1
|
Yoon KW, Chu KB, Eom GD, Mao J, Heo SI, Quan FS. Dose sparing enabled by immunization with influenza vaccine using orally dissolving film. Int J Pharm 2024; 667:124945. [PMID: 39550013 DOI: 10.1016/j.ijpharm.2024.124945] [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: 09/13/2024] [Revised: 11/02/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Influenza vaccine delivered by orally dissolving film vaccine (ODFV) is a promising approach. In this study, we generated three ODFVs each comprising pulluan and trehalose with different doses of inactivated A/Puerto Rico/8/34, H1N1 virus (ODFV I, II, III) to evaluate their dose-sparing effect in mice. The ODFVs were placed on the tongues of mice to elicit immunization and after 3 immunizations at 4-week intervals, mice were challenged with a lethal dose of A/PR/8/34 to assess vaccine-induced protection. The 3 ODFVs containing 50, 250, or 750 μg of inactivated viruses elicited virus-specific antibody responses and virus neutralization in a dose-dependent manner. Dose-dependent antibody responses were also observed from the mucosal tissue samples, and also from antibody-secreting cells of the lungs and spleens. ODFV-induced cellular immunity, particularly germinal center B cells and T cells were also dose-dependent. Importantly, all 3 ODFVs evaluated in this study provided complete protection by strongly suppressing the pro-inflammatory cytokine production and lung virus titers. None of the immunized mice underwent noticeable weight loss nor succumbed to death, a phenomenon that was only observed in the infection challenge controls. These results indicated that the protection conferred by a low dose influenza vaccine formulated in ODF is comparable to that of a high-dose vaccine, thereby enabling vaccine dose sparing effect.
Collapse
Affiliation(s)
- Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ki Back Chu
- Department of Parasitology, Inje University College of Medicine, Busan 47392, Republic of Korea; Department of Infectious Disease and Malaria, Paik Institute of Clinical Research, Inje University, Busan 47392, Republic of Korea
| | - Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Su In Heo
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Mandviwala AS, Huckriede ALW, Arankalle VA, Patil HP. Mucosal delivery of a prefusogenic-F, glycoprotein, and matrix proteins-based virus-like particle respiratory syncytial virus vaccine induces protective immunity as evidenced by challenge studies in mice. Virology 2024; 598:110194. [PMID: 39096774 DOI: 10.1016/j.virol.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.
Collapse
Affiliation(s)
- Ahmedali S Mandviwala
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anke L W Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vidya A Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Harshad P Patil
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
3
|
Sallard E, Aydin M. Cutting-edge research frontiers in oral cavity vaccines for respiratory diseases: a roadmap for scientific advancement. Front Cell Infect Microbiol 2024; 14:1388222. [PMID: 38988815 PMCID: PMC11234472 DOI: 10.3389/fcimb.2024.1388222] [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: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Intramuscular vaccines present limitations in eliciting robust mucosal immunity and preventing respiratory pathogens transmission. Sublingual vaccine administration offers promising advantages, including interconnected mucosal protection. Despite these advantages, only a few clinical trials have explored sublingual vaccines, underscoring the necessity of optimizing next-generation vaccine formulas. Critical research priorities include understanding vector behavior in the oral environment, understanding their interactions with mucosal immunity and developing formulations enabling sustained mucosal contact to facilitate efficient transduction. Consequently, tonsil organoids, as representative human mucosal models, could offer critical insights into sublingual immunization. Thus, a multi-disciplinary approach integrating pharmacological, immunological, and manufacturing considerations is pivotal for sublingual vaccines in targeting pathogen-aggravated prevalent respiratory diseases including asthma, COPD and lung cancer, as well as the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Erwan Sallard
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| |
Collapse
|
4
|
Jang H, Matsuoka M, Freire M. Oral mucosa immunity: ultimate strategy to stop spreading of pandemic viruses. Front Immunol 2023; 14:1220610. [PMID: 37928529 PMCID: PMC10622784 DOI: 10.3389/fimmu.2023.1220610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Abstract
Global pandemics are most likely initiated via zoonotic transmission to humans in which respiratory viruses infect airways with relevance to mucosal systems. Out of the known pandemics, five were initiated by respiratory viruses including current ongoing coronavirus disease 2019 (COVID-19). Striking progress in vaccine development and therapeutics has helped ameliorate the mortality and morbidity by infectious agents. Yet, organism replication and virus spread through mucosal tissues cannot be directly controlled by parenteral vaccines. A novel mitigation strategy is needed to elicit robust mucosal protection and broadly neutralizing activities to hamper virus entry mechanisms and inhibit transmission. This review focuses on the oral mucosa, which is a critical site of viral transmission and promising target to elicit sterile immunity. In addition to reviewing historic pandemics initiated by the zoonotic respiratory RNA viruses and the oral mucosal tissues, we discuss unique features of the oral immune responses. We address barriers and new prospects related to developing novel therapeutics to elicit protective immunity at the mucosal level to ultimately control transmission.
Collapse
Affiliation(s)
- Hyesun Jang
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
| | - Michele Matsuoka
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
| | - Marcelo Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
5
|
Nair VV, Cabrera P, Ramírez-Lecaros C, Jara MO, Brayden DJ, Morales JO. Buccal delivery of small molecules and biologics: Of mucoadhesive polymers, films, and nanoparticles - An update. Int J Pharm 2023; 636:122789. [PMID: 36868332 DOI: 10.1016/j.ijpharm.2023.122789] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/08/2023] [Accepted: 02/25/2023] [Indexed: 03/05/2023]
Abstract
Buccal delivery of small and large molecules is an attractive route of administration that has been studied extensively over the past few decades. This route bypasses first-pass metabolism and can be used to deliver therapeutics directly to systemic circulation. Moreover, buccal films are efficient dosage forms for drug delivery due to their simplicity, portability, and patient comfort. Films have traditionally been formulated using conventional techniques, including hot-melt extrusion and solvent casting. However, newer methods are now being exploited to improve the delivery of small molecules and biologics. This review discusses recent advances in buccal film manufacturing, using the latest technologies, such as 2D and 3D printing, electrospraying, and electrospinning. This review also focuses on the excipients used in the preparation of these films, with emphasis on mucoadhesive polymers and plasticizers. Along with advances in manufacturing technology, newer analytical tools have also been used for the assessment of permeation of the active agents across the buccal mucosa, the most critical biological barrier and limiting factor of this route. Additionally, preclinical and clinical trial challenges are discussed, and some small molecule products already on the market are explored.
Collapse
Affiliation(s)
- Varsha V Nair
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Pablo Cabrera
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Independencia, Santiago 8380494, Chile; Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile
| | | | - Miguel O Jara
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, Belfield, Dublin D04 V1W8, Ireland
| | - Javier O Morales
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Independencia, Santiago 8380494, Chile; Center of New Drugs for Hypertension (CENDHY), Santiago 8380492, Chile; Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8380492, Chile.
| |
Collapse
|
6
|
Sublingual immunisation with GBS serotype III capsular polysaccharide-tetanus toxoid conjugate vaccine induces systemic and mucosal antibody responses which are opsonophagocytic and inhibit GBS colonisation of vaginal epithelial cells. Vaccine 2022; 40:6055-6063. [PMID: 36096970 DOI: 10.1016/j.vaccine.2022.08.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022]
Abstract
No vaccines are currently licensed against Group B streptococcus (GBS), an important cause of morbidity and mortality in babies and adults. Using a mouse model, and in vitro opsonophagocytosis and colonisation assays, we evaluated the potential of a sublingually-administered polysaccharide-conjugate vaccine against GBS serotype III. Sublingual immunisation of mice with 10 µg of GBS conjugate vaccine once a week for 5 weeks induced a substantial systemic IgG anti-polysaccharide response which was similar to the level induced by subcutaneous immunsation. In addition, sublingual immunisation also induced mucosal (IgA) antibody responses in the mouth, intestines and vagina. Immune sera and intestinal washes were functionally active at mediating killing of the homologous GBS serotype III in an opsonophagocytosis assay. In addition, intestinal and vaginal washes inhibited the colonisation of mouse vaginal epithelial cells by the vaccine homologous strain. These results suggest that, in addition to the induction of high levels of IgG antibodies that could be transduced from the immunised mother to the foetus to protect the newborn against GBS infection, sublingual immunisation can elicit a substantial mucosal antibody response which might play an important role in the prevention of GBS colonisation in immunised women, thereby eliminating the risk of GBS transmission from the mother to the baby during pregnancy or at birth.
Collapse
|
7
|
Garcia‐del Rio L, Diaz‐Rodriguez P, Pedersen GK, Christensen D, Landin M. Sublingual Boosting with a Novel Mucoadhesive Thermogelling Hydrogel Following Parenteral CAF01 Priming as a Strategy Against Chlamydia trachomatis. Adv Healthc Mater 2022; 11:e2102508. [PMID: 35124896 PMCID: PMC11468966 DOI: 10.1002/adhm.202102508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/18/2022] [Indexed: 01/13/2023]
Abstract
Chlamydia trachomatis is the most prevalent sexually transmitted disease of bacterial origin. The high number of asymptomatic cases makes it difficult to stop the transmission, requiring vaccine development. Herein, a strategy is proposed to obtain local genital tract immunity against C. trachomatis through parenteral prime and sublingual boost. Subcutaneous administration of chlamydia CTH522 subunit vaccine loaded in the adjuvant CAF01 is combined with sublingual administration of CTH522 loaded in a novel thermosensitive and mucoadhesive hydrogel. Briefly, a ternary optimized hydrogel (OGEL) with desirable biological and physicochemical properties is obtained using artificial intelligence techniques. This formulation exhibits a high gel strength and a strong mucoadhesive, adhesive and cohesive nature. The thermosensitive properties of the hydrogel facilitate application under the tongue. Meanwhile the fast gelation at body temperature together with rapid antigen release should avoid CTH522 leakage by swallowing and increase the contact with sublingual tissue, thus promoting absorption. In vivo studies demonstrate that parenteral-sublingual prime-boost immunization, using CAF01 and OGEL as CTH522 vaccine carriers, shows a tendency to increase cellular (Th1/Th17) immune responses when compared to mucosal or parenteral vaccination alone. Furthermore, parenteral prime with CAF01/CTH522 followed by sublingual boosting with OGEL/CTH522 elicits a local IgA response in the genital tract.
Collapse
Affiliation(s)
- Lorena Garcia‐del Rio
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Patricia Diaz‐Rodriguez
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Dennis Christensen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Mariana Landin
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| |
Collapse
|
8
|
Trincado V, Gala RP, Morales JO. Buccal and Sublingual Vaccines: A Review on Oral Mucosal Immunization and Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101177. [PMID: 34696284 PMCID: PMC8539688 DOI: 10.3390/vaccines9101177] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, most vaccines available on the market are for parental use; however, this may not be the best option on several occasions. Mucosal routes of administration such as intranasal, sublingual, and buccal generate great interest due to the benefits they offer. These range from increasing patient compliance to inducing a more effective immune response than that achieved through conventional routes. Due to the activation of the common mucosal immune system, it is possible to generate an effective systemic and local immune response, which is not achieved through parenteral administration. Protection against pathogens that use mucosal entry routes is provided by an effective induction of mucosal immunity. Mucosal delivery systems are being developed, such as films and microneedles, which have proven to be effective, safe, and easy to administer. These systems have multiple advantages over commonly used injections, which are simple to manufacture, stable at room temperature, painless for the patient since they do not require puncture. Therefore, these delivery systems do not require to be administered by medical personnel; in fact, they could be self-administered.
Collapse
Affiliation(s)
- Valeria Trincado
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
| | - Rikhav P. Gala
- Biotechnology Division, Center Mid-Atlantic, Fraunhofer USA, Newark, DE 19702, USA;
| | - Javier O. Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
- Correspondence:
| |
Collapse
|
9
|
Ciccolella M, Andreone S, Mancini J, Sestili P, Negri D, Pacca AM, D’Urso MT, Macchia D, Canese R, Pang K, SaiYing Ko T, Decadt Y, Schiavoni G, Mattei F, Belardelli F, Aricò E, Bracci L. Anticancer Effects of Sublingual Type I IFN in Combination with Chemotherapy in Implantable and Spontaneous Tumor Models. Cells 2021; 10:845. [PMID: 33917958 PMCID: PMC8068355 DOI: 10.3390/cells10040845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Salivary gland tumors are a heterogeneous group of neoplasms representing less than 10% of all head and neck tumors. Among salivary gland tumors, salivary duct carcinoma (SDC) is a rare, but highly aggressive malignant tumor resembling ductal breast carcinoma. Sublingual treatments are promising for SDC due to the induction of both local and systemic biological effects and to reduced systemic toxicity compared to other administration routes. In the present study, we first established that the sublingual administration of type I IFN (IFN-I) is safe and feasible, and exerts antitumor effects both as monotherapy and in combination with chemotherapy in transplantable tumor models, i.e., B16-OVA melanoma and EG.7-OVA lymphoma. Subsequently, we proved that sublingual IFN-I in combination with cyclophosphamide (CTX) induces a long-lasting reduction of tumor mass in NeuT transgenic mice that spontaneously develop SDC. Most importantly, tumor shrinkage in NeuT transgenic micewas accompanied by the emergence of tumor-specific cellular immune responses both in the blood and in the tumor tissue. Altogether, these results provide evidence that sublingual IFN holds promise in combination with chemotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Maria Ciccolella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Paola Sestili
- National Center for the Control and Evaluation of Medicines, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anna Maria Pacca
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Maria Teresa D’Urso
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Daniele Macchia
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Rossella Canese
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ken Pang
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Thomas SaiYing Ko
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Yves Decadt
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Filippo Belardelli
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 00133 Rome, Italy;
| | - Eleonora Aricò
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| |
Collapse
|
10
|
Sheng A, Lin L, Zhu J, Zhuang J, Li J, Chang L, Cheng H. Micro/nanodevices for assessment and treatment in stomatology and ophthalmology. MICROSYSTEMS & NANOENGINEERING 2021; 7:11. [PMID: 33532080 PMCID: PMC7844113 DOI: 10.1038/s41378-021-00238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/19/2020] [Accepted: 12/09/2020] [Indexed: 05/09/2023]
Abstract
Micro/nanodevices have been widely applied for the real-time monitoring of intracellular activities and the delivery of exogenous substances in the past few years. This review focuses on miniaturized micro/nanodevices for assessment and treatment in stomatology and ophthalmology. We first summarize the recent progress in this field by examining the available materials and fabrication techniques, device design principles, mechanisms, and biosafety aspects of micro/nanodevices. Following a discussion of biochemical sensing technology from the cellular level to the tissue level for disease assessment, we then summarize the use of microneedles and other micro/nanodevices in the treatment of oral and ocular diseases and conditions, including oral cancer, eye wrinkles, keratitis, and infections. Along with the identified key challenges, this review concludes with future directions as a small fraction of vast opportunities, calling for joint efforts between clinicians and engineers with diverse backgrounds to help facilitate the rapid development of this burgeoning field in stomatology and ophthalmology.
Collapse
Affiliation(s)
- An’an Sheng
- The Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering; School of Biological Science and Medical Engineering, Beihang University, 100191 Beijing, China
- Department of Stomatology, Xiang’An Hospital of Xiamen University, 361100 Xiamen, China
- School of Stomatology, North China University of Science and Technology, 063210 Tangshan, China
| | - Long Lin
- The Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering; School of Biological Science and Medical Engineering, Beihang University, 100191 Beijing, China
- Institute of Plastic Machinery and Plastic Engineering, School of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Jia Zhu
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA 16802 USA
| | - Jian Zhuang
- Institute of Plastic Machinery and Plastic Engineering, School of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Jian Li
- Department of Stomatology, Xiang’An Hospital of Xiamen University, 361100 Xiamen, China
| | - Lingqian Chang
- The Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering; School of Biological Science and Medical Engineering, Beihang University, 100191 Beijing, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, 230032 Hefei, China
| | - Huanyu Cheng
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
11
|
Fragoso-Saavedra M, Vega-López MA. Induction of mucosal immunity against pathogens by using recombinant baculoviral vectors: Mechanisms, advantages, and limitations. J Leukoc Biol 2020; 108:835-850. [PMID: 32392638 DOI: 10.1002/jlb.4mr0320-488r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/19/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Over 90% of pathogens of medical importance invade the organism through mucosal surfaces, which makes it urgent to develop safe and effective mucosal vaccines and mucosal immunization protocols. Besides, parenteral immunization does not provide adequate protective immunity in mucosal surfaces. Effective mucosal vaccination could protect local and systemic compartments and favor herd immunity. Although various mucosal adjuvants and Ag-delivery systems have been developed, none has filled the gap to control diseases caused by complex mucosal pathogens. Among the strategies to counteract them, recombinant virions from the baculovirus Autographa californica multiple nucleopolyhedrovirus (rAcMNPV) are useful vectors, given their safety and efficacy to produce mucosal and systemic immunity in animal infection models. Here, we review the immunogenic properties of rAcMNPV virions from the perspectives of mucosal immunology and vaccinology. Some features, which are analyzed and extrapolated from studies with different particulate antigens, include size, shape, surface molecule organization, and danger signals, all needed to break the tolerogenic responses of the mucosal immune tissues. Also, we present a condensed discussion on the immunity provided by rAcMNPV virions against influenza virus and human papillomavirus in animal models. Through the text, we highlight the advantages and limitations of this experimental immunization platform.
Collapse
Affiliation(s)
- Mario Fragoso-Saavedra
- Laboratorio de Inmunobiología de las Mucosas, Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Colonia Zacatenco, Ciudad de México, México
| | - Marco A Vega-López
- Laboratorio de Inmunobiología de las Mucosas, Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Colonia Zacatenco, Ciudad de México, México
| |
Collapse
|
12
|
Development of an Orodispersible Film Containing Stabilized Influenza Vaccine. Pharmaceutics 2020; 12:pharmaceutics12030245. [PMID: 32182676 PMCID: PMC7150837 DOI: 10.3390/pharmaceutics12030245] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/29/2022] Open
Abstract
Most influenza vaccines are administered via injection, which is considered as user-unfriendly. Vaccination via oral cavity using an orodispersible film (ODF) might be a promising alternative. To maintain the antigenicity of the vaccine during preparation and subsequent storage of these ODFs, sugars such as trehalose and pullulan can be employed as stabilizing excipients for the antigens. In this study, first, β-galactosidase was used as a model antigen. Solutions containing β-galactosidase and sugar (trehalose or trehalose/pullulan blends) were pipetted onto plain ODFs and then either air- or vacuum-dried. Subsequently, sugar ratios yielding the highest β-galactosidase stability were used to prepare ODFs containing H5N1 whole inactivated influenza virus vaccine (WIV). The stability of the H5N1 hemagglutinin was assessed by measuring its hemagglutination activity. Overall, various compositions of trehalose and pullulan successfully stabilized β-galactosidase and WIV in ODFs. WIV incorporated in ODFs showed excellent stability even at challenging storage conditions (60 °C/0% relative humidity or 30 °C/56% relative humidity) for 4 weeks. Except for sugars, the polymeric component of ODFs, i.e., hypromellose, possibly improved stability of WIV as well. In conclusion, ODFs may be suitable for delivering of WIV to the oral cavity and can possibly serve as an alternative for injections.
Collapse
|
13
|
Lomeli-Martinez SM, Valentin-Goméz E, Varela-Hernández JJ, Alvarez-Zavala M, Sanchez-Reyes K, Ramos-Solano M, Cabrera-Silva RI, Ramirez-Anguiano VM, Lomeli-Martinez MA, Martinez-Salazar SY, González-Hernández LA, Andrade-Villanueva JF. Candida spp. Determination and Th1/Th2 Mixed Cytokine Profile in Oral Samples From HIV+ Patients With Chronic Periodontitis. Front Immunol 2019; 10:1465. [PMID: 31316513 PMCID: PMC6610488 DOI: 10.3389/fimmu.2019.01465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Chronic periodontitis (CP), caused by bacteria and fungi, appears in up to 66% of HIV-patients. The impact and association of HIV-treatment (HAART) and Candida itself has not been properly evaluated in the development and progression of CP. The immunopathogenesis is characterized by CD4+ T-cells activation and the balance between the T-helper 1 (Th1) and T-helper 2 (Th2) or a mixed cytokine profile. Currently, the associated causes of an immune response in HIV-patients with CP is controversial. Our aims were the determination of Candida spp. and cytokine profile in oral samples from HIV-positive patients with CP, considering the CD4+ T cells levels and HAART use. Methods: From 500 HIV-positive patients evaluated, 228 patients were enrolled. Patients were separated in groups: (A) n = 53 (≤200 CD4+ T-cells on HAART); (B) n = 57 (≤200 CD4+ T-cells without HAART); (C) n = 50 (>200 CD4+ T-cells without HAART); (D) n = 68 (>200 CD4+ T-cells on HAART). Candida spp. were isolated from the oral biofilm and crevicular fluid in CHROMagar and confirmed by endpoint PCR. Cytokine levels were measured by beads-based immunoassay in saliva by flow cytometry. Results: 147 patients (64.5%) were positive to Candida spp. and 204 strains were isolated; 138 (67.6%) were C. albicans and the remaining C. non-albicans species (C. glabrata>C. tropicalis>C. krusei>C. dubliniensis). In this study, CHROMagar showed good sensitivity (95%) but poor specificity (68%); since of the 152 samples identified as C. albicans, only 131 were confirmed by PCR; from the 10 samples identified as C. glabrata, only six were confirmed. Finally, of the 42 samples detected as C. tropicalis, only five were confirmed. When evaluating Candida spp. presence, group A and D had higher isolation, while group B had the highest species diversity. Whereas, group C had a significant reduction of Candida spp. Despite the presence of Candida and HAART, we found a Th1/Th2 hybrid profile in the saliva of patients with low CD4+ T-cell count (group A). Conclusion: Abundance and diversity of the Candida spp. detected in HIV-patients with CP could be related to HAART and low CD4+ T-cells levels. Also, the immunosuppression might promote a local Th1/Th2 hybrid cytokine profile.
Collapse
Affiliation(s)
- Sarah M Lomeli-Martinez
- Department of Wellbeing and Sustainable Development, Centro Universitario del Norte, University of Guadalajara, Colotlán, Mexico.,Biological and Agricultural Sciences Ph.D. Program, Centro Universitario de la Ciénega, University of Guadalajara, Ocotlán, Mexico
| | - Eulogio Valentin-Goméz
- GMCA Research Unit, Department of Microbiology and Ecology, University of Valencia, Valencia, Spain.,Severe Infection Group, Health Research Institute "La Fe,", Valencia, Spain
| | - Juan J Varela-Hernández
- Department of Medical and Life Sciences, Centro Universitario de la Ciénega, University of Guadalajara, Ocotlán, Mexico
| | - Monserrat Alvarez-Zavala
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico
| | - Karina Sanchez-Reyes
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico
| | - Moises Ramos-Solano
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico
| | - Rodolfo I Cabrera-Silva
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico
| | - Victor M Ramirez-Anguiano
- Department of Integrated Dentistry Clinics, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara, Mexico
| | - Manuel A Lomeli-Martinez
- Department of Wellbeing and Sustainable Development, Centro Universitario del Norte, University of Guadalajara, Colotlán, Mexico
| | - Silvia Y Martinez-Salazar
- Department of Medical and Life Sciences, Centro Universitario de la Ciénega, University of Guadalajara, Ocotlán, Mexico
| | - Luz A González-Hernández
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico.,HIV Unit Department, University Hospital "Fray Antonio Alcalde," University of Guadalajara, Guadalajara, Mexico
| | - Jaime F Andrade-Villanueva
- HIV and Immunodeficiencies Research Institute, Clinical Medicine Department, Centro Universitario de Ciencias de la Salud-University of Guadalajara, Guadalajara, Mexico.,HIV Unit Department, University Hospital "Fray Antonio Alcalde," University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
14
|
Soria I, López-Relaño J, Viñuela M, Tudela JI, Angelina A, Benito-Villalvilla C, Díez-Rivero CM, Cases B, Manzano AI, Fernández-Caldas E, Casanovas M, Palomares O, Subiza JL. Oral myeloid cells uptake allergoids coupled to mannan driving Th1/Treg responses upon sublingual delivery in mice. Allergy 2018; 73:875-884. [PMID: 29319882 PMCID: PMC5947296 DOI: 10.1111/all.13396] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Polymerized allergoids coupled to nonoxidized mannan (PM-allergoids) may represent novel vaccines targeting dendritic cells (DCs). PM-allergoids are better captured by DCs than native allergens and favor Th1/Treg cell responses upon subcutaneous injection. Herein we have studied in mice the in vivo immunogenicity of PM-allergoids administered sublingually in comparison with native allergens. METHODS Three immunization protocols (4-8 weeks long) were used in Balb/c mice. Serum antibody levels were tested by ELISA. Cell responses (proliferation, cytokines, and Tregs) were assayed by flow cytometry in spleen and lymph nodes (LNs). Allergen uptake was measured by flow cytometry in myeloid sublingual cells. RESULTS A quick antibody response and higher IgG2a/IgE ratio were observed with PM-allergoids. Moreover, stronger specific proliferative responses were seen in both submandibular LNs and spleen cells assayed in vitro. This was accompanied by a higher IFNγ/IL-4 ratio with a quick IL-10 production by submandibular LN cells. An increase in CD4+ CD25high FOXP3+ Treg cells was detected in LNs and spleen of mice treated with PM-allergoids. These allergoids were better captured than native allergens by antigen-presenting (CD45+ MHC-II+ ) cells obtained from the sublingual mucosa, including DCs (CD11b+ ) and macrophages (CD64+ ). Importantly, all the differential effects induced by PM-allergoids were abolished when using oxidized instead of nonoxidized PM-allergoids. CONCLUSION Our results demonstrate for the first time that PM-allergoids administered through the sublingual route promote the generation of Th1 and FOXP3+ Treg cells in a greater extent than native allergens by mechanisms that might well involve their better uptake by oral antigen-presenting cells.
Collapse
Affiliation(s)
- I. Soria
- Inmunotek; Alcalá de Henares Spain
| | - J. López-Relaño
- Inmunotek; Alcalá de Henares Spain
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
- Department of Immunology; School of Medicine; Complutense University of Madrid; Madrid Spain
| | - M. Viñuela
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
| | | | - A. Angelina
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | - C. Benito-Villalvilla
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | | - B. Cases
- Inmunotek; Alcalá de Henares Spain
| | | | | | | | - O. Palomares
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | |
Collapse
|
15
|
Busignies V, Simon G, Mollereau G, Bourry O, Mazel V, Rosa-Calatrava M, Tchoreloff P. Development and pre-clinical evaluation in the swine model of a mucosal vaccine tablet for human influenza viruses: A proof-of-concept study. Int J Pharm 2018; 538:87-96. [DOI: 10.1016/j.ijpharm.2018.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 01/09/2023]
|
16
|
Abstract
The prostate secretes immunoglobulin (Ig) A (IgA) and IgG; however, how immunoglobulins reach the secretion, where the plasma cells are located, whether immunoglobulins are antigen-specific and where activation of the adaptive response occurs are still unknown. Immune cells, including CD45RA+ cells, were scattered in the stroma and not organized mucosae-associated lymphoid-tissue. IgA (but not IgG) immunostaining identified stromal plasma cells and epithelial cells in non-immunized rats. Injected tetramethylrhodamine-IgA transcytosed the epithelium along with polymeric immunoglobulin receptor. Oral immunization with ovalbumin/mesopourous SBA-15 silica adjuvant resulted in more stromal CD45RA+/IgA+ cells, increased content of ovalbumin-specific IgA and IgG, and the appearance of intraepithelial CD45RA+/IgG+ cells. An increased number of dendritic cells that cooperate in other sites with transient immunocompetent lymphocytes, and the higher levels of interleukin-1β, interferon-γ and transforming growth factor-β, explain the levels of specific antibodies. Nasal immunization produced similar results except for the increase in dendritic cells. This immunomodulatory strategy seems useful to boost immunity against genitourinary infections and, perhaps, cancer.
Collapse
|
17
|
Morales JO, Brayden DJ. Buccal delivery of small molecules and biologics: of mucoadhesive polymers, films, and nanoparticles. Curr Opin Pharmacol 2017; 36:22-28. [PMID: 28800417 DOI: 10.1016/j.coph.2017.07.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 01/28/2023]
Abstract
Buccal delivery of macromolecules (biologics) sets a great challenge for researchers. Although several niche small molecule products have been approved as simple sprays, tablets and oral films, it is not simply a case of adapting existing technologies to biologics. Buccal delivery of insulin has reached clinical trials with two approaches: oromucosal sprays of the peptide with permeation enhancers, and embedded gold nanoparticles in a dissolvable film. However, neither of these approaches have led to FDA approvals likely due to poor efficacy, submaximal peptide loading in the dosage form, and to wide intra-subject variability in pharmacokinetics and pharmacodynamics. It is likely however that printed film designs with lower molecular weight stable biotech payloads including lipophilic glucagon-like 1 (GLP-1) agonists and macrocycles with long half-lives will generate greater efficacy than was achieved to date for insulin.
Collapse
Affiliation(s)
- Javier O Morales
- Department of Pharmaceutical Science and Technology, School of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380494, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile; Pharmaceutical Biomaterial Research Group, Department of Health Sciences, Luleå University of Technology, Luleå 97187, Sweden.
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
18
|
Schulze K, Ebensen T, Riese P, Prochnow B, Lehr CM, Guzmán CA. New Horizons in the Development of Novel Needle-Free Immunization Strategies to Increase Vaccination Efficacy. Curr Top Microbiol Immunol 2017; 398:207-234. [PMID: 27370343 DOI: 10.1007/82_2016_495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The young twenty-first century has already brought several medical advances, such as a functional artificial human liver created from stem cells, improved antiviral (e.g., against HIV) and cancer (e.g., against breast cancer) therapies, interventions controlling cardiovascular diseases, and development of new and optimized vaccines (e.g., HPV vaccine). However, despite this substantial progress and the achievements of the last century, humans still suffer considerably from diseases, especially from infectious diseases. Thus, almost one-fourth of all deaths worldwide are caused directly or indirectly by infectious agents. Although vaccination has led to the control of many diseases, including smallpox, diphtheria, and tetanus, emerging diseases are still not completely contained. Furthermore, pathogens such as Bordetella pertussis undergo alterations making adaptation of the respective vaccine necessary. Moreover, insufficient implementation of vaccination campaigns leads to re-emergence of diseases which were believed to be already under control (e.g., poliomyelitis). Therefore, novel vaccination strategies need to be developed in order to meet the current challenges including lack of compliance, safety issues, and logistic constraints. In this context, mucosal and transdermal approaches constitute promising noninvasive vaccination strategies able to match these demands.
Collapse
Affiliation(s)
- Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Blair Prochnow
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Braunschweig, Germany.,Department of Pharmacy, Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| |
Collapse
|
19
|
Martin TL, Jee J, Kim E, Steiner HE, Cormet-Boyaka E, Boyaka PN. Sublingual targeting of STING with 3'3'-cGAMP promotes systemic and mucosal immunity against anthrax toxins. Vaccine 2017; 35:2511-2519. [PMID: 28343781 DOI: 10.1016/j.vaccine.2017.02.064] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
Anthrax is caused by Bacillus anthracis, a zoonotic bacterial pathogen affecting humans and livestock worldwide. The current human anthrax vaccine, anthrax vaccine adsorbed (AVA), is an injected vaccine with a cumbersome administration schedule and fails to promote mucosal immunity. Bacterial enterotoxins, which stimulate production of the cyclic nucleotide cAMP are effective experimental mucosal vaccine adjuvants, but their inherent toxicity has precluded their use in humans. We investigated whether cyclic dinucleotides that target Stimulator of Interferon Gamma Genes (STING) in mammalian cells could represent an alternative to bacterial enterotoxins as adjuvant for sublingual immunization and promotion of mucosal immunity and secretory IgA responses in addition to systemic immunity. We found that sublingual immunization of mice with Bacillus anthracis protective antigen (PA) and the STING ligand 3'3'-cGAMP promotes PA-specific serum IgG Ab responses of the same magnitude as those induced after immunization with PA and the experimental adjuvant cholera toxin (CT). Interestingly, this STING ligand also promoted serum anti-PA IgA and IgA-producing cells in the bone marrow. Furthermore, the saliva of mice immunized with the STING ligand exhibited similar levels of PA-specific IgA Abs as groups immunized with CT as adjuvant. The adjuvant activity of 3'3'-cGAMP was associated with mixed Th1, Th2, and Th17 responses. This STING ligand also induced rapid IFN-β and IL-10 responses in sublingual tissues and cervical lymph nodes, and TGF-β responses in the cervical lymph nodes, which could contribute to promoting IgA responses after sublingual immunization.
Collapse
Affiliation(s)
- Tara L Martin
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Junbae Jee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Haley E Steiner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
20
|
Gala RP, Popescu C, Knipp GT, McCain RR, Ubale RV, Addo R, Bhowmik T, Kulczar CD, D’Souza MJ. Physicochemical and Preclinical Evaluation of a Novel Buccal Measles Vaccine. AAPS PharmSciTech 2017; 18:283-292. [PMID: 27357420 DOI: 10.1208/s12249-016-0566-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/03/2016] [Indexed: 11/30/2022] Open
Abstract
The aim of this study is to develop an orally disintegrating film (ODF) containing a microparticulate measles vaccine formulation for buccal delivery. The measles vaccine microparticles were made with biocompatible and biodegradable bovine serum albumin (BSA) and processed by spray drying. These vaccine microparticles were incorporated in the ODF, consisting of Lycoat RS720®, Neosorb P60W® and Tween 80. The yield of the microparticles was approximately 85-95%, w/w. The mean size of the vaccine microparticles was 3.65 ± 1.89 μm and had a slightly negative surface charge of 32.65 ± 2.4 mV. The vaccine particles were nontoxic to normal cells at high concentrations (500 μg/2.5 × 105 cells) of vaccine particles. There was a significant induction of innate immune response by vaccine microparticles which was observed in vitro when compared to blank microparticles (P < 0.05). The vaccine microparticles also significantly increased the antigen presentation and co-stimulatory molecules expression on antigen presenting cells, which is a prerequisite for Th1 and Th2 immune responses. When the ODF vaccine formulation was dosed in juvenile pigs, significantly higher antibody titers were observed after week 2, with a significant increase at week 4 and plateauing through week 6 comparative to naïve predose titers. The results suggest that the ODF measles vaccine formulation is a viable dosage form alternative to noninvasive immunization that may increase patient compliance and commercial distribution.
Collapse
|
21
|
Bekri S, Bourdely P, Luci C, Dereuddre-Bosquet N, Su B, Martinon F, Braud VM, Luque I, Mateo PL, Crespillo S, Conejero-Lara F, Moog C, Le Grand R, Anjuère F. Sublingual Priming with a HIV gp41-Based Subunit Vaccine Elicits Mucosal Antibodies and Persistent B Memory Responses in Non-Human Primates. Front Immunol 2017; 8:63. [PMID: 28203239 PMCID: PMC5285372 DOI: 10.3389/fimmu.2017.00063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/16/2017] [Indexed: 01/01/2023] Open
Abstract
Persistent B cell responses in mucosal tissues are crucial to control infection against sexually transmitted pathogens like human immunodeficiency virus 1 (HIV-1). The genital tract is a major site of infection by HIV. Sublingual (SL) immunization in mice was previously shown to generate HIV-specific B cell immunity that disseminates to the genital tract. We report here the immunogenicity in female cynomolgus macaques of a SL vaccine based on a modified gp41 polypeptide coupled to the cholera toxin B subunit designed to expose hidden epitopes and to improve mucosal retention. Combined SL/intramuscular (IM) immunization with such mucoadhesive gp41-based vaccine elicited mucosal HIV-specific IgG and IgA antibodies more efficiently than IM immunization alone. This strategy increased the number and duration of gp41-specific IgA secreting cells. Importantly, combined immunization improved the generation of functional antibodies 3 months after vaccination as detected in HIV-neutralizing assays. Therefore, SL immunization represents a promising vaccine strategy to block HIV-1 transmission.
Collapse
Affiliation(s)
- Selma Bekri
- Université Côte d'Azur, Nice, France; CNRS UMR7275, IPMC, Valbonne, France
| | - Pierre Bourdely
- Université Côte d'Azur, Nice, France; CNRS UMR7275, IPMC, Valbonne, France
| | - Carmelo Luci
- Université Côte d'Azur, Nice, France; CNRS UMR7275, IPMC, Valbonne, France; INSERM, Paris, France
| | - Nathalie Dereuddre-Bosquet
- CEA, Université Paris Sud, INSERM U1184 "Immunology of Viral Infections and Autoimmune Diseases" , Fontenay-aux-Roses , France
| | - Bin Su
- INSERM, Unit 1109 INSERM/UNISTRA, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France; Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Frédéric Martinon
- CEA, Université Paris Sud, INSERM U1184 "Immunology of Viral Infections and Autoimmune Diseases" , Fontenay-aux-Roses , France
| | - Véronique M Braud
- Université Côte d'Azur, Nice, France; CNRS UMR7275, IPMC, Valbonne, France
| | - Irene Luque
- Departamento de Química Física e Instituto de Biotecnología, Universidad de Granada , Granada , Spain
| | - Pedro L Mateo
- Departamento de Química Física e Instituto de Biotecnología, Universidad de Granada , Granada , Spain
| | - Sara Crespillo
- Departamento de Química Física e Instituto de Biotecnología, Universidad de Granada , Granada , Spain
| | - Francisco Conejero-Lara
- Departamento de Química Física e Instituto de Biotecnología, Universidad de Granada , Granada , Spain
| | - Christiane Moog
- INSERM, Unit 1109 INSERM/UNISTRA, Fédération de Médecine Translationnelle de Strasbourg , Strasbourg , France
| | - Roger Le Grand
- CEA, Université Paris Sud, INSERM U1184 "Immunology of Viral Infections and Autoimmune Diseases" , Fontenay-aux-Roses , France
| | - Fabienne Anjuère
- Université Côte d'Azur, Nice, France; CNRS UMR7275, IPMC, Valbonne, France; INSERM, Paris, France
| |
Collapse
|
22
|
Preparation of Multifunctional Liposomes as a Stable Vaccine Delivery-Adjuvant System by Procedure of Emulsification-Lyophilization. Methods Mol Biol 2016; 1404:635-649. [PMID: 27076327 DOI: 10.1007/978-1-4939-3389-1_41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Liposomes have been proven to be useful carriers for vaccine antigens and can be modified as a versatile vaccine adjuvant-delivery system (VADS). To fulfill efficiently both functions of adjuvant and delivery, the liposomes are often modified with different functional molecules, such as lipoidal immunopotentiators, APC (antigen-presenting cell) targeting ligands, steric stabilization polymers, and charged lipids. Also, to overcome the weakness of instability, vaccines are often lyophilized as a dry product. In this chapter the procedure of emulsification-lyophilization (PEL) is introduced as an efficient method for preparing a stable anhydrous precursor to the multifunctional liposomes which bear dual modifications with APC targeting molecule of the mannosylated cholesterol and the adjuvant material of monophosphoryl lipid A. The techniques and procedures for synthesis of APC targeting molecule, i.e., the mannosylated cholesterol, and for characterization of the multifunctional liposomes are also described.
Collapse
|
23
|
Seth A, Kong IG, Lee SH, Yang JY, Lee YS, Kim Y, Wibowo N, Middelberg AP, Lua LH, Kweon MN. Modular virus-like particles for sublingual vaccination against group A streptococcus. Vaccine 2016; 34:6472-6480. [DOI: 10.1016/j.vaccine.2016.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/28/2016] [Accepted: 11/04/2016] [Indexed: 02/05/2023]
|
24
|
Sjökvist Ottsjö L, Jeverstam F, Yrlid L, Wenzel AU, Walduck AK, Raghavan S. Induction of mucosal immune responses against Helicobacter pylori infection after sublingual and intragastric route of immunization. Immunology 2016; 150:172-183. [PMID: 27676456 DOI: 10.1111/imm.12676] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/04/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022] Open
Abstract
There is a current lack of effective mucosal vaccines against major gastroenteric pathogens and particularly against Helicobacter pylori, which causes a chronic infection that can lead to peptic ulcers and gastric cancer in a subpopulation of infected individuals. Mucosal CD4+ T-cell responses have been shown to be essential for vaccine-induced protection against H. pylori infection. The current study addresses the influence of the adjuvant and site of mucosal immunization on early CD4+ T-cell priming to H. pylori antigens. The vaccine formulation consisted of H. pylori lysate antigens and mucosal adjuvants, cholera toxin (CT) or a detoxified double-mutant heat-labile enterotoxin from Escherichia coli (dmLT), which were administered by either the sublingual or intragastric route. We report that in vitro, adjuvants CT and dmLT induce up-regulation of pro-inflammatory gene expression in purified dendritic cells and enhance the H. pylori-specific CD4+ T-cell response including interleukin-17A (IL-17A), interferon-γ (IFN-γ) and tumour necrosis factor-α (TNF-α) secretion. In vivo, sublingual immunization led to an increased frequency of IL-17A+ , IFN-γ+ and TNF-α+ secreting CD4+ T cells in the cervical lymph nodes compared with in the mesenteric lymph nodes after intragastric immunization. Subsequently, IL-17A+ cells were visualized in the stomach of sublingually immunized and challenged mice. In summary, our results suggest that addition of an adjuvant to the vaccine clearly activated dendritic cells, which in turn, enhanced CD4+ T-cell cytokines IL-17A, IFN-γ and TNF-α responses, particularly in the cervical lymph nodes after sublingual vaccination.
Collapse
Affiliation(s)
- Louise Sjökvist Ottsjö
- Department of Microbiology & Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Frida Jeverstam
- Department of Microbiology & Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linda Yrlid
- Department of Microbiology & Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexander U Wenzel
- Department of Microbiology & Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna K Walduck
- School of Science, RMIT University, Bundoora, Vic., Australia
| | - Sukanya Raghavan
- Department of Microbiology & Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Zhang M, Sun C, Gu J, Yan X, Wang B, Cui Z, Sun X, Tong C, Feng X, Lei L, Han W. Salmonella Typhimurium strain expressing OprF-OprI protects mice against fatal infection by Pseudomonas aeruginosa. Microbiol Immunol 2016; 59:533-44. [PMID: 26249788 DOI: 10.1111/1348-0421.12291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/29/2015] [Accepted: 08/02/2015] [Indexed: 01/16/2023]
Abstract
Pseudomonas aeruginosa poses a major threat to human health and to the mink industry. Thus, development of vaccines that elicit robust humoral and cellular immunity against P. aeruginosa is greatly needed. In this study, a recombinant attenuated Salmonella vaccine (RASV) that expresses the outer membrane proteins fusion OprF190-342 -OprI21-83 (F1I2) from P. aeruginosa was constructed and the potency of this vaccine candidate assessed by measuring F1I2-specific humoral immune responses upon vaccination through s.c. or oral routes. S.C. administration achieved higher serum IgG titers and IgA titers in the intestine and induced stronger F1I2-specific IgG and IgA titers in lung homogenate than did oral administration, which resulted in low IgG titers and no local IgA production. High titers of IFN-γ, IL-4, and T-lymphocyte subsets induced a mixed Th1/Th2 response in mice immunized s.c., indicating elicitation of cellular immunity. Importantly, when immunized mice were challenged with P. aeruginosa by the intranasal route 30 days after the initial immunization, s.c. vaccination achieved 77.78% protection, in contrast to 41.18% via oral administration and 66.67% via Escherichia coli-expressed F1I2 (His-F1I2) vaccination. These results indicate that s.c. vaccination provides a better protective response against P. aeruginosa infection than do oral administration and the His-F1I2 vaccine.
Collapse
Affiliation(s)
| | | | | | - Xinwu Yan
- College of Animal Science, Jilin University, No. 5333, Xi'an Street, Changchun, Jilin, 130062
| | | | | | | | | | | | | | - Wenyu Han
- College of Veterinary Medicine.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, People's Republic of China
| |
Collapse
|
26
|
van der Meulen TA, Harmsen HJM, Bootsma H, Spijkervet FKL, Kroese FGM, Vissink A. The microbiome-systemic diseases connection. Oral Dis 2016; 22:719-734. [DOI: 10.1111/odi.12472] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 12/28/2022]
Affiliation(s)
- TA van der Meulen
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - HJM Harmsen
- Department of Medical Microbiology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - H Bootsma
- Department of Rheumatology and Clinical Immunology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - FKL Spijkervet
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - FGM Kroese
- Department of Rheumatology and Clinical Immunology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - A Vissink
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
27
|
Preparation of the Multifunctional Liposome-Containing Microneedle Arrays as an Oral Cavity Mucosal Vaccine Adjuvant-Delivery System. Methods Mol Biol 2016; 1404:651-667. [PMID: 27076328 DOI: 10.1007/978-1-4939-3389-1_42] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, the multifunctional liposome-constituted microneedle arrays (LiposoMAs) have been proven to be an interesting vaccine adjuvant-delivery system (VADS) that are stable and can be vaccinated via oral cavity mucosal route. When given to mice at oral mucosa, the LiposoMAs can effectively eliminate the ingredient loss caused by chewing, swallowing, and saliva flowing and can, thus, elicit robust systemic as well as mucosal immunoresponses against the loaded antigens. In addition, the LiposoMAs can induce a mixed Th1/Th2 immunoresponse and strong cellular/humoral immunity due to special adjuvanticity and targeting delivery functions of the nanoparticulate VADS. In this chapter, the preparation, characterization as well as mucosal vaccination of the LiposoMAs are introduced. In addition, the methods for sampling mouse organs, tissues, and cells and for evaluation of the immunization efficacy are mainly included.
Collapse
|
28
|
Multifunctional liposomes constituting microneedles induced robust systemic and mucosal immunoresponses against the loaded antigens via oral mucosal vaccination. Vaccine 2015; 33:4330-40. [PMID: 25858854 DOI: 10.1016/j.vaccine.2015.03.081] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 03/12/2015] [Accepted: 03/24/2015] [Indexed: 11/20/2022]
Abstract
To develop effective, convenient and stable mucosal vaccines, mannose-PEG-cholesterol (MPC)/lipid A-liposomes (MLLs) entrapping model antigen bovine serum albumin (BSA) were prepared by the procedure of emulsification-lyophilization and used to constitute microneedles, forming the proMLL-filled microneedle arrays (proMMAs). The proMMAs were rather stable and hard enough to pierce porcine skin and, upon rehydration, dissolved rapidly recovering the MLLs without size and entrapment change. The proMMAs given to mice via oral mucosal (o.m.) route, rather than routine intradermal administration, elicited robust systemic and mucosal immunoresponses against the loaded antigens as evidenced by high levels of BSA-specific IgG in the sera and IgA in the salivary, intestinal and vaginal secretions of mice. Enhanced levels of IgG2a and IFN-γ in treated mice revealed that proMMAs induced a mixed Th1/Th2 immunoresponse. Moreover, a significant increase in CD8(+) T cells confirmed that strong cellular immunity had also been established by the immunization of the proMMAs. Thus, the proMMAs can be immunized via o.m. route to set up an effective multiple defense against pathogen invasion and may be an effective vaccine adjuvant-delivery system (VADS) applicable in the controlled temperature chain.
Collapse
|
29
|
Vacher G, Sublet E, Gurny R, Borchard G. Establishment and first characterization of a sublingual epithelial and immune cell co-culture model. Int J Pharm 2015; 482:61-7. [PMID: 25448555 DOI: 10.1016/j.ijpharm.2014.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/11/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
We describe here the establishment and first characterization of a co-culture model of human epithelial sublingual cells (HO-1-u-1 cell line) and human dendritic cells derived from human peripheral blood monocytes (PBMC). Cell culture conditions for HO-1-u-1 cells were optimized. First characterization of phenotypic features by electron microscopy and fluorescence imaging revealed resemblance to sublingual tissue specimen from healthy donors. Successful co-culturing of epithelial and dendritic cells (DCs) was confirmed by confocal laser scanning microscopy. Stimulation of HO-1-u1 cells alone and the epithelial/DC co-culture by incubation with liposomes, virosomes and influenza virus lead reproducibly to the release of inflammatory cytokine GM-CSF. This co-culture model may be suitable for elucidation of mechanisms involved in the immune response at the sublingual epithelium as well as for the evaluation of novel topical vaccines, potentially replacing cumbersome ex vivo and in vivo methods currently in place.
Collapse
Affiliation(s)
- Gaëlle Vacher
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Emmanuelle Sublet
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Robert Gurny
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, 1211 Geneva, Switzerland.
| |
Collapse
|
30
|
Wang T, Zhen Y, Ma X, Wei B, Li S, Wang N. Mannosylated and lipid A-incorporating cationic liposomes constituting microneedle arrays as an effective oral mucosal HBV vaccine applicable in the controlled temperature chain. Colloids Surf B Biointerfaces 2015; 126:520-30. [PMID: 25612819 DOI: 10.1016/j.colsurfb.2015.01.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/11/2014] [Accepted: 01/04/2015] [Indexed: 10/24/2022]
Abstract
To develop an effective, convenient and stable mucosal vaccine against hepatitis B virus (HBV), the mannose-PEG-cholesterol/lipid A-liposomes (MLLs) loaded with HBsAg were prepared by the procedure of emulsification-lyophilization and, subsequently, filled into the microholes of microneedle array reverse molds and dried to form the proHBsAg-MLLs microneedle arrays (proHMAs). The proHMAs were stable even at 40 °C for up to 3 days and hard enough to pierce porcine skin but, upon rehydration, rapidly dissolved recovering the HBsAg-MLLs without obvious changes in size and antigen association efficiency. Notably, immunization of mice only once with the proHMAs at oral mucosa induced robust systemic and widespread mucosal immunoresponses, as evidenced by the high levels of HBsAg-specific IgG in the sera and IgA in the salivary, intestinal and vaginal secretions. In addition, a strong cellular immunity against HBV had been established through a mixed Th1/Th2 response, as confirmed by a significant increase in CD8(+) T cells as well as the enhanced levels of IgG2a and IFN-γ in the treated mice. Thus, the proHMAs can be conveniently vaccinated via oral mucosal route to set up a multiple immune defense against HBV invasion and, in addition, may be a stable HBV vaccine applicable in the controlled temperature chain for wide distribution.
Collapse
Affiliation(s)
- Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| | - Yuanyuan Zhen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Xiaoyu Ma
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Biao Wei
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Shuqin Li
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ning Wang
- School of Medical Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui Province 230009, China.
| |
Collapse
|
31
|
Singh S, Yang G, Byrareddy SN, Barry MA, Sastry KJ. Natural killer T cell and TLR9 agonists as mucosal adjuvants for sublingual vaccination with clade C HIV-1 envelope protein. Vaccine 2014; 32:6934-6940. [PMID: 25444819 DOI: 10.1016/j.vaccine.2014.10.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/22/2014] [Accepted: 10/23/2014] [Indexed: 02/06/2023]
Abstract
The vast majority of HIV-1 infections occur at mucosa during sexual contact. It may therefore be advantageous to provide mucosal barrier protection against this entry by mucosal vaccination. While a number of mucosal routes of vaccination are possible, many like enteric oral vaccines or intranasal vaccines have significant impediments that limit vaccine efficacy or pose safety risks. In contrast, immunogens applied to the sublingual region of the mouth could provide a simple route for mucosal vaccination. While sublingual immunization is appealing, this site does not always drive strong immune responses, particularly when using protein antigens. To address this issue, we have tested the ability of two mucosal adjuvants: alpha-galactosylceramide (αGalCer) that is a potent stimulator of natural killer T cells and CpG-oligodeoxynucleotide (CpG-ODN) a TLR9 agonist for their ability to amplify immune responses against clade C gp140 HIV-1 envelope protein antigen. Immunization with envelope protein alone resulted in a weak T cell and antibody responses. In contrast, CD4(+) and CD8(+) T cells responses in systemic and mucosal tissues were significantly higher in mice immunized with gp140 in the presence of either αGalCer or CpG-ODN and these responses were further augmented when the two adjuvants were used together. While both the adjuvants effectively increased gp140-specific serum IgG and vaginal IgA antibody levels, combining both significantly improved these responses. Memory T cell responses 60 days after immunization revealed αGalCer to be more potent than CpG-ODN and the combination of the αGalCer and CpG-ODN adjuvants was more effective than either alone. Serum and vaginal washes collected 60 days after immunization with gp140 with both αGalCer and CpG-ODN adjuvants had significant neutralization activity against Tier 1 and Tier 2 SHIVs. These data support the utility of the sublingual route for mucosal vaccination particularly in combination with αGalCer and CpG-ODN adjuvants.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Guojun Yang
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Siddappa N Byrareddy
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Michael A Barry
- Department of Internal Medicine, Division of Infectious Diseases, Translational Immunovirology Program, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States; Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX, United States.
| |
Collapse
|
32
|
Zhang C, Ohno T, Kang S, Takai T, Azuma M. Repeated antigen painting and sublingual immunotherapy in mice convert sublingual dendritic cell subsets. Vaccine 2014; 32:5669-76. [PMID: 25168308 DOI: 10.1016/j.vaccine.2014.08.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 07/21/2014] [Accepted: 08/08/2014] [Indexed: 12/24/2022]
Abstract
The sublingual mucosa (SLM) is utilized as the site for sublingual immunotherapy (SLIT) to induce tolerance against allergens. The contribution of SLM-dendritic cells (SLM-DCs) has not been clarified. The aim of this study was to examine the dynamics and phenotype of SLM-DCs after topical antigen painting and SLIT. SLM-DCs were histologically evaluated after FITC painting. A novel murine Japanese cedar pollinosis (JCP) model was generated and change in SLM-DCs after SLIT was examined. The density of SLM-DCs was clearly lower compared with the buccal mucosa and dorsal surface of the tongue. Topical FITC painting on the SLM induced maximal recruitment of submucosal DCs (smDCs) at 6h, but most smDCs had vanished at 24h. Repeated painting on the SLM induced exhaustion and conversion of the smDC phenotype. CD206(high)CD11c(low) round-type cells with fewer dendrites and less lymph node migration capacity became dominant. In the murine model of JCP, SLIT efficiently inhibited clinical symptoms and allergen-mediated immunological responses. SLIT markedly reduced the number of SLM-DCs, converted to the round-type dominant phenotype and inhibited the activation of regional lymph node DCs. Topical antigen painting on the SLM induced rapid exhaustion and conversion of smDCs. The unique dynamics of SLM-DCs may contribute to tolerance induction in SLIT.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Tatsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Siwen Kang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan
| | - Toshiro Takai
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8547, Japan.
| |
Collapse
|
33
|
Oral transmucosal drug delivery for pediatric use. Adv Drug Deliv Rev 2014; 73:50-62. [PMID: 23999459 DOI: 10.1016/j.addr.2013.08.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 07/13/2013] [Accepted: 08/22/2013] [Indexed: 12/25/2022]
Abstract
The formulation of medicines for children remains a challenge. An ideal pediatric formulation must allow accurate dose administration and be in a dosage form that can be handled by the target age group. It is also important to consider the choices and the amount of excipients used in the formulation for this vulnerable age group. Although oral formulations are generally acceptable to most pediatric patients, they are not suitable for drugs with poor oral bioavailability or when a rapid clinical effect is required. In recent years, oral transmucosal delivery has emerged as an attractive route of administration for pediatric patients. With this route of administration, a drug is absorbed through the oral mucosa, therefore bypassing hepatic first pass metabolism and thus avoiding drug degradation or metabolism in the gastrointestinal tract. The high blood flow and relatively high permeability of the oral mucosa allow a quick onset of action to be achieved. It is a simple and non-invasive route of drug administration. However, there are several barriers that need to be overcome in the development of oral transmucosal products. This article aims to provide a comprehensive review of the current development of oral transmucosal delivery specifically for the pediatric population in order to achieve systemic drug delivery. The anatomical and physiological properties of the oral mucosa of infants and young children are carefully examined. The different dosage forms and formulation strategies that are suitable for young patients are discussed.
Collapse
|
34
|
Wang N, Wang T, Zhang M, Chen R, Niu R, Deng Y. Mannose derivative and lipid A dually decorated cationic liposomes as an effective cold chain free oral mucosal vaccine adjuvant-delivery system. Eur J Pharm Biopharm 2014; 88:194-206. [PMID: 24769065 DOI: 10.1016/j.ejpb.2014.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/14/2014] [Accepted: 04/16/2014] [Indexed: 01/24/2023]
Abstract
To develop convenient, effective cold chain-free subunit vaccines, a mannose-PEG-cholesterol conjugate (MPC) was synthesized as a lectin binding molecule and anchored onto liposomes which entrapped lipid A and model antigen to form a vaccine adjuvant-delivery system targeting antigen presenting cells. With MPC, soy phosphatidylcholine, stearylamine and monophosphoryl lipid A as emulsifiers dissolved in oil phase (O), and sucrose and BSA in water phase (W), the O/W emulsions were prepared and subsequently lyophilized. The lyophilized product was stable enough to be stored at room temperature and, upon rehydration, formed MPC-/lipid A-liposomes (MLLs) with a size under 300 nm and antigen association rates of around 36%. The MLLs given to mice via oral mucosal (o.m.) administration showed no side effects but induced potent immune responses as evidenced by the high levels of IgG in the sera and IgA in the salivary, intestinal and vaginal secretions of mice. High levels of IgG2a and IFN-γ in treated mice revealed that MLLs via o.m. vaccination induced a mixed Th1/Th2 response against antigens, establishing both humoral and cellular immunity. Thus, the MLLs may be a potent cold chain-free oral mucosal vaccine adjuvant-delivery system.
Collapse
Affiliation(s)
- Ning Wang
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China
| | - Ting Wang
- Department of Pharmacy, Anhui Medical University, Hefei, China; Department of Pharmacy, Jining Medical College, Sunshine City, China.
| | - Meiling Zhang
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Ruonan Chen
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Ruowen Niu
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Yihui Deng
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
35
|
Baptista AAS, Donato TC, Garcia KCOD, Gonçalves GAM, Coppola MP, Okamoto AS, Sequeira JL, Andreatti Filho RL. Immune response of broiler chickens immunized orally with the recombinant proteins flagellin and the subunit B of cholera toxin associated with Lactobacillus spp. Poult Sci 2014; 93:39-45. [PMID: 24570421 DOI: 10.3382/ps.2013-03372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study investigated the immune response of broiler chickens with oral treatment of a Lactobacillus spp. pool (PL) associated with microencapsulated recombinant proteins flagellin (FliC) and the subunit B of cholera toxin (CTB). Immune responses were evaluated by measuring IgA from intestinal fluid, serum IgY, and immunostaining of CD8(+) T lymphocytes present in the cecum. The evaluations were performed on d 0, 7, 14, 21, and 28 posttreatment. A significant increase (P < 0.05) was observed in IgA levels in all immunized groups, especially 3 wk after immunization. Treatments 2 (recombinant CTB) and 3 (recombinant FliC+CTB) showed the highest concentrations. Similarly, serum concentrations IgY (μg/mL) increased along the experiment, and the means for treatments 2 and 3 showed significant differences (P < 0.05) compared with controls, reaching concentrations of 533 and 540 μg/mL, respectively. The number of CD8(+) T lymphocytes in all treatments greatly differed (P < 0.05) compared with the negative control at 21 d posttreatment. However, only treatment 2 (recombinant CTB), 4 (PL), and 5 (recombinant FliC+ recombinant CTB + PL) remained significantly (P < 0.05) different from the control at 28 d posttreatment. Thus, it is concluded that the microencapsulated recombinant proteins administered orally to broiler chickens are capable of stimulating humoral and cellular immune response, and the combinations of these antigens with Lactobacillus spp. can influence the population of CD8(+) T cells residing in the cecum.
Collapse
Affiliation(s)
- A A S Baptista
- College of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Sao Paulo, Brazil 18618-970
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Wang N, Wang T, Zhang M, Chen R, Deng Y. Using procedure of emulsification-lyophilization to form lipid A-incorporating cochleates as an effective oral mucosal vaccine adjuvant-delivery system (VADS). Int J Pharm 2014; 468:39-49. [PMID: 24704308 DOI: 10.1016/j.ijpharm.2014.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 03/07/2014] [Accepted: 04/02/2014] [Indexed: 12/22/2022]
Abstract
Using a procedure of emulsification-lyophilization (PEL), adjuvant lipid A-cochleates (LACs) were prepared as a carrier for model antigen bovine serum albumin (BSA). With phosphatidylserine and lipid A as emulsifiers dissolved in oil phase (O), sucrose and CaCl2 in the inner water phase (W1), and BSA, sucrose and PEG2000 in the outer water phase (W2), the W1/O/W2 emulsions were prepared and subsequently lyophilized to form a dry product which was stable enough to be stored at room temperature. Upon rehydration of the dry products, cochleates formed with a size of 800 nm and antigen association rates of 38%. After vaccination of mice through oral mucosal (o.m.) administration, LACs showed no side effects but induced potent immune responses as evidenced by high levels of IgG in the sera and IgA in the salivary, intestinal and vaginal secretions of mice. In addition, high levels of IgG2a and IFN-γ in the sera or culture supernatants of splenocytes of the immunized mice were also detected. These results revealed that LACs induced a mixed Th1/Th2 response against the loaded antigens. Thus, the LACs prepared by PEL were able to induce both systemic and mucosal immune responses and may act as a potent cold-chain-free oral mucosal vaccine adjuvant delivery system (VADS).
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/chemistry
- Administration, Oral
- Animals
- Cells, Cultured
- Chemistry, Pharmaceutical
- Drug Carriers
- Drug Stability
- Emulsions
- Excipients/chemistry
- Female
- Freeze Drying
- Immunity, Humoral/drug effects
- Immunity, Mucosal/drug effects
- Immunoglobulin A, Secretory/metabolism
- Immunoglobulin G/blood
- Interferon-gamma/metabolism
- Lipid A/administration & dosage
- Lipid A/chemistry
- Lipid A/immunology
- Mice
- Mouth Mucosa/drug effects
- Mouth Mucosa/immunology
- Particle Size
- Phagocytosis/drug effects
- Powders
- Serum Albumin, Bovine/administration & dosage
- Serum Albumin, Bovine/chemistry
- Serum Albumin, Bovine/immunology
- Technology, Pharmaceutical/methods
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th2 Cells/drug effects
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Ning Wang
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Ting Wang
- Department of Pharmacy, Anhui Medical University, 81 Mei Hill Road, Hefei, Anhui Province 230032, China; Department of Pharmacy, Jining Medical College, 669 Xueyuan Road, Sunshine City, Shandong Province 276826, China.
| | - Meiling Zhang
- Department of Pharmacy, Anhui Medical University, 81 Mei Hill Road, Hefei, Anhui Province 230032, China
| | - Ruonan Chen
- Department of Pharmacy, Anhui Medical University, 81 Mei Hill Road, Hefei, Anhui Province 230032, China
| | - Yihui Deng
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| |
Collapse
|
37
|
Singh S, Yang G, Schluns KS, Anthony SM, Sastry KJ. Sublingual vaccination induces mucosal and systemic adaptive immunity for protection against lung tumor challenge. PLoS One 2014; 9:e90001. [PMID: 24599269 PMCID: PMC3943861 DOI: 10.1371/journal.pone.0090001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/25/2014] [Indexed: 12/26/2022] Open
Abstract
Sublingual route offers a safer and more practical approach for delivering vaccines relative to other systemic and mucosal immunization strategies. Here we present evidence demonstrating protection against ovalbumin expressing B16 (B16-OVA) metastatic melanoma lung tumor formation by sublingual vaccination with the model tumor antigen OVA plus synthetic glycolipid alpha-galactosylceramide (aGalCer) for harnessing the adjuvant potential of natural killer T (NKT) cells, which effectively bridge innate and adaptive arms of the immune system. The protective efficacy of immunization with OVA plus aGalCer was antigen-specific as immunized mice challenged with parental B16 tumors lacking OVA expression were not protected. Multiple sublingual immunizations in the presence, but not in the absence of aGalCer, resulted in repeated activation of NKT cells in the draining lymph nodes, spleens, and lungs of immunized animals concurrent with progressively increasing OVA-specific CD8+ T cell responses as well as serum IgG and vaginal IgA levels. Furthermore, sublingual administration of the antigen only in the presence of the aGalCer adjuvant effectively boosted the OVA-specific immune responses. These results support potential clinical utility of sublingual route of vaccination with aGalCer-for prevention of pulmonary metastases.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Guojun Yang
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Kimberly S. Schluns
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Immunology Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States of America
| | - Scott M. Anthony
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Immunology Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States of America
| | - K. Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Immunology Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Antigen-bearing dendritic cells from the sublingual mucosa recirculate to distant systemic lymphoid organs to prime mucosal CD8 T cells. Mucosal Immunol 2014; 7:280-91. [PMID: 23801305 DOI: 10.1038/mi.2013.45] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 05/28/2013] [Indexed: 02/04/2023]
Abstract
Effector T cells are described to be primed in the lymph nodes draining the site of immunization and to recirculate to effector sites. Sublingual immunization generates effector T cells able to disseminate to the genital tract. Herein, we report an alternative mechanism that involves the recirculation of antigen-bearing dendritic cells (DCs) in remote lymphoid organs to prime T cells. Sublingual immunization with a muco-adhesive model antigen unable to diffuse through lymphatic or blood vessels induced genital CD8 T cells. The sublingual draining lymph nodes were not mandatory to generate these lymphocytes, and antigen-bearing DCs from distant lymph nodes and spleen were able to prime specific CD8 T cells in a time- and dose-dependent manner. This study demonstrates, for the first time, that antigen-bearing DCs originating from the site of immunization recirculate to distant lymphoid organs and provides insights into the mechanism of distant CD8 T-cell generation by sublingual immunization.
Collapse
|
39
|
Murugappan S, Patil HP, Frijlink HW, Huckriede A, Hinrichs WLJ. Simplifying influenza vaccination during pandemics: sublingual priming and intramuscular boosting of immune responses with heterologous whole inactivated influenza vaccine. AAPS JOURNAL 2014; 16:342-9. [PMID: 24482005 DOI: 10.1208/s12248-014-9565-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/06/2014] [Indexed: 11/30/2022]
Abstract
The best approach to control the spread of influenza virus during a pandemic is vaccination. Yet, an appropriate vaccine is not available early in the pandemic since vaccine production is time consuming. For influenza strains with a high pandemic potential like H5N1, stockpiling of vaccines has been considered but is hampered by rapid antigenic drift of the virus. It has, however, been shown that immunization with a given H5N1 strain can prime the immune system for a later booster with a drifted variant. Here, we investigated whether whole inactivated virus (WIV) vaccine can be processed to tablets suitable for sublingual (s.l.) use and whether s.l. vaccine administration can prime the immune system for a later intramuscular (i.m.) boost with a heterologous vaccine. In vitro results demonstrate that freeze-drying and tableting of WIV did not affect the integrity of the viral proteins or the hemagglutinating properties of the viral particles. Immunization experiments revealed that s.l. priming with WIV (prepared from the H5N1 vaccine strain NIBRG-14) 4 weeks prior to i.m. booster immunization with the same virus strongly enhanced hemagglutination-inhibition (HI) titers against NIBRG-14 and the drifted variant NIBRG-23. Moreover, s.l. (and i.m.) immunization with NIBRG-14 also primed for a subsequent heterologous i.m. booster immunization with NIBRG-23 vaccine. In addition to HI serum antibodies, s.l. priming enhanced lung and nose IgA responses, while i.m. priming enhanced lung IgA but not nose IgA levels. Our results identify s.l. vaccination as a user-friendly method to prime for influenza-specific immune responses toward homologous and drifted variants.
Collapse
Affiliation(s)
- Senthil Murugappan
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands,
| | | | | | | | | |
Collapse
|
40
|
White JA, Blum JS, Hosken NA, Marshak JO, Duncan L, Zhu C, Norton EB, Clements JD, Koelle DM, Chen D, Weldon WC, Steven Oberste M, Lal M. Serum and mucosal antibody responses to inactivated polio vaccine after sublingual immunization using a thermoresponsive gel delivery system. Hum Vaccin Immunother 2014; 10:3611-21. [PMID: 25483682 PMCID: PMC4514067 DOI: 10.4161/hv.32253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/23/2014] [Accepted: 08/04/2014] [Indexed: 01/27/2023] Open
Abstract
Administering vaccines directly to mucosal surfaces can induce both serum and mucosal immune responses. Mucosal responses may prevent establishment of initial infection at the port of entry and subsequent dissemination to other sites. The sublingual route is attractive for mucosal vaccination, but both a safe, potent adjuvant and a novel formulation are needed to achieve an adequate immune response. We report the use of a thermoresponsive gel (TRG) combined with a double mutant of a bacterial heat-labile toxin (dmLT) for sublingual immunization with a trivalent inactivated poliovirus vaccine (IPV) in mice. This TRG delivery system, which changes from aqueous solution to viscous gel upon contact with the mucosa at body temperature, helps to retain the formulation at the site of delivery and has functional adjuvant activity from the inclusion of dmLT. IPV was administered to mice either sublingually in the TRG delivery system or intramuscularly in phosphate-buffered saline. We measured poliovirus type-specific serum neutralizing antibodies as well as polio-specific serum Ig and IgA antibodies in serum, saliva, and fecal samples using enzyme-linked immunosorbent assays. Mice receiving sublingual vaccination via the TRG delivery system produced both mucosal and serum antibodies, including IgA. Intramuscularly immunized animals produced only serum neutralizing and binding Ig but no detectable IgA. This study provides proof of concept for sublingual immunization using the TRG delivery system, comprising a thermoresponsive gel and dmLT adjuvant.
Collapse
Key Words
- CT, cholera toxin
- DPBS, Dulbecco's phosphate-buffered saline
- DU, D-antigen units
- ELISA, enzyme-linked immunosorbent assay
- IM, intramuscular
- IPV, inactivated poliovirus vaccine
- IgA, immunoglobulin A
- IgG, immunoglobulin G
- OPV, oral poliovirus vaccine
- PBS, phosphate-buffered saline
- RT, room temperature
- SL, sublingual
- SSI, Staten Serum Institute
- TMB, tetramethylbenzidine
- TRG, thermoresponsive gel
- adjuvants
- dmLT
- dmLT, double mutant heat-labile toxin
- mucosal immune response
- poliovirus
- sublingual immunization
- thermoresponsive gel
- vaccine delivery
Collapse
Affiliation(s)
| | | | - Nancy A Hosken
- Department of Medicine; University of Washington; Seattle, WA USA
| | - Joshua O Marshak
- Department of Medicine; University of Washington; Seattle, WA USA
| | | | | | - Elizabeth B Norton
- Department of Microbiology and Immunology; Tulane University School of Medicine; New Orleans, LA USA
| | - John D Clements
- Department of Microbiology and Immunology; Tulane University School of Medicine; New Orleans, LA USA
| | - David M Koelle
- Department of Medicine; University of Washington; Seattle, WA USA
- Department of Laboratory Medicine; University of Washington; Seattle, WA USA
- Vaccine and Infectious Diseases Division; Fred Hutchinson Cancer Research Institute; Seattle, WA USA
- Department of Global Health; University of Washington; Seattle, WA USA
| | | | - William C Weldon
- Division of Viral Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | - M Steven Oberste
- Division of Viral Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | | |
Collapse
|
41
|
Abstract
The oral cavity contains distinct mucosal surfaces, each with its own unique distribution of dendritic cell (DC) subsets. In addition to tissue-specific properties, such organization might confer differential immune outcomes guided by tissue-resident DCs, which translate in the lymph node into an overall immune response. This process is further complicated by continual exposure and colonization of the oral cavity with enormous numbers of diverse microbes, some of which might induce destructive immunity. As a central cell type constantly monitoring changes in oral microbiota and orchestrating T-cell function, oral DCs are of major importance in deciding whether to induce immunity or tolerance. In this review, an overview of the phenotype and distribution of DCs in the oral mucosa is provided. In addition, the role of the various oral DC subsets in inducing immunity vs. tolerance, as well as their involvement in several oral pathologies is discussed.
Collapse
|
42
|
Kim JY, Choi Y, Nguyen HH, Song MK, Chang J. Mucosal immunization with recombinant adenovirus encoding soluble globular head of hemagglutinin protects mice against lethal influenza virus infection. Immune Netw 2013; 13:275-82. [PMID: 24385946 PMCID: PMC3875786 DOI: 10.4110/in.2013.13.6.275] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/06/2013] [Accepted: 12/06/2013] [Indexed: 12/15/2022] Open
Abstract
Influenza virus is one of the major sources of respiratory tract infection. Due to antigenic drift in surface glycoproteins the virus causes annual epidemics with severe morbidity and mortality. Although hemagglutinin (HA) is one of the highly variable surface glycoproteins of the influenza virus, it remains the most attractive target for vaccine development against seasonal influenza infection because antibodies generated against HA provide virus neutralization and subsequent protection against the virus infection. Combination of recombinant adenovirus (rAd) vector-based vaccine and mucosal administration is a promising regimen for safe and effective vaccination against influenza. In this study, we constructed rAd encoding the globular head region of HA from A/Puerto Rico/8/34 virus as vaccine candidate. The rAd vaccine was engineered to express high level of the protein in secreted form. Intranasal or sublingual immunization of mice with the rAd-based vaccine candidates induced significant levels of sustained HA-specific mucosal IgA and IgG. When challenged with lethal dose of homologous virus, the vaccinated mice were completely protected from the infection. The results demonstrate that intranasal or sublingual vaccination with HA-encoding rAd elicits protective immunity against infection with homologous influenza virus. This finding underlines the potential of our recombinant adenovirus-based influenza vaccine candidate for both efficacy and rapid production.
Collapse
Affiliation(s)
- Joo Young Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Youngjoo Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Huan H Nguyen
- Laboratory Science Division, International Vaccine Institute, Seoul 151-919, Korea
| | - Man Ki Song
- Laboratory Science Division, International Vaccine Institute, Seoul 151-919, Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| |
Collapse
|
43
|
Hernández-Santos N, Huppler AR, Peterson AC, Khader SA, McKenna KC, Gaffen SL. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol 2013; 6:900-10. [PMID: 23250275 PMCID: PMC3608691 DOI: 10.1038/mi.2012.128] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 11/13/2012] [Indexed: 02/04/2023]
Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection caused by Candida albicans. Despite its prevalence, little is known about C. albicans-specific immunity in the oral mucosa. Vaccines against Candida generate both T helper type 1 (Th1) and Th17 responses, and considerable evidence implicates interleukin (IL)-17 in immunity to OPC. However, IL-17 is also produced by innate immune cells that are remarkably similar to Th17 cells, expressing the same markers and localizing to similar mucosal sites. To date, the relative contribution(s) of Th1, Th17, and innate IL-17-producing cells in OPC have not been clearly defined. Here, we sought to determine the nature and function of adaptive T-cell responses to OPC, using a new recall infection model. Mice subjected to infection and re-challenge with Candida mounted a robust and stable antigen-specific IL-17 response in CD4+ but not CD8+ T cells. There was little evidence for Th1 or Th1/Th17 responses. The Th17 response promoted accelerated fungal clearance, and Th17 cells could confer protection in Rag1-/- mice upon adoptive transfer. Surprisingly, CD4 deficiency did not cause OPC but was instead associated with compensatory IL-17 production by Tc17 and CD3+CD4-CD8- cells. Therefore, classic CD4+Th17 cells protect from OPC but can be compensated by other IL-17-producing cells in CD4-deficient hosts.
Collapse
Affiliation(s)
| | - Anna R. Huppler
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh
| | - Alanna C. Peterson
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh
| | | | | | - Sarah L. Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh,Correspondence: Division of Rheumatology & Clinical Immunology, BST S703, 3500 Terrace St, Pittsburgh PA 15261, USA. 412-383-8903, Fax: 412-383-8864,
| |
Collapse
|
44
|
Choi JH, Schafer SC, Zhang L, Juelich T, Freiberg AN, Croyle MA. Modeling pre-existing immunity to adenovirus in rodents: immunological requirements for successful development of a recombinant adenovirus serotype 5-based ebola vaccine. Mol Pharm 2013; 10:3342-55. [PMID: 23915419 DOI: 10.1021/mp4001316] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pre-existing immunity (PEI) to human adenovirus serotype 5 (Ad5) worldwide is the primary limitation to routine clinical use of Ad5-based vectors in immunization platforms. Using systemic and mucosal PEI induction models in rodents (mice and guinea pigs), we assessed the influence of PEI on the type of adaptive immune response elicited by an Ad5-based vaccine for Ebola with respect to immunization route. Splenocytes isolated from vaccinated animals revealed that immunization by the same route in which PEI was induced significantly compromised Ebola Zaire glycoprotein (ZGP)-specific IFN-γ+ CD8+ T cells and ZGP-specific multifunctional CD8+ T cell populations. ZGP-specific IgG1 antibody levels were also significantly reduced and a sharp increase in serum anti-Ad5 neutralizing antibody (NAB) titers were noted following immunization. These immune parameters correlated with poor survival after lethal challenge with rodent-adapted Ebola Zaire virus (ZEBOV). Although the number of IFN-γ+ CD8+ T cells was reduced in animals given the vaccine by a different route from that used for PEI induction, the multifunctional CD8+ T cell response was not compromised. Survival rates in these groups were higher than when PEI was induced by the same route as immunization. These results suggest that antigen-specific multifunctional CD8(+) T cell and Th2 type antibody responses compromised by PEI to Ad5 are required for protection from Ebola. They also illustrate that methods for induction of PEI used in preclinical studies must be carefully evaluated for successful development of novel Ad5-based vaccines.
Collapse
Affiliation(s)
- Jin Huk Choi
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | | | | | | | | | | |
Collapse
|
45
|
Brandtzaeg P. Secretory IgA: Designed for Anti-Microbial Defense. Front Immunol 2013; 4:222. [PMID: 23964273 PMCID: PMC3734371 DOI: 10.3389/fimmu.2013.00222] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/16/2013] [Indexed: 01/30/2023] Open
Abstract
Prevention of infections by vaccination remains a compelling goal to improve public health. Mucosal vaccines would make immunization procedures easier, be better suited for mass administration, and most efficiently induce immune exclusion - a term coined for non-inflammatory antibody shielding of internal body surfaces, mediated principally by secretory immunoglobulin A (SIgA). The exported antibodies are polymeric, mainly IgA dimers (pIgA), produced by local plasma cells (PCs) stimulated by antigens that target the mucose. SIgA was early shown to be complexed with an epithelial glycoprotein - the secretory component (SC). A common SC-dependent transport mechanism for pIgA and pentameric IgM was then proposed, implying that membrane SC acts as a receptor, now usually called the polymeric Ig receptor (pIgR). From the basolateral surface, pIg-pIgR complexes are taken up by endocytosis and then extruded into the lumen after apical cleavage of the receptor - bound SC having stabilizing and innate functions in the secretory antibodies. Mice deficient for pIgR show that this is the only receptor responsible for epithelial export of IgA and IgM. These knockout mice show a variety of defects in their mucosal defense and changes in their intestinal microbiota. In the gut, induction of B-cells occurs in gut-associated lymphoid tissue, particularly the Peyer's patches and isolated lymphoid follicles, but also in mesenteric lymph nodes. PC differentiation is accomplished in the lamina propria to which the activated memory/effector B-cells home. The airways also receive such cells from nasopharynx-associated lymphoid tissue but by different homing receptors. This compartmentalization is a challenge for mucosal vaccination, as are the mechanisms used by the mucosal immune system to discriminate between commensal symbionts (mutualism), pathobionts, and overt pathogens (elimination).
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
46
|
Li H, Reeves RK. Functional perturbation of classical natural killer and innate lymphoid cells in the oral mucosa during SIV infection. Front Immunol 2013; 3:417. [PMID: 23316201 PMCID: PMC3539714 DOI: 10.3389/fimmu.2012.00417] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/20/2012] [Indexed: 12/25/2022] Open
Abstract
Despite the fact that the majority of human pathogens are transmitted across mucosal surfaces, including the oral mucosae, oral immunity is poorly understood. Furthermore, because the normal flora of the oral cavity is vast and significantly diverse, host immunity must balance a complex system of tolerance and pathogen recognition. Due to the rapid recognition and response to pathogens, the innate immune system, including natural killer (NK) cells, likely plays a critical role in mediating this balance. Because logistical and ethical restraints limit access to significant quantities of human mucosal tissues, non-human primate models offer one of the best opportunities to study mucosal NK cells. In this study we have identified both classical NK cells, as well as innate lymphoid cells (ILCs) in tonsillar and buccal tissues and oral-draining lymph nodes. Identified by mutually exclusive expression of NKG2A and NKp44, NK cells, and ILCs in the oral mucosa are generally phenotypically and functionally analogous to their gut counterparts. NKG2A+ NK cells were more cytotoxic while NKp44+ ILCs produced copious amounts of IL-17 and TNF-α. However, in contrast to gut, oral NK cells and ILCs both produced large quantities of IFN-γ and the beta-chemokine, MIP-1β. Also in contrast to what we have previously found in gut tissues of SIV-infected macaques, we found no reduction in NK cells during chronic SIV infection, but rather an expansion of ILCs in oral-draining lymph nodes and tonsils. These data suggest that the lentivirus-induced depletion of the NK cell/ILC compartment in the gut may be absent in the oral mucosa, but the inherent differences and SIV-induced alterations are likely to have significant impact on preventing oral opportunistic infections in lentiviral disease. Furthermore, these data extend our understanding of the oral innate immune system in general and could aid future studies evaluating the regulation of both normal oral flora and limiting transmission of oral mucosal pathogens.
Collapse
Affiliation(s)
- Haiying Li
- Division of Immunology, New England Primate Research Center, Harvard Medical School, One Pine Hill Drive Southborough, MA, USA
| | | |
Collapse
|
47
|
Dhama K, Kumar A, Kumar Verm A, Rahal A, Kumar Panw P. Recent Trends in Development of Adjuvant of Vaccine. ACTA ACUST UNITED AC 2013. [DOI: 10.3923/tmr.2013.32.35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
48
|
Buffa V, Klein K, Fischetti L, Shattock RJ. Evaluation of TLR agonists as potential mucosal adjuvants for HIV gp140 and tetanus toxoid in mice. PLoS One 2012; 7:e50529. [PMID: 23272062 PMCID: PMC3521731 DOI: 10.1371/journal.pone.0050529] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 10/23/2012] [Indexed: 11/19/2022] Open
Abstract
In the present study we investigate the impact of a range of TLR ligands and chitosan as potential adjuvants for different routes of mucosal immunisation (sublingual (SL), intranasal (IN), intravaginal (IVag) and a parenteral route (subcutaneous (SC)) in the murine model. We assess their ability to enhance antibody responses to HIV-1 CN54gp140 (gp140) and Tetanus toxoid (TT) in systemic and vaginal compartments. A number of trends were observed by route of administration. For non-adjuvanted antigen, SC>SL>IN immunisation with respect to systemic IgG responses, where endpoint titres were greater for TT than for gp140. In general, co-administration with adjuvants increased specific IgG responses where IN = SC>SL, while in the vaginal compartment IN>SL>SC for specific IgA. In contrast, for systemic and mucosal IgA responses to antigen alone SL>IN = SC. A number of adjuvants increased specific systemic IgA responses where in general IN>SL>SC immunisation, while for mucosal responses IN = SL>SC. In contrast, direct intravaginal immunisation failed to induce any detectable systemic or mucosal responses to gp140 even in the presence of adjuvant. However, significant systemic IgG responses to TT were induced by intravaginal immunisation with or without adjuvant, and detectable mucosal responses IgG and IgA were observed when TT was administered with FSL-1 or Poly I∶C. Interestingly some TLRs displayed differential activity dependent upon the route of administration. MPLA (TLR4) suppressed systemic responses to SL immunisation while enhancing responses to IN or SC immunisation. CpG B enhanced SL and IN responses, while having little or no impact on SC immunisation. These data demonstrate important route, antigen and adjuvant effects that need to be considered in the design of mucosal vaccine strategies.
Collapse
Affiliation(s)
- Viviana Buffa
- Clinical Sciences, St. George's University of London, London, United Kingdom
| | - Katja Klein
- Clinical Sciences, St. George's University of London, London, United Kingdom
| | - Lucia Fischetti
- Clinical Sciences, St. George's University of London, London, United Kingdom
| | - Robin J. Shattock
- Clinical Sciences, St. George's University of London, London, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Shin MK, Kang ML, Jung MH, Cha SB, Lee WJ, Kim JM, Kim DH, Yoo HS. Induction of protective immune responses against challenge of Actinobacillus pleuropneumoniae by oral administration with Saccharomyces cerevisiae expressing Apx toxins in pigs. Vet Immunol Immunopathol 2012. [PMID: 23206402 DOI: 10.1016/j.vetimm.2012.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Actinobacillus pleuropneumoniae is a causative agent of porcine pleuropneumonia, a highly contagious endemic disease of pigs worldwide, inducing significant economic losses worldwide. Apx toxins, which are correlated with the virulence of A. pleuropneumoniae, were expressed in Saccharomyces cerevisiae and its possible use as an oral vaccine has been confirmed in our previous studies using a murine model. The present study was undertaken to test the hypothesis that oral immunization using S. cerevisiae expressing either ApxI or ApxII could protect pigs against A. pleuropneumoniae as an effective way of inducing both mucosal and systemic immune responses. The surface-displayed ApxIIA#5 expressing S. cerevisiae was selected as an oral vaccine candidate by finding on induction of higher immune responses in mice after oral vaccination. The surface-displayed ApxIIA#5 expressing S. cerevisiae and the ApxIA expressing S. cerevisiae were developed to serve as an oral vaccine in pigs. The vaccinated pigs showed higher specific IgG- and IgA-related antibody activities than the non-treated control and vector control pigs. Additionally, the induced immune responses were found to protect pigs infected with A. pleuropneumoniae according to the analysis of clinical signs and the gross and microscopic pulmonary lesions. These results suggested that the surface-displayed ApxIIA#5 and ApxIA in S. cerevisiae might be a potential oral vaccine to protect pigs against porcine pleuropneumonia. Thus the present study is expected to contribute to the development of a live oral vaccine against porcine pleuropneumonia as an alternative to current conventional vaccines.
Collapse
Affiliation(s)
- Min-Kyoung Shin
- Department of Infectious Disease, College of Veterinary Medicine and Brain Korea 21 Program for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Signarovitz AL, Ray HJ, Yu JJ, Guentzel MN, Chambers JP, Klose KE, Arulanandam BP. Mucosal immunization with live attenuated Francisella novicida U112ΔiglB protects against pulmonary F. tularensis SCHU S4 in the Fischer 344 rat model. PLoS One 2012; 7:e47639. [PMID: 23118885 PMCID: PMC3484155 DOI: 10.1371/journal.pone.0047639] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 09/18/2012] [Indexed: 12/17/2022] Open
Abstract
The need for an efficacious vaccine against Francisella tularensis is a consequence of its low infectious dose and high mortality rate if left untreated. This study sought to characterize a live attenuated subspecies novicida-based vaccine strain (U112ΔiglB) in an established second rodent model of pulmonary tularemia, namely the Fischer 344 rat using two distinct routes of vaccination (intratracheal [i.t.] and oral). Attenuation was verified by comparing replication of U112ΔiglB with wild type parental strain U112 in F344 primary alveolar macrophages. U112ΔiglB exhibited an LD50>107 CFU compared to the wild type (LD50 = 5×106 CFU i.t.). Immunization with 107 CFU U112ΔiglB by i.t. and oral routes induced antigen-specific IFN-γ and potent humoral responses both systemically (IgG2a>IgG1 in serum) and at the site of mucosal vaccination (respiratory/intestinal compartment). Importantly, vaccination with U112ΔiglB by either i.t. or oral routes provided equivalent levels of protection (50% survival) in F344 rats against a subsequent pulmonary challenge with ∼25 LD50 (1.25×104 CFU) of the highly human virulent strain SCHU S4. Collectively, these results provide further evidence on the utility of a mucosal vaccination platform with a defined subsp. novicida U112ΔiglB vaccine strain in conferring protective immunity against pulmonary tularemia.
Collapse
Affiliation(s)
- Aimee L. Signarovitz
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Heather J. Ray
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - M. N. Guentzel
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - James P. Chambers
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Karl E. Klose
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Disease and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|