1
|
Kim JH, Lee Y, Ahn S, Koh D, Lim Y, Lee YH, Bae DH, Shin SY. Design, Synthesis, and Biological Evaluation of Aryl Pyrazolopyrimidines as Toll-Like Receptor 7 Agonists. Chem Biol Drug Des 2025; 105:e70056. [PMID: 39887539 DOI: 10.1111/cbdd.70056] [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: 07/05/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Compounds containing pyrazolopyrimidine scaffolds were designed and synthesized as toll-like receptor 7 (TLR7) agonists. Thirty-three compounds, including 22 novel compounds, were prepared, and their structures were identified using nuclear magnetic resonance spectroscopy and mass spectrometry. TLR7 agonist activity was determined in HEK-Blue hTLR7 reporter cells. Among the compounds tested, 2-((4-methoxyphenyl)amino)-7-(pyridin-2-yl)pyrazolo[1,5-a]pyrimidine-3-carbonitrile showed the highest activity, and further in vitro biological experiments were performed using this compound. Treatment with the title compound activated the TLR7-mediated NF-κB pathway, triggering the IRAK4-IKKα/β-IκBα-p65 NF-κB signaling cascade, which led to an increase in the expression of NF-κB-regulated innate cytokines such as TNFα and IL-1β in RAW264.7 macrophages. These findings suggest that the title compound acts as a TLR7 agonist and enhances the innate immune response.
Collapse
Affiliation(s)
- Ji Hwan Kim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Korea
| | - Youngshim Lee
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Korea
| | - Seunghyun Ahn
- Department of Applied Chemistry, Dongduk Women's University, Seoul, Korea
| | - Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul, Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, Korea
| | - Young Han Lee
- Department of Biological Sciences, Konkuk University, Seoul, Korea
| | - Dong-Ho Bae
- Department of Food Sciences and Biotechnology of Animal Resources, Konkuk University, Seoul, Korea
| | - Soon Young Shin
- Department of Biological Sciences, Konkuk University, Seoul, Korea
- Cancer and Metabolism Institute, Konkuk University, Seoul, Korea
| |
Collapse
|
2
|
Kim M, Noh K, Kim P, Kim JH, Choi BW, Singh R, Choi JH, Han SB, Kim SS, Lee EY, Bae MA, Shin D, Kim M, Ahn JH. Design, Synthesis, and Biological Evaluation of New 2,6,7-Substituted Purine Derivatives as Toll-like Receptor 7 Agonists for Intranasal Vaccine Adjuvants. J Med Chem 2024; 67:9389-9405. [PMID: 38787938 DOI: 10.1021/acs.jmedchem.4c00489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
TLR7/8 agonists are versatile immune stimulators capable of treating various diseases such as viral infections, autoimmune, and cancer. Despite the structural similarity of TLR7/8, their immune stimulation mechanisms and time-course responses significantly differ. In this study, a new series of TLR7-selective agonists was synthesized utilizing the economical building block 2,6-dichloropurine. Compound 27b showed the most potent activity on hTLR7 with an EC50 of 17.53 nM and demonstrated high hTLR7 selectivity (224 folds against TLR8). 27b effectively stimulated the secretion of proinflammatory cytokines in mouse macrophages and enhanced intranasal vaccine efficacy against influenza A virus in vivo. Assessment of humoral and mucosal antibody titers confirmed that 27b elevates IgG and IgA levels, protecting against both homologous and heterologous influenza viral infections. These findings suggest that 27b is a promising candidate as a vaccine adjuvant to prevent viral infections or as a robust immunomodulator with prolonged activity for treating immune-suppressed diseases.
Collapse
Affiliation(s)
- Morgan Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kyungseob Noh
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Pyeongkeun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jae Ho Kim
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Byeong Wook Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ravi Singh
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jun-Ho Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seong Soon Kim
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Eun-Young Lee
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Myung Ae Bae
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Daeho Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
3
|
Saleki K, Alijanizadeh P, Javanmehr N, Rezaei N. The role of Toll-like receptors in neuropsychiatric disorders: Immunopathology, treatment, and management. Med Res Rev 2024; 44:1267-1325. [PMID: 38226452 DOI: 10.1002/med.22012] [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: 04/08/2022] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Neuropsychiatric disorders denote a broad range of illnesses involving neurology and psychiatry. These disorders include depressive disorders, anxiety, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder, autism spectrum disorders, headaches, and epilepsy. In addition to their main neuropathology that lies in the central nervous system (CNS), lately, studies have highlighted the role of immunity and neuroinflammation in neuropsychiatric disorders. Toll-like receptors (TLRs) are innate receptors that act as a bridge between the innate and adaptive immune systems via adaptor proteins (e.g., MYD88) and downstream elements; TLRs are classified into 13 families that are involved in normal function and illnesses of the CNS. TLRs expression affects the course of neuropsychiatric disorders, and is influenced during their pharmacotherapy; For example, the expression of multiple TLRs is normalized during the major depressive disorder pharmacotherapy. Here, the role of TLRs in neuroimmunology, treatment, and management of neuropsychiatric disorders is discussed. We recommend longitudinal studies to comparatively assess the cell-type-specific expression of TLRs during treatment, illness progression, and remission. Also, further research should explore molecular insights into TLRs regulation and related pathways.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
- Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
4
|
Song Y, Fan W, Yao C, Wang H, Lu X, Wang Y, Liu P, Ma Y, Zhang Z, Wang J, Chu B, Shi L, Yang G, Wang M. Design, synthesis and biological evaluation of quinazoline and pyrrolo[3,2- d]pyrimidine derivatives as TLR7 agonists for antiviral agents. Org Biomol Chem 2024; 22:2764-2773. [PMID: 38497199 DOI: 10.1039/d4ob00048j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Pattern recognition receptors (PRRs) play a critical role in the innate immune response, and toll-like receptor 7 (TLR7) is an important member of PRRs. Although several TLR7 agonists are available, most of them are being tested clinically, with only one available on the market. Thus, it is imperative to develop new TLR7 agonists. In this study, we designed and synthesized three kinds of quinazoline derivatives and five kinds of pyrrolo[3,2-d]pyrimidine derivatives targeting TLR7. The antiviral efficacy of these compounds was evaluated in vitro and in vivo. Our findings indicated that four kinds of compounds showed exceptional antiviral activity. Furthermore, molecular docking studies confirmed that compound 11 successfully positioned itself in the pocket of the TLR7 guanosine loading site with a binding energy of -4.45 kcal mol-1. These results suggested that these compounds might be potential antiviral agents.
Collapse
Affiliation(s)
- Yue Song
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Molecule Biology Laboratory of Zhengzhou Normal University, Zhengzhou, Henan, 450044, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Wenjie Fan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Chen Yao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Heng Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Xiuxiang Lu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Yumin Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Pengxiang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Yanjie Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Zhen Zhang
- Molecule Biology Laboratory of Zhengzhou Normal University, Zhengzhou, Henan, 450044, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - BeiBei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Lijun Shi
- College of Sciences, Henan Agricultural University, Zhengzhou, Henan, 450046, China.
| | - Guoyu Yang
- College of Food and Bioengineering, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, 450046, China.
- Key Laboratories of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, Henan Province, 450046, China
| | - Mengdi Wang
- College of Food and Bioengineering, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
5
|
Liu YS, Wang JX, Jin GY, Hu MH, Wang XD. Combination Therapy with a TLR7 Agonist and a BRD4 Inhibitor Suppresses Tumor Growth via Enhanced Immunomodulation. Int J Mol Sci 2024; 25:663. [PMID: 38203835 PMCID: PMC10779224 DOI: 10.3390/ijms25010663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
JQ-1 is a typical BRD4 inhibitor with the ability to directly fight tumor cells and evoke antitumor immunity via reducing the expression of PD-L1. However, problems arise with the development of JQ-1 in clinical trials, such as marked lymphoid and hematopoietic toxicity, leading to the investigation of combination therapy. SZU-101 is a TLR7 agonist designed and synthesized by our group with potent immunostimulatory activity. Therefore, we hypothesized that combination therapy of SZU-101 and JQ-1 would target innate immunity and adaptive immunity simultaneously, to achieve a better antitumor efficacy than monotherapy. In this study, the repressive effects of the combination administration on tumor growth and metastasis were demonstrated in both murine breast cancer and melanoma models. In 4T1 tumor-bearing mice, i.t. treatment with SZU-101 in combination with i.p. treatment with JQ-1 suppressed the growth of tumors at both injected and uninjected sites. Combination therapy increased M1/M2 ratio in TAMs, decreased PD-L1 expression and promoted the recruitment of activated CD8+ T cells in the TME. In summary, the improved therapeutic efficacy of the novel combination therapy appears to be feasible for the treatment of a diversity of cancers.
Collapse
Affiliation(s)
| | | | | | - Ming-Hao Hu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen 518060, China; (Y.-S.L.); (J.-X.W.); (G.-Y.J.)
| | - Xiao-Dong Wang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen 518060, China; (Y.-S.L.); (J.-X.W.); (G.-Y.J.)
| |
Collapse
|
6
|
Li F, Song B, Zhou WF, Chu LJ. Toll-Like Receptors 7/8: A Paradigm for the Manipulation of Immunologic Reactions for Immunotherapy. Viral Immunol 2023; 36:564-578. [PMID: 37751284 DOI: 10.1089/vim.2023.0077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
The innate immune system recognizes conserved features of viral and microbial pathogens through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are one type of PRR used by the innate immune system to mediate the secretion of proinflammatory cytokines and promote innate and adaptive immune responses. TLR family members TLR7 and TLR8 (referred to as TLR7/8 from herein) are endosomal transmembrane receptors that recognize purine-rich single-stranded RNA (ssRNA) and bacterial DNA, eliciting an immunologic reaction to pathogens. TLR7/8 were discovered to mediate the secretion of proinflammatory cytokines by activating immune cells. In addition, accumulating evidence has indicated that TLR7/8 may be closely related to numerous immune-mediated disorders, specifically several types of cancer, autoimmune disease, and viral disease. TLR7/8 agonists and antagonists, which are used as drugs or adjuvants, have been identified in preclinical studies and clinical trials as promising immune stimulators for the immunotherapy of these immune-mediated disorders. These results provided reasoning to further explore immunotherapy for the treatment of immune-mediated disorders. Nevertheless, numerous needs remain unmet, and the therapeutic effects of TLR7/8 agonists and antagonists are poor and exert strong immune-related toxicities. The present review aimed to provide an overview of the TLR family members, particularly TLR7/8, and address the underlying molecular mechanisms and clinical implications of TLR7/8 in immune-mediated disorders. The aim of the work is to discuss the underlying molecular mechanisms and clinical implications of TLR7/8 in immune-mediated disorders.
Collapse
Affiliation(s)
- Fang Li
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Biao Song
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei-Feng Zhou
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Li-Jin Chu
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| |
Collapse
|
7
|
3-(5-Hydroxyphenyl)-5-Phenyl-2-Pyrazolines as Toll-Like Receptor 7 Agonists. J CHEM-NY 2023. [DOI: 10.1155/2023/2151669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is an attractive target for developing immune modulators to enhance innate immunity against ssRNA virus infections, including hepatitis C and COVID-19. Ten 3-(5-hydroxyphenyl)-5-phenyl-2-pyrazolines were tested using TLR7 reporter cells, overexpressing TLR7 and the NF-κB-inducible SEAP reporter gene to discover a novel TLR7 agonist enhancing innate immunity. Of these, 2-(3-(2-hydroxynaphthalen-1-yl)-5-(4-methoxyphenyl)-4,5-dihydro-1H-pyrazol-1-yl)thiazol-4(5H)-one (compound 6) showed the best TLR7 agonistic activity, and further experiments were carried out to study the immune-modulatory capability of compound 6. Treatment with compound 6 rapidly induced phosphorylation of IRAK4, IKKα/β, IκBα, and p65/RelA in THP1 monocytic cells. In addition, it increased the expression of NF-κB-regulated innate cytokines, such as TNFα and IL1β, in THP1 monocytic cells. These data suggest that compound 6 induces an innate immune response by agonizing TLR7 activity in THP1 human monocytic cells. Therefore, compound 6 can be used as an innate immune modulator to develop antiviral agents and vaccine adjuvants.
Collapse
|
8
|
Wu LS, Hu Y, Gane EJ, Slaets L, De Creus A, Ding Y, Niu J, Schwabe C, Goeyvaerts N, Xu Z, Huo D, Tuefferd M, Verbrugge I, Van Remoortere P, Schwertschlag U, Vandenbossche J. Population pharmacokinetic/pharmacodynamic models of JNJ-64794964, a toll-like receptor 7 agonist, in healthy adult participants. Antivir Ther 2023; 28:13596535231151626. [PMID: 36691849 DOI: 10.1177/13596535231151626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND JNJ-4964 is a TLR7 agonist, which, via a type I interferon (IFN)-dependent mechanism, may enhance host immunity suppressed by persistent exposure to hepatitis B antigens in chronic hepatitis B. METHODS PK and PD data were pooled from 2 studies involving 90 participants (n = 74 JNJ-4964, dose range 0.2-1.8 mg; n = 16 placebo) in a fasted state. Food effects on PK were studied in 24 participants (1.2 or 1.25 mg). A population PK model and PK/PD models were developed to characterize the effect of JNJ-4964 plasma levels on the time course of IFN-α, IFN-γ-inducible protein 10 (IP-10 or CXCL10), IFN-stimulated gene 15 (ISG15), neopterin and lymphocytes following single and weekly dosing in healthy adults. Covariate effects, circadian rhythms and negative feedback were incorporated in the models. RESULTS A 3-compartment linear PK model with transit absorption adequately described JNJ-4964 PK. Bioavailability was 44.2% in fed state relative to fasted conditions. Indirect response models with maximum effect (Emax) stimulation on production rate constant (kin) described IFN-α, IP-10, ISG15 and neopterin, while a precursor-dependent indirect response model with inhibitory effect described the transient lymphocyte reduction. Emax, EC50 and γ (steepness) estimates varied according to PD markers, with EC50 displaying substantial between-subject variability. Female and Asian race exhibited lower EC50, suggesting higher responsiveness. CONCLUSIONS PK/PD models well characterized the time course of immune system markers in healthy adults. Our results supported sex and race as covariates on JNJ-4964 responsiveness, as well as circadian rhythms and negative feedback as homeostatic mechanisms that are relevant in TLR7-induced type I IFN responses.
Collapse
Affiliation(s)
| | - Yue Hu
- 117971The First Hospital of Jilin University, Department of Hepatology, Changchun, Jilin, China
| | - Edward J Gane
- New Zealand Liver Transplant Unit, Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | - Leen Slaets
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| | - An De Creus
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| | - Yanhua Ding
- 117971The First Hospital of Jilin University, Department of Hepatology, Changchun, Jilin, China
| | - Junqi Niu
- 117971The First Hospital of Jilin University, Department of Hepatology, Changchun, Jilin, China
| | - Christian Schwabe
- Auckland Clinical Studies, New Zealand Clinical Research, Auckland, New Zealand
| | - Nele Goeyvaerts
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| | - Zhongnan Xu
- Chia Tai-Tianqing Pharmaceutical Group Co., Ltd, Nanjing, Jiangsu, China
| | - Dandan Huo
- Chia Tai-Tianqing Pharmaceutical Group Co., Ltd, Nanjing, Jiangsu, China
| | - Marianne Tuefferd
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| | - Inge Verbrugge
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | - Joris Vandenbossche
- Janssen Research & Development, 50148Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
9
|
Janku F, Han SW, Doi T, Amatu A, Ajani JA, Kuboki Y, Cortez A, Cellitti SE, Mahling PC, Subramanian K, Schoenfeld HA, Choi SM, Iaconis LA, Lee LH, Pelletier MR, Dranoff G, Askoxylakis V, Siena S. Preclinical Characterization and Phase I Study of an Anti-HER2-TLR7 Immune-Stimulator Antibody Conjugate in Patients with HER2+ Malignancies. Cancer Immunol Res 2022; 10:1441-1461. [PMID: 36129967 DOI: 10.1158/2326-6066.cir-21-0722] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 04/29/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023]
Abstract
Immune-stimulator antibody conjugates (ISAC) combining tumor-targeting monoclonal antibodies with immunostimulatory agents allow targeted delivery of immune activators into tumors. NJH395 is a novel, first-in-class ISAC comprising a Toll-like receptor 7 (TLR7) agonist conjugated to an anti-HER2 antibody via a noncleavable linker payload. Preclinical characterization showed ISAC-mediated activation of myeloid cells in the presence of antigen-expressing cancer cells, with antigen targeting and TLR7 agonism contributing to antitumor activity. Safety, efficacy, immunogenicity, pharmacokinetics, and pharmacodynamics were investigated in a phase I, multicenter, open-label study in patients with HER2+ non-breast advanced malignancies (NCT03696771). Data from 18 patients enrolled in single ascending dose escalation demonstrated delivery of the TLR7-agonist payload in HER2+ tumor cells and induction of type I IFN responses, which correlated with immune modulation in the tumor microenvironment. Cytokine release syndrome was a common, but manageable, drug-related adverse event. Antidrug antibodies and neuroinflammation at high doses represented significant clinical challenges. Data provide proof-of-mechanism and critical insights for novel immunotherapies.
Collapse
Affiliation(s)
- Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University Hospital and Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | | | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Alex Cortez
- Novartis Institutes for BioMedical Research, San Diego, California
| | - Susan E Cellitti
- Novartis Institutes for BioMedical Research, San Diego, California
| | | | | | | | - Sarah M Choi
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Lori A Iaconis
- Novartis Institutes for BioMedical Research, San Diego, California
| | - Lang Ho Lee
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Marc R Pelletier
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Glenn Dranoff
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | | | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
10
|
Liu ZM, Yang MH, Yu K, Lian ZX, Deng SL. Toll-like receptor (TLRs) agonists and antagonists for COVID-19 treatments. Front Pharmacol 2022; 13:989664. [PMID: 36188605 PMCID: PMC9518217 DOI: 10.3389/fphar.2022.989664] [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: 07/08/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) rapidly infects humans and animals which make coronavirus disease 2019 (COVID-19) a grievous epidemic worldwide which broke out in 2020. According to data analysis of the other coronavirus family, for instance severe acute respiratory syndrome SARS coronavirus (SARS-CoV), can provide experience for the mutation of SARS-CoV-2 and the prevention and treatment of COVID-19. Toll-like receptors (TLRs) as a pattern recognition receptor (PRRs), have an indispensable function in identifying the invader even activate the innate immune system. It is possible for organism to activate different TLR pathways which leads to secretion of proinflammatory cytokines such as Interleukin 1 (IL-1), Interleukin 6 (IL-6), Tumor necrosis factor α (TNFα) and type Ⅰ interferon. As a component of non-specific immunity, TLRs pathway may participate in the SARS-CoV-2 pathogenic processes, due to previous works have proved that TLRs are involved in the invasion and infection of SARS-CoV and MERS to varying degrees. Different TLR, such as TLR2, TLR4, TLR7, TLR8 and TLR9 probably have a double-sided in COVID-19 infection. Therefore, it is of great significance for a correctly acknowledging how TLR take part in the SARS-CoV-2 pathogenic processes, which will be the development of treatment and prevention strategies.
Collapse
Affiliation(s)
- Zhi-Mei Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ming-Hui Yang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, China
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| |
Collapse
|
11
|
Girkin JLN, Maltby S, Bartlett NW. Toll-like receptor-agonist-based therapies for respiratory viral diseases: thinking outside the cell. Eur Respir Rev 2022; 31:210274. [PMID: 35508333 PMCID: PMC9488969 DOI: 10.1183/16000617.0274-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
Respiratory virus infections initiate in the upper respiratory tract (URT). Innate immunity is critical for initial control of infection at this site, particularly in the absence of mucosal virus-neutralising antibodies. If the innate immune response is inadequate, infection can spread to the lower respiratory tract (LRT) causing community-acquired pneumonia (as exemplified by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/coronavirus disease 2019). Vaccines for respiratory viruses (influenza and SARS-CoV-2) leverage systemic adaptive immunity to protect from severe lung disease. However, the URT remains vulnerable to infection, enabling viral transmission and posing an ongoing risk of severe disease in populations that lack effective adaptive immunity.Innate immunity is triggered by host cell recognition of viral pathogen-associated molecular patterns via molecular sensors such as Toll-like receptors (TLRs). Here we review the role of TLRs in respiratory viral infections and the potential of TLR-targeted treatments to enhance airway antiviral immunity to limit progression to severe LRT disease and reduce person-to-person viral transmission. By considering cellular localisation and antiviral mechanisms of action and treatment route/timing, we propose that cell surface TLR agonist therapies are a viable strategy for preventing respiratory viral diseases by providing immediate, durable pan-viral protection within the URT.
Collapse
Affiliation(s)
- Jason L N Girkin
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
12
|
Chiang CY, Lane DJ, Zou Y, Hoffman T, Pan J, Hampton J, Maginnis J, Nayak BP, D'Oro U, Valiante N, Miller AT, Cooke M, Wu T, Bavari S, Panchal RG. A Novel Toll-Like Receptor 2 Agonist Protects Mice in a Prophylactic Treatment Model Against Challenge With Bacillus anthracis. Front Microbiol 2022; 13:803041. [PMID: 35369443 PMCID: PMC8965344 DOI: 10.3389/fmicb.2022.803041] [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: 10/27/2021] [Accepted: 02/02/2022] [Indexed: 11/23/2022] Open
Abstract
Current therapies for anthrax include the use of antibiotics (i.e., doxycycline, and ciprofloxacin), an anthrax vaccine (BioThrax) and Bacillus anthracis-specific, monoclonal antibody (mAb) (i.e., Raxibacumab and obiltoxaximab). In this study, we investigated the activity of immunomodulators, which potentiate inflammatory responses through innate immune receptors. The rationale for the use of innate immune receptor agonists as adjunctive immunomodulators for infectious diseases is based on the concept that augmentation of host defense should promote the antimicrobial mechanism of the host. Our aim was to explore the anti-B. anthracis effector function of Toll-like receptor (TLR) agonists using a mouse model. Amongst the six TLR ligands tested, Pam3CSK4 (TLR1/2 ligand) was the best at protecting mice from lethal challenge of B. anthracis. We then evaluated the activity of a novel TLR2 ligand, DA-98-WW07. DA-98-WW07 demonstrated enhanced protection in B. anthracis infected mice. The surviving mice that received DA-98-WW07 when re-challenged with B. anthracis 20 days post the first infection showed increased survival rate. Moreover, ciprofloxacin, when treated in adjunct with a suboptimal concentration of DA-98-WW07 demonstrated augmented activity in protecting mice from B. anthracis infection. Taken together, we report the prophylactic treatment potential of DA-98-WW07 for anthrax and the utility of immunomodulators in combination with an antibiotic to treat infections caused by the B. anthracis bacterium.
Collapse
Affiliation(s)
- Chih-Yuan Chiang
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Douglas J Lane
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Yefen Zou
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Tim Hoffman
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Jianfeng Pan
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Janice Hampton
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Jillian Maginnis
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Bishnu P Nayak
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Ugo D'Oro
- Novartis Vaccines and Diagnostics, Siena, Italy
| | | | - Andrew T Miller
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Michael Cooke
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Tom Wu
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Sina Bavari
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Rekha G Panchal
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| |
Collapse
|
13
|
Li X, Sun X, Guo X, Li X, Peng S, Mu X. Chemical reagents modulate nucleic acid-activated toll-like receptors. Biomed Pharmacother 2022; 147:112622. [PMID: 35008000 DOI: 10.1016/j.biopha.2022.112622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 02/08/2023] Open
Abstract
Nucleic acid-mediated interferon signaling plays a pivotal role in defense against microorganisms, especially during viral infection. Receptors sensing exogenous nucleic acid molecules are localized in the cytosol and endosomes. Cytosolic sensors, including cGAS, RIG-I, and MDA5, and endosome-anchored receptors are toll-like receptors (TLR3, TLR7, TLR8, and TLR9). These TLRs share the same domain architecture and have similar structures, facing the interior of endosomes so their binding to nucleic acids of invading pathogens via endocytosis is possible. The correct function of these receptors is crucial for cell homeostasis and effective response against pathogen invasion. A variety of endogenous mechanisms modulates their activities. Nevertheless, naturally occurring mutations lead to aberrant TLR-mediated interferon (IFN) signaling. Furthermore, certain pathogens require a more robust defense against control. Thus, manipulating these TLR activities has a profound impact. High-throughput virtual screening followed by experimental validation led to the discovery of numerous chemicals that can change these TLR-mediated IFN signaling activities. Many of them are unique in selectivity, while others regulate more than one TLR due to commonalities in these receptors. We summarized these nucleic acid-sensing TLR-mediated IFN signaling pathways and the corresponding chemicals activating or deactivating their signaling.
Collapse
Affiliation(s)
- Xiao Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xinyuan Sun
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xuemin Guo
- Meizhou People's Hospital, Meizhou 514031, China; Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translation Research of Hakka Population, Meizhou 514031, China
| | - Xueren Li
- Department of Respiratory Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, China
| | - Shouchun Peng
- Department of Respiratory Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, China.
| | - Xin Mu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; Tianjin University and Health-Biotech United Group Joint Laboratory of Innovative Drug Development and Translational Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
14
|
El Sahly HM, Atmar RL, Sendra E, Wegel A, Keitel WA. Topical Imiquimod Does Not Provide an Adjuvant Effect When Administered With Inactivated Influenza A/H5N1 Vaccine in Healthy Young Adults. J Infect Dis 2021; 224:1712-1719. [PMID: 33852718 PMCID: PMC9633716 DOI: 10.1093/infdis/jiab206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/13/2021] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Safe, effective, and easy to deploy adjuvants are needed for influenza prepandemic preparedness. Based on recent reports, we hypothesized that preapplication of topical imiquimod followed by intradermal (ID) vaccination with monovalent inactivated influenza A/H5N1 vaccine (MIV A/H5N1) results in improved serologic responses. METHODS We randomized 50 healthy adults in a 1:1 ratio to receive topical imiquimod (group 1) or control cream (group 2) followed by ID injection of 9 µg of the hemagglutinin MIV A/H5N1 in 2 doses, 21 days apart. Subjects were followed for safety and serologic responses as measured by the hemagglutination inhibition (HAI) and microneutralization (MN) assays. RESULTS Solicited and unsolicited adverse events were comparable between groups 1 and 2, and were mostly mild to moderate in severity. At 21 days after dose 2, the geometric mean titers (GMTs) of HAI antibodies against the vaccine strain were 16.2 and 24.3 in groups 1 and 2, respectively. The MN antibody GMTs were 9.3 and 10.7 in groups 1 and 2, respectively. There were no significant differences in antibody levels between groups at study time points. CONCLUSIONS Topical imiquimod administration combined with ID MIV A/H5N1 was safe but did not result in improved serologic responses to the vaccine.Clinical Trials Registration. NCT03472976.
Collapse
Affiliation(s)
- Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Eli Sendra
- The Emmes Company, Rockville, Maryland, USA
| | | | - Wendy A Keitel
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
15
|
Gane E, Pastagia M, Schwertschlag U, De Creus A, Schwabe C, Vandenbossche J, Slaets L, Fevery B, Smyej I, Wu LS, Li R, Siddiqui S, Oey A, Musto C, Van Remoortere P. Safety, tolerability, pharmacokinetics, and pharmacodynamics of oral JNJ-64794964, a TLR-7 agonist, in healthy adults. Antivir Ther 2021; 26:58-68. [DOI: 10.1177/13596535211056581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background This Phase I, two-part, first-in-human study assessed safety/tolerability and pharmacokinetics/pharmacodynamics of single-ascending doses (SAD) and multiple doses (MD) of the oral toll-like receptor-7 agonist, JNJ-64794964 (JNJ-4964) in healthy adults. Methods In the SAD phase, participants received JNJ-4964 0.2 ( N = 6), 0.6 ( N = 6), 1.25 ( N = 8) or 1.8 mg ( N = 6) or placebo ( N = 2/dose cohort) in a fasted state. Food effect was evaluated for the 1.25 mg cohort following ≥6 weeks washout. In the MD phase, participants received JNJ-4964 1.25 mg ( N = 6) or placebo ( N = 2) weekly (fasted) for 4 weeks. Participants were followed-up for 4 weeks. Results No serious adverse events (AEs) occurred. 10/34 (SAD) and 5/8 (MD) participants reported mild-to-moderate (≤Grade 2), transient, reversible AEs possibly related to JNJ-4964. Five (SAD) participants had fever/flu-like AEs, coinciding with interferon-α serum levels ≥100 pg/mL and lymphopenia (<1 × 109/L), between 24–48 h after dosing and resolving approximately 96 h after dosing. One participant (MD) had an asymptomatic Grade 1 AE of retinal exudates (cotton wool spots) during follow-up, resolving 6 weeks after observation. JNJ-4964 exhibited dose-proportional pharmacokinetics, with rapid absorption (tmax 0.5–0.75 h) and distribution, and a long terminal half-life (150–591 h). Overall, no significant differences in JNJ-4964 pharmacokinetic parameters were observed in the fed versus fasted state. JNJ-4964 dose-dependently and transiently induced cytokines with potential anti-HBV activity, including interferon-α, IP-10, IL-1 RA, and/or MCP-1, and interferon-stimulated genes (ISG15, MX1, and OAS1) in serum. Conclusions In healthy adults, JNJ-4964 was generally well-tolerated, exhibited dose-proportional pharmacokinetics and induced cytokines/ISGs, with possible anti-HBV activity.
Collapse
Affiliation(s)
- Edward Gane
- New Zealand Liver Transplant Unit, University of Auckland, Auckland, New Zealand
| | - Mina Pastagia
- Janssen BioPharma Inc., South San Francisco, CA, USA
| | | | - An De Creus
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | - Leen Slaets
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Bart Fevery
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ilham Smyej
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - Rui Li
- Janssen Research & Development LLC, Titusville, NJ, USA
| | | | - Abbie Oey
- Janssen BioPharma Inc., South San Francisco, CA, USA
| | - Clark Musto
- Janssen BioPharma Inc., South San Francisco, CA, USA
| | | |
Collapse
|
16
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
17
|
Grippo JF, Folitar I, Passe S, Jiang Q, Rodriguez I, Fettner SH, Calleja E. Safety, tolerability, pharmacokinetics, and pharmacodynamics of a TLR7 agonist prodrug RO6870868 in healthy volunteers. Clin Transl Sci 2021; 14:1524-1534. [PMID: 33742764 PMCID: PMC8301559 DOI: 10.1111/cts.13016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
RO6870868 is an oral prodrug of the toll‐like receptor 7 (TLR7) specific agonist, RO6871765. TLR7 agonists augment host immune activity and are in development to treat hepatitis B infection. We evaluated the safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of RO6870868 in a first‐in‐human, phase I, randomized, single ascending oral dose study in 60 healthy volunteers at 6 dose levels (200–2000 mg). Single oral doses were generally well‐tolerated with a predictable safety profile associated with dose‐dependent increases in systemic interferon. No serious adverse events (AEs) were reported and no subject withdrew from the study due to an AE. No clinically significant changes were observed in vital signs, electrocardiograms, or laboratory parameters. Following oral RO6870868 doses, plasma RO6871765 concentrations increased rapidly, exhibiting mean terminal half‐life ranging 2–6 h across all cohorts, with area under the plasma concentration versus time curve extrapolated to infinity (AUC0‐∞) increasing proportionally with dose. A pattern of dose and time‐dependent PD activity was demonstrated consistent with engagement of the TLR7 system. Single RO6870868 doses activated components of the TLR innate immune system in a dose‐dependent manner with adequate safety and tolerability. Single‐dose data in healthy volunteers are useful to evaluate safety, PK, and PD activity of TLR7 agonists and help to guide dose and regimen selection for further trials in patients with chronic hepatitis B.
Collapse
Affiliation(s)
| | | | - Sharon Passe
- Roche Innovation Center, New York, New York, USA
| | - Qiudi Jiang
- Roche Innovation Center Shanghai, Shanghai, China
| | | | | | | |
Collapse
|
18
|
Man RJ, Jeelani N, Zhou C, Yang YS. Recent Progress in the Development of Quinoline Derivatives for the Exploitation of Anti-Cancer Agents. Anticancer Agents Med Chem 2021; 21:825-838. [PMID: 32416703 DOI: 10.2174/1871520620666200516150345] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/23/2020] [Accepted: 02/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Along with the progress in medicine and therapies, the exploitation of anti-cancer agents focused more on the vital signaling pathways and key biological macromolecules. With rational design and advanced synthesis, quinoline derivatives have been utilized frequently in medicinal chemistry, especially in developing anti-cancer drugs or candidates. METHODS Using DOI searching, articles published before 2020 all over the world have been reviewed as comprehensively as possible. RESULTS In this review, we selected the representative quinoline derivate drugs in market or clinical trials, classified them into five major categories with detailed targets according to their main mechanisms, discussed the relationship within the same mechanism, and generated a summative discussion with prospective expectations. For each mechanism, the introduction of the target was presented, with the typical examples of quinoline derivate drugs. CONCLUSION This review has highlighted the quinoline drugs or candidates, suited them into corresponding targets in their pathways, summarized and discussed. We hope that this review may help the researchers who are interested in discovering quinoline derivate anti-cancer agents obtain considerable understanding of this specific topic. Through the flourishing period and the vigorous strategies in clinical trials, quinoline drugs would be potential but facing new challenges in the future.
Collapse
Affiliation(s)
- Ruo-Jun Man
- College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China
| | - Nasreen Jeelani
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Chongchen Zhou
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Yu-Shun Yang
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
19
|
Frega G, Wu Q, Le Naour J, Vacchelli E, Galluzzi L, Kroemer G, Kepp O. Trial Watch: experimental TLR7/TLR8 agonists for oncological indications. Oncoimmunology 2020; 9:1796002. [PMID: 32934889 PMCID: PMC7466852 DOI: 10.1080/2162402x.2020.1796002] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Resiquimod (R848) and motolimod (VTX-2337) are second-generation experimental derivatives of imiquimod, an imidazoquinoline with immunostimulatory properties originally approved by the US Food and Drug Administration for the topical treatment of actinic keratosis and genital warts more than 20 years ago. Both resiquimod and motolimod operate as agonists of Toll-like receptor 7 (TLR7) and/or TLR8, in thus far delivering adjuvant-like signals to antigen-presenting cells (APCs). In line with such an activity, these compounds are currently investigated as immunostimulatory agents for the treatment of various malignancies, especially in combination with peptide-based, dendritic cell-based, cancer cell lysate-based, or DNA-based vaccines. Here, we summarize preclinical and clinical evidence recently collected to support the development of resiquimod and motolimod and other TLR7/TLR8 agonists as anticancer agents.
Collapse
Affiliation(s)
- Giorgio Frega
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Qi Wu
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Julie Le Naour
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Erika Vacchelli
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Oliver Kepp
- Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| |
Collapse
|
20
|
Flórez-Álvarez L, Ruiz-Perez L, Taborda N, Hernandez JC. Toll-like receptors as a therapeutic target in cancer, infections and inflammatory diseases. Immunotherapy 2020; 12:311-322. [PMID: 32237938 DOI: 10.2217/imt-2019-0096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Toll-like receptors (TLRs) are widely expressed pattern recognition receptors that bind to conserved molecular patterns expressed by pathogens and damaged cells. After recognition, activated TLRs induce the expression of various proinflammatory and antiviral molecules. Thus, TLRs are potential targets for treatment strategies aimed at boosting the adaptive immune response to vaccines, controlling infections, enhancing immune responses during tumor treatment and attenuating immune responses in inflammatory disorders. This Special Report examines the potential of TLRs as targets for the treatment of cancer, infections and inflammatory diseases. Here, we make a particular emphasis on molecules capable of modulating TLRs and their therapeutic applications.
Collapse
Affiliation(s)
- Lizdany Flórez-Álvarez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia.,Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA; Calle 70 No. 52-21, Medellín, Colombia
| | - Lanie Ruiz-Perez
- School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth WA 6845, Australia
| | - Natalia Taborda
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA; Calle 70 No. 52-21, Medellín, Colombia.,Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Juan C Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| |
Collapse
|
21
|
McGowan DC. Latest Advances in Small Molecule TLR 7/8 Agonist Drug Research. Curr Top Med Chem 2019; 19:2228-2238. [DOI: 10.2174/1568026619666191009165418] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Toll-like receptors (TLRs) 7 and 8 play an important role in the activation of innate immune
cells in mammals. These evolutionarily conserved receptors serve as important sentinels in response to
infection. Activation of TLRs 7 and 8 triggers induction of a Th1 type innate immune response. The
emergence of new structural and small molecule information generated in the last decade has contributed
enormously to our understanding of this highly sophisticated process of innate immunity signaling.
This review will focus on recent developments in the small molecule activation of TLR 7 and 8.
Collapse
Affiliation(s)
- David C. McGowan
- Janssen Pharmaceutica, N.V., Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
22
|
Udgata A, Dolasia K, Ghosh S, Mukhopadhyay S. Dribbling through the host defence: targeting the TLRs by pathogens. Crit Rev Microbiol 2019; 45:354-368. [PMID: 31241370 DOI: 10.1080/1040841x.2019.1608904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Atul Udgata
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Komal Dolasia
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| |
Collapse
|
23
|
Urbanowicz A, Zagożdżon R, Ciszek M. Modulation of the Immune System in Chronic Hepatitis C and During Antiviral Interferon-Free Therapy. Arch Immunol Ther Exp (Warsz) 2018; 67:79-88. [PMID: 30443787 PMCID: PMC6420452 DOI: 10.1007/s00005-018-0532-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
The treatment of patients with chronic hepatitis C virus (HCV) infection has changed tremendously over the past 2 years, with an increasing variety of all-oral direct-acting antiviral (DAA) treatment regimens available for different HCV genotypes and distinct clinical settings. These treatments have significantly improved safety in patients with advanced liver disease compared with interferon (IFN)-based regimens. HCV modifies the human immune system to escape immunosurveillance via several mechanisms. One of the basic mechanisms of HCV is the ability to “switch” the immune response by reducing the activity of cells responsible for the elimination of virus-infected cells. IFN-free DAA treatment regimens provide a unique opportunity to assess the effect of HCV elimination on the immune system. Abrupt changes in the immune system can in some cases be responsible for two alarming processes: viral reactivation in patients with chronic hepatitis B and recurrence of hepatocellular carcinoma in patients with previous successful cancer treatment.
Collapse
Affiliation(s)
- Arkadiusz Urbanowicz
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Radosław Zagożdżon
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, Warsaw, Poland.,Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michał Ciszek
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
24
|
Bazin HG, Bess LS, Livesay MT. Synthesis and Applications of Imidazoquinolines: A Review. ORG PREP PROCED INT 2018. [DOI: 10.1080/00304948.2018.1433427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Hélène G. Bazin
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| | - Laura S. Bess
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| | - Mark T. Livesay
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| |
Collapse
|
25
|
Burt AJ, Hantho JD, Nielsen AE, Mancini RJ. An Enzyme-Directed Imidazoquinoline Activated by Drug Resistance. Biochemistry 2018; 57:2184-2188. [DOI: 10.1021/acs.biochem.8b00095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Anthony J. Burt
- Department of Chemistry, Washington State University, 1470 East College Avenue, Pullman, Washington 99164, United States
| | - Joseph D. Hantho
- Department of Chemistry, Washington State University, 1470 East College Avenue, Pullman, Washington 99164, United States
| | - Amy E. Nielsen
- Department of Chemistry, Washington State University, 1470 East College Avenue, Pullman, Washington 99164, United States
| | - Rock J. Mancini
- Department of Chemistry, Washington State University, 1470 East College Avenue, Pullman, Washington 99164, United States
| |
Collapse
|
26
|
Camateros P, Kanagaratham C, Najdekr L, Holub D, Vrbkova J, Coté L, Fournier J, Gourdon J, Creery D, Olivenstein R, Kopriva F, Adam T, Friedecký D, Džubák P, Hajdúch M, Radzioch D. Toll-Like Receptor 7/8 Ligand, S28463, Suppresses Ascaris suum-induced Allergic Asthma in Nonhuman Primates. Am J Respir Cell Mol Biol 2018; 58:55-65. [PMID: 28850259 DOI: 10.1165/rcmb.2017-0184oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
S28463 (S28), a ligand for Toll-like receptor 7/8, has been shown to have antiinflammatory properties in rodent models of allergic asthma. The principle goal of this study was to assess whether these antiinflammatory effects can also be observed in a nonhuman primate (NHP) model of allergic asthma. NHPs were sensitized then challenged with natural allergen, Ascaris suum extract. The animals were treated with S28 orally before each allergen challenge. The protective effect of S28 in NHPs was assessed by measuring various asthma-related phenotypes. We also characterized the metabolomic and proteomic signatures of the lung environment and plasma to identify markers associated with the disease and treatment. Our data demonstrate that clinically relevant parameters, such as wheal and flare response, blood IgE levels, recruitment of white blood cells to the bronchoalveolar space, and lung responsiveness, are decreased in the S28-treated allergic NHPs compared with nontreated allergic NHPs. Furthermore, we also identified markers that can distinguish allergic from nonallergic or allergic and drug-treated NHPs, such as metabolites, phosphocreatine and glutathione, in the plasma and BAL fluid, respectively; and inflammatory cytokines, IL-5 and IL-13, in the bronchoalveolar lavage fluid. Our preclinical study demonstrates that S28 has potential as a treatment for allergic asthma in primate species closely related to humans. Combined with our previous findings, we demonstrate that S28 is effective in different models of asthma and in different species, and has the antiinflammatory properties clinically relevant for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Pierre Camateros
- 1 Faculty of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Cynthia Kanagaratham
- 1 Faculty of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.,2 The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Lukáš Najdekr
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Dušan Holub
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Jana Vrbkova
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Lucie Coté
- 2 The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.,4 Institut National de la Recherche Scientifique at the Armand Frappier, Laval, Quebec, Canada
| | - Jocelyn Fournier
- 5 Sir Frederick G. Banting Research Centre, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Jim Gourdon
- 6 Comparative Medicine, McGill University, Montreal, Quebec, Canada
| | - David Creery
- 7 Faculty of Medicine, University of Ottawa, Pediatric Critical Care, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Ron Olivenstein
- 8 Respiratory Division, McGill University, Montreal, Quebec, Canada
| | - Frantisek Kopriva
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic.,9 Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic; and
| | - Tomáš Adam
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - David Friedecký
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Petr Džubák
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Marian Hajdúch
- 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Danuta Radzioch
- 1 Faculty of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.,2 The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.,10 Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Morgan P, Brown DG, Lennard S, Anderton MJ, Barrett JC, Eriksson U, Fidock M, Hamrén B, Johnson A, March RE, Matcham J, Mettetal J, Nicholls DJ, Platz S, Rees S, Snowden MA, Pangalos MN. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat Rev Drug Discov 2018; 17:167-181. [DOI: 10.1038/nrd.2017.244] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Jackson S, Candia AF, Delaney S, Floettmann S, Wong C, Campbell JD, Kell S, Lum J, Hessel EM, Traquina P, McHale M, Robinson I, Bell J, Fuhr R, Keeling D, Coffman RL. First-in-Human Study With the Inhaled TLR9 Oligonucleotide Agonist AZD1419 Results in Interferon Responses in the Lung, and Is Safe and Well-Tolerated. Clin Pharmacol Ther 2017; 104:335-345. [DOI: 10.1002/cpt.938] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Sam Jackson
- Dynavax Technologies; Berkeley California USA
| | | | - Stephen Delaney
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
| | | | | | | | - Sariah Kell
- Dynavax Technologies; Berkeley California USA
| | - Jeremy Lum
- Dynavax Technologies; Berkeley California USA
| | | | | | - Mark McHale
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
| | - Ian Robinson
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
| | - John Bell
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
| | | | - David Keeling
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
| | | |
Collapse
|
29
|
Bell J, Dymond M, Biffen M, Delaney S, Keeling D, Zhang H, Robinson I. Temporal cytokine and lymphoid responses to an inhaled TLR7 antedrug agonist in the cynomolgus monkey demonstrates potential safety and tolerability of this approach. Toxicol Appl Pharmacol 2017; 338:9-19. [PMID: 29122671 DOI: 10.1016/j.taap.2017.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/30/2017] [Accepted: 11/03/2017] [Indexed: 01/04/2023]
Abstract
AZD8848 is a TLR7 agonist antedrug developed for administration by inhalation dosing for the treatment of allergic diseases, such as asthma. Allergic asthma is associated with increased levels of Th2 cytokines which are suppressed for extended periods by TLR7 agonists in a number of preclinical models of allergic airway inflammation. However, TLRs form a central part of innate immunity and their activation often results in proinflammatory responses. Whilst AZD8848's antedrug mechanism is designed to restrict its pharmacological action beyond the lung, the effect of chronic, supramaximal dosing to the target tissue has yet to be defined. To support clinical development of this potentially disease modifying approach the nonclinical safety and pharmacodynamics of AZD8848 were evaluated in cynomolgus monkeys in studies examining single or multiple weekly inhaled doses. Here we show that following a single dose nearly all responses returned to baseline within a week. During multiple dosing serum biomarkers were quantified over the dosing period and indicated a limited systemic response. The dose at which maximal interferon responses were seen was dependent on dose. Thorough histopathological examination revealed a dose related increase of size and cells of lymphoid tissues in the lung and nose. Local lymphoid responses were recovered after the treatment free period. These studies are the first to evaluate safety of an inhaled TLR7 agonist and demonstrate AZD8848 is safe with a no observed adverse effect level at 26μg/kg allowing progression to man with weekly inhalation dosing.
Collapse
Affiliation(s)
- John Bell
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Mike Dymond
- Plus-Project Ltd, Alderley Park, Cheshire, United Kingdom
| | - Mark Biffen
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Stephen Delaney
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - David Keeling
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Hui Zhang
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ian Robinson
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom.
| |
Collapse
|
30
|
Sato-Kaneko F, Yao S, Ahmadi A, Zhang SS, Hosoya T, Kaneda MM, Varner JA, Pu M, Messer KS, Guiducci C, Coffman RL, Kitaura K, Matsutani T, Suzuki R, Carson DA, Hayashi T, Cohen EE. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight 2017; 2:93397. [PMID: 28931759 PMCID: PMC5621908 DOI: 10.1172/jci.insight.93397] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/10/2017] [Indexed: 12/28/2022] Open
Abstract
Checkpoint inhibitors have demonstrated efficacy in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, the majority of patients do not benefit from these agents. To improve the efficacy of checkpoint inhibitors, intratumoral (i.t.) injection with innate immune activators, TLR7 and TLR9 agonists, were tested along with programmed death-1 receptor (PD-1) blockade. The combination therapy suppressed tumor growth at the primary injected and distant sites in human papillomavirus-negative (HPV-negative) SCC7 and MOC1, and HPV-positive MEER syngeneic mouse models. Abscopal effects and suppression of secondary challenged tumor suggest that local treatment with TLR agonists in combination with anti-PD-1 provided systemic adaptive immunity. I.t. treatment with a TLR7 agonist increased the ratio of M1 to M2 tumor-associated macrophages (TAMs) and promoted the infiltration of tumor-specific IFNγ-producing CD8+ T cells. Anti-PD-1 treatment increased T cell receptor (TCR) clonality of CD8+ T cells in tumors and spleens of treated mice. Collectively, these experiments demonstrate that combination therapy with i.t. delivery of TLR agonists and PD-1 blockade activates TAMs and induces tumor-specific adaptive immune responses, leading to suppression of primary tumor growth and prevention of metastasis in HNSCC models.
Collapse
Affiliation(s)
| | - Shiyin Yao
- Moores Cancer Center, UCSD, La Jolla, California, USA
| | - Alast Ahmadi
- Moores Cancer Center, UCSD, La Jolla, California, USA
| | | | | | | | | | - Minya Pu
- Moores Cancer Center, UCSD, La Jolla, California, USA
| | | | | | | | - Kazutaka Kitaura
- Repertoire Genesis Inc., Saito Bioincubator, Saito-Asagai, Ibaraki-shi, Osaka, Japan
| | - Takaji Matsutani
- Repertoire Genesis Inc., Saito Bioincubator, Saito-Asagai, Ibaraki-shi, Osaka, Japan
| | - Ryuji Suzuki
- Repertoire Genesis Inc., Saito Bioincubator, Saito-Asagai, Ibaraki-shi, Osaka, Japan
| | | | | | | |
Collapse
|
31
|
Ellis AK, Tsitoura DC, Quint D, Powley W, Lee LA. Safety and pharmacodynamics of intranasal GSK2245035, a TLR7 agonist for allergic rhinitis: A randomized trial. Clin Exp Allergy 2017; 47:1193-1203. [PMID: 28681506 DOI: 10.1111/cea.12974] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/04/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Toll-like receptor 7 (TLR7) stimulation in the airways may reduce responses to aeroallergens by induction of type 1 interferons (IFNs). GSK2245035 is a novel selective TLR7 agonist in pharmaceutical development. OBJECTIVE Assessment of safety, pharmacodynamics and nasal allergic reactivity following repeated weekly intranasal (i.n.) GSK2245035. METHODS This randomized, double-blind, placebo-controlled study (TL7116958) was conducted over two pollen seasons (2013-2014) and follow-up study (204509) conducted 1 year later. Participants with allergic rhinitis (n=42) were randomized to receive eight weekly doses of i.n. GSK2245035 (20 ng [2014 Cohort; n=14] or 80 ng [2013 Cohort; n=14]) or placebo (n=14). Adverse events (AEs) including cytokine release syndrome AEs (CytoRS-AEs) and nasal symptoms were assessed. Nasal and serum IFN-inducible protein 10 (IP-10) were measured after doses 1 and 8, then 1 (follow-up visit [FUV] 1) and 3 (FUV2) weeks after final dose. Nasal allergen challenges (NACs) and allergic biomarker assessment (nasal, serum) were conducted at baseline, FUV1, FUV2 and at a FUV 1 year after final dose (FUV3; 2014 Cohort only). A Bayesian framework enabled probability statements for mean effect sizes. RESULTS GSK2245035 induced CytoRS-AEs (most commonly headache, median duration <1 day) in 93% of participants at 80 ng, while AE incidence at 20 ng was similar to placebo. There was no evidence of nasal inflammation. Dose-related increases in nasal and serum IP-10 were observed 24 hours after doses 1 and 8 (>95% certainty). Both doses showed a trend in reducing total nasal symptom score 15 minutes post-NAC at FUV1 and FUV2, but there was no reduction evident at FUV3. Nasal levels of selected allergic biomarkers demonstrated trends for reductions at FUV1, FUV2 and FUV3. CONCLUSIONS AND CLINICAL RELEVANCE Weekly i.n. GSK2245035 20 ng was well tolerated and reduced allergic reactivity to nasal challenge for 3 weeks post-treatment.
Collapse
MESH Headings
- Adenine/administration & dosage
- Adenine/adverse effects
- Adenine/analogs & derivatives
- Adenine/pharmacokinetics
- Adenine/therapeutic use
- Administration, Intranasal
- Adult
- Aged
- Allergens/immunology
- Biomarkers
- Drug Monitoring
- Female
- Follow-Up Studies
- Humans
- Immunization
- Male
- Middle Aged
- Piperidines/administration & dosage
- Piperidines/adverse effects
- Piperidines/pharmacokinetics
- Piperidines/therapeutic use
- Rhinitis, Allergic/diagnosis
- Rhinitis, Allergic/drug therapy
- Rhinitis, Allergic/immunology
- Rhinitis, Allergic/metabolism
- Rhinitis, Allergic, Seasonal/diagnosis
- Rhinitis, Allergic, Seasonal/drug therapy
- Rhinitis, Allergic, Seasonal/immunology
- Rhinitis, Allergic, Seasonal/metabolism
- Seasons
- Toll-Like Receptor 7/antagonists & inhibitors
- Toll-Like Receptor 7/metabolism
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- A K Ellis
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - D Quint
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - W Powley
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - L A Lee
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
32
|
Athari SS, Athari SM, Beyzay F, Movassaghi M, Mortaz E, Taghavi M. Critical role of Toll-like receptors in pathophysiology of allergic asthma. Eur J Pharmacol 2016; 808:21-27. [PMID: 27894811 DOI: 10.1016/j.ejphar.2016.11.047] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 11/21/2016] [Accepted: 11/25/2016] [Indexed: 12/11/2022]
Abstract
Allergic asthma is an airway disease, characterized by reversible bronchoconstriction, chronic inflammation of the airway, and thickness of smooth muscle in the respiratory tract. Asthma is orchestrated by an excessive Th2-adaptive immune response, in which innate immunity plays a key role. Recently TLRs have received more and more attention as they are central to orchestrate the innate immune responses. TLRs are localized as integral membrane or intracellular glycoproteins with those on the cell surface sensing microbial antigens and the ones, localized in intracellular vesicles, sensing microbial nucleic acid species. Having recognized microbial antigens, TLRs conduct the immune response towards a pro- or anti-allergy response. As a double-edged sword, they could initiate either harmful or helpful responses by the immune system in case of allergic asthma. In the current review, we will describe the role of TLRs and their signaling pathways in allergic asthma.
Collapse
Affiliation(s)
- Seyyed Shamsadin Athari
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Health policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Fateme Beyzay
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Movassaghi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehdi Taghavi
- Mycology Research Center, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
33
|
Hantho JD, Strayer TA, Nielsen AE, Mancini RJ. An Enzyme-Directed Imidazoquinoline for Cancer Immunotherapy. ChemMedChem 2016; 11:2496-2500. [PMID: 27726302 DOI: 10.1002/cmdc.201600443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Indexed: 01/01/2023]
Abstract
Herein we report the synthesis and activity of an enzyme-directed immunostimulant with immune cell activation mediated by β-galactosidase, either exogenously added, or on B16 melanoma cells. Covalent attachment of a β-galactopyranoside to an imidazoquinoline immunostimulant at a position critical for activity resulted in a pro-immunostimulant that could be selectively converted by β-galactosidase into an active immunostimulant. The pro-immunostimulant exhibited β-galactosidase-directed immune cell activation as measured by NF-κB transcription in RAW-Blue macrophages or cytokine production (TNF, IL-6, IL-12) in JAWSII monocytes. Conversion of the pro-immunostimulant into an active immunostimulant was also found to occur using β-galactosidase-enriched B16 melanoma cells. In co-culture experiments with either immune cell line, β-galactosidase-enriched B16 cells effected activation of bystander immune cells.
Collapse
Affiliation(s)
- Joseph D Hantho
- Department of Chemistry, Washington State University, 100 Dairy Road, Pullman, WA, 99164, USA
| | - Timothy A Strayer
- Department of Chemistry, Washington State University, 100 Dairy Road, Pullman, WA, 99164, USA
| | - Amy E Nielsen
- Department of Chemistry, Washington State University, 100 Dairy Road, Pullman, WA, 99164, USA
| | - Rock J Mancini
- Department of Chemistry, Washington State University, 100 Dairy Road, Pullman, WA, 99164, USA
| |
Collapse
|
34
|
Wu TYH. Strategies for designing synthetic immune agonists. Immunology 2016; 148:315-25. [PMID: 27213842 DOI: 10.1111/imm.12622] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 12/16/2022] Open
Abstract
Enhancing the immune system is a validated strategy to combat infectious disease, cancer and allergy. Nevertheless, the development of immune adjuvants has been hampered by safety concerns. Agents that can stimulate the immune system often bear structural similarities with pathogen-associated molecular patterns found in bacteria or viruses and are recognized by pattern recognition receptors (PRRs). Activation of these PRRs results in the immediate release of inflammatory cytokines, up-regulation of co-stimulatory molecules, and recruitment of innate immune cells. The distribution and duration of these early inflammatory events are crucial in the development of antigen-specific adaptive immunity in the forms of antibody and/or T cells capable of searching for and destroying the infectious pathogens or cancer cells. However, systemic activation of these PRRs is often poorly tolerated. Hence, different strategies have been employed to modify or deliver immune agonists in an attempt to control the early innate receptor activation through temporal or spatial restriction. These approaches include physicochemical manipulation, covalent conjugation, formulation and conditional activation/deactivation. This review will describe recent examples of discovery and optimization of synthetic immune agonists towards clinical application.
Collapse
|
35
|
Shah M, Anwar MA, Kim JH, Choi S. Advances in Antiviral Therapies Targeting Toll-like Receptors. Expert Opin Investig Drugs 2016; 25:437-53. [DOI: 10.1517/13543784.2016.1154040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Delaney S, Biffen M, Maltby J, Bell J, Asimus S, Aggarwal A, Kraan M, Keeling D. Tolerability in man following inhalation dosing of the selective TLR7 agonist, AZD8848. BMJ Open Respir Res 2016; 3:e000113. [PMID: 26933507 PMCID: PMC4769423 DOI: 10.1136/bmjresp-2015-000113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 01/22/2023] Open
Abstract
Background Many patients with asthma have a T-helper type 2 (Th2) driven inflammation of the lung, whereas toll-like receptor 7 (TLR7) agonists, by inducing type I interferons, inhibit Th2 responses. In man, oral or parenteral TLR7 agonists can induce influenza-like symptoms through systemic induction of type I interferons. Design of a TLR7 agonist that is only active in the lung could reduce the risk of side effects and offer a new means for treating asthma. We developed a TLR7 agonist antedrug, AZD8848, to determine its local and systemic effects in preclinical models and man. Methods In vitro cellular potencies for the TLR7 antedrug agonist, AZD8848, were determined along with pharmacokinetics and efficacy in a rat allergy model. Sputum and blood biomarkers were measured in single ascending and multiple ascending dose clinical studies following inhalation delivery of AZD8848 and tolerability assessed. Results AZD8848 was potent in cellular assays and pharmacokinetics confirmed lack of systemic exposure to AZD8848. Weekly lung dosing in an animal model showed efficacy 26 days beyond the final dose. In healthy volunteers, AZD8848 was initially well tolerated with target engagement being demonstrated by induction of CXCL10 in sputum. A second inhaled dose, given 1 week later, amplified the systemic interferon signal in more than half the participants and resulted in significant influenza-like symptoms. Conclusions The antedrug design restricted the direct actions of AZD8848 to the lung. However, the type I interferon induced locally by TLR7 spilled over systemically, limiting the utility of this inhaled antedrug approach. Trial registration number NCT01560234, NCT01818869.
Collapse
Affiliation(s)
- Stephen Delaney
- Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development, AstraZeneca , Mölndal , Sweden
| | - Mark Biffen
- Bisocience, AstraZeneca R&D Charnwood , Loughborough , UK
| | - Justine Maltby
- Personalised Healthcare and Biomarkers, Innovative Medicines and Early Development, Alderley Park , Macclesfield , UK
| | - John Bell
- Bisocience, AstraZeneca R&D Charnwood , Loughborough , UK
| | - Sara Asimus
- Quantitative Clinical Pharmacology, Early Clinical Development, AstraZeneca , Mölndal , Sweden
| | - Ajay Aggarwal
- Early Clinical Development, Innovative Medicines and Early Development, AstraZeneca , Waltham, Massachusetts , USA
| | - Maarten Kraan
- Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development, AstraZeneca , Mölndal , Sweden
| | - David Keeling
- Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development, AstraZeneca , Mölndal , Sweden
| |
Collapse
|
37
|
Biggadike K, Ahmed M, Ball DI, Coe DM, Dalmas Wilk DA, Edwards CD, Gibbon BH, Hardy CJ, Hermitage SA, Hessey JO, Hillegas AE, Hughes SC, Lazarides L, Lewell XQ, Lucas A, Mallett DN, Price MA, Priest FM, Quint DJ, Shah P, Sitaram A, Smith SA, Stocker R, Trivedi NA, Tsitoura DC, Weller V. Discovery of 6-Amino-2-{[(1S)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one (GSK2245035), a Highly Potent and Selective Intranasal Toll-Like Receptor 7 Agonist for the Treatment of Asthma. J Med Chem 2016; 59:1711-26. [PMID: 26861551 DOI: 10.1021/acs.jmedchem.5b01647] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Induction of IFNα in the upper airways via activation of TLR7 represents a novel immunomodulatory approach to the treatment of allergic asthma. Exploration of 8-oxoadenine derivatives bearing saturated oxygen or nitrogen heterocycles in the N-9 substituent has revealed a remarkable selective enhancement in IFNα inducing potency in the nitrogen series. Further potency enhancement was achieved with the novel (S)-pentyloxy substitution at C-2 leading to the selection of GSK2245035 (32) as an intranasal development candidate. In human cell cultures, compound 32 resulted in suppression of Th2 cytokine responses to allergens, while in vivo intranasal administration at very low doses led to local upregulation of TLR7-mediated cytokines (IP-10). Target engagement was confirmed in humans following single intranasal doses of 32 of ≥20 ng, and reproducible pharmacological response was demonstrated following repeat intranasal dosing at weekly intervals.
Collapse
Affiliation(s)
- Keith Biggadike
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Mahbub Ahmed
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Doug I Ball
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Diane M Coe
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Deidre A Dalmas Wilk
- GlaxoSmithKline R&D, UpperMerion , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Chris D Edwards
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Bob H Gibbon
- GlaxoSmithKline R&D, David Jack Centre , Park Road, Ware, Hertfordshire SG12 ODP, U.K
| | - Charlotte J Hardy
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Stephen A Hermitage
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Joanne O Hessey
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Aimee E Hillegas
- GlaxoSmithKline R&D, UpperMerion , 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Stephen C Hughes
- GlaxoSmithKline R&D, David Jack Centre , Park Road, Ware, Hertfordshire SG12 ODP, U.K
| | - Linos Lazarides
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Xiao Q Lewell
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Amanda Lucas
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - David N Mallett
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Mark A Price
- GlaxoSmithKline R&D, David Jack Centre , Park Road, Ware, Hertfordshire SG12 ODP, U.K
| | - Fiona M Priest
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Diana J Quint
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Poonam Shah
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Anesh Sitaram
- GlaxoSmithKline R&D, David Jack Centre , Park Road, Ware, Hertfordshire SG12 ODP, U.K
| | - Stephen A Smith
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Richard Stocker
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Naimisha A Trivedi
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Daphne C Tsitoura
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Victoria Weller
- GlaxoSmithKline R&D, Medicines Research Centre , Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
38
|
Tsitoura D, Ambery C, Price M, Powley W, Garthside S, Biggadike K, Quint D. Early clinical evaluation of the intranasal TLR7 agonist GSK2245035: Use of translational biomarkers to guide dosing and confirm target engagement. Clin Pharmacol Ther 2015; 98:369-80. [PMID: 26044169 DOI: 10.1002/cpt.157] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/29/2015] [Indexed: 12/30/2022]
Abstract
Modulation of the airways' immune milieu is a key therapeutic goal for remission from respiratory allergies. To explore this hypothesis, GSK2245035, a selective Toll-like receptor 7 (TLR7) agonist with preferential Type-1 interferon (IFN)-stimulating properties, was developed for intranasal application. Doses for clinical assessment were extrapolated from translational biomarker studies in primates. Randomized, double-blind, placebo-controlled trials in healthy volunteers and patients with allergic rhinitis demonstrated that intranasal GSK2245035 doses <100 ng were tolerated and did not cause nasal inflammation. Higher doses were not tested due to considerable cytokine release syndrome-related symptoms observed at 100 ng. Clear target engagement, reflected by local and peripheral increase of IFN-gamma-inducible protein-10, was observed at 20 ng, indicating IFN-stimulated immune changes at tolerated doses. Repeat intranasal administration at weekly intervals did not tolerize or amplify the pharmacological response. Intranasal GSK2245035 has an acceptable safety profile at doses that induce local TLR7-mediated immune responses.
Collapse
Affiliation(s)
- D Tsitoura
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - C Ambery
- GlaxoSmithKline, Uxbridge, Middlesex, UK
| | - M Price
- GlaxoSmithKline, Ware, Hertfordshire, UK
| | - W Powley
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | | | - K Biggadike
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - D Quint
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
39
|
Koga-Yamakawa E, Murata M, Dovedi SJ, Wilkinson RW, Ota Y, Umehara H, Sugaru E, Hirose Y, Harada H, Jewsbury PJ, Yamamoto S, Robinson DT, Li CJ. TLR7 tolerance is independent of the type I IFN pathway and leads to loss of anti-tumor efficacy in mice. Cancer Immunol Immunother 2015; 64:1229-39. [PMID: 26091797 PMCID: PMC11029383 DOI: 10.1007/s00262-015-1730-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 05/30/2015] [Indexed: 02/05/2023]
Abstract
Systemic administration of small molecule toll-like receptor (TLR)-7 agonists leads to potent activation of innate immunity and to the generation of anti-tumor immune responses. However, activation of TLRs with small molecule agonists may lead to the induction of TLR tolerance, defined as a state of hyporesponsiveness to subsequent agonism, which may limit immune activation, the generation of anti-tumor responses and clinical response. Our data reveal that dose scheduling impacts on the efficacy of systemic therapy with the selective TLR7 agonist, 6-amino-2-(butylamino)-9-((6-(2-(dimethylamino)ethoxy)pyridin-3-yl)methyl)-7,9-dihydro-8H-purin-8-one (DSR-6434). In a preclinical model of renal cell cancer, systemic administration of DSR-6434 dosed once weekly resulted in a significant anti-tumor response. However, twice weekly dosing of DSR-6434 led to the induction of TLR tolerance, and no anti-tumor response was observed. We show that TLR7 tolerance was independent of type I interferon (IFN) negative feedback because induction of TLR7 tolerance was also observed in IFN-α/β receptor knockout mice treated with DSR-6434. Moreover, our data demonstrate that treatment of bone marrow-derived plasmacytoid dendritic cells (BM-pDC) with DSR-6434 led to downregulation of TLR7 expression. From our data, dose scheduling of systemically administered TLR7 agonists can impact on anti-tumor activity through the induction of TLR tolerance. Furthermore, TLR7 expression on pDC may be a useful biomarker of TLR7 tolerance and aid in the optimization of dosing schedules involving systemically administered TLR7 agonists.
Collapse
Affiliation(s)
- Erina Koga-Yamakawa
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - Masashi Murata
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - Simon J. Dovedi
- Oncology Innovative Medicines and Early Development (iMed), AstraZeneca, Alderley Park, Macclesfield, SK10 4TG UK
- Present Address: Manchester Cancer Research Centre, Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - Robert W. Wilkinson
- Oncology Innovative Medicines and Early Development (iMed), AstraZeneca, Alderley Park, Macclesfield, SK10 4TG UK
- Present Address: MedImmune Ltd, Milstein Building, Granta Park, Cambridge, UK
| | - Yosuke Ota
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - Hiroki Umehara
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
- Boston Biomedical, Inc., 640 Memorial Drive, Cambridge, MA USA
| | - Eiji Sugaru
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - Yuko Hirose
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - Hideyuki Harada
- Drug Research Division, Sumitomo Dainippon Pharma, 33-94, Enoki-cho, Suita, Osaka 564-0053 Japan
| | - Philip J. Jewsbury
- Oncology Innovative Medicines and Early Development (iMed), AstraZeneca, Alderley Park, Macclesfield, SK10 4TG UK
| | - Setsuko Yamamoto
- Sumitomo Dainippon Pharma (DSP) Cancer Institute, Sumitomo Dainippon Pharma, 3-1-98, Kasugade Naka, Konohana-ku, Osaka, 554-0022 Japan
| | - David T. Robinson
- Oncology Innovative Medicines and Early Development (iMed), AstraZeneca, Alderley Park, Macclesfield, SK10 4TG UK
| | - Chiang J. Li
- Boston Biomedical, Inc., 640 Memorial Drive, Cambridge, MA USA
| |
Collapse
|
40
|
Rajaram S, Bright H. Commentary on articles published on 2010 Southern Hemisphere Trivalent Influenza Vaccine association with adverse events. Vaccine 2015; 33:3271-2. [PMID: 25239485 DOI: 10.1016/j.vaccine.2014.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/04/2014] [Accepted: 09/07/2014] [Indexed: 10/24/2022]
|
41
|
Hoffmann HH, Schneider WM, Rice CM. Interferons and viruses: an evolutionary arms race of molecular interactions. Trends Immunol 2015; 36:124-38. [PMID: 25704559 DOI: 10.1016/j.it.2015.01.004] [Citation(s) in RCA: 308] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 12/24/2022]
Abstract
Over half a century has passed since interferons (IFNs) were discovered and shown to inhibit virus infection in cultured cells. Since then, researchers have steadily brought to light the molecular details of IFN signaling, catalogued their pleiotropic effects on cells, and harnessed their therapeutic potential for a variety of maladies. While advances have been plentiful, several fundamental questions have yet to be answered and much complexity remains to be unraveled. We explore the current knowledge surrounding four main questions: are type I IFN subtypes differentially produced in response to distinct pathogens? How are IFN subtypes distinguished by cells? What are the mechanisms and consequences of viral antagonism? Lastly, how can the IFN response be harnessed to improve vaccine efficacy?
Collapse
Affiliation(s)
- Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - William M Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
42
|
Eyre NS, Helbig KJ, Beard MR. Current and future targets of antiviral therapy in the hepatitis C virus life cycle. Future Virol 2014. [DOI: 10.2217/fvl.14.83] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT Advances in our understanding of the hepatitis C virus (HCV) life cycle have enabled the development of numerous clinically advanced direct-acting antivirals. Indeed, the recent approval of first-generation direct-acting antivirals that target the viral NS3–4A protease and NS5B RNA-dependent RNA polymerase brings closer the possibility of universally efficacious and well-tolerated antiviral therapies for this insidious infection. However, the complexities of comorbidities, unforeseen side effects or drug–drug interactions, viral diversity, the high mutation rate of HCV RNA replication and the elegant and constantly evolving mechanisms employed by HCV to evade host and therapeutically implemented antiviral strategies remain as significant obstacles to this goal. Here, we review advances in our understanding of the HCV life cycle and associated opportunities for antiviral therapy.
Collapse
Affiliation(s)
- Nicholas S Eyre
- School of Molecular & Biomedical Science, The University of Adelaide & Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Karla J Helbig
- School of Molecular & Biomedical Science, The University of Adelaide & Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Michael R Beard
- School of Molecular & Biomedical Science, The University of Adelaide & Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| |
Collapse
|
43
|
Patel MC, Shirey KA, Pletneva LM, Boukhvalova MS, Garzino-Demo A, Vogel SN, Blanco JC. Novel drugs targeting Toll-like receptors for antiviral therapy. Future Virol 2014; 9:811-829. [PMID: 25620999 DOI: 10.2217/fvl.14.70] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Toll-like receptors (TLRs) are sentinel receptors of the host innate immune system that recognize conserved 'pathogen-associated molecular patterns' of invading microbes, including viruses. The activation of TLRs establishes antiviral innate immune responses and coordinates the development of long-lasting adaptive immunity in order to control viral pathogenesis. However, microbe-induced damage to host tissues may release 'danger-associated molecular patterns' that also activate TLRs, leading to an overexuberant inflammatory response and, ultimately, to tissue damage. Thus, TLRs have proven to be promising targets as therapeutics for the treatment of viral infections that result in inflammatory damage or as adjuvants in order to enhance the efficacy of vaccines. Here, we explore recent advances in TLR biology with a focus on novel drugs that target TLRs (agonists and antagonists) for antiviral therapy.
Collapse
Affiliation(s)
- Mira C Patel
- Department of Microbiology & Immunology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Kari Ann Shirey
- Department of Microbiology & Immunology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | | | | | - Alfredo Garzino-Demo
- Department of Microbiology & Immunology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA ; Institute of Human Virology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Stefanie N Vogel
- Department of Microbiology & Immunology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
44
|
Ireton RC, Gale M. Pushing to a cure by harnessing innate immunity against hepatitis C virus. Antiviral Res 2014; 108:156-64. [PMID: 24907428 DOI: 10.1016/j.antiviral.2014.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 05/08/2014] [Accepted: 05/23/2014] [Indexed: 02/08/2023]
Abstract
Hepatitis C virus (HCV) causes 350,000 deaths and infects at least 3million people worldwide every year. Currently no vaccine has been developed. Direct-acting antiviral (DAA) drugs with high efficacy for suppressing HCV infection have recently been introduced into the clinic. While DAAs initially required combination therapy with type-1 interferon (IFN) administration for full efficacy and to avoid viral resistance to treatment, new DAA combinations show promise as an IFN-free regimen. However, IFN-free DAA therapy is in its infancy, still to be proven and today is cost-prohibitive for the patient. A major goal in HCV therapy to remove or replace IFN with DAAs or an alternative therapeutic to render virologic response with continued virus sensitivity to DAAs, thus facilitating a cure for infection. Recent advances in our understanding of innate immune responses to HCV have identified new therapeutic targets to combat HCV infection. We discuss how the targeting of innate immune response factors can be harnessed with DAAs to produce new generations of DAA-based HCV therapeutics. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."
Collapse
Affiliation(s)
- Reneé C Ireton
- Center for the Study of Innate Immunity to Hepatitis C Virus, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, United States.
| | - Michael Gale
- Center for the Study of Innate Immunity to Hepatitis C Virus, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, United States.
| |
Collapse
|
45
|
Funk E, Kottilil S, Gilliam B, Talwani R. Tickling the TLR7 to cure viral hepatitis. J Transl Med 2014; 12:129. [PMID: 24884741 PMCID: PMC4039542 DOI: 10.1186/1479-5876-12-129] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/16/2014] [Indexed: 01/01/2023] Open
Abstract
Chronic hepatitis B and C are the leading causes of liver disease and liver transplantation worldwide. Ability to mount an effective immune response against both HBV and HCV is associated with spontaneous clearance of both infections, while an inability to do so leads to chronicity of both infections. To mount an effective immune response, both innate and adaptive immune responses must work in tandem. Hence, developing protective immunity to hepatitis viruses is an important goal in order to reduce the global burden of these two infections and prevent development of long-term complications. In this regard, the initial interactions between the pathogen and immune system are pivotal in determining the effectiveness of immune response and subsequent elimination of pathogens. Toll-like receptors (TLRs) are important regulators of innate and adaptive immune responses to various pathogens and are often involved in initiating and augmenting effective antiviral immunity. Immune-based therapeutic strategies that specifically induce type I interferon responses are associated with functional cure for both chronic HBV and HCV infections. Precisely, TLR7 stimulation mediates an endogenous type I interferon response, which is critical in development of a broad, effective and protective immunity against hepatitis viruses. This review focuses on anti-viral strategies that involve targeting TLR7 that may lead to development of protective immunity and eradication of hepatitis B.
Collapse
Affiliation(s)
- Emily Funk
- Critical Care Medicine Department, Clinical Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shyam Kottilil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bruce Gilliam
- Institute of Human Virology at the University of Maryland School of Medicine, 725 West Lombard St. N151, Baltimore, MD 21201, USA
| | - Rohit Talwani
- Institute of Human Virology at the University of Maryland School of Medicine, 725 West Lombard St. N151, Baltimore, MD 21201, USA
| |
Collapse
|
46
|
Mifsud EJ, Tan ACL, Jackson DC. TLR Agonists as Modulators of the Innate Immune Response and Their Potential as Agents Against Infectious Disease. Front Immunol 2014; 5:79. [PMID: 24624130 PMCID: PMC3939722 DOI: 10.3389/fimmu.2014.00079] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/13/2014] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies that can either activate or suppress innate immune responses are being investigated as treatments against infectious diseases and the pathology they can cause. The objective of these therapies is to elicit protective immune responses thereby limiting the harm inflicted by the pathogen. The Toll-like receptor (TLR) signaling pathway plays critical roles in numerous host immune defenses and has been identified as an immunotherapeutic target against the consequences of infectious challenge. This review focuses on some of the recent advances being made in the development of TLR-ligands as potential prophylactic and/or therapeutic agents.
Collapse
Affiliation(s)
- Edin J. Mifsud
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Amabel C. L. Tan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David C. Jackson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Sajadian A, Tabarraei A, Soleimanjahi H, Fotouhi F, Gorji A, Ghaemi A. Comparing the effect of Toll-like receptor agonist adjuvants on the efficiency of a DNA vaccine. Arch Virol 2014; 159:1951-60. [PMID: 24573220 DOI: 10.1007/s00705-014-2024-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/08/2014] [Indexed: 11/29/2022]
Abstract
We have investigated whether poly(I:C) Toll-like receptor 3 (TLR3) and resiquimod Toll-like receptor 7 (TLR7) agonists can serve as vaccine adjuvants and promote the efficiency of therapeutic DNA vaccination against tumors expressing the human papilloma virus 16 (HPV-16) E7 protein. For this purpose, C57BL/6 mice were inoculated with 2 × 10(5) TC-1 cells, and they were then immunized with HPV-16 E7 DNA vaccine alone or with 50 μg of resiquimod or poly(I:C) individually. We found that poly(I:C) and resiquimod could induce more antigen-specific lymphocyte proliferation and cytolytic activity compared to vaccination with E7 DNA alone. While E7 DNA had no significant inhibitory effect on tumor growth, co-administration of poly(I:C) and resiquimod with E7 DNA induced significant tumor regression. Peripheral and local cytokine assays demonstrated that co-administration of poly(I:C) and resiquimod with E7 DNA induced circulating antigen-specific IFN-γ and nonspecific intratumoral IL-12. TLR3 and TLR7 agonists can be used to enhance the immune response to DNA vaccine immunogens. Taken together, these data indicate that combined vaccination with DNA encoding HPV-16 E7 plus TLR agonists provides a strategy for improving the efficacy of a vaccine as a possible immunotherapeutic strategy for cervical cancer.
Collapse
|
48
|
Hussein WM, Liu TY, Skwarczynski M, Toth I. Toll-like receptor agonists: a patent review (2011 - 2013). Expert Opin Ther Pat 2014; 24:453-70. [PMID: 24456079 DOI: 10.1517/13543776.2014.880691] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Toll-like receptors (TLRs) are a crucial part of the innate immunity and present the first line of defense against pathogens. In humans, there are ten TLRs, with TLR3, 7, 8 and 9 located in intracellular vesicles and the remaining expressed on the cell surface. These transmembrane protein receptors recognize a wide range of pathogen components. A large number of TLR agonists, either derived from pathogen components or modified synthetic molecules, were developed and investigated for their ability to stimulate an immune response. AREAS COVERED This review includes an updated summary (2011 - 2013) of TLR agonists that have been published in patent applications and/or progressed to clinical studies, with an emphasis on their chemical structure, immune response, prophylactic and therapeutic outcomes. EXPERT OPINION A number of factors have contributed to the design and development of TLR agonists such as solving the crystal structures of TLR bound to their ligands, improvements in our understanding of the signaling pathway activated after TLR stimulation and the identification of the native ligands of all human TLRs. Some of the TLR agonists have been approved for human use by the FDA while others have reached clinical studies in Phases I, II and III. Generally, immunotherapy based on TLR agonists is very promising for the prevention and/or treatment of several disorders including cancer, allergy and microbial infections. However, many TLR agonists were withdrawn from further studies as they either lacked efficacy or caused serious side effects.
Collapse
Affiliation(s)
- Waleed M Hussein
- The University of Queensland, School of Chemistry and Molecular Biosciences , St. Lucia, Brisbane, Qld 4072 , Australia
| | | | | | | |
Collapse
|
49
|
Savva A, Roger T. Targeting toll-like receptors: promising therapeutic strategies for the management of sepsis-associated pathology and infectious diseases. Front Immunol 2013; 4:387. [PMID: 24302927 PMCID: PMC3831162 DOI: 10.3389/fimmu.2013.00387] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/05/2013] [Indexed: 01/04/2023] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors playing a fundamental role in sensing microbial invasion and initiating innate and adaptive immune responses. TLRs are also triggered by danger signals released by injured or stressed cells during sepsis. Here we focus on studies developing TLR agonists and antagonists for the treatment of infectious diseases and sepsis. Positioned at the cell surface, TLR4 is essential for sensing lipopolysaccharide of Gram-negative bacteria, TLR2 is involved in the recognition of a large panel of microbial ligands, while TLR5 recognizes flagellin. Endosomal TLR3, TLR7, TLR8, TLR9 are specialized in the sensing of nucleic acids produced notably during viral infections. TLR4 and TLR2 are favorite targets for developing anti-sepsis drugs, and antagonistic compounds have shown efficient protection from septic shock in pre-clinical models. Results from clinical trials evaluating anti-TLR4 and anti-TLR2 approaches are presented, discussing the challenges of study design in sepsis and future exploitation of these agents in infectious diseases. We also report results from studies suggesting that the TLR5 agonist flagellin may protect from infections of the gastrointestinal tract and that agonists of endosomal TLRs are very promising for treating chronic viral infections. Altogether, TLR-targeted therapies have a strong potential for prevention and intervention in infectious diseases, notably sepsis.
Collapse
Affiliation(s)
- Athina Savva
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne , Lausanne , Switzerland
| | | |
Collapse
|
50
|
Roethle PA, McFadden RM, Yang H, Hrvatin P, Hui H, Graupe M, Gallagher B, Chao J, Hesselgesser J, Duatschek P, Zheng J, Lu B, Tumas DB, Perry J, Halcomb RL. Identification and Optimization of Pteridinone Toll-like Receptor 7 (TLR7) Agonists for the Oral Treatment of Viral Hepatitis. J Med Chem 2013; 56:7324-33. [DOI: 10.1021/jm400815m] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Paul A. Roethle
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Ryan M. McFadden
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Hong Yang
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Paul Hrvatin
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Hon Hui
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Michael Graupe
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Brian Gallagher
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jessica Chao
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Joseph Hesselgesser
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Paul Duatschek
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jim Zheng
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Bing Lu
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Daniel B. Tumas
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jason Perry
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| | - Randall L. Halcomb
- Departments of †Medicinal Chemistry, ‡Clinical Virology, §Drug Metabolism, ∥Biology, and ⊥Structural Chemistry, Gilead Sciences, 333 Lakeside Drive, Foster City, California 94404, United States
| |
Collapse
|