1
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [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] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
2
|
Du JJ, Zhou SH, Cheng ZR, Xu WB, Zhang RY, Wang LS, Guo J. MUC1 Specific Immune Responses Enhanced by Coadministration of Liposomal DDA/MPLA and Lipoglycopeptide. Front Chem 2022; 10:814880. [PMID: 35186882 PMCID: PMC8854779 DOI: 10.3389/fchem.2022.814880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Mucin 1 (MUC1), a well-known tumor-associated antigen and attractive target for tumor immunotherapy, is overexpressed in most human epithelial adenomas with aberrant glycosylation. However, its low immunogenicity impedes the development of MUC1-targeted antitumor vaccines. In this study, we investigated three liposomal adjuvant systems containing toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) and auxiliary lipids of different charges: cationic lipid dimethyldioctadecylammonium (DDA), neutral lipid distearoylglycerophosphocholine (DSPC) or anionic lipid dioleoylphosphatidylglycerol (DOPG), respectively. ELISA assay evidenced that the positively charged DDA/MPLA liposomes are potent immune activators, which induced remarkable levels of anti-MUC1 antibodies and exhibited robust Th1-biased immune responses. Importantly, the antibodies induced by DDA/MPLA liposomes efficiently recognized and killed MUC1-positive tumor cells through complement-mediated cytotoxicity. In addition, antibody titers in mice immunized with P2-MUC1 vaccine were significantly higher than those from mice immunized with P1-MUC1 or MUC1 vaccine, which indicated that the lipid conjugated on MUC1 antigen also played important role for immunomodulation. This study suggested that the liposomal DDA/MPLA with lipid-MUC1 is a promising antitumor vaccine, which can be used for the immunotherapy of various epithelial carcinomas represented by breast cancer.
Collapse
Affiliation(s)
- Jing-Jing Du
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi, China
| | - Shi-Hao Zhou
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Zi-Ru Cheng
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi, China
| | - Wen-Bo Xu
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Long-Sheng Wang
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, China
- *Correspondence: Long-Sheng Wang, ; Jun Guo,
| | - Jun Guo
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
- *Correspondence: Long-Sheng Wang, ; Jun Guo,
| |
Collapse
|
3
|
The Critical Role of Toll-like Receptor-mediated Signaling in Cancer Immunotherapy. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
4
|
Kingstad-Bakke B, Toy R, Lee W, Pradhan P, Vogel G, Marinaik CB, Larsen A, Gates D, Luu T, Pandey B, Kawaoka Y, Roy K, Suresh M. Polymeric Pathogen-Like Particles-Based Combination Adjuvants Elicit Potent Mucosal T Cell Immunity to Influenza A Virus. Front Immunol 2021; 11:559382. [PMID: 33767689 PMCID: PMC7986715 DOI: 10.3389/fimmu.2020.559382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Eliciting durable and protective T cell-mediated immunity in the respiratory mucosa remains a significant challenge. Polylactic-co-glycolic acid (PLGA)-based cationic pathogen-like particles (PLPs) loaded with TLR agonists mimic biophysical properties of microbes and hence, simulate pathogen-pattern recognition receptor interactions to safely and effectively stimulate innate immune responses. We generated micro particle PLPs loaded with TLR4 (glucopyranosyl lipid adjuvant, GLA) or TLR9 (CpG) agonists, and formulated them with and without a mucosal delivery enhancing carbomer-based nanoemulsion adjuvant (ADJ). These adjuvants delivered intranasally to mice elicited high numbers of influenza nucleoprotein (NP)-specific CD8+ and CD4+ effector and tissue-resident memory T cells (TRMs) in lungs and airways. PLPs delivering TLR4 versus TLR9 agonists drove phenotypically and functionally distinct populations of effector and memory T cells. While PLPs loaded with CpG or GLA provided immunity, combining the adjuvanticity of PLP-GLA and ADJ markedly enhanced the development of airway and lung TRMs and CD4 and CD8 T cell-dependent immunity to influenza virus. Further, balanced CD8 (Tc1/Tc17) and CD4 (Th1/Th17) recall responses were linked to effective influenza virus control. These studies provide mechanistic insights into vaccine-induced pulmonary T cell immunity and pave the way for the development of a universal influenza and SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Randall Toy
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Pallab Pradhan
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Gabriela Vogel
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Chandranaik B Marinaik
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Autumn Larsen
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Daisy Gates
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Tracy Luu
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Bhawana Pandey
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Yoshihoro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
5
|
Birmpilis AI, Karachaliou CE, Samara P, Ioannou K, Selemenakis P, Kostopoulos IV, Kavrochorianou N, Kalbacher H, Livaniou E, Haralambous S, Kotsinas A, Farzaneh F, Trougakos IP, Voelter W, Dimopoulos MA, Bamias A, Tsitsilonis O. Antitumor Reactive T-Cell Responses Are Enhanced In Vivo by DAMP Prothymosin Alpha and Its C-Terminal Decapeptide. Cancers (Basel) 2019; 11:cancers11111764. [PMID: 31717548 PMCID: PMC6896021 DOI: 10.3390/cancers11111764] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Prothymosin α (proTα) and its C-terminal decapeptide proTα(100-109) were shown to pleiotropically enhance innate and adaptive immune responses. Their activities have been broadly studied in vitro, focusing primarily on the restoration of the deficient immunoreactivity of cancer patients' leukocytes. Previously, we showed that proTα and proTα(100-109) act as danger-associated molecular patterns (DAMPs), ligate Toll-like receptor-4, signal through TRIF- and MyD88-dependent pathways, promote the maturation of dendritic cells and elicit T-helper type 1 (Th1) immune responses in vitro, leading to the optimal priming of tumor antigen-reactive T-cell functions. Herein, we assessed their activity in a preclinical melanoma model. Immunocompetent mice bearing B16.F1 tumors were treated with two cycles of proTα or proTα(100-109) together with a B16.F1-derived peptide vaccine. Coadministration of proTα or proTα(100-109) and the peptide vaccine suppressed melanoma-cell proliferation, as evidenced by reduced tumor-growth rates. Higher melanoma infiltration by CD3+ T cells was observed, whereas ex vivo analysis of mouse total spleen cells verified the in vivo induction of melanoma-reactive cytotoxic responses. Additionally, increased levels of proinflammatory and Th1-type cytokines were detected in mouse serum. We propose that, in the presence of tumor antigens, DAMPs proTα and proTα(100-109) induce Th1-biased immune responses in vivo. Their adjuvant ability to orchestrate antitumor immunoreactivities can eventually be exploited therapeutically in humans.
Collapse
Affiliation(s)
- Anastasios I. Birmpilis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Pinelopi Samara
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Kyriaki Ioannou
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Platon Selemenakis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Ioannis V. Kostopoulos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Nadia Kavrochorianou
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Sylva Haralambous
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Athanasios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Farzin Farzaneh
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Ioannis P. Trougakos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Ourania Tsitsilonis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- Correspondence: ; Tel.: +30-210-727-4215; Fax: +30-210-727-4635
| |
Collapse
|
6
|
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
|
7
|
Zom GG, Willems MMJHP, Meeuwenoord NJ, Reintjens NRM, Tondini E, Khan S, Overkleeft HS, van der Marel GA, Codee JDC, Ossendorp F, Filippov DV. Dual Synthetic Peptide Conjugate Vaccine Simultaneously Triggers TLR2 and NOD2 and Activates Human Dendritic Cells. Bioconjug Chem 2019; 30:1150-1161. [PMID: 30865430 DOI: 10.1021/acs.bioconjchem.9b00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Simultaneous triggering of Toll-like receptors (TLRs) and NOD-like receptors (NLRs) has previously been shown to synergistically activate monocytes, dendritic cells, and macrophages. We applied these properties in a T-cell vaccine setting by conjugating the NOD2-ligand muramyl-dipeptide (MDP) and TLR2-ligand Pam3CSK4 to a synthetic peptide derived from a model antigen. Stimulation of human DCs with the MDP-peptide-Pam3CSK4 conjugate led to a strongly increased secretion of pro-inflammatory and Th1-type cytokines and chemokines. We further show that the conjugated ligands retain their ability to trigger their respective receptors, while even improving NOD2-triggering. Also, activation of murine DCs was enhanced by the dual triggering, ultimately leading to effective induction of vaccine-specific T cells expressing IFNγ, IL-2, and TNFα. Together, these data indicate that the dual MDP-SLP-Pam3CSK4 conjugate constitutes a chemically well-defined vaccine approach that holds promise for the use in the treatment of virus infections and cancer.
Collapse
Affiliation(s)
- Gijs G Zom
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Marian M J H P Willems
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Nico J Meeuwenoord
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Niels R M Reintjens
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Selina Khan
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Herman S Overkleeft
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Gijsbert A van der Marel
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Jeroen D C Codee
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , P.O. Box 9600, 2300 RC Leiden , The Netherlands
| | - Dmitri V Filippov
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2333 CC Leiden , The Netherlands
| |
Collapse
|
8
|
Traini G, Ruiz-de-Angulo A, Blanco-Canosa JB, Zamacola Bascarán K, Molinaro A, Silipo A, Escors D, Mareque-Rivas JC. Cancer Immunotherapy of TLR4 Agonist-Antigen Constructs Enhanced with Pathogen-Mimicking Magnetite Nanoparticles and Checkpoint Blockade of PD-L1. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1803993. [PMID: 30569516 DOI: 10.1002/smll.201803993] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/07/2018] [Indexed: 05/10/2023]
Abstract
Despite the tremendous potential of Toll-like receptor 4 (TLR4) agonists in vaccines, their efficacy as monotherapy to treat cancer has been limited. Only some lipopolysaccharides (LPS) isolated from particular bacterial strains or structures like monophosphoryl lipid A (MPLA) derived from lipooligosaccharide (LOS), avoid toxic overactivation of innate immune responses while retaining adequate immunogenicity to act as adjuvants. Here, different LOS structures are incorporated into nanoparticle-filled phospholipid micelles for efficient vaccine delivery and more potent cancer immunotherapy. The structurally unique LOS of the plant pathogen Xcc is incorporated into phospholipid micelles encapsulating iron oxide nanoparticles, producing stable pathogen-mimicking nanostructures suitable for targeting antigen presenting cells in the lymph nodes. The antigen is conjugated via a hydrazone bond, enabling rapid, easy-to-monitor and high-yield antigen ligation at low concentrations. The protective effect of these constructs is investigated against a highly aggressive model for tumor immunotherapy. The results show that the nanovaccines lead to a higher-level antigen-specific cytotoxic T lymphocyte (CTL) effector and memory responses, which when combined with abrogation of the immunosuppressive programmed death-ligand 1 (PD-L1), provide 100% long-term protection against repeated tumor challenge. This nanovaccine platform in combination with checkpoint inhibition of PD-L1 represents a promising approach to improve the cancer immunotherapy of TLR4 agonists.
Collapse
Affiliation(s)
- Giordano Traini
- CIC biomaGUNE, Paseo Miramón 182, 20014, San Sebastián, Spain
| | | | | | | | - Antonio Molinaro
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte Sant' Angelo, Via Cintia 4, 80126, Napoli, Italy
| | - Alba Silipo
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte Sant' Angelo, Via Cintia 4, 80126, Napoli, Italy
| | - David Escors
- Navarrabiomed-Biomedical Research Centre, Fundación Miguel Servet-IdISNA, Complejo Hospitalario de Navarra, 31008, Pamplona, Spain
| | - Juan C Mareque-Rivas
- Department of Chemistry and Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| |
Collapse
|
9
|
Brooks N, Hsu J, Esparon S, Pouniotis D, Pietersz GA. Immunogenicity of a Tripartite Cell Penetrating Peptide Containing a MUC1 Variable Number of Tandem Repeat (VNTR) and A T Helper Epitope. Molecules 2018; 23:molecules23092233. [PMID: 30200528 PMCID: PMC6225367 DOI: 10.3390/molecules23092233] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
Peptide-based vaccines for cancer have many advantages however, for optimization these immunogens should incorporate peptide epitopes that induce CD8, as well as CD4 responses, antibody and long term immunity. Cell penetrating peptides (CPP) with a capacity of cytosolic delivery have been used to deliver antigenic peptides and proteins to antigen presenting cells to induce cytotoxic T cell, helper T cell and humoral responses in mice. For this study, a tripartite CPP including a mucin 1 (MUC1) variable number of tandem repeat (VNTR) containing multiple T cell epitopes and tetanus toxoid universal T helper epitope peptide (tetCD4) was synthesised (AntpMAPMUC1tet) and immune responses investigated in mice. Mice vaccinated with AntpMAPMUC1tet + CpG show enhanced antigen-specific interferon-gamma (IFN-γ) and IL-4 T cell responses compared with AntpMAPMUC1tet vaccination alone and induced a Th1 response, characterised by a higher ratio of IgG2a antibody/IgG1 antibodies. Furthermore, vaccination generated long term MUC1-specific antibody and T cell responses and delayed growth of MUC1+ve tumours in mice. This data demonstrates the efficient delivery of branched multiple antigen peptides incorporating CPP and that the addition of CpG augments immune responses.
Collapse
Affiliation(s)
- Nicole Brooks
- School of Medical Sciences, RMIT University, Plenty Road, Bundoora 3083, Victoria, Australia.
| | - Jennifer Hsu
- Bio-Organic and Medicinal Chemistry Laboratory, Burnet Institute, 85 Commercial Rd, Melbourne 3004, Australia.
- Dendritic Cell Biology and Therapeutics Group, ANZAC Medical Research Institute, Institute of Haematology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, NSW 2050, Australia.
| | - Sandra Esparon
- Bio-Organic and Medicinal Chemistry Laboratory, Burnet Institute, 85 Commercial Rd, Melbourne 3004, Australia.
| | - Dodie Pouniotis
- School of Medical Sciences, RMIT University, Plenty Road, Bundoora 3083, Victoria, Australia.
| | - Geoffrey A Pietersz
- Bio-Organic and Medicinal Chemistry Laboratory, Burnet Institute, 85 Commercial Rd, Melbourne 3004, Australia.
- Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia.
- Department of Immunology, Monash University, Clayton, Victoria 3800, Australia.
- Baker Heart and Diabetes Institute, Melbourne 3004, Australia.
- College of Health and Biomedicine, Victoria University, Melbourne 3021, Australia.
| |
Collapse
|
10
|
McCormick AA, Shakeel A, Yi C, Kaur H, Mansour AM, Bakshi CS. Intranasal administration of a two-dose adjuvanted multi-antigen TMV-subunit conjugate vaccine fully protects mice against Francisella tularensis LVS challenge. PLoS One 2018; 13:e0194614. [PMID: 29684046 PMCID: PMC5912714 DOI: 10.1371/journal.pone.0194614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/06/2018] [Indexed: 12/03/2022] Open
Abstract
Tularemia is a fatal human disease caused by Francisella tularensis, a Gram-negative encapsulated coccobacillus bacterium. Due to its low infectious dose, ease of aerosolized transmission, and lethal effects, the CDC lists F. tularensis as a Category A pathogen, the highest level for a potential biothreat agent. Previous vaccine studies have been conducted with live attenuated, inactivated, and subunit vaccines, which have achieved partial or full protection from F. tularensis live vaccine strain (LVS) challenge, but no vaccine has been approved for human use. We demonstrate the improved efficacy of a multi-antigen subunit vaccine by using Tobacco Mosaic virus (TMV) as an antigen carrier for the F. tularensis SchuS4 proteins DnaK, OmpA, SucB and Tul4 (DOST). The magnitude and quality of immune responses were compared after mice were immunized by subcutaneous or intranasal routes of administration with a TMV-DOST mixture, with or without four different adjuvants. Immune responses varied in magnitude and isotype profile, by antigen, by route of administration, and by protection in an F. tularensis LVS challenge model of disease. Interestingly, our analysis demonstrates an overwhelming IgG2 response to SucB after intranasal dosing, as well as a robust cellular response, which may account for the improved two-dose survival imparted by the tetravalent vaccine, compared to a previous study that tested efficacy of TMV-DOT. Our study provides evidence that potent humoral, cellular and mucosal immunity can be achieved by optimal antigen combination, delivery, adjuvant and appropriate route of administration, to improve vaccine potency and provide protection from pathogen challenge.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/analysis
- Antibodies, Bacterial/metabolism
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Bacterial Vaccines/immunology
- Disease Models, Animal
- Female
- Francisella tularensis/immunology
- Immunity, Cellular
- Immunoglobulin G/analysis
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Immunoglobulin Isotypes/immunology
- Immunoglobulin Isotypes/metabolism
- Mice
- Mice, Inbred C57BL
- Survival Rate
- Tobacco Mosaic Virus/genetics
- Tobacco Mosaic Virus/metabolism
- Tularemia/immunology
- Tularemia/microbiology
- Tularemia/prevention & control
- Vaccines, Conjugate/immunology
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
| | - Aisha Shakeel
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Chris Yi
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Hardeep Kaur
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Ahd M. Mansour
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
11
|
Ibrahim M, Scozzi D, Toth KA, Ponti D, Kreisel D, Menna C, De Falco E, D'Andrilli A, Rendina EA, Calogero A, Krupnick AS, Gelman AE. Naive CD4 + T Cells Carrying a TLR2 Agonist Overcome TGF-β-Mediated Tumor Immune Evasion. THE JOURNAL OF IMMUNOLOGY 2017; 200:847-856. [PMID: 29212908 DOI: 10.4049/jimmunol.1700396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 11/01/2017] [Indexed: 01/09/2023]
Abstract
TLR agonists are effective at treating superficial cancerous lesions, but their use internally for other types of tumors remains challenging because of toxicity. In this article, we report that murine and human naive CD4+ T cells that sequester Pam3Cys4 (CD4+ TPam3) become primed for Th1 differentiation. CD4+ TPam3 cells encoding the OVA-specific TCR OT2, when transferred into mice bearing established TGF-β-OVA-expressing thymomas, produce high amounts of IFN-γ and sensitize tumors to PD-1/programmed cell death ligand 1 blockade-induced rejection. In contrast, naive OT2 cells without Pam3Cys4 cargo are prone to TGF-β-dependent inducible regulatory Foxp3+ CD4+ T cell conversion and accelerate tumor growth that is largely unaffected by PD-1/programmed cell death ligand 1 blockade. Ex vivo analysis reveals that CD4+ TPam3 cells are resistant to TGF-β-mediated gene expression through Akt activation controlled by inputs from the TCR and a TLR2-MyD88-dependent PI3K signaling pathway. These data show that CD4+ TPam3 cells are capable of Th1 differentiation in the presence of TGF-β, suggesting a novel approach to adoptive cell therapy.
Collapse
Affiliation(s)
- Mohsen Ibrahim
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63108.,Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Davide Scozzi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63108.,Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Kelsey A Toth
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63108
| | - Donatella Ponti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Rome, Italy
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63108.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63108; and
| | - Cecilia Menna
- Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Rome, Italy
| | - Antonio D'Andrilli
- Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Erino A Rendina
- Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Rome, Italy
| | - Alexander S Krupnick
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22098
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63108; .,Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63108; and
| |
Collapse
|
12
|
Waki K, Yamada A. Blockade of high mobility group box 1 augments antitumor T-cell response induced by peptide vaccination as a co-adjuvant. Cancer Sci 2016; 107:1721-1729. [PMID: 27717108 PMCID: PMC5198958 DOI: 10.1111/cas.13084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a member of the family of damage-associated molecular patterns, which cause inflammation and trigger innate immunity through Toll-like receptors 2/4 and the receptor for advanced glycation end products. We examined the effect of glycyrrhizin, a selective inhibitor of HMGB1, on the induction of CTLs in mice. B6 mice, either OT-1 spleen cell-transferred or untransferred, were immunized with an s.c. injection of OVA257-264 peptide with topical imiquimod, and glycyrrhizin was mixed with the antigen peptide. Proliferation of OT-1 cells after immunization was enhanced by glycyrrhizin. The effect of glycyrrhizin was confirmed in other adjuvant systems, such as CpG oligonucleotide and monophosphoryl lipid A, but glycyrrhizin was not effective in Freund's incomplete adjuvant system. The augmenting effects of glycyrrhizin were also observed in other synthetic HMGB1 inhibitors, gabexate mesilate, nafamostat, and sivelestat. Thus, the effects are common to the HMGB1 inhibitors. Induction of CTLs detected by γ-interferon enzyme-linked immunospot assay was similarly augmented by glycyrrhizin. In a therapeutic vaccine model, glycyrrhizin inhibited the growth of s.c. transplanted EG.7 tumors. Expression of inflammatory cytokines in the skin inoculation site was downregulated by glycyrrhizin. These results suggest that HMGB1 inhibitors might be useful as a co-adjuvant for peptide vaccination with an innate immunity receptor-related adjuvant.
Collapse
Affiliation(s)
- Kayoko Waki
- Cancer Vaccine Development Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Akira Yamada
- Cancer Vaccine Development Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| |
Collapse
|
13
|
Liu L, Yi H, Wang C, He H, Li P, Pan H, Sheng N, Ji M, Cai L, Ma Y. Integrated Nanovaccine with MicroRNA-148a Inhibition Reprograms Tumor-Associated Dendritic Cells by Modulating miR-148a/DNMT1/SOCS1 Axis. THE JOURNAL OF IMMUNOLOGY 2016; 197:1231-41. [DOI: 10.4049/jimmunol.1600182] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/08/2016] [Indexed: 12/26/2022]
|
14
|
Elster JD, Krishnadas DK, Lucas KG. Dendritic cell vaccines: A review of recent developments and their potential pediatric application. Hum Vaccin Immunother 2016; 12:2232-9. [PMID: 27245943 DOI: 10.1080/21645515.2016.1179844] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
For many cancers the use of conventional chemotherapy has been maximized, and further intensification of chemotherapy generally results in excess toxicity with little long-term benefit for cure. Many tumors become resistant to chemotherapy, making the investigation of novel approaches such as immunotherapy of interest. Because the tumor microenvironment is known to promote immune tolerance and down regulate the body's natural defense mechanisms, modulating the immune system with the use of dendritic cell (DC) therapy is an attractive approach. Thousands of patients with diverse tumor types have been treated with DC vaccines. While antigen specific immune responses have been reported, the duration and magnitude of these responses are typically weak, and objective clinical responses have been limited. DC vaccine generation and administration is a multi-step process with opportunities for improvement in source of DC for vaccine, selection of target antigen, and boosting effector cell response via administration of vaccine adjuvant or concomitant pharmacologic immunomodulation. In this review we will discuss recent developments in each of these areas and highlight elements that could be moved into pediatric clinical trials.
Collapse
Affiliation(s)
- Jennifer D Elster
- a Department of Pediatrics , Hematology/Oncology, University of Louisville , Louisville , KY , USA
| | - Deepa K Krishnadas
- a Department of Pediatrics , Hematology/Oncology, University of Louisville , Louisville , KY , USA
| | - Kenneth G Lucas
- a Department of Pediatrics , Hematology/Oncology, University of Louisville , Louisville , KY , USA
| |
Collapse
|
15
|
Schölch S, Rauber C, Tietz A, Rahbari NN, Bork U, Schmidt T, Kahlert C, Haberkorn U, Tomai MA, Lipson KE, Carretero R, Weitz J, Koch M, Huber PE. Radiotherapy combined with TLR7/8 activation induces strong immune responses against gastrointestinal tumors. Oncotarget 2016; 6:4663-76. [PMID: 25609199 PMCID: PMC4467106 DOI: 10.18632/oncotarget.3081] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 12/26/2014] [Indexed: 12/28/2022] Open
Abstract
In addition to local cytotoxic activity, radiotherapy may also elicit local and systemic antitumor immunity, which may be augmented by immunotherapeutic agents including Toll-like receptor (TLR) 7/8 agonists. Here, we investigated the ability of 3M-011 (854A), a TLR7/8 agonist, to boost the antigen-presenting activity of dendritic cells (DC) as an adjuvant to radiotherapy. The combined treatment induced marked local and systemic responses in subcutaneous and orthotopic mouse models of colorectal and pancreatic cancer. In vitro cytotoxicity assays as well as in vivo depletion experiments with monoclonal antibodies identified NK and CD8 T cells as the cell populations mediating the cytotoxic effects of the treatment, while in vivo depletion of CD11c+ dendritic cells (DC) in CD11c-DTR transgenic mice revealed DC as the pivotal immune hub in this setting. The specificity of the immune reaction was confirmed by ELISPOT assays. TLR7/8 agonists therefore seem to be potent adjuvants to radiotherapy, inducing strong local and profound systemic immune responses to tumor antigens released by conventional therapy.
Collapse
Affiliation(s)
- Sebastian Schölch
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,CCU Molecular and Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Center, 69120 Heidelberg, Germany
| | - Conrad Rauber
- Department of General, Gastrointestinal and Transplant Surgery, University Hospital Heidelberg, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany.,CCU Molecular and Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Center, 69120 Heidelberg, Germany
| | - Alexandra Tietz
- CCU Molecular and Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Center, 69120 Heidelberg, Germany
| | - Nuh N Rahbari
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ulrich Bork
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Thomas Schmidt
- Department of General, Gastrointestinal and Transplant Surgery, University Hospital Heidelberg, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Kahlert
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Uwe Haberkorn
- Division of Nuclear Medicine, Department of Radiology, University Hospital Heidelberg, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | | | | | - Rafael Carretero
- Division of Molecular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Jürgen Weitz
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Moritz Koch
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Peter E Huber
- CCU Molecular and Radiation Oncology, German Cancer Research Center, 69120 Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Center, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Liang TJ, Block TM, McMahon BJ, Ghany MG, Urban S, Guo JT, Locarnini S, Zoulim F, Chang KM, Lok AS. Present and future therapies of hepatitis B: From discovery to cure. Hepatology 2015; 62:1893-908. [PMID: 26239691 PMCID: PMC4681668 DOI: 10.1002/hep.28025] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 07/31/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatitis B virus (HBV) is a significant global pathogen, infecting more than 240 million people worldwide. While treatment for HBV has improved, HBV patients often require lifelong therapies and cure is still a challenging goal. Recent advances in technologies and pharmaceutical sciences have heralded a new horizon of innovative therapeutic approaches that are bringing us closer to the possibility of a functional cure of chronic HBV infection. In this article, we review the current state of science in HBV therapy and highlight new and exciting therapeutic strategies spurred by recent scientific advances. Some of these therapies have already entered into clinical phase, and we will likely see more of them moving along the development pipeline. CONCLUSION With growing interest in developing and efforts to develop more effective therapies for HBV, the challenging goal of a cure may be well within reach in the near future.
Collapse
Affiliation(s)
- T. Jake Liang
- Liver Diseases Branch, NIDDK, NIH, Bethesda, MD. USA
| | | | - Brian J. McMahon
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Anchorage, AK. USA
| | - Marc G. Ghany
- Liver Diseases Branch, NIDDK, NIH, Bethesda, MD. USA
| | - Stephan Urban
- Dept of Infectious Diseases, Molecular Virology and German Center for Infection Diseases (DZIF), Univ Hospital Heidelberg, Heidelberg, Germany
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Doylestown, PA. USA
| | | | - Fabien Zoulim
- Victorian Infectious Diseases Reference Laboratory, Doherty Institute, Melbourne, VIC, Australia
| | - Kyong-Mi Chang
- Dept of Medicine, Philadelphia VAMC & University of Pennsylvania, Philadelphia, PA. USA
| | - Anna S. Lok
- Div of Gastroenterology and Hepatology, Univ of Michigan, Ann Arbor, MI. USA
| |
Collapse
|
17
|
Fitton JH, Stringer DN, Karpiniec SS. Therapies from Fucoidan: An Update. Mar Drugs 2015; 13:5920-46. [PMID: 26389927 PMCID: PMC4584361 DOI: 10.3390/md13095920] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/30/2022] Open
Abstract
Fucoidans are a class of sulfated fucose-rich polysaccharides found in brown marine algae and echinoderms. Fucoidans have an attractive array of bioactivities and potential applications including immune modulation, cancer inhibition, and pathogen inhibition. Research into fucoidan has continued to gain pace over the last few years and point towards potential therapeutic or adjunct roles. The source, extraction, characterization and detection of fucoidan is discussed.
Collapse
Affiliation(s)
- Janet Helen Fitton
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Damien N Stringer
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Samuel S Karpiniec
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| |
Collapse
|
18
|
Wang XD, Gao NN, Diao YW, Liu Y, Gao D, Li W, Wan YY, Zhong JJ, Jin GY. Conjugation of toll-like receptor-7 agonist to gastric cancer antigen MG7-Ag exerts antitumor effects. World J Gastroenterol 2015; 21:8052-8060. [PMID: 26185376 PMCID: PMC4499347 DOI: 10.3748/wjg.v21.i26.8052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/01/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of our tumor vaccines on reversing immune tolerance and generating therapeutic response.
METHODS: Vaccines were synthesized by solid phase using an Fmoc strategy, where a small molecule toll-like receptor-7 agonist (T7) was conjugated to a monoclonal gastric cancer 7 antigen mono-epitope (T7-MG1) or tri-epitope (T7-MG3). Cytokines were measured in both mouse bone marrow dendritic cells and mouse spleen lymphocytes after exposed to the vaccines. BALB/c mice were intraperitoneally immunized with the vaccines every 2 wk for a total of three times, and then subcutaneously challenged with Ehrlich ascites carcinoma (EAC) cells. Three weeks later, the mice were killed, and the tumors were surgically removed and weighed. Serum samples were collected from the mice, and antibody titers were determined by ELISA using an alkaline phosphate-conjugated detection antibody for total IgG. Antibody-dependent cell-mediated cytotoxicity was detected by the lactate dehydrogenase method using natural killer cells as effectors and antibody-labeled EAC cells as targets. Cytotoxic T lymphocyte activities were also detected by the lactate dehydrogenase method using lymphocytes as effectors and EAC cells as targets.
RESULTS: Vaccines were successfully synthesized and validated by analytical high performance liquid chromatography and electrospray mass spectrometry, including T7, T7-MG1, and T7-MG3. Rapid inductions of tumor necrosis factor-α and interleukin-12 in bone marrow dendritic cells and interferon γ and interleukin-12 in lymphocytes occurred in vitro after T7, T7-MG1, and T7-MG3 treatment. Immunization with T7-MG3 reduced the EAC tumor burden in BALB/c mice to 62.64% ± 5.55% compared with PBS control (P < 0.01). Six or nine weeks after the first immunization, the monoclonal gastric cancer 7 antigen antibody increased significantly in the T7-MG3 group compared with the PBS control (P < 0.01). As for antibody-dependent cell-mediated cytotoxicity, antisera obtained by immunization with T7-MG3 were able to markedly enhance cell lysis compared to PBS control (31.58% ± 2.94% vs 18.02% ± 2.26%; P < 0.01). As for cytotoxic T lymphocytes, T7-MG3 exhibited obviously greater cytotoxicity compared with PBS control (40.92% ± 4.38% vs 16.29% ± 1.90%; P < 0.01).
CONCLUSION: A successful method is confirmed for the design of gastric cancer vaccines by chemical conjugation of T7 and multi-repeat-epitope of monoclonal gastric cancer 7 antigen.
Collapse
MESH Headings
- Animals
- Antibody-Dependent Cell Cytotoxicity
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/chemical synthesis
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Carcinoma, Ehrlich Tumor/drug therapy
- Carcinoma, Ehrlich Tumor/immunology
- Carcinoma, Ehrlich Tumor/pathology
- Cells, Cultured
- Cytokines/metabolism
- Epitopes
- Female
- Immunization Schedule
- Immunoconjugates/administration & dosage
- Immunoconjugates/pharmacology
- Injections, Intraperitoneal
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Membrane Glycoproteins/agonists
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred BALB C
- Signal Transduction/drug effects
- Superantigens
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Time Factors
- Toll-Like Receptor 7/agonists
- Toll-Like Receptor 7/immunology
- Toll-Like Receptor 7/metabolism
- Tumor Burden
- Tumor Escape/drug effects
Collapse
|
19
|
Abstract
Newer immunotherapy agents may break the barrier that tumors create to evade the attack from the immune system. Dendritic cell vaccination has shown encouraging clinical activity and a favorable safety profile in advanced tumor stages. However, optimal cell maturation status, choice of tumor antigens and route of administration have not been established. Single or multiple peptides derived from tumor-associated antigens may also be used for cancer vaccination. Intratumoral delivery of oncolytic viruses expressing immunostimulating cytokines like GM-CSF have produced stimulating clinical results that need further verification. But it is probably T-cell checkpoint modulation with monoclonal antibodies that has attracted the highest expectations. Promising activity has been reported for tremelimumab, a CTLA-4 inhibitor, and a clinical trial testing the PD-1 antibody nivolumab is underway. Future progress will probably come from a better understanding of the mechanisms of cancer-related immunosuppression, improvement in agents and strategies and combination of the available therapeutic tools.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain.,Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain
| | - Daniel Palmer
- The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK.,The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - Ignacio Melero
- Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain.,Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain
| |
Collapse
|
20
|
Luo Z, Wang C, Yi H, Li P, Pan H, Liu L, Cai L, Ma Y. Nanovaccine loaded with poly I:C and STAT3 siRNA robustly elicits anti-tumor immune responses through modulating tumor-associated dendritic cells in vivo. Biomaterials 2014; 38:50-60. [PMID: 25457983 DOI: 10.1016/j.biomaterials.2014.10.050] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/18/2014] [Indexed: 12/16/2022]
Abstract
Although cancer vaccine-based immunotherapy holds great potential for cancer treatment, tumor-induced dendritic cell (DC) dysfunction remains to be the major obstacle for developing effective vaccines. Compared with normal DCs, tumor-associated DCs (TADCs) are less matured with poor responsiveness to Toll-like receptor (TLR) stimulation, which has been related with STAT3 hyperactivity. In the present study, Poly I:C (PIC, a TLR3 agonist), STAT3 siRNA and OVA antigen were co-encapsulated by poly (ethylene glycol)-b-poly (L-lysine)-b-poly (L-leucine) (PEG-PLL-PLLeu) polypeptide micelles to generate PMP/OVA/siRNA nanovaccine, which was aimed to effectively overcome DC dysfunction in vivo by deleting STAT3 gene in situ. The results showed that PMP/OVA/siRNA simultaneously facilitated the cellular uptake of OVA antigen and siRNA about 3-200 folds, and decreased STAT3 expression in TADCs over 50% both in vitro and in vivo. PMP/OVA/siRNA also elevated CD86 and CD40 expression as well as IL-12 production by TADCs more effectively than PMP/OVA did, indicating its strong potency of inducing TADC maturation and activation. Moreover, the immunization of PMP/OVA/siRNA rather than PMP/OVA effectively abrogated immunosuppression in the tumor microenvironment by increasing mature DCs and decreasing immunosuppressive cells in tumor-draining lymph nodes, which thereby led to potent anti-tumor immune responses and dramatic tumor regression with prolonged survival. Hence, in vivo co-delivery of immunopotentiator (PIC) and immunosuppressive gene silencer (STAT3 siRNA) by nanovaccines are expected to be a promising strategy to improve the therapeutic efficacy of cancer vaccines by modulating TADCs and overcoming tumor immunosupression.
Collapse
Affiliation(s)
- Zichao Luo
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Ce Wang
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Huqiang Yi
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Ping Li
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Hong Pan
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Lanlan Liu
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Lintao Cai
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Yifan Ma
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China.
| |
Collapse
|
21
|
Molinaro A, Holst O, Di Lorenzo F, Callaghan M, Nurisso A, D'Errico G, Zamyatina A, Peri F, Berisio R, Jerala R, Jiménez-Barbero J, Silipo A, Martín-Santamaría S. Chemistry of lipid A: at the heart of innate immunity. Chemistry 2014; 21:500-19. [PMID: 25353096 DOI: 10.1002/chem.201403923] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In many Gram-negative bacteria, lipopolysaccharide (LPS) and its lipid A moiety are pivotal for bacterial survival. Depending on its structure, lipid A carries the toxic properties of the LPS and acts as a potent elicitor of the host innate immune system via the Toll-like receptor 4/myeloid differentiation factor 2 (TLR4/MD-2) receptor complex. It often causes a wide variety of biological effects ranging from a remarkable enhancement of the resistance to the infection to an uncontrolled and massive immune response resulting in sepsis and septic shock. Since the bioactivity of lipid A is strongly influenced by its primary structure, a broad range of chemical syntheses of lipid A derivatives have made an enormous contribution to the characterization of lipid A bioactivity, providing novel pharmacological targets for the development of new biomedical therapies. Here, we describe and discuss the chemical aspects regarding lipid A and its role in innate immunity, from the (bio)synthesis, isolation and characterization to the molecular recognition at the atomic level.
Collapse
Affiliation(s)
- Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II via Cinthia 4, 80126 Napoli (Italy).
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Innate adjuvant receptor Toll-like receptor 3 can promote breast cancer through cell surface. Tumour Biol 2014; 36:1261-71. [PMID: 25348355 DOI: 10.1007/s13277-014-2737-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 10/13/2014] [Indexed: 02/03/2023] Open
Abstract
Toll-like receptor 3 has been targeted in different cancers for adjuvant therapy. The ligand-mediated effects of TLR-3 on cancer cells are discordant. In the present work, we have addressed the hypothesis possibility of cell membrane-bound action of TLR-3 in breast cancer to justify its pro-tumor effect. TLR-3 was stimulated by Poly (I:C) on the surface of human breast cancer cells MCF-7 and MDA-MB-231 for up to 72 h. To check the cell survival and growth, thiazol blue tetrazolium bromide (MTT) assay, apoptosis assay, and cell cycle analysis were carried out. For changes in the metastatic properties, in vitro colony formation assay, scratch-wound healing assay and adhesion assay were also done. Using real-time PCR and immunocytochemistry, expression of E-cadherin, was studied. To determine the affect of cytoplasmic stimulation, Poly (I:C) was delivered with lipid transfection reagent. The results of the aforesaid experiments showed that there was a gradual increase of cellular survivability, growth, and metastasis after the cell surface stimulation of TLR-3 with Poly (I:C). Interestingly, E-cadherin expression was increased both at transcriptional and translational level. On the other hand, when Poly (I:C) was delivered in the cytoplasm by lipid transfecting medium, the cells survivability was decreased. For the first time, in the present work, we are convincingly reporting the functional evidence that TLR-3 induces cell survivability and metastasis through cell surface. The present work may help for the proper understanding of the adjuvant therapy of breast cancer.
Collapse
|
23
|
Lai R, Jeyanathan M, Shaler CR, Damjanovic D, Khera A, Horvath C, Ashkar AA, Xing Z. Restoration of innate immune activation accelerates Th1-cell priming and protection following pulmonary mycobacterial infection. Eur J Immunol 2014; 44:1375-86. [PMID: 24519467 DOI: 10.1002/eji.201344300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Abstract
The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.
Collapse
Affiliation(s)
- Rocky Lai
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang M, Luo F, Zhang Y, Wang L, Lin W, Yang M, Hu D, Wu X, Chu Y. Pseudomonas aeruginosa mannose-sensitive hemagglutinin promotes T-cell response via toll-like receptor 4-mediated dendritic cells to slow tumor progression in mice. J Pharmacol Exp Ther 2014; 349:279-87. [PMID: 24623801 DOI: 10.1124/jpet.113.212316] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pseudomonas aeruginosa-mannose-sensitive hemagglutinin (PA-MSHA) as a drug may kill tumor cells and has been used clinically. However, the antitumor immune response of PA-MSHA is not completely understood. In this study, we found that treating Lewis lung carcinoma (3LL)-bearing mice with PA-MSHA plus 3LL antigen led to slower tumor progression and longer survival. After PA-MSHA treatment, T-cell number and dendritic cell maturation were both increased significantly at the tumor site. In addition, PA-MSHA in vitro stimulation resulted in the maturation of bone marrow-derived dendritic cells (BMDCs) from naive mice, showing higher costimulatory molecule expression, more cytokine secretion, lower endocytic activity, and stronger capacity to enhance T-cell activation. Toll-like receptor (TLR)4 but not TLR2 was required in the maturation process. More importantly, PA-MSHA-induced DCs were essential for PA-MSHA to enhance activation, expansion, and interferon (IFN)-γ secretion of TLR4-mediated T cells, which play a role in the antitumor effect of PA-MSHA. Thus, this study reveals PA-MSHA as a novel TLR4 agonist that elicits antitumor immune response to slow tumor progression.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center (M.Z., F.L., Y.Z., L.W., W.L., Y.C.) and Department of General Surgery, Zhongshan Hospital (M.Y.), Fudan University, Shanghai, China; and Beijing Wanter Biopharmaceutical Co., Ltd., Huairou Yanqi Economic-Technical Development Area, Beijing, China (D.H., X.W.)
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ioannou K, Derhovanessian E, Tsakiri E, Samara P, Kalbacher H, Voelter W, Trougakos IP, Pawelec G, Tsitsilonis OE. Prothymosin α and a prothymosin α-derived peptide enhance T(H)1-type immune responses against defined HER-2/neu epitopes. BMC Immunol 2013; 14:43. [PMID: 24053720 PMCID: PMC3852324 DOI: 10.1186/1471-2172-14-43] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/17/2013] [Indexed: 12/31/2022] Open
Abstract
Background Active cancer immunotherapies are beginning to yield clinical benefit, especially those using peptide-pulsed dendritic cells (DCs). Different adjuvants, including Toll-like receptor (TLR) agonists, commonly co-administered to cancer patients as part of a DC-based vaccine, are being widely tested in the clinical setting. However, endogenous DCs in tumor-bearing individuals are often dysfunctional, suggesting that ex vivo educated DCs might be superior inducers of anti-tumor immune responses. We have previously shown that prothymosin alpha (proTα) and its immunoreactive decapeptide proTα(100–109) induce the maturation of human DCs in vitro. The aim of this study was to investigate whether proTα- or proTα(100–109)-matured DCs are functionally competent and to provide preliminary evidence for the mode of action of these agents. Results Monocyte-derived DCs matured in vitro with proTα or proTα(100–109) express co-stimulatory molecules and secrete pro-inflammatory cytokines. ProTα- and proTα(100–109)-matured DCs pulsed with HER-2/neu peptides induce TH1-type immune responses, prime autologous naïve CD8-positive (+) T cells to lyse targets expressing the HER-2/neu epitopes and to express a polyfunctional profile, and stimulate CD4+ T cell proliferation in an HER-2/neu peptide-dependent manner. DC maturation induced by proTα and proTα(100–109) is likely mediated via TLR-4, as shown by assessing TLR-4 surface expression and the levels of the intracellular adaptor molecules TIRAP, MyD88 and TRIF. Conclusions Our results suggest that proTα and proTα(100–109) induce both the maturation and the T cell stimulatory capacity of DCs. Although further studies are needed, evidence for a possible proTα and proTα(100–109) interaction with TLR-4 is provided. The initial hypothesis that proTα and the proTα-derived immunoactive decapeptide act as “alarmins”, provides a rationale for their eventual use as adjuvants in DC-based anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Kyriaki Ioannou
- Department of Animal and Human Physiology, Faculty of Biology, University of Athens, Athens 15784, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Development of an ELISA for the quantification of the C-terminal decapeptide prothymosin α(100-109) in sera of mice infected with bacteria. J Immunol Methods 2013; 395:54-62. [PMID: 23831611 DOI: 10.1016/j.jim.2013.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 11/20/2022]
Abstract
Apoptosis is characterized by a series of discrete biochemical events, among which is the truncation of the nuclear polypeptide prothymosin alpha (proTα) by activated caspase-3. This early apoptotic event results in the generation of a carboxy-terminal fragment of proTα, the immunoactive decapeptide proTα(100-109). We hypothesized that the detection of increased levels of proTα(100-109) in serum can be directly correlated with the induction of massive cell apoptosis, resulting from a severe bacterial infection. Thus, using high-affinity-purified polyclonal antibodies (Abs), raised in rabbits and a prototype antibody-capture system, we developed a highly sensitive and specific competitive ELISA for proTα(100-109). The sensitivity of the ELISA (0.1ng/mL to 10μg/mL) is acceptable for the quantification of the decapeptide in serum samples. To assess our initial hypothesis, we determined the concentration of proTα(100-109) in the sera of mice infected with the bacterium Streptococcus pyogenes over the course of the infection. We show that serum concentration of proTα(100-109) was marginal to undetectable before infection, increased over time and peaked at 72h postinfection. In silico analysis suggests that the Abs generated are unlikely to cross-react with any other unrelated mouse or bacterial protein. Further validation of our ELISA using serum samples from humans, infected with bacteria, may provide a useful tool to differentiate the causative agent of a potentially lethal septic infection.
Collapse
|