokine production was higher for H-1PV-infected cells but was not significantly enhanced by tremelimumab treatment alone or in combination. Addition of tremelimumab did not interfere with the maturation process as measured by markers of maturation as well as by determination of cytokine levels. Conclusion By enhancing both cell death and immunogenicity of tumors, H-1PV is of special interest for tumor-directed therapy. These features make it a promising candidate for clinical application in human colorectal cancer. As tremelimumab does not significantly interfere with this process, an interesting therapeutic combination of active enhancement of tumor immunogenicity and independent masking of the CTLA-4 silencing process on tumor cells is highlighted.
Collapse
Affiliation(s)
- Bernd Heinrich
- University Medical Center of the Johannes Gutenberg University Mainz, 1st Department of Internal Medicine, Langenbeckstrasse, Mainz, Germany
| | | | | | | | | |
Collapse
|
26
|
Anguille S, Lion E, Tel J, de Vries IJM, Couderé K, Fromm PD, Van Tendeloo VF, Smits EL, Berneman ZN. Interleukin-15-induced CD56(+) myeloid dendritic cells combine potent tumor antigen presentation with direct tumoricidal potential. PLoS One 2012; 7:e51851. [PMID: 23284789 PMCID: PMC3532168 DOI: 10.1371/journal.pone.0051851] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/07/2012] [Indexed: 11/18/2022] Open
Abstract
Dendritic cells (DCs) are the quintessential antigen-presenting cells of the human immune system and play a prime role in coordinating innate and adaptive immune responses, explaining the strong and still growing interest in their application for cancer immunotherapy. Much current research in the field of DC-based immunotherapy focuses on optimizing the culture conditions for in vitro DC generation in order to assure that DCs with the best possible immunogenic qualities are being used for immunotherapy. In this context, monocyte-derived DCs that are alternatively induced by interleukin-15 (IL-15 DCs) have attracted recent attention due to their superior immunostimulatory characteristics. In this study, we show that IL-15 DCs, in addition to potent tumor antigen-presenting function, possess tumoricidal potential and thus qualify for the designation of killer DCs. Notwithstanding marked expression of the natural killer (NK) cell marker CD56 on a subset of IL-15 DCs, we found no evidence of a further phenotypic overlap between IL-15 DCs and NK cells. Allostimulation and antigen presentation assays confirmed that IL-15 DCs should be regarded as bona fide myeloid DCs not only from the phenotypic but also from the functional point of view. Concerning their cytotoxic activity, we demonstrate that IL-15 DCs are able to induce apoptotic cell death of the human K562 tumor cell line, while sparing tumor antigen-specific T cells. The cytotoxicity of IL-15 DCs is predominantly mediated by granzyme B and, to a small extent, by tumor necrosis factor-α (TNF-α)-related apoptosis-inducing ligand (TRAIL) but is independent of perforin, Fas ligand and TNF-α. In conclusion, our data provide evidence of a previously unappreciated role for IL-15 in the differentiation of human monocytes towards killer DCs. The observation that IL-15 DCs have killer DC capacity lends further support to their implementation in DC-based immunotherapy protocols.
Collapse
Affiliation(s)
- Sébastien Anguille
- University of Antwerp, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), Laboratory of Experimental Hematology, Antwerp, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sieben M, Schäfer P, Dinsart C, Galle PR, Moehler M. Activation of the human immune system via toll-like receptors by the oncolytic parvovirus H-1. Int J Cancer 2012; 132:2548-56. [PMID: 23151948 DOI: 10.1002/ijc.27938] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 10/05/2012] [Indexed: 01/09/2023]
Abstract
This study aimed to investigate the function of toll-like receptors (TLRs) during oncolytic parvovirus H-1 (H-1PV)-induced human immune responses. First, the role of TLRs in the activation of the NFκB transcription factor was characterized; second, the immunologic effects of H-1PV-induced tumor cell lysates (TCL) on human antitumor immune responses were evaluated. A human ex vivo model was used to study immune responses with dendritic cells (DCs). Human embryonic kidney cells (HEK293) transfected to stably express TLRs were used as potential human DC equivalents to further investigate the role of specific TLRs during immune activation. TLR3 and TLR9 were activated by H-1PV infection, which correlated with NFκB translocation to the nucleus and a reduced cytoplasmic IκB expression. Using a TLR-signaling reporter plasmid (pNiFty-Luc), NFκB activity was increased following H-1PV infection. In addition, human DCs coincubated with H-1PV-induced TCL demonstrated increased TLR3 and TLR9 expression. These data suggest that H-1PV-induced TCL stimulate human DCs at least in part through TLR-dependent signaling pathways. Thus, DC maturation occurred through exposure to H-1PV-induced TCL through TLR-signaling leading to NFκB-dependent activation of the adaptive immune system as indicated by the increased expression of CD86, TLR3 and TLR9. Furthermore, the transcription of various cytokines indicates the activation of immune response, therefore the production of the proinflammatory cytokine TNF-α was determined. Here, H-1PV-induced TCL significantly enhanced the TNF-α level by DCs after coculture. H-1PV oncolytic virotherapy enhances immune priming by different effects on DCs and generates antitumor immunity. These findings potentially offer a new approach to tumor therapy.
Collapse
Affiliation(s)
- Maike Sieben
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | | | | | | |
Collapse
|
28
|
Cancer Immunotherapy by Retargeting of Immune Effector Cells via Recombinant Bispecific Antibody Constructs. Antibodies (Basel) 2012. [DOI: 10.3390/antib1020172] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
29
|
Abstract
Senescence is a stable cell cycle arrest program that contributes to tumor suppression, organismal aging and certain wound healing responses. During liver fibrosis, for example, hepatic stellate cells initially proliferate and secrete extracellular matrix components that produce fibrosis; however, these cells eventually senesce and are cleared by immune cells, including natural killer (NK) cells. Here, we examine how NK cells target senescent cells and assess the impact of this process on liver fibrosis. We show that granule exocytosis, but not death-receptor-mediated apoptosis, is required for NK-cell-mediated killing of senescent cells. This pathway bias is due to upregulation of the decoy death receptor, Dcr2, an established senescence marker that attenuates NK-mediated cell death. Accordingly, mice with defects in granule exocytosis accumulate senescent stellate cells and display more liver fibrosis in response to a fibrogenic agent. Our results thus provide new insights into the immune surveillance of senescent cells and reveal how granule exocytosis has a protective role against liver fibrosis.
Collapse
|
30
|
Activation of the human immune system by chemotherapeutic or targeted agents combined with the oncolytic parvovirus H-1. BMC Cancer 2011; 11:464. [PMID: 22029859 PMCID: PMC3234202 DOI: 10.1186/1471-2407-11-464] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/26/2011] [Indexed: 01/08/2023] Open
Abstract
Background Parvovirus H-1 (H-1PV) infects and lyses human tumor cells including melanoma, hepatoma, gastric, colorectal, cervix and pancreatic cancers. We assessed whether the beneficial effects of chemotherapeutic agents or targeted agents could be combined with the oncolytic and immunostimmulatory properties of H-1PV. Methods Using human ex vivo models we evaluated the biological and immunological effects of H-1PV-induced tumor cell lysis alone or in combination with chemotherapeutic or targeted agents in human melanoma cells +/- characterized human cytotoxic T-cells (CTL) and HLA-A2-restricted dendritic cells (DC). Results H-1PV-infected MZ7-Mel cells showed a clear reduction in cell viability of >50%, which appeared to occur primarily through apoptosis. This correlated with viral NS1 expression levels and was enhanced by combination with chemotherapeutic agents or sunitinib. Tumor cell preparations were phagocytosed by DC whose maturation was measured according to the treatment administered. Immature DC incubated with H-1PV-induced MZ7-Mel lysates significantly increased DC maturation compared with non-infected or necrotic MZ7-Mel cells. Tumor necrosis factor-α and interleukin-6 release was clearly increased by DC incubated with H-1PV-induced SK29-Mel tumor cell lysates (TCL) and was also high with DC-CTL co-cultures incubated with H-1PV-induced TCL. Similarly, DC co-cultures with TCL incubated with H-1PV combined with cytotoxic agents or sunitinib enhanced DC maturation to a greater extent than cytotoxic agents or sunitinib alone. Again, these combinations increased pro-inflammatory responses in DC-CTL co-cultures compared with chemotherapy or sunitinib alone. Conclusions In our human models, chemotherapeutic or targeted agents did not only interfere with the pronounced immunomodulatory properties of H-1PV, but also reinforced drug-induced tumor cell killing. H-1PV combined with cisplatin, vincristine or sunitinib induced effective immunostimulation via a pronounced DC maturation, better cytokine release and cytotoxic T-cell activation compared with agents alone. Thus, the clinical assessment of H-1PV oncolytic tumor therapy not only alone but also in combination strategies is warranted.
Collapse
|
31
|
Jensen TO, Schmidt H, Møller HJ, Donskov F, Høyer M, Sjoegren P, Christensen IJ, Steiniche T. Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer 2011; 118:2476-85. [PMID: 21953023 DOI: 10.1002/cncr.26511] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Revised: 05/01/2011] [Accepted: 06/14/2011] [Indexed: 11/08/2022]
Abstract
BACKGROUND Tumor cell and host immune cell interaction plays a key role in carcinogenesis. Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in cancer and believed to be an important mediator of tumor-induced immunosuppression. This paper aims to describe the prognostic impact of neutrophil and dendritic cell infiltration in primary melanoma and the association of this infiltration with activated STAT3 (pSTAT3) in primary melanoma cells. METHODS Formalin-fixed, paraffin-embedded primary melanomas from 186 stage-I/II melanoma patients surgically resected from 1997 to 2000. Infiltrating neutrophils (CD66b), dendritic cells (CD123+ and DC-LAMP+), T-lymphocytes (CD8) and pSTAT3 melanoma cell expression were studied by immunohistochemistry and evaluated as present or absent. DC-LAMP+ cell infiltration was evaluated as absent/few versus dense. Study endpoints: relapse-free survival, melanoma-specific, and overall survival. RESULTS The median observation time was 12.2 years (range, 10.4-14.2 years). Fifty-one deaths were observed of which 38 (20%) were melanoma-specific. In a multivariate Cox proportional hazards model including ulceration and melanoma thickness, neutrophil and CD123+ dendritic cell infiltration were independently associated with poor prognosis (CD66b: hazard ratio [HR] = 3.13; 95% confidence interval [CI], 1.43-6.83; P = .004; CD123: HR = 2.45; 95% CI, 1.22-4.92; P = .012). The association between melanoma cell pSTAT3 expression and immune infiltration (neutrophils and CD123+ cells) was strong. pSTAT3 expression, CD8 and DC-LAMP infiltration were not independently associated with poor prognosis. CONCLUSIONS Neutrophil infiltration and CD123+ dendritic cell infiltration in primary melanoma are independently associated with poor prognosis. Melanoma cell expression of pSTAT3 is strongly associated with the surrounding immune infiltrate.
Collapse
Affiliation(s)
- Trine O Jensen
- Cancer Immunotherapy Group, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kees T, Egeblad M. Innate immune cells in breast cancer--from villains to heroes? J Mammary Gland Biol Neoplasia 2011; 16:189-203. [PMID: 21789554 DOI: 10.1007/s10911-011-9224-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 07/18/2011] [Indexed: 12/13/2022] Open
Abstract
The innate immune system ensures effective protection against foreign pathogens and plays important roles in tissue remodeling. There are many types of innate immune cells, including monocytes, macrophages, dendritic cells, and granulocytes. Interestingly, these cells accumulate in most solid tumors, including those of the breast. There, they play a tumor-promoting role through secretion of growth and angiogenic factors, as well as immunosuppressive molecules. This is in strong contrast to the tumor-suppressing effects that innate immune cells exert in vitro upon proper activation. Therapeutic approaches have been developed with the aim of achieving similar suppressive activities in vivo. However, multiple factors in the tumor microenvironment, many of which are immunosuppressive, represent a major obstacle to effective treatment. Here, we discuss the potential of combating breast cancer through activation of the innate immune system, including possible strategies to enhance the success of immunotherapy.
Collapse
Affiliation(s)
- Tim Kees
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
33
|
Dong LF, Jameson VJA, Tilly D, Prochazka L, Rohlena J, Valis K, Truksa J, Zobalova R, Mahdavian E, Kluckova K, Stantic M, Stursa J, Freeman R, Witting PK, Norberg E, Goodwin J, Salvatore BA, Novotna J, Turanek J, Ledvina M, Hozak P, Zhivotovsky B, Coster MJ, Ralph SJ, Smith RAJ, Neuzil J. Mitochondrial targeting of α-tocopheryl succinate enhances its pro-apoptotic efficacy: a new paradigm for effective cancer therapy. Free Radic Biol Med 2011; 50:1546-55. [PMID: 21402148 DOI: 10.1016/j.freeradbiomed.2011.02.032] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 02/16/2011] [Accepted: 02/25/2011] [Indexed: 12/20/2022]
Abstract
Mitochondria are emerging as intriguing targets for anti-cancer agents. We tested here a novel approach, whereby the mitochondrially targeted delivery of anti-cancer drugs is enhanced by the addition of a triphenylphosphonium group (TPP(+)). A mitochondrially targeted analog of vitamin E succinate (MitoVES), modified by tagging the parental compound with TPP(+), induced considerably more robust apoptosis in cancer cells with a 1-2 log gain in anti-cancer activity compared to the unmodified counterpart, while maintaining selectivity for malignant cells. This is because MitoVES associates with mitochondria and causes fast generation of reactive oxygen species that then trigger mitochondria-dependent apoptosis, involving transcriptional modulation of the Bcl-2 family proteins. MitoVES proved superior in suppression of experimental tumors compared to the untargeted analog. We propose that mitochondrially targeted delivery of anti-cancer agents offers a new paradigm for increasing the efficacy of compounds with anti-cancer activity.
Collapse
Affiliation(s)
- Lan-Feng Dong
- School of Medical Science, Griffith University, Southport, QLD 4222, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ramdial PK, Sing Y, Naicker S, Calonje E, Sewram V, Singh B. Langerhans cells in anaplastic Kaposi sarcoma with a paucivascular phenotype: A potential diagnostic pitfall. Pathol Int 2011; 61:221-7. [DOI: 10.1111/j.1440-1827.2011.02658.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Cytokeratin-Positive Cells (CK+) as Potential Dendritic Cells. FOOTMARKS OF INNATE IMMUNITY IN THE OVARY AND CYTOKERATIN-POSITIVE CELLS AS POTENTIAL DENDRITIC CELLS 2011. [DOI: 10.1007/978-3-642-16077-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
36
|
Pajtasz-Piasecka E, Indrová M. Dendritic cell-based vaccines for the therapy of experimental tumors. Immunotherapy 2010; 2:257-68. [PMID: 20635932 DOI: 10.2217/imt.10.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are believed to be the most potent antigen-presenting cells able to link the innate and adaptive immune systems. Many studies have focused on different immunotherapeutic approaches to applying DCs as tools to improve anticancer therapy. Although a number of investigations suggesting the benefit of DC-based vaccination during anticancer therapy have been reported, the general knowledge regarding the ultimate methods of DC-vaccine preparation is still unsatisfactory. In this article, the perspectives of DC-based anti-tumor immunotherapy and optimizing strategies of DC vaccination in humans in light of results obtained in mouse models are discussed.
Collapse
Affiliation(s)
- Elzbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.
| | | |
Collapse
|
37
|
Wang PH, Shen TY, Ge HY. Role of PA-MSHA vaccine in adjuvant therapy for malignant tumors. Shijie Huaren Xiaohua Zazhi 2010; 18:3171-3174. [DOI: 10.11569/wcjd.v18.i30.3171] [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
Nowadays, malignant tumors have increasingly tended to be treated in a comprehensive manner. Pseudomonas aeruginosa-mannose sensitive hemagglutinin (PA-MSHA) vaccine is a class of immunotherapeutic agents that are being trialed for tumor treatment. This paper focuses on the research on the effect of PA-MSHA vaccine in adjuvant therapy for malignant tumors.
Collapse
|
38
|
Dendritic Cells Generated in Clinical Grade Bags Strongly Differ in Immune Functionality When Compared With Classical DCs Generated in Plates. J Immunother 2010; 33:352-63. [DOI: 10.1097/cji.0b013e3181cc266b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
39
|
Larmonier N, Fraszczak J, Lakomy D, Bonnotte B, Katsanis E. Killer dendritic cells and their potential for cancer immunotherapy. Cancer Immunol Immunother 2010; 59:1-11. [PMID: 19618185 PMCID: PMC11031008 DOI: 10.1007/s00262-009-0736-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 07/01/2009] [Indexed: 12/25/2022]
Abstract
Known for years as the principal messengers of the immune system, dendritic cells (DC) represent a heterogeneous population of antigen presenting cells critically located at the nexus between innate and adaptive immunity. DC play a central role in the initiation of tumor-specific immune responses as they are endowed with the unique ability to take up, process and present tumor antigens to naïve CD4(+) or CD8(+) effector T lymphocytes. By virtue of the cytokines they produce, DC also regulate the type, strength and duration of T cell immune responses. In addition, they can participate in anti-tumoral NK and NKT cell activation and in the orchestration of humoral immunity. More recent studies have documented that besides their primary role in the induction and regulation of adaptive anti-tumoral immune responses, DC are also endowed with the capacity to directly kill cancer cells. This dual role of DC as killers and messengers may have important implications for tumor immunotherapy. First, the direct killing of malignant cells by DC may foster the release and thereby the immediate availability of specific tumor antigens for presentation to cytotoxic or helper T lymphocytes. Second, DC may participate in the effector phase of the immune response, potentially augmenting the diversity of the killing mechanisms leading to tumor elimination. This review focuses on this non-conventional cytotoxic function of DC as it relates to the promotion of cancer immunity and discusses the potential application of killer DC (KDC) in tumor immunotherapy.
Collapse
Affiliation(s)
- Nicolas Larmonier
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona, 1501 N. Campbell Ave., PO Box 245073, Tucson, AZ 85724-5073 USA
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724 USA
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724 USA
| | | | - Daniela Lakomy
- Faculty of Medicine, INSERM UMR 866, IFR 100, Dijon, France
| | | | - Emmanuel Katsanis
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona, 1501 N. Campbell Ave., PO Box 245073, Tucson, AZ 85724-5073 USA
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724 USA
- BIO5 Institute and Arizona Cancer Center, University of Arizona, Tucson, AZ 85724 USA
| |
Collapse
|
40
|
Al-Shibli K, Al-Saad S, Donnem T, Persson M, Bremnes RM, Busund LT. The prognostic value of intraepithelial and stromal innate immune system cells in non-small cell lung carcinoma. Histopathology 2009; 55:301-12. [PMID: 19723145 DOI: 10.1111/j.1365-2559.2009.03379.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS The major value of prognostic markers in potentially curable non-small cell lung carcinoma (NSCLC) should be to guide therapy after surgical resection. The prognostic significance of tumour-infiltrating macrophages, their growth factor, macrophage colony-stimulating factor (M-CSF), and its receptor, colony-stimulating factor-1 receptor (CSF-1R), as well as natural killer cells and dendritic cells, is controversial. The aim of this study was to elucidate the prognostic significance of these markers in the epithelial and stromal compartments of NSCLC. METHODS AND RESULTS Tissue microarrays from 335 resected NSCLC, stage I-IIIA were constructed from duplicate cores of epithelial and stromal areas. Immunohistochemistry was used to evaluate epithelial and stromal areas for CD68, M-CSF, CSF-1R, CD56 and CD1a. On univariate analysis, increasing numbers of stromal CD1a+ (P = 0.011) and CD56+ cells (P = 0.014) correlated significantly with improved disease-specific survival (DSS). On multivariate analysis, stromal CD56+ cells were an independent prognostic factor for DSS (hazard ratio = 2.3, confidence interval = 1.1, 5.0, P = 0.031). CONCLUSIONS High density of stromal CD56+ cells is an independent factor associated with improved prognosis in resected NSCLC, suggesting that these cells mediate an antitumour immune response in the tumour stroma.
Collapse
Affiliation(s)
- Khalid Al-Shibli
- Department of Pathology, Nordland Central Hospital, Bodo 8092, Norway.
| | | | | | | | | | | |
Collapse
|
41
|
Pletneva M, Fan H, Park JJ, Radojcic V, Jie C, Yu Y, Chan C, Redwood A, Pardoll D, Housseau F. IFN-producing killer dendritic cells are antigen-presenting cells endowed with T-cell cross-priming capacity. Cancer Res 2009; 69:6607-14. [PMID: 19679552 DOI: 10.1158/0008-5472.can-09-0508] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IFN-producing killer dendritic cells (IKDC) represent a recently discovered cell type in the immune system that possesses a number of functions contributing to innate and adaptive immunity, including production of type 1 and 2 IFNs, interleukin (IL)-12, natural killing, and ultimately antigen presentation to naïve T cells. Here, we compared in vitro and in vivo responses of mouse IKDC, conventional dendritic cells (DC), and natural killer (NK) cells to murine cytomegalovirus infection and found distinct functions among these cell subsets. Upon recognition of infected fibroblasts, IKDC, as well as NK, produced high level of IFN-gamma, but unlike NK, IKDC simultaneously produced IL-12p40 and up-regulated MHC class II (MHC-II) and costimulatory molecules. Using MHC-II molecule expression as a phenotypic marker to distinguish activated IKDC from activated NK, we further showed that highly purified MHC-II(+) IKDC but not NK cross-present MHC class I-restricted antigens derived from MCMV-infected targets to CD8(+) T cells in vitro and in vivo. Our findings emphasize the unique nature of IKDC as a killer antigen-presenting cell directly linking innate and adaptive immunity.
Collapse
Affiliation(s)
- Maria Pletneva
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gi M, Im W, Hong S. Dendritic cells as danger-recognizing biosensors. SENSORS 2009; 9:6730-51. [PMID: 22399974 PMCID: PMC3290479 DOI: 10.3390/s90906730] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 08/19/2009] [Accepted: 08/24/2009] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are antigen presenting cells that are characterized by a potent capacity to initiate immune responses. DCs comprise several subsets with distinct phenotypes. After sensing any danger(s) to the host via their innate immune receptors such as Toll-like receptors, DCs become mature and subsequently present antigens to CD4+ T cells. Since DCs possess the intrinsic capacity to polarize CD4+ helper cells, it is critical to understand the immunological roles of DCs for clinical applications. Here, we review the different DC subsets, their danger-sensing receptors and immunological functions. Furthermore, the cytokine reporter mouse model for studying DC activation is introduced.
Collapse
Affiliation(s)
- Mia Gi
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Seoul, 143-747, Korea; E-Mails: (M.G.); (W.I.)
| | | | | |
Collapse
|
43
|
Zhao Y, Li R, Xia W, Neuzil J, Lu Y, Zhang H, Zhao X, Zhang X, Sun C, Wu K. Bid integrates intrinsic and extrinsic signaling in apoptosis induced by alpha-tocopheryl succinate in human gastric carcinoma cells. Cancer Lett 2009; 288:42-9. [PMID: 19640637 DOI: 10.1016/j.canlet.2009.06.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2009] [Revised: 06/04/2009] [Accepted: 06/22/2009] [Indexed: 12/13/2022]
Abstract
The underlying mechanisms of alpha-tocopheryl succinate (alpha-TOS)-mediated apoptosis are not understood in detail, although the redox-silent vitamin E analog is a potent apoptogen and anti-cancer agent. Our previous studies showed the important role of Fas signaling in apoptosis induced by the mitocan. The objective of the present study was to investigate whether apoptosis triggered by alpha-TOS in gastric carcinomas cells involves both mitochondria- and death receptor-dependent pathways. alpha-TOS induced apoptosis and mitochondrial permeability transition in a concentration- and time-dependent manner. As a consequence, cytochrome c and the apoptosis-inducing factor were released and caspases were activated. Bax was translocated from the cytosol to mitochondria and Bid was cleaved into its truncated form, tBid. Knocking down Bid by RNAi and Fas antisense oligodeoxynucleotides resulted in a decreased release and cleavage. The results imply that Bid may serve as a critical integrating factor of the death receptor and mitochondrial pathway in alpha-TOS-mediated apoptosis.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Nutrition and Food, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hua G, Wang S, Zhong C, Xue P, Fan Z. Ignition of p53 bomb sensitizes tumor cells to granzyme K-mediated cytolysis. THE JOURNAL OF IMMUNOLOGY 2009; 182:2152-9. [PMID: 19201868 DOI: 10.4049/jimmunol.0802307] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inactivation of tumor suppressor p53 results in loss of the apoptosis-regulating function of the p53 protein in tumor cells. Restoration of wild-type p53 expression in p53 mutant tumor cells increases tumor susceptibility to CTL-mediated cytolysis. However, the direct role of p53 in regulating tumor sensitivity to NK cell-mediated lysis and the functional relationship between p53 and granzymes in the control of tumor killing are still poorly documented. In this study, we found that p53 can sensitize tumor-killing susceptibility to NK and granzyme K-mediated cytolysis. Granzyme K is constitutively expressed in high levels in NK cells and induces rapid caspase-independent cell death. Granzyme K may exert a critical role in NK cell-mediated tumor clearance. p53 associates with granzyme K and is a physiological substrate of granzyme K. p53 was processed to three cleavage products of p40, p35, and p13 fragments at Lys(24) and Lys(305). These three cleavage products harbor strong proapoptotic activities that amplify the proapoptotic action of p53 to potentiate tumor-killing sensitivity. Therefore, p53 is as a cytotoxic bomb that can be triggered by granzyme K, leading to potentiating killing efficacy.
Collapse
Affiliation(s)
- Guoqiang Hua
- National Laboratory of Biomacromolecules and Center for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | |
Collapse
|
45
|
Burton DGA. Cellular senescence, ageing and disease. AGE (DORDRECHT, NETHERLANDS) 2009; 31:1-9. [PMID: 19234764 PMCID: PMC2645988 DOI: 10.1007/s11357-008-9075-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 07/29/2008] [Indexed: 05/27/2023]
Abstract
Cellular senescence is the irreversible growth arrest of individual mitotic cells, which as a consequence display a radically altered phenotype that is thought to impair tissue function and predispose tissues to disease development and/or progression as they gradually accumulate. However, in the past, research into mechanisms of ageing has commonly been researched and treated separately from disease development. This may partly be due to the lack of understanding concerning mechanisms of ageing and the difficulty in implementing what was known into models of disease development. Only in the last 10 years, with increasing knowledge of the senescent phenotype and the ability to detect senescent cells in human tissues, have biologists been able to investigate the relationship between cellular senescence and disease. This review therefore brings together and discusses recent findings which suggest that cellular senescence does contribute to ageing and the development/progression of disease.
Collapse
Affiliation(s)
- D G A Burton
- School of Pharmacy and Biomolecular Sciences, Cockcroft Building, University of Brighton, Brighton, East Sussex, BN2 4GJ, UK.
| |
Collapse
|
46
|
Zobalova R, McDermott L, Stantic M, Prokopova K, Dong LF, Neuzil J. CD133-positive cells are resistant to TRAIL due to up-regulation of FLIP. Biochem Biophys Res Commun 2008; 373:567-71. [DOI: 10.1016/j.bbrc.2008.06.073] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 06/18/2008] [Indexed: 12/22/2022]
|
47
|
Zitvogel L, Kroemer G. The immune response against dying tumor cells: avoid disaster, achieve cure. Cell Death Differ 2008; 15:1-2. [PMID: 18084310 DOI: 10.1038/sj.cdd.4402267] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|