1
|
Hwang S, Hicks A, Hoo CZ, Kwon YS, Cho YE, Moore J, Gao B. Novel treatment of acute and acute-on-chronic liver failure: Interleukin-22. Liver Int 2025; 45:e15619. [PMID: 37208937 PMCID: PMC10657333 DOI: 10.1111/liv.15619] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/15/2023] [Accepted: 05/09/2023] [Indexed: 05/21/2023]
Abstract
Acute liver failure (ALF) is a life-threatening medical condition, characterized by rapidly progressive hepatic dysfunction, coagulopathy and hepatic encephalopathy in patients without chronic liver disease, while acute-on-chronic liver failure (ACLF) occurs in patients with existing chronic liver disease. ALF and ACLF are often associated with multiple organ failure and a high short-term mortality. In this review, we briefly discuss the causes and pathogenesis of ALF and ACLF, the current options available for the treatment of both deadly maladies and interleukin-22 (IL-22), a novel promising drug that may have great therapeutic potential for ALF and ACLF treatment. IL-22 is a cytokine produced by immune cells but mainly targets epithelial cells including hepatocytes. IL-22 has been shown to protect against organ damage and reduce bacterial infection in many preclinical models and several clinical trials including alcohol-associated hepatitis. The potential application of IL-22 for the treatment of ALF and ACLF is also elaborated.
Collapse
Affiliation(s)
- Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Amy Hicks
- Leeds Liver Unit, St James’s University Hospital, UK
| | - Chai Zhen Hoo
- Leeds Liver Unit, St James’s University Hospital, UK
| | - Yong Seong Kwon
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Ye Eun Cho
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Joanna Moore
- Leeds Liver Unit, St James’s University Hospital, UK
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Lim J, Lee HK. Engineering interferons for cancer immunotherapy. Biomed Pharmacother 2024; 179:117426. [PMID: 39243429 DOI: 10.1016/j.biopha.2024.117426] [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: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024] Open
Abstract
Interferons are a family of cytokines that are famously known for their involvement in innate and adaptive immunity. Type I interferons (IFNs) exert pleiotropic effects on various immune cells and contribute to tumor-intrinsic and extrinsic mechanisms. Their pleiotropic effects and ubiquitous expression on nucleated cells have made them attractive candidates for cytokine engineering to deliver to largely immunosuppressive tumors. Type III interferons were believed to play overlapping roles with type I IFNs because they share a similar signaling pathway and induce similar transcriptional programs. However, type III IFNs are unique in their cell specific receptor expression and their antitumor activity is specific to a narrow range of cell types. Thus, type III IFN based therapies may show reduced toxic side effects compared with type I IFN based treatment. In this review, we focus on the development of IFN-based therapeutics used to treat different tumors. We highlight how the development in cytokine engineering has allowed for efficient delivery of type I and type III IFNs to tumor sites and look ahead to the obstacles that are still associated with IFN-based therapies before they can be fully and safely integrated into clinical settings.
Collapse
Affiliation(s)
- Juhee Lim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; KAIST Institute of Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
3
|
Wang MJ, Xia Y, Gao QL. DNA Damage-driven Inflammatory Cytokines: Reprogramming of Tumor Immune Microenvironment and Application of Oncotherapy. Curr Med Sci 2024; 44:261-272. [PMID: 38561595 DOI: 10.1007/s11596-024-2859-1] [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: 12/15/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
DNA damage occurs across tumorigenesis and tumor development. Tumor intrinsic DNA damage can not only increase the risk of mutations responsible for tumor generation but also initiate a cellular stress response to orchestrate the tumor immune microenvironment (TIME) and dominate tumor progression. Accumulating evidence documents that multiple signaling pathways, including cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) and ataxia telangiectasia-mutated protein/ataxia telangiectasia and Rad3-related protein (ATM/ATR), are activated downstream of DNA damage and they are associated with the secretion of diverse cytokines. These cytokines possess multifaced functions in the anti-tumor immune response. Thus, it is necessary to deeply interpret the complex TIME reshaped by damaged DNA and tumor-derived cytokines, critical for the development of effective tumor therapies. This manuscript comprehensively reviews the relationship between the DNA damage response and related cytokines in tumors and depicts the dual immunoregulatory roles of these cytokines. We also summarize clinical trials targeting signaling pathways and cytokines associated with DNA damage and provide future perspectives on emerging technologies.
Collapse
Affiliation(s)
- Meng-Jie Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Qing-Lei Gao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Miroshnichenko S, Pykhtina M, Kotliarova A, Chepurnov A, Beklemishev A. Engineering a New IFN-ApoA-I Fusion Protein with Low Toxicity and Prolonged Action. Molecules 2023; 28:8014. [PMID: 38138504 PMCID: PMC10745500 DOI: 10.3390/molecules28248014] [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: 10/28/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Recombinant human interferon alpha-2b (rIFN) is widely used in antiviral and anticancer immunotherapy. However, the high efficiency of interferon therapy is accompanied by a number of side effects; this problem requires the design of a new class of interferon molecules with reduced cytotoxicity. In this work, IFN was modified via genetic engineering methods by merging it with the blood plasma protein apolipoprotein A-I in order to reduce acute toxicity and improve the pharmacokinetics of IFN. The chimeric protein was obtained via biosynthesis in the yeast P. pastoris. The yield of ryIFN-ApoA-I protein when cultivated on a shaker in flasks was 30 mg/L; protein purification was carried out using reverse-phase chromatography to a purity of 95-97%. The chimeric protein demonstrated complete preservation of the biological activity of IFN in the model of vesicular stomatitis virus and SARS-CoV-2. In addition, the chimeric form had reduced cytotoxicity towards Vero cells and increased cell viability under viral load conditions compared with commercial IFN-a2b preparations. Analysis of the pharmacokinetic profile of ryIFN-ApoA-I after a single subcutaneous injection in mice showed a 1.8-fold increased half-life of the chimeric protein compared with ryIFN.
Collapse
Affiliation(s)
- Svetlana Miroshnichenko
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Mariya Pykhtina
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Anastasiia Kotliarova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia;
| | - Alexander Chepurnov
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| | - Anatoly Beklemishev
- Federal Research Center of Fundamental and Translational Medicine (FRC FTM), Timakova str., 2, 630117 Novosibirsk, Russia; (S.M.); (A.C.); (A.B.)
| |
Collapse
|
5
|
Tran NL, Ferreira LM, Alvarez-Moya B, Buttiglione V, Ferrini B, Zordan P, Monestiroli A, Fagioli C, Bezzecchi E, Scotti GM, Esposito A, Leone R, Gnasso C, Brendolan A, Guidotti LG, Sitia G. Continuous sensing of IFNα by hepatic endothelial cells shapes a vascular antimetastatic barrier. eLife 2022; 11:e80690. [PMID: 36281643 PMCID: PMC9596162 DOI: 10.7554/elife.80690] [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: 06/01/2022] [Accepted: 09/18/2022] [Indexed: 11/21/2022] Open
Abstract
Hepatic metastases are a poor prognostic factor of colorectal carcinoma (CRC) and new strategies to reduce the risk of liver CRC colonization are highly needed. Herein, we used mouse models of hepatic metastatization to demonstrate that the continuous infusion of therapeutic doses of interferon-alpha (IFNα) controls CRC invasion by acting on hepatic endothelial cells (HECs). Mechanistically, IFNα promoted the development of a vascular antimetastatic niche characterized by liver sinusoidal endothelial cells (LSECs) defenestration extracellular matrix and glycocalyx deposition, thus strengthening the liver vascular barrier impairing CRC trans-sinusoidal migration, without requiring a direct action on tumor cells, hepatic stellate cells, hepatocytes, or liver dendritic cells (DCs), Kupffer cells (KCs) and liver capsular macrophages (LCMs). Moreover, IFNα endowed LSECs with efficient cross-priming potential that, along with the early intravascular tumor burden reduction, supported the generation of antitumor CD8+ T cells and ultimately led to the establishment of a protective long-term memory T cell response. These findings provide a rationale for the use of continuous IFNα therapy in perioperative settings to reduce CRC metastatic spreading to the liver.
Collapse
Affiliation(s)
- Ngoc Lan Tran
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Lorena Maria Ferreira
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Blanca Alvarez-Moya
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Valentina Buttiglione
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Barbara Ferrini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Paola Zordan
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
- Vita-Salute San Raffaele UniversityMilanItaly
| | - Andrea Monestiroli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Claudio Fagioli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| | | | | | - Antonio Esposito
- Vita-Salute San Raffaele UniversityMilanItaly
- Experimental Imaging Center, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Riccardo Leone
- Vita-Salute San Raffaele UniversityMilanItaly
- Experimental Imaging Center, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Chiara Gnasso
- Vita-Salute San Raffaele UniversityMilanItaly
- Experimental Imaging Center, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Andrea Brendolan
- Division of Experimental Oncology, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Luca G Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
- Vita-Salute San Raffaele UniversityMilanItaly
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific InstituteMilanItaly
| |
Collapse
|
6
|
Antczak M, Cañete PF, Chen Z, Belle C, Yu D. Evolution of γ chain cytokines: Mechanisms, methods and applications. Comput Struct Biotechnol J 2022; 20:4746-4755. [PMID: 36147674 PMCID: PMC9465101 DOI: 10.1016/j.csbj.2022.08.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/12/2022] Open
Abstract
The common γ chain family of cytokines and their receptors play fundamental roles in the immune system. Evolutionary studies of γ chain cytokines have elegantly illustrated how the immune system adapts to ever-changing environmental conditions. Indeed, these studies have revealed the uniqueness of cytokine evolution, which exhibits strong positive selection pressure needed to adapt to rapidly evolving threats whilst still conserving their receptor binding capabilities. In this review, we summarise the evolutionary mechanisms that gave rise to the characteristically diverse family of γ chain cytokines. We also speculate on the benefits of studying cytokine evolution, which may provide alternative ways to design novel cytokine therapeutic strategies. Additionally, we discuss current evolutionary models that elucidate the emergence of distinct cytokines (IL-4 and IL-13) and cytokine receptors (IL-2Rα and IL-15Rα). Finally, we address and reflect on the difficulties associated with evolutionary studies of rapidly evolving genes and describe a variety of computational methods that have revealed numerous aspects of cytokine evolution.
Collapse
Affiliation(s)
- Magdalena Antczak
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Pablo F. Cañete
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Clémence Belle
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
7
|
Suresh M, Menne S. Recent Drug Development in the Woodchuck Model of Chronic Hepatitis B. Viruses 2022; 14:v14081711. [PMID: 36016334 PMCID: PMC9416195 DOI: 10.3390/v14081711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/24/2022] Open
Abstract
Infection with hepatitis B virus (HBV) is responsible for the increasing global hepatitis burden, with an estimated 296 million people being carriers and living with the risk of developing chronic liver disease and cancer. While the current treatment options for chronic hepatitis B (CHB), including oral nucleos(t)ide analogs and systemic interferon-alpha, are deemed suboptimal, the path to finding an ultimate cure for this viral disease is rather challenging. The lack of suitable laboratory animal models that support HBV infection and associated liver disease progression is one of the major hurdles in antiviral drug development. For more than four decades, experimental infection of the Eastern woodchuck with woodchuck hepatitis virus has been applied for studying the immunopathogenesis of HBV and developing new antiviral therapeutics against CHB. There are several advantages to this animal model that are beneficial for performing both basic and translational HBV research. Previous review articles have focused on the value of this animal model in regard to HBV replication, pathogenesis, and immune response. In this article, we review studies of drug development and preclinical evaluation of direct-acting antivirals, immunomodulators, therapeutic vaccines, and inhibitors of viral entry, gene expression, and antigen release in the woodchuck model of CHB since 2014 until today and discuss their significance for clinical trials in patients.
Collapse
|
8
|
Di Trani CA, Cirella A, Arrizabalaga L, Fernandez-Sendin M, Bella A, Aranda F, Melero I, Berraondo P. Overcoming the limitations of cytokines to improve cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:107-141. [PMID: 35777862 DOI: 10.1016/bs.ircmb.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytokines are pleiotropic soluble proteins used by immune cells to orchestrate a coordinated response against pathogens and malignancies. In cancer immunotherapy, cytokine-based drugs can be developed potentiating pro-inflammatory cytokines or blocking immunosuppressive cytokines. However, the complexity of the mechanisms of action of cytokines requires the use of biotechnological strategies to minimize systemic toxicity, while potentiating the antitumor response. Sequence mutagenesis, fusion proteins and gene therapy strategies are employed to enhance the half-life in circulation, target the desired bioactivity to the tumor microenvironment, and to optimize the therapeutic window of cytokines. In this review, we provide an overview of the different strategies currently being pursued in pre-clinical and clinical studies to make the most of cytokines for cancer immunotherapy.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Angela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
9
|
Recent Advances and Next Breakthrough in Immunotherapy for Cancer Treatment. J Immunol Res 2022; 2022:8052212. [PMID: 35340585 PMCID: PMC8956433 DOI: 10.1155/2022/8052212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
With the huge therapeutic potential, cancer immunotherapy is expected to become the mainstream of cancer treatment. In the current field of cancer immunotherapy, there are mainly five types. Immune checkpoint blockade therapy is one of the most promising directions. Adoptive cell therapy is an important component of cancer immunotherapy. The therapy with the cancer vaccine is promising cancer immunotherapy capable of cancer prevention. Cytokine therapy is one of the pillars of cancer immunotherapy. Oncolytic immunotherapy is a promising novel component of cancer immunotherapy, which with significantly lower incidence of serious adverse reactions. The recent positive results of many clinical trials with cancer immunotherapy may herald good clinical prospects. But there are still many challenges in the broad implementation of immunotherapy. Such as the immunotherapy cannot act on all tumors, and it has serious adverse effects including but not limited to nonspecific and autoimmunity inflammation. Here, we center on recent progress made within the last 5 years in cancer immunotherapy. And we discuss the theoretical background, as well as the opportunities and challenges of cancer immunotherapy.
Collapse
|
10
|
Ye J, Chen J. Interferon and Hepatitis B: Current and Future Perspectives. Front Immunol 2021; 12:733364. [PMID: 34557195 PMCID: PMC8452902 DOI: 10.3389/fimmu.2021.733364] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection remains a major health burden worldwide for which there is still no effective curative treatment. Interferon (IFN) consists of a group of cytokines with antiviral activity and immunoregulatory and antitumor effects, that play crucial roles in both innate and adaptive immune responses. IFN-α and its pegylated form have been used for over thirty years to treat chronic hepatitis B (CHB) with advantages of finite treatment duration and sustained virologic response, however, the efficacy is limited and side effects are common. Here, we summarize the status and unique advantages of IFN therapy against CHB, review the mechanisms of IFN-α action and factors affecting IFN response, and discuss the possible improvement of IFN-based therapy and the rationale of combinations with other antiviral agents in seeking an HBV cure.
Collapse
Affiliation(s)
- Jianyu Ye
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Research Unit of Cure of Chronic Hepatitis B Virus Infection, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
11
|
Nash A, Aghlara-Fotovat S, Hernandez A, Scull C, Veiseh O. Clinical translation of immunomodulatory therapeutics. Adv Drug Deliv Rev 2021; 176:113896. [PMID: 34324885 PMCID: PMC8567306 DOI: 10.1016/j.addr.2021.113896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Immunomodulatory therapeutics represent a unique class of drug products that have tremendous potential to rebalance malfunctioning immune systems and are quickly becoming one of the fastest-growing areas in the pharmaceutical industry. For these drugs to become mainstream medicines, they must provide greater therapeutic benefit than the currently used treatments without causing severe toxicities. Immunomodulators, cell-based therapies, antibodies, and viral therapies have all achieved varying amounts of success in the treatment of cancers and/or autoimmune diseases. However, many challenges related to precision dosing, off-target effects, and manufacturing hurdles will need to be addressed before we see widespread adoption of these therapies in the clinic. This review provides a perspective on the progress of immunostimulatory and immunosuppressive therapies to date and discusses the opportunities and challenges for clinical translation of the next generation of immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Amanda Nash
- Rice University, Department of Bioengineering, Houston TX, United States
| | | | - Andrea Hernandez
- Rice University, Department of Bioengineering, Houston TX, United States
| | | | - Omid Veiseh
- Rice University, Department of Bioengineering, Houston TX, United States.
| |
Collapse
|
12
|
Tenesaca S, Vasquez M, Alvarez M, Otano I, Fernandez-Sendin M, Di Trani CA, Ardaiz N, Gomar C, Bella A, Aranda F, Medina-Echeverz J, Melero I, Berraondo P. Statins act as transient type I interferon inhibitors to enable the antitumor activity of modified vaccinia Ankara viral vectors. J Immunother Cancer 2021; 9:jitc-2020-001587. [PMID: 34321273 PMCID: PMC8320251 DOI: 10.1136/jitc-2020-001587] [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] [Accepted: 06/27/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Modified vaccinia virus Ankara (MVA) are genetically engineered non-replicating viral vectors. Intratumoral administration of MVA induces a cyclic GMP-AMP synthase-mediated type I interferon (IFN) response and the production of high levels of the transgenes engineered into the viral genome such as tumor antigens to construct cancer vaccines. Although type I IFNs are essential for establishing CD8-mediated antitumor responses, this cytokine family may also give rise to immunosuppressive mechanisms. METHODS In vitro assays were performed to evaluate the activity of simvastatin and atorvastatin on type I IFN signaling and on antigen presentation. Surface levels of IFN α/β receptor 1, endocytosis of bovine serum albumin-fluorescein 5 (6)-isothiocyanate, signal transducer and activator of transcription (STAT) phosphorylation, and real-time PCR of IFN-stimulated genes were assessed in the murine fibroblast cell line L929. In vivo experiments were performed to characterize the effect of simvastatin on the MVA-induced innate immune response and on the antitumor effect of MVA-based antitumor vaccines in B16 melanoma expressing ovalbumin (OVA) and Lewis lung carcinoma (LLC)-OVA tumor models. RNAseq analysis, depleting monoclonal antibodies, and flow cytometry were used to evaluate the MVA-mediated immune response. RESULTS In this work, we identified commonly prescribed statins as potent IFNα pharmacological inhibitors due to their ability to reduce surface expression levels of IFN-α/β receptor 1 and to reduce clathrin-mediated endocytosis. Simvastatin and atorvastatin efficiently abrogated for 8 hours the transcriptomic response to IFNα and enhanced the number of dendritic cells presenting an OVA-derived peptide bound to major histocompatibility complex (MHC) class I. In vivo, intraperitoneal or intramuscular administration of simvastatin reduced the inflammatory response mediated by peritumoral administration of MVA and enhanced the antitumor activity of MVA encoding tumor-associated antigens. The synergistic antitumor effects critically depend on CD8+ cells, whereas they were markedly improved by depletion of CD4+ lymphocytes, T regulatory cells, or NK cells. Either MVA-OVA alone or combined with simvastatin augmented B cells, CD4+ lymphocytes, CD8+ lymphocytes, and tumor-specific CD8+ in the tumor-draining lymph nodes. However, only the treatment combination increased the numbers of these lymphocyte populations in the tumor microenvironment and in the spleen. CONCLUSION In conclusion, blockade of IFNα functions by simvastatin markedly enhances lymphocyte infiltration and the antitumor activity of MVA, prompting a feasible drug repurposing.
Collapse
Affiliation(s)
- Shirley Tenesaca
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Marcos Vasquez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Maite Alvarez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Itziar Otano
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Angela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | | | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain .,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
13
|
Hoteit M, Oneissi Z, Reda R, Wakim F, Zaidan A, Farran M, Abi-Khalil E, El-Sibai M. Cancer immunotherapy: A comprehensive appraisal of its modes of application. Oncol Lett 2021; 22:655. [PMID: 34386077 DOI: 10.3892/ol.2021.12916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022] Open
Abstract
Conventional cancer treatments such as chemotherapy and radiation therapy have reached their therapeutic potential, leaving a gap for developing more effective cancer therapeutics. Cancer cells evade the immune system using various mechanisms of immune tolerance, underlying the potential impact of immunotherapy in the treatment of cancer. Immunotherapy includes several approaches such as activating the immune system in a cytokine-dependent manner, manipulating the feedback mechanisms involved in the immune response, enhancing the immune response via lymphocyte expansion and using cancer vaccines to elicit long-lasting, robust responses. These techniques can be used as monotherapies or combination therapies. The present review describes the immune-based mechanisms involved in tumor cell proliferation and maintenance and the rationale underlying various treatment methods. In addition, the present review provides insight into the potential of immunotherapy used alone or in combination with various types of therapeutics.
Collapse
Affiliation(s)
- Mira Hoteit
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Zeina Oneissi
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Ranim Reda
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Fadi Wakim
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Amar Zaidan
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Mohammad Farran
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Elie Abi-Khalil
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| |
Collapse
|
14
|
Construction of Recombinant Human GM-CSF and GM-CSF-ApoA-I Fusion Protein and Evaluation of Their Biological Activity. Pharmaceuticals (Basel) 2021; 14:ph14050459. [PMID: 34068113 PMCID: PMC8152757 DOI: 10.3390/ph14050459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/01/2021] [Accepted: 05/07/2021] [Indexed: 12/04/2022] Open
Abstract
In this study, two strains of the yeast P. pastoris were constructed, one of which produced authentic recombinant human granulocyte-macrophage colony-stimulating factor (ryGM-CSF), and the other was a chimera consisting of ryGM-CSF genetically fused with mature human apolipoprotein A-I (ApoA-I) (ryGM-CSF-ApoA-I). Both forms of the cytokine were secreted into the culture medium. The proteins’ yield during cultivation in flasks was 100 and 60 mg/L for ryGM-CSF and ryGM-CSF-ApoA-I, respectively. Both forms of recombinant GM-CSF stimulated the proliferation of human TF-1 erythroleukemia cells; however, the amount of chimera required was 10-fold that of authentic GM-CSF to induce a similar proliferative effect. RyGM-CSF exhibited a 2-fold proliferative effect on BFU-E (burst-forming units—erythroid) at a concentration 1.7 fold less than non-glycosylated E. coli-derived GM-CSF. The chimera together with authentic ryGM-CSF increased the number of both erythroid precursors and BMC granulocytes after 48 h of incubation of human bone marrow cells (BMCs). In addition, the chimeric form of ryGM-CSF was more effective at increasing the viability of the total amount of BMCs, decreasing apoptosis compared to the authentic form. ryGM-CSF-ApoA-I normalized the proliferation, maturation, and segmentation of neutrophils within the physiological norm, preserving the pool of blast cells under conditions of impaired granulopoiesis. The chimera form of GM-CSF exhibited the properties of a multilinear growth factor, modulating the activity of GM-CSF and, perhaps, it may be more suitable for the normalization of granulopoiesis.
Collapse
|
15
|
Filin IY, Solovyeva VV, Kitaeva KV, Rutland CS, Rizvanov AA. Current Trends in Cancer Immunotherapy. Biomedicines 2020; 8:biomedicines8120621. [PMID: 33348704 PMCID: PMC7766207 DOI: 10.3390/biomedicines8120621] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
The search for an effective drug to treat oncological diseases, which have become the main scourge of mankind, has generated a lot of methods for studying this affliction. It has also become a serious challenge for scientists and clinicians who have needed to invent new ways of overcoming the problems encountered during treatments, and have also made important discoveries pertaining to fundamental issues relating to the emergence and development of malignant neoplasms. Understanding the basics of the human immune system interactions with tumor cells has enabled new cancer immunotherapy strategies. The initial successes observed in immunotherapy led to new methods of treating cancer and attracted the attention of the scientific and clinical communities due to the prospects of these methods. Nevertheless, there are still many problems that prevent immunotherapy from calling itself an effective drug in the fight against malignant neoplasms. This review examines the current state of affairs for each immunotherapy method, the effectiveness of the strategies under study, as well as possible ways to overcome the problems that have arisen and increase their therapeutic potentials.
Collapse
Affiliation(s)
- Ivan Y. Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Catrin S. Rutland
- Faculty of Medicine and Health Science, University of Nottingham, Nottingham NG7 2QL, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
- Republic Clinical Hospital, 420064 Kazan, Russia
- Correspondence: ; Tel.: +7-905-316-7599
| |
Collapse
|
16
|
GSDMD membrane pore is critical for IL-1β release and antagonizing IL-1β by hepatocyte-specific nanobiologics is a promising therapeutics for murine alcoholic steatohepatitis. Biomaterials 2020; 227:119570. [DOI: 10.1016/j.biomaterials.2019.119570] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
|
17
|
Vasquez M, Consuegra-Fernández M, Aranda F, Jimenez A, Tenesaca S, Fernandez-Sendin M, Gomar C, Ardaiz N, Di Trani CA, Casares N, Lasarte JJ, Lozano F, Berraondo P. Treatment of Experimental Autoimmune Encephalomyelitis by Sustained Delivery of Low-Dose IFN-α. THE JOURNAL OF IMMUNOLOGY 2019; 203:696-704. [PMID: 31209101 DOI: 10.4049/jimmunol.1801462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 05/31/2019] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease with no curative treatment. The immune regulatory properties of type I IFNs have led to the approval of IFN-β for the treatment of relapsing-remitting MS. However, there is still an unmet need to improve the tolerability and efficacy of this therapy. In this work, we evaluated the sustained delivery of IFN-α1, either alone or fused to apolipoprotein A-1 by means of an adeno-associated viral (AAV) system in the mouse model of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. These in vivo experiments demonstrated the prophylactic and therapeutic efficacy of the AAV-IFN-α or AAV-IFN-α fused to apolipoprotein A-1 vectors in experimental autoimmune encephalomyelitis, even at low doses devoid of hematological or neurologic toxicity. The sustained delivery of such low-dose IFN-α resulted in immunomodulatory effects, consisting of proinflammatory monocyte and T regulatory cell expansion. Moreover, encephalitogenic T lymphocytes from IFN-α-treated mice re-exposed to the myelin oligodendrocyte glycoprotein peptide in vitro showed a reduced proliferative response and cytokine (IL-17A and IFN-γ) production, in addition to upregulation of immunosuppressive molecules, such as IL-10, IDO, or PD-1. In conclusion, the results of the present work support the potential of sustained delivery of low-dose IFN-α for the treatment of MS and likely other T cell-dependent chronic autoimmune disorders.
Collapse
Affiliation(s)
- Marcos Vasquez
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Marta Consuegra-Fernández
- Institut d'Investigacions Biomédiques August Pi i Sunyer, Barcelona 08036, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona 08036, Spain.,Departament de Biomedicina, Universitat de Barcelona, Barcelona 08007, Spain; and
| | - Fernando Aranda
- Institut d'Investigacions Biomédiques August Pi i Sunyer, Barcelona 08036, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona 08036, Spain.,Departament de Biomedicina, Universitat de Barcelona, Barcelona 08007, Spain; and
| | - Aitor Jimenez
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Shirley Tenesaca
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Noelia Casares
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Juan Jose Lasarte
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain.,Navarra Institute for Health Research, Pamplona 31008, Spain
| | - Francisco Lozano
- Institut d'Investigacions Biomédiques August Pi i Sunyer, Barcelona 08036, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona 08036, Spain.,Departament de Biomedicina, Universitat de Barcelona, Barcelona 08007, Spain; and
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima University of Navarra, Pamplona 31008, Spain; .,Navarra Institute for Health Research, Pamplona 31008, Spain.,Centro de Investigación Biomédica en Red de Cáncer, Madrid 28029, Spain
| |
Collapse
|
18
|
Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, Rodríguez-Ruiz ME, Ponz-Sarvise M, Castañón E, Melero I. Cytokines in clinical cancer immunotherapy. Br J Cancer 2019; 120:6-15. [PMID: 30413827 PMCID: PMC6325155 DOI: 10.1038/s41416-018-0328-y] [Citation(s) in RCA: 779] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Cytokines are soluble proteins that mediate cell-to-cell communication. Based on the discovery of the potent anti-tumour activities of several pro-inflammatory cytokines in animal models, clinical research led to the approval of recombinant interferon-alpha and interleukin-2 for the treatment of several malignancies, even if efficacy was only modest. These early milestones in immunotherapy have been followed by the recent addition to clinical practice of antibodies that inhibit immune checkpoints, as well as chimeric antigen receptor T cells. A renewed interest in the anti-tumour properties of cytokines has led to an exponential increase in the number of clinical trials that explore the safety and efficacy of cytokine-based drugs, not only as single agents, but also in combination with other immunomodulatory drugs. These second-generation drugs under clinical development include known molecules with novel mechanisms of action, new targets, and fusion proteins that increase half-life and target cytokine activity to the tumour microenvironment or to the desired effector immune cells. In addition, the detrimental activity of immunosuppressive cytokines can be blocked by antagonistic antibodies, small molecules, cytokine traps or siRNAs. In this review, we provide an overview of the novel trends in the cytokine immunotherapy field that are yielding therapeutic agents for clinical trials.
Collapse
Affiliation(s)
- Pedro Berraondo
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| | - Miguel F Sanmamed
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - María C Ochoa
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Iñaki Etxeberria
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria A Aznar
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - José Luis Pérez-Gracia
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - María E Rodríguez-Ruiz
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Eduardo Castañón
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
19
|
Pastor F, Berraondo P, Etxeberria I, Frederick J, Sahin U, Gilboa E, Melero I. An RNA toolbox for cancer immunotherapy. Nat Rev Drug Discov 2018; 17:751-767. [DOI: 10.1038/nrd.2018.132] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Shan Z, Alvarez-Sola G, Uriarte I, Arechederra M, Fernández-Barrena MG, Berasain C, Ju C, Avila MA. Fibroblast growth factors 19 and 21 in acute liver damage. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:257. [PMID: 30069459 DOI: 10.21037/atm.2018.05.26] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Currently there are very few pharmacological options available to treat acute liver injury. Because its natural exposure to noxious stimuli the liver has developed a strong endogenous hepatoprotective capacity. Indeed, experimental evidence exposed a variety of endogenous hepatic and systemic responses naturally activated to protect the hepatic parenchyma and to foster liver regeneration, therefore preserving individual's survival. The fibroblast growth factor (FGF) family encompasses a range of polypeptides with important effects on cellular differentiation, growth survival and metabolic regulation in adult organisms. Among these FGFs, FGF19 and FGF21 are endocrine hormones that profoundly influence systemic metabolism but also exert important hepatoprotective activities. In this review, we revisit the biology of these factors and highlight their potential application for the clinical management of acute liver injury.
Collapse
Affiliation(s)
- Zhao Shan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, USA
| | - Gloria Alvarez-Sola
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain
| | - María Arechederra
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain
| | - Maite G Fernández-Barrena
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain
| | - Carmen Berasain
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| | - Cynthia Ju
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, USA
| | - Matías A Avila
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERehd, Carlos III Institute of Health, Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IDISNA), Pamplona, Spain
| |
Collapse
|
21
|
Chen W, Zhang X, Fan J, Zai W, Luan J, Li Y, Wang S, Chen Q, Wang Y, Liang Y, Ju D. Tethering Interleukin-22 to Apolipoprotein A-I Ameliorates Mice from Acetaminophen-induced Liver Injury. Theranostics 2017; 7:4135-4148. [PMID: 29158815 PMCID: PMC5695002 DOI: 10.7150/thno.20955] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/14/2017] [Indexed: 11/22/2022] Open
Abstract
Increasing evidence indicates that interleukin-22 (IL-22) holds tremendous potential as a protective agent in preventing liver injury, but its pleiotropic effects and pathogenic role in carcinogenesis, rheumatoid arthritis and psoriasis restrict its systemic application. Here, we first developed a nanoparticle (liposIA) as a liver-targeted agent through IL-22 tethered to apolipoprotein A-I (ApoA-I) in a gene therapy vector. LiposIA was prepared using thin film dispersion method and the complexes exhibited desirable nanoparticle size, fine polydisperse index, highly efficient transfection, and excellent serum and storage stability. Biodistribution and hepatic STAT3 phosphorylation studies revealed that IL-22 tethered to ApoA-I led to highly efficient liver targeting. More importantly, our studies showed that a single-dose of liposIA was able to protect mice against acetaminophen-induced liver injury and did not initiate inflammatory response or systemic toxicity in vivo. During this process, activated STAT3/Erk and Akt/mTOR signaling transductions were observed, as well as inhibition of reactive oxygen species (ROS) generation, which prevented mitochondrial dysfunction. These studies demonstrated that IL-22 tethered to apolipoprotein A-I could target and ameliorate acetaminophen-induced acute liver injury, which highlighted that a targeted strategy for IL-22 delivery might have broad utility for the protection of hepatocellular damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dianwen Ju
- Department of Microbiological and Biochemical Pharmacy & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| |
Collapse
|
22
|
Alvarez-Sola G, Uriarte I, Latasa MU, Jimenez M, Barcena-Varela M, Santamaría E, Urtasun R, Rodriguez-Ortigosa C, Prieto J, Berraondo P, Fernandez-Barrena MG, Berasain C, Avila MA. Bile acids, FGF15/19 and liver regeneration: From mechanisms to clinical applications. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1326-1334. [PMID: 28709961 DOI: 10.1016/j.bbadis.2017.06.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022]
Abstract
The liver has an extraordinary regenerative capacity rapidly triggered upon injury or resection. This response is intrinsically adjusted in its initiation and termination, a property termed the "hepatostat". Several molecules have been involved in liver regeneration, and among them bile acids may play a central role. Intrahepatic levels of bile acids rapidly increase after resection. Through the activation of farnesoid X receptor (FXR), bile acids regulate their hepatic metabolism and also promote hepatocellular proliferation. FXR is also expressed in enterocytes, where bile acids stimulate the expression of fibroblast growth factor 15/19 (FGF15/19), which is released to the portal blood. Through the activation of FGFR4 on hepatocytes FGF15/19 regulates bile acids synthesis and finely tunes liver regeneration as part of the "hepatostat". Here we review the experimental evidences supporting the relevance of the FXR-FGF15/19-FGFR4 axis in liver regeneration and discuss potential therapeutic applications of FGF15/19 in the prevention of liver failure. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Collapse
Affiliation(s)
- Gloria Alvarez-Sola
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain
| | - Iker Uriarte
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain
| | - Maria U Latasa
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Maddalen Jimenez
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Marina Barcena-Varela
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Eva Santamaría
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain
| | - Raquel Urtasun
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Carlos Rodriguez-Ortigosa
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain; Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Jesús Prieto
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain; Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Pedro Berraondo
- Immunology and Immunotherapy Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Maite G Fernandez-Barrena
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain; Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain
| | - Carmen Berasain
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain; Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain.
| | - Matías A Avila
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda. Pio XII, n 36, 31008 Pamplona, Spain; Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, 31008 Pamplona, Spain.
| |
Collapse
|
23
|
Vasquez M, Paredes-Cervantes V, Aranda F, Ardaiz N, Gomar C, Berraondo P. Antitumor effect of an adeno-associated virus expressing apolipoprotein A-1 fused to interferon alpha in an interferon alpha-resistant murine tumor model. Oncotarget 2017; 8:5247-5255. [PMID: 28029653 PMCID: PMC5354905 DOI: 10.18632/oncotarget.14127] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/22/2016] [Indexed: 11/25/2022] Open
Abstract
Interferon alpha (IFNα) is a cytokine approved for the treatment of several types of cancer. However, the modest effect on overall survival and the high toxicity associated with the treatment has reduced the clinical use of this cytokine. In this study, we have developed a tumor model that reproduces this clinical setting. A high dose of an adeno-associated virus encoding IFNα (AAV-IFNα) was able to eradicate a liver metastases model of colon cancer but induced lethal pancytopenia. On the other hand, a safe dose of AAV-IFNα was not able to eliminate the liver metastases of colon cancer. In this IFNα-resistant tumor model, administration of an adeno-associated vector encoding apolipoprotein A-1 fused to IFNα was able to fully eradicate the tumor in 43% of mice without toxicity. This antitumor effect was limited by suboptimal long-term CD8+ T cell activation and the expansion of T regulatory cells. In contrast, IFNα upregulated suppressor molecules such as PD-1 and interleukin 10 on CD8+ T lymphocytes. In conclusion, we show that apolipoprotein A-1 fused to IFNα is a novel antitumor drug that differs from IFNα in the modulation of suppressor mechanisms of the immune response. These differential properties pave the way for rational combinations with other immunomodulatory drugs.
Collapse
Affiliation(s)
- Marcos Vasquez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Vladimir Paredes-Cervantes
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
- Centro Médico Nacional La Raza, IMSS, México DF, Mexico
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| |
Collapse
|
24
|
Nistal-Villan E, Poutou J, Rodríguez-Garcia E, Buñuales M, Carte-Abad B, Prieto J, Gonzalez-Aseguinolaza G, Hernandez-Alcoceba R, Larrea E. A Versatile Vector for In Vivo Monitoring of Type I Interferon Induction and Signaling. PLoS One 2016; 11:e0152031. [PMID: 27007218 PMCID: PMC4805199 DOI: 10.1371/journal.pone.0152031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/08/2016] [Indexed: 12/02/2022] Open
Abstract
Development of reporter systems for in vivo examination of IFN-β induction or signaling of type I interferon (IFN-I) pathways is of great interest in order to characterize biological responses to different inducers such as viral infections. Several reporter mice have been developed to monitor the induction of both pathways in response to different agonists. However, alternative strategies that do not require transgenic mice breeding have to date not been reported. In addition, detection of these pathways in vivo in animal species other than mice has not yet been addressed. Herein we describe a simple method based on the use of an adeno-associated viral vector (AAV8-3xIRF-ISRE-Luc) containing an IFN-β induction and signaling-sensitive promoter sequence controlling the expression of the reporter gene luciferase. This vector is valid for monitoring IFN-I responses in vivo elicited by diverse stimuli in different organs. Intravenous administration of the vector in C57BL/6 mice and Syrian hamsters was able to detect activation of the IFN pathway in the liver upon systemic treatment with different pro-inflammatory agents and infection with Newcastle disease virus (NDV). In addition, intranasal instillation of AAV8-3xIRF-ISRE-Luc showed a rapid and transient IFN-I response in the respiratory tract of mice infected with the influenza A/PR8/34 virus lacking the NS1 protein. In comparison, this response was delayed and exacerbated in mice infected with influenza A/PR/8 wild type virus. In conclusion, the AAV8-3xIRF-ISRE-Luc vector offers the possibility of detecting IFN-I activation in response to different stimuli and in different animal models with no need for reporter transgenic animals.
Collapse
Affiliation(s)
- Estanislao Nistal-Villan
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Joanna Poutou
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Estefania Rodríguez-Garcia
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Maria Buñuales
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Beatriz Carte-Abad
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Jesus Prieto
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Ruben Hernandez-Alcoceba
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
- * E-mail: (EL); (RHA)
| | - Esther Larrea
- Instituto de Salud Tropical, University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, Pamplona, Spain
- * E-mail: (EL); (RHA)
| |
Collapse
|
25
|
Dysfunctional High-Density Lipoprotein: An Innovative Target for Proteomics and Lipidomics. CHOLESTEROL 2015; 2015:296417. [PMID: 26634153 PMCID: PMC4655037 DOI: 10.1155/2015/296417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
Abstract
High-Density Lipoprotein-Cholesterol (HDL-C) is regarded as an important protective factor against cardiovascular disease, with abundant evidence of an inverse relationship between its serum levels and risk of cardiovascular disease, as well as various antiatherogenic, antioxidant, and anti-inflammatory properties. Nevertheless, observations of hereditary syndromes featuring scant HDL-C concentration in absence of premature atherosclerotic disease suggest HDL-C levels may not be the best predictor of cardiovascular disease. Indeed, the beneficial effects of HDL may not depend solely on their concentration, but also on their quality. Distinct subfractions of this lipoprotein appear to be constituted by specific protein-lipid conglomerates necessary for different physiologic and pathophysiologic functions. However, in a chronic inflammatory microenvironment, diverse components of the HDL proteome and lipid core suffer alterations, which propel a shift towards a dysfunctional state, where HDL-C becomes proatherogenic, prooxidant, and proinflammatory. This heterogeneity highlights the need for further specialized molecular studies in this aspect, in order to achieve a better understanding of this dysfunctional state; with an emphasis on the potential role for proteomics and lipidomics as valuable methods in the search of novel therapeutic approaches for cardiovascular disease.
Collapse
|
26
|
Berraondo P, Di Scala M, Korolowicz K, Thampi LM, Otano I, Suarez L, Fioravanti J, Aranda F, Ardaiz N, Yang J, Kallakury BV, Tucker RD, Vasquez M, Menne S, Prieto J, González-Aseguinolaza G. Liver-directed gene therapy of chronic hepadnavirus infection using interferon alpha tethered to apolipoprotein A-I. J Hepatol 2015; 63:329-36. [PMID: 25772035 PMCID: PMC4508219 DOI: 10.1016/j.jhep.2015.02.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 02/13/2015] [Accepted: 02/23/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Current hepatitis B virus (HBV) management is challenging as treatment with nucleos(t)ide analogues needs to be maintained indefinitely and because interferon (IFN)-α therapy is associated with considerable toxicity. Previously, we showed that linking IFNα to apolipoprotein A-I generates a molecule (IA) with distinct antiviral and immunostimulatory activities which lacks the hematological toxicity of IFNα. METHODS Here, we analyse the antiviral potential of an adeno-associated vector encoding IFNα fused to apolipoprotein A-I (AAV-IA) in comparison to a vector encoding only IFNα (AAV-IFN) in two animal models of chronic hepadnavirus infection. RESULTS In HBV transgenic mice, we found that both vectors induced marked reductions in serum and liver HBV DNA and in hepatic HBV RNA but AAV-IFN caused lethal pancytopenia. Woodchucks with chronic hepatitis virus (WHV) infection that were treated by intrahepatic injection of vectors encoding the woodchuck sequences (AAV-wIFN or AAV-wIA), experienced only a slight reduction of viremia which was associated with hematological toxicity and high mortality when using AAV-wIFN, while AAV-wIA was well tolerated. However, when we tested AAV-wIA or a control vector encoding woodchuck apolipoprotein A-I (AAV-wApo) in combination with entecavir, we found that AAV-wApo-treated animals exhibited an immediate rebound of viral load upon entecavir withdrawal while, in AAV-wIA-treated woodchucks, viremia and antigenemia remained at low levels for several weeks following entecavir interruption. CONCLUSIONS Treatment with AAV-IA is safe and elicits antiviral effects in animal models with difficult to treat chronic hepadnavirus infection. AAV-IA in combination with nucleos(t)ide analogues represents a promising approach for the treatment of HBV infection in highly viremic patients.
Collapse
Affiliation(s)
- Pedro Berraondo
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain.
| | - Marianna Di Scala
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Kyle Korolowicz
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Linta M Thampi
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Itziar Otano
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Lester Suarez
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Jessica Fioravanti
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Fernando Aranda
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Nuria Ardaiz
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Junming Yang
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Bhaskar V Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Robin D Tucker
- Division of Comparative Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Marcos Vasquez
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Stephan Menne
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - Gloria González-Aseguinolaza
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
27
|
Medina-Echeverz J, Vasquez M, Gomar C, Ardaiz N, Berraondo P. Overexpression of apolipoprotein A-I fused to an anti-transforming growth factor beta peptide modulates the tumorigenicity and immunogenicity of mouse colon cancer cells. Cancer Immunol Immunother 2015; 64:717-25. [PMID: 25795134 PMCID: PMC11028610 DOI: 10.1007/s00262-015-1681-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/06/2015] [Indexed: 02/01/2023]
Abstract
Transforming growth factor beta (TGF-β) promotes tumor growth, invasion and metastasis in established tumors. In this study, we analyzed the effect of overexpressing an anti-TGF-β peptide fused to apolipoprotein A-I (ApoA-I) as a scaffold molecule. We generated and characterized stable MC38 colon carcinoma clones expressing ApoA-I fused to the anti-TGF-β peptide P144 and ApoA-I as control cells. We evaluated in vitro the gene expression profile, cell cycle and anchorage-independent growth. The in vivo tumorigenic potential and immunogenicity were analyzed inoculating the MC38 clones into C57BL/6 mice, recombination-activating gene 1 knockout mice or mice deficient in NK cells either subcutaneously or intrasplenically to generate hepatic metastases. While overexpression of ApoA-I had no effect on the parameters analyzed, ApoA-I fused to P144 markedly diminished the tumorigenic capacity and metastatic potential of MC38 in vitro and in vivo, thus generating a highly immunogenic cell line. MC38 cells transfected with ApoA-I fused to P144 triggered memory T cell responses able to eliminate the parental cell line upon re-challenge. In summary, expression of ApoA-I fused to P144 is a novel strategy to modulate TGF-β in tumor cells. These results highlight the potential of TGF-β as a target in the development of new antitumor treatments.
Collapse
Affiliation(s)
- José Medina-Echeverz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Avda. Pio XII 55, 31008 Pamplona, Spain
| | - Marcos Vasquez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Avda. Pio XII 55, 31008 Pamplona, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Avda. Pio XII 55, 31008 Pamplona, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Avda. Pio XII 55, 31008 Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Avda. Pio XII 55, 31008 Pamplona, Spain
| |
Collapse
|
28
|
Ochoa MC, Melero I, Berraondo P. High-density lipoproteins delivering interleukin-15. Oncoimmunology 2014; 2:e23410. [PMID: 23734302 PMCID: PMC3654572 DOI: 10.4161/onci.23410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 12/23/2012] [Indexed: 12/03/2022] Open
Abstract
Circulating lipoproteins may offer interesting properties as therapeutic carriers for cytokines and hormones, in terms of both stability and bio-distribution. The fusion of apolipoprotein A-I with interleukin-15 (IL-15) targets the latter to high-density lipoproteins (HDLs). The bioactivity of this chimera can be further enhanced by creating triple fusions with IL-15 receptor α domain involved in IL-15 trans-presentation.
Collapse
Affiliation(s)
- Maria C Ochoa
- Division of Hepatology and Gene Therapy; Center for Applied Medical Research, and University Clinic; University of Navarra, Pamplona; Navarra, Spain
| | | | | |
Collapse
|
29
|
Medina-Echeverz J, Fioravanti J, Díaz-Valdés N, Frank K, Aranda F, Gomar C, Ardaiz N, Dotor J, Umansky V, Prieto J, Berraondo P. Harnessing high density lipoproteins to block transforming growth factor beta and to inhibit the growth of liver tumor metastases. PLoS One 2014; 9:e96799. [PMID: 24797128 PMCID: PMC4010484 DOI: 10.1371/journal.pone.0096799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/11/2014] [Indexed: 11/30/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a powerful promoter of cancer progression and a key target for antitumor therapy. As cancer cells exhibit active cholesterol metabolism, high density lipoproteins (HDLs) appear as an attractive delivery system for anticancer TGFβ-inhibitory molecules. We constructed a plasmid encoding a potent TGF-β-blocking peptide (P144) linked to apolipoprotein A-I (ApoA-I) through a flexible linker (pApoLinkerP144). The ApoLinkerP144 sequence was then incorporated into a hepatotropic adeno-associated vector (AAVApoLinkerP144). The aim was to induce hepatocytes to produce HDLs containing a modified ApoA-I capable of blocking TGF-β. We observed that transduction of the murine liver with pApoLinkerP144 led to the appearance of a fraction of circulating HDL containing the fusion protein. These HDLs were able to attenuate TGF-β signaling in the liver and to enhance IL-12 -mediated IFN-γ production. Treatment of liver metastasis of MC38 colorectal cancer with AAVApoLinkerP144 resulted in a significant reduction of tumor growth and enhanced expression of IFN-γ and GM-CSF in cancerous tissue. ApoLinkerP144 also delayed MC38 liver metastasis in Rag2−/−IL2rγ−/− immunodeficient mice. This effect was associated with downregulation of TGF-β target genes essential for metastatic niche conditioning. Finally, in a subset of ret transgenic mice, a model of aggressive spontaneous metastatic melanoma, AAVApoLinkerP144 delayed tumor growth in association with increased CD8+ T cell numbers in regional lymph nodes. In conclusion, modification of HDLs to transport TGF-β-blocking molecules is a novel and promising approach to inhibit the growth of liver metastases by immunological and non-immunological mechanisms.
Collapse
Affiliation(s)
- José Medina-Echeverz
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Jessica Fioravanti
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Nancy Díaz-Valdés
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Skin Cancer Unit, German Cancer Research Center, Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Heidelberg, Germany
| | - Kathrin Frank
- Skin Cancer Unit, German Cancer Research Center, Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Heidelberg, Germany
| | - Fernando Aranda
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Celia Gomar
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Nuria Ardaiz
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center, Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Heidelberg, Germany
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Liver Unit, University of Navarra Clinic, Networked Biomedical Research Center of Hepatic and Digestive Diseases, Pamplona, Spain
- * E-mail: (PB); (JP)
| | - Pedro Berraondo
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- * E-mail: (PB); (JP)
| |
Collapse
|
30
|
Evaluation of adverse effects of mutein forms of recombinant human interferon alpha-2b in female swiss webster mice. BIOMED RESEARCH INTERNATIONAL 2013; 2013:943687. [PMID: 23738335 PMCID: PMC3659462 DOI: 10.1155/2013/943687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/30/2013] [Indexed: 12/02/2022]
Abstract
Purpose. We successfully developed recombinant human interferon alpha-2b (rhIFN-α2b) and mutein forms through the site-directed mutagenesis technique. The mutein forms were developed by substituting cysteins at positions 2 and 99 with aspartic acids. The potential adverse effects of these rhIFN-α2bs were assessed by acute and subchronic studies. Methods. In the acute study, rhIFN-α2bs were subcutaneously administered to mice at a single dose of 97.5 μg/kg, 975 μg/kg, and 9.75 mg/kg BW and were observed for 14 days. In the subchronic study, single dose of 1.95 μg/kg and 19.5 μg/kg, respectively, was given subcutaneously every 3 days for 45 days. Results. No death as well as abnormality in body weight, behavior, presentation of main organs, and value of plasma SGPT and SGOT was observed. Wild type and mutein rhIFN-α2bs did not show significant adverse effects at dose up to 9.75 mg/kg BW. Administration of these rhIFN-α2bs given repeatedly did not induce any adverse effect. Conclusion. These results suggest that our rhIFN-α2bs are safe. However, further study is still needed to clarify the safety issue before use in clinical trial.
Collapse
|
31
|
Hernandez-Alcoceba R, Sangro B, Berraondo P, Gonzalez-Aseguinolaza G, Prieto J. Cytokines for the treatment of gastrointestinal cancers: clinical experience and new perspectives. Expert Opin Investig Drugs 2013; 22:827-41. [PMID: 23594171 DOI: 10.1517/13543784.2013.793307] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Cytokines are key mediators of the immune system and have been proposed as therapeutic agents against cancer, either as recombinant proteins, or as transgenes in gene therapy approaches. Stimulation of immune responses against cancer cells is an appealing method to treat tumors with high risk of relapse and systemic dissemination. AREAS COVERED We provide a critical overview of clinical trials involving the use of cytokines for the treatment of liver, colon and pancreatic cancers. Special attention has been paid to advances in the field of gene therapy and oncolytic viruses. The potential of new developments still in a pre-clinical stage is also discussed. We have revised public sources of information (PubMed, US National Institutes of Health clinical trials database) up to January 2013. EXPERT OPINION The complexity of the immune system and the unfavorable pharmacokinetic properties of cytokines limit the efficacy of these molecules as single agents for the treatment of cancer. Expression from gene therapy vectors, together with new methods of targeting and stabilization, may overcome these hurdles. We believe cytokines will play a crucial role as part of combined approaches, enhancing the action of adoptive cell immunotherapy, oncolytic viruses or biological therapies.
Collapse
Affiliation(s)
- Ruben Hernandez-Alcoceba
- CIMA, University of Navarra, Division of Hepatology and Gene Therapy, Foundation for Applied Medical Research, Pamplona, Spain
| | | | | | | | | |
Collapse
|
32
|
Liver gene transfer of interkeukin-15 constructs that become part of circulating high density lipoproteins for immunotherapy. PLoS One 2012; 7:e52370. [PMID: 23285013 PMCID: PMC3528770 DOI: 10.1371/journal.pone.0052370] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 11/13/2012] [Indexed: 12/11/2022] Open
Abstract
Apolipoprotein A-I (Apo A-I) is a major component of high density lipoproteins (HDL) that transport cholesterol in circulation. We have constructed an expression plasmid encoding a chimeric molecule encompassing interleukin-15 (IL-15) and Apo A-I (pApo-hIL15) that was tested by hydrodynamic injections into mice and was co-administered with a plasmid encoding the sushi domain of IL-15Rα (pSushi) in order to enhance IL-15 trans-presentation and thereby bioactivity. The pharmacokinetics of the Apo A-I chimeric protein were much longer than non-stabilized IL-15 and its bioactivity was enhanced in combination with IL-15Rα Sushi. Importantly, the APO-IL-15 fusion protein was incorporated in part into circulating HDL. Liver gene transfer of these constructs increased NK and memory-phenotype CD8 lymphocyte numbers in peripheral blood, spleen and liver as a result of proliferation documented by CFSE dilution and BrdU incorporation. Moreover, the gene transfer procedure partly rescued the NK and memory T-cell deficiency observed in IL-15Rα−/− mice. pApo-hIL15+ pSushi gene transfer to the liver showed a modest therapeutic activity against subcutaneously transplanted MC38 colon carcinoma tumors, that was more evident when tumors were set up as liver metastases. The improved pharmacokinetic profile and the strong biological activity of APO-IL-15 fusion protein holds promise for further development in combination with other immunotherapies.
Collapse
|
33
|
Ochoa MC, Fioravanti J, Rodriguez I, Hervas-Stubbs S, Azpilikueta A, Mazzolini G, Gúrpide A, Prieto J, Pardo J, Berraondo P, Melero I. Antitumor Immunotherapeutic and Toxic Properties of an HDL-Conjugated Chimeric IL-15 Fusion Protein. Cancer Res 2012; 73:139-49. [DOI: 10.1158/0008-5472.can-12-2660] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Parra-Guillen ZP, Fioravanti J, Medina-Echeverz J, Gomar C, Ardaiz N, Troconiz IF, Berraondo P. Kinetic and dynamic computational model-based characterization of new proteins in mice: application to interferon alpha linked to apolipoprotein A-I. PLoS One 2012; 7:e42100. [PMID: 22848716 PMCID: PMC3407104 DOI: 10.1371/journal.pone.0042100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/02/2012] [Indexed: 02/06/2023] Open
Abstract
Interferon alpha linked to apolipoprotein A-I has been recently proposed as an improved interferon-based therapy. In the present study, we aimed to develop a computational model to gain further insight into the in vivo behaviour of this new fusion protein. In order to facilitate in vivo evaluation of interferon and the fusion protein without altering their biological properties, green fluorescent protein was incorporated into their structures. Kinetic and dynamic behaviour of both compounds was successfully described after plasmid hydrodynamic administration and in situ synthesis of the studied proteins. Results from the modelling exercise showed that apolipoprotein A-I conferred a modified kinetic behaviour, varying molecule distribution and prolonging half-life without altering liver dynamic performance. However, differences in the gene expression activity were observed at brain level between both compounds. Those differences could be explained by modifications in the dynamic, but also in the biodistribution properties, which would be worth evaluating in future experiments. Therefore, the modelling approach provided a global comprehension of a complex system and allowed us to compare the in vivo behaviour of both compounds and to identify critical aspects that might be important to understand the system better and suggests a need for new model-based experiments.
Collapse
Affiliation(s)
- Zinnia Patricia Parra-Guillen
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Navarra, Spain
| | - Jessica Fioravanti
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research (CIMA), Pamplona, Navarra, Spain
| | - Jose Medina-Echeverz
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research (CIMA), Pamplona, Navarra, Spain
| | - Celia Gomar
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research (CIMA), Pamplona, Navarra, Spain
| | - Nuria Ardaiz
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research (CIMA), Pamplona, Navarra, Spain
| | - Iñaki F. Troconiz
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Navarra, Spain
| | - Pedro Berraondo
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research (CIMA), Pamplona, Navarra, Spain
- * E-mail:
| |
Collapse
|
35
|
Fioravanti J, Medina-Echeverz J, Ardaiz N, Gomar C, Parra-Guillén ZP, Prieto J, Berraondo P. The fusion protein of IFN-α and apolipoprotein A-I crosses the blood-brain barrier by a saturable transport mechanism. THE JOURNAL OF IMMUNOLOGY 2012; 188:3988-92. [PMID: 22422884 DOI: 10.4049/jimmunol.1101598] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-α is widely used for the treatment of chronic viral hepatitis and malignancies. However, systemic IFN-α treatment causes severe neuropsychiatric complications in humans, including depression, anxiety, and cognitive impairments. We have previously reported that the fusion protein formed by IFN-α and apolipoprotein A-I (IA) circulates bound to high-density lipoproteins (HDLs) and exhibits liver targeting, increased half-life, enhanced immunostimulatory activity, and reduced cytotoxicity. As the transport of HDLs across the blood-brain barrier is a highly complex and regulated process, in this study, we examine the effects of IA on the brain. Determination of IFN-α in brain and serum after hydrodynamic administration of different doses of a plasmid encoding IFN-α or IA showed that IA penetrated into the brain by a saturable transport mechanism. Thus, at high serum levels of the transgenes, the induction of IFN-sensitive genes and the number of phospho-STAT1(+) cell nuclei in the brain were substantially higher with IFN-α than with IA. This was associated with attenuation of neurodepression in mice given IA, as manifested by shorter immobility time in the tail suspension test. However, when given low doses of rIFN-α or the same antiviral units of HDLs containing IA, the induction of IFN-stimulated genes in the brain was significantly greater with the latter. In conclusion, IA crosses the blood-brain barrier not by diffusion, as is the case of IFN-α, but by a facilitated saturable transport mechanism. Thus, linkage to apolipoprotein A-I may serve to modulate the effects of IFN-α on the CNS.
Collapse
Affiliation(s)
- Jessica Fioravanti
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra 31008, Spain
| | | | | | | | | | | | | |
Collapse
|