1
|
Strobl S, Zucchetta D, Vašíček T, Monti A, Ruda A, Widmalm G, Heine H, Zamyatina A. Nonreducing Sugar Scaffold Enables the Development of Immunomodulatory TLR4-specific LPS Mimetics with Picomolar Potency. Angew Chem Int Ed Engl 2024; 63:e202408421. [PMID: 38870340 DOI: 10.1002/anie.202408421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Innate immune defense mechanisms against infection and cancer encompass the modulation of pattern recognition receptor (PRR)-mediated inflammation, including upregulation of various transcription factors and the activation of pro-inflammatory pathways important for immune surveillance. Dysfunction of PRRs-mediated signaling has been implicated in cancer and autoimmune diseases, while the overactivation of PRRs-driven responses during infection can lead to devastating consequences such as acute lung injury or sepsis. We used crystal structure-based design to develop immunomodulatory lipopolysaccharide (LPS) mimetics targeting one of the ubiquitous PRRs, Toll-like Receptor 4 (TLR4). Taking advantage of an exo-anomeric conformation and specific molecular shape of synthetic nonreducing β,β-diglucosamine, which was investigated by NMR, we developed two sets of lipid A mimicking glycolipids capable of either potently activating innate immune responses or inhibiting pro-inflammatory signaling. Stereoselective 1,1'-glycosylation towards fully orthogonally protected nonreducing GlcNβ(1↔1')βGlcN followed by stepwise assembly of differently functionalised phosphorylated glycolipids provided biologically active molecules that were evaluated for their ability to trigger or to inhibit cellular innate immune responses. Two LPS mimetics, identified as potent TLR4-specific inducers of the intracellular signaling pathways, serve as vaccine adjuvant- and immunotherapy candidates, while anionic glycolipids with TLR4-inhibitory potential hold therapeutic promise for the management of acute or chronic inflammation.
Collapse
Affiliation(s)
- Sebastian Strobl
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Daniele Zucchetta
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Tomáš Vašíček
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Alessandro Monti
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Alessandro Ruda
- Department of Organic Chemistry, Stockholm University, S-106 91, Stockholm, Sweden
| | - Göran Widmalm
- Department of Organic Chemistry, Stockholm University, S-106 91, Stockholm, Sweden
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, Borstel, 23845, Germany
| | - Alla Zamyatina
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| |
Collapse
|
2
|
Tsukidate T, Hespen CW, Hang HC. Small molecule modulators of immune pattern recognition receptors. RSC Chem Biol 2023; 4:1014-1036. [PMID: 38033733 PMCID: PMC10685800 DOI: 10.1039/d3cb00096f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/03/2023] [Indexed: 12/02/2023] Open
Abstract
Pattern recognition receptors (PRRs) represent a re-emerging class of therapeutic targets for vaccine adjuvants, inflammatory diseases and cancer. In this review article, we summarize exciting developments in discovery and characterization of small molecule PRR modulators, focusing on Toll-like receptors (TLRs), NOD-like receptors (NLRs) and the cGAS-STING pathway. We also highlight PRRs that are currently lacking small molecule modulators and opportunities for chemical biology and therapeutic discovery.
Collapse
Affiliation(s)
- Taku Tsukidate
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
| | - Charles W Hespen
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
| | - Howard C Hang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
- Department of Immunology and Microbiology and Department of Chemistry, Scripps Research, La Jolla California 92037 USA
| |
Collapse
|
3
|
Nguyen DM, Poveda C, Pollet J, Gusovsky F, Bottazzi ME, Hotez PJ, Jones KM. The impact of vaccine-linked chemotherapy on liver health in a mouse model of chronic Trypanosoma cruzi infection. PLoS Negl Trop Dis 2023; 17:e0011519. [PMID: 37988389 PMCID: PMC10697595 DOI: 10.1371/journal.pntd.0011519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/05/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Chagas disease, chronic infection with Trypanosoma cruzi, mainly manifests as cardiac disease. However, the liver is important for both controlling parasite burdens and metabolizing drugs. Notably, high doses of anti-parasitic drug benznidazole (BNZ) causes liver damage. We previously showed that combining low dose BNZ with a prototype therapeutic vaccine is a dose sparing strategy that effectively reduced T. cruzi induced cardiac damage. However, the impact of this treatment on liver health is unknown. Therefore, we evaluated several markers of liver health after treatment with low dose BNZ plus the vaccine therapy in comparison to a curative dose of BNZ. METHODOLOGY Female BALB/c mice were infected with a bioluminescent T. cruzi H1 clone for approximately 70 days, then randomly divided into groups of 15 mice each. Mice were treated with a 25mg/kg BNZ, 25μg Tc24-C4 protein/ 5μg E6020-SE (Vaccine), 25mg/kg BNZ followed by vaccine, or 100mg/kg BNZ (curative dose). At study endpoints we evaluated hepatomegaly, parasite burden by quantitative PCR, cellular infiltration by histology, and expression of B-cell translocation gene 2(BTG2) and Peroxisome proliferator-activated receptor alpha (PPARα) by RT-PCR. Levels of alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP) and lactate dehydrogenase (LDH) were quantified from serum. RESULTS Curative BNZ treatment significantly reduced hepatomegaly, liver parasite burdens, and the quantity of cellular infiltrate, but significantly elevated serum levels of ALT, AST, and LDH. Low BNZ plus vaccine did not significantly affect hepatomegaly, parasite burdens or the quantity of cellular infiltrate, but only elevated ALT and AST. Low dose BNZ significantly decreased expression of both BTG2 and PPARα, and curative BNZ reduced expression of BTG2 while low BNZ plus vaccine had no impact. CONCLUSIONS These data confirm toxicity associated with curative doses of BNZ and suggest that while dose sparing low BNZ plus vaccine treatment does not reduce parasite burdens, it better preserves liver health.
Collapse
Affiliation(s)
- Duc Minh Nguyen
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Cristina Poveda
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fabian Gusovsky
- Global Health Research, Eisai, Inc., Cambridge, Massachusetts, United States of America
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
- James A. Baker III Institute for Public Policy, Rice University, Houston, Texas, United States of America
- Hagler Institute for Advanced Study at Texas A&M University, College Station, Texas, United States of America
| | - Kathryn Marie Jones
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
4
|
Syntin P, Piras-Douce F, Dalençon F, Garinot M, Haensler J. Nonclinical safety assessments of a novel synthetic toll-like receptor 4 agonist and saponin based adjuvant. Toxicol Appl Pharmacol 2023; 460:116358. [PMID: 36572229 DOI: 10.1016/j.taap.2022.116358] [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: 11/03/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
A full nonclinical safety package was performed to support the clinical use of SPA14, a novel liposome-based vaccine adjuvant containing the synthetic toll-like receptor 4 agonist E6020 and saponin QS21. E6020 and QS21 were tested negative for their potential genotoxic effects in Ames, micronucleus, or mouse-lymphoma TK (thymidine kinase) assay. To evaluate the potential local and systemic effects of SPA14, two toxicity studies were performed in rabbits. In the first dose range finding toxicity study, rabbits received two intramuscular injections of SPA14 at increasing doses of E6020 combined with two antigens, a control (saline), the two antigens alone, or the antigens adjuvanted with a liposome-based adjuvant AS01B. No systemic toxicity was detected, supporting the dose of 5 μg of E6020 for the subsequent pivotal study. In the second repeated dose toxicity study, rabbits received four intramuscular injections of SPA14 alone, a control (saline), SPA14 combined with two antigens, the two antigens alone, or the antigens combined with AF03 adjuvant, which is a squalene-based emulsion. SPA14 alone or in combination with the antigens was well tolerated and did not cause any systemic toxicity. Finally, two safety pharmacology studies were conducted to assess potential cardiovascular and respiratory effects of E6020 and SPA14 in conscious telemetered cynomolgus monkeys and beagle dogs, respectively. One subcutaneous injection of E6020 in monkeys and one intramuscular injection of SPA14 in dogs had no consequences on respiratory and cardiovascular functions. Altogether these results support the clinical development of SPA14.
Collapse
|
5
|
Heine H, Zamyatina A. Therapeutic Targeting of TLR4 for Inflammation, Infection, and Cancer: A Perspective for Disaccharide Lipid A Mimetics. Pharmaceuticals (Basel) 2022; 16:23. [PMID: 36678520 PMCID: PMC9864529 DOI: 10.3390/ph16010023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
The Toll-like receptor 4 (TLR4) signaling pathway plays a central role in the prompt defense against infectious challenge and provides immediate response to Gram-negative bacterial infection. The TLR4/MD-2 complex can sense and respond to various pathogen-associated molecular patterns (PAMPs) with bacterial lipopolysaccharide (LPS) being the most potent and the most frequently occurring activator of the TLR4-mediated inflammation. TLR4 is believed to be both a friend and foe since improperly regulated TLR4 signaling can result in the overactivation of immune responses leading to sepsis, acute lung injury, or pathologic chronic inflammation involved in cancer and autoimmune disease. TLR4 is also considered a legitimate target for vaccine adjuvant development since its activation can boost the adaptive immune responses. The dual action of the TLR4 complex justifies the efforts in the development of both TLR4 antagonists as antisepsis drug candidates or remedies for chronic inflammatory diseases and TLR4 agonists as vaccine adjuvants or immunotherapeutics. In this review, we provide a brief overview of the biochemical evidences for possible pharmacologic applications of TLR4 ligands as therapeutics and report our systematic studies on the design, synthesis, and immunobiological evaluation of carbohydrate-based TLR4 antagonists with nanomolar affinity for MD-2 as well as disaccharide-based TLR4 agonists with picomolar affinity for the TLR4/MD-2 complex.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel—Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, 23845 Borstel, Germany
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
6
|
Dzul-Huchim VM, Ramirez-Sierra MJ, Martinez-Vega PP, Rosado-Vallado ME, Arana-Argaez VE, Ortega-Lopez J, Gusovsky F, Dumonteil E, Cruz-Chan JV, Hotez P, Bottazzi ME, Villanueva-Lizama LE. Vaccine-linked chemotherapy with a low dose of benznidazole plus a bivalent recombinant protein vaccine prevents the development of cardiac fibrosis caused by Trypanosoma cruzi in chronically-infected BALB/c mice. PLoS Negl Trop Dis 2022; 16:e0010258. [PMID: 36095001 PMCID: PMC9499242 DOI: 10.1371/journal.pntd.0010258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/22/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background Chagas disease (CD) is caused by Trypanosoma cruzi and affects 6–7 million people worldwide. Approximately 30% of chronic patients develop chronic chagasic cardiomyopathy (CCC) after decades. Benznidazole (BNZ), one of the first-line chemotherapy used for CD, induces toxicity and fails to halt the progression of CCC in chronic patients. The recombinant parasite-derived antigens, including Tc24, Tc24-C4, TSA-1, and TSA-1-C4 with Toll-like receptor 4 (TLR-4) agonist-adjuvants reduce cardiac parasite burdens, heart inflammation, and fibrosis, leading us to envision their use as immunotherapy together with BNZ. Given genetic immunization (DNA vaccines) encoding Tc24 and TSA-1 induce protective immunity in mice and dogs, we propose that immunization with the corresponding recombinant proteins offers an alternative and feasible strategy to develop these antigens as a bivalent human vaccine. We hypothesized that a low dose of BNZ in combination with a therapeutic vaccine (TSA-1-C4 and Tc24-C4 antigens formulated with a synthetic TLR-4 agonist-adjuvant, E6020-SE) given during early chronic infection, could prevent cardiac disease progression and provide antigen-specific T cell immunity. Methodology/ Principal findings We evaluated the therapeutic vaccine candidate plus BNZ (25 mg/kg/day/7 days) given on days 72 and 79 post-infection (p.i) (early chronic phase). Fibrosis, inflammation, and parasite burden were quantified in heart tissue at day 200 p.i. (late chronic phase). Further, spleen cells were collected to evaluate antigen-specific CD4+ and CD8+ T cell immune response, using flow cytometry. We found that vaccine-linked BNZ treated mice had lower cardiac fibrosis compared to the infected untreated control group. Moreover, cells from mice that received the immunotherapy had higher stimulation index of antigen-specific CD8+Perforin+ T cells as well as antigen-specific central memory T cells compared to the infected untreated control. Conclusions Our results suggest that the bivalent immunotherapy together with BNZ treatment given during early chronic infection protects BALB/c mice against cardiac fibrosis progression and activates a strong CD8+ T cell response by in vitro restimulation, evidencing the induction of a long-lasting T. cruzi-immunity. Chagas disease (CD) is a neglected tropical disease caused by the parasite Trypanosoma cruzi, transmitted through contact with infected feces of vectors bugs. CD can induce cardiac abnormalities including the development of fibrosis and eventually death. Benznidazole (BNZ) is the first-line drug approved against CD, however, its toxicity and lack of efficacy in the chronic phase have limited its use. Previous studies have demonstrated the feasibility of reducing doses of BNZ given in combination with therapeutic vaccines during the acute phase of CD, which increases its tolerability and reduces adverse side effects. Considering that patients are often diagnosed until more advanced stages of the disease, its necessary to evaluate therapies given in the chronic phase of CD. In this study, we evaluated a vaccine formulated with the recombinant T. cruzi-antigens TSA-1-C4 and Tc24-C4 and the adjuvant E6020-SE in combination with a low dose of BNZ given during the chronic phase of T. cruzi-infection using a murine model. The authors found that the combination therapy protects mice against cardiac fibrosis progression, allows the activation of a CD8+ T cell response, and induces a prolonged memory response against T. cruzi. This study supports the development of the vaccine-linked chemotherapy approach to prevent T. cruzi chronic infection.
Collapse
Affiliation(s)
- Victor Manuel Dzul-Huchim
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Maria Jesus Ramirez-Sierra
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Pedro Pablo Martinez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Miguel Enrique Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Victor Ermilo Arana-Argaez
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Jaime Ortega-Lopez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Fabian Gusovsky
- Eisai, Inc., Eisai Inc, Andover, Massachusetts, United States of America
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Los Angeles, United States of America
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Peter Hotez
- Texas Children’s Center for Vaccine Development, Departments of Pediatrics and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - María Elena Bottazzi
- Texas Children’s Center for Vaccine Development, Departments of Pediatrics and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Liliana Estefania Villanueva-Lizama
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
- * E-mail:
| |
Collapse
|
7
|
Haile CN, Varner KJ, Huijing X, Arora R, Orson FM, Kosten TR, Kosten TA. Active and Passive Immunization with an Anti-Methamphetamine Vaccine Attenuates the Behavioral and Cardiovascular Effects of Methamphetamine. Vaccines (Basel) 2022; 10:vaccines10091508. [PMID: 36146588 PMCID: PMC9503672 DOI: 10.3390/vaccines10091508] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Methamphetamine use disorder (MUD) is a growing health concern with no FDA-approved treatment. The present series of studies build upon our previous work developing an anti-methamphetamine (MA) vaccine for MUD. We determined the effects of a formulation that included tetanus-toxoid (TT) conjugated to succinyl-methamphetamine (TT-SMA) adsorbed onto aluminum hydroxide (alum) in combination with the novel Toll-Like Receptor-5 agonist, entolimod. METHODS Mice were vaccinated (0, 3, 6 weeks) with TT-SMA+alum and various doses of entolimod to determine an optimal dose for enhancing immunogenicity against MA. Functional effects were then assessed using MA-induced locomotor activation in mice. Experiments using passive immunization of antibodies generated by the vaccine tested its ability to attenuate MA-induced cardiovascular effects and alter the reinforcing effects of MA in an MA-induced reinstatement of a drug seeking model of relapse in male and female rats. RESULTS Antibody levels peaked at 10 weeks following vaccination with TT-SMA+alum combined with entolimod (1, 3 and 10 μg). MA-induced locomotor activation was significantly attenuated in vaccinated vs. unvaccinated mice and antibody levels significantly correlated with ambulation levels. Passive immunization decreased mean arterial pressure following MA dosing in rats of both sexes but did not alter heart rate. Passive immunization also attenuated the ability of MA to reinstate extinguished drug-seeking behavior in male and female rats. Results support further development of this vaccine for relapse prevention for individuals with MUD.
Collapse
Affiliation(s)
- Colin N. Haile
- Department of Psychology/TIMES, University of Houston, Houston, TX 77204, USA
| | - Kurt J. Varner
- Department of Pharmacology and Experimental Therapeutics and the Cardiovascular Center of Excellence, LSUHSC, New Orleans, LA 70112, USA
| | - Xia Huijing
- Department of Pharmacology and Experimental Therapeutics and the Cardiovascular Center of Excellence, LSUHSC, New Orleans, LA 70112, USA
| | - Reetakshi Arora
- The Michael E DeBakey Veteran’s Affairs Medical Center, Houston, TX 77030, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank M. Orson
- The Michael E DeBakey Veteran’s Affairs Medical Center, Houston, TX 77030, USA
- Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas R. Kosten
- The Michael E DeBakey Veteran’s Affairs Medical Center, Houston, TX 77030, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Therese A. Kosten
- Department of Psychology/TIMES, University of Houston, Houston, TX 77204, USA
- Correspondence:
| |
Collapse
|
8
|
Strobl S, Hofbauer K, Heine H, Zamyatina A. Lipid A Mimetics Based on Unnatural Disaccharide Scaffold as Potent TLR4 Agonists for Prospective Immunotherapeutics and Adjuvants. Chemistry 2022; 28:e202200547. [PMID: 35439332 PMCID: PMC9325513 DOI: 10.1002/chem.202200547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Indexed: 11/11/2022]
Abstract
TLR4 is a key pattern recognition receptor that can sense pathogen- and danger- associated molecular patterns to activate the downstream signaling pathways which results in the upregulation of transcription factors and expression of interferons and cytokines to mediate protective pro-inflammatory responses involved in immune defense. Bacterial lipid A is the primary TLR4 ligand with very complex, species-specific, and barely predictable structure-activity relationships. Given that therapeutic targeting of TLR4 is an emerging tool for management of a variety of human diseases, the development of novel TLR4 activating biomolecules other than lipid A is of vast importance. We report on design, chemical synthesis and immunobiology of novel glycan-based lipid A-mimicking molecules that can activate human and murine TLR4-mediated signaling with picomolar affinity. Exploiting crystal structure - based design we have created novel disaccharide lipid A mimetics (DLAMs) where the inherently flexible β(1→6)-linked diglucosamine backbone of lipid A is exchanged with a conformationally restrained non-reducing βGlcN(1↔1')βGlcN scaffold. Excellent stereoselectivity in a challenging β,β-1,1' glycosylation was achieved by tuning the reactivities of donor and acceptor molecules using protective group manipulation strategy. Divergent streamlined synthesis of β,β-1,1'-linked diglucosamine-derived glycolipids entailing multiple long-chain (R)-3- acyloxyacyl residues and up two three phosphate groups was developed. Specific 3D-molecular shape and conformational rigidity of unnatural β,β-1,1'-linked diglucosamine combined with carefully optimized phosphorylation and acylation pattern ensured efficient induction of the TLR4-mediated signaling in a species-independent manner.
Collapse
Affiliation(s)
- Sebastian Strobl
- Department of ChemistryUniversity of Natural Resources and Life SciencesMuthgasse 18Vienna1190Austria
| | - Karin Hofbauer
- Department of ChemistryUniversity of Natural Resources and Life SciencesMuthgasse 18Vienna1190Austria
| | - Holger Heine
- Research Group Innate ImmunityResearch Center Borstel-Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Disease (DZL)Parkallee 22Borstel23845Germany
| | - Alla Zamyatina
- Department of ChemistryUniversity of Natural Resources and Life SciencesMuthgasse 18Vienna1190Austria
| |
Collapse
|
9
|
Wu X, Li W. The Applications of
β
‐Keto
Amides for Heterocycle Synthesis. J Heterocycl Chem 2022. [DOI: 10.1002/jhet.4497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Xiaoqiang Wu
- School of Materials and Chemistry University of Shanghai for Science and Technology Shanghai P. R. China
| | - Wanfang Li
- School of Materials and Chemistry University of Shanghai for Science and Technology Shanghai P. R. China
| |
Collapse
|
10
|
Garcia-Vello P, Di Lorenzo F, Zucchetta D, Zamyatina A, De Castro C, Molinaro A. Lipopolysaccharide lipid A: A promising molecule for new immunity-based therapies and antibiotics. Pharmacol Ther 2022; 230:107970. [PMID: 34454000 DOI: 10.1016/j.pharmthera.2021.107970] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022]
Abstract
Lipopolysaccharides (LPS) are the main components of the external leaflet of the Gram-negative outer membrane and consist of three different moieties: lipid A, core oligosaccharide, and O-polysaccharide. The lipid A is a glucosamine disaccharide with different levels of acylation and phosphorylation, beside carrying, in certain cases, additional substituents on the sugar backbone. It is also the main immunostimulatory part of the LPS, as its recognition by the host immune system represents a fundamental event for detection of perilous microorganisms. Moreover, an uncontrolled immune response caused by a large amount of circulating LPS can lead to dramatic outcomes for human health, such as septic shock. The immunostimulant properties of an LPS incredibly vary depending on lipid A chemical structure, and for this reason, natural and synthetic variants of the lipid A are under study to develop new drugs that mimic or antagonise its natural effects. Here, we review past and recent findings on the lipid A as an antibiotic target and immune-therapeutic molecule, with a special attention on the crucial role of the chemical structure and its exploitation for conceiving novel strategies for treatment of several immune-related pathologies.
Collapse
Affiliation(s)
- Pilar Garcia-Vello
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| | - Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| | - Daniele Zucchetta
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| |
Collapse
|
11
|
Schijns V, Majhen D, van der Ley P, Thakur A, Summerfield A, Berisio R, Nativi C, Fernández-Tejada A, Alvarez-Dominguez C, Gizurarson S, Zamyatina A, Molinaro A, Rosano C, Jakopin Ž, Gursel I, McClean S. Rational Vaccine Design in Times of Emerging Diseases: The Critical Choices of Immunological Correlates of Protection, Vaccine Antigen and Immunomodulation. Pharmaceutics 2021; 13:501. [PMID: 33917629 PMCID: PMC8067490 DOI: 10.3390/pharmaceutics13040501] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023] Open
Abstract
Vaccines are the most effective medical intervention due to their continual success in preventing infections and improving mortality worldwide. Early vaccines were developed empirically however, rational design of vaccines can allow us to optimise their efficacy, by tailoring the immune response. Establishing the immune correlates of protection greatly informs the rational design of vaccines. This facilitates the selection of the best vaccine antigens and the most appropriate vaccine adjuvant to generate optimal memory immune T cell and B cell responses. This review outlines the range of vaccine types that are currently authorised and those under development. We outline the optimal immunological correlates of protection that can be targeted. Finally we review approaches to rational antigen selection and rational vaccine adjuvant design. Harnessing current knowledge on protective immune responses in combination with critical vaccine components is imperative to the prevention of future life-threatening diseases.
Collapse
Affiliation(s)
- Virgil Schijns
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
- Epitopoietic Research Corporation (ERC), 5374 RE Schaijk, The Netherlands
| | - Dragomira Majhen
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Instiute, HR-10000 Zagreb, Croatia;
| | - Peter van der Ley
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
| | - Aneesh Thakur
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Artur Summerfield
- Institute of Virology and Immunology, 3147 Mittelhausern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, I-80134 Naples, Italy;
| | - Cristina Nativi
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy;
| | - Alberto Fernández-Tejada
- Chemical Immunology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Biscay Science and Technology Park, 48160 Derio-Bilbao, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Carmen Alvarez-Dominguez
- Facultativo en plantilla (Research Faculty), Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, 107 Reykjavik, Iceland;
- Department of Pharmacy, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, I-80126 Napoli, Italy;
- Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Camillo Rosano
- Proteomics and Mass Spectrometry Unit, IRCCS Policlinico San Martino, 16132 Genova-1, Italy;
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubiljana, Slovenia;
| | - Ihsan Gursel
- Molecular Biology and Genetics Department, Science Faculty, Bilkent University, Bilkent, 06800 Ankara, Turkey;
| | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
12
|
Heine H, Adanitsch F, Peternelj TT, Haegman M, Kasper C, Ittig S, Beyaert R, Jerala R, Zamyatina A. Tailored Modulation of Cellular Pro-inflammatory Responses With Disaccharide Lipid A Mimetics. Front Immunol 2021; 12:631797. [PMID: 33815382 PMCID: PMC8012497 DOI: 10.3389/fimmu.2021.631797] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
Pro-inflammatory signaling mediated by Toll-like receptor 4 (TLR4)/myeloid differentiation-2 (MD-2) complex plays a crucial role in the instantaneous protection against infectious challenge and largely contributes to recovery from Gram-negative infection. Activation of TLR4 also boosts the adaptive immunity which is implemented in the development of vaccine adjuvants by application of minimally toxic TLR4 activating ligands. The modulation of pro-inflammatory responses via the TLR4 signaling pathway was found beneficial for management of acute and chronic inflammatory disorders including asthma, allergy, arthritis, Alzheimer disease pathology, sepsis, and cancer. The TLR4/MD-2 complex can recognize the terminal motif of Gram-negative bacterial lipopolysaccharide (LPS)—a glycophospholipid lipid A. Although immense progress in understanding the molecular basis of LPS-induced TLR4-mediated signaling has been achieved, gradual, and predictable TLR4 activation by structurally defined ligands has not yet been attained. We report on controllable modulation of cellular pro-inflammatory responses by application of novel synthetic glycolipids—disaccharide-based lipid A mimetics (DLAMs) having picomolar affinity for TLR4/MD-2. Using crystal structure inspired design we have developed endotoxin mimetics where the inherently flexible β(1 → 6)-linked diglucosamine backbone of lipid A is replaced by a conformationally restricted α,α-(1↔1)-linked disaccharide scaffold. The tertiary structure of the disaccharide skeleton of DLAMs mirrors the 3-dimensional shape of TLR4/MD-2 bound E. coli lipid A. Due to exceptional conformational rigidity of the sugar scaffold, the specific 3D organization of DLAM must be preserved upon interaction with proteins. These structural factors along with specific acylation and phosphorylation pattern can ensure picomolar affinity for TLR4 and permit efficient dimerization of TLR4/MD-2/DLAM complexes. Since the binding pose of lipid A in the binding pocket of MD-2 (±180°) is crucial for the expression of biological activity, the chemical structure of DLAMs was designed to permit a predefined binding orientation in the binding groove of MD-2, which ensured tailored and species-independent (human and mice) TLR4 activation. Manipulating phosphorylation and acylation pattern at the sugar moiety facing the secondary dimerization interface allowed for adjustable modulation of the TLR4-mediated signaling. Tailored modulation of cellular pro-inflammatory responses by distinct modifications of the molecular structure of DLAMs was attained in primary human and mouse immune cells, lung epithelial cells and TLR4 transfected HEK293 cells.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel - Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Florian Adanitsch
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Tina Tinkara Peternelj
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Mira Haegman
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | | | - Simon Ittig
- Biozentrum University of Basel, Basel, Switzerland
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Roman Jerala
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
13
|
Beatty P, Ranganathan S, Finn OJ. Prevention of colitis-associated colon cancer using a vaccine to target abnormal expression of the MUC1 tumor antigen. Oncoimmunology 2021; 1:263-270. [PMID: 22737601 PMCID: PMC3382848 DOI: 10.4161/onci.18950] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Association between chronic inflammation and cancer development is exemplified by inflammatory bowel disease (IBD) where patients with chronic uncontrolled colitis have a significantly increased risk of developing colitis-associated colorectal cancer (CACC). CACC appears to progresses through the inflammation-dysplasia-carcinoma sequence. This highlights the need to identify targets and interventions that reduce inflammation and prevent development of dysplasia in the context of IBD. Using the dextran sulfate sodium (DSS) mouse model of chronic colitis and CACC, we show that an important target of intervention in human disease would be the epithelial cell molecule MUC1 that is aberrantly expressed on inflamed colonocytes and promotes inflammation and progression to CACC. We show that a MUC1 vaccine can ameliorate chronic colitis and prevent development of dysplasia in the colon and thus extend survival in human MUC1 transgenic mice. This study supports the potential of prophylactic vaccines to target antigens that become aberrantly expressed in chronic inflammation (e.g., IBD) and continue to be expressed on the associated cancers (e.g., colon cancer), to prevent and/or treat both diseases.
Collapse
Affiliation(s)
- Pamela Beatty
- Department of Immunology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; Division of Pediatric Pathology; Children's Hospital of Pittsburgh; Pittsburgh, PA USA
| | | | | |
Collapse
|
14
|
Lee JC, Janda KD. Immunopharmacotherapeutic advancements in addressing methamphetamine abuse. RSC Chem Biol 2021; 2:77-93. [PMID: 34458776 PMCID: PMC8341824 DOI: 10.1039/d0cb00165a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/01/2020] [Indexed: 12/26/2022] Open
Abstract
Methamphetamine (METH) is an illicit psychostimulant that is known to account for substance abuse disorders globally, second only to opioids, yet has no approved pharmacotherapies. Traditional therapies employ small molecule agonists or antagonists for substance use disorders or overdose reversal by targeting drug-specific receptors in the brain. However, the comprehensive mechanism of METH on multiple sites within the central nervous system (CNS) implies its receptors lack the high affinity and specificity required for an "ideal" drug target. The alternative to pharmacotherapies is to sequester abused drugs in the periphery, effectively eliminating the effects from CNS receptor occupation through pharmacokinetic antagonism. This review presents updates on immunopharmacotherapeutic advancements in addressing methamphetamine abuse by focusing on the cultivation of research optimization strategies regarding hapten chemistry, carrier proteins, and adjuvants implemented in active immunization. Furthermore, we discuss necessary developments for each component of active immunopharmacotherapies and the future of active vaccines in treating METH use disorder.
Collapse
Affiliation(s)
- Jinny Claire Lee
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute 10550 North Torrey Pines Rd La Jolla CA 92037 USA
| | - Kim D Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute 10550 North Torrey Pines Rd La Jolla CA 92037 USA
| |
Collapse
|
15
|
Zamyatina A, Heine H. Lipopolysaccharide Recognition in the Crossroads of TLR4 and Caspase-4/11 Mediated Inflammatory Pathways. Front Immunol 2020; 11:585146. [PMID: 33329561 PMCID: PMC7732686 DOI: 10.3389/fimmu.2020.585146] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
The innate immune response to lipopolysaccharide is essential for host defense against Gram-negative bacteria. In response to bacterial infection, the TLR4/MD-2 complex that is expressed on the surface of macrophages, monocytes, dendritic, and epithelial cells senses picomolar concentrations of endotoxic LPS and triggers the production of various pro-inflammatory mediators. In addition, LPS from extracellular bacteria which is either endocytosed or transfected into the cytosol of host cells or cytosolic LPS produced by intracellular bacteria is recognized by cytosolic proteases caspase-4/11 and hosts guanylate binding proteins that are involved in the assembly and activation of the NLRP3 inflammasome. All these events result in the initiation of pro-inflammatory signaling cascades directed at bacterial eradication. However, TLR4-mediated signaling and caspase-4/11-induced pyroptosis are largely involved in the pathogenesis of chronic and acute inflammation. Both extra- and intracellular LPS receptors-TLR4/MD-2 complex and caspase-4/11, respectively-are able to directly bind the lipid A motif of LPS. Whereas the structural basis of lipid A recognition by the TLR4 complex is profoundly studied and well understood, the atomic mechanism of LPS/lipid A interaction with caspase-4/11 is largely unknown. Here we describe the LPS-induced TLR4 and caspase-4/11 mediated signaling pathways and their cross-talk and scrutinize specific structural features of the lipid A motif of diverse LPS variants that have been reported to activate caspase-4/11 or to induce caspase-4/11 mediated activation of NLRP3 inflammasome (either upon transfection of LPS in vitro or upon infection of cell cultures with intracellular bacteria or by LPS as a component of the outer membrane vesicles). Generally, inflammatory caspases show rather similar structural requirements as the TLR4/MD-2 complex, so that a "basic" hexaacylated bisphosphorylated lipid A architecture is sufficient for activation. However, caspase-4/11 can sense and respond to much broader variety of lipid A variants compared to the very "narrow" specificity of TLR4/MD-2 complex as far as the number and the length of lipid chains attached at the diglucosamine backbone of lipid A is concerned. Besides, modification of the lipid A phosphate groups with positively charged appendages such as phosphoethanolamine or aminoarabinose could be essential for the interaction of lipid A/LPS with inflammatory caspases and related proteins.
Collapse
Affiliation(s)
- Alla Zamyatina
- Institute of Organic Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel—Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Disease (DZL), Borstel, Germany
| |
Collapse
|
16
|
Villanueva-Lizama LE, Cruz-Chan JV, Versteeg L, Teh-Poot CF, Hoffman K, Kendricks A, Keegan B, Pollet J, Gusovsky F, Hotez PJ, Bottazzi ME, Jones KM. TLR4 agonist protects against Trypanosoma cruzi acute lethal infection by decreasing cardiac parasite burdens. Parasite Immunol 2020; 42:e12769. [PMID: 32592180 DOI: 10.1111/pim.12769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/11/2023]
Abstract
E6020 is a synthetic agonist of Toll-like receptor-4 (TLR4). The purpose of this study was to evaluate the effect of different doses of E6020-SE on Trypanosoma cruzi-specific immune responses and its ability to confer protection against acute lethal infection in mice. Forty female BALB/c were infected with 500 trypomastigotes of T cruzi H1 strain, divided into four groups (n = 10) and treated at 7- and 14-day post-infection (dpi) with different doses of E6020-SE or PBS (control). Survival was followed for 51 days, mice were euthanized and hearts were collected to evaluate parasite burden, inflammation and fibrosis. We found significantly higher survival and lower parasite burdens in mice injected with E6020-SE at all doses compared to the control group. However, E6020-SE treatment did not significantly reduce cardiac inflammation or fibrosis. On the other hand, E6020-SE modulated Th1 and Th2 cytokines, decreasing IFN-γ and IL-4 in a dose-dependent manner after stimulation with parasite antigens. We conclude that E6020-SE alone increased survival by decreasing cardiac parasite burdens in BALB/c mice acutely infected with T cruzi but failed to prevent cardiac damage. Our results suggest that for optimal protection, a vaccine antigen is necessary to balance and orient a protective immune response.
Collapse
Affiliation(s)
- Liliana E Villanueva-Lizama
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Julio V Cruz-Chan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Leroy Versteeg
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Christian F Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Kristyn Hoffman
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - April Kendricks
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Brian Keegan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | | | - Peter J Hotez
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA.,James A. Baker III Institute for Public Policy, Rice University, Houston, TX, USA.,Hagler Institute for Advanced Study at Texas A&M University, College Station, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA
| | - Kathryn M Jones
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
17
|
Richard K, Perkins DJ, Harberts EM, Song Y, Gopalakrishnan A, Shirey KA, Lai W, Vlk A, Mahurkar A, Nallar S, Hawkins LD, Ernst RK, Vogel SN. Dissociation of TRIF bias and adjuvanticity. Vaccine 2020; 38:4298-4308. [PMID: 32389496 PMCID: PMC7302928 DOI: 10.1016/j.vaccine.2020.04.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/03/2020] [Accepted: 04/18/2020] [Indexed: 02/08/2023]
Abstract
Toll-like receptors (TLRs), a family of "pattern recognition receptors," bind microbial and host-derived molecules, leading to intracellular signaling and proinflammatory gene expression. TLR4 is unique in that ligand-mediated activation requires the co-receptor myeloid differentiation 2 (MD2) to initiate two signaling cascades: the MyD88-dependent pathway is initiated at the cell membrane, and elicits rapid MAP kinase and NF-κB activation, while the TIR-domain containing adaptor inducing interferon-β (TRIF)-dependent pathway is initiated from TLR4-containing endosomes and results in IRF3 activation. Previous studies associated inflammation with the MyD88 pathway and adjuvanticity with the TRIF pathway. Gram-negative lipopolysaccharide (LPS) is a potent TLR4 agonist, and structurally related molecules signal through TLR4 to differing extents. Herein, we compared monophosphoryl lipid A (sMPL) and E6020, two synthetic, non-toxic LPS lipid A analogs used as vaccine adjuvants, for their capacities to activate TLR4-mediated innate immune responses and to enhance antibody production. In mouse macrophages, high dose sMPL activates MyD88-dependent signaling equivalently to E6020, while E6020 exhibits significantly more activation of the TRIF pathway (a "TRIF bias") than sMPL. Eritoran, a TLR4/MD2 antagonist, competitively inhibited sMPL more strongly than E6020. Despite these differences, sMPL and E6020 adjuvants enhanced antibody responses to comparable extents, with balanced immunoglobulin (Ig) isotypes in two immunization models. These data indicate that a TRIF bias is not necessarily predictive of superior adjuvanticity.
Collapse
Affiliation(s)
- Katharina Richard
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Darren J Perkins
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Erin M Harberts
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry (UMSOD), Baltimore, MD, United States
| | - Yang Song
- Genome Informatics Core, Institute for Genome Sciences (IGS), UMSOM, Baltimore, MD, United States
| | - Archana Gopalakrishnan
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Wendy Lai
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Alexandra Vlk
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | - Anup Mahurkar
- Genome Informatics Core, Institute for Genome Sciences (IGS), UMSOM, Baltimore, MD, United States
| | - Shreeram Nallar
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States
| | | | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry (UMSOD), Baltimore, MD, United States
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, United States.
| |
Collapse
|
18
|
Dumonteil E, Herrera C, Tu W, Goff K, Fahlberg M, Haupt E, Kaur A, Marx PA, Ortega-Lopez J, Hotez PJ, Bottazzi ME. Safety and immunogenicity of a recombinant vaccine against Trypanosoma cruzi in Rhesus macaques. Vaccine 2020; 38:4584-4591. [PMID: 32417142 DOI: 10.1016/j.vaccine.2020.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 11/30/2022]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi is one of the most important neglected parasitic diseases in the Americas. Vaccines represent an attractive complementary strategy for the control of T. cruzi infection and pre-clinical studies in mice demonstrated that trypomastigote surface antigen (TSA-1) and the flagellar calcium-binding (Tc24) parasite antigens are promising candidates for vaccine development. We performed here the first evaluation of the safety and immunogenicity of two recombinant vaccine antigens (named TSA1-C4 and Tc24-C4) in naïve non-human primates. Three rhesus macaques received 3 doses of each recombinant protein, formulated with E6020 (Eisai Co., Ltd.), a novel Toll-like receptor-4 agonist, in a stable emulsion. All parameters from blood chemistry and blood cell counts were stable over the course of the study and unaffected by the vaccine. A specific IgG response against both antigens was detectable after the first vaccine dose, and increased with the second dose. After three vaccine doses, stimulation of PBMCs with a peptide pool derived from TSA1-C4 resulted in the induction of TSA1-C4-specific TNFα-, IL-2- and IFNγ-producing CD4+ in one or two animals while stimulation with a peptide pool derived from Tc24-C4 only activated IFNγ-producing CD4+T cells in one animal. In two animals there was also activation of TSA1-C4-specific IL2-producing CD8+ T cells. This is the first report of the immunogenicity of T. cruzi-derived recombinant antigens formulated as an emulsion with a TLR4 agonist in a non-human primate model. Our results strongly support the need for further evaluation of the preventive efficacy of this type of vaccine in non-human primates and explore the effect of the vaccine in a therapeutic model of naturally-infected Chagasic non-human primates, which would strengthen the rationale for the clinical development as a human vaccine against Chagas disease.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA.
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Weihong Tu
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Marissa Fahlberg
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Erin Haupt
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Preston A Marx
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Jaime Ortega-Lopez
- Departmento de Biotecnología y Bioingeniería, CINVESTAV, Mexico, D.F., Mexico
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Wang ZB, Xu J. Better Adjuvants for Better Vaccines: Progress in Adjuvant Delivery Systems, Modifications, and Adjuvant-Antigen Codelivery. Vaccines (Basel) 2020; 8:vaccines8010128. [PMID: 32183209 PMCID: PMC7157724 DOI: 10.3390/vaccines8010128] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Traditional aluminum adjuvants can trigger strong humoral immunity but weak cellular immunity, limiting their application in some vaccines. Currently, various immunomodulators and delivery carriers are used as adjuvants, and the mechanisms of action of some of these adjuvants are clear. However, customizing targets of adjuvant action (cellular or humoral immunity) and action intensity (enhancement or inhibition) according to different antigens selected is time-consuming. Here, we review the adjuvant effects of some delivery systems and immune stimulants. In addition, to improve the safety, effectiveness, and accessibility of adjuvants, new trends in adjuvant development and their modification strategies are discussed.
Collapse
Affiliation(s)
| | - Jing Xu
- Correspondence: ; Tel.: +86-(10)-5224-5008
| |
Collapse
|
20
|
Leitner GR, Wenzel TJ, Marshall N, Gates EJ, Klegeris A. Targeting toll-like receptor 4 to modulate neuroinflammation in central nervous system disorders. Expert Opin Ther Targets 2019; 23:865-882. [PMID: 31580163 DOI: 10.1080/14728222.2019.1676416] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Adverse immune activation contributes to many central nervous system (CNS) disorders. All main CNS cell types express toll-like receptor 4 (TLR 4). This receptor is critical for a myriad of immune functions such as cytokine secretion and phagocytic activity of microglia; however, imbalances in TLR 4 activation can contribute to the progression of neurodegenerative diseases. Areas covered: We considered available evidence implicating TLR 4 activation in the following CNS pathologies: Alzheimer's disease, Parkinson's disease, ischemic stroke, traumatic brain injury, multiple sclerosis, multiple systems atrophy, and Huntington's disease. We reviewed studies reporting effects of TLR 4-specific antagonists and agonists in models of peripheral and CNS diseases from the perspective of possible future use of TLR 4 ligands in CNS disorders. Expert opinion: TLR 4-specific antagonists could suppress neuroinflammation by reducing overproduction of inflammatory mediators; however, they may interfere with protein clearance mechanisms and myelination. Agonists that specifically activate myeloid differentiation primary-response protein 88 (MyD88)-independent pathway of TLR 4 signaling could facilitate beneficial glial phagocytic activity with limited activity as inducers of proinflammatory mediators. Deciphering the disease stage-specific involvement of TLR 4 in CNS pathologies is crucial for the future clinical development of TLR 4 agonists and antagonists.
Collapse
Affiliation(s)
- Gunnar R Leitner
- Department of Biology, University of British Columbia Okanagan Campus , Kelowna , British Columbia , Canada
| | - Tyler J Wenzel
- Department of Biology, University of British Columbia Okanagan Campus , Kelowna , British Columbia , Canada
| | - Nick Marshall
- Department of Biology, University of British Columbia Okanagan Campus , Kelowna , British Columbia , Canada
| | - Ellen J Gates
- Department of Biology, University of British Columbia Okanagan Campus , Kelowna , British Columbia , Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus , Kelowna , British Columbia , Canada
| |
Collapse
|
21
|
Psychopharmacology: neuroimmune signaling in psychiatric disease-developing vaccines against abused drugs using toll-like receptor agonists. Psychopharmacology (Berl) 2019; 236:2899-2907. [PMID: 30726515 DOI: 10.1007/s00213-019-5176-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/16/2019] [Indexed: 01/05/2023]
Abstract
RATIONALE Since substance use disorders have few or no effective pharmacotherapies, researchers have developed vaccines as immune-therapies against nicotine, cocaine, methamphetamine, and opioids including fentanyl. OBJECTIVES We focus on enhancing antibody (AB) production through stimulation of toll-like receptor-5 (TLR5) during active vaccination. The stimulating adjuvant is Entolimod, a novel protein derivative of flagellin. We review the molecular and cellular mechanisms underlying Entolimod's actions on TLR5. RESULTS Entolimod shows excellent efficacy for increasing AB levels to levels well beyond those produced by anti-addiction vaccines alone in animal models and humans. These ABs also significantly block the behavioral effects of the targeted drug of abuse. The TLR5 stimulation involves a wide range of immune cell types such as dendritic, antigen presenting, T and B cells. Entolimod binding to TLR5 initiates an intracellular signaling cascade that stimulates cytokine production of tumor necrosis factor and two interleukins (IL-6 and IL-12). While cytokine release can be catastrophic in cytokine storm, Entolimod produces a modulated release with few side effects even at doses 30 times greater than doses needed in these vaccine studies. Entolimod has markedly increased AB responses to all of our anti-addiction vaccines in rodent models, and in normal humans. CONCLUSIONS Entolimod and TLR5 stimulation has broad application to vaccines and potentially to other psychiatric disorders like depression, which has critical inflammatory contributions that Entolimod could reduce.
Collapse
|
22
|
Arora S, Ahmad S, Irshad R, Goyal Y, Rafat S, Siddiqui N, Dev K, Husain M, Ali S, Mohan A, Syed MA. TLRs in pulmonary diseases. Life Sci 2019; 233:116671. [PMID: 31336122 PMCID: PMC7094289 DOI: 10.1016/j.lfs.2019.116671] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
Abstract
Toll-like receptors (TLRs) comprise a clan of proteins involved in identification and triggering a suitable response against pathogenic attacks. As lung is steadily exposed to multiple infectious agents, antigens and host-derived danger signals, the inhabiting stromal and myeloid cells of the lung express an aggregate of TLRs which perceive the endogenously derived damage-associated molecular patterns (DAMPs) along with pathogen associated molecular patterns (PAMPs) and trigger the TLR-associated signalling events involved in host defence. Thus, they form an imperative component of host defence activation in case of microbial infections as well as non-infectious pulmonary disorders such as interstitial lung disease, acute lung injury and airways disease, such as COPD and asthma. They also play an equally important role in lung cancer. Targeting the TLR signalling network would pave ways to the design of more reliable and effective vaccines against infectious agents and control deadly infections, desensitize allergens and reduce inflammation. Moreover, TLR agonists may act as adjuvants by increasing the efficiency of cancer vaccines, thereby contributing their role in treatment of lung cancer too. Overall, TLRs present a compelling and expeditiously bolstered area of research and addressing their signalling events would be of significant use in pulmonary diseases.
Collapse
Affiliation(s)
- Shweta Arora
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shaniya Ahmad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Rasha Irshad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Yamini Goyal
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Sahar Rafat
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Neha Siddiqui
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Mohammad Husain
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.
| | - Anant Mohan
- Department of Pulmonary Medicine, AIIMS, New Delhi, India.
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
23
|
Rudicell RS, Garinot M, Kanekiyo M, Kamp HD, Swanson K, Chou TH, Dai S, Bedel O, Simard D, Gillespie RA, Yang K, Reardon M, Avila LZ, Besev M, Dhal PK, Dharanipragada R, Zheng L, Duan X, Dinapoli J, Vogel TU, Kleanthous H, Mascola JR, Graham BS, Haensler J, Wei CJ, Nabel GJ. Comparison of adjuvants to optimize influenza neutralizing antibody responses. Vaccine 2019; 37:6208-6220. [PMID: 31493950 DOI: 10.1016/j.vaccine.2019.08.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/26/2019] [Accepted: 08/17/2019] [Indexed: 12/14/2022]
Abstract
Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.
Collapse
Affiliation(s)
| | | | - Masaru Kanekiyo
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Rebecca A Gillespie
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
24
|
Yang J, Zhang H, Zhu Z, Gao Y, Xiang B, Wei Q. The immunostimulatory effects and pro-apoptotic activity of rhCNB against Lewis lung cancer is mediated by Toll-like receptor 4. Cancer Med 2019; 8:4441-4453. [PMID: 31218844 PMCID: PMC6675711 DOI: 10.1002/cam4.2158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Background Recombinant human calcineurin B subunit (rhCNB) has been shown to be an immune‐stimulatory protein promoting cytokine production and inducing phenotypic maturation of Dendritic cells (DCs). In vivo, it has good antitumor efficacy, and has potential as an antitumor drug. Exogenous rhCNB was found to be internalized into tumor cells via the Toll‐like receptor 4 (TLR4) complex, but it was not known whether its immuno‐modulatory and antitumor functions involved entry by this same route. Methods The production and secretion of the cytokines and chemokines in innate immune cells induced by rhCNB were determined by ELISA, and the expression of CD40, CD80, CD86, and MHCII was analyzed by FACs. Experimental Lewis lung cancer (LLC) model was prepared in C57 BL/6 wild‐type (WT) mice, TLR4−/− mice or their littermates by the inoculation of LLCs in their right armpit, and then administrated daily intraperitoneal injections (0.2 mL) of normal saline, rhCNB 20 mg/kg, and rhCNB 40 mg/kg, respectively. Results Recombinant human calcineurin B subunit promoted the production of antitumor cytokines by innate immune cells, and culture supernatants of rhCNB‐stimulated immune cells induced apoptosis of LLCs. In addition, rhCNB up‐regulated CD40, CD80, CD86, and MHCII expression in macrophages and DCs in TLR4+ cells but failed to do so in TLR4 deficient cells. rhCNB also induced the formation of CD4+ and CD8+T cells in splenocytes from WT mice, but not from TLR4‐deficient littermates. Intraperitoneal administration of WT C57BL/6 mice with rhCNB resulted in a 50% reduction in LLC tumor growth, but failed to inhibit tumor growth in TLR4−/− littermates. Conclusions The in vivo antitumor and immunomodulatory effects of rhCNB are mediated by the TLR4. This conclusion is important for the further understanding and development of rhCNB as an antitumor drug.
Collapse
Affiliation(s)
- Jinju Yang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China.,National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Hongwei Zhang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China
| | - Ziwei Zhu
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China
| | - Yadan Gao
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China
| | - Benqiong Xiang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China
| | - Qun Wei
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, P. R. China
| |
Collapse
|
25
|
Barry MA, Versteeg L, Wang Q, Pollet J, Zhan B, Gusovsky F, Bottazzi ME, Hotez PJ, Jones KM. A therapeutic vaccine prototype induces protective immunity and reduces cardiac fibrosis in a mouse model of chronic Trypanosoma cruzi infection. PLoS Negl Trop Dis 2019; 13:e0007413. [PMID: 31145733 PMCID: PMC6542517 DOI: 10.1371/journal.pntd.0007413] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/25/2019] [Indexed: 02/06/2023] Open
Abstract
Chagas disease, caused by the parasite Trypanosoma cruzi, develops into chronic Chagas’ cardiomyopathy in ~30% of infected individuals, characterized by conduction disorders, arrhythmias, heart failure, and even sudden cardiac death. Current anti-parasitic treatments are plagued by significant side effects and poor efficacy in the chronic phase of disease; thus, there is a pressing need for new treatment options. A therapeutic vaccine could bolster the protective TH1-mediated immune response, thereby slowing or halting the progression of chronic Chagas’ cardiomyopathy. Prior work in mice has demonstrated therapeutic efficacy of a Tc24 recombinant protein vaccine in the acute phase of Chagas disease. However, it is anticipated that humans will be vaccinated therapeutically when in the chronic phase of disease. This study investigates the therapeutic efficacy of a vaccine prototype containing recombinant protein Tc24, formulated with an emulsion containing the Toll-like receptor 4 agonist E6020 as an immunomodulatory adjuvant in a mouse model of chronic T. cruzi infection. Among outbred ICR mice vaccinated during chronic T. cruzi infection, there is a significant increase in the number of animals with undetectable systemic parasitemia (60% of vaccinated mice compared to 0% in the sham vaccine control group), and a two-fold reduction in cardiac fibrosis over the control group. The vaccinated mice produce a robust protective TH1-biased immune response to the vaccine, as demonstrated by a significant increase in antigen-specific IFNγ-production, the number of antigen-specific IFNγ-producing cells, and IgG2a antibody titers. Importantly, therapeutic vaccination significantly reduced cardiac fibrosis in chronically infected mice. This is a first study demonstrating therapeutic efficacy of the prototype Tc24 recombinant protein and E6020 stable emulsion vaccine against cardiac fibrosis in a mouse model of chronic T. cruzi infection. Chagas disease is a parasitic infection that can cause severe heart disease. Current treatments do not work well and have significant side effects. Because of this, the authors created a new vaccine prototype with the goal that it could be given to infected people to prevent Chagas-associated heart disease. The vaccine contains a manufactured protein identical to a protein in the parasite (called Tc24) as well as a component to help the body produce a protective immune response (a vaccine adjuvant called E6020). The vaccine would boost the body’s natural immune response to the parasite infection, reducing the number of parasites in the body, and protecting the heart. Frequently, people are not diagnosed until later in the infection, because the early (or acute) stage of disease can be mistaken for a common cold. Because of this, it is important to test the vaccine when given in the later (or chronic) stage of infection. The authors tested the vaccine in a mouse model of chronic T. cruzi infection and found that the vaccinated mice had lower levels of parasites in their body and less damage to their hearts. This research shows promising value of a therapeutic vaccine to prevent Chagas-associated heart disease in a mouse model, with the hope that the same effect could be found in humans one day.
Collapse
Affiliation(s)
- Meagan A. Barry
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, United States of America
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (MB); (KJ)
| | - Leroy Versteeg
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Qian Wang
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jeroen Pollet
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bin Zhan
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fabian Gusovsky
- Eisai, Inc., Eisai Inc, Andover, Massachusetts, United States of America
| | - Maria Elena Bottazzi
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Peter J. Hotez
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Kathryn M. Jones
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (MB); (KJ)
| |
Collapse
|
26
|
Human Toll-Like Receptor 4 (hTLR4): Structural and functional dynamics in cancer. Int J Biol Macromol 2019; 122:425-451. [DOI: 10.1016/j.ijbiomac.2018.10.142] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/10/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
|
27
|
Arora R, Haile CN, Kosten TA, Wu Y, Ramakrishnan M, Hawkins LD, Orson FM, Kosten TR. Preclinical efficacy of an anti-methamphetamine vaccine using E6020 adjuvant. Am J Addict 2019; 28:119-126. [PMID: 30701618 DOI: 10.1111/ajad.12867] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/02/2019] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Methamphetamine (MA) substance use disorder (SUD) does not have an efficacious pharmacotherapy. We developed a MA vaccine and investigated its potential to attenuate MA induced responses. METHODS We examined a novel adjuvant, E6020, a Toll-like receptor-4 (TLR-4) agonist combined with tetanus-toxoid conjugated to succinyl-methamphetamine (TT-SMA) adsorbed on aluminum hydroxide (alum). Adult BALB/c female mice received the vaccine and booster injections at weeks 0, 3, and 6. The efficacy of the vaccine was assessed by the level and affinity of anti-MA antibodies elicited, its ability to attenuate MA induced locomotor activation and its reduction in the amount of MA entering the brains of vaccinated mice. RESULTS The TT-SMA vaccine containing alum and E6020 adjuvant produced anti-MA antibodies with nanomolar affinities and showed threefold greater peak titer levels than without E6020 (700 vs 250 μg/ml). These antibodies significantly decreased MA-induced locomotor activation (p < .05), and reduced the brain (p < .005) MA levels following MA administration in actively immunized mice. CONCLUSIONS Thus, this anti-MA vaccine formulated with E6020 demonstrated effective functional protection against behavioral disruptions induced by MA. SCIENTIFIC SIGNIFICANCE Together, anti-MA vaccine showing a promising improvement in the efficacy of the vaccine that could be an effective candidate vaccine for methamphetamine use disorder (MUD). Furthermore, combinations of adjuvants may be a tool to design vaccines for MA dependence in humans. (Am J Addict 2019;XX:1-8).
Collapse
Affiliation(s)
- Reetakshi Arora
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Colin N Haile
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Therese A Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Yan Wu
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Muthu Ramakrishnan
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Frank M Orson
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Thomas R Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| |
Collapse
|
28
|
de la Cruz JJ, Villanueva-Lizama L, Dzul-Huchim V, Ramírez-Sierra MJ, Martinez-Vega P, Rosado-Vallado M, Ortega-Lopez J, Flores-Pucheta CI, Gillespie P, Zhan B, Bottazzi ME, Hotez PJ, Dumonteil E. Production of recombinant TSA-1 and evaluation of its potential for the immuno-therapeutic control of Trypanosoma cruzi infection in mice. Hum Vaccin Immunother 2018; 15:210-219. [PMID: 30192702 PMCID: PMC6363145 DOI: 10.1080/21645515.2018.1520581] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A therapeutic vaccine for human Chagas disease (American trypanosomiasis caused by Trypanosoma cruzi) is under development based on the success of vaccinating mice with DNA constructs expressing the antigens Tc24 and Tc-TSA-1. However, because DNA and nucleic acid vaccines produce less than optimal responses in humans, our strategy relies on administering a recombinant protein-based vaccine, together with adjuvants that promote Th1-type immunity. Here we describe a process for the purification and refolding of recombinant TSA-1 expressed in Escherichia coli. The overall yield (20–25%) and endotoxin level of the purified recombinant TSA-1 (rTSA-1) is suitable for pilot scale production of the antigen for use in phase 1 clinical trials. Mice infected with T. cruzi were treated with rTSA-1, either alone or with Toll-like receptor 4 (TLR-4) agonist adjuvants including monophosphoryl lipid A (MPLA), glucopyranosyl lipid A (GLA, IDRI), and E6020 (EISEI, Inc). TSA-1 with the TLR-4 agonists was effective at reducing parasitemia relative to rTSA-1 alone, although it was difficult to discern a therapeutic effect compared to treatment with TLR-4 agonists alone. However, rTSA-1 with a 10 ug dose of MPLA optimized reductions in cardiac tissue inflammation, which were significantly reduced compared to MPLA alone. It also elicited the lowest parasite burden and the highest levels of TSA-1-specific IFN-gamma levels and IFN-gamma/IL-4 ratios. These results warrant the further evaluation of rTSA-1 in combination with rTc24 in order to maximize the therapeutic effect of vaccine-linked chemotherapy in both mice and non-human primates before advancing to clinical development.
Collapse
Affiliation(s)
- Juan Jose de la Cruz
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - Liliana Villanueva-Lizama
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - Victor Dzul-Huchim
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - María-Jesus Ramírez-Sierra
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - Pedro Martinez-Vega
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - Miguel Rosado-Vallado
- a Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi , Universidad Autónoma de Yucatán , Mérida , Yucatán , México
| | - Jaime Ortega-Lopez
- b Departamento de Biotecnología y Bioingeniería , CINVESTAV-IPN , Ciudad de México , México
| | | | - Portia Gillespie
- c Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology , National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA
| | - Bin Zhan
- c Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology , National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA
| | - Maria Elena Bottazzi
- c Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology , National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA
| | - Peter J Hotez
- c Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology , National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA
| | - Eric Dumonteil
- d Department of Tropical Medicine , Vector-Borne and Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University , New Orleans , LA , USA
| |
Collapse
|
29
|
Pinheiro RO, Schmitz V, Silva BJDA, Dias AA, de Souza BJ, de Mattos Barbosa MG, de Almeida Esquenazi D, Pessolani MCV, Sarno EN. Innate Immune Responses in Leprosy. Front Immunol 2018; 9:518. [PMID: 29643852 PMCID: PMC5882777 DOI: 10.3389/fimmu.2018.00518] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/27/2018] [Indexed: 12/20/2022] Open
Abstract
Leprosy is an infectious disease that may present different clinical forms depending on host immune response to Mycobacterium leprae. Several studies have clarified the role of various T cell populations in leprosy; however, recent evidences suggest that local innate immune mechanisms are key determinants in driving the disease to its different clinical manifestations. Leprosy is an ideal model to study the immunoregulatory role of innate immune molecules and its interaction with nervous system, which can affect homeostasis and contribute to the development of inflammatory episodes during the course of the disease. Macrophages, dendritic cells, neutrophils, and keratinocytes are the major cell populations studied and the comprehension of the complex networking created by cytokine release, lipid and iron metabolism, as well as antimicrobial effector pathways might provide data that will help in the development of new strategies for leprosy management.
Collapse
Affiliation(s)
- Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Veronica Schmitz
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - André Alves Dias
- Cellular Microbiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | | | | | | | - Euzenir Nunes Sarno
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Scott BA, Yarchoan M, Jaffee EM. Prophylactic Vaccines for Nonviral Cancers. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Blake Alan Scott
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;, ,
- Cellular and Molecular Medicine Program, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;, ,
| | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;, ,
- Cellular and Molecular Medicine Program, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
31
|
Zamyatina A. Aminosugar-based immunomodulator lipid A: synthetic approaches. Beilstein J Org Chem 2018; 14:25-53. [PMID: 29379577 PMCID: PMC5769089 DOI: 10.3762/bjoc.14.3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
The immediate immune response to infection by Gram-negative bacteria depends on the structure of a lipopolysaccharide (LPS, also known as endotoxin), a complex glycolipid constituting the outer leaflet of the bacterial outer membrane. Recognition of picomolar quantities of pathogenic LPS by the germ-line encoded Toll-like Receptor 4 (TLR4) complex triggers the intracellular pro-inflammatory signaling cascade leading to the expression of cytokines, chemokines, prostaglandins and reactive oxygen species which manifest an acute inflammatory response to infection. The "endotoxic principle" of LPS resides in its amphiphilic membrane-bound fragment glycophospholipid lipid A which directly binds to the TLR4·MD-2 receptor complex. The lipid A content of LPS comprises a complex mixture of structural homologs varying in the acylation pattern, the length of the (R)-3-hydroxyacyl- and (R)-3-acyloxyacyl long-chain residues and in the phosphorylation status of the β(1→6)-linked diglucosamine backbone. The structural heterogeneity of the lipid A isolates obtained from bacterial cultures as well as possible contamination with other pro-inflammatory bacterial components makes it difficult to obtain unambiguous immunobiological data correlating specific structural features of lipid A with its endotoxic activity. Advanced understanding of the therapeutic significance of the TLR4-mediated modulation of the innate immune signaling and the central role of lipid A in the recognition of LPS by the innate immune system has led to a demand for well-defined materials for biological studies. Since effective synthetic chemistry is a prerequisite for the availability of homogeneous structurally distinct lipid A, the development of divergent and reproducible approaches for the synthesis of various types of lipid A has become a subject of considerable importance. This review focuses on recent advances in synthetic methodologies toward LPS substructures comprising lipid A and describes the synthesis and immunobiological properties of representative lipid A variants corresponding to different bacterial species. The main criteria for the choice of orthogonal protecting groups for hydroxyl and amino functions of synthetically assembled β(1→6)-linked diglucosamine backbone of lipid A which allows for a stepwise introduction of multiple functional groups into the molecule are discussed. Thorough consideration is also given to the synthesis of 1,1'-glycosyl phosphodiesters comprising partial structures of 4-amino-4-deoxy-β-L-arabinose modified Burkholderia lipid A and galactosamine-modified Francisella lipid A. Particular emphasis is put on the stereoselective construction of binary glycosyl phosphodiester fragments connecting the anomeric centers of two aminosugars as well as on the advanced P(III)-phosphorus chemistry behind the assembly of zwitterionic double glycosyl phosphodiesters.
Collapse
Affiliation(s)
- Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
32
|
Innate transcriptional effects by adjuvants on the magnitude, quality, and durability of HIV envelope responses in NHPs. Blood Adv 2017; 1:2329-2342. [PMID: 29296883 DOI: 10.1182/bloodadvances.2017011411] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/01/2017] [Indexed: 12/15/2022] Open
Abstract
Adjuvants have a critical role for improving vaccine efficacy against many pathogens, including HIV. Here, using transcriptional RNA profiling and systems serology, we assessed how distinct innate pathways altered HIV-specific antibody responses in nonhuman primates (NHPs) using 8 clinically based adjuvants. NHPs were immunized with a glycoprotein 140 HIV envelope protein (Env) and insoluble aluminum salts (alum), MF59, or adjuvant nanoemulsion (ANE) coformulated with or without Toll-like receptor 4 (TLR4) and 7 agonists. These were compared with Env administered with polyinosinic-polycytidylic acid:poly-L-lysine, carboxymethylcellulose (pIC:LC) or immune-stimulating complexes. Addition of the TLR4 agonist to alum enhanced upregulation of a set of inflammatory genes, whereas the TLR7 agonist suppressed expression of alum-responsive inflammatory genes and enhanced upregulation of antiviral and interferon (IFN) genes. Moreover, coformulation of the TLR4 or 7 agonists with alum boosted Env-binding titers approximately threefold to 10-fold compared with alum alone, but remarkably did not alter gene expression or enhance antibody titers when formulated with ANE. The hierarchy of adjuvant potency was established after the second of 4 immunizations. In terms of antibody durability, antibody titers decreased ∼10-fold after the final immunization and then remained stable after 65 weeks for all adjuvants. Last, Env-specific Fc-domain glycan structures and a series of antibody effector functions were assessed by systems serology. Antiviral/IFN gene signatures correlated with Fc-receptor binding across all adjuvant groups. This study defines the potency and durability of 8 different clinically based adjuvants in NHPs and shows how specific innate pathways can alter qualitative aspects of Env antibody function.
Collapse
|
33
|
Pollet J, Versteeg L, Rezende W, Strych U, Gusovsky F, Hotez PJ, Bottazzi ME. A simple fluorescence-based assay for quantification of the Toll-Like Receptor agonist E6020 in vaccine formulations. Vaccine 2017; 35:1410-1416. [PMID: 28190745 DOI: 10.1016/j.vaccine.2017.01.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 11/16/2022]
Abstract
Despite the generally accepted immunostimulatory effect of Toll-Like Receptor 4 (TLR4) agonists and their value as vaccine adjuvants, there remains a demand for fast and easy quantification assays for these TLR4 agonists in order to accelerate and improve vaccine formulation studies. A new medium-throughput method was developed for the quantification of the TLR4 agonist, E6020, independent of the formulation composition. The assay uses a fluorescent hydrazide (DCCH) to label the synthetic lipopolysaccharide (LPS) analog E6020 through its diketone groups. This novel, low-cost, and fluorescence based assay may obviate the need for traditional approaches that primarily rely on Fourier transform infrared spectroscopy (FTIR) or mass spectrometry. The experiments were performed in a wide diversity of vaccine formulations containing E6020 to assess method robustness and accuracy. The assay was also expanded to evaluate the loading efficiency of E6020 in poly(lactic-co-glycolic acid) (PLGA) micro-particles.
Collapse
Affiliation(s)
- Jeroen Pollet
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA
| | - Leroy Versteeg
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA
| | - Wanderson Rezende
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA
| | - Ulrich Strych
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA
| | | | - Peter J Hotez
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA; Department of Biology, Baylor University, Waco, TX, USA
| | - Maria Elena Bottazzi
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Houston, TX 77030, USA; Department of Biology, Baylor University, Waco, TX, USA.
| |
Collapse
|
34
|
Church JS, Milich LM, Lerch JK, Popovich PG, McTigue DM. E6020, a synthetic TLR4 agonist, accelerates myelin debris clearance, Schwann cell infiltration, and remyelination in the rat spinal cord. Glia 2017; 65:883-899. [PMID: 28251686 DOI: 10.1002/glia.23132] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 12/26/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are present throughout the adult brain and spinal cord and can replace oligodendrocytes lost to injury, aging, or disease. Their differentiation, however, is inhibited by myelin debris, making clearance of this debris an important step for cellular repair following demyelination. In models of peripheral nerve injury, TLR4 activation by lipopolysaccharide (LPS) promotes macrophage phagocytosis of debris. Here we tested whether the novel synthetic TLR4 agonist E6020, a Lipid A mimetic, promotes myelin debris clearance and remyelination in spinal cord white matter following lysolecithin-induced demyelination. In vitro, E6020 induced TLR4-dependent cytokine expression (TNFα, IL1β, IL-6) and NF-κB signaling, albeit at ∼10-fold reduced potency compared to LPS. Microinjection of E6020 into the intact rat spinal cord gray/white matter border induced macrophage activation, OPC proliferation, and robust oligodendrogenesis, similar to what we described previously using an intraspinal LPS microinjection model. Finally, a single co-injection of E6020 with lysolecithin into spinal cord white matter increased axon sparing, accelerated myelin debris clearance, enhanced Schwann cell infiltration into demyelinated lesions, and increased the number of remyelinated axons. In vitro assays confirmed that direct stimulation of macrophages by E6020 stimulates myelin phagocytosis. These data implicate TLR4 signaling in promoting repair after CNS demyelination, likely by stimulating phagocytic activity of macrophages, sparing axons, recruiting myelinating cells, and promoting remyelination. This work furthers our understanding of immune-myelin interactions and identifies a novel synthetic TLR4 agonist as a potential therapeutic avenue for white matter demyelinating conditions such as spinal cord injury and multiple sclerosis.
Collapse
Affiliation(s)
- Jamie S Church
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Lindsay M Milich
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Jessica K Lerch
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA.,Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA.,Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Dana M McTigue
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA.,Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
35
|
Marshall JD, Heeke DS, Rao E, Maynard SK, Hornigold D, McCrae C, Fraser N, Tovchigrechko A, Yu L, Williams N, King S, Cooper ME, Hajjar AM, Woo JC. A Novel Class of Small Molecule Agonists with Preference for Human over Mouse TLR4 Activation. PLoS One 2016; 11:e0164632. [PMID: 27736941 PMCID: PMC5063506 DOI: 10.1371/journal.pone.0164632] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/28/2016] [Indexed: 01/09/2023] Open
Abstract
The best-characterized Toll-like receptor 4 (TLR4) ligands are lipopolysaccharide (LPS) and its chemically modified and detoxified variant, monophosphoryl lipid A (MPL). Although both molecules are active for human TLR4, they demonstrate a potency preference for mouse TLR4 based on data from transfected cell lines and primary cells of both species. After a high throughput screening process of small molecule libraries, we have discovered a new class of TLR4 agonist with a species preference profile differing from MPL. Products of the 4-component Ugi synthesis reaction were demonstrated to potently trigger human TLR4-transfected HEK cells but not mouse TLR4, although inclusion of the human MD2 with mTLR4 was able to partially recover activity. Co-expression of CD14 was not required for optimal activity of Ugi compounds on transfected cells, as it is for LPS. The species preference profile for the panel of Ugi compounds was found to be strongly active for human and cynomolgus monkey primary cells, with reduced but still substantial activity for most Ugi compounds on guinea pig cells. Mouse, rat, rabbit, ferret, and cotton rat cells displayed little or no activity when exposed to Ugi compounds. However, engineering the human versions of TLR4 and MD2 to be expressed in mTLR4/MD2 deficient mice allowed for robust activity by Ugi compounds both in vitro and in vivo. These findings extend the range of compounds available for development as agonists of TLR4 and identify novel molecules which reverse the TLR4 triggering preference of MPL for mouse TLR4 over human TLR4. Such compounds may be amenable to formulation as more potent human-specific TLR4L-based adjuvants than typical MPL-based adjuvants.
Collapse
Affiliation(s)
- Jason D. Marshall
- Vaccine Platform Group, MedImmune, Gaithersburg, Maryland, United States of America
- * E-mail:
| | - Darren S. Heeke
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| | - Eileen Rao
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| | - Sean K. Maynard
- Vaccine Platform Group, MedImmune, Gaithersburg, Maryland, United States of America
| | - David Hornigold
- Department of Cardiovascular and Metabolic Diseases, MedImmune, Cambridge, United Kingdom
| | - Christopher McCrae
- Translational Science, Respiratory, Inflammation and Autoimmunity Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Neil Fraser
- Biology Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Andrey Tovchigrechko
- Research Bioinformatics, MedImmune, Gaithersburg, Maryland, United States of America
| | - Li Yu
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, United States of America
| | - Nicola Williams
- Biology Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Sarah King
- Medicinal Chemistry Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Martin E. Cooper
- Translational Science, Respiratory, Inflammation and Autoimmunity Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jennifer C. Woo
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| |
Collapse
|
36
|
Wu TYH. Strategies for designing synthetic immune agonists. Immunology 2016; 148:315-25. [PMID: 27213842 DOI: 10.1111/imm.12622] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 12/16/2022] Open
Abstract
Enhancing the immune system is a validated strategy to combat infectious disease, cancer and allergy. Nevertheless, the development of immune adjuvants has been hampered by safety concerns. Agents that can stimulate the immune system often bear structural similarities with pathogen-associated molecular patterns found in bacteria or viruses and are recognized by pattern recognition receptors (PRRs). Activation of these PRRs results in the immediate release of inflammatory cytokines, up-regulation of co-stimulatory molecules, and recruitment of innate immune cells. The distribution and duration of these early inflammatory events are crucial in the development of antigen-specific adaptive immunity in the forms of antibody and/or T cells capable of searching for and destroying the infectious pathogens or cancer cells. However, systemic activation of these PRRs is often poorly tolerated. Hence, different strategies have been employed to modify or deliver immune agonists in an attempt to control the early innate receptor activation through temporal or spatial restriction. These approaches include physicochemical manipulation, covalent conjugation, formulation and conditional activation/deactivation. This review will describe recent examples of discovery and optimization of synthetic immune agonists towards clinical application.
Collapse
|
37
|
Visan L, Sanchez V, Kania M, de Montfort A, de la Maza LM, Ausar SF. Phosphate substitution in an AlOOH - TLR4 adjuvant system (SPA08) modulates the immunogenicity of Serovar E MOMP from Chlamydia trachomatis. Hum Vaccin Immunother 2016; 12:2341-50. [PMID: 27104338 DOI: 10.1080/21645515.2016.1168958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Chlamydia trachomatis is one of the most common sexually transmitted pathogens and the development of an effective vaccine is highly desirable. The Major Outer Membrane Protein (MOMP) is one of the most abundant and immunogenic chlamydial proteins. Here we investigated the effects of phosphate substitution on the physicochemical and immunochemical properties of an experimental vaccine composed of serovar E recombinant MOMP (rMOMP) and a proprietary adjuvant system SPA08, consisting of aluminum oxyhydroxide (AlOOH) containing the TLR4 agonist E6020. An increase in phosphate substitution in the AlOOH component of the adjuvant markedly decreased the adsorptive coefficient and adsorptive capacity for both Ser E rMOMP and E6020. In vaccine formulations used for immunizations, phosphate substitution induced a decrease in the % adsorption of Ser E rMOMP without affecting the % adsorption of E6020. Immunogenicity studies in CD1 mice showed that an increase in phosphate substitution of the SPA08 adjuvant resulted in an increase in Ser E rMOMP-specific serum total IgG and IgG1 but not IgG2a titers. The degree of phosphate substitution in SPA08 also significantly increased in vitro neutralization concomitant with a decrease in proinflammatory cytokines secreted by Ser E rMOMP-restimulated splenocytes. Taken together, the results of these studies suggest that the degree of phosphate substitution in AlOOH greatly affects the adsorption of E6020 and Ser E rMOMP to AlOOH resulting in significant effects on vaccine-induced cellular and humoral responses.
Collapse
Affiliation(s)
- Lucian Visan
- a Research & Non Clinical Safety Department , Sanofi Pasteur , Marcy l'Etoile , France
| | - Violette Sanchez
- a Research & Non Clinical Safety Department , Sanofi Pasteur , Marcy l'Etoile , France
| | - Margaux Kania
- b Bioprocess Research and Development, Sanofi Pasteur , Toronto , ON , Canada
| | - Aymeric de Montfort
- a Research & Non Clinical Safety Department , Sanofi Pasteur , Marcy l'Etoile , France
| | - Luis M de la Maza
- c Department of Pathology and Laboratory Medicine , Medical Sciences I, University of California , Irvine , CA , USA
| | | |
Collapse
|
38
|
Haensler J, Probeck P, Su J, Piras F, Dalençon F, Cotte JF, Chambon V, Iqbal SM, Hawkins L, Burdin N. Design and preclinical characterization of a novel vaccine adjuvant formulation consisting of a synthetic TLR4 agonist in a thermoreversible squalene emulsion. Int J Pharm 2015; 486:99-111. [PMID: 25794609 DOI: 10.1016/j.ijpharm.2015.03.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/22/2022]
Abstract
We describe the development, analytical characterization, stability and preclinical efficacy of AF04, a combination adjuvant comprising the synthetic toll-like receptor 4 (TLR4) agonist, E6020, formulated in AF03, a thermoreversible squalene emulsion. By using AF04 with the recombinant major outer membrane protein of Chlamydia trachomatis (Ct-MOMP) and with the recombinant surface glycoprotein gB from human cytomegalovirus (CMV-gB) as model antigens, we show that AF03 and E6020 can synergize to augment specific antibody and Th-1 cellular immune responses in mice. In terms of formulation, we observe that the method used to incorporate E6020 into AF03 affects its partition between the oil and water phases of the emulsion which in turn has a significant impact on the tolerability (IV pyrogenicity test in rabbits) of this novel adjuvant combination.
Collapse
Affiliation(s)
| | | | - Jin Su
- Sanofi Pasteur R&D, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Khalaf JK, Bowen WS, Bazin HG, Ryter KT, Livesay MT, Ward JR, Evans JT, Johnson DA. Characterization of TRIF selectivity in the AGP class of lipid A mimetics: role of secondary lipid chains. Bioorg Med Chem Lett 2014; 25:547-53. [PMID: 25553892 DOI: 10.1016/j.bmcl.2014.12.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
TLR4 agonists that favor TRIF-dependent signaling and the induction of type 1 interferons may have potential as vaccine adjuvants with reduced toxicity. CRX-547 (4), a member of the aminoalkyl glucosaminide 4-phosphate (AGP) class of lipid A mimetics possessing three (R)-3-decanoyloxytetradecanoyl groups and d-relative configuration in the aglycon, selectively reduces MyD88-dependent signaling resulting in TRIF-selective signaling, whereas the corresponding secondary ether lipid 6a containing (R)-3-decyloxytetradecanoyl groups does not. In order to determine which secondary acyl groups are important for the reduction in MyD88-dependent signaling activity of 4, the six possible ester/ether hybrid derivatives of 4 and 6a were synthesized and evaluated for their ability to induce NF-κB in a HEK293 cell reporter assay. An (R)-3-decanoyloxytetradecanoyl group on the 3-position of the d-glucosamine unit was found to be indispensable for maintaining low NF-κB activity irrespective of the substitutions (decyl or decanoyl) on the other two secondary positions. These results suggest that the carbonyl group of the 3-secondary lipid chain may impede homodimerization and/or conformational changes in the TLR4-MD2 complex necessary for MyD88 binding and pro-inflammatory cytokine induction.
Collapse
Affiliation(s)
- Juhienah K Khalaf
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - William S Bowen
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Donald E. Baxter Biomedical Research Building, 570 South Preston Street, Louisville, KY 40202, USA
| | - Hélène G Bazin
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Kendal T Ryter
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Mark T Livesay
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Jon R Ward
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Jay T Evans
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - David A Johnson
- GlaxoSmithKline Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA.
| |
Collapse
|
40
|
Adanitsch F, Ittig S, Stöckl J, Oblak A, Haegman M, Jerala R, Beyaert R, Kosma P, Zamyatina A. Development of αGlcN(1↔1)αMan-based lipid A mimetics as a novel class of potent Toll-like receptor 4 agonists. J Med Chem 2014; 57:8056-71. [PMID: 25252784 PMCID: PMC4191062 DOI: 10.1021/jm500946r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
The endotoxic portion of lipopolysaccharide
(LPS), a glycophospholipid
Lipid A, initiates the activation of the Toll-like Receptor 4 (TLR4)–myeloid
differentiation factor 2 (MD-2) complex, which results in pro-inflammatory
immune signaling. To unveil the structural requirements for TLR4·MD-2-specific
ligands, we have developed conformationally restricted Lipid A mimetics
wherein the flexible βGlcN(1→6)GlcN backbone of Lipid
A is exchanged for a rigid trehalose-like αGlcN(1↔1)αMan scaffold
resembling the molecular shape of TLR4·MD-2-bound E.
coli Lipid A disclosed in the X-ray structure. A convergent
synthetic route toward orthogonally protected αGlcN(1↔1)αMan
disaccharide has been elaborated. The α,α-(1↔1)
linkage was attained by the glycosylation of 2-N-carbamate-protected
α-GlcN-lactol with N-phenyl-trifluoroacetimidate
of 2-O-methylated mannose. Regioselective acylation
with (R)-3-acyloxyacyl fatty acids and successive
phosphorylation followed by global deprotection afforded bis- and
monophosphorylated hexaacylated Lipid A mimetics. αGlcN(1↔1)αMan-based
Lipid A mimetics (α,α-GM-LAM) induced potent activation
of NF-κB signaling in hTLR4/hMD-2/CD14-transfected HEK293 cells
and robust LPS-like cytokines expression in macrophages and dendritic
cells. Thus, restricting the conformational flexibility of Lipid A
by fixing the molecular shape of its carbohydrate backbone in the
“agonistic” conformation attained by a rigid αGlcN(1↔1)αMan scaffold
represents
an efficient approach toward powerful and adjustable TLR4 activation.
Collapse
Affiliation(s)
- Florian Adanitsch
- Department of Chemistry, University of Natural Resources and Life Sciences , Muthgasse 18, A-1190 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Awasthi S. Toll-like receptor-4 modulation for cancer immunotherapy. Front Immunol 2014; 5:328. [PMID: 25120541 PMCID: PMC4110442 DOI: 10.3389/fimmu.2014.00328] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022] Open
Affiliation(s)
- Shanjana Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| |
Collapse
|
42
|
Hayashi T, Crain B, Yao S, Caneda CD, Cottam HB, Chan M, Corr M, Carson DA. Novel synthetic toll-like receptor 4/MD2 ligands attenuate sterile inflammation. J Pharmacol Exp Ther 2014; 350:330-40. [PMID: 24893985 DOI: 10.1124/jpet.114.214312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Toll-like receptor (TLR) stimulation has been implicated as a major contributor to chronic inflammation. Among these receptors, TLR4 has been described as a key regulator of endogenous inflammation and has been proposed as a therapeutic target. Previously, we discovered by high-throughput screening a group of substituted pyrimido[5,4-b]indoles that activated a nuclear factor-κB reporter in THP-1 human monocytic cells. A biologically active hit compound was resynthesized, and derivatives were prepared to assess structure-activity relationships. The derived compounds activated cells in a TLR4/myeloid differentiation protein 2 (MD2)-dependent and CD14-independent manner, using the myeloid differentiation primary response 88 and Toll/IL-1 receptor domain-containing adapter-inducing interferon-β pathways. Two lead compounds, 1Z105 and 1Z88, were selected for further analysis based on favorable biologic properties and lack of toxicity. In vivo pharmacokinetics indicated that 1Z105 was orally bioavailable, whereas 1Z88 was not. Oral or parenteral doses of 1Z105 and 1Z88 induced undetectable or negligible levels of circulating cytokines and did not induce hepatotoxicity when administered to galactosamine-conditioned mice, indicating good safety profiles. Both compounds were very effective in preventing lethal liver damage in lipopolysaccharide treated galatosamine-conditioned mice. Orally administered 1Z105 and parenteral 1Z88 prevented arthritis in an autoantibody-driven murine model. Hence, these low molecular weight molecules that target TLR4/MD2 were well tolerated and effective in reducing target organ damage in two different mouse models of sterile inflammation.
Collapse
Affiliation(s)
- Tomoko Hayashi
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Brian Crain
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Shiyin Yao
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Christa D Caneda
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Howard B Cottam
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Michael Chan
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Maripat Corr
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Dennis A Carson
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| |
Collapse
|
43
|
Psychostimulant addiction treatment. Neuropharmacology 2014; 87:150-60. [PMID: 24727297 DOI: 10.1016/j.neuropharm.2014.04.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/14/2014] [Accepted: 04/01/2014] [Indexed: 01/29/2023]
Abstract
Treatment of psychostimulant addiction has been a major, and not fully met, challenge. For opioid addiction, there is strong evidence for the effectiveness of several medications. For psychostimulants, there is no corresponding form of agonist maintenance that has met criteria for regulatory approval or generally accepted use. Stimulant-use disorders remain prevalent and can result in both short-term and long-term adverse consequences. The mainstay of treatment remains behavioral interventions. In this paper, we discuss those interventions and some promising candidates in the search for pharmacological interventions. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
44
|
Ireton GC, Reed SG. Adjuvants containing natural and synthetic Toll-like receptor 4 ligands. Expert Rev Vaccines 2014; 12:793-807. [PMID: 23885824 DOI: 10.1586/14760584.2013.811204] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The last decade has seen an increased focus on the development of adjuvants for vaccines, and several novel adjuvants are now in licensed products or in late-stage clinical development. These advancements have been aided by the discovery of receptors and signaling pathways of the innate immune system and an increased understanding of how these innate responses influence the adaptive immune response. Successful vaccine development relies on knowledge of which adjuvants to use and the proper formulation of adjuvants and antigens to achieve safe, stable and immunogenic vaccines. In this review, the authors focus on the current use of natural and synthetic lipopolysaccharide analogues that retain their adjuvant properties with reduced toxicity compared with the parent compound for use in emerging vaccines. The authors review how these compounds initiate signal transduction through Toll-like receptor 4, insights from structure-function studies and how formulation parameters can influence their effectiveness as vaccine adjuvants.
Collapse
Affiliation(s)
- Gregory C Ireton
- Infectious Disease Research Institute, 1124 Columbia St., Ste 400, Seattle, WA 98104, USA
| | | |
Collapse
|
45
|
|
46
|
Han JE, Wui SR, Kim KS, Cho YJ, Cho WJ, Lee NG. Characterization of the structure and immunostimulatory activity of a vaccine adjuvant, de-O-acylated lipooligosaccharide. PLoS One 2014; 9:e85838. [PMID: 24465739 PMCID: PMC3899070 DOI: 10.1371/journal.pone.0085838] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria. LPS elicits strong immunopathological responses during bacterial infection, and the lipid A moiety of LPS is responsible for this immunostimulatory activity. Lipid A exerts its biological activity by sending signals via TLR4 present on immune cells, and TLR4 agonists have been a target for vaccine adjuvant. Previously, we demonstrated an adjuvant activity of deacylated lipooligosaccharide (dLOS) to viral and bacterial antigens. In this study, we characterized the chemical structure of dLOS and evaluated its immunostimulatory activity on mouse and human immune cells in comparison with monophosphoryl lipid A (MPL). dLOS consists of a core oligosaccharide lacking the terminal glucose residue, a glucosamine disaccharide with two phosphate groups, and two N-linked acyl groups. dLOS was similar to MPL in induction of cytokine production in mouse peritoneal macrophages, but was a more potent activator in human monocytes and dendritic cells (DCs). Results of an analysis of allogeneic T cell responses revealed that dLOS induces Th1, Th2, and Th17-type immune responses in a dose-dependent manner. The immunostimulatory activities of dLOS were completely abrogated in TLR4−/− mice, which confirms its TLR4-dependency. These results suggest that in the presence of the core oligosaccharide, O-linked acyl groups of LPS are dispensable for activating the TLR4 signaling pathway. dLOS did not cause any pathological effects or death at 0.25, 0.5, or 1 mg per kg body weight in mice in the acute toxicity tests. This result suggests that dLOS has a low toxicity. dLOS should be considered for further development as a safe and effective adjuvant for human vaccines.
Collapse
MESH Headings
- Acylation
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/pharmacology
- Animals
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/blood
- Cytokines/immunology
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dose-Response Relationship, Drug
- Female
- Flow Cytometry
- Humans
- Immunoblotting
- Lipopolysaccharides/chemistry
- Lipopolysaccharides/immunology
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Structure
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Vaccines/immunology
Collapse
Affiliation(s)
- Ji Eun Han
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Seo Ri Wui
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Kwang Sung Kim
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Yang Je Cho
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Wan Je Cho
- Yonsei University Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Na Gyong Lee
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
47
|
Sanders H, Feavers IM. Adjuvant properties of meningococcal outer membrane vesicles and the use of adjuvants inNeisseria meningitidisprotein vaccines. Expert Rev Vaccines 2014; 10:323-34. [DOI: 10.1586/erv.11.10] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
48
|
Abstract
Vaccines are being developed against substance abuse and most progress has been made with anti-cocaine, nicotine and opiate vaccines, but new ones are being developed for methamphetamine and may be in humans within 18 - 24 months. These haptenated vaccines share a common problem in that only about one-third of those vaccinated get a sufficiently robust antibody titer to enable them to effectively block drug use. This problem is being addressed with better carrier proteins and new adjuvants beyond alum. This review provides details about these developing vaccines that act through pharmacokinetic rather than pharmacodynamics blockade. Due to this pharmacokinetic mechanism of keeping abused drugs in the bloodstream and not allowing them entry into the brain or other organs, these vaccines have very few side effects compared to other blockers used in addictions treatment.
Collapse
Affiliation(s)
- Thomas R Kosten
- Michael E DeBakey VA Medical Center, Baylor College of Medicine, Departments of Psychiatry, Neuroscience, Pharmacology & Immunology, 2002 Holcombe, VA Hospital Bldg 110, # 229 Houston, TX 77030, USA.
| | | |
Collapse
|
49
|
Singh M, Kazzaz J, Ugozzoli M, Baudner B, Pizza M, Giuliani M, Hawkins LD, Otten G, O'Hagan DT. MF59 oil-in-water emulsion in combination with a synthetic TLR4 agonist (E6020) is a potent adjuvant for a combination Meningococcus vaccine. Hum Vaccin Immunother 2013; 8:486-90. [PMID: 22832252 DOI: 10.4161/hv.19229] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The inclusion of a potent TLR4 immune potentiator to a recombinant antigen vaccine formulation enhances both the magnitude and the breadth of the engendered immune response. One such immune potentiator (TLR4 agonist E6020) was evaluated with recombinant Men B antigens delivered in MF59 sub-micron adjuvant emulsion. The ability of this formulation to enhance serum antibody and bactercidal titers was investigated. The co-delivery of E6020 within MF59 enhanced both the serum and bactericidal titers for Men B antigens and for Men B antigens combined with Men ACWY-CRM conjugate vaccine. The delivery of TLR4 agonist within MF59 emulsion oil droplets leads to a more potent response in comparison to the TLR4 when admixed with MF59 emulsion.
Collapse
|
50
|
Dumonteil E, Bottazzi ME, Zhan B, Heffernan MJ, Jones K, Valenzuela JG, Kamhawi S, Ortega J, de Leon Rosales SP, Lee BY, Bacon KM, Fleischer B, Slingsby BT, Cravioto MB, Tapia-Conyer R, Hotez PJ. Accelerating the development of a therapeutic vaccine for human Chagas disease: rationale and prospects. Expert Rev Vaccines 2012; 11:1043-55. [PMID: 23151163 PMCID: PMC3819810 DOI: 10.1586/erv.12.85] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chagas disease is a leading cause of heart disease affecting approximately 10 million people in Latin America and elsewhere worldwide. The two major drugs available for the treatment of Chagas disease have limited efficacy in Trypanosoma cruzi-infected adults with indeterminate (patients who have seroconverted but do not yet show signs or symptoms) and determinate (patients who have both seroconverted and have clinical disease) status; they require prolonged treatment courses and are poorly tolerated and expensive. As an alternative to chemotherapy, an injectable therapeutic Chagas disease vaccine is under development to prevent or delay Chagasic cardiomyopathy in patients with indeterminate or determinate status. The bivalent vaccine will be comprised of two recombinant T. cruzi antigens, Tc24 and TSA-1, formulated on alum together with the Toll-like receptor 4 agonist, E6020. Proof-of-concept for the efficacy of these antigens was obtained in preclinical testing at the Autonomous University of Yucatan. Here the authors discuss the potential for a therapeutic Chagas vaccine as well as the progress made towards such a vaccine, and the authors articulate a roadmap for the development of the vaccine as planned by the nonprofit Sabin Vaccine Institute Product Development Partnership and Texas Children's Hospital Center for Vaccine Development in collaboration with an international consortium of academic and industrial partners in Mexico, Germany, Japan, and the USA.
Collapse
Affiliation(s)
- Eric Dumonteil
- Laboratorio de Parasitología Centro De Investigaciones Regional, “Dr. Hideo Noguchi” Autonomous University of Yucatan (UADY), Merida, Mexico
| | - Maria Elena Bottazzi
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Bin Zhan
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael J Heffernan
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn Jones
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jaime Ortega
- Departamento de Biotecnología y Bioingeniería, Centro de Investigacion y de Estudios Avanzados - Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Bruce Y Lee
- Public Health Computational and Operations Research (PHICOR), University of Pittsburgh, Pittsburgh PA, USA
| | - Kristina M Bacon
- Public Health Computational and Operations Research (PHICOR), University of Pittsburgh, Pittsburgh PA, USA
| | | | | | | | | | - Peter J Hotez
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|