1
|
Lim YJ, Lim MS, Lee JJ, Bae H, Baek YJ, Kim GS, An Y, Kim SK, Yu D. Evaluation of clinical and immunological responses to recombinant canine interleukin-15 therapy in dogs with cancer: A pilot study. Vet Immunol Immunopathol 2025; 283:110923. [PMID: 40203669 DOI: 10.1016/j.vetimm.2025.110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
Interleukin-15 (IL-15) is a pleiotropic cytokine that plays a pivotal role in innate and adaptive immunity. Therefore, it is a promising therapeutic agent for cancer treatment. Despite growing interest in the use of IL-15 as an immunotherapeutic agent, there have been very few reports on its immunological and clinical effects in canine cancers. In this study, we generated recombinant canine IL-15 (rcIL-15) and evaluated its clinical and immunomodulatory effects in combination with metronomic cyclophosphamide in 15 canines with various tumor types. The treatment outcomes were assessed in a prospective clinical trial. Low-dose cyclophosphamide (12.5 mg/m2, PO, SID) was continuously administered for 8 weeks. Starting on day 14, after administering cyclophosphamide, rcIL-15 (20 μg/kg daily) was injected intravenously for 8 days. The disease control rate for combination therapy was 66.6 %, with the most notable partial response accounting for 33.3 % of hematological malignancies. The adverse events were minimal and primarily of grade 1 severity. Moreover, rcIL-15 administration led to significant elevations in anticancer lymphocyte subsets, such as natural killer and cytotoxic T cells, along with increased Ki-67 expression, indicating cellular proliferation. These changes were correlated with improved clinical outcomes. Our findings underscore the therapeutic potential and safety of combining rcIL-15 and metronomic cyclophosphamide for the treatment of various canine cancers.
Collapse
Affiliation(s)
- Y J Lim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - M S Lim
- Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea.
| | - J J Lee
- Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.
| | - H Bae
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea.
| | - Y J Baek
- Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - G S Kim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - Y An
- Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - S K Kim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea; Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - D Yu
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
2
|
Subhi-Issa N, Tovar Manzano D, Pereiro Rodriguez A, Sanchez Ramon S, Perez Segura P, Ocaña A. γδ T Cells: Game Changers in Immune Cell Therapy for Cancer. Cancers (Basel) 2025; 17:1063. [PMID: 40227601 PMCID: PMC11987767 DOI: 10.3390/cancers17071063] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 04/15/2025] Open
Abstract
Gamma delta (γδ) T cells are a unique subset of T lymphocytes with distinctive features that make them highly promising candidates for cancer therapy. Their MHC-independent recognition of tumor antigens, ability to mediate direct cytotoxicity, and role in modulating the tumor microenvironment position them as versatile agents in cancer immunotherapy. This review integrates and synthesizes the existing data on γδ T cells, with an emphasis on the development and optimization of in vitro expansion protocols. Critical aspects are detailed such as activation strategies, co-culture systems, cytokine use, and other parameters to ensure robust cell proliferation and functionality, which may be valuable for those developing or optimizing clinical practices. Finally, we discuss current advancements in γδ T cell research, clinical experience, and highlight areas needing further exploration. Considering these data, we hypothesize and propose potential new applications such as engineering γδ T cells for enhanced resistance to immune checkpoint pathways or for localized cytokine delivery within the tumor microenvironment, which could broaden their therapeutic impact in the treatment of cancer and beyond.
Collapse
Affiliation(s)
- Nabil Subhi-Issa
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - Daniel Tovar Manzano
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | | | - Silvia Sanchez Ramon
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology, and ORL, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Pedro Perez Segura
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| | - Alberto Ocaña
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| |
Collapse
|
3
|
Rani D, Kaur S, Shahjahan, Dey JK, Dey SK. Engineering immune response to regulate cardiovascular disease and cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:381-417. [PMID: 38762276 DOI: 10.1016/bs.apcsb.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Cardiovascular disease (CVD) and cancer are major contributors to global morbidity and mortality. This book chapter delves into the intricate relationship between the immune system and the pathogenesis of both cardiovascular and cancer diseases, exploring the roles of innate and adaptive immunities, immune regulation, and immunotherapy in these complex conditions. The innate immune system acts as the first line of defense against tissue damage and infection, with a significant impact on the initiation and progression of CVD and cancer. Endothelial dysfunction, a hallmark in CVD, shares commonalities with the tumor microenvironment in cancer, emphasizing the parallel involvement of the immune system in both conditions. The adaptive immune system, particularly T cells, contributes to prolonged inflammation in both CVD and cancer. Regulatory T cells and the intricate balance between different T cell subtypes influence disease progression, wound healing, and the outcomes of ischemic injury and cancer immunosurveillance. Dysregulation of immune homeostasis can lead to chronic inflammation, contributing to the development and progression of both CVD and cancer. Thus, immunotherapy emerged as a promising avenue for preventing and managing these diseases, with strategies targeting immune cell modulation, cytokine manipulation, immune checkpoint blockade, and tolerance induction. The impact of gut microbiota on CVD and cancer too is explored in this chapter, highlighting the role of gut leakiness, microbial metabolites, and the potential for microbiome-based interventions in cardiovascular and cancer immunotherapies. In conclusion, immunomodulatory strategies and immunotherapy hold promise in reshaping the landscape of cardiovascular and cancer health. Additionally, harnessing the gut microbiota for immune modulation presents a novel approach to prevent and manage these complex diseases, emphasizing the importance of personalized and precision medicine in healthcare. Ongoing research and clinical trials are expected to further elucidate the complex immunological underpinnings of CVD and cancer thereby refining these innovative approaches.
Collapse
Affiliation(s)
- Diksha Rani
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Smaranjot Kaur
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India.
| |
Collapse
|
4
|
Cheng Q, Kang Y, Yao B, Dong J, Zhu Y, He Y, Ji X. Genetically Engineered-Cell-Membrane Nanovesicles for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302131. [PMID: 37409429 PMCID: PMC10502869 DOI: 10.1002/advs.202302131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
The advent of immunotherapy has marked a new era in cancer treatment, offering significant clinical benefits. Cell membrane as drug delivery materials has played a crucial role in enhancing cancer therapy because of their inherent biocompatibility and negligible immunogenicity. Different cell membranes are prepared into cell membrane nanovesicles (CMNs), but CMNs have limitations such as inefficient targeting ability, low efficacy, and unpredictable side effects. Genetic engineering has deepened the critical role of CMNs in cancer immunotherapy, enabling genetically engineered-CMN (GCMN)-based therapeutics. To date, CMNs that are surface modified by various functional proteins have been developed through genetic engineering. Herein, a brief overview of surface engineering strategies for CMNs and the features of various membrane sources is discussed, followed by a description of GCMN preparation methods. The application of GCMNs in cancer immunotherapy directed at different immune targets is addressed as are the challenges and prospects of GCMNs in clinical translation.
Collapse
Affiliation(s)
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jinrui Dong
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yalan Zhu
- Jinhua Municipal Central HospitalJinhua321000China
| | - Yiling He
- Jinhua Municipal Central HospitalJinhua321000China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
5
|
Lee SH, Lim YJ, Kim CJ, Yu D, Lee JJ, Won Hong J, Baek YJ, Jung JY, Shin DJ, Kim SK. Safety and immunological effects of recombinant canine IL-15 in dogs. Cytokine 2021; 148:155599. [PMID: 34103211 DOI: 10.1016/j.cyto.2021.155599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/03/2021] [Accepted: 05/26/2021] [Indexed: 11/19/2022]
Abstract
Interleukin-15 (IL-15) is a pleiotropic cytokine that plays pivotal roles in innate and adaptive immunity. It is also a promising cytokine for treating cancer. Despite growing interest in its use as an immunotherapeutic, its safety and immunological effects in dogs have not been reported. In this study, healthy dogs were given recombinant canine IL-15 (rcIL-15) intravenously at a daily dose of 20 μg/kg for 8 days and monitored for 32 days to determine the safety and immunological effects of rcIL-15. The repeated administration of rcIL-15 was well tolerated, did not cause any serious side effects, and promoted the selective proliferation and activation of canine anti-cancer effector cells, including CD3+CD8+ cytotoxic T lymphocytes, CD3+CD5dimCD21-, and non-B/non-T NK cell populations, without stimulating Treg lymphocytes. The rcIL-15 injections also stimulated the expression of molecules and transcription factors associated with the activation and effector functions of NK cells, including CD16, NKG2D, NKp30, NKp44, NKp46, perforin, granzyme B, Ly49, T-bet, and Eomes. These results suggest that rcIL-15 might be a valuable therapeutic adjuvant to improve immunity against cancer in dogs.
Collapse
Affiliation(s)
- Soo-Hyeon Lee
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Yu-Jin Lim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Cheol-Jung Kim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Dohyeon Yu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Je-Jung Lee
- Department of Hemotology-Oncology, Chonnam National Univresity Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Jeong Won Hong
- Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Yeon-Ju Baek
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Ji-Youn Jung
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Dong-Jun Shin
- Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; SD Medic Co, Gwangju, Republic of Korea.
| | - Sang-Ki Kim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| |
Collapse
|
6
|
Coulibaly A, Velásquez SY, Kassner N, Schulte J, Barbarossa MV, Lindner HA. STAT3 governs the HIF-1α response in IL-15 primed human NK cells. Sci Rep 2021; 11:7023. [PMID: 33782423 PMCID: PMC8007797 DOI: 10.1038/s41598-021-84916-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/01/2021] [Indexed: 02/01/2023] Open
Abstract
Natural killer (NK) cells mediate innate host defense against microbial infection and cancer. Hypoxia and low glucose are characteristic for these tissue lesions but do not affect early interferon (IFN) γ and CC chemokine release by interleukin 15 (IL-15) primed human NK cells in vitro. Hypoxia inducible factor 1α (HIF-1α) mediates cellular adaption to hypoxia. Its production is supported by mechanistic target of rapamycin complex 1 (mTORC1) and signal transducer and activator of transcription 3 (STAT3). We used chemical inhibition to probe the importance of mTORC1 and STAT3 for the hypoxia response and of STAT3 for the cytokine response in isolated and IL-15 primed human NK cells. Cellular responses were assayed by magnetic bead array, RT-PCR, western blotting, flow cytometry, and metabolic flux analysis. STAT3 but not mTORC1 activation was essential for HIF-1α accumulation, glycolysis, and oxygen consumption. In both primed normoxic and hypoxic NK cells, STAT3 inhibition reduced the secretion of CCL3, CCL4 and CCL5, and it interfered with IL-12/IL-18 stimulated IFNγ production, but it did not affect cytotoxic granule degranulation up on target cell contact. We conclude that IL-15 priming promotes the HIF-1α dependent hypoxia response and the early cytokine response in NK cells predominantly through STAT3 signaling.
Collapse
Affiliation(s)
- Anna Coulibaly
- grid.7700.00000 0001 2190 4373Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Sonia Y. Velásquez
- grid.7700.00000 0001 2190 4373Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nina Kassner
- grid.7700.00000 0001 2190 4373Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Jutta Schulte
- grid.7700.00000 0001 2190 4373Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Maria Vittoria Barbarossa
- grid.7700.00000 0001 2190 4373Interdisciplinary Center for Scientific Computing, Heidelberg University, 69120 Heidelberg, Germany ,grid.417999.bFrankfurt Institute of Advanced Studies, 60438 Frankfurt, Germany
| | - Holger A. Lindner
- grid.7700.00000 0001 2190 4373Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
7
|
Giehl E, Kosaka H, Liu Z, Feist M, Kammula US, Lotze MT, Ma C, Guo ZS, Bartlett DL. In Vivo Priming of Peritoneal Tumor-Reactive Lymphocytes With a Potent Oncolytic Virus for Adoptive Cell Therapy. Front Immunol 2021; 12:610042. [PMID: 33679747 PMCID: PMC7930493 DOI: 10.3389/fimmu.2021.610042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/04/2021] [Indexed: 01/07/2023] Open
Abstract
Adoptive cell therapy (ACT) using autologous tumor infiltrating lymphocytes (TIL) achieves durable clinical benefit for patients from whom these cells can be derived in advanced metastatic melanoma but is limited in most solid tumors as a result of immune escape and exclusion. A tumor microenvironment (TME) priming strategy to improve the quantity and quality of TIL represents an important tactic to explore. Oncolytic viruses expressing immune stimulatory cytokines induce a potent inflammatory response that may enhance infiltration and activation of T cells. In this study, we examined the ability of an attenuated oncolytic vaccinia virus expressing IL15/IL15Rα (vvDD-IL15/Rα) to enhance recovery of lavage T cells in peritoneal carcinomatosis (PC). We found that intraperitoneal (IP) vvDD-IL15/Rα treatment of animals bearing PC resulted in a significant increase in cytotoxic function and memory formation in CD8+ T cells in peritoneal fluid. Using tetramers for vaccinia virus B8R antigen and tumor rejection antigen p15E, we found that the expanded population of peritoneal CD8+ T cells are specific for vaccinia or tumor with increased tumor-specificity over time, reinforced with viral clearance. Application of these vvDD-IL15/Rα induced CD8+ T cells in ACT of a lethal model of PC significantly increased survival. In addition, we found in patients with peritoneal metastases from various primary solid tumors that peritoneal T cells could be recovered but were exhausted with infrequent tumor-reactivity. If clinically translatable, vvDD-IL15/Rα in vivo priming would greatly expand the number of patients with advanced metastatic cancers responsive to T cell therapy.
Collapse
Affiliation(s)
- Esther Giehl
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Hiromichi Kosaka
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Oncology Research Laboratories Oncology R&D Unit, Kyowa Kirin Co., Ltd., Shizuoka, Japan
| | - Zuqiang Liu
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Mathilde Feist
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Surgery, CCM/CVK, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Udai S. Kammula
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Michael T. Lotze
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Congrong Ma
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Zong Sheng Guo
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - David L. Bartlett
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Liao YM, Hung TH, Tung JK, Yu J, Hsu YL, Hung JT, Yu AL. Low Expression of IL-15 and NKT in Tumor Microenvironment Predicts Poor Outcome of MYCN-Non-Amplified Neuroblastoma. J Pers Med 2021; 11:jpm11020122. [PMID: 33668573 PMCID: PMC7918138 DOI: 10.3390/jpm11020122] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Immune tumor microenvironment (TME) in neuroblastoma (NBL) contributes to tumor behavior and treatment response. T cells and natural killer (NK) cells have been shown to play important roles in the neuroblastoma TME. However, few reports address the clinical relevance of natural killer T cells (NKTs) and interleukin-15 (IL-15), one of the crucial cytokines controlling the activation and expansion of NK/NKT cells, in NBL. In this study, we examined NKT immunoscores and IL-15 expression in both MYCN-amplified and MYCN-non-amplified NBL to correlate with clinical outcomes such as event-free survival (EFS) and overall survival (OS). From Gene Expression Omnibus (GEO) datasets GSE45480 (n = 643) and GSE49711 (n = 493), we found that NKT immunoscore and IL-15 expression were both significantly lower in MYCN-amplified NBL, and similar results were observed using our clinical NBL samples (n = 53). Moreover, NBL patients (GEO dataset GSE49711 and our clinical samples) with both lower NKT immunoscore and IL-15 expression exhibited decreased EFS and OS regardless of MYCN gene amplification status. Multivariate analysis further showed that the combination of low NKT immunoscore and low IL-15 expression level was an independent prognostic factor for poor EFS and OS in our NBL patients. These findings provide the rationale for the development of strategy to incorporate IL-15 and NKT cell therapy into the treatment regimen for neuroblastoma.
Collapse
Affiliation(s)
- Yu-Mei Liao
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Program in Translational Medicine, Kaohsiung Medical University, Kaohsiung, and Academia Sinica, Taipei 115, Taiwan
- Division of Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
| | - John K. Tung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Correspondence: (J.-T.H.); (A.L.Y.); Tel.: +886-3328-1200 (ext. 7813) (J.-T.H.); +886-3328-1200 (ext. 7805) (A.L.Y.); Fax: +886-3328-1200 (A.L.Y. & J.-T.H.)
| | - Alice L. Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Department of Pediatrics, University of California in San Diego, San Diego, CA 92103, USA
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
- Correspondence: (J.-T.H.); (A.L.Y.); Tel.: +886-3328-1200 (ext. 7813) (J.-T.H.); +886-3328-1200 (ext. 7805) (A.L.Y.); Fax: +886-3328-1200 (A.L.Y. & J.-T.H.)
| |
Collapse
|
9
|
Arora P, Moll JM, Andersen D, Workman CT, Williams AR, Kristiansen K, Brix S. Body fluid from the parasitic worm Ascaris suum inhibits broad-acting pro-inflammatory programs in dendritic cells. Immunology 2020; 159:322-334. [PMID: 31705653 PMCID: PMC7011627 DOI: 10.1111/imm.13151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) are essential for generating T-cell-based immune responses through sensing of potential inflammatory and metabolic cues in the local environment. However, there is still limited insight into the processes defining the resultant DC phenotype, including the type of early transcriptional changes in pro-inflammatory cues towards regulatory or type 2 immune-based cues induced by a variety of exogenous and endogenous molecules. Here we compared the ability of a selected number of molecules to modulate the pro-inflammatory phenotype of lipopolysaccharide (LPS) and interferon-γ (IFN-γ)-stimulated human monocyte-derived DCs towards an anti-inflammatory or regulatory phenotype, including Ascaris suum body fluid [helminth pseudocoelomic fluid (PCF)], the metabolites succinate and butyrate, and the type 2 cytokines thymic stromal lymphopoietin and interleukin-25. Our data show that helminth PCF and butyrate treatment suppress the T helper type 1 (Th1)-inducing pro-inflammatory DC phenotype through induction of different transcriptional programs in DCs. RNA sequencing indicated that helminth PCF treatment strongly inhibited the Th1 and Th17 polarizing ability of LPS + IFN-γ-matured DCs by down-regulating myeloid differentiation primary response gene 88 (MyD88)-dependent and MyD88-independent pathways in Toll-like receptor 4 signaling. By contrast, butyrate treatment had a strong Th1-inhibiting action, and transcripts encoding important gut barrier defending factors such as IL18, IL1B and CXCL8 were up-regulated. Collectively, our results further understanding of how compounds from parasites and gut microbiota-derived butyrate may exert immunomodulatory effects on the host immune system.
Collapse
Affiliation(s)
- Pankaj Arora
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Janne Marie Moll
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Daniel Andersen
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | | | - Andrew R. Williams
- Parasitology and Aquatic PathobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular BiomedicineDepartment of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Susanne Brix
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| |
Collapse
|
10
|
Owen KL, Brockwell NK, Parker BS. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers (Basel) 2019; 11:E2002. [PMID: 31842362 PMCID: PMC6966445 DOI: 10.3390/cancers11122002] [Citation(s) in RCA: 414] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling mediates almost all immune regulatory processes, including those that are involved in tumor cell recognition and tumor-driven immune escape. Antitumor immune responses are largely driven by STAT1 and STAT2 induction of type I and II interferons (IFNs) and the downstream programs IFNs potentiate. Conversely, STAT3 has been widely linked to cancer cell survival, immunosuppression, and sustained inflammation in the tumor microenvironment. The discovery of JAK-STAT cross-regulatory mechanisms, post-translational control, and non-canonical signal transduction has added a new level of complexity to JAK-STAT governance over tumor initiation and progression. Endeavors to better understand the vast effects of JAK-STAT signaling on antitumor immunity have unearthed a wide range of targets, including oncogenes, miRNAs, and other co-regulatory factors, which direct specific phenotypical outcomes subsequent to JAK-STAT stimulation. Yet, the rapidly expanding field of therapeutic developments aimed to resolve JAK-STAT aberrations commonly reported in a multitude of cancers has been marred by off-target effects. Here, we discuss JAK-STAT biology in the context of immunity and cancer, the consequences of pathway perturbations and current therapeutic interventions, to provide insight and consideration for future targeting innovations.
Collapse
Affiliation(s)
- Katie L. Owen
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| | - Natasha K. Brockwell
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| | - Belinda S. Parker
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| |
Collapse
|
11
|
Coulibaly A, Bettendorf A, Kostina E, Figueiredo AS, Velásquez SY, Bock HG, Thiel M, Lindner HA, Barbarossa MV. Interleukin-15 Signaling in HIF-1α Regulation in Natural Killer Cells, Insights Through Mathematical Models. Front Immunol 2019; 10:2401. [PMID: 31681292 PMCID: PMC6805776 DOI: 10.3389/fimmu.2019.02401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/25/2019] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells belong to the first line of host defense against infection and cancer. Cytokines, including interleukin-15 (IL-15), critically regulate NK cell activity, resulting in recognition and direct killing of transformed and infected target cells. NK cells have to adapt and respond in inflamed and often hypoxic areas. Cellular stabilization and accumulation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) is a key mechanism of the cellular hypoxia response. At the same time, HIF-1α plays a critical role in both innate and adaptive immunity. While the HIF-1α hydroxylation and degradation pathway has been recently described with the help of mathematical methods, less is known concerning the mechanistic mathematical description of processes regulating the levels of HIF-1α mRNA and protein. In this work we combine mathematical modeling with experimental laboratory analysis and examine the dynamic relationship between HIF-1α mRNA, HIF-1α protein, and IL-15-mediated upstream signaling events in NK cells from human blood. We propose a system of non-linear ordinary differential equations with positive and negative feedback loops for describing the complex interplay of HIF-1α regulators. The experimental design is optimized with the help of mathematical methods, and numerical optimization techniques yield reliable parameter estimates. The mathematical model allows for the investigation and prediction of HIF-1α stabilization under different inflammatory conditions and provides a better understanding of mechanisms mediating cellular enrichment of HIF-1α. Thanks to the combination of in vitro experimental data and in silico predictions we identified the mammalian target of rapamycin (mTOR), the nuclear factor-κB (NF-κB), and the signal transducer and activator of transcription 3 (STAT3) as central regulators of HIF-1α accumulation. We hypothesize that the regulatory pathway proposed here for NK cells can be extended to other types of immune cells. Understanding the molecular mechanisms involved in the dynamic regulation of the HIF-1α pathway in immune cells is of central importance to the immune cell function and could be a promising strategy in the design of treatments for human inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Anna Coulibaly
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Anja Bettendorf
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Ekaterina Kostina
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany.,Institute for Applied Mathematics, Heidelberg University, Heidelberg, Germany
| | - Ana Sofia Figueiredo
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Sonia Y Velásquez
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans-Georg Bock
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Manfred Thiel
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Holger A Lindner
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
12
|
Beha N, Harder M, Ring S, Kontermann RE, Müller D. IL15-Based Trifunctional Antibody-Fusion Proteins with Costimulatory TNF-Superfamily Ligands in the Single-Chain Format for Cancer Immunotherapy. Mol Cancer Ther 2019; 18:1278-1288. [PMID: 31040163 DOI: 10.1158/1535-7163.mct-18-1204] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/08/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
IL15 and costimulatory receptors of the tumor necrosis superfamily (TNFRSF) have shown great potential to support and drive an antitumor immune response. However, their efficacy as monotherapy is limited. Here, we present the development of a novel format for a trifunctional antibody-fusion protein that combines and focuses the activity of IL15/TNFSF-ligand in a targeting-mediated manner to the tumor site. The previously reported format consisted of a tumor-directed antibody (scFv), IL15 linked to an IL15Rα-fragment (RD), and the extracellular domain of 4-1BBL, where noncovalent trimerization of 4-1BBL into its functional unit led to a homotrimeric molecule with 3 antibody and 3 IL15-RD units. To reduce the size and complexity of the molecule, we have now designed a second format, where 4-1BBL is introduced as single-chain (sc), that is 3 consecutively linked 4-1BBL ectodomains. Thus, a monomeric trifunctional fusion protein presenting only 1 functional unit of each component was generated. Interestingly, the in vitro activity on T-cell stimulation was conserved or even enhanced for the soluble and target-bound molecule, respectively. Also, in a lung tumor mouse model, comparable antitumor effects were observed. Furthermore, corroborating the concept, OX40L and GITRL were also successfully incorporated into the novel single-chain format and the advantage of target-bound trifunctional versus corresponding combined bifunctional fusion proteins demonstrated by measuring T-cell proliferation and cytotoxic potential in vitro and antitumor effects of RD_IL15_scFv_scGITRL in a lung tumor mouse model in vivo Thus, the trifunctional antibody-fusion protein single-chain format constitutes a promising innovative platform for further therapeutic developments.
Collapse
Affiliation(s)
- Nadine Beha
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Markus Harder
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sarah Ring
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
13
|
Yang H, Kureshi R, Spangler JB. Structural Basis for Signaling Through Shared Common γ Chain Cytokines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1172:1-19. [PMID: 31628649 DOI: 10.1007/978-981-13-9367-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The common γ chain (γc) family of hematopoietic cytokines consists of six distinct four α-helix bundle soluble ligands that signal through receptors which include the shared γc subunit to coordinate a wide range of physiological processes, in particular, those related to innate and adaptive immune function. Since the first crystallographic structure of a γc family cytokine/receptor signaling complex (the active Interleukin-2 [IL-2] quaternary complex) was determined in 2005 [1], tremendous progress has been made in the structural characterization of this protein family, transforming our understanding of the molecular mechanisms underlying immune activity. Although many conserved features of γc family cytokine complex architecture have emerged, distinguishing details have been observed for individual cytokine complexes that rationalize their unique functional properties. Much work remains to be done in the molecular characterization of γc family signaling, particularly with regard to intracellular activation events, and looking forward, new technologies in structural biophysics will offer further insight into the biology of cytokine signaling to inform the design of targeted therapeutics for treatment of immune-linked diseases such as cancer, infection, and autoimmune disorders.
Collapse
Affiliation(s)
- Huilin Yang
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rakeeb Kureshi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Liu X, Li Y, Sun X, Muftuoglu Y, Wang B, Yu T, Hu Y, Ma L, Xiang M, Guo G, You C, Gao X, Wei Y. Powerful anti-colon cancer effect of modified nanoparticle-mediated IL-15 immunogene therapy through activation of the host immune system. Theranostics 2018; 8:3490-3503. [PMID: 30026861 PMCID: PMC6037032 DOI: 10.7150/thno.24157] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/20/2018] [Indexed: 02/05/2023] Open
Abstract
Rationale: Colorectal cancer (CRC) is the third most commonly diagnosed cancer around the world. Over the past several years, immunotherapy has demonstrated considerable clinical benefit in CRC therapy, and the number of immunologic therapies for cancer treatment continues to climb each year. Interleukin-15 (IL15), a potent pro-inflammatory cytokine, has emerged as a candidate immunomodulator for the treatment of CRC. Methods: In this study, we developed a novel gene delivery system with a self-assembly method using DOTAP and MPEG-PLA (DMA) to carry pIL15, denoted as DMA-pIL15 which was used to treat tumor-bearing mice. Results: Supernatant from lymphocytes treated with supernatant derived from CT26 cells transfected with DMA-pIL15 inhibited the growth of CT26 cells and induced cell apoptosis in vitro. Treatment of tumor-bearing mice with DMA-pIL15 complex significantly inhibited tumor growth in both subcutaneous and peritoneal models in vivo by inhibiting angiogenesis, promoting apoptosis, and reducing proliferation through activation of the host immune system. Conclusion: The IL-15 plasmid and DMA complex showed promise for treating CRC clinically as an experimental new drug.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
- Department of Radiation Oncology, Cancer Center, Affiliated Hospital of Xuzhou Medical University; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, China
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Yanyan Li
- Department of radiation oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaodong Sun
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | | | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yuzhu Hu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lu Ma
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Mingli Xiang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Gang Guo
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Chao You
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yuquan Wei
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| |
Collapse
|
15
|
Van Acker HH, Campillo-Davo D, Roex G, Versteven M, Smits EL, Van Tendeloo VF. The role of the common gamma-chain family cytokines in γδ T cell-based anti-cancer immunotherapy. Cytokine Growth Factor Rev 2018; 41:54-64. [PMID: 29773448 DOI: 10.1016/j.cytogfr.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/28/2022]
Abstract
Cytokines of the common gamma-chain receptor family, comprising interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15 and IL-21, are vital with respect to organizing and sustaining healthy immune cell functions. Supporting the anti-cancer immune response, these cytokines inspire great interest for their use as vaccine adjuvants and cancer immunotherapies. It is against this background that gamma delta (γδ) T cells, as special-force soldiers and natural contributors of the tumor immunosurveillance, also received a lot of attention the last decade. As γδ T cell-based cancer trials are coming of age, this present review focusses on the effects of the different cytokines of the common gamma-chain receptor family on γδ T cells with respect to boosting γδ T cells as a therapeutic target in cancer immunotherapy. This review also gathers data that IL-15 in particular exhibits key features for augmenting the anti-tumor activity of effector killer γδ T cells whilst overcoming the myriad of immune escape mechanisms used by cancer cells.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
16
|
Discovery of a novel IL-15 based protein with improved developability and efficacy for cancer immunotherapy. Sci Rep 2018; 8:7675. [PMID: 29769573 PMCID: PMC5955975 DOI: 10.1038/s41598-018-25987-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/30/2018] [Indexed: 12/22/2022] Open
Abstract
Interleukin-15 (IL-15) can promote both innate and adaptive immune reactions by stimulating CD8+/CD4+ T cells and natural killer cells (NK) while showing no effect in activating T-regulatory (Treg) cells or inducing activation-associated death among effector T cells and NK cells. Thus, IL-15 is considered as one of the most promising molecules for antitumor immune therapy. To improve the drug-like properties of natural IL-15, we create an IL-15-based molecule, named P22339, with the following characteristics: 1) building a complex of IL-15 and the Sushi domain of IL-15 receptor α chain to enhance the agonist activity of IL-15 via transpresentation; 2) through a rational structure-based design, creating a disulfide bond linking the IL-15/Sushi domain complex with an IgG1 Fc to augment its half-life. P22339 demonstrates excellent developability, pharmacokinetic and pharmacodynamic properties as well as antitumor efficacy in both in vitro assessments and in vivo studies. It significantly suppresses tumor growth and metastasis in rodent models, and activates T effector cells and NK cells in cynomolgus monkey. Overall, these data suggest that P22339 has a great potential for cancer immunotherapy.
Collapse
|
17
|
Van den Bergh JMJ, Smits ELJM, Berneman ZN, Hutten TJA, De Reu H, Van Tendeloo VFI, Dolstra H, Lion E, Hobo W. Monocyte-Derived Dendritic Cells with Silenced PD-1 Ligands and Transpresenting Interleukin-15 Stimulate Strong Tumor-Reactive T-cell Expansion. Cancer Immunol Res 2017. [PMID: 28637876 DOI: 10.1158/2326-6066.cir-16-0336] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although allogeneic stem cell transplantation (allo-SCT) can elicit graft-versus-tumor (GVT) immunity, patients often relapse due to residual tumor cells. As essential orchestrators of the immune system, vaccination with dendritic cells (DC) is an appealing strategy to boost the GVT response. Nevertheless, durable clinical responses after DC vaccination are still limited, stressing the need to improve current DC vaccines. Aiming to empower DC potency, we engineered monocyte-derived DCs to deprive them of ligands for the immune checkpoint regulated by programmed death 1 (PD-1). We also equipped them with interleukin (IL)-15 "transpresentation" skills. Transfection with short interfering (si)RNA targeting the PD-1 ligands PD-L1 and PD-L2, in combination with IL15 and IL15Rα mRNA, preserved their mature DC profile and rendered the DCs superior in inducing T-cell proliferation and IFNγ and TNFα production. Translated into an ex vivo hematological disease setting, DCs deprived of PD-1 ligands (PD-L), equipped with IL15/IL15Rα expression, or most effectively, both, induced superior expansion of minor histocompatibility antigen-specific CD8+ T cells from transplanted cancer patients. These data support the combinatorial approach of in situ suppression of the PD-L inhibitory checkpoints with DC-mediated IL15 transpresentation to promote antigen-specific T-cell responses and, ultimately, contribute to GVT immunity. Cancer Immunol Res; 5(8); 710-5. ©2017 AACR.
Collapse
Affiliation(s)
- Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| | - Evelien L J M Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Tim J A Hutten
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Harry Dolstra
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Willemijn Hobo
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Van den Bergh J, Willemen Y, Lion E, Van Acker H, De Reu H, Anguille S, Goossens H, Berneman Z, Van Tendeloo V, Smits E. Transpresentation of interleukin-15 by IL-15/IL-15Rα mRNA-engineered human dendritic cells boosts antitumoral natural killer cell activity. Oncotarget 2016; 6:44123-33. [PMID: 26675759 DOI: 10.18632/oncotarget.6536] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/28/2015] [Indexed: 01/20/2023] Open
Abstract
In cancer immunotherapy, the use of dendritic cell (DC)-based vaccination strategies can improve overall survival, but until now durable clinical responses remain scarce. To date, DC vaccines are designed primarily to induce effective T-cell responses, ignoring the antitumor activity potential of natural killer (NK) cells. Aiming to further improve current DC vaccination outcome, we engineered monocyte-derived DC to produce interleukin (IL)-15 and/or IL-15 receptor alpha (IL-15Rα) using mRNA electroporation. The addition of IL-15Rα to the protocol, enabling IL-15 transpresentation to neighboring NK cells, resulted in significantly better NK-cell activation compared to IL-15 alone. Next to upregulation of NK-cell membrane activation markers, IL-15 transpresentation resulted in increased NK-cell secretion of IFN-γ, granzyme B and perforin. Moreover, IL-15-transpresenting DC/NK cell cocultures from both healthy donors and acute myeloid leukemia (AML) patients in remission showed markedly enhanced cytotoxic activity against NK cell sensitive and resistant tumor cells. Blocking IL-15 transpresentation abrogated NK cell-mediated cytotoxicity against tumor cells, pointing to a pivotal role of IL-15 transpresentation by IL-15Rα to exert its NK cell-activating effects. In conclusion, we report an attractive approach to improve antitumoral NK-cell activity in DC-based vaccine strategies through the use of IL-15/IL-15Rα mRNA-engineered designer DC.
Collapse
Affiliation(s)
- Johan Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Heleen Van Acker
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Evelien Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
19
|
Oncolytic influenza A virus expressing interleukin-15 decreases tumor growth in vivo. Surgery 2016; 161:735-746. [PMID: 27776794 DOI: 10.1016/j.surg.2016.08.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Interleukin-15 has become a promising molecule in the context of eliciting an effective, antitumor immune response because it is able to stimulate cells of the innate and adaptive immune system. METHODS We generated an interleukin-15-expressing oncolytic influenza A virus for the treatment of an established murine tumor model. RESULTS Our oncolytic influenza A virus produced large amounts of interleukin-15 and induced proliferation and activation of human T cells in vitro. Intraperitoneal administration increased the amount of mouse natural killer cells and effector memory T cells, as well as T cell reactivity in vivo. Moreover, intratumoral injection induced a profound decrease in growth of established tumors in mice and increased the amount of tumor-infiltrating T cells and natural killer cells. CONCLUSION We established a stable, IL-15-producing oncolytic influenza A virus with promising immunostimulatory and antitumor attributes.
Collapse
|
20
|
IL-15 receptor alpha as the magic wand to boost the success of IL-15 antitumor therapies: The upswing of IL-15 transpresentation. Pharmacol Ther 2016; 170:73-79. [PMID: 27777088 DOI: 10.1016/j.pharmthera.2016.10.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-15 as a stand-alone therapy can activate the antitumor functions of immune effector cells resulting in significant tumor regression. Interestingly, combining IL-15 with the α-moiety of its receptor (IL-15Rα), also called IL-15 transpresentation, increases the in vivo half-life of IL-15 and enhances binding of IL-15 with cells expressing the IL-15Rβγ, such as NK cells and CD8+ T cells. These features enlarge the signal transmission of IL-15, resulting in improved proliferation and antitumor activities of both NK cells and CD8+ T cells, eventually leading to enhanced killing of tumor cells. In this review, we discuss the antitumor strategies in which this IL-15 transpresentation mechanism is implemented, that are currently under preclinical investigation. Furthermore, we give an overview of the studies in which the IL-15/IL-15Rα complexes are combined with other antitumor therapies. The promising results in these preclinical studies have incited several clinical trials to test the safety and efficacy of IL-15 transpresentation strategies to treat both hematological and advanced solid tumors.
Collapse
|
21
|
Van Acker HH, Anguille S, Willemen Y, Van den Bergh JM, Berneman ZN, Lion E, Smits EL, Van Tendeloo VF. Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J Hematol Oncol 2016; 9:101. [PMID: 27686372 PMCID: PMC5041439 DOI: 10.1186/s13045-016-0329-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022] Open
Abstract
Background Adoptive immunotherapy is gaining momentum to fight malignancies, whereby γδ T cells have received recent attention as an alternative cell source as to natural killer cells and αβ T cells. The advent of γδ T cells is largely due to their ability to recognize and target tumor cells using both innate characteristic and T cell receptor (TCR)-mediated mechanisms, their capacity to enhance the generation of antigen-specific T cell responses, and their potential to be used in an autologous or allogeneic setting. Methods In this study, we explored the beneficial effect of the immunostimulatory cytokine interleukin (IL)-15 on purified γδ T cells and its use as a stimulatory signal in the ex vivo expansion of γδ T cells for adoptive transfer. The expansion protocol was validated both with immune cells of healthy individuals and acute myeloid leukemia patients. Results We report that the addition of IL-15 to γδ T cell cultures results in a more activated phenotype, a higher proliferative capacity, a more pronounced T helper 1 polarization, and an increased cytotoxic capacity of γδ T cells. Moreover γδ T cell expansion starting with peripheral blood mononuclear cells from healthy individuals and acute myeloid leukemia patients is boosted in the presence of IL-15, whereby the antitumor properties of the γδ T cells are strengthened as well. Conclusions Our results support the rationale to explore the use of IL-15 in clinical adoptive therapy protocols exploiting γδ T cells.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| | - Johan M Van den Bergh
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Antwerp, Belgium
| |
Collapse
|
22
|
TRAIL-mediated killing of acute lymphoblastic leukemia by plasmacytoid dendritic cell-activated natural killer cells. Oncotarget 2016; 6:29440-55. [PMID: 26320191 PMCID: PMC4745738 DOI: 10.18632/oncotarget.4984] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/12/2015] [Indexed: 01/24/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) still frequently recurs after hematopoietic stem cell transplantation (HSCT), underscoring the need to improve the graft-versus-leukemia (GvL) effect. Natural killer (NK) cells reconstitute in the first months following HSCT when leukemia burden is at its lowest, but ALL cells have been shown to be resistant to NK cell-mediated killing. We show here that this resistance is overcome by NK cell stimulation with TLR-9-activated plasmacytoid dendritic cells (pDCs). NK cell priming with activated pDCs resulted in TRAIL and CD69 up-regulation on NK cells and IFN-γ production. NK cell activation was dependent on IFN-α produced by pDCs, but was not reproduced by IFN-α alone. ALL killing was further enhanced by inhibition of KIR engagement. We showed that ALL lysis was mainly mediated by TRAIL engagement, while the release of cytolytic granules was involved when ALL expressed NK cell activating receptor ligands. Finally, adoptive transfers of activated-pDCs in ALL-bearing humanized mice delayed the leukemia onset and cure 30% of mice. Our data therefore demonstrate that TLR-9 activated pDCs are a powerful tool to overcome ALL resistance to NK cell-mediated killing and to reinforce the GvL effect of HSCT. These results open new therapeutic avenues to prevent relapse in children with ALL.
Collapse
|
23
|
Song Y, Liu Y, Hu R, Su M, Rood D, Lai L. In Vivo Antitumor Activity of a Recombinant IL7/IL15 Hybrid Cytokine in Mice. Mol Cancer Ther 2016; 15:2413-2421. [PMID: 27474151 DOI: 10.1158/1535-7163.mct-16-0111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/15/2016] [Indexed: 11/16/2022]
Abstract
Both IL7 and IL15 have become important candidate immunomodulators for cancer treatment. However, IL7 or IL15 used alone suffers from shortcomings, such as short serum half-life and limited antitumor effect. We have cloned and expressed a recombinant (r) IL7/IL15 fusion protein in which IL7 and IL15 are linked by a flexible linker. We then compared the antitumor effect of rIL7/IL15 with the individual factors rIL7 and/or rIL15. We show here that rIL7/IL15 has a higher antitumor activity than the combination of the individual factors in both murine B16F10 melanoma and CT-26 colon cancer models. This was associated with a significant increase in tumor infiltration of T cells, DCs, and NK cells and a decrease in regulatory T cells (Tregs). In addition, rIL7/IL15-treated DCs had higher expression of costimulatory molecules CD80 and CD86. The higher antitumor activity of rIL7/IL15 is likely due to its longer in vivo half-life and different effects on immune cells. Our results suggest that rIL7/IL15 may offer a new tool to enhance antitumor immunity and treat cancer. Mol Cancer Ther; 15(10); 2413-21. ©2016 AACR.
Collapse
Affiliation(s)
- Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. Medical College, Three Gorges University, Yichang, China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
24
|
Read KA, Powell MD, McDonald PW, Oestreich KJ. IL-2, IL-7, and IL-15: Multistage regulators of CD4(+) T helper cell differentiation. Exp Hematol 2016; 44:799-808. [PMID: 27423815 DOI: 10.1016/j.exphem.2016.06.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/20/2022]
Abstract
Cytokines represent a class of environmental factors that are critical drivers of immune cell development. Cytokines of the common gamma-chain family, including interleukin (IL)-2, IL-7, and IL-15, have been the subject of intense experimental scrutiny and have well-defined roles as regulators of diverse immune cell types including CD4(+) T helper cells. Because of their pleiotropic effects on T-cell development and function, researchers and clinicians have attempted to harness the capabilities of these cytokines for therapeutic benefit. In this review, we summarize the recent progress in our understanding of the molecular mechanisms underlying the effects of these cytokines on CD4(+) T cell development and briefly discuss how these immunomodulatory cytokines are being used in efforts to treat human disease.
Collapse
Affiliation(s)
| | - Michael D Powell
- Virginia Tech Carilion Research Institute, Roanoke, VA; Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA
| | | | - Kenneth J Oestreich
- Virginia Tech Carilion Research Institute, Roanoke, VA; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA; Virginia Tech Carilion School of Medicine, Roanoke, VA.
| |
Collapse
|
25
|
Velásquez SY, Killian D, Schulte J, Sticht C, Thiel M, Lindner HA. Short Term Hypoxia Synergizes with Interleukin 15 Priming in Driving Glycolytic Gene Transcription and Supports Human Natural Killer Cell Activities. J Biol Chem 2016; 291:12960-77. [PMID: 27129235 DOI: 10.1074/jbc.m116.721753] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells induce apoptosis in infected and transformed cells and are important producers of immunoregulatory cytokines. Therefore, they operate under low oxygen conditions (hypoxia) in inflammatory and tumor environments. In vitro studies of NK cells are, however, commonly performed in ambient air (normoxia). We used global gene expression profiling to evaluate changes in transcriptional pathways in primary human NK cells following short term culture under hypoxia compared with normoxia and in response to interleukin 15 (IL-15) priming using a 2 × 2 factorial design. The largest contrasts observed were priming dependences for associations between hypoxia and the hypoxia-inducible factor (Hif) 1 signaling and glycolysis pathways. RT-PCR confirmed positive synergistic hypoxia/IL-15 interactions for genes of key regulatory and metabolic enzymes. IL-15 primes NK cells for effector functions, which were recently demonstrated to depend on glycolytic switching. We did not, however, observe important increases in glycolytic flux through hypoxia and priming alone. Chemical Hif-1α inhibition suggested equal importance of this transcription factor for glycolysis and energy production under normoxia and hypoxia. Hypoxia promoted secretion of CC chemokines Ccl3/4/5 and macrophage migration inhibitory factor. Unexpectedly, hypoxia also stimulated migration of NK cells through the extracellular matrix and shifted amounts of susceptible leukemia target cells toward late apoptosis in a cell killing assay. We conclude that short term hypoxia supports these activities by positively interacting with NK cell priming at the level of glycolytic gene transcription. Hypoxic conditioning of NK cells may thus benefit their use in cell-based immunotherapy of cancer.
Collapse
Affiliation(s)
- Sonia Y Velásquez
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Doreen Killian
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Jutta Schulte
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Carsten Sticht
- Medical Research Center, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Manfred Thiel
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Holger A Lindner
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| |
Collapse
|
26
|
Anguille S, Smits EL, Bryant C, Van Acker HH, Goossens H, Lion E, Fromm PD, Hart DN, Van Tendeloo VF, Berneman ZN. Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy. Pharmacol Rev 2015; 67:731-53. [PMID: 26240218 DOI: 10.1124/pr.114.009456] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Although the earliest—rudimentary—attempts at exploiting the immune system for cancer therapy can be traced back to the late 18th Century, it was not until the past decade that cancer immunotherapeutics have truly entered mainstream clinical practice. Given their potential to stimulate both adaptive and innate antitumor immune responses, dendritic cells (DCs) have come under intense scrutiny in recent years as pharmacological tools for cancer immunotherapy. Conceptually, the clinical effectiveness of this form of active immunotherapy relies on the completion of three critical steps: 1) the DCs used as immunotherapeutic vehicles must properly activate the antitumor immune effector cells of the host, 2) these immune effector cells must be receptive to stimulation by the DCs and be competent to mediate their antitumor effects, which 3) requires overcoming the various immune-inhibitory mechanisms used by the tumor cells. In this review, following a brief overview of the pivotal milestones in the history of cancer immunotherapy, we will introduce the reader to the basic immunobiological and pharmacological principles of active cancer immunotherapy using DCs. We will then discuss how current research is trying to define the optimal parameters for each of the above steps to realize the full clinical potential of DC therapeutics. Given its high suitability for immune interventions, acute myeloid leukemia was chosen here to showcase the latest research trends driving the field of DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Sébastien Anguille
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Evelien L Smits
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Christian Bryant
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Heleen H Van Acker
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Herman Goossens
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Eva Lion
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Phillip D Fromm
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | | | - Viggo F Van Tendeloo
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| | - Zwi N Berneman
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, Tumor Immunology Group (S.A., H.H.V.A., H.G., E.L., V.F.V.T., Z.N.B.), and Faculty of Medicine and Health Sciences, Center for Oncological Research (E.L.S.), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium (S.A., E.L.S., Z.N.B.); and ANZAC Research Institute, Dendritic Cell Biology and Therapeutics Group, University of Sydney, Sydney, New South Wales, Australia (C.B., P.D.F.)
| |
Collapse
|
27
|
Yan C, Jie L, Yongqi W, Weiming X, Juqun X, Yanbing D, Li Q, Xingyuan P, Mingchun J, Weijuan G. Delivery of human NKG2D-IL-15 fusion gene by chitosan nanoparticles to enhance antitumor immunity. Biochem Biophys Res Commun 2015; 463:336-43. [PMID: 26022121 DOI: 10.1016/j.bbrc.2015.05.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 01/22/2023]
Abstract
Nanoparticles are becoming promising carriers for gene delivery because of their high capacity in gene loading and low cell cytotoxicity. In this study, a chitosan-based nanoparticle encapsulated within a recombinant pcDNA3.1-dsNKG2D-IL-15 plasmid was generated. The fused dsNKG2D-IL-15 gene fragment consisted of double extracellular domains of NKG2D with IL-15 gene at downstream. The average diameter of the gene nanoparticles ranged from 200 nm to 400 nm, with mean zeta potential value of 53.8 ± 6.56 mV. The nanoparticles which were loaded with the dsNKG2D-IL-15 gene were uptaken by tumor cells with low cytotoxicity. Tumor cells pre-transfected by gene nanopartilces stimulated NK and T cells in vitro. Intramuscular injection of gene nanoparticles suppressed tumor growth and prolonged survival of tumor-bearing mice through activation of NK and CD8(+) T cells. Thus, chitosan-based nanoparticle delivery of dsNKG2D-IL-15 gene vaccine can be potentially used for tumor therapy.
Collapse
Affiliation(s)
- Chen Yan
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Leng Jie
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Wang Yongqi
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Xiao Weiming
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China
| | - Xi Juqun
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225009, PR China
| | - Ding Yanbing
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China
| | - Qian Li
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Pan Xingyuan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, PR China
| | - Ji Mingchun
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Gong Weijuan
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China; Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China.
| |
Collapse
|
28
|
Van Acker HH, Anguille S, Van Tendeloo VF, Lion E. Empowering gamma delta T cells with antitumor immunity by dendritic cell-based immunotherapy. Oncoimmunology 2015; 4:e1021538. [PMID: 26405575 PMCID: PMC4570126 DOI: 10.1080/2162402x.2015.1021538] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/13/2015] [Accepted: 02/14/2015] [Indexed: 12/16/2022] Open
Abstract
Gamma delta (γδ) T cells are the all-rounders of our immune-system with their major histocompatibility complex-unrestricted cytotoxicity, capacity to secrete immunosti-mulatory cytokines and ability to promote the generation of tumor antigen-specific CD8+ and CD4+ T cell responses. Dendritic cell (DC)-based vaccine therapy has the prospective to harness these unique features of the γδ T cells in the fight against cancer. In this review, we will discuss our current knowledge on DC-mediated γδ T cell activation and related opportunities for tumor immunologists.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); Vaccine & Infectious Disease Institute (VAXINFECTIO); Faculty of Medicine and Health Sciences; University of Antwerp ; Antwerp, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); Vaccine & Infectious Disease Institute (VAXINFECTIO); Faculty of Medicine and Health Sciences; University of Antwerp ; Antwerp, Belgium ; Center for Cell Therapy & Regenerative Medicine; Antwerp University Hospital ; Edegem, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); Vaccine & Infectious Disease Institute (VAXINFECTIO); Faculty of Medicine and Health Sciences; University of Antwerp ; Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology; Tumor Immunology Group (TIGR); Vaccine & Infectious Disease Institute (VAXINFECTIO); Faculty of Medicine and Health Sciences; University of Antwerp ; Antwerp, Belgium ; Center for Cell Therapy & Regenerative Medicine; Antwerp University Hospital ; Edegem, Belgium
| |
Collapse
|