1
|
Scapin G, Cagdas E, Grav LM, Lewis NE, Goletz S, Hafkenscheid L. Implications of glycosylation for the development of selected cytokines and their derivatives for medical use. Biotechnol Adv 2024; 77:108467. [PMID: 39447666 DOI: 10.1016/j.biotechadv.2024.108467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cytokines are important regulators of immune responses, making them attractive targets for autoimmune diseases and cancer therapeutics. Yet, the significance of cytokine glycosylation remains underestimated. Many cytokines carry N- and O-glycans and some even undergo C-mannosylation. Recombinant cytokines produced in heterologous host cells may lack glycans or exhibit a different glycosylation pattern such as varying levels of galactosylation, sialylation, fucosylation or xylose addition compared to their human counterparts, potentially impacting critical immune interactions. We focused on cytokines that are currently utilized or designed in advanced therapeutic formats, including immunocytokines, fusokines, engager cytokines, and genetically engineered 'supercytokines.' Despite the innovative designs of these cytokine derivatives, their glycosylation patterns have not been extensively studied. By examining the glycosylation of the human native cytokines, G-CSF and GM-CSF, interferons β and γ, TNF-α and interleukins-2, -3 -4, -6, -7, -9, -12, -13, -15, -17A, -21, and - 22, we aim to assess its potential impact on their therapeutic derivatives. Understanding the glycosylation of the native cytokines could provide critical insights into the safety, efficacy, and functionality of these next-generation cytokine therapies, affecting factors such as stability, bioactivity, antigenicity, and half-life. This knowledge can guide the choice of optimal expression hosts for production and advance the development of effective cytokine-based therapeutics and synthetic immunology drugs.
Collapse
Affiliation(s)
- Giulia Scapin
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Ece Cagdas
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| |
Collapse
|
2
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
3
|
Beig Parikhani A, Dehghan R, Talebkhan Y, Bayat E, Biglari A, Shokrgozar MA, Ahangari Cohan R, Mirabzadeh E, Ajdary S, Behdani M. A novel nanobody-based immunocytokine of a mutant interleukin-2 as a potential cancer therapeutic. AMB Express 2024; 14:19. [PMID: 38337114 PMCID: PMC10857990 DOI: 10.1186/s13568-023-01648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/12/2024] Open
Abstract
The immunotherapeutic application of interleukin-2 (IL-2) in cancer treatment is limited by its off-target effects on different cell populations and insufficient activation of anti-tumor effector cells at the site of the tumor upon tolerated doses. Targeting IL-2 to the tumor microenvironment by generating antibody-cytokine fusion proteins (immunocytokine) would be a promising approach to increase efficacy without associated toxicity. In this study, a novel nanobody-based immunocytokine is developed by the fusion of a mutant (m) IL-2 with a decreased affinity toward CD25 to an anti-vascular endothelial growth factor receptor-2 (VEGFR2) specific nanobody, denoted as VGRmIL2-IC. The antigen binding, cell proliferation, IFN-γ-secretion, and cytotoxicity of this new immunocytokine are evaluated and compared to mIL-2 alone. Furthermore, the pharmacokinetic properties are analyzed. Flow cytometry analysis shows that the VGRmIL2-IC molecule can selectively target VEGFR2-positive cells. The results reveal that the immunocytokine is comparable to mIL-2 alone in the stimulation of Primary Peripheral Blood Mononuclear Cells (PBMCs) and cytotoxicity in in vitro conditions. In vivo studies demonstrate improved pharmacokinetic properties of VGRmIL2-IC in comparison to the wild or mutant IL-2 proteins. The results presented here suggest VGRmIL2-IC could be considered a candidate for the treatment of VEGFR2-positive tumors.
Collapse
Affiliation(s)
- Arezoo Beig Parikhani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Rada Dehghan
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Yeganeh Talebkhan
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Bayat
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Biglari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat Mirabzadeh
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Dehghan R, Parikhani AB, Cohan RA, Shokrgozar MA, Mirabzadeh E, Ajdary S, Zeinali S, Ghaderi H, Talebkhan Y, Behdani M. Specific Targeting of Zinc Transporter LIV-1 with Immunocytokine Containing Anti-LIV-1 VHH and Human IL-2 and Evaluation of its In vitro Antitumor Activity. Curr Pharm Des 2024; 30:868-876. [PMID: 38482625 DOI: 10.2174/0113816128295195240305060103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/16/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Interleukin 2 (IL-2) is a vital cytokine in the induction of T and NK cell responses, the proliferation of CD8+ T cells, and the effective treatment of human cancers such as melanoma and renal cell carcinoma. However, widespread use of this cytokine is limited due to its short half-life, severe toxicity, lack of specific tumor targeting, and activation of Treg cells mediated by high-affinity interleukin-2 receptors. OBJECTIVE In this study, a tumor-targeting LIV-1 VHH-mutIL2 immunocytokine with reduced CD25 (α chain of the high-affinity IL-2 receptor) binding activity was developed to improve IL-2 half-life by decreasing its renal infiltration in comparison with wild and mutant IL-2 molecules. METHODS The recombinant immunocytokine was designed and expressed. The biological activity of the purified fusion protein was investigated in in vitro and in vivo experiments. RESULTS The fusion protein represented specific binding to MCF7 (the breast cancer cell line) and more efficient cytotoxicity than wild-type IL-2 and mutant IL-2. The PK parameters of the recombinant immunocytokine were also improved in comparison to the IL-2 molecules. CONCLUSION The observed results showed that LIV1-mIL2 immunocytokine could be considered as an effective agent in the LIV-1-targeted treatment of cancers due to its longer half-life and stronger cytotoxicity.
Collapse
Affiliation(s)
- Rada Dehghan
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Arezoo Beig Parikhani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | | | - Esmat Mirabzadeh
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sirous Zeinali
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Hajarossadat Ghaderi
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Yeganeh Talebkhan
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
Jeong KY. Challenges to addressing the unmet medical needs for immunotherapy targeting cold colorectal cancer. World J Gastrointest Oncol 2023; 15:215-224. [PMID: 36908316 PMCID: PMC9994045 DOI: 10.4251/wjgo.v15.i2.215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/14/2023] Open
Abstract
With the establishment of the immune surveillance mechanism since the 1950s, attempts have been made to activate the immune system for cancer treatment through the discovery of various cytokines or the development of antibodies up to now. The fruits of these efforts have contributed to the recognition of the 3rd generation of anticancer immunotherapy as the mainstream of cancer treatment. However, the limitations of cancer immunotherapy are also being recognized through the conceptual establishment of cold tumors recently, and colorectal cancer (CRC) has become a major issue from this therapeutic point of view. Here, it is emphasized that non-clinical strategies to overcome the immunosuppressive environment and clinical trials based on these basic investigations are being made on the journey to achieve better treatment outcomes for the treatment of cold CRC.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Research and Development Center, PearlsinMires, Seoul 03690, South Korea
| |
Collapse
|
6
|
Casadesús AV, Cruz BM, Díaz W, González MÁ, Gómez T, Fernández B, González A, Ledón N, Sosa K, Castro K, López A, Plasencia C, Ramírez Y, Teillaud JL, Hernández C, León K, Hernández T. Potent immunomodulatory and antitumor effect of anti-CD20-IL2no-alpha tri-functional immunocytokine for cancer therapy. Front Immunol 2022; 13:1021828. [PMID: 36569901 PMCID: PMC9780377 DOI: 10.3389/fimmu.2022.1021828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction The anti-CD20 antibody rituximab (RTX) has substantially improved outcomes of patients with B-cell lymphomas, although more efficient therapies are needed for refractory or relapsing lymphomas. An approach to increase the clinical effectiveness of anti-tumor therapy is the use of antibody-cytokine fusion proteins (immunocytokines (ICKs)) to deliver at the tumor site the antibody effector functions and cytokines that trigger anti-tumor activities. In particular, IL-2-based ICKs have shown significant results in preclinical studies but not in clinical trials due to the toxicity profile associated to high doses IL-2 and the undesired expansion of Tregs. Methods To improve the efficacy of RTX therapy, we fused a murine (mIgG2a) or a human (hIgG1) version of RTX to a mutated IL-2 (no-alpha mutein), which has a disrupted affinity for the high affinity IL-2 receptor (IL-2R) to prevent the stimulation of Tregs and reduce the binding to endothelial cells expressing CD25, the α chain of high affinity IL-2R. Characterization of anti-CD20-IL2no-alpha ICKs was performed by SDS-PAGE, Western-blotting and SEC-HPLC and also by several functional in vitro techniques like T-cell proliferation assays, apoptosis, CDC and ADCC assays. The in vivo activity was assessed by using murine tumor cells expressing huCD20 in C57/Bl6 mice. Results Both ICKs exhibited similar in vitro specific activity of their IL2no-alpha mutein moieties and kept CD20-binding capacity. Anti-CD20-IL2no-alpha (hIgG1) retained antibody effector functions as complement-dependent cytotoxicity and enhanced direct apoptosis, NK cell activation and antibody-dependent cellular cytotoxicity relative to RTX. In addition, both ICKs demonstrated a higher antitumor efficacy than parental molecules or their combination in an EL4-huCD20 tumor model in immunocompetent mice. Anti-CD20-IL2no-alpha (hIgG1) strongly expanded NK and CD8+ T cells but not Tregs in tumor-bearing mice. Discussion These findings suggest that anti-CD20-IL2no-alpha could represent an alternative treatment for B cell lymphoma patients, mainly those refractory to RTX therapy.
Collapse
Affiliation(s)
- Ana Victoria Casadesús
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Beatriz María Cruz
- Quality Control Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Wilden Díaz
- Quality Control Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Miguel Ángel González
- Department of Animal Facilities, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Tania Gómez
- Quality Control Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Briandy Fernández
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Addys González
- Department of Animal Facilities, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Nuris Ledón
- Department of Innovation´s Management, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Katya Sosa
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Kathleen Castro
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Armando López
- Department of Animal Facilities, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Claudia Plasencia
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Yaima Ramírez
- Development Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Jean-Luc Teillaud
- Laboratory of Immune Microenvironment and Immunotherapy, Centre d’immunologie et des maladies infectieuses (CIMI-Paris), Inserm UMRS1135, Sorbonne University, Paris, France
| | | | - Kalet León
- Research Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Tays Hernández
- Department of Chimeric Proteins, Immunobiology Division, Center of Molecular Immunology (CIM), Havana, Cuba,*Correspondence: Tays Hernández,
| |
Collapse
|
7
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
8
|
Interleukin 2-Based Fusion Proteins for the Treatment of Cancer. J Immunol Res 2021; 2021:7855808. [PMID: 34790830 PMCID: PMC8592747 DOI: 10.1155/2021/7855808] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Interleukin 2 (IL-2) plays a fundamental role in both immune activation and tolerance and has revolutionized the field of cancer immunotherapy since its discovery. The ability of IL-2 to mediate tumor regression in preclinical and clinical settings led to FDA approval for its use in the treatment of metastatic renal cell carcinoma and metastatic melanoma in the 1990s. Although modest success is observed in the clinic, cancer patients receiving IL-2 therapy experience a wide array of side effects ranging from flu-like symptoms to life-threatening conditions such as vascular leak syndrome. Over the past three decades, efforts have focused on circumventing IL-2-related toxicities by engineering methods to localize IL-2 to the tumor or secondary lymphoid tissue, preferentially activate CD8+ T cells and NK cells, and alter pharmacokinetic properties to increase bioavailability. This review summarizes the various IL-2-based strategies that have emerged, with a focus on chimeric fusion methods.
Collapse
|
9
|
Runbeck E, Crescioli S, Karagiannis SN, Papa S. Utilizing Immunocytokines for Cancer Therapy. Antibodies (Basel) 2021; 10:antib10010010. [PMID: 33803078 PMCID: PMC8006145 DOI: 10.3390/antib10010010] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Cytokine therapy for cancer has indicated efficacy in certain diseases but is generally accompanied by severe toxicity. The field of antibody-cytokine fusion proteins (immunocytokines) arose to target these effector molecules to the tumor environment in order to expand the therapeutic window of cytokine therapy. Pre-clinical evidence has shown the increased efficacy and decreased toxicity of various immunocytokines when compared to their cognate unconjugated cytokine. These anti-tumor properties are markedly enhanced when combined with other treatments such as chemotherapy, radiotherapy, and checkpoint inhibitor antibodies. Clinical trials that have continued to explore the potential of these biologics for cancer therapy have been conducted. This review covers the in vitro, in vivo, and clinical evidence for the application of immunocytokines in immuno-oncology.
Collapse
Affiliation(s)
- Erin Runbeck
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Studies, King’s College London, London SE19RT, UK;
| | - Silvia Crescioli
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London SE1 9RT, UK; (S.C.); (S.N.K.)
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London SE1 9RT, UK; (S.C.); (S.N.K.)
| | - Sophie Papa
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Studies, King’s College London, London SE19RT, UK;
- Correspondence:
| |
Collapse
|
10
|
Chen Y, Wang G, Lin B, Huang J. MicroRNA‐93‐5p expression in tumor tissue and its tumor suppressor function via targeting programmed death ligand‐1 in colorectal cancer. Cell Biol Int 2020; 44:1224-1236. [PMID: 32068322 DOI: 10.1002/cbin.11323] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/16/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Yi‐Lin Chen
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical University Quanzhou 362000 Fujian China
| | - Gao‐Xiong Wang
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical University Quanzhou 362000 Fujian China
| | - Bei‐An Lin
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical University Quanzhou 362000 Fujian China
| | - Jing‐Shan Huang
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical University Quanzhou 362000 Fujian China
| |
Collapse
|
11
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Hu Z, Ye L, Xing Y, Hu J, Xi T. Combined SEP and anti-PD-L1 antibody produces a synergistic antitumor effect in B16-F10 melanoma-bearing mice. Sci Rep 2018; 8:217. [PMID: 29317734 PMCID: PMC5760644 DOI: 10.1038/s41598-017-18641-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 11/06/2017] [Indexed: 12/29/2022] Open
Abstract
The increased PD-L1 induces poorer prognosis in melanoma. The treatment with PD-1/PD-L1 antibodies have a low response rate. The combination immunotherapies are the encouraging drug development strategy to receive maximal therapeutic benefit. In this study, we investigated the enhanced antitumor and immunomodulatory activity of combined SEP and αPD-L1 in B16-F10 melanoma-bearing mice. The results shown that combined SEP and αPD-L1 presented significant synergistic antitumor effects, increased the frequency of CD8+ and CD4+ T cells in spleen and tumor, cytotoxic activity of CTL in spleen, and IL-2 and IFN-γ levels in splenocytes and tumor. The combination treatment also produced synergistic increase in P-ERK1/2 level in spleen. Immunohistochemistry shown that SEP induced the PD-L1 expression in melanoma tissue possibly by promoting IFN-γ excretion, which led to the synergistic anti-tumor effects of aPD-L1 and SEP. Furthermore, in the purified T lymphocyte from the naive mice, the combination of SEP and αPD-L1 had more potent than SEP or αPD-L1 in promoting T lymphocyte proliferation and cytokines secretion including IL-2 and IFN-γ, at least partially by activating MEK/ERK pathway. Our study provides the scientific basis for a clinical trial that would involve combination of anti-PD-L1 mAb and SEP for sustained melanoma control.
Collapse
Affiliation(s)
- Zhengping Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Liang Ye
- School of Public Health and Management, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jinhang Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
13
|
Kosmidis C, Sapalidis K, Koletsa T, Kosmidou M, Efthimiadis C, Anthimidis G, Varsamis N, Michalopoulos N, Koulouris C, Atmatzidis S, Liavas L, Strati TM, Koimtzis G, Tsakalidis A, Mantalovas S, Zarampouka K, Florou M, Giannakidis DE, Georgakoudi E, Baka S, Zarogoulidis P, Man YG, Kesisoglou I. Interferon-γ and Colorectal Cancer: an up-to date. J Cancer 2018; 9:232-238. [PMID: 29344268 PMCID: PMC5771329 DOI: 10.7150/jca.22962] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer still remains the third cause of cancer death among cancer patients. Early diagnosis is crucial and they can be either endoscopic or with blood biomarkers. Endoscopic methods consist of gastroscopy and colonoscopy, however; in recent years, endoscopic ultrasound is being used. The microenvironment is very important for the successful delivery of the treatment. Several proteins and hormones play a crucial role in the efficiency of the treatment. In the current mini review we will focus on interferon-γ.
Collapse
Affiliation(s)
- Christoforos Kosmidis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Konstantinos Sapalidis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Triantafyllia Koletsa
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Kosmidou
- 1st Internal Medicine Division, University Hospital of Ioannina, University of Ioaninna, Medical School
| | | | - George Anthimidis
- Surgery Department, "Interbalkan" European Medical Center, Thessaloniki, Greece
| | - Nikolaos Varsamis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Nikolaos Michalopoulos
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Charilaos Koulouris
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Stefanos Atmatzidis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Lazaros Liavas
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Titika-Marina Strati
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Georgios Koimtzis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Alexandros Tsakalidis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Stylianos Mantalovas
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Katerina Zarampouka
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Florou
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Dimitrios E Giannakidis
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Eleni Georgakoudi
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| | - Sofia Baka
- Oncology Department, "Interbalkan" European Medical Center, Thessaloniki, Greece
| | - Paul Zarogoulidis
- Pulmonary - Oncology Department, "Theageneio" Anticancer Hospital, Thessaloniki, Greece
| | - Yan-Gao Man
- Research Laboratory and International Collaboration, Bon Secours Cancer Institute, VA, USA
| | - Isaac Kesisoglou
- 3rd Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, Medical School
| |
Collapse
|
14
|
Liu B, Guo H, Xu J, Qin T, Guo Q, Gu N, Zhang D, Qian W, Dai J, Hou S, Wang H, Guo Y. Elimination of tumor by CD47/PD-L1 dual-targeting fusion protein that engages innate and adaptive immune responses. MAbs 2017; 10:315-324. [PMID: 29182441 DOI: 10.1080/19420862.2017.1409319] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The host immune system generally serves as a barrier against tumor formation. Programmed death-ligand 1 (PD-L1) is a critical "don't find me" signal to the adaptive immune system, whereas CD47 transmits an anti-phagocytic signal, known as the "don't eat me" signal, to the innate immune system. These and similar immune checkpoints are often overexpressed on human tumors. Thus, dual targeting both innate and adaptive immune checkpoints would likely maximize anti-tumor therapeutic effect and elicit more durable responses. Herein, based on the variable region of atezolizumab and consensus variant 1 (CV1) monomer, we constructed a dual-targeting fusion protein targeting both CD47 and PD-L1 using "Knobs-into-holes" technology, denoted as IAB. It was effective in inducing phagocytosis of tumor cells, stimulating T-cell activation and mediating antibody-dependent cell-mediated cytotoxicity in vitro. No obvious sign of hematological toxicity was observed in mice administered IAB at a dose of 100 mg/kg, and IAB exhibited potent antitumor activity in an immune-competent mouse model of MC38. Additionally, the anti-tumor effect of IAB was impaired by anti-CD8 antibody or clodronate liposomes, which implied that both CD8+ T cells and macrophages were required for the anti-tumor efficacy of IAB and IAB plays an essential role in the engagement of innate and adaptive immune responses. Collectively, these results demonstrate the capacity of an elicited endogenous immune response against tumors and elucidate essential characteristics of synergistic innate and adaptive immune response, and indicate dual blockade of CD47 and PD-L1 by IAB may be a synergistic therapy that activates both innate and adaptive immune response against tumors.
Collapse
Affiliation(s)
- Boning Liu
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,b Department of Molecular Biology, School of Bioscience and Bioengineering , South China University of Technology , Guangzhou , China.,f Department of Pharmacy for Biologics , Center for Drug Evaluation, China Food and Drug Administration , Beijing , China
| | - Huaizu Guo
- c R&D Department , Shanghai Sinomab Biotechnology Co. , Shanghai , China
| | - Jin Xu
- c R&D Department , Shanghai Sinomab Biotechnology Co. , Shanghai , China
| | - Ting Qin
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,b Department of Molecular Biology, School of Bioscience and Bioengineering , South China University of Technology , Guangzhou , China
| | - Qingcheng Guo
- c R&D Department , Shanghai Sinomab Biotechnology Co. , Shanghai , China
| | - Nana Gu
- d R&D Department , Shanghai Zhangjiang Biotechnology Co. , Shanghai , China
| | - Dapeng Zhang
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,d R&D Department , Shanghai Zhangjiang Biotechnology Co. , Shanghai , China
| | - Weizhu Qian
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,g Department of AIMBL , Institute of Molecular and Cell Biology , Proteos , Singapore
| | - Jianxin Dai
- c R&D Department , Shanghai Sinomab Biotechnology Co. , Shanghai , China
| | - Sheng Hou
- c R&D Department , Shanghai Sinomab Biotechnology Co. , Shanghai , China
| | - Hao Wang
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,d R&D Department , Shanghai Zhangjiang Biotechnology Co. , Shanghai , China
| | - Yajun Guo
- a Department of Molecular Biology, State Key Laboratory of Antibody Medicine and Targeted Therapy , Shanghai , China.,e Department of Biopharmaceutical Sciences , School of Pharmacy, Liaocheng University , Liaocheng , China.,g Department of AIMBL , Institute of Molecular and Cell Biology , Proteos , Singapore
| |
Collapse
|
15
|
Sun H, Han X, Yan X, Xu J, Huang Q, Meng F, Zhang H, Li S. A novel mimovirus encoding ChgA 10-19 peptide with PD-L1 induces T cell tolerance and ameliorates the severity of diabetes. Cell Immunol 2017; 320:56-61. [PMID: 28916112 DOI: 10.1016/j.cellimm.2017.09.001] [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: 06/06/2017] [Revised: 07/23/2017] [Accepted: 09/03/2017] [Indexed: 10/18/2022]
Abstract
Related studies demonstrate that type 1 diabetes (T1D) is caused by β-cell antigen specific autoreactive CD8+ T cells. ChgA has recently been identified as the autoantigen in NOD mice and T1D patients. Therefore, attenuating the activation of ChgA specific CD8+ T cells might be a promising target for T1D therapy. The negative co-stimulatory PD-L1 inhibits T cell mediated alloimmunity and induces tolerance. In this experiment, a novel mimovirus encoding ChgA10-19 peptide with PD-L1 was constructed. The NOD.β2m null HHD mice were administrated with mimovirus transduced DCs. After immunization, the activation and proliferation of CD8+ T cells were detected, diabetes incidence and pancreatic tissue destruction were also analyzed. The results demonstrated that transduced DCs attenuated CD8+ T cell activation and proliferation. In addition, transduced DCs inhibited CD8+ T response to ChgA stimulation, and ameliorated the severity of diabetes. These data suggested that mimovirus transduced DCs might provide novel clues for T1D therapy.
Collapse
Affiliation(s)
- Hong Sun
- Department of Outpatient, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Xiaoguang Han
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Xiuhui Yan
- Department of Obstetrics and Gynecology, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Jingli Xu
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Qiujing Huang
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Fanqing Meng
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Hongjin Zhang
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China
| | - Shufa Li
- Department of Endocrinology and Metabolism, The Third People's Hospital of Linyi, Linyi, Shangdong 276000, China.
| |
Collapse
|
16
|
王 维, 王 丹, 秦 国, 陈 新, 张 毅. Immune-checkpoint blockade in colorectal cancer: Current research and future perspectives. Shijie Huaren Xiaohua Zazhi 2017; 25:1714. [DOI: 10.11569/wcjd.v25.i19.1714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|