1
|
Zuo X, Cheng Q, Wang Z, Liu J, Lu W, Wu G, Zhu S, Liu X, Lv T, Song Y. A novel oral TLR7 agonist orchestrates immune response and synergizes with PD-L1 blockade via type I IFN pathway in lung cancer. Int Immunopharmacol 2024; 137:112478. [PMID: 38901243 DOI: 10.1016/j.intimp.2024.112478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/21/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Despite the groundbreaking impact of immune checkpoint blockade (ICB), response rates in non-small cell lung cancer remain modest, particularly in immune-excluded or immune-desert microenvironments. Toll-like receptor 7 (TLR7) emerges as a latent target bridging innate and adaptive immunity, offering a promising avenue for combination therapies to augment ICB efficacy. Here, we explored the anti-tumor activity of the novel oral TLR7 agonist TQ-A3334 and its potential to enhance anti-programmed death ligand 1 (PD-L1) therapy through a combination strategy in a syngeneic murine lung cancer model. Oral administration of TQ-A3334 significantly alleviated tumor burden in C57BL/6J mice, modulated by type I interferon (IFN), and exhibited low toxicity. This therapy elicited activation of both innate and adaptive immune cells in tumor tissue, particularly increasing the abundance of CD8+ TILs through type I IFN pathway and subsequent CXCL10 expression. In vitro examinations validated that IFN-α-stimulated tumor cells exhibited increased secretion of CXCL10, conducive to the promoted trafficking of CD8+ T cells. Furthermore, combining TQ-A3334 with anti-PD-L1 treatment exceeded tumor control, with a further increase in CD8+ TIL frequency compared to monotherapy. These findings suggest that TQ-A3334 can mobilize innate immunity and promote T cell recruitment into the tumor microenvironment; a combination of TQ-A3334 and anti-PD-L1 antibodies can intensify the sensitivity of tumors to anti-PD-L1 therapy, which demonstrates significant potential for treating poorly immune-infiltrated lung cancer.
Collapse
Affiliation(s)
- Xueying Zuo
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Zimu Wang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210008, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210008, Jiangsu, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Guannan Wu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Xin Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China.
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu, China.
| |
Collapse
|
2
|
Li MY, Ye W, Luo KW. Immunotherapies Targeting Tumor-Associated Macrophages (TAMs) in Cancer. Pharmaceutics 2024; 16:865. [PMID: 39065562 PMCID: PMC11280177 DOI: 10.3390/pharmaceutics16070865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are one of the most plentiful immune compositions in the tumor microenvironment, which are further divided into anti-tumor M1 subtype and pro-tumor M2 subtype. Recent findings found that TAMs play a vital function in the regulation and progression of tumorigenesis. Moreover, TAMs promote tumor vascularization, and support the survival of tumor cells, causing an impact on tumor growth and patient prognosis. Numerous studies show that reducing the density of TAMs, or modulating the polarization of TAMs, can inhibit tumor growth, indicating that TAMs are a promising target for tumor immunotherapy. Recently, clinical trials have found that treatments targeting TAMs have achieved encouraging results, and the U.S. Food and Drug Administration has approved a number of drugs for use in cancer treatment. In this review, we summarize the origin, polarization, and function of TAMs, and emphasize the therapeutic strategies targeting TAMs in cancer treatment in clinical studies and scientific research, which demonstrate a broad prospect of TAMs-targeted therapies in tumor immunotherapy.
Collapse
Affiliation(s)
- Mei-Ye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
| | - Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
| | - Ke-Wang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
- People’s Hospital of Longhua, The affiliated hospital of Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
3
|
Yu Z, Zou J, Xu F. Tumor-associated macrophages affect the treatment of lung cancer. Heliyon 2024; 10:e29332. [PMID: 38623256 PMCID: PMC11016713 DOI: 10.1016/j.heliyon.2024.e29332] [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: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
As one of the most common malignant tumors in the world, lung cancer has limited benefits for patients despite its diverse treatment methods due to factors such as personalized medicine targeting histological type, immune checkpoint expression, and driver gene mutations. The high mortality rate of lung cancer is partly due to the immune-suppressive which limits the effectiveness of anti-cancer drugs and induces tumor cell resistance. The currently widely recognized TAM phenotypes include the anti-tumor M1 and pro-tumor M2 phenotypes. M2 macrophages promote the formation of an immune-suppressive microenvironment and hinder immune cell infiltration, thereby inhibiting activation of the anti-tumor immune system and aiding tumor cells in resisting treatment. Analyzing the relationship between different treatment methods and macrophages in the TME can help us better understand the impact of TAMs on lung cancer and confirm the feasibility of targeted TAM therapy. Targeting TAMs to reduce the M2/M1 ratio and reverse the immune-suppressive microenvironment can improve the clinical efficacy of conventional treatment methods and potentially open up more efficient combination treatment strategies, maximizing the benefit for lung cancer patients.
Collapse
Affiliation(s)
- Zhuchen Yu
- Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Juntao Zou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Fei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
4
|
Zhang F, Huang B, Utturkar SM, Luo W, Cresswell G, Herr SA, Zheng S, Napoleon JV, Jiang R, Zhang B, Liu M, Lanman N, Srinivasarao M, Ratliff TL, Low PS. Tumor-specific activation of folate receptor beta enables reprogramming of immune cells in the tumor microenvironment. Front Immunol 2024; 15:1354735. [PMID: 38384467 PMCID: PMC10879311 DOI: 10.3389/fimmu.2024.1354735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Folate receptors can perform folate transport, cell adhesion, and/or transcription factor functions. The beta isoform of the folate receptor (FRβ) has attracted considerable attention as a biomarker for immunosuppressive macrophages and myeloid-derived suppressor cells, however, its role in immunosuppression remains uncharacterized. We demonstrate here that FRβ cannot bind folate on healthy tissue macrophages, but does bind folate after macrophage incubation in anti-inflammatory cytokines or cancer cell-conditioned media. We further show that FRβ becomes functionally active following macrophage infiltration into solid tumors, and we exploit this tumor-induced activation to target a toll-like receptor 7 agonist specifically to immunosuppressive myeloid cells in solid tumors without altering myeloid cells in healthy tissues. We then use single-cell RNA-seq to characterize the changes in gene expression induced by the targeted repolarization of tumor-associated macrophages and finally show that their repolarization not only changes their own phenotype, but also induces a proinflammatory shift in all other immune cells of the same tumor mass, leading to potent suppression of tumor growth. Because this selective reprogramming of tumor myeloid cells is accompanied by no systemic toxicity, we propose that it should constitute a safe method to reprogram the tumor microenvironment.
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Bo Huang
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Sagar M. Utturkar
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Weichuan Luo
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Gregory Cresswell
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Seth A. Herr
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Suilan Zheng
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - John V. Napoleon
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Rina Jiang
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Boning Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Muyi Liu
- University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, United States
- Department of Computer Sciences, Purdue University, West Lafayette, IN, United States
| | - Nadia Lanman
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Timothy L. Ratliff
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Philip S. Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
5
|
Mukherjee S, Patra R, Behzadi P, Masotti A, Paolini A, Sarshar M. Toll-like receptor-guided therapeutic intervention of human cancers: molecular and immunological perspectives. Front Immunol 2023; 14:1244345. [PMID: 37822929 PMCID: PMC10562563 DOI: 10.3389/fimmu.2023.1244345] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 10/13/2023] Open
Abstract
Toll-like receptors (TLRs) serve as the body's first line of defense, recognizing both pathogen-expressed molecules and host-derived molecules released from damaged or dying cells. The wide distribution of different cell types, ranging from epithelial to immune cells, highlights the crucial roles of TLRs in linking innate and adaptive immunity. Upon stimulation, TLRs binding mediates the expression of several adapter proteins and downstream kinases, that lead to the induction of several other signaling molecules such as key pro-inflammatory mediators. Indeed, extraordinary progress in immunobiological research has suggested that TLRs could represent promising targets for the therapeutic intervention of inflammation-associated diseases, autoimmune diseases, microbial infections as well as human cancers. So far, for the prevention and possible treatment of inflammatory diseases, various TLR antagonists/inhibitors have shown to be efficacious at several stages from pre-clinical evaluation to clinical trials. Therefore, the fascinating role of TLRs in modulating the human immune responses at innate as well as adaptive levels directed the scientists to opt for these immune sensor proteins as suitable targets for developing chemotherapeutics and immunotherapeutics against cancer. Hitherto, several TLR-targeting small molecules (e.g., Pam3CSK4, Poly (I:C), Poly (A:U)), chemical compounds, phytocompounds (e.g., Curcumin), peptides, and antibodies have been found to confer protection against several types of cancers. However, administration of inappropriate doses of such TLR-modulating therapeutics or a wrong infusion administration is reported to induce detrimental outcomes. This review summarizes the current findings on the molecular and structural biology of TLRs and gives an overview of the potency and promises of TLR-directed therapeutic strategies against cancers by discussing the findings from established and pipeline discoveries.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Ritwik Patra
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Alessandro Paolini
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| |
Collapse
|
6
|
Margul D, Yu C, AlHilli MM. Tumor Immune Microenvironment in Gynecologic Cancers. Cancers (Basel) 2023; 15:3849. [PMID: 37568665 PMCID: PMC10417375 DOI: 10.3390/cancers15153849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Gynecologic cancers have varying response rates to immunotherapy due to the heterogeneity of each cancer's molecular biology and features of the tumor immune microenvironment (TIME). This article reviews key features of the TIME and its role in the pathophysiology and treatment of ovarian, endometrial, cervical, vulvar, and vaginal cancer. Knowledge of the role of the TIME in gynecologic cancers has been rapidly developing with a large body of preclinical studies demonstrating an intricate yet dichotomous role that the immune system plays in either supporting the growth of cancer or opposing it and facilitating effective treatment. Many targets and therapeutics have been identified including cytokines, antibodies, small molecules, vaccines, adoptive cell therapy, and bacterial-based therapies but most efforts in gynecologic cancers to utilize them have not been effective. However, with the development of immune checkpoint inhibitors, we have started to see the rapid and successful employment of therapeutics in cervical and endometrial cancer. There remain many challenges in utilizing the TIME, particularly in ovarian cancer, and further studies are needed to identify and validate efficacious therapeutics.
Collapse
Affiliation(s)
| | | | - Mariam M. AlHilli
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cleveland Clinic, Cleveland, OH 44195, USA; (D.M.); (C.Y.)
| |
Collapse
|
7
|
Bhagchandani SH, Vohidov F, Milling LE, Tong EY, Brown CM, Ramseier ML, Liu B, Fessenden TB, Nguyen HVT, Kiel GR, Won L, Langer RS, Spranger S, Shalek AK, Irvine DJ, Johnson JA. Engineering kinetics of TLR7/8 agonist release from bottlebrush prodrugs enables tumor-focused immune stimulation. SCIENCE ADVANCES 2023; 9:eadg2239. [PMID: 37075115 PMCID: PMC10115420 DOI: 10.1126/sciadv.adg2239] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Imidazoquinolines (IMDs), such as resiquimod (R848), are of great interest as potential cancer immunotherapies because of their ability to activate Toll-like receptor 7 (TLR7) and/or TLR8 on innate immune cells. Nevertheless, intravenous administration of IMDs causes severe immune-related toxicities, and attempts to improve their tissue-selective exposure while minimizing acute systemic inflammation have proven difficult. Here, using a library of R848 "bottlebrush prodrugs" (BPDs) that differ only by their R848 release kinetics, we explore how the timing of R848 exposure affects immune stimulation in vitro and in vivo. These studies led to the discovery of R848-BPDs that exhibit optimal activation kinetics to achieve potent stimulation of myeloid cells in tumors and substantial reductions in tumor growth following systemic administration in mouse syngeneic tumor models without any observable systemic toxicity. These results suggest that release kinetics can be tuned at the molecular level to provide safe yet effective systemically administered immunostimulant prodrugs for next-generation cancer immunotherapies.
Collapse
Affiliation(s)
- Sachin H. Bhagchandani
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Farrukh Vohidov
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Lauren E. Milling
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Evelyn Yuzhou Tong
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Christopher M. Brown
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Michelle L. Ramseier
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Bin Liu
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Timothy B. Fessenden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Hung V.-T. Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Gavin R. Kiel
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Lori Won
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Robert S. Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Alex K. Shalek
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
8
|
Truxova I, Cibula D, Spisek R, Fucikova J. Targeting tumor-associated macrophages for successful immunotherapy of ovarian carcinoma. J Immunother Cancer 2023; 11:jitc-2022-005968. [PMID: 36822672 PMCID: PMC9950980 DOI: 10.1136/jitc-2022-005968] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is among the top five causes of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant cells to the peritoneum. Despite improvements in medical therapies, particularly with the implementation of novel drugs targeting homologous recombination deficiency, the survival rates of patients with EOC remain low. Unlike other neoplasms, EOC remains relatively insensitive to immune checkpoint inhibitors, which is correlated with a tumor microenvironment (TME) characterized by poor infiltration by immune cells and active immunosuppression dominated by immune components with tumor-promoting properties, especially tumor-associated macrophages (TAMs). In recent years, TAMs have attracted interest as potential therapeutic targets by seeking to reverse the immunosuppression in the TME and enhance the clinical efficacy of immunotherapy. Here, we review the key biological features of TAMs that affect tumor progression and their relevance as potential targets for treating EOC. We especially focus on the therapies that might modulate the recruitment, polarization, survival, and functional properties of TAMs in the TME of EOC that can be harnessed to develop superior combinatorial regimens with immunotherapy for the clinical care of patients with EOC.
Collapse
Affiliation(s)
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic .,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
9
|
Sun H, Li Y, Zhang P, Xing H, Zhao S, Song Y, Wan D, Yu J. Targeting toll-like receptor 7/8 for immunotherapy: recent advances and prospectives. Biomark Res 2022; 10:89. [PMID: 36476317 PMCID: PMC9727882 DOI: 10.1186/s40364-022-00436-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are a large family of proteins that are expressed in immune cells and various tumor cells. TLR7/8 are located in the intracellular endosomes, participate in tumor immune surveillance and play different roles in tumor growth. Activation of TLRs 7 and 8 triggers induction of a Th1 type innate immune response in the highly sophisticated process of innate immunity signaling with the recent research advances involving the small molecule activation of TLR 7 and 8. The wide range of expression and clinical significance of TLR7/TLR8 in different kinds of cancers have been extensively explored. TLR7/TLR8 can be used as novel diagnostic biomarkers, progression and prognostic indicators, and immunotherapeutic targets for various tumors. Although the mechanism of action of TLR7/8 in cancer immunotherapy is still incomplete, TLRs on T cells are involved in the regulation of T cell function and serve as co-stimulatory molecules and activate T cell immunity. TLR agonists can activate T cell-mediated antitumor responses with both innate and adaptive immune responses to improve tumor therapy. Recently, novel drugs of TLR7 or TLR8 agonists with different scaffolds have been developed. These agonists lead to the induction of certain cytokines and chemokines that can be applied to the treatment of some diseases and can be used as good adjutants for vaccines. Furthermore, TLR7/8 agonists as potential therapeutics for tumor-targeted immunotherapy have been developed. In this review, we summarize the recent advances in the development of immunotherapy strategies targeting TLR7/8 in patients with various cancers and chronic hepatitis B.
Collapse
Affiliation(s)
- Hao Sun
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yingmei Li
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Peng Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Haizhou Xing
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yongping Song
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Dingming Wan
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004 Henan China
| |
Collapse
|
10
|
Yang Y, Li H, Fotopoulou C, Cunnea P, Zhao X. Toll-like receptor-targeted anti-tumor therapies: Advances and challenges. Front Immunol 2022; 13:1049340. [PMID: 36479129 PMCID: PMC9721395 DOI: 10.3389/fimmu.2022.1049340] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors, originally discovered to stimulate innate immune reactions against microbial infection. TLRs also play essential roles in bridging the innate and adaptive immune system, playing multiple roles in inflammation, autoimmune diseases, and cancer. Thanks to the immune stimulatory potential of TLRs, TLR-targeted strategies in cancer treatment have proved to be able to regulate the tumor microenvironment towards tumoricidal phenotypes. Quantities of pre-clinical studies and clinical trials using TLR-targeted strategies in treating cancer have been initiated, with some drugs already becoming part of standard care. Here we review the structure, ligand, signaling pathways, and expression of TLRs; we then provide an overview of the pre-clinical studies and an updated clinical trial watch targeting each TLR in cancer treatment; and finally, we discuss the challenges and prospects of TLR-targeted therapy.
Collapse
Affiliation(s)
- Yang Yang
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Hongyi Li
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Christina Fotopoulou
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Paula Cunnea
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xia Zhao
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Gianni C, Palleschi M, Schepisi G, Casadei C, Bleve S, Merloni F, Sirico M, Sarti S, Cecconetto L, Di Menna G, Schettini F, De Giorgi U. Circulating inflammatory cells in patients with metastatic breast cancer: Implications for treatment. Front Oncol 2022; 12:882896. [PMID: 36003772 PMCID: PMC9393759 DOI: 10.3389/fonc.2022.882896] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
Adaptive and innate immune cells play a crucial role as regulators of cancer development. Inflammatory cells in blood flow seem to be involved in pro-tumor activities and contribute to breast cancer progression. Circulating lymphocyte ratios such as the platelet-lymphocytes ratio (PLR), the monocyte-lymphocyte ratio (MLR) and the neutrophil-lymphocyte ratio (NLR) are new reproducible, routinely feasible and cheap biomarkers of immune response. These indexes have been correlated to prognosis in many solid tumors and there is growing evidence on their clinical applicability as independent prognostic markers also for breast cancer. In this review we give an overview of the possible value of lymphocytic indexes in advanced breast cancer prognosis and prediction of outcome. Furthermore, targeting the immune system appear to be a promising therapeutic strategy for breast cancer, especially macrophage-targeted therapies. Herein we present an overview of the ongoing clinical trials testing systemic inflammatory cells as therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Caterina Gianni,
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Samanta Sarti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lorenzo Cecconetto
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giandomenico Di Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Francesco Schettini
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
12
|
Luo W, Napoleon JV, Zhang F, Lee YG, Wang B, Putt KS, Low PS. Repolarization of Tumor-Infiltrating Myeloid Cells for Augmentation of CAR T Cell Therapies. Front Immunol 2022; 13:816761. [PMID: 35250995 PMCID: PMC8889096 DOI: 10.3389/fimmu.2022.816761] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
Although CAR T cell therapies have proven to be effective in treating hematopoietic cancers, their abilities to regress solid tumors have been less encouraging. Mechanisms to explain these disparities have focused primarily on differences in cancer cell heterogeneity, barriers to CAR T cell penetration of solid tumors, and immunosuppressive microenvironments. To evaluate the contributions of immunosuppressive tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) on CAR T cell efficacies, we have exploited the ability of a folate-targeted Toll-like receptor 7 agonist (FA-TLR7-1A) to specifically reactivate TAMs and MDSCs from an immunosuppressive to pro-inflammatory phenotype without altering the properties of other immune cells. We report here that FA-TLR7-1A significantly augments standard CAR T cell therapies of 4T1 solid tumors in immune competent mice. We further show that co-administration of the FA-TLR7-1A with the CAR T cell therapy not only repolarizes TAMs and MDSCs from an M2-like anti-inflammatory to M1-like pro-inflammatory phenotype, but also enhances both CAR T cell and endogenous T cell accumulation in solid tumors while concurrently increasing their states of activation. Because analogous myeloid cells in healthy tissues ar not altered by administration of FA-TLR7-1A, no systemic activation of the immune system nor accompanying weight loss is observed. These data argue that immunosuppressive myeloid cells contribute prominently to the failure of CAR T cells to eradicate solid tumors and suggest that methods to reprogram tumor associated myeloid cells to a more inflammatory phenotype could significantly augment the potencies of CAR T cell therapies.
Collapse
Affiliation(s)
- Weichuan Luo
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - John V Napoleon
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Fenghua Zhang
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Yong Gu Lee
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Bingbing Wang
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Karson S Putt
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Philip S Low
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
13
|
Roberto Raúl SG, Damaris IA, Ángel de Jesús JC, Leticia MF. Cry1Ac Protoxin Confers Antitumor Adjuvant Effect in a Triple-Negative Breast Cancer Mouse Model by Improving Tumor Immunity. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234211065154. [PMID: 35002244 PMCID: PMC8738886 DOI: 10.1177/11782234211065154] [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: 11/22/2021] [Accepted: 11/17/2021] [Indexed: 12/07/2022] Open
Abstract
The Cry1Ac protoxin from Bacillus thuringiensis is a systemic
and mucosal adjuvant, able to confer protective immunity in different infection
murine models and induce both Th1 and TCD8+ cytotoxic lymphocyte responses,
which are required to induce antitumor immunity. The Cry1Ac toxin, despite
having not being characterized as an adjuvant, has also proved to be immunogenic
and able to activate macrophages. Here, we investigated the potential antitumor
adjuvant effect conferred by the Cry1Ac protoxin and Cry1Ac toxin in a triple
negative breast cancer (TNBC) murine model. First, we evaluated the ability of
Cry1Ac proteins to improve dendritic cell (DC) activation and cellular response
through intraperitoneal (i.p.) coadministration with the 4T1 cellular lysate.
Mice coadministered with the Cry1Ac protoxin showed an increase in the number
and activation of CD11c+MHCII- and CD11c+MHCII+low in the peritoneal
cavity and an increase in DC activation (CD11c+MHCII+) in the spleen. Cry1Ac
protoxin increased the proliferation of TCD4+ and TCD8+ lymphocytes in the
spleen and mesenteric lymph nodes (MLN), while the Cry1Ac toxin only increased
the proliferation of TCD4+ and TCD8+ in the MLN. Remarkably, when tested in the
in vivo TNBC mouse model, prophylactic immunizations with 4T1 lysates plus the
Cry1Ac protoxin protected mice from developing tumors. The antitumor effect
conferred by the Cry1Ac protoxin also increased specific cytotoxic T cell
responses, and prevented the typical tumor-related decrease of T cells
(TCD3+ and TCD4+) as well the increase of myeloid-derived suppressor cells
(MDSC) in spleen. Also in the tumor microenvironment of mice coadministered
twice with Cry1Ac protoxin immunological improvements were found such as
reductions in immunosupressive populations (T regulatory lymphocytes and MDSC)
along with increases in macrophages upregulating CD86. These results show a
differential antitumor adjuvant capability of Cry1Ac proteins, highlighting the
ability of Cry1Ac protoxin to enhance local and systemic tumor immunity in TNBC.
Finally, using a therapeutic approach, we evaluated the coadministration of
Cry1Ac protoxin with doxorubicin. A significant reduction in tumor volume and
lung metastasis was found, with increased intratumoral levels of tumor necrosis
factor-α and IL-6 with respect to the vehicle group, further supporting its
antitumor applicability.
Collapse
Affiliation(s)
- Servin-Garrido Roberto Raúl
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Ilhuicatzi-Alvarado Damaris
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Jiménez-Chávez Ángel de Jesús
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Moreno-Fierros Leticia
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| |
Collapse
|
14
|
Huang X, Zhang X, Lu M. Recent trends in the development of Toll-like receptor 7/8-targeting therapeutics. Expert Opin Drug Discov 2021; 16:869-880. [PMID: 33678093 DOI: 10.1080/17460441.2021.1898369] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Toll-like receptor (TLR) 7 and TLR8 are functionally localized to endosomes and recognize specific RNA sequences. They play crucial roles in initiating innate and adaptive immune responses. TLR7/8 activation protects the host against invading pathogens and enhances immune responses. In contrast, sustained TLR7/8 signaling leads to immune overreaction. Therefore, agonists or antagonists targeting TLR7/8 signaling are favorable drug candidates for the treatment of immune disorders.Areas covered: Basic knowledge about TLR7 and TLR8 and their signaling pathways are briefly reviewed. Various therapeutic agents have been designed to activate or antagonize TLR7/8 signaling pathways, and their safety and efficacy for the treatment of multiple diseases have been investigated in preclinical animal models and clinical trials. TLR7/8 agonists exhibit potent antiviral activity and regulate anti-tumor immune responses. TLR7 agonists have also been used as adjuvants to improve vaccine immunogenicity and generate greater seroprotection. TLR7/8 antagonists are promising candidates for the treatment of autoimmune and inflammatory diseases.Expert opinion: TLR7/8 pathways are favorable targets for immunological therapies. Future research should concentrate on the optimization of drug safety, efficiency, and specificity. Detailed mechanistic studies will contribute to the development of TLR7/8 immunomodulators and novel therapeutic strategies.
Collapse
Affiliation(s)
- Xuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, Essen, Germany
| |
Collapse
|
15
|
Zhang J, Zhang Y, Qu B, Yang H, Hu S, Dong X. If small molecules immunotherapy comes, can the prime be far behind? Eur J Med Chem 2021; 218:113356. [PMID: 33773287 DOI: 10.1016/j.ejmech.2021.113356] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Anti-cancer immunotherapy, which includes cellular immunotherapy, immune checkpoint inhibitors and cancer vaccines, has transformed the treatment strategies of several malignancies in the past decades. Immune checkpoints blockade (ICB) is the most commonly tested therapy and has the potential to induce a durable immune response in different types of cancers. However, all approved immune checkpoint inhibitors (ICIs) are monoclonal antibodies (mAbs), which are fraught with disadvantages including lack of oral bioavailability, prolonged tissue retention and poor membrane permeability. Therefore, the research focus has shifted to developing small molecule inhibitors to obviate the limitations of mAbs. Given the complexity of the tumor micro-environment (TME), the combination of ICIs with various small molecule agonists/inhibitors are currently being tested in clinical trials to improve treatment outcomes and prevent tumor recurrence. In this review, we have summarized the mechanisms and therapeutic potential of several molecular targets, along with the current status of small molecule inhibitors.
Collapse
Affiliation(s)
- Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Bingxue Qu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Haiyan Yang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), PR China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, PR China
| | - Shengquan Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310058, PR China; Cancer Center of Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
16
|
Hingorani DV, Camargo MF, Quraishi MA, Adams SR, Advani SJ. Tumor Activated Cell Penetrating Peptides to Selectively Deliver Immune Modulatory Drugs. Pharmaceutics 2021; 13:pharmaceutics13030365. [PMID: 33801967 PMCID: PMC8000974 DOI: 10.3390/pharmaceutics13030365] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/01/2021] [Accepted: 03/08/2021] [Indexed: 01/15/2023] Open
Abstract
Recent advances in immunotherapy have revolutionized cancer therapy. Immunotherapies can engage the adaptive and innate arms of the immune system. Therapeutics targeting immune checkpoint inhibitors (i.e., CTLA-4; PD-1, and PD-L1) have shown efficacy for subsets of cancer patients by unleashing an adaptive antitumor immune response. Alternatively, small molecule immune modulators of the innate immune system such as toll-like receptor (TLR) agonists are being developed for cancer therapy. TLRs function as pattern recognition receptors to microbial products and are also involved in carcinogenesis. Reisquimod is a TLR 7/8 agonist that has antitumor efficacy. However, systemic delivery free resiquimod has proven to be challenging due to toxicity of nonspecific TLR 7/8 activation. Therefore, we developed a targeted peptide-drug conjugate strategy for systemic delivery of resiquimod. We designed an activatable cell penetrating peptide to deliver resiquimod specifically to the tumor tissue while avoiding normal tissues. The activatable cell penetrating peptide (ACPP) scaffold undergoes enzymatic cleavage by matrix metalloproteinases 2/9 in the extracellular matrix followed by intracellular lysosomal cathepsin B mediated release of the free resiquimod. Importantly, when conjugated to ACPP; the tumor tissue concentration of resiquimod was more than 1000-fold greater than that of surrounding non-cancerous tissue. Moreover, systemic ACPP-resiquimod delivery produced comparable therapeutic efficacy to localized free resiquimod in syngeneic murine tumors. These results highlight a precision peptide-drug conjugate delivery.
Collapse
Affiliation(s)
- Dina V. Hingorani
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.V.H.); (M.F.C.); (M.A.Q.)
| | - Maria F. Camargo
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.V.H.); (M.F.C.); (M.A.Q.)
| | - Maryam A. Quraishi
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.V.H.); (M.F.C.); (M.A.Q.)
| | - Stephen R. Adams
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA;
| | - Sunil J. Advani
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.V.H.); (M.F.C.); (M.A.Q.)
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, USA
- Correspondence: ; Tel.: +85-8822-6046; Fax: +85-8822-5568
| |
Collapse
|
17
|
Díaz-Carballo D, Saka S, Acikelli AH, Homp E, Erwes J, Demmig R, Klein J, Schröer K, Malak S, D'Souza F, Noa-Bolaño A, Menze S, Pano E, Andrioff S, Teipel M, Dammann P, Klein D, Nasreen A, Tannapfel A, Grandi N, Tramontano E, Ochsenfarth C, Strumberg D. Enhanced antitumoral activity of TLR7 agonists via activation of human endogenous retroviruses by HDAC inhibitors. Commun Biol 2021; 4:276. [PMID: 33658617 PMCID: PMC7930250 DOI: 10.1038/s42003-021-01800-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
In this work, we are reporting that "Shock and Kill", a therapeutic approach designed to eliminate latent HIV from cell reservoirs, is extrapolatable to cancer therapy. This is based on the observation that malignant cells express a spectrum of human endogenous retroviral elements (HERVs) which can be transcriptionally boosted by HDAC inhibitors. The endoretroviral gene HERV-V2 codes for an envelope protein, which resembles syncytins. It is significantly overexpressed upon exposure to HDAC inhibitors and can be effectively targeted by simultaneous application of TLR7/8 agonists, triggering intrinsic apoptosis. We demonstrated that this synergistic cytotoxic effect was accompanied by the functional disruption of the TLR7/8-NFκB, Akt/PKB, and Ras-MEK-ERK signalling pathways. CRISPR/Cas9 ablation of TLR7 and HERV-V1/V2 curtailed apoptosis significantly, proving the pivotal role of these elements in driving cell death. The effectiveness of this new approach was confirmed in ovarian tumour xenograft studies, revealing a promising avenue for future cancer therapies.
Collapse
Affiliation(s)
- David Díaz-Carballo
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany.
| | - Sahitya Saka
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Ali H Acikelli
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Ekaterina Homp
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Julia Erwes
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Rebecca Demmig
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Jacqueline Klein
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Katrin Schröer
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Sascha Malak
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Flevy D'Souza
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Adrien Noa-Bolaño
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Saskia Menze
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Emilio Pano
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Swetlana Andrioff
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Marc Teipel
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Philip Dammann
- Central Animal Laboratory, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute of Cell Biology, Cancer Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Amber Nasreen
- Visceral Surgery Department, Marien Hospital Herne, Ruhr University Bochum Medical School, Herne, Germany
| | | | - Nicole Grandi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Crista Ochsenfarth
- Department of Anesthesia, Intensive Care, Pain and Palliative Medicine, Marien Hospital Herne, Ruhr-University Bochum Medical School, Herne, Germany
| | - Dirk Strumberg
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| |
Collapse
|
18
|
Hotz C, Bourquin C. Systemic cancer immunotherapy with Toll-like receptor 7 agonists: Timing is everything. Oncoimmunology 2021; 1:227-228. [PMID: 22720251 PMCID: PMC3376978 DOI: 10.4161/onci.1.2.18169] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Toll-like receptor (TLR) 7 agonists represent a promising strategy for the immunotherapy of cancer. We have recently investigated the influence of TLR tolerance on the efficacy of systemic tumor treatment with TLR7 ligands. We propose that considering the kinetics of receptor sensitivity highly improves the outcome of cancer immunotherapy.
Collapse
Affiliation(s)
- Christian Hotz
- Center for Integrated Protein Science Munich (CIPSM); Division of Clinical Pharmacology; Department of Internal Medicine; Ludwig-Maximilian University of Munich; Munich, Germany ; Department of Medicine; University of Fribourg; Fribourg, Switzerland
| | | |
Collapse
|
19
|
Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2021; 1:699-716. [PMID: 22934262 PMCID: PMC3429574 DOI: 10.4161/onci.20696] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are prototypic pattern recognition receptors (PRRs) best known for their ability to activate the innate immune system in response to conserved microbial components such as lipopolysaccharide and double-stranded RNA. Accumulating evidence indicates that the function of TLRs is not restricted to the elicitation of innate immune responses against invading pathogens. TLRs have indeed been shown to participate in tissue repair and injury-induced regeneration as well as in adaptive immune responses against cancer. In particular, TLR4 signaling appears to be required for the efficient processing and cross-presentation of cell-associated tumor antigens by dendritic cells, which de facto underlie optimal therapeutic responses to some anticancer drugs. Thus, TLRs constitute prominent therapeutic targets for the activation/intensification of anticancer immune responses. In line with this notion, long-used preparations such as the Coley toxin (a mixture of killed Streptococcus pyogenes and Serratia marcescens bacteria) and the bacillus Calmette-Guérin (BCG, an attenuated strain of Mycobacterium bovis originally developed as a vaccine against tuberculosis), both of which have been associated with consistent anticancer responses, potently activate TLR2 and TLR4 signaling. Today, besides BCG, only one TLR agonist is FDA-approved for therapeutic use in cancer patients: imiquimod. In this Trial Watch, we will briefly present the role of TLRs in innate and cognate immunity and discuss the progress of clinical studies evaluating the safety and efficacy of experimental TLR agonists as immunostimulatory agents for oncological indications.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang Y, Yang H, Li H, Zhao S, Zeng Y, Zhang P, Lin X, Sun X, Wang L, Fu G, Gao Y, Wang P, Gao D. Development of a novel TLR8 agonist for cancer immunotherapy. MOLECULAR BIOMEDICINE 2020; 1:6. [PMID: 35006413 PMCID: PMC8607422 DOI: 10.1186/s43556-020-00007-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/14/2020] [Indexed: 01/12/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of proteins that recognize pathogen associated molecular patterns (PAMPs). Their primary function is to activate innate immune responses while also involved in facilitating adaptive immune responses. Different TLRs exert distinct functions by activating varied immune cascades. Several TLRs are being pursued as cancer drug targets. We discovered a novel, highly potent and selective small molecule TLR8 agonist DN052. DN052 exhibited strong in vitro cellular activity with EC50 at 6.7 nM and was highly selective for TLR8 over other TLRs including TLR4, 7 and 9. DN052 displayed excellent in vitro ADMET and in vivo PK profiles. DN052 potently inhibited tumor growth as a single agent. Moreover, combination of DN052 with the immune checkpoint inhibitor, selected targeted therapeutics or chemotherapeutic drugs further enhanced efficacy of single agents. Mechanistically, treatment with DN052 resulted in strong induction of pro-inflammatory cytokines in ex vivo human PBMC assay and in vivo monkey study. GLP toxicity studies in rats and monkeys demonstrated favorable safety profile. This led to the advancement of DN052 into phase 1 clinical trials.
Collapse
Affiliation(s)
- Yuxun Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China.
| | - Heping Yang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Huanping Li
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Shuda Zhao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Yikun Zeng
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Panpan Zhang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Xiaoqin Lin
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Xiaoxiang Sun
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Longsheng Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Guangliang Fu
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Yaqiao Gao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Pei Wang
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| | - Daxin Gao
- Shanghai Denovo Pharmatech Co., Ltd., 576 Libing Road, Shanghai Zhangjiang High-Tech Park, Pudong New District, Shanghai, 201203, China
| |
Collapse
|
21
|
Yang Y, Yang Y, Yang J, Zhao X, Wei X. Tumor Microenvironment in Ovarian Cancer: Function and Therapeutic Strategy. Front Cell Dev Biol 2020; 8:758. [PMID: 32850861 PMCID: PMC7431690 DOI: 10.3389/fcell.2020.00758] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is one of the leading causes of death in patients with gynecological malignancy. Despite optimal cytoreductive surgery and platinum-based chemotherapy, ovarian cancer disseminates and relapses frequently, with poor prognosis. Hence, it is urgent to find new targeted therapies for ovarian cancer. Recently, the tumor microenvironment has been reported to play a vital role in the tumorigenesis of ovarian cancer, especially with discoveries from genome-, transcriptome- and proteome-wide studies; thus tumor microenvironment may present potential therapeutic target for ovarian cancer. Here, we review the interactions between the tumor microenvironment and ovarian cancer and various therapies targeting the tumor environment.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Zhang F, Ayaub EA, Wang B, Puchulu‐Campanella E, Li Y, Hettiarachchi SU, Lindeman SD, Luo Q, Rout S, Srinivasarao M, Cox A, Tsoyi K, Nickerson‐Nutter C, Rosas IO, Low PS. Reprogramming of profibrotic macrophages for treatment of bleomycin-induced pulmonary fibrosis. EMBO Mol Med 2020; 12:e12034. [PMID: 32597014 PMCID: PMC7411553 DOI: 10.15252/emmm.202012034] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022] Open
Abstract
Fibrotic diseases cause organ failure that lead to ~45% of all deaths in the United States. Activated macrophages stimulate fibrosis by secreting cytokines that induce fibroblasts to synthesize collagen and extracellular matrix proteins. Although suppression of macrophage-derived cytokine production can halt progression of fibrosis, therapeutic agents that prevent release of these cytokines (e.g., TLR7 agonists) have proven too toxic to administer systemically. Based on the expression of folate receptor β solely on activated myeloid cells, we have created a folate-targeted TLR7 agonist (FA-TLR7-54) that selectively accumulates in profibrotic macrophages and suppresses fibrosis-inducing cytokine production. We demonstrate that FA-TLR7-54 reprograms M2-like fibrosis-inducing macrophages into fibrosis-suppressing macrophages, resulting in dramatic declines in profibrotic cytokine release, hydroxyproline biosynthesis, and collagen deposition, with concomitant increases in alveolar airspaces. Although nontargeted TLR7-54 is lethal at fibrosis-suppressing doses, FA-TLR7-54 halts fibrosis without evidence of toxicity. Taken together, FA-TLR7-54 is shown to constitute a novel and potent approach for treating fibrosis without causing dose-limiting systemic toxicities.
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Ehab A Ayaub
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Bingbing Wang
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | | | - Yen‐Hsing Li
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Suraj U Hettiarachchi
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Spencer D Lindeman
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Qian Luo
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Sasmita Rout
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Abigail Cox
- Department of Comparative PathobiologyPurdue College of Veterinary MedicineWest LafayetteINUSA
| | - Konstantin Tsoyi
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | | | - Ivan O Rosas
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Philip S Low
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
23
|
Irvine DJ, Aung A, Silva M. Controlling timing and location in vaccines. Adv Drug Deliv Rev 2020; 158:91-115. [PMID: 32598970 PMCID: PMC7318960 DOI: 10.1016/j.addr.2020.06.019] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.
Collapse
Affiliation(s)
- Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 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.
| | - Aereas Aung
- 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
| | - Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
24
|
Vohidov F, Milling LE, Chen Q, Zhang W, Bhagchandani S, Nguyen HVT, Irvine DJ, Johnson JA. ABC triblock bottlebrush copolymer-based injectable hydrogels: design, synthesis, and application to expanding the therapeutic index of cancer immunochemotherapy. Chem Sci 2020; 11:5974-5986. [PMID: 34094088 PMCID: PMC8159417 DOI: 10.1039/d0sc02611e] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/16/2020] [Indexed: 12/29/2022] Open
Abstract
Bottlebrush copolymers are a versatile class of macromolecular architectures with broad applications in the fields of drug delivery, self-assembly, and polymer networks. Here, the modular nature of graft-through ring-opening metathesis polymerization (ROMP) is exploited to synthesize "ABC" triblock bottlebrush copolymers (TBCs) from polylactic acid (PLA), polyethylene glycol (PEG), and poly(N-isopropylacrylamide) (PNIPAM) macromonomers. Due to the hydrophobicity of their PLA domains, these TBCs self-assemble in aqueous media at room temperature to yield uniform ∼100 nm micelles that can encapsulate a wide range of therapeutic agents. Heating these micellar solutions above the lower critical solution temperature (LCST) of PNIPAM (∼32 °C) induces the rapid formation of multi-compartment hydrogels with PLA and PNIPAM domains acting as physical crosslinks. Following the synthesis and characterization of these materials in vitro, TBC micelles loaded with various biologically active small molecules were investigated as injectable hydrogels for sustained drug release in vivo. Specifically, intratumoral administration of TBCs containing paclitaxel and resiquimod-the latter a potent Toll-like receptor (TLR) 7/8 agonist-into mice bearing subcutaneous CT26 tumors resulted in a significantly enhanced therapeutic index compared to the administration of these two drugs alone. This effect is attributed to the TBC hydrogel maintaining a high local drug concentration, thus reducing systemic immune activation and local inflammation. Collectively, this work represents, to our knowledge, the first example of thermally-responsive TBCs designed for multi-compartment hydrogel formation, establishing these materials as versatile scaffolds for self-assembly and drug delivery.
Collapse
Affiliation(s)
- Farrukh Vohidov
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
| | - Lauren E Milling
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology Cambridge Massachusetts 02139 USA
| | - Qixian Chen
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
| | - Wenxu Zhang
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
| | - Sachin Bhagchandani
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
| | - Hung V-T Nguyen
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology Cambridge Massachusetts 02139 USA
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology Massachusetts 02139 USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology Cambridge Massachusetts 02139 USA
| |
Collapse
|
25
|
Bourquin C, Pommier A, Hotz C. Harnessing the immune system to fight cancer with Toll-like receptor and RIG-I-like receptor agonists. Pharmacol Res 2020; 154:104192. [PMID: 30836160 DOI: 10.1016/j.phrs.2019.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/23/2019] [Accepted: 03/01/2019] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has come of age with the advent of immune checkpoint inhibitors. In this article we review how agonists for receptors of the innate immune system, the Toll-like receptors and the RIG-I-like receptors, impact anticancer immune responses. Treatment with these agonists enhances the activity of anticancer effector cells, such as cytotoxic T cells and NK cells, and at the same time blocks the activity of immunosuppressive cell types such as regulatory T cells and myeloid-derived suppressor cells. These compounds also impact the recruitment of immune cells to the tumor. The phenomena of pattern-recognition receptor tolerance and reprogramming and their implications for immunotherapy are discussed. Finally, novel delivery systems that target the immune-stimulating drugs to the tumor or the tumor-draining lymph nodes to enhance their efficacy and safety are presented.
Collapse
Affiliation(s)
- Carole Bourquin
- Chair of Pharmacology, Faculty of Science, University of Fribourg, 1700, Fribourg, Switzerland; Institute of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211, Geneva, Switzerland; Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland.
| | - Aurélien Pommier
- Institute of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211, Geneva, Switzerland
| | - Christian Hotz
- Chair of Pharmacology, Faculty of Science, University of Fribourg, 1700, Fribourg, Switzerland
| |
Collapse
|
26
|
Patinote C, Karroum NB, Moarbess G, Cirnat N, Kassab I, Bonnet PA, Deleuze-Masquéfa C. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem 2020; 193:112238. [PMID: 32203790 PMCID: PMC7173040 DOI: 10.1016/j.ejmech.2020.112238] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
The discovery of the TLRs family and more precisely its functions opened a variety of gates to modulate immunological host responses. TLRs 7/8 are located in the endosomal compartment and activate a specific signaling pathway in a MyD88-dependant manner. According to their involvement into various autoimmune, inflammatory and malignant diseases, researchers have designed diverse TLRs 7/8 ligands able to boost or block the inherent signal transduction. These modulators are often small synthetic compounds and most act as agonists and to a much lesser extent as antagonists. Some of them have reached preclinical and clinical trials, and only one has been approved by the FDA and EMA, imiquimod. The key to the success of these modulators probably lies in their combination with other therapies as recently demonstrated. We gather in this review more than 360 scientific publications, reviews and patents, relating the extensive work carried out by researchers on the design of TLRs 7/8 modulators, which are classified firstly by their biological activities (agonist or antagonist) and then by their chemical structures, which total syntheses are not discussed here. This review also reports about 90 clinical cases, thereby showing the biological interest of these modulators in multiple pathologies.
Collapse
Affiliation(s)
- Cindy Patinote
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Nour Bou Karroum
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Natalina Cirnat
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | | | | |
Collapse
|
27
|
Toll-Like Receptors Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:81-97. [PMID: 32030686 DOI: 10.1007/978-3-030-35582-1_5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The involvement of inflammation in cancer progression is well-established. The immune system can play both tumor-promoting and -suppressive roles, and efforts to harness the immune system to help fight tumor growth are at the forefront of research. Of particular importance is the inflammatory profile at the site of the tumor, with respect to both the leukocyte population numbers, the phenotype of these cells, as well as the contribution of the tumor cells themselves. In this regard, the pro-inflammatory effects of pattern recognition receptor expression and activation in the tumor microenvironment have emerged as a relevant issue both for therapy and to understand tumor development.Pattern recognition receptors (PRRs) were originally recognized as components of immune cells, particularly innate immune cells, as detectors of pathogens. PRR signaling in immune cells activates them, inducing robust antimicrobial responses. In particular, toll-like receptors (TLRs) constitute a family of membrane-bound PRRs which can recognize pathogen-associated molecular patterns (PAMPs) carried by bacteria, virus, and fungi. In addition, PRRs can recognize products generated by stressed cells or damaged tissues, namely damage-associated molecular patterns or DAMPS. Taking into account the role of the immune system in fighting tumors together with the presence of immune cells in the microenvironment of different types of tumors, strategies to activate immune cells via PRR ligands have been envisioned as an anticancer therapeutic approach.In the last decades, it has been determined that PRRs are present and functional on nonimmune cells and that their activation in these cells contributes to the inflammation in the tumor microenvironment. Both tumor-promoting and antitumor effects have been observed when tumor cell PRRs are activated. This argues against nonspecific activation of PRR ligands in the tumor microenvironment as a therapeutic approach. Therefore, the use of PRR ligands for anticancer therapy might benefit from strategies that specifically deliver these ligands to immune cells, thus avoiding tumor cells in some settings. This review focuses on these aspects of TLR signaling in the tumor microenvironment.
Collapse
|
28
|
Vascotto F, Petschenka J, Walzer KC, Vormehr M, Brkic M, Strobl S, Rösemann R, Diken M, Kreiter S, Türeci Ö, Sahin U. Intravenous delivery of the toll-like receptor 7 agonist SC1 confers tumor control by inducing a CD8+ T cell response. Oncoimmunology 2019; 8:1601480. [PMID: 31143525 DOI: 10.1080/2162402x.2019.1601480] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 10/27/2022] Open
Abstract
TLR7 agonists are considered promising drugs for cancer therapy. The currently available compounds are not well tolerated when administered intravenously and therefore are restricted to disease settings amenable for topical application. Here we present the preclinical characterization of SC1, a novel synthetic agonist with exquisite specificity for TLR7. We found that intravenously administered SC1 mediates systemic release of type I interferon, but not of proinflammatory cytokines such as TNFα and IL6, and results in activation of circulating immune cells. Tumors of SC1-treated mice have brisk immune cell infiltrates and are polarized towards a Th1 type signature. Intratumoral CD8+ T cells and CD11b+ conventional dendritic cells (cDCs) are significantly increased, plasmacytoid dendritic cells (pDCs) are strongly activated and macrophages are M1 phenotype polarized, whereas myeloid-derived suppressor cells (MDSCs) are decreased. We further show that treatment of mice with SC1 profoundly inhibits the growth of established syngeneic tumors and results in significantly prolonged survival. Mice, which are tumor-free after SC1 treatment are protected from subsequent tumor rechallenge. The antitumor effect of SC1 depends on antigen-specific CD8+ T cells, which we found to be strongly enriched in the tumors of SC1-treated mice. In conclusion, this study shows that systemically administered SC1 mobilizes innate and adaptive immunity and is highly potent as single agent in mice and thereby provides a rationale for clinical testing of this compound.
Collapse
Affiliation(s)
- Fulvia Vascotto
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Jutta Petschenka
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Kerstin C Walzer
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Mathias Vormehr
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Magdalena Brkic
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany
| | | | | | - Mustafa Diken
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Sebastian Kreiter
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Özlem Türeci
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Ugur Sahin
- TRON - Translational Oncology, University Medical Center of Johannes Gutenberg University gGmbH, Mainz, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center at the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
29
|
Moradi-Marjaneh R, Hassanian SM, Hasanzadeh M, Rezayi M, Maftouh M, Mehramiz M, Ferns GA, Khazaei M, Avan A. Therapeutic potential of toll-like receptors in treatment of gynecological cancers. IUBMB Life 2019; 71:549-564. [PMID: 30729633 DOI: 10.1002/iub.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/28/2022]
Abstract
Toll-like receptors (TLRs) play an important role in the innate and adaptive immune system. They are expressed in various regions of the female reproductive tract, and their regulation may be involved in the pathogenesis of gynecological lesions. There is growing evidence that ligands for several TLRs are potentially anticancer agents, some of which have already been approved by the FDA, and these compounds are now undergoing clinical evaluation. There is a rationale for using these ligands as adjuvants in the treatment or prevention of gynecological cancer. Some TLR agonists that are of potential interest in the treatment of gynecological lesions include imiquimod, motolimod, cervarix, and CpG-oligodeoxynucleotides (ODNs). In this review, we outline the different functions of TLRs in gynecological cancer with particular emphasis on the value of TLR agonists as a potential therapeutic target in the treatment of gynecological cancer. © 2019 IUBMB Life, 71(5):549-564, 2019.
Collapse
Affiliation(s)
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezayi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrane Mehramiz
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
The effect of intranasally administered TLR3 agonist larifan on metabolic profile of microglial cells in rat with C6 glioma. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.06.110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
31
|
Qiu SQ, Waaijer SJH, Zwager MC, de Vries EGE, van der Vegt B, Schröder CP. Tumor-associated macrophages in breast cancer: Innocent bystander or important player? Cancer Treat Rev 2018; 70:178-189. [PMID: 30227299 DOI: 10.1016/j.ctrv.2018.08.010] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 02/05/2023]
Abstract
Tumor-associated macrophages (TAMs) are important tumor-promoting cells in the breast tumor microenvironment. Preclinically TAMs stimulate breast tumor progression, including tumor cell growth, invasion and metastasis. TAMs also induce resistance to multiple types of treatment in breast cancer models. The underlying mechanisms include: induction and maintenance of tumor-promoting phenotype in TAMs, inhibition of CD8+ T cell function, degradation of extracellular matrix, stimulation of angiogenesis and inhibition of phagocytosis. Several studies reported that high TAM infiltration of breast tumors is correlated with a worse patient prognosis. Based on these findings, macrophage-targeted treatment strategies have been developed and are currently being evaluated in clinical breast cancer trials. These strategies include: inhibition of macrophage recruitment, repolarization of TAMs to an antitumor phenotype, and enhancement of macrophage-mediated tumor cell killing or phagocytosis. This review summarizes the functional aspects of TAMs and the rationale and current evidence for TAMs as a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Si-Qi Qiu
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands; The Breast Center, Cancer Hospital of Shantou University Medical College, Raoping 7, 515041 Shantou, China
| | - Stijn J H Waaijer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Mieke C Zwager
- Department of Pathology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
32
|
Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int Immunopharmacol 2018; 59:391-412. [PMID: 29730580 PMCID: PMC7106078 DOI: 10.1016/j.intimp.2018.03.002] [Citation(s) in RCA: 417] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
The immune system is a very diverse system of the host that evolved during evolution to cope with various pathogens present in the vicinity of environmental surroundings inhabited by multicellular organisms ranging from achordates to chordates (including humans). For example, cells of immune system express various pattern recognition receptors (PRRs) that detect danger via recognizing specific pathogen-associated molecular patterns (PAMPs) and mount a specific immune response. Toll-like receptors (TLRs) are one of these PRRs expressed by various immune cells. However, they were first discovered in the Drosophila melanogaster (common fruit fly) as genes/proteins important in embryonic development and dorso-ventral body patterning/polarity. Till date, 13 different types of TLRs (TLR1-TLR13) have been discovered and described in mammals since the first discovery of TLR4 in humans in late 1997. This discovery of TLR4 in humans revolutionized the field of innate immunity and thus the immunology and host-pathogen interaction. Since then TLRs are found to be expressed on various immune cells and have been targeted for therapeutic drug development for various infectious and inflammatory diseases including cancer. Even, Single nucleotide polymorphisms (SNPs) among various TLR genes have been identified among the different human population and their association with susceptibility/resistance to certain infections and other inflammatory diseases. Thus, in the present review the current and future importance of TLRs in immunity, their pattern of expression among various immune cells along with TLR based therapeutic approach is reviewed. TLRs are first described PRRs that revolutionized the biology of host-pathogen interaction and immune response The discovery of different TLRs in humans proved milestone in the field of innate immunity and inflammation The pattern of expression of all the TLRs expressed by human immune cells An association of various TLR SNPs with different inflammatory diseases Currently available drugs or vaccines based on TLRs and their future in drug targeting along with the role in reproduction, and regeneration
Collapse
|
33
|
Stromal cells in breast cancer as a potential therapeutic target. Oncotarget 2018; 9:23761-23779. [PMID: 29805773 PMCID: PMC5955086 DOI: 10.18632/oncotarget.25245] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Breast cancer in the United States is the second most commonly diagnosed cancer in women. About 1 in 8 women will develop invasive breast cancer over the course of her lifetime and breast cancer remains the second leading cause of cancer-related death. In pursuit of novel therapeutic strategies, researchers have examined the tumor microenvironment as a potential anti-cancer target. In addition to neoplastic cells, the tumor microenvironment is composed of several critical normal cell types, including fibroblasts, vascular and lymph endothelial cells, osteoclasts, adipocytes, and immune cells. These cells have important roles in healthy tissue stasis, which frequently are altered in tumors. Indeed, tumor-associated stromal cells often contribute to tumorigenesis, tumor progression, and metastasis. Consequently, these host cells may serve as a possible target in anti-tumor and anti-metastatic therapeutic strategies. Targeting the tumor associated host cells offers the benefit that such cells do not mutate and develop resistance in response to treatment, a major cause of failure in cancer therapeutics targeting neoplastic cells. This review discusses the role of host cells in the tumor microenvironment during tumorigenesis, progression, and metastasis, and provides an overview of recent developments in targeting these cell populations to enhance cancer therapy efficacy.
Collapse
|
34
|
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: 0.5] [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
|
35
|
Toogood PL. Small molecule immuno-oncology therapeutic agents. Bioorg Med Chem Lett 2017; 28:319-329. [PMID: 29326017 DOI: 10.1016/j.bmcl.2017.12.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022]
Abstract
Treatment of cancer by activation of an antitumor immune response is now a widely practiced and well-accepted approach to therapy. However, despite dramatic responses in some patients, the high proportion of unresponsive patients points to a considerable unmet medical need. Although antibody therapies have led the way, small molecule immuno-oncology agents are close behind. This perspective provides an overview of some of the many small molecule approaches being explored. It encompasses small molecule modulators of validated targets such as programed cell death 1 (PD-1) as well as novel approaches still to be proven clinically.
Collapse
Affiliation(s)
- Peter L Toogood
- Lycera Corp., 1350 Highland Drive, Ann Arbor, MI, United States.
| |
Collapse
|
36
|
Chi H, Li C, Zhao FS, Zhang L, Ng TB, Jin G, Sha O. Anti-tumor Activity of Toll-Like Receptor 7 Agonists. Front Pharmacol 2017; 8:304. [PMID: 28620298 PMCID: PMC5450331 DOI: 10.3389/fphar.2017.00304] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/10/2017] [Indexed: 01/04/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that play a bridging role in innate immunity and adaptive immunity. The activated TLRs not only induce inflammatory responses, but also elicit the development of antigen specific immunity. TLR7, a member of TLR family, is an intracellular receptor expressed on the membrane of endosomes. TLR7 can be triggered not only by ssRNA during viral infections, but also by immune modifiers that share a similar structure to nucleosides. Its powerful immune stimulatory action can be potentially used in the anti-tumor therapy. This article reviewed the anti-tumor activity and mechanism of TLR7 agonists that are frequently applied in preclinical and clinical investigations, and mainly focused on small synthetic molecules, including imiquimod, resiquimod, gardiquimod, and 852A, etc.
Collapse
Affiliation(s)
- Huju Chi
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Chunman Li
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Flora Sha Zhao
- School of Life Sciences, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Li Zhang
- Department of Physiology and Neurology, University of ConnecticutStorrs, CT, United States
| | - Tzi Bun Ng
- Departmet of Biochemistry, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Guangyi Jin
- Department of Pharmacy, Shenzhen University Health Science CentreShenzhen, China
| | - Ou Sha
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| |
Collapse
|
37
|
Fehri E, Ennaifer E, Ardhaoui M, Ouerhani K, Laassili T, Bel Haj Rhouma R, Guizani I, Boubaker S. Expression of Toll-like receptor 9 increases with progression of cervical neoplasia in Tunisian women--a comparative analysis of condyloma, cervical intraepithelial neoplasia and invasive carcinoma. Asian Pac J Cancer Prev 2017; 15:6145-50. [PMID: 25124588 DOI: 10.7314/apjcp.2014.15.15.6145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Toll-like receptors (TLRs) are expressed in immune and tumor cells and recognize pathogen-associated molecular patterns. Cervical cancer (CC) is directly linked to a persistent infection with high risk human papillomaviruses (HR-HPVs) and could be associated with alteration of TLRs expression. TLR9 plays a key role in the recognition of DNA viruses and better understanding of this signaling pathway in CC could lead to the development of novel immunotherapeutic approaches. The present study was undertaken to determine the level of TLR9 expression in cervical neoplasias from Tunisian women with 53 formalin-fixed and paraffin-embedded specimens, including 22 samples of invasive cervical carcinoma (ICC), 18 of cervical intraepithelial neoplasia (CIN), 7 of condyloma and 6 normal cervical tissues as control cases. Quantification of TLR9 expression was based on scoring four degrees of extent and intensity of immunostaining in squamous epithelial cells. TLR9 expression gradually increased from CIN1 (80% weak intensity) to CIN2 (83.3% moderate), CIN3 (57.1% strong) and ICC (100% very strong). It was absent in normal cervical tissue and weak in 71.4% of condyloma. The mean scores of TLR9 expression were compared using the Kruskall-Wallis test and there was a statistical significance between normal tissue and condyloma as well as between condyloma, CINs and ICC. These results suggest that TLR9 may play a role in progression of cervical neoplasia in Tunisian patients and could represent a useful biomarker for malignant transformation of cervical squamous cells.
Collapse
Affiliation(s)
- Emna Fehri
- HPV Unit Research. Laboratory of Molecular Epidemiology and Experimental Pathology Applied to Infectious Diseases, le Belvedere, Tunis, Tunisia E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Imprime PGG-Mediated Anti-Cancer Immune Activation Requires Immune Complex Formation. PLoS One 2016; 11:e0165909. [PMID: 27812183 PMCID: PMC5094785 DOI: 10.1371/journal.pone.0165909] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/19/2016] [Indexed: 11/19/2022] Open
Abstract
Imprime PGG (Imprime), an intravenously-administered, soluble β-glucan, has shown compelling efficacy in multiple phase 2 clinical trials with tumor targeting or anti-angiogenic antibodies. Mechanistically, Imprime acts as pathogen-associated molecular pattern (PAMP) directly activating innate immune effector cells, triggering a coordinated anti-cancer immune response. Herein, using whole blood from healthy human subjects, we show that Imprime-induced anti-cancer functionality is dependent on immune complex formation with naturally-occurring, anti-β glucan antibodies (ABA). The formation of Imprime-ABA complexes activates complement, primarily via the classical complement pathway, and is opsonized by iC3b. Immune complex binding depends upon Complement Receptor 3 and Fcg Receptor IIa, eliciting phenotypic activation of, and enhanced chemokine production by, neutrophils and monocytes, enabling these effector cells to kill antibody-opsonized tumor cells via the generation of reactive oxygen species and antibody-dependent cellular phagocytosis. Importantly, these innate immune cell changes were not evident in subjects with low ABA levels but could be rescued with exogenous ABA supplementation. Together, these data indicate that pre-existing ABA are essential for Imprime-mediated anti-cancer immune activation and suggest that pre-treatment ABA levels may provide a plausible patient selection biomarker to delineate patients most likely to benefit from Imprime-based therapy.
Collapse
|
39
|
Abstract
It has been established that a high degree of tumor-infiltrating T cells is associated with ovarian cancer prognosis. We hypothesized that tumors display an immune-related program of transcription that can act in a stimulatory or a regulatory manner. We analyzed transcriptome-wide gene expression data from 503 ovarian tumors from the Cancer Genome Atlas to identify genes that show differential prognoses when stratified by CD3 expression. Genes with immunological functions and tumor antigen genes were selected for analysis. We repeated our analysis in an independent validation study. Five genes showed stimulatory/regulatory patterns at a high level of confidence (Bonferroni p < 0.05). Three of these (MAGEA8, MPL, AMHR2) were validated and one (WT1) could not be evaluated. These patterns show specific prognostic effect only in conjunction with CD3 expression. When patients express multiple transcripts in poor prognosis directions, there is a dose response: increasingly regulatory type tumors are associated with higher stage, lower treatment response and shorter overall survival and progression free survival. The high-confidence set of transcripts (MAGEA8, MPL, AMHR2, WT1) and selected low-confidence hits (EPOR, TLR7) alone or in combination represent candidate prognosis markers for further investigation.
Collapse
|
40
|
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: 14] [Impact Index Per Article: 1.6] [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
|
41
|
Kobold S, Wiedemann G, Rothenfußer S, Endres S. Modes of action of TLR7 agonists in cancer therapy. Immunotherapy 2015; 6:1085-95. [PMID: 25428647 DOI: 10.2217/imt.14.75] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
From the numerous Toll-like receptor agonists, only TLR7 agonists have been approved for cancer treatment, although they are current restricted to topical application. The main target cells of TLR7 agonists are plasmacytoid dendritic cells, producing IFN-α and thus acting on other immune cells. Thereby dendritic cells acquire enhanced costimulatory and antigen-presenting capacity, priming an adaptive immune response. Besides NK cells, antigen-specific T cells are the main terminal effectors of TLR7 agonists in tumor therapy. This qualifies TLR7 agonists as vaccine adjuvants, which is currently being tested in clinical trials. However, the systemic application of TLR7 agonists shows insufficient efficacy, most likely owing to toxicity-limited dosing. The use of TLR7 agonists in combinational therapy holds the promise of synergistic activity and lower required doses.
Collapse
Affiliation(s)
- Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Department of Internal Medicine IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | |
Collapse
|
42
|
Husseinzadeh N, Davenport SM. Role of Toll-like receptors in cervical, endometrial and ovarian cancers: A review. Gynecol Oncol 2014; 135:359-63. [DOI: 10.1016/j.ygyno.2014.08.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/06/2014] [Accepted: 08/09/2014] [Indexed: 12/30/2022]
|
43
|
Muccioli M, Benencia F. Toll-like Receptors in Ovarian Cancer as Targets for Immunotherapies. Front Immunol 2014; 5:341. [PMID: 25101083 PMCID: PMC4105689 DOI: 10.3389/fimmu.2014.00341] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/04/2014] [Indexed: 01/21/2023] Open
Abstract
In the last decade, it has become apparent that toll-like receptor (TLR) signaling can play an important role in ovarian cancer (OC) progression. Interestingly, TLR activation in immune cells can help activate an anti-tumor response, while TLR signaling in tumor cells themselves is often associated with cancer-promoting inflammation. For example, it has been shown that TLR activation in dendritic cells can result in more effective antigen presentation to T cells, thereby favoring tumor eradication. However, aberrant TLR expression in OC cells is associated with more aggressive disease (likely due to recruitment of pro-tumoral leukocytes to the tumor site) and has also been implicated in resistance to mainstream chemotherapy. The delicate balance of TLR activation in the tumor microenvironment in different cell types altogether help shape the inflammatory profile and outcome of tumor growth or regression. With further studies, specific activation or repression of TLRs may be harnessed to offer novel immunotherapies or adjuvants to traditional chemotherapy for some OC patients. Herewith, we review recent literature on basic and translational research concerning therapeutic targeting of TLR pathways for the treatment of OC.
Collapse
Affiliation(s)
- Maria Muccioli
- Molecular and Cell Biology Program, Ohio University , Athens, OH , USA
| | - Fabian Benencia
- Molecular and Cell Biology Program, Ohio University , Athens, OH , USA ; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University , Athens, OH , USA
| |
Collapse
|
44
|
Zhou CX, Li D, Chen YL, Lu ZJ, Sun P, Cao YM, Bao HF, Fu YF, Li PH, Bai XW, Xie BX, Liu ZX. Resiquimod and polyinosinic-polycytidylic acid formulation with aluminum hydroxide as an adjuvant for foot-and-mouth disease vaccine. BMC Vet Res 2014; 10:2. [PMID: 24386990 PMCID: PMC3892093 DOI: 10.1186/1746-6148-10-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/31/2013] [Indexed: 11/10/2022] Open
Abstract
Background Toll-like receptor (TLR) agonists reportedly have potent antiviral and antitumor activities and may be a new kind of adjuvant for enhancing immune efficacy. Resiquimod (R848) is an imidazoquinoline compound with potent antiviral activity and functions through the TLR7/TLR8 MyD88-dependent signaling pathway. Polyinosinic-polycytidylic acid [poly(I:C)] is a synthetic analog of double-stranded RNA that induces the production of pro-inflammatory cytokines by the activation of NF-κB through TLR3. This study investigated the potential of R848 and poly(I:C) as an adjuvant 146S foot-and-mouth disease virus (FMDV) vaccine formulated with aluminum hydroxide (Al(OH)3). Results Antibody titers to FMDV and CD8+ T cells were markedly enhanced in mice immunized to 146S FMDV + Al(OH)3 + R848 + poly(I:C) compared with mice immunized to FMDV + ISA206. IFN-γ secretion substantially increased compared with IL-4 secretion by splenic T cells stimulated with FMDV antigens in vitro, suggesting that R848, poly(I:C), and with Al(OH)3 together biased the immune response toward a Th1-type direction. Conclusions These results indicated that the R848 and poly(I:C) together with Al(OH)3 enhanced humoral and cellular immune responses to immunization with 146S FMDV antigens. Thus, this new vaccine formulation can be used for FMDV prevention.
Collapse
Affiliation(s)
| | - Dong Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, OIE/National Foot-and-Mouth Disease Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Combinations of TLR ligands: a promising approach in cancer immunotherapy. Clin Dev Immunol 2013; 2013:271246. [PMID: 24371445 PMCID: PMC3859257 DOI: 10.1155/2013/271246] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/29/2013] [Accepted: 10/02/2013] [Indexed: 12/14/2022]
Abstract
Toll-like receptors (TLRs), a family of pattern recognition receptors recognizing molecules expressed by pathogens, are typically expressed by immune cells. However, several recent studies revealed functional TLR expression also on tumor cells. Their expression is a two-sided coin for tumor cells. Not only tumor-promoting effects of TLR ligands are described but also direct oncopathic and immunostimulatory effects. To clarify TLRs' role in colorectal cancer (CRC), we tested the impact of the TLR ligands LPS, Poly I:C, R848, and Taxol on primary human CRC cell lines (HROC40, HROC60, and HROC69) in vitro and in vivo (CT26). Taxol, not only a potent tumor-apoptosis-inducing, but also TLR4-activating chemotherapeutic compound, inhibited growth and viability of all cell lines, whereas the remaining TLR ligands had only marginal effects (R848 > LPS > Poly I:C). Combinations of the substances here did not improve the results, whereas antitumoral effects were dramatically boosted when human lymphocytes were added. Here, combining the TLR ligands often diminished antitumoral effects. In vivo, best tumor growth control was achieved by the combination of Taxol and R848. However, when combined with LPS, Taxol accelerated tumor growth. These data generally prove the potential of TLR ligands to control tumor growth and activate immune cells, but they also demonstrate the importance of choosing the right combinations.
Collapse
|
46
|
Toll-like receptors as therapeutic targets for cancer. Drug Discov Today 2013; 19:379-82. [PMID: 24012797 DOI: 10.1016/j.drudis.2013.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/21/2013] [Accepted: 08/23/2013] [Indexed: 12/28/2022]
Abstract
Stimulation of Toll-like receptors (TLRs) to activate the innate immune system has been a legitimate therapeutic strategy for some years. TLRs 3, 4, 7, 8 and 9 are all validated targets for cancer and a number of companies are developing agonists and vaccine adjuvants. TLR7 in particular has established proof-of-concept as a target in the topical treatment of bladder and skin cancers. However, the development of systemic treatments targeting TLR7 for most other cancers has proved difficult owing to cardiotoxicity or myelosuppression. Tantalisingly, recent animal data have demonstrated that a new class of modified TLR7 agonists can be administered systemically with a good toxicology profile, opening up this target in therapeutic interventions for systemic cancers.
Collapse
|
47
|
Therapy with TLR7 agonists induces lymphopenia: correlating pharmacology to mechanism in a mouse model. J Clin Immunol 2012; 32:1082-92. [PMID: 22476912 DOI: 10.1007/s10875-012-9687-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/16/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Synthetic TLR7 agonists have been proposed as oral replacements for interferonα (IFNα) therapy in the treatment of hepatitis C virus infection. However, adverse effects, such as lymphopenia and cardiovascular irregularities, have been observed in the clinical following treatment with TLR7 agonists. We wished to understand and characterise the relationship between TLR7 agonism and adverse effects. METHODS We compared responses to two prototypic TLR7 agonists (Resiquimod: R-848; and PF-04878691) in a mouse model and compared the responses to treatment with IFNα. We measured clinically relevant adverse effects such as lymphopenia and cardiovascular irregularities and related them to plasma drug levels and clinically relevant efficacy biomarkers such as the pro-inflammatory cytokine IP-10, 2'5'OAS and TLR7 receptor expression. RESULTS By 2 h post dose all agents had induced a dose-dependent transient lymphopenia. IFNα increased heart rate immediately following dosing, persisting for 5 h, whilst PF-04878691 induced significant reductions in blood pressure. Lymphopenia co-incided with maximum plasma drug levels, raised levels of IP-10 and the auto-induction of TLR7 expression in the blood and lymph nodes. Peak levels of 2'5'OAS occurred at 24 h post-dose and only at doses which also induced lymphopenia. CONCLUSIONS We conclude that systemic delivery of TLR7 agonists or IFNα induces similar exaggerated pharmacology, consistent with there being a narrow therapeutic window between efficacy and safety. This clinically validated mouse model will help to investigate whether more potent agonists or optimised dosing schedules, will be successful strategies for targeting TLR7 in patients.
Collapse
|
48
|
Seiffert M, Dietrich S, Jethwa A, Glimm H, Lichter P, Zenz T. Exploiting biological diversity and genomic aberrations in chronic lymphocytic leukemia. Leuk Lymphoma 2011; 53:1023-31. [PMID: 22023519 DOI: 10.3109/10428194.2011.631638] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is remarkable heterogeneity in the clinical course and biological characteristics of patient subgroups with chronic lymphocytic leukemia (CLL). Mutations of key tumor suppressors (ATM, miR-15a/16-1 and TP53) have been identified in CLL, and these aberrations are important "drivers" of the disease and some of its clinical characteristics. While some mutations are associated with poor outcome [particularly del(17p) and TP53 mutation], others are linked to a favorable clinical course [e.g. del(13q) as sole aberration]. In addition to genetic aberrations, antigen drive and microenvironmental interactions contribute to the pathogenesis of CLL. How the genetic aberrations impact on the process of antigen drive or microenvironmental interactions is currently unclear. Our improved understanding of the biology and clinical course of specific genetic subgroups is beginning to be translated into more specific and targeted treatment approaches. As a result, genetic subgroups are treated in distinct protocols. This review summarizes the contribution of the microenvironment and the most important genetic aberrations in CLL and how our improved knowledge of the biology of CLL may translate into improved treatment results.
Collapse
Affiliation(s)
- Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Smirnov D, Schmidt JJ, Capecchi JT, Wightman PD. Vaccine adjuvant activity of 3M-052: an imidazoquinoline designed for local activity without systemic cytokine induction. Vaccine 2011; 29:5434-42. [PMID: 21641953 DOI: 10.1016/j.vaccine.2011.05.061] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/04/2011] [Accepted: 05/19/2011] [Indexed: 10/18/2022]
Abstract
The human Toll-like receptors (TLRs) are a family of receptors, which sense the presence of various structural elements of pathogens and damaged or effete components in the host. As they do so, they activate two critical arms of host defense, the rapid innate immune response and an adaptive immune response. The innate immune response is typified by the generation of Th1 cytokines, chemokines and type 1 interferons. As such, agonists for the TLRs have potential as antiviral and anticancer therapeutics. They are also well suited to function as vaccine adjuvants. 3M imidazoquinoline (IRM) molecules were the first synthetic small molecules identified as TLR agonists and can affect their biological activities through TLR7, TLR8, or both. The breadth of therapeutic opportunities for this family of molecules can require formulations tailored to the specific application. One consideration is specific formulations to avoid a systemic distribution of these TLR agonists and resulting cytokine storm-like effects on the host. 3M-052 is an IRM bearing a C18 lipid moiety and designed for slow dissemination from the site of application. In the present study 3M-052 has been evaluated for its in vitro TLR activity and for its efficacy as a vaccine adjuvant using a recombinant hemagglutinin from H1N1 A/Puerto Rico/8/34. Given subcutaneously, 3M-052 drives a strong Th1 response to hemagglutinin and serum neutralization of viable H1N1 A/Puerto Rico/8/34 virus in the absence of circulating TNFα or the induction of Th1 cytokines.
Collapse
Affiliation(s)
- Dmitri Smirnov
- 3M Drug Delivery Systems, 3M Center, St. Paul, MN 55144, USA
| | | | | | | |
Collapse
|
50
|
Chiang CLL, Kandalaft LE, Coukos G. Adjuvants for enhancing the immunogenicity of whole tumor cell vaccines. Int Rev Immunol 2011; 30:150-82. [PMID: 21557641 DOI: 10.3109/08830185.2011.572210] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Whole tumor cell lysates can serve as excellent multivalent vaccines for priming tumor-specific CD8(+) and CD4(+) T cells. Whole cell vaccines can be prepared with hypochlorous acid oxidation, UVB-irradiation and repeat cycles of freeze and thaw. One major obstacle to successful immunotherapy is breaking self-tolerance to tumor antigens. Clinically approved adjuvants, including Montanide™ ISA-51 and 720, and keyhole-limpet proteins can be used to enhance tumor cell immunogenicity by stimulating both humoral and cellular anti-tumor responses. Other potential adjuvants, such as Toll-like receptor agonists (e.g., CpG, MPLA and PolyI:C), and cytokines (e.g., granulocyte-macrophage colony stimulating factor), have also been investigated.
Collapse
Affiliation(s)
- Cheryl Lai-Lai Chiang
- Ovarian Cancer Research Center, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104-6142, USA
| | | | | |
Collapse
|