1
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
2
|
Minute L, Bergón-Gutiérrez M, Mata-Martínez P, Fernández-Pascual J, Terrón V, Bravo-Robles L, Bıçakcıoğlu G, Zapata-Fernández G, Aguiló N, López-Collazo E, del Fresno C. Heat-killed Mycobacterium tuberculosis induces trained immunity in vitro and in vivo administered systemically or intranasally. iScience 2024; 27:108869. [PMID: 38318361 PMCID: PMC10838711 DOI: 10.1016/j.isci.2024.108869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/03/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Trained immunity (TI) represents a memory-like process of innate immune cells. TI can be initiated with various compounds such as fungal β-glucan or the tuberculosis vaccine, Bacillus Calmette-Guérin. Nevertheless, considering the clinical applications of harnessing TI against infections and cancer, there is a growing need for new, simple, and easy-to-use TI inducers. Here, we demonstrate that heat-killed Mycobacterium tuberculosis (HKMtb) induces TI both in vitro and in vivo. In human monocytes, this effect represents a truly trained process, as HKMtb confers boosted inflammatory responses against various heterologous challenges, such as lipopolysaccharide (Toll-like receptor [TLR] 4 ligand) and R848 (TLR7/8 ligand). Mechanistically, HKMtb-induced TI relies on epigenetic mechanisms in a Syk/HIF-1α-dependent manner. In vivo, HKMtb induced TI when administered both systemically and intranasally, with the latter generating a more robust TI response. Summarizing, our research has demonstrated that HKMtb has the potential to act as a mucosal immunotherapy that can successfully induce trained responses.
Collapse
Affiliation(s)
- Luna Minute
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Marta Bergón-Gutiérrez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Pablo Mata-Martínez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Jaime Fernández-Pascual
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Verónica Terrón
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumor Immunology Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Laura Bravo-Robles
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Gülce Bıçakcıoğlu
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Gabriela Zapata-Fernández
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Nacho Aguiló
- Department of Microbiology, Pediatrics, Radiology, and Public Health, University of Zaragoza/IIS Aragon, Zaragoza, Spain
- CIBERES, CIBERINFEC, Carlos III Health Institute, Madrid, Spain
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumor Immunology Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
- CIBERES, CIBERINFEC, Carlos III Health Institute, Madrid, Spain
| | - Carlos del Fresno
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Immunomodulation Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
3
|
Hartmann AK, Bartneck J, Pielenhofer J, Meiser SL, Arnold-Schild D, Klein M, Stassen M, Schild H, Muth S, Probst HC, Langguth P, Grabbe S, Radsak MP. Optimized dithranol-imiquimod-based transcutaneous immunization enables tumor rejection. Front Immunol 2023; 14:1238861. [PMID: 37727790 PMCID: PMC10505723 DOI: 10.3389/fimmu.2023.1238861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
Introduction Transcutaneous immunization (TCI) is a non-invasive vaccination method promoting strong cellular immune responses, crucial for the immunological rejection of cancer. Previously, we reported on the combined application of the TLR7 agonist imiquimod (IMQ) together with the anti-psoriatic drug dithranol as novel TCI platform DIVA (dithranol/IMQ based vaccination). In extension of this work, we further optimized DIVA in terms of drug dose, application pattern and established a new IMQ formulation. Methods C57BL/6 mice were treated on the ear skin with dithranol and IMQ-containing ointments together with ovalbumin-derived peptides. T cell responses were determined by flow cytometry and IFN-ɤ ELISpot assay, local skin inflammation was characterized by ear swelling. Results Applying the adjuvants on separate skin sites, a reduced number of specific CD8+ T cells with effector function was detectable, indicating that the local concurrence of adjuvants and peptide antigens is required for optimal vaccination. Likewise, changing the order of dithranol and IMQ resulted in an increased skin inflammatory reaction, but lower frequencies of antigen-specific CD8+ T cells indicating that dithranol is essential for superior T cell priming upon DIVA. Dispersing nanocrystalline IMQ in a spreadable formulation (IMI-Sol+) facilitated storage and application rendering comparable immune responses. DIVA applied one or two weeks after the first immunization resulted in a massive increase in antigen-specific T cells and up to a ten-fold increased memory response. Finally, in a prophylactic tumor setting, double but no single DIVA treatment enabled complete control of tumor growth, resulting in full tumor protection. Discussion Taken together, the described optimized transcutaneous vaccination method leads to the generation of a strong cellular immune response enabling the effective control of tumor growth and has the potential for clinical development as a novel non-invasive vaccination method for peptide-based cancer vaccines in humans.
Collapse
Affiliation(s)
- Ann-Kathrin Hartmann
- IIIrd Department of Medicine – Hematology and Oncology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Joschka Bartneck
- IIIrd Department of Medicine – Hematology and Oncology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jonas Pielenhofer
- Biopharmaceutics and Pharmaceutical Technology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Sophie Luise Meiser
- Biopharmaceutics and Pharmaceutical Technology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Danielle Arnold-Schild
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Klein
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael Stassen
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Mainz Research School of Translational Biomedicine (TransMed), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sabine Muth
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Peter Langguth
- Biopharmaceutics and Pharmaceutical Technology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Stephan Grabbe
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Mainz Research School of Translational Biomedicine (TransMed), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Markus P. Radsak
- IIIrd Department of Medicine – Hematology and Oncology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Mainz Research School of Translational Biomedicine (TransMed), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
4
|
Hirobe S, Yamasaki T, Ito S, Quan YS, Kamiyama F, Tachibana M, Okada N. Transcutaneous Administration of Imiquimod Promotes T and B Cell Differentiation into Effector Cells or Plasma Cells. Pharmaceutics 2022; 14:pharmaceutics14020385. [PMID: 35214117 PMCID: PMC8878978 DOI: 10.3390/pharmaceutics14020385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
We are interested in promoting the development of transcutaneous immunization using microneedle technology and attempting to apply an adjuvant with transcutaneous immunization to improve the efficacy and reduce the amount of antigen and number of administrations needed. In this study, we collected basic information to help elucidate the mechanism responsible for the transcutaneous adjuvant activity of imiquimod (IMQ), which is a ligand of toll-like receptor (TLR) 7. In mouse groups administered ovalbumin (OVA), the OVA-specific IgG antibody titer of the IMQ-adjuvanted group was higher than that of the group administered OVA alone. No immune response bias due to transcutaneous IMQ administration was observed in terms of IgG1 (T helper cell [Th]2-type IgG subclass) and IgG2c (Th1-type IgG subclass) antibody titers. After the initial immunization, the IMQ-adjuvanted group showed increased migration of Langerhans cells to draining lymph nodes (dLNs) and active proliferation of OVA-specific CD4+ T cells. Transcutaneously administered IMQ did not affect the direction of CD4+ T cell differentiation, while promoted B cell activation and germinal center (GC) B cell differentiation. Immune staining revealed greater GC formation in the dLNs with the IMQ-adjuvanted group than in the OVA-alone group. In the secondary immune response, effector T cells increased in the dLNs and spleen, and effector memory T cells also increased in the spleen in the IMQ-adjuvanted group. In addition, our results suggested that the administration of IMQ enhanced B cell differentiation into plasma cells and GC B cells in the dLNs and spleen. In this study, we partially clarified the mechanism underlying the adjuvant activity of transcutaneously administered IMQ, which is required for the practical application of transcutaneous immunization with IMQ.
Collapse
Affiliation(s)
- Sachiko Hirobe
- Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan;
- Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
- Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Taki Yamasaki
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.Y.); (S.I.); (M.T.)
| | - Sayami Ito
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.Y.); (S.I.); (M.T.)
| | - Ying-Shu Quan
- CosMED Pharmaceutical Co., Ltd., 32 Higashikujokawanishi-cho, Minami-ku, Kyoto 601-8014, Kyoto, Japan; (Y.-S.Q.); (F.K.)
| | - Fumio Kamiyama
- CosMED Pharmaceutical Co., Ltd., 32 Higashikujokawanishi-cho, Minami-ku, Kyoto 601-8014, Kyoto, Japan; (Y.-S.Q.); (F.K.)
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.Y.); (S.I.); (M.T.)
- Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan; (T.Y.); (S.I.); (M.T.)
- Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita 565-0871, Osaka, Japan
- Correspondence: ; Tel.: +81-668-794-412; Fax: +81-661-056-102
| |
Collapse
|
5
|
Telomeres and Cancer. Life (Basel) 2021; 11:life11121405. [PMID: 34947936 PMCID: PMC8704776 DOI: 10.3390/life11121405] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
Telomeres cap the ends of eukaryotic chromosomes and are indispensable chromatin structures for genome protection and replication. Telomere length maintenance has been attributed to several functional modulators, including telomerase, the shelterin complex, and the CST complex, synergizing with DNA replication, repair, and the RNA metabolism pathway components. As dysfunctional telomere maintenance and telomerase activation are associated with several human diseases, including cancer, the molecular mechanisms behind telomere length regulation and protection need particular emphasis. Cancer cells exhibit telomerase activation, enabling replicative immortality. Telomerase reverse transcriptase (TERT) activation is involved in cancer development through diverse activities other than mediating telomere elongation. This review describes the telomere functions, the role of functional modulators, the implications in cancer development, and the future therapeutic opportunities.
Collapse
|
6
|
Barati M, Mohebali M, Khamesipour A, Bahrami F, Darabi H, Khaze V, Riazi-Rad F, Habibi G, Ajdary S, Alimohammadian MH. Evaluation of Cellular Immune Responses in Dogs Immunized with Alum-Precipitated Autoclaved Leishmania major along with BCG and Imiquimod. IRANIAN JOURNAL OF PARASITOLOGY 2021; 16:348-356. [PMID: 34630579 PMCID: PMC8476724 DOI: 10.18502/ijpa.v16i3.7087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/05/2020] [Indexed: 11/24/2022]
Abstract
Background: We aimed to investigate the potential effects of BCG and imiquimod on improvement of current experimental L. major vaccine against dogs in an endemic area of Zoonotic visceral leishmaniasis (ZVL) in Iran. Methods: During 2012 till 2014, seven mixed-breed shepherd dogs with no anti-Leishmania antibodies and no response to Leishmanin reagent were immunized with 2 doses of alum-precipitated autoclaved L. major (Alum-AML) while BCG and imiquimod (for skin pre-treatment) were used as adjuvants. The productions of a few characteristic cytokines of T-helper immune responses and the development of delayed-type hypersensitivity (DTH) of the immunized animals were then evaluated, up to 300 days. Blood samples were collected at 0, 30, 80 and 300 d post-vaccination and the concentrations of IFN-γ, IL10, IL-12 and TGF-β cytokines secreted from PBMCs at these time-points were quantified by ELISA. DTH was evaluated by Leishmanin skin test (LST). Results: Although a similar LST conversion was observed at all time-points, the cytokine measurement results indicated significantly higher levels of IFN-γ at day 80 and elevated levels of IL-10 at days 80 and 300, post-vaccination. Moreover, a significantly higher IFN-γ/IL-10 ratio was observed at day 30 post-vaccination compared to the other time-points. Conclusion: Although a Th1-like response could be observed at day 30 post-vaccination, the development of cytokine profiles was inclined toward mixed Th1 and Th2 responses at days 80 and 300 post-vaccination. This situation may indicate the requirement of an additional boosting by this Alum-AML formula, in order to induce long-lasting protection against ZVL.
Collapse
Affiliation(s)
- Mohammad Barati
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Haiedeh Darabi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Khaze
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Farhad Riazi-Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
7
|
Hamie M, Najm R, Deleuze-Masquefa C, Bonnet PA, Dubremetz JF, El Sabban M, El Hajj H. Imiquimod Targets Toxoplasmosis Through Modulating Host Toll-Like Receptor-MyD88 Signaling. Front Immunol 2021; 12:629917. [PMID: 33767699 PMCID: PMC7986122 DOI: 10.3389/fimmu.2021.629917] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/11/2021] [Indexed: 12/22/2022] Open
Abstract
Toxoplasma gondii is a prevalent parasite of medical and veterinary importance. Tachyzoïtes and bradyzoïtes are responsible for acute and chronic toxoplasmosis (AT and CT), respectively. In immunocompetent hosts, AT evolves into a persistent CT, which can reactivate in immunocompromised patients with dire consequences. Imiquimod is an efficient immunomodulatory drug against certain viral and parasitic infections. In vivo, treatment with Imiquimod, throughout AT, reduces the number of brain cysts while rendering the remaining cysts un-infectious. Post-establishment of CT, Imiquimod significantly reduces the number of brain cysts, leading to a delay or abortion of reactivation. At the molecular level, Imiquimod upregulates the expression of Toll-like receptors 7, 11, and 12, following interconversion from bradyzoïtes to tachyzoïtes. Consequently, MyD88 pathway is activated, resulting in the induction of the immune response to control reactivated Toxoplasma foci. This study positions Imiquimod as a potent drug against toxoplasmosis and elucidates its mechanism of action particularly against chronic toxoplasmosis, which is the most prevalent form of the disease.
Collapse
Affiliation(s)
- Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rania Najm
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Relitti N, Saraswati AP, Federico S, Khan T, Brindisi M, Zisterer D, Brogi S, Gemma S, Butini S, Campiani G. Telomerase-based Cancer Therapeutics: A Review on their Clinical Trials. Curr Top Med Chem 2020; 20:433-457. [PMID: 31894749 DOI: 10.2174/1568026620666200102104930] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022]
Abstract
Telomeres are protective chromosomal ends that shield the chromosomes from DNA damage, exonucleolytic degradation, recombination, and end-to-end fusion. Telomerase is a ribonucleoprotein that adds TTAGGG tandem repeats to the telomeric ends. It has been observed that 85 to 90% of human tumors express high levels of telomerase, playing a crucial role in the development of cancers. Interestingly, the telomerase activity is generally absent in normal somatic cells. This selective telomerase expression has driven scientists to develop novel anti-cancer therapeutics with high specificity and potency. Several advancements have been made in this area, which is reflected by the enormous success of the anticancer agent Imetelstat. Since the discovery of Imetelstat, several research groups have contributed to enrich the therapeutic arsenal against cancer. Such contributions include the application of new classes of small molecules, peptides, and hTERT-based immunotherapeutic agents (p540, GV1001, GRNVAC1 or combinations of these such as Vx-001). Many of these therapeutic tools are under different stages of clinical trials and have shown promising outcomes. In this review, we highlight the current status of telomerase-based cancer therapeutics and the outcome of these investigations.
Collapse
Affiliation(s)
- Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Akella P Saraswati
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Tuhina Khan
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, University of Napoli Federico II, via D. Montesano 49, I-80131 Napoli, Italy
| | - Daniela Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, I-56126 Pisa, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, via Aldo Moro 2, I- 53100 Siena, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Anti-cancer Immunotherapies Targeting Telomerase. Cancers (Basel) 2020; 12:cancers12082260. [PMID: 32806719 PMCID: PMC7465444 DOI: 10.3390/cancers12082260] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
Telomerase is a reverse transcriptase that maintains telomeres length, compensating for the attrition of chromosomal ends that occurs during each replication cycle. Telomerase is expressed in germ cells and stem cells, whereas it is virtually undetectable in adult somatic cells. On the other hand, telomerase is broadly expressed in the majority of human tumors playing a crucial role in the replicative behavior and immortality of cancer cells. Several studies have demonstrated that telomerase-derived peptides are able to bind to HLA (human leukocyte antigen) class I and class II molecules and effectively activate both CD8+ and CD4+ T cells subsets. Due to its broad and selective expression in cancer cells and its significant immunogenicity, telomerase is considered an ideal universal tumor-associated antigen, and consequently, a very attractive target for anti-cancer immunotherapy. To date, different telomerase targeting immunotherapies have been studied in pre-clinical and clinical settings, these approaches include peptide vaccination and cell-based vaccination. The objective of this review paper is to discuss the role of human telomerase in cancer immunotherapy analyzing recent developments and future perspectives in this field.
Collapse
|
10
|
Gandhapudi SK, Ward M, Bush JPC, Bedu-Addo F, Conn G, Woodward JG. Antigen Priming with Enantiospecific Cationic Lipid Nanoparticles Induces Potent Antitumor CTL Responses through Novel Induction of a Type I IFN Response. THE JOURNAL OF IMMUNOLOGY 2019; 202:3524-3536. [PMID: 31053626 DOI: 10.4049/jimmunol.1801634] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
Certain types of cationic lipids have shown promise in cancer immunotherapy, but their mechanism of action is poorly understood. In this study, we describe the properties of an immunotherapeutic consisting of the pure cationic lipid enantiomer R-1,2-dioleoyl-3-trimethyl-ammonium-propane (R-DOTAP) formulated with modified viral or self-peptide Ags. R-DOTAP formulations with peptide Ags stimulate strong cross-presentation and potent CD8 T cell responses associated with a high frequency of polyfunctional CD8 T cells. In a human papillomavirus tumor model system, a single s.c. injection of tumor-bearing mice with R-DOTAP plus human papillomavirus Ags induces complete regression of large tumors associated with an influx of Ag-specific CD8 T cells and a reduction of the ratio of regulatory/Ag-specific CD8 T cells. R-DOTAP also synergizes with an anti-PD1 checkpoint inhibitor, resulting in a significant inhibition of B16 melanoma tumor growth. We found that R-DOTAP stimulates type I IFN production by dendritic cells in vivo and in vitro. s.c. injection of R-DOTAP results in an IFN-dependent increase in draining lymph node size and a concomitant increase in CD69 expression. Using knockout mice, we show that type I IFN is required for the induction of CD8 T cell activity following administration of R-DOTAP plus Ag. This response requires Myd88 but not TRIF or STING. We also show that R-DOTAP stimulates both TLR7 and 9. Collectively, these studies reveal that R-DOTAP stimulates endosomal TLRs, resulting in a Myd88-dependent production of type I IFN. When administered with Ag, this results in potent Ag-specific CD8 T cell responses and antitumor activity.
Collapse
Affiliation(s)
- Siva K Gandhapudi
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | - Martin Ward
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | - John Peyton C Bush
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | | | - Greg Conn
- PDS Biotechnology Corporation, Princeton, NJ 08540
| | - Jerold G Woodward
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| |
Collapse
|
11
|
Cytokine Effect of TLR3, TLR4, and TLR7 Agonists Alone or Associated with Leishmania infantum Antigen on Blood from Dogs. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5693736. [PMID: 30539014 PMCID: PMC6260531 DOI: 10.1155/2018/5693736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/30/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Activation of toll-like receptors (TLRs) has been shown to play an important role in leishmaniosis by enhancing the parasite specific immune responses to control infection. However, the role of TLR agonists has not been studied in detail in dogs. The aim of this study was to determine the effect of TLR3, TLR4, and TLR7 agonists (TLR3a, TLR4a, and TLR7a) alone or in combination with Leishmania infantum antigen (LSA) on TNF-α and IL-6 production in blood from dogs living in endemic areas of canine leishmaniosis (CanL). Twenty-four healthy dogs from Catalonia (n=14) and Ibizan hound dogs from the island of Mallorca (n=10) were enrolled. Whole blood with TLR3a, TLR4a, and TLR7a alone or combined with LSA were cultured separately, and IFN-γ, TNF-α, and IL-6 were measured by ELISA. A significant increase of TNF-α was found for all conditions studied compared to medium alone. Stimulation with TLR4a (p=0.0001) and TLR7a (p=0.005) presented a significantly marked increase in TNF-α and IL-6 production compared to TLR3a. Importantly, significantly higher TNF-α production was found in LSA+TLR4a (p=0.0001) stimulated blood and LSA+TLR7a (p=0.005) compared to LSA alone. All dogs showed higher TNF-α production after LSA+TLR7a compared to TLR7a (p=0.047) and LSA+TLR3a compared to TLR3a (p=0.052). These data indicate a marked inflammatory cytokine effect of TLR4a and TLR7a on blood from healthy dogs living in endemic areas of CanL. Additionally, LSA+TLR7a promoted a synergistic proinflammatory effect with TNF-α in all dogs. Those findings suggest an active role of TLRs in proinflammatory responses, which might be strongly involved in the process of disease resolution.
Collapse
|
12
|
Wilkinson A, Lattmann E, Roces CB, Pedersen GK, Christensen D, Perrie Y. Lipid conjugation of TLR7 agonist Resiquimod ensures co-delivery with the liposomal Cationic Adjuvant Formulation 01 (CAF01) but does not enhance immunopotentiation compared to non-conjugated Resiquimod+CAF01. J Control Release 2018; 291:1-10. [PMID: 30291987 DOI: 10.1016/j.jconrel.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 01/12/2023]
Abstract
Pattern recognition receptors, including the Toll-like receptors (TLRs), are important in the induction and activation of two critical arms of the host defence to pathogens and microorganisms: the rapid innate immune response (as characterised by the production of Th1 promoting cytokines and type 1 interferons) and the adaptive immune response. Through this activation, ligands and agonists of TLRs can enhance immunotherapeutic efficacy. Resiquimod is a small (water-soluble) agonist of the endosome-located Toll-like receptors 7 and 8 (TLR7/8). However due to its molecular attributes it rapidly distributes throughout the body after injection. To circumvent this, these TLR agonists can be incorporated within delivery systems, such as liposomes, to promote the co-delivery of both antigen and agonists to antigen presenting cells. In this present study, resiquimod has been chemically conjugated to a lipid to form a lipid-TLR7/8 agonist conjugate which can be incorporated within immunogenic cationic liposomes composed of dimethyldioctadecylammonium bromide (DDA) and the immunostimulatory glycolipid trehalose 6,6' - dibehenate (TDB). This DDA:TDB-TLR7/8 formulation offers similar vesicle characteristics to DDA:TDB (size and charge) and offers high retention of both resiquimod and the electrostatically adsorbed TB subunit antigen Ag85B-ESAT6-Rv2660c (H56). Following immunisation through the intramuscular (i.m.) route, these cationic DDA:TDB-TLR7/8 liposomes form a vaccine depot at the injection site. However, immunisation studies have shown that this biodistribution does not translate into notably increased antibody nor Th1 responses at the spleen and draining popliteal lymph node compared to DDA:TDB liposomes. This work demonstrates that the conjugation of TLR7/8 agonists to cationic liposomes can promote co-delivery but the immune responses stimulated do not merit the added complexity considerations of the formulation.
Collapse
Affiliation(s)
| | - Eric Lattmann
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Carla B Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow G4 0RE, UK
| | - Gabriel K Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow G4 0RE, UK.
| |
Collapse
|
13
|
de Groot AM, Platteel ACM, Kuijt N, van Kooten PJS, Vos PJ, Sijts AJAM, van der Maaden K. Nanoporous Microneedle Arrays Effectively Induce Antibody Responses against Diphtheria and Tetanus Toxoid. Front Immunol 2017; 8:1789. [PMID: 29375544 PMCID: PMC5770646 DOI: 10.3389/fimmu.2017.01789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/29/2017] [Indexed: 01/06/2023] Open
Abstract
The skin is immunologically very potent because of the high number of antigen-presenting cells in the dermis and epidermis, and is therefore considered to be very suitable for vaccination. However, the skin's physical barrier, the stratum corneum, prevents foreign substances, including vaccines, from entering the skin. Microneedles, which are needle-like structures with dimensions in the micrometer range, form a relatively new approach to circumvent the stratum corneum, allowing for minimally invasive and pain-free vaccination. In this study, we tested ceramic nanoporous microneedle arrays (npMNAs), representing a novel microneedle-based drug delivery technology, for their ability to deliver the subunit vaccines diphtheria toxoid (DT) and tetanus toxoid (TT) intradermally. First, the piercing ability of the ceramic (alumina) npMNAs, which contained over 100 microneedles per array, a length of 475 µm, and an average pore size of 80 nm, was evaluated in mouse skin. Then, the hydrodynamic diameters of DT and TT and the loading of DT, TT, and imiquimod into, and subsequent release from the npMNAs were assessed in vitro. It was shown that DT and TT were successfully loaded into the tips of the ceramic nanoporous microneedles, and by using near-infrared fluorescently labeled antigens, we found that DT and TT were released following piercing of the antigen-loaded npMNAs into ex vivo murine skin. Finally, the application of DT- and TT-loaded npMNAs onto mouse skin in vivo led to the induction of antigen-specific antibodies, with titers similar to those obtained upon subcutaneous immunization with a similar dose. In conclusion, we show for the first time, the potential of npMNAs for intradermal (ID) immunization with subunit vaccines, which opens possibilities for future ID vaccination designs.
Collapse
Affiliation(s)
- Anne Marit de Groot
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
| | - Anouk C. M. Platteel
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
| | - Nico Kuijt
- MyLife Technologies, Leiden, Netherlands
| | - Peter J. S. van Kooten
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Alice J. A. M. Sijts
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
| | | |
Collapse
|
14
|
Telò I, Favero ED, Cantù L, Frattini N, Pescina S, Padula C, Santi P, Sonvico F, Nicoli S. Gel-like TPGS-Based Microemulsions for Imiquimod Dermal Delivery: Role of Mesostructure on the Uptake and Distribution into the Skin. Mol Pharm 2017; 14:3281-3289. [DOI: 10.1021/acs.molpharmaceut.7b00348] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Isabella Telò
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Elena Del Favero
- Department
of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Via F.lli Cervi, 93, 20090 Segrate, Italy
| | - Laura Cantù
- Department
of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Via F.lli Cervi, 93, 20090 Segrate, Italy
| | - Noemi Frattini
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Silvia Pescina
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Cristina Padula
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Patrizia Santi
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Fabio Sonvico
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Sara Nicoli
- Food
and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| |
Collapse
|
15
|
The adjuvant effect of TLR7 agonist conjugated to a meningococcal serogroup C glycoconjugate vaccine. Eur J Pharm Biopharm 2016; 107:110-9. [DOI: 10.1016/j.ejpb.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/11/2016] [Accepted: 07/03/2016] [Indexed: 02/03/2023]
|
16
|
Kaurav M, Minz S, Sahu K, Kumar M, Madan J, Pandey RS. Nanoparticulate mediated transcutaneous immunization: Myth or reality. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1063-1081. [PMID: 26767517 DOI: 10.1016/j.nano.2015.12.372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/02/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED Transcutaneous immunization (TCI) is a promising route of vaccine delivery through skin due to many well documented advantages. The main obstacle in TCI is the skin's top dead layer i.e. stratum corneum which is difficult to penetrate. Efficiently delivery of antigen to the immune competent cells of epidermis or dermis in TCI might elicit an effective immune response. In this review, skin immunology with a particular focus on potential of immunological active receptors in influencing adaptive immune responses is highlighted. The challenges with TCI and methods to improve it using different adjuvants, chemical and physical approaches, delivery systems, and combination of above methods to further improve immune response following skin application of antigen are elaborately discussed. Nanoparticulate vaccine delivery systems with reference to their applications in TCI are classified according to their chronological development. Conclusively, clinical translations of above methods are also briefly reviewed. FROM THE CLINICAL EDITOR Transcutaneous immunization has been investigated by many as a promising route of vaccination. In this comprehensive review article, the authors described and discussed the existing knowledge and difficulties in this approach. Furthermore, ways of improving transcutaneous delivery were also reviewed.
Collapse
Affiliation(s)
- Monika Kaurav
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Sunita Minz
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Kantrol Sahu
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Manoj Kumar
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | | | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| |
Collapse
|
17
|
Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines 2015; 14:1509-23. [PMID: 26313239 DOI: 10.1586/14760584.2015.1081067] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The many factors impacting the efficacy of a vaccine can be broadly divided into three categories: features of the vaccine itself, including immunogen design, vaccine type, formulation, adjuvant and dosing; individual variations among vaccine recipients and vaccine administration-related parameters. While much literature exists related to vaccines, and recently systems biology has started to dissect the impact of individual subject variation on vaccine efficacy, few studies have focused on the role of vaccine administration-related parameters on vaccine efficacy. Parenteral and mucosal vaccinations are traditional approaches for licensed vaccines; novel vaccine delivery approaches, including needless injection and adjuvant formulations, are being developed to further improve vaccine safety and efficacy. This review provides a brief summary of vaccine administration-related factors, including vaccination approach, delivery route and method of administration, to gain a better understanding of their potential impact on the safety and immunogenicity of candidate vaccines.
Collapse
Affiliation(s)
- Lu Zhang
- a 1 Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.,b 2 China-US Vaccine Research Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wei Wang
- c 3 Wang Biologics, LLC, Chesterfield, MO 63017, USA ; Current affiliation: Bayer HealthCare, Berkeley, CA 94710, USA
| | - Shixia Wang
- d 4 Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
18
|
Conjugation of a TLR7 agonist and antigen enhances protection in the S. pneumoniae murine infection model. Eur J Pharm Biopharm 2014; 87:310-7. [PMID: 24434202 DOI: 10.1016/j.ejpb.2014.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 12/04/2013] [Accepted: 01/07/2014] [Indexed: 11/21/2022]
Abstract
Next generation vaccine adjuvants include Toll like receptor agonists, which are mostly extracted from microorganisms, but synthetic small molecule TLR agonists have also been identified. However, their delivery systems have not been optimized for effective administration in conjunction with antigens. Here, we describe a novel approach in which a small molecule TLR agonist was directly conjugated to antigen to ensure effective co-delivery. We describe the conjugation of a recombinant protective antigen from Streptococcus pneumoniae linked to a TLR7 agonist. Following thorough characterization to ensure no aggregation, the conjugate was evaluated in a murine infection model. Results showed that the conjugate extended the animals' survival after lethal challenge with S. pneumoniae. Comparable results were obtained with a dose 10-fold lower than that of the native unconjugated antigen. Notably, the animals immunized with the same dose of unconjugated TLR7 agonist and antigen showed no adjuvant effect. The increased immunogenicity was likely a consequence of the co-localization of TLR7 agonist and antigen by chemical binding and was more effective than simple co-administration. This approach can be adopted to increase potency of a broad variety of antigens and reduce the dose of antigen required to induce protective immunity.
Collapse
|
19
|
Igartua M, Pedraz JL. Topical resiquimod: a promising adjuvant for vaccine development? Expert Rev Vaccines 2014; 9:23-7. [DOI: 10.1586/erv.09.135] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Topical CpG adjuvantation of a protein-based vaccine induces protective immunity to Listeria monocytogenes. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:329-39. [PMID: 24391136 DOI: 10.1128/cvi.00734-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Robust CD8(+) T cell responses are essential for immune protection against intracellular pathogens. Using parenteral administration of ovalbumin (OVA) protein as a model antigen, the effect of the Toll-like receptor 9 (TLR9) agonist, CpG oligodeoxynucleotide (ODN) 1826, as an adjuvant delivered either topically, subcutaneously, or intramuscularly on antigen-specific CD8(+) T cell responses in a mouse model was evaluated. Topical CpG adjuvant increased the frequency of OVA-specific CD8(+) T cells in the peripheral blood and in the spleen. The more effective strategy to administer topical CpG adjuvant to enhance CD8(+) T cell responses was single-dose administration at the time of antigen injection with a prime-boost regimen. Topical CpG adjuvant conferred both rapid and long-lasting protection against systemic challenge with recombinant Listeria monocytogenes expressing the cytotoxic T lymphocyte (CTL) epitope of OVA(257-264) (strain Lm-OVA) in a TLR9-dependent manner. Topical CpG adjuvant induced a higher proportion of CD8(+) effector memory T cells than parenteral administration of the adjuvant. Although traditional vaccination strategies involve coformulation of antigen and adjuvant, split administration using topical adjuvant is effective and has advantages of safety and flexibility. Split administration of topical CpG ODN 1826 with parenteral protein antigen is superior to other administration strategies in enhancing both acute and memory protective CD8(+) T cell immune responses to subcutaneous protein vaccines. This vaccination strategy induces rapid and persistent protective immune responses against the intracellular organism L. monocytogenes.
Collapse
|
21
|
Abstract
Vaccination via skin often induces stronger immune responses than via muscle. This, in line with potential needle-free, painless delivery, makes skin a very attractive site for immunization. Yet, despite decades of effort, effective skin delivery is still in its infant stage and safe and potent adjuvants for skin vaccination remain largely undefined. We have shown that laser technologies including both fractional and non-fractional lasers can greatly augment vaccine-induced immune response without incurring any significant local and systemic side effects. Laser illumination at specific settings can accelerate the motility of antigen-presenting cells or trigger release of 'danger' signals stimulating the immune system. Moreover, several other groups including the authors explore laser technologies for needle-free transcutaneous vaccine delivery. As these laser-mediated resurfacing technologies are convenient, safe and cost-effective, their new applications in vaccination warrant clinical studies in the very near future.
Collapse
Affiliation(s)
- Xinyuan Chen
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School, Massachusetts General Hospital, 50 Boston Street, Edwards 222, Boston, MA 02114, USA
| | - Ji Wang
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School, Massachusetts General Hospital, 50 Boston Street, Edwards 222, Boston, MA 02114, USA
| | - Dilip Shah
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School, Massachusetts General Hospital, 50 Boston Street, Edwards 222, Boston, MA 02114, USA
| | - Mei X Wu
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
22
|
Fenoglio D, Traverso P, Parodi A, Tomasello L, Negrini S, Kalli F, Battaglia F, Ferrera F, Sciallero S, Murdaca G, Setti M, Sobrero A, Boccardo F, Cittadini G, Puppo F, Criscuolo D, Carmignani G, Indiveri F, Filaci G. A multi-peptide, dual-adjuvant telomerase vaccine (GX301) is highly immunogenic in patients with prostate and renal cancer. Cancer Immunol Immunother 2013; 62:1041-52. [PMID: 23591981 PMCID: PMC11029691 DOI: 10.1007/s00262-013-1415-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 03/07/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anti-tumor vaccination is a new frontier in cancer treatment applicable to immunogenic neoplasms such as prostate and renal cancers. GX301 is a vaccine constituted by four telomerase peptides and two adjuvants, Montanide ISA-51 and Imiquimod. OBJECTIVE The aim of this study was to analyze safety and tolerability of GX301 in an open-label, phase I/II trial. Immunological and clinical responses were also evaluated as secondary endpoints. EXPERIMENTAL DESIGN GX301 was administered by intradermally injecting 500 μg of each peptide (dissolved in Montanide ISA-51) in the skin of the abdomen. Imiquimod was applied as a cream at the injection sites. The protocol included 8 administrations at days 1, 3, 5, 7, 14, 21, 35, 63. Eligible patients were affected with stage IV prostate or renal cancer resistant to conventional treatments. Patients were clinically and immunologically monitored up to 6 months from the first immunization. RESULTS No grade 3-4 adverse events were observed. Evidence of vaccine-specific immunological responses was detected in 100 % of patients. Disease stabilization occurred in 4 patients. Prolonged progression-free survival and overall survival were observed in patients showing a full pattern of vaccine-specific immunological responses. CONCLUSION GX301 demonstrated to be safe and highly immunogenic. Further studies are needed to determine its clinical efficacy.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Surgical Sciences, University of Genoa, Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Laura Tomasello
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Simone Negrini
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Florinda Battaglia
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Francesca Ferrera
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Stefania Sciallero
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Maurizio Setti
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Alberto Sobrero
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Francesco Boccardo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Giuseppe Cittadini
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Francesco Puppo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Domenico Criscuolo
- Genovax srl, Colleretto Giacosa, Italy
- Present Address: Mediolanum Farmaceutici Spa, Milan, Italy
| | | | - Francesco Indiveri
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
23
|
Schully KL, Sharma S, Peine KJ, Pesce J, Elberson MA, Fonseca ME, Prouty AM, Bell MG, Borteh H, Gallovic M, Bachelder EM, Keane-Myers A, Ainslie KM. Rapid vaccination using an acetalated dextran microparticulate subunit vaccine confers protection against triplicate challenge by bacillus anthracis. Pharm Res 2013; 30:1349-61. [PMID: 23354770 DOI: 10.1007/s11095-013-0975-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/04/2013] [Indexed: 01/20/2023]
Abstract
PURPOSE A rapid immune response is required to prevent death from Anthrax, caused by Bacillus anthracis. METHOD We formulated a vaccine carrier comprised of acetalated dextran microparticles encapsulating recombinant protective antigen (rPA) and resiquimod (a toll-like receptor 7/8 agonist). RESULTS We were able to protect against triplicate lethal challenge by vaccinating twice (Days 0, 7) and then aggressively challenging on Days 14, 21, 28. A significantly higher level of antibodies was generated by day 14 with the encapsulated group compared to the conventional rPA and alum group. Antibodies produced by the co-encapsulated group were only weakly-neutralizing in toxin neutralization; however, survival was not dependent on toxin neutralization, as all vaccine formulations survived all challenges except control groups. Post-mortem culture swabs taken from the hearts of vaccinated groups that did not produce significant neutralizing titers failed to grow B. anthracis. CONCLUSIONS Results indicate that protective antibodies are not required for rapid protection; indeed, cytokine results indicate that T cell protection may play a role in protection from anthrax. We report the first instance of use of a particulate carrier to generate a rapid protective immunity against anthrax.
Collapse
Affiliation(s)
- Kevin L Schully
- Vaccine and Medical Countermeasures Department Biological Defense Research Directorate Naval Medical Research Center, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hirobe S, Okada N, Nakagawa S. Transcutaneous vaccines--current and emerging strategies. Expert Opin Drug Deliv 2013; 10:485-98. [PMID: 23316778 DOI: 10.1517/17425247.2013.760542] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Vaccination, which is the major fundamental prophylaxis against illness and death from infectious disease, has greatly contributed to the global improvement of human health. However, the disadvantages of conventional injection systems hamper the delivery of vaccination technologies to developing countries. The imminent practice of easy-to-use vaccination methods is expected to overcome certain issues associated with injectable vaccinations. One innovative method is the transcutaneous immunization (TCI) system. AREAS COVERED Two major strategies for TCI are discussed in this review. One is to promote antigen permeation of the skin barrier by patch systems or nanoparticles. The other is the delivery of antigens into the skin by electroporation and microneedles in order to physically overcome the skin barrier. Moreover, adjuvant development for TCI is discussed. EXPERT OPINION Many different approaches have been developed for TCI, which have the potential to be effective, easy-to-use and painless methods of vaccination. However, in practical terms, the guidelines concerning the manufacturing processes and clinical trial evaluation of the procedures have not kept pace with the development of these novel formulations. The accumulation of information regarding skin characteristics and the properties of TCI devices will help refine TCI system development guidelines and thus lead to the improvement of transcutaneous vaccination.
Collapse
Affiliation(s)
- Sachiko Hirobe
- Osaka University, Graduate School of Pharmaceutical Sciences, Laboratory of Biotechnology and Therapeutics, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
25
|
Crespo MI, Zacca ER, Núñez NG, Ranocchia RP, Maccioni M, Maletto BA, Pistoresi-Palencia MC, Morón G. TLR7 triggering with polyuridylic acid promotes cross-presentation in CD8α+ conventional dendritic cells by enhancing antigen preservation and MHC class I antigen permanence on the dendritic cell surface. THE JOURNAL OF IMMUNOLOGY 2013; 190:948-60. [PMID: 23284054 DOI: 10.4049/jimmunol.1102725] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
ssRNA can interact with dendritic cells (DCs) through binding to TLR7, inducing secretion of proinflammatory cytokines and type I IFN. Triggering TLR7 enhances cross-priming of CD8(+) T cells, which requires cross-presentation of exogenous Ag to DCs. However, how TLR triggering can affect Ag cross-presentation is still not clear. Using OVA as an Ag model, we observed that stimulation of TLR7 in DCs by polyuridylic acid (polyU), a synthetic ssRNA analog, generates a strong specific cytotoxic response in C57BL/6 mice. PolyU stimulate CD8α(+) DCs to cross-prime naive CD8(+) T cells in a type I IFN-dependent fashion. This enhanced cross-priming is accompanied by a higher density of OVA(256-264)/H-2K(b) complexes on CD8α(+) DCs treated with polyU, as well as by upregulation of costimulatory molecules and increased secretion of proinflammatory cytokines by DCs. Cross-priming of CD8(+) T cells by DCs treated with polyU requires proteasome and Ag translocation to cytosol through the Sec61 channel in DCs. The observed enhancement in OVA cross-presentation with polyU in DCs could be mediated by a limited Ag degradation in endophagosomal compartments and a higher permanence of OVA peptide/MHC class I complexes on DCs. These observations clearly reveal that key steps of Ag processing for cross-presentation can be modulated by TLR ligands, opening new avenues for understanding their mechanisms as adjuvants of the immune response.
Collapse
Affiliation(s)
- María I Crespo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Li N, Peng LH, Chen X, Nakagawa S, Gao JQ. Transcutaneous vaccines: Novel advances in technology and delivery for overcoming the barriers. Vaccine 2011; 29:6179-90. [DOI: 10.1016/j.vaccine.2011.06.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 06/19/2011] [Accepted: 06/22/2011] [Indexed: 12/17/2022]
|
27
|
Trepiakas R, Berntsen A, Hadrup SR, Bjørn J, Geertsen PF, Straten PT, Andersen MH, Pedersen AE, Soleimani A, Lorentzen T, Johansen JS, Svane IM. Vaccination with autologous dendritic cells pulsed with multiple tumor antigens for treatment of patients with malignant melanoma: results from a phase I/II trial. Cytotherapy 2011; 12:721-34. [PMID: 20429791 DOI: 10.3109/14653241003774045] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIM Dendritic cells are regarded as the most effective antigen presenting cells and coordinators of the immune response and therefore suitable as vaccine basis. Here we present results from a clinical study in which patients with malignant melanoma (MM) with verified progressive disease received vaccination with autologous monocyte-derived mature dendritic cells (DC) pulsed with p53, survivin and telomerase-derived peptides (HLA-A2+ patients) or with autologous/allogeneic tumor lysate (HLA-A2(−) patients) in combination with low-dose interleukin (IL)-2 and interferon (IFN)-alpha2b. RESULTS Of 46 patients who initiated treatment, 10 stopped treatment within 1-4 weeks because of rapid disease progression and deterioration. After 8 weeks, 36 patients were evaluable: no patient had an objective response, 11 patients had stable disease (SD); six had continued SD after 4 months, and three patients had prolonged SD for more than 6 months. The mean overall survival time was 9 months, with a significantly longer survival (18.4 months) of patients who attained SD compared with patients with progressive disease (PD) (5 months). Induction of antigen-specific T-cell responses was analyzed by multidimensional encoding of T cells using HLA-A2 major histocompatibility complex (MHC) multimers. Immune responses against five high-affinity vaccine peptides were detectable in the peripheral blood of six out of 10 analyzed HLA-A2+ patients. There was no observed correlation between the induction of immune responses and disease stabilization. A significant lower blood level of regulatory T cells (CD25(high) CD4 T cells) was demonstrable after six vaccinations in patients with SD compared with PD. CONCLUSIONS Vaccination was feasible and safe. Treatment-associated SD was observed in 24% of the patients. SD correlated with prolonged survival suggesting a clinical benefit. Differences in the level of regulatory T cells among SD and PD patients could indicate a significant role of these immune suppressive cells.
Collapse
Affiliation(s)
- Redas Trepiakas
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev Hospital, Herlev, Denmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
van den Berg JH, Nuijen B, Schumacher TN, Haanen JBAG, Storm G, Beijnen JH, Hennink WE. Synthetic vehicles for DNA vaccination. J Drug Target 2010; 18:1-14. [PMID: 19814658 DOI: 10.3109/10611860903278023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
DNA vaccination is an attractive immunization method able to induce robust cellular immune responses in pre-clinical models. However, clinical DNA vaccination trials performed thus far have resulted in marginal responses. Consequently, strategies are currently under development to improve the efficacy of DNA vaccines. A promising strategy is the use of synthetic particle formulations as carrier systems for DNA vaccines. This review discusses commonly used synthetic carriers for DNA vaccination and provides an overview of in vivo studies that use this strategy. Future recommendations on particle characteristics, target cell types and evaluation models are suggested for the potential improvement of current and novel particle delivery systems. Finally, hurdles which need to be tackled for clinical evaluation of these systems are discussed.
Collapse
Affiliation(s)
- Joost H van den Berg
- Department of Pharmacy & Pharmacology, Slotervaart Hospital, Louwesweg 6, 1066 EC Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
30
|
Wei J, Waithman J, Lata R, Mifsud NA, Cebon J, Kay T, Smyth MJ, Sadler AJ, Chen W. Influenza A infection enhances cross-priming of CD8+ T cells to cell-associated antigens in a TLR7- and type I IFN-dependent fashion. THE JOURNAL OF IMMUNOLOGY 2010; 185:6013-22. [PMID: 20956347 DOI: 10.4049/jimmunol.1002129] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The initiation of antitumor immunity relies on dendritic cells (DCs) to cross-present cell-associated tumor Ag to CD8(+) T cells (T(CD8+)) due to a lack of costimulatory molecules on tumor cells. Innate danger signals have been demonstrated to enhance cross-priming of T(CD8+) to soluble as well as virally encoded Ags; however, their effect on enhancing T(CD8+) cross-priming to cell genome-encoded Ags remains unknown. Furthermore, influenza A virus (IAV) has not been shown to enhance antitumor immunity. Using influenza-infected allogeneic cell lines, we show in this study that T(CD8+) responses to cell-associated Ags can be dramatically enhanced due to enhanced T(CD8+) expansion. This enhanced cross-priming in part involves TLR7- but not TLR3-mediated sensing of IAV and is entirely dependent on MyD88 and IFN signaling pathways. We also showed that the inflammasome-induced IL-1 and IFN-γ did not play a role in enhancing cross-priming in our system. We further demonstrated in our ex vivo system that CD8(+) DCs are the only APCs able to prime TCR-transgenic T(CD8+). Importantly, plasmacytoid DCs and CD8(-) DCs were both able to enhance such priming when provided in coculture. These observations suggest that IAV infection of tumor cells may facilitate improved cross-presentation of tumor Ags and may be used to augment clinical vaccine efficacy.
Collapse
Affiliation(s)
- Joe Wei
- Ludwig Institute for Cancer Research, Austin Health, Heidelberg, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Roukens AH, Vossen AC, Boland GJ, Verduyn W, van Dissel JT, Visser LG. Intradermal hepatitis B vaccination in non-responders after topical application of imiquimod (Aldara). Vaccine 2010; 28:4288-93. [PMID: 20433806 DOI: 10.1016/j.vaccine.2010.04.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 04/07/2010] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Five to ten percent of immunocompetent persons fail to develop a protective immune response to hepatitis B vaccination, and are defined non-responders (NR). We investigated the immune response to intradermal hepatitis B vaccination after pre-treatment of the skin with the TLR7 agonist imiquimod. METHODS Twenty-one non-responders (anti-HBs <10 IU/l after at least 6 intramuscular hepatitis B vaccinations) were randomly assigned to the control group (N=11) or the experimental group (N=10). Participants in both groups received 3 intradermal (ID) vaccinations with 5 microg HBsAg (0.125 mL) at 0, 1 and 6 months. In the experimental group, the dermal site of injection was pre-treated with 250 mg imiquimod ointment. Anti-HBs antibodies were determined at 0, 1, 2, 6 and 7 months. RESULTS In both study groups, 70% of the participants developed a protective immune response (anti-HBs >or=10 IU/l), after the 3rd intradermal vaccination. CONCLUSION The application of imiquimod on the skin prior to intradermal vaccination did not enhance the humoral response to hepatitis B vaccine. However, irrespective of imiquimod application, 70% of the NR who had not responded to 6 previous intramuscular vaccinations, developed a protective immune response with high affinity antibodies after 3 ID hepatitis B vaccinations with 5 microg HBsAg.
Collapse
Affiliation(s)
- Anna H Roukens
- Department of Infectious Diseases, Leiden University Medical Center, The Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Lu H, Wagner WM, Gad E, Yang Y, Duan H, Amon LM, Van Denend N, Larson ER, Chang A, Tufvesson H, Disis ML. Treatment failure of a TLR-7 agonist occurs due to self-regulation of acute inflammation and can be overcome by IL-10 blockade. THE JOURNAL OF IMMUNOLOGY 2010; 184:5360-7. [PMID: 20308630 DOI: 10.4049/jimmunol.0902997] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple TLR agonists have been shown to have antitumor effects in animal models. However, the therapeutic efficacy of TLR agonist monotherapy in cancer treatment has been limited, and the mechanisms of failure remain unknown. We demonstrate that topical treatment with a TLR-7 agonist, imiquimod, can elicit significant regression of spontaneous breast cancers in neu transgenic mice, a model of human HER-2/neu(+) breast cancer. However, tumor growth progressed once imiquimod therapy was ended. Gene expression analysis using tumor-derived RNA demonstrated that imiquimod induced high levels of IL-10 in addition to TNF-alpha and IFN-gamma. Elevated levels of circulating IL-10 were also detected in sera from imiquimod-treated mice. Elevated serum IL-10 appeared to be derived from IL-10 and dual cytokine secreting (IFN-gamma(+) and IL-10(+)) CD4(+) T cells rather than CD4(+)CD25(+)Foxp3(+) T regulatory cells, which were also induced by imiquimod treatment. Blockade of IL-10, but not TGF-beta, enhanced the antitumor effect of imiquimod by significantly prolonging survival in treated mice. These data suggest that the excessive inflammation induced by TLR agonists may result in a self-regulatory immunosuppression via IL-10 induction and that blocking IL-10 could enhance the therapeutic efficacy of these agents.
Collapse
Affiliation(s)
- Hailing Lu
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, 815 Mercer Street, Room 220, Box 358050, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chang BA, Cross JL, Najar HM, Dutz JP. Topical resiquimod promotes priming of CTL to parenteral antigens. Vaccine 2009; 27:5791-9. [PMID: 19660592 DOI: 10.1016/j.vaccine.2009.07.062] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 07/14/2009] [Accepted: 07/19/2009] [Indexed: 01/12/2023]
Abstract
We explored the topical use of resiquimod (R-848), a Toll-like receptor (TLR) 7/8 agonist, in gel formulation, to enhance cross-priming to subcutaneously administered protein antigen in a murine model. Resiquimod application at the time of subcutaneous administration of ovalbumin generated robust antigen-specific CTL as detected by tetramers, IFN-gamma ELISPOT assays and standard cytotoxicity assays. Induced CTL were capable of mediating antigen-specific killing in vivo as measured by in vivo cytotoxicity assays and an ability to protect against B16-OVA tumor challenge. Multiple serial applications of topical resiquimod increased the frequency of antigen-specific CTL when compared to single application. This enhanced frequency was noted despite a marked inhibition of adjuvant mediated pro-inflammatory cytokine release following repeated administration. Topical resiquimod is a potent adjuvant for locally administered subcutaneous vaccines, inducing clinically relevant CTL responses following single application at the time of subcutaneous vaccination.
Collapse
Affiliation(s)
- Brent A Chang
- Child and Family Research Institute, Department of Dermatology & Skin Science, University of British Columbia, 835 West Tenth Ave, Vancouver, BC, Canada V5Z 4E8
| | | | | | | |
Collapse
|
34
|
Vandermeulen G, Daugimont L, Richiardi H, Vanderhaeghen ML, Lecouturier N, Ucakar B, Préat V. Effect of Tape Stripping and Adjuvants on Immune Response After Intradermal DNA Electroporation. Pharm Res 2009; 26:1745-51. [DOI: 10.1007/s11095-009-9885-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 03/27/2009] [Indexed: 10/20/2022]
|
35
|
Skountzou I, Kang SM. Transcutaneous Immunization with Influenza Vaccines. Curr Top Microbiol Immunol 2009; 333:347-68. [DOI: 10.1007/978-3-540-92165-3_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
Enhanced immunogenicity of Plasmodium falciparum peptide vaccines using a topical adjuvant containing a potent synthetic Toll-like receptor 7 agonist, imiquimod. Infect Immun 2008; 77:739-48. [PMID: 19047411 DOI: 10.1128/iai.00974-08] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Plasmodium sporozoites injected into the skin by malaria-infected mosquitoes can be effectively targeted by antibodies that block parasite invasion of host hepatocytes and thus prevent the subsequent development of blood stage infections responsible for clinical disease. Malaria subunit vaccines require potent adjuvants, as they lack known pathogen-associated molecular patterns found in attenuated viral or bacterial vaccines that function as Toll-like receptor (TLR) agonists to stimulate dendritic cells and initiate strong adaptive immune responses. A synthetic TLR7 agonist, imiquimod, which is FDA approved for topical treatment of various skin conditions, can function as a potent adjuvant for eliciting T-cell responses to intracellular pathogens and model protein antigens. In the current studies, the topical application of imiquimod at the site of subcutaneously injected Plasmodium falciparum circumsporozoite (CS) peptides elicited strong parasite-specific humoral immunity that protected against challenge with transgenic rodent parasites that express P. falciparum CS repeats. In addition, injection of a simple linear peptide followed by topical imiquimod elicited strong Th1 CD4(+) T-cell responses, as well as high antibody titers. The correlation of high anti-repeat antibody titers with resistance to sporozoite challenge in vivo and in vitro supports use of this topical TLR7 agonist adjuvant to elicit protective humoral immunity. The safety, simplicity, and economic advantages of a topical synthetic TLR7 agonist adjuvant also apply to other vaccines requiring high antibody titers, such as malaria asexual or sexual blood stage antigens to prevent red blood cell invasion and block transmission to the mosquito vector, and to vaccines to other extracellular pathogens.
Collapse
|
37
|
Therapeutic Dendritic Cell Vaccination of Patients With Metastatic Renal Cell Carcinoma. J Immunother 2008; 31:771-80. [DOI: 10.1097/cji.0b013e3181833818] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
|
39
|
Adams S, O'Neill DW, Nonaka D, Hardin E, Chiriboga L, Siu K, Cruz CM, Angiulli A, Angiulli F, Ritter E, Holman RM, Shapiro RL, Berman RS, Berner N, Shao Y, Manches O, Pan L, Venhaus RR, Hoffman EW, Jungbluth A, Gnjatic S, Old L, Pavlick AC, Bhardwaj N. Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant. THE JOURNAL OF IMMUNOLOGY 2008; 181:776-84. [PMID: 18566444 DOI: 10.4049/jimmunol.181.1.776] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell-mediated immunity to microbes and to cancer can be enhanced by the activation of dendritic cells (DCs) via TLRs. In this study, we evaluated the safety and feasibility of topical imiquimod, a TLR7 agonist, in a series of vaccinations against the cancer/testis Ag NY-ESO-1 in patients with malignant melanoma. Recombinant, full-length NY-ESO-1 protein was administered intradermally into imiquimod preconditioned sites followed by additional topical applications of imiquimod. The regimen was very well tolerated with only mild and transient local reactions and constitutional symptoms. Secondarily, we examined the systemic immune response induced by the imiquimod/NY-ESO-1 combination, and show that it elicited both humoral and cellular responses in a significant fraction of patients. Skin biopsies were assessed for imiquimod's in situ immunomodulatory effects. Compared with untreated skin, topical imiquimod induced dermal mononuclear cell infiltrates in all patients composed primarily of T cells, monocytes, macrophages, myeloid DCs, NK cells, and, to a lesser extent, plasmacytoid DCs. DC activation was evident. This study demonstrates the feasibility and excellent safety profile of a topically applied TLR7 agonist used as a vaccine adjuvant in cancer patients. Imiquimod's adjuvant effects require further evaluation and likely need optimization of parameters such as formulation, dose, and timing relative to Ag exposure for maximal immunogenicity.
Collapse
Affiliation(s)
- Sylvia Adams
- Department of Medicine, Division of Medical Oncology, New York University Cancer Institute, School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Immunization with a Toll-like receptor 7 and/or 8 agonist vaccine adjuvant increases protective immunity against Leishmania major in BALB/c mice. Infect Immun 2008; 76:3777-83. [PMID: 18474642 DOI: 10.1128/iai.01527-07] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Activation of Toll-like receptors (TLRs) on antigen-presenting cells of the innate immune system initiates, amplifies, and directs the antigen-specific acquired immune response. Ligands that stimulate TLRs therefore represent potential vaccine adjuvants. In the present study, we determined whether imiquimod and its related compound R848, which are TLR7 and/or TLR8 agonists, represent potential vaccine adjuvants when delivered topically, subcutaneously, or intramuscularly. Using the Leishmania major infection model in BALB/c mice, vaccination with crude Leishmania antigen was not protective against subsequent challenge infection unless it was administered with R848 or a topical application of imiquimod containing cream on the skin. Subcutaneous vaccination with these adjuvants mediated a TH1 response against L. major antigen, which appeared to suppress the TH2 response following a challenge infection. Protective immunity was generated following subcutaneous vaccination but not intramuscular vaccination. These observations suggest that topically administered imiquimod or subcutaneously injected R848 represent potential vaccine adjuvants to enhance the TH1 response, which can be used with existing or new vaccine formulations.
Collapse
|
41
|
Zhong J, Hadis U, De Kluyver R, Leggatt GR, Fernando GJP, Frazer IH. TLR7 stimulation augments T effector-mediated rejection of skin expressing neo-self antigen in keratinocytes. Eur J Immunol 2008; 38:73-81. [PMID: 18157820 DOI: 10.1002/eji.200737599] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Immunotherapy generally fails to induce tumour regression in spontaneously arising tumours. Failure is attributed to both tumour-related factors and an ineffective immune response. As a model of tumour immunotherapy, without the confounding effects of potential tumour-determined mechanisms of immune evasion, we studied the requirements for rejection of skin grafts expressing a neo-self antigen in somatic cells and not in antigen-presenting cells. When antigen expression was restricted to somatic cells, both CD4(+) and CD8(+) effector cells were required for graft rejection. Although freshly placed grafts were spontaneously rejected, healed grafts established under the cover of T cell depletion were not rejected even after T cell numbers recovered to a level where freshly placed grafts on the same animal were rejected, suggesting that healed skin grafts expressing a neo-self antigen only in somatic cells could not be rejected by primed recipients with functional effector T cells. Local TLR7 ligation induced inflammatory responses and rejection of healed grafts exposed to the TLR agonist but did not induce rejection of untreated healed grafts on the same animal. Thus, local pro-inflammatory signalling via TLR7 can promote effector T cell function against skin cells displaying their nominal antigen.
Collapse
Affiliation(s)
- Jie Zhong
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, University of Queensland, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Tomai MA, Miller RL, Lipson KE, Kieper WC, Zarraga IE, Vasilakos JP. Resiquimod and other immune response modifiers as vaccine adjuvants. Expert Rev Vaccines 2007; 6:835-47. [PMID: 17931162 DOI: 10.1586/14760584.6.5.835] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synthetic immune response modifiers, such as resiquimod, are Toll-like receptor 7 and 8 agonists that act as vaccine adjuvants, enhancing antigen-specific antibody production and skewing immunity towards a Th1 response. These compounds stimulate dendritic cells to secrete cytokines, upregulate costimulatory molecule expression and enhance antigen presentation to T cells. The compounds have demonstrated vaccine adjuvant properties in a number of animal models. The adjuvant effects can be enhanced by measures that allow the drug to stay localized with the vaccine without quickly entering the systemic circulation. Clinical studies demonstrate that topical application of resiquimod and analogs is safe and effective at activating the local immune response. For injection, resiquimod or a similar compound may need to be formulated to allow for local immune activation without induction of systemic cytokines.
Collapse
Affiliation(s)
- Mark A Tomai
- 3M Drug Delivery Systems, 3M Center, 275-3E-10 St Paul, MN 55144, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Vuk-Pavlovic S. Rebuilding immunity in cancer patients. Blood Cells Mol Dis 2007; 40:94-100. [PMID: 17827037 PMCID: PMC2225479 DOI: 10.1016/j.bcmd.2007.06.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 06/30/2007] [Accepted: 06/30/2007] [Indexed: 10/22/2022]
Abstract
Rebuilding and maintaining immunity are paramount to the success of cancer immunotherapy and hematopoietic stem cell transplantation. If immune surveillance indeed can protect from cancer, the very manifestation of malignancy means that the disease has prevailed over immunity. Yet, often, tumor-specific T cells can be found in cancer patients irrespective of vaccination. Interestingly, patients suffering from malignancy often harbor unexpectedly high levels of immature CD14(+)HLA-DR(-) monocytes, although the abundance of CD4(+) cells, CD8(+) cells and CD4(+)CD25(high) cells may be normal. It is plausible that in cancer such cells suppress T cell function, analogous to CD14(+)HLA-DR(-) cells in sepsis and major trauma, in addition to their likely failure to re-present tumor-associated antigens once dendritic cells have initiated the T cell response. Recent evidence indicates that tumor-borne adenosine, lactate and hypoxia in the tumor environment may modulate tumor-specific immunity to a significant extent, but their effects on myeloid cell function are unclear. Thus, understanding and controlling these factors may appreciably impact the success of rebuilding and maintaining immunity in cancer patients.
Collapse
Affiliation(s)
- Stanimir Vuk-Pavlovic
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| |
Collapse
|
44
|
Johnston D, Zaidi B, Bystryn JC. TLR7 imidazoquinoline ligand 3M-019 is a potent adjuvant for pure protein prototype vaccines. Cancer Immunol Immunother 2007; 56:1133-41. [PMID: 17139492 PMCID: PMC11030820 DOI: 10.1007/s00262-006-0262-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 11/14/2006] [Indexed: 12/15/2022]
Abstract
Cancer vaccines, while theoretically attractive, present difficult challenges that must be overcome to be effective. Cancer vaccines are often poorly immunogenic and may require augmentation of immunogenicity through the use of adjuvants and/or immune response modifiers. Toll-like receptor (TLR) ligands are a relatively new class of immune response modifiers that may have great potential in inducing and augmenting both cellular and humoral immunity to vaccines. TLR7 ligands produce strong cellular responses and specific IgG2a and IgG2b antibody responses to protein immunogens. This study shows that a new TLR7 ligand, 3M-019, in combination with liposomes produces very strong immune responses to a pure protein prototype vaccine in mice. Female C57BL/6 mice were immunized subcutaneously with ovalbumin (OVA, 0.1 mg/dose) weekly 4x. Some groups were immunized to OVA plus 3M-019 or to OVA plus 3M-019 encapsulated in liposomes. Both antibody and cellular immune responses against OVA were measured after either two or four immunizations. Anti-OVA IgG antibody responses were significantly increased after two immunizations and were substantially higher after four immunizations in mice immunized with OVA combined with 3M-019. Encapsulation in liposomes further augmented antibody responses. IgM responses, on the other hand, were lowered by 3M-019. OVA-specific IgG2a levels were increased 625-fold by 3M-019 in liposomes compared to OVA alone, while anti-OVA IgG2b levels were over 3,000 times higher. In both cases encapsulation of 3M-019 in liposomes was stronger than either liposomes alone or 3M-019 without liposomes. Cellular immune responses were likewise increased by 3M-019 but further enhanced when it was encapsulated in liposomes. The lack of toxicity also indicates that this combination may by safe, effective method to boost immune response to cancer vaccines.
Collapse
Affiliation(s)
- Dean Johnston
- Hunter College School of Health Sciences, New York, NY, USA.
| | | | | |
Collapse
|