1
|
Gong X, Wani MY, Al-Bogami AS, Ahmad A, Robinson K, Khan A. The Road Ahead: Advancing Antifungal Vaccines and Addressing Fungal Infections in the Post-COVID World. ACS Infect Dis 2024; 10:3475-3495. [PMID: 39255073 DOI: 10.1021/acsinfecdis.4c00245] [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] [Indexed: 09/12/2024]
Abstract
In impoverished nations, the COVID-19 pandemic has led to a widespread occurrence of deadly fungal diseases like mucormycosis. The limited availability of effective antifungal treatments and the emergence of drug-resistant fungal strains further exacerbate the situation. Factors such as systemic steroid use, intravenous drug misuse, and overutilization of broad-spectrum antimicrobials contribute to the prevalence of hospital-acquired infections caused by drug-resistant fungi. Fungal infections exploit compromised immune status and employ intricate mechanisms to evade immune surveillance. The immune response involves the innate and adaptive immune systems, leading to phagocytic and complement-mediated elimination of fungi. However, resistance to antifungals poses a challenge, highlighting the importance of antifungal prophylaxis and therapeutic vaccination. Understanding the host-fungal immunological interactions and developing vaccines are vital in combating fungal infections. Further research is needed to address the high mortality and morbidity associated with multidrug-resistant fungal pathogens and to develop innovative treatment drugs and vaccines. This review focuses on the global epidemiological burden of fungal infections, host-fungal immunological interactions, recent advancements in vaccine development and the road ahead.
Collapse
Affiliation(s)
- Xiaolong Gong
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, 21589, Jeddah, Saudi Arabia
| | - Abdullah Saad Al-Bogami
- Department of Chemistry, College of Science, University of Jeddah, 21589, Jeddah, Saudi Arabia
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, United States
| | - Keven Robinson
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, United States
| | - Amber Khan
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
2
|
Diez A, Arrieta-Aguirre I, Carrano G, Fernandez-de-Larrinoa I, Moragues MD. A novel Candida albicans Als3, Hwp1 and Met6 derived complex peptide protects mice against hematogenously induced candidiasis. Vaccine 2024; 42:125990. [PMID: 38789371 DOI: 10.1016/j.vaccine.2024.05.038] [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: 07/18/2023] [Revised: 04/22/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024]
Abstract
Candida albicans can cause superficial or systemic infections in humans, particularly in immunocompromised individuals. Vaccination strategies targeting specific antigens of C. albicans have shown promise in providing protection against invasive candidiasis. This study aimed to evaluate the immuno-protective capacity of a KLH conjugated complex peptide, 3P-KLH, containing epitopes from C. albicans antigens Als3, Hwp1, and Met6 in a murine model of hematogenously induced candidiasis. Mice immunized with 3P-KLH raised a specific antibody response, and protection against C. albicans infection was assessed. Immunized mice exhibited significantly lower fungal load in their kidneys compared to the control group. Moreover, 37.5 % of immunized mice survived 21 days after the infection, while all control animals died within the first nine days. These findings suggest that the 3P-KLH complex peptide, targeting C. albicans key antigens, elicits a protective immune response and reduces the severity of systemic Candida infection. In addition, the high binding affinity of the selected epitopes with MHC II alleles further supports the potential immunogenicity of this peptide in humans. This research provides insights into the development of novel immunotherapeutic approaches against invasive candidiasis.
Collapse
Affiliation(s)
- Ander Diez
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country UPV/EHU, Leioa, Spain; Department of Nursing I, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Ines Arrieta-Aguirre
- Department of Nursing I, University of the Basque Country UPV/EHU, Leioa, Spain.
| | - Giulia Carrano
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | | | | |
Collapse
|
3
|
Bromuro C, Posteraro B, Murri R, Fantoni M, Tumbarello M, Sanguinetti M, Dattilo R, Cauda R, Cassone A, Torosantucci A. Identification of two anti- Candida antibodies associated with the survival of patients with candidemia. mBio 2024; 15:e0276923. [PMID: 38088540 PMCID: PMC10790786 DOI: 10.1128/mbio.02769-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
IMPORTANCE Candidemia (bloodstream invasion by Candida species) is a major fungal disease in humans. Despite the recent progress in diagnosis and treatment, therapeutic options are limited and under threat of antimicrobial resistance. The disease mortality remains high (around 40%). In contrast with deep-seated invasive candidiasis, particularly that occurring in patients with hematologic malignancies and organ transplants, patients with candidemia are often not immunocompromised and therefore able to mount memory anticandidal immune responses, perhaps primed by Candida commensalism. We investigated antibody immunity in candidemia patients and report here on the ability of these patients to produce antibodies that react with Candida antigens. In particular, the patients with high titers of IgG reactive with two immunodominant, virulence-associated antigens (Als3 and MP65) had a higher 30-day survival. If confirmed by controlled, prospective clinical studies, our data could inform the development of antibody therapy to better treat a severe fungal infection such as candidiasis.
Collapse
Affiliation(s)
- Carla Bromuro
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Brunella Posteraro
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rita Murri
- Dipartimento Salute e Bioetica, Sezione Malattie Infettive, Università Cattolica del Sacro Cuore, Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Massimo Fantoni
- Dipartimento Salute e Bioetica, Sezione Malattie Infettive, Università Cattolica del Sacro Cuore, Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Mario Tumbarello
- Dipartimento Biotecnologie Mediche, University of Siena, Siena, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rosanna Dattilo
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Cauda
- Dipartimento Salute e Bioetica, Sezione Malattie Infettive, Università Cattolica del Sacro Cuore, Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Antonio Cassone
- Polo d'Innovazione della Genomica, Genetica e Biologia, Siena, Italy
| | | |
Collapse
|
4
|
Gupta SK, Osmanoglu Ö, Minocha R, Bandi SR, Bencurova E, Srivastava M, Dandekar T. Genome-wide scan for potential CD4+ T-cell vaccine candidates in Candida auris by exploiting reverse vaccinology and evolutionary information. Front Med (Lausanne) 2022; 9:1008527. [PMID: 36405591 PMCID: PMC9669072 DOI: 10.3389/fmed.2022.1008527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2023] Open
Abstract
Candida auris is a globally emerging fungal pathogen responsible for causing nosocomial outbreaks in healthcare associated settings. It is known to cause infection in all age groups and exhibits multi-drug resistance with high potential for horizontal transmission. Because of this reason combined with limited therapeutic choices available, C. auris infection has been acknowledged as a potential risk for causing a future pandemic, and thus seeking a promising strategy for its treatment is imperative. Here, we combined evolutionary information with reverse vaccinology approach to identify novel epitopes for vaccine design that could elicit CD4+ T-cell responses against C. auris. To this end, we extensively scanned the family of proteins encoded by C. auris genome. In addition, a pathogen may acquire substitutions in epitopes over a period of time which could cause its escape from the immune response thus rendering the vaccine ineffective. To lower this possibility in our design, we eliminated all rapidly evolving genes of C. auris with positive selection. We further employed highly conserved regions of multiple C. auris strains and identified two immunogenic and antigenic T-cell epitopes that could generate the most effective immune response against C. auris. The antigenicity scores of our predicted vaccine candidates were calculated as 0.85 and 1.88 where 0.5 is the threshold for prediction of fungal antigenic sequences. Based on our results, we conclude that our vaccine candidates have the potential to be successfully employed for the treatment of C. auris infection. However, in vivo experiments are imperative to further demonstrate the efficacy of our design.
Collapse
Affiliation(s)
- Shishir K. Gupta
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
- Evolutionary Genomics Group, Center for Computational and Theoretical Biology, University of Würzburg, Würzburg, Germany
| | - Özge Osmanoglu
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
| | - Rashmi Minocha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sourish Reddy Bandi
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Elena Bencurova
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
| | - Mugdha Srivastava
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
- Core Unit Systems Medicine, University of Würzburg, Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, Functional Genomics and Systems Biology Group, University of Würzburg, Würzburg, Germany
- BioComputing Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| |
Collapse
|
5
|
The Role of B-Cells and Antibodies against Candida Vaccine Antigens in Invasive Candidiasis. Vaccines (Basel) 2021; 9:vaccines9101159. [PMID: 34696267 PMCID: PMC8540628 DOI: 10.3390/vaccines9101159] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic candidiasis is an invasive fungal infection caused by members of the genus Candida. The recent emergence of antifungal drug resistance and increased incidences of infections caused by non-albicans Candida species merit the need for developing immune therapies against Candida infections. Although the role of cellular immune responses in anti-Candida immunity is well established, less is known about the role of humoral immunity against systemic candidiasis. This review summarizes currently available information on humoral immune responses induced by several promising Candida vaccine candidates, which have been identified in the past few decades. The protective antibody and B-cell responses generated by polysaccharide antigens such as mannan, β-glucan, and laminarin, as well as protein antigens like agglutinin-like sequence gene (Als3), secreted aspartyl proteinase (Sap2), heat shock protein (Hsp90), hyphally-regulated protein (Hyr1), hyphal wall protein (Hwp1), enolase (Eno), phospholipase (PLB), pyruvate kinase (Pk), fructose bisphosphate aldolase (Fba1), superoxide dismutase gene (Sod5) and malate dehydrogenase (Mdh1), are outlined. As per studies reviewed, antibodies induced in response to leading Candida vaccine candidates contribute to protection against systemic candidiasis by utilizing a variety of mechanisms such as opsonization, complement fixation, neutralization, biofilm inhibition, direct candidacidal activity, etc. The contributions of B-cells in controlling fungal infections are also discussed. Promising results using anti-Candida monoclonal antibodies for passive antibody therapy reinforces the need for developing antibody-based therapeutics including anti-idiotypic antibodies, single-chain variable fragments, peptide mimotopes, and antibody-derived peptides. Future research involving combinatorial immunotherapies using humanized monoclonal antibodies along with antifungal drugs/cytokines may prove beneficial for treating invasive fungal infections.
Collapse
|
6
|
dos Santos Dias L, Dobson HE, Bakke BK, Kujoth GC, Huang J, Kohn EM, Taira CL, Wang H, Supekar NT, Fites JS, Gates D, Gomez CL, Specht CA, Levitz SM, Azadi P, Li L, Suresh M, Klein BS, Wüthrich M. Structural basis of Blastomyces Endoglucanase-2 adjuvancy in anti-fungal and -viral immunity. PLoS Pathog 2021; 17:e1009324. [PMID: 33735218 PMCID: PMC8009368 DOI: 10.1371/journal.ppat.1009324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 03/30/2021] [Accepted: 01/19/2021] [Indexed: 11/20/2022] Open
Abstract
The development of safe subunit vaccines requires adjuvants that augment immunogenicity of non-replicating protein-based antigens. Current vaccines against infectious diseases preferentially induce protective antibodies driven by adjuvants such as alum. However, the contribution of antibody to host defense is limited for certain classes of infectious diseases such as fungi, whereas animal studies and clinical observations implicate cellular immunity as an essential component of the resolution of fungal pathogens. Here, we decipher the structural bases of a newly identified glycoprotein ligand of Dectin-2 with potent adjuvancy, Blastomyces endoglucanase-2 (Bl-Eng2). We also pinpoint the developmental steps of antigen-specific CD4+ and CD8+ T responses augmented by Bl-Eng2 including expansion, differentiation and tissue residency. Dectin-2 ligation led to successful systemic and mucosal vaccination against invasive fungal infection and Influenza A infection, respectively. O-linked glycans on Bl-Eng2 applied at the skin and respiratory mucosa greatly augment vaccine subunit- induced protective immunity against lethal influenza and fungal pulmonary challenge. Fungal disease remains a challenging clinical and public health problem in part because there is no commercial vaccine available. The lack of suitable adjuvants is a critical barrier to developing safe and effective vaccines against fungal pathogens. Current adjuvants such as alum preferentially induce antibody responses which may be limited in mediating protection against fungi. Clinical observations and animal studies implicate cellular immunity as the essential component for the resolution of fungal infections. We have recently discovered an adjuvant that augments cell mediated immune responses and vaccine induced protection against fungi. Here, we identified the structural and mechanistic requirements by which this newly discovered adjuvant induces cell mediated immunity against fungi. As a proof of principle we also demonstrate that the adjuvant drives cellular immune responses against viruses such as influenza. We anticipate that our adjuvant can be used for vaccination with safe subunit vaccines against many microbial pathogens including viruses, intracellular bacteria, fungi and parasites that require cell mediated immune responses.
Collapse
Affiliation(s)
- Lucas dos Santos Dias
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hannah E. Dobson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brock Kingstad Bakke
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gregory C. Kujoth
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Junfeng Huang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elaine M. Kohn
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cleison Ledesma Taira
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Huafeng Wang
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nitin T. Supekar
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, United States of America
| | - J. Scott Fites
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Daisy Gates
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christina L. Gomez
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Charles A. Specht
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, United States of America
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Marulasiddappa Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Bruce S. Klein
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Deparment of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Marcel Wüthrich
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
7
|
Advances in Fungal Peptide Vaccines. J Fungi (Basel) 2020; 6:jof6030119. [PMID: 32722452 PMCID: PMC7558412 DOI: 10.3390/jof6030119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/09/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the greatest public health achievements in the past century, protecting and improving the quality of life of the population worldwide. However, a safe and effective vaccine for therapeutic or prophylactic treatment of fungal infections is not yet available. The lack of a vaccine for fungi is a problem of increasing importance as the incidence of diverse species, including Paracoccidioides, Aspergillus, Candida, Sporothrix, and Coccidioides, has increased in recent decades and new drug-resistant pathogenic fungi are emerging. In fact, our antifungal armamentarium too frequently fails to effectively control or cure mycoses, leading to high rates of mortality and morbidity. With this in mind, many groups are working towards identifying effective and safe vaccines for fungal pathogens, with a particular focus of generating vaccines that will work in individuals with compromised immunity who bear the major burden of infections from these microbes. In this review, we detail advances in the development of vaccines for pathogenic fungi, and highlight new methodologies using immunoproteomic techniques and bioinformatic tools that have led to new vaccine formulations, like peptide-based vaccines.
Collapse
|
8
|
Youssef EG, Zhang L, Alkhazraji S, Gebremariam T, Singh S, Yount NY, Yeaman MR, Uppuluri P, Ibrahim AS. Monoclonal IgM Antibodies Targeting Candida albicans Hyr1 Provide Cross-Kingdom Protection Against Gram-Negative Bacteria. Front Immunol 2020; 11:76. [PMID: 32153560 PMCID: PMC7045048 DOI: 10.3389/fimmu.2020.00076] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/13/2020] [Indexed: 02/03/2023] Open
Abstract
Recent years have seen an unprecedented rise in the incidence of multidrug-resistant (MDR) Gram-negative bacteria (GNBs) such as Acinetobacter and Klebsiella species. In view of the shortage of novel drugs in the pipeline, alternative strategies to prevent, and treat infections by GNBs are urgently needed. Previously, we have reported that the Candida albicans hypha-regulated protein Hyr1 shares striking three-dimensional structural homology with cell surface proteins of Acinetobacter baumannii. Moreover, active vaccination with rHyr1p-N or passive immunization with anti-Hyr1p polyclonal antibody protects mice from Acinetobacter infection. In the present study, we use molecular modeling to guide design of monoclonal antibodies (mAbs) generated against Hyr1p and show them to bind to priority surface antigens of Acinetobacter and Klebsiella pneumoniae. The anti-Hyr1 mAbs block damage to primary endothelial cells induced by the bacteria and protect mice from lethal pulmonary infections mediated by A. baumannii or K. pneumoniae. Our current studies emphasize the potential of harnessing Hyr1p mAbs as a cross-kingdom immunotherapeutic strategy against MDR GNBs.
Collapse
Affiliation(s)
- Eman G. Youssef
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Lina Zhang
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Sondus Alkhazraji
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Teclegiorgis Gebremariam
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Shakti Singh
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Nannette Y. Yount
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Michael R. Yeaman
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Priya Uppuluri
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Ashraf S. Ibrahim
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
9
|
NDV-3A vaccination prevents C. albicans colonization of jugular vein catheters in mice. Sci Rep 2019; 9:6194. [PMID: 30996274 PMCID: PMC6470131 DOI: 10.1038/s41598-019-42517-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
NDV-3A, a novel fungal vaccine undergoing clinical trials, contains a recombinant version of the Candida albicans rAls3 N-terminus protein (rAls3p-N) in aluminum hydroxide. In a Phase 1b/2a clinical trial, NDV-3A protected women from recurrent vulvovaginal candidiasis. Here, we reveal that active immunization in mice with NDV-3A induces high titers of anti-rAls3p-N antibodies that interfere with C. albicans ability to adhere to and invade endothelial cells, and form biofilm in vitro. Anti-rAls3p-N antibodies also significantly inhibit yeast dispersal from the hyphal layers of biofilms. Compared to placebo, NDV-3A vaccination inhibited C. albicans dissemination to kidneys and prevented colonization of central venous catheters in mice. Overall, these preclinical studies suggest that NDV-3A may serve as an immunotherapeutic strategy for prevention of infections on indwelling medical devices.
Collapse
|
10
|
Nami S, Mohammadi R, Vakili M, Khezripour K, Mirzaei H, Morovati H. Fungal vaccines, mechanism of actions and immunology: A comprehensive review. Biomed Pharmacother 2018; 109:333-344. [PMID: 30399567 DOI: 10.1016/j.biopha.2018.10.075] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/02/2018] [Accepted: 10/14/2018] [Indexed: 11/28/2022] Open
Abstract
Fungal infections include a wide range of opportunistic and invasive diseases. Two of four major fatal diseases in patients with human immunodeficiency virus (HIV) infection are related to the fungal infections, cryptococcosis, and pneumocystosis. Disseminated candidiasis and different clinical forms of aspergillosis annually impose expensive medical costs to governments and hospitalized patients and ultimately lead to high mortality rates. Therefore, urgent implementations are necessary to prevent the expansion of these diseases. Designing an effective vaccine is one of the most important approaches in this field. So far, numerous efforts have been carried out in developing an effective vaccine against fungal infections. Some of these challenges engaged in different stages of clinical trials but none of them could be approved by the United States Food and Drug Administration (FDA). Here, in addition to have a comprehensive overview on the data from studied vaccine programs, we will discuss the immunology response against fungal infections. Moreover, it will be attempted to clarify the underlying immune mechanisms of vaccines targeting different fungal infections that are crucial for designing an effective vaccination strategy.
Collapse
Affiliation(s)
- Sanam Nami
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasoul Mohammadi
- Department of Medical Parasitology and Mycology, School of Medicine/Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Vakili
- Department of Medical Mycology and Parasitology/Invasive Fungi Research Center (IFRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kimia Khezripour
- Department of Pharmacotherapy, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Morovati
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Zhang J, Cui N, Wang H, Han W, Li Y, Xiao M, Liu D. Invasive Fungal Disease in Critically Ill Patients at High Risk: Usefulness of Lymphocyte Subtyping. J Intensive Care Med 2018; 35:909-918. [PMID: 30231674 DOI: 10.1177/0885066618800690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES This study aimed to investigate the distinguishing ability of lymphocyte subtyping for diagnosis and prognosis of invasive fungal disease (IFD). METHODS We assessed lymphocyte subtyping and evaluated the quantitative changes in key immunological parameters at intensive care unit (ICU) admission in critically ill patients at high risk and their potential influence on diagnosis and outcome of IFD. The primary outcome was 28-day mortality. RESULTS Among the 124 critically ill patients with mean Candida score 3.89 (0.76), 19 (15.3%) were in the IFD group. CD28+CD8+ T-cell counts (area under the curve [AUC] 0.899, 95% confidence interval [CI], 0.834-0.964, P < .001) had better distinguishing ability than other immune parameters for IFD diagnosis. The cutoff value of CD28+CD8+ T-cell counts at ICU admission for IFD diagnosis was 59.5 cells/mm3, with 83.3% sensitivity and 86.4% specificity. Multivariate logistic regression analysis identified CD28+CD8+ T-cell count <59.5 cells/mm3 (odds ratio 59.7, 95% CI, 7.33-486.9, P < .001) as an independent predictor for IFD diagnosis. CD28+CD8+ T-cell counts could also predict 28-day mortality (AUC 0.656, 95% CI, 0.525-0.788, P = .045). Kaplan-Meier survival analysis provided evidence that natural killer cell count <76.0 cells/mm3 (log-rank test; P = .001), CD8+ T-cell count <321.5 cells/mm3 (log-rank test; P = .04), and CD28+CD8+ T-cell count <129.0 cells/mm3 (log-rank test; P = .02) at ICU admission were associated with lower survival probabilities. CONCLUSION CD28+CD8+ T-cell counts play an important role in early diagnosis of IFD. Low counts are associated with early mortality in critically ill patients at high risk of IFD. Our findings add evidence to the utility of lymphocyte subtyping in a diagnostic algorithm to better define IFD in critically ill patients at high risk.
Collapse
Affiliation(s)
- Jiahui Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wen Han
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanfei Li
- Department of Critical Care Medicine, Changsha Central Hospital, Changsha, China
| | - Meng Xiao
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Dawei Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Uppuluri P, Singh S, Alqarihi A, Schmidt CS, Hennessey JP, Yeaman MR, Filler SG, Edwards JE, Ibrahim AS. Human Anti-Als3p Antibodies Are Surrogate Markers of NDV-3A Vaccine Efficacy Against Recurrent Vulvovaginal Candidiasis. Front Immunol 2018; 9:1349. [PMID: 29963049 PMCID: PMC6013566 DOI: 10.3389/fimmu.2018.01349] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022] Open
Abstract
A Phase 1b/2a clinical trial of NDV-3A vaccine containing a Candida albicans recombinant Als3 protein formulated with alum protected women <40 years old from recurrent vulvovaginal candidiasis (RVVC). We investigated the potential use of anti-Als3p sera as surrogate marker of NDV-3A efficacy. Pre- and post-vaccination sera from subjects who experienced recurrence of vulvovaginal candidiasis (R) vs. those who were recurrence-free [non-recurrent (NR)] were evaluated. Anti-Als3p antisera obtained were evaluated for (1) titer and subclass profile and (2) their ability to influence C. albicans virulence traits including hyphal elongation, adherence to plastic, invasion of vaginal epithelial cells, biofilm formation on plastic and catheter material, and susceptibility to neutrophil killing in vitro. Serum IgG titers in NR patients were consistently higher than in R patients, particularly for anti-Als3 subclass IgG2. Sera from vaccinated NR patients reduced hyphal elongation, adhesion to plastic, invasion of vaginal epithelial cells, and biofilm formation significantly more than pre-immune sera, or sera from R- or placebo-group subjects. Pre-adsorption of sera with C. albicans germ tubes eliminated these effects, while heat inactivation did not. Finally, sera from NR subjects enhanced neutrophil-mediated killing of C. albicans relative to pre-immune sera or sera from R patients. Our results suggest that higher Als3p antibody titers are associated with protection from RVVC, attenuate C. albicans virulence, and augment immune clearance of the fungus in vitro. Thus, Als3p serum IgG antibodies are likely useful markers of efficacy in RVVC patients vaccinated with NDV-3A.
Collapse
Affiliation(s)
- Priya Uppuluri
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Shakti Singh
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Abdullah Alqarihi
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States
| | | | | | - Michael R Yeaman
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Scott G Filler
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - John E Edwards
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Ashraf S Ibrahim
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Uppuluri P, Lin L, Alqarihi A, Luo G, Youssef EG, Alkhazraji S, Yount NY, Ibrahim BA, Bolaris MA, Edwards JE, Swidergall M, Filler SG, Yeaman MR, Ibrahim AS. The Hyr1 protein from the fungus Candida albicans is a cross kingdom immunotherapeutic target for Acinetobacter bacterial infection. PLoS Pathog 2018; 14:e1007056. [PMID: 29746596 PMCID: PMC5963808 DOI: 10.1371/journal.ppat.1007056] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/22/2018] [Accepted: 04/26/2018] [Indexed: 11/21/2022] Open
Abstract
Different pathogens share similar medical settings and rely on similar virulence strategies to cause infections. We have previously applied 3-D computational modeling and bioinformatics to discover novel antigens that target more than one human pathogen. Active and passive immunization with the recombinant N-terminus of Candida albicans Hyr1 (rHyr1p-N) protect mice against lethal candidemia. Here we determine that Hyr1p shares homology with cell surface proteins of the multidrug resistant Gram negative bacterium, Acinetobacter baumannii including hemagglutinin (FhaB) and outer membrane protein A (OmpA). The A. baumannii OmpA binds to C. albicans Hyr1p, leading to a mixed species biofilm. Deletion of HYR1, or blocking of Hyr1p using polyclonal antibodies, significantly reduce A. baumannii binding to C. albicans hyphae. Furthermore, active vaccination with rHyr1p-N or passive immunization with polyclonal antibodies raised against specific peptide motifs of rHyr1p-N markedly improve survival of diabetic or neutropenic mice infected with A. baumannii bacteremia or pneumonia. Antibody raised against one particular peptide of the rHyr1p-N sequence (peptide 5) confers majority of the protection through blocking A. baumannii invasion of host cells and inducing death of the bacterium by a putative iron starvation mechanism. Anti-Hyr1 peptide 5 antibodies also mitigate A. baumannii /C. albicans mixed biofilm formation in vitro. Consistent with our bioinformatic analysis and structural modeling of Hyr1p, anti-Hyr1p peptide 5 antibodies bound to A. baumannii FhaB, OmpA, and an outer membrane siderophore binding protein. Our studies highlight the concept of cross-kingdom vaccine protection against high priority human pathogens such as A. baumannii and C. albicans that share similar ecological niches in immunocompromised patients.
Collapse
Affiliation(s)
- Priya Uppuluri
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Lin Lin
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Abdullah Alqarihi
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Guanpingsheng Luo
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Eman G. Youssef
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Sondus Alkhazraji
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Nannette Y. Yount
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Belal A. Ibrahim
- Portola High School, Irvine, California, United States of America
| | - Michael Anthony Bolaris
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - John E. Edwards
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Marc Swidergall
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Scott G. Filler
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Michael R. Yeaman
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ashraf S. Ibrahim
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
14
|
Kumaresan PR, da Silva TA, Kontoyiannis DP. Methods of Controlling Invasive Fungal Infections Using CD8 + T Cells. Front Immunol 2018; 8:1939. [PMID: 29358941 PMCID: PMC5766637 DOI: 10.3389/fimmu.2017.01939] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Invasive fungal infections (IFIs) cause high rates of morbidity and mortality in immunocompromised patients. Pattern-recognition receptors present on the surfaces of innate immune cells recognize fungal pathogens and activate the first line of defense against fungal infection. The second line of defense is the adaptive immune system which involves mainly CD4+ T cells, while CD8+ T cells also play a role. CD8+ T cell-based vaccines designed to prevent IFIs are currently being investigated in clinical trials, their use could play an especially important role in acquired immune deficiency syndrome patients. So far, none of the vaccines used to treat IFI have been approved by the FDA. Here, we review current and future antifungal immunotherapy strategies involving CD8+ T cells. We highlight recent advances in the use of T cells engineered using a Sleeping Beauty vector to treat IFIs. Recent clinical trials using chimeric antigen receptor (CAR) T-cell therapy to treat patients with leukemia have shown very promising results. We hypothesized that CAR T cells could also be used to control IFI. Therefore, we designed a CAR that targets β-glucan, a sugar molecule found in most of the fungal cell walls, using the extracellular domain of Dectin-1, which binds to β-glucan. Mice treated with D-CAR+ T cells displayed reductions in hyphal growth of Aspergillus compared to the untreated group. Patients suffering from IFIs due to primary immunodeficiency, secondary immunodeficiency (e.g., HIV), or hematopoietic transplant patients may benefit from bioengineered CAR T cell therapy.
Collapse
Affiliation(s)
- Pappanaicken R. Kumaresan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thiago Aparecido da Silva
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
15
|
Abstract
Glycosylation is an important post-translational modification that is required for structural and stability purposes and functional roles such as signalling, attachment and shielding. Many human pathogens such as bacteria display an array of carbohydrates on their surface that are non-self to the host; others such as viruses highjack the host-cell machinery and present self-carbohydrates sometimes arranged in a non-self more immunogenic manner. In combination with carrier proteins, these glycan structures can be highly immunogenic. During natural infection, glycan-binding antibodies are often elicited that correlate with long-lasting protection. A great amount of research has been invested in carbohydrate vaccine design to elicit such an immune response, which has led to the development of vaccines against the bacterial pathogens Haemophilus influenzae type b, Streptococcus pneumonia and Neisseria meningitidis. Other vaccines, e.g. against HIV-1, are still in development, but promising progress has been made with the isolation of broadly neutralizing glycan-binding antibodies and the engineering of stable trimeric envelope glycoproteins. Carbohydrate vaccines against other pathogens such as viruses (Dengue, Hepatitis C), parasites (Plasmodium) and fungi (Candida) are at different stages of development. This chapter will discuss the challenges in inducing cross-reactive carbohydrate-targeting antibodies and progress towards carbohydrate vaccines.
Collapse
|
16
|
Gerwien F, Skrahina V, Kasper L, Hube B, Brunke S. Metals in fungal virulence. FEMS Microbiol Rev 2018; 42:4562650. [PMID: 29069482 PMCID: PMC5812535 DOI: 10.1093/femsre/fux050] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/19/2017] [Indexed: 12/25/2022] Open
Abstract
Metals are essential for life, and they play a central role in the struggle between infecting microbes and their hosts. In fact, an important aspect of microbial pathogenesis is the 'nutritional immunity', in which metals are actively restricted (or, in an extended definition of the term, locally enriched) by the host to hinder microbial growth and virulence. Consequently, fungi have evolved often complex regulatory networks, uptake and detoxification systems for essential metals such as iron, zinc, copper, nickel and manganese. These systems often differ fundamentally from their bacterial counterparts, but even within the fungal pathogens we can find common and unique solutions to maintain metal homeostasis. Thus, we here compare the common and species-specific mechanisms used for different metals among different fungal species-focusing on important human pathogens such as Candida albicans, Aspergillus fumigatus or Cryptococcus neoformans, but also looking at model fungi such as Saccharomyces cerevisiae or A. nidulans as well-studied examples for the underlying principles. These direct comparisons of our current knowledge reveal that we have a good understanding how model fungal pathogens take up iron or zinc, but that much is still to learn about other metals and specific adaptations of individual species-not the least to exploit this knowledge for new antifungal strategies.
Collapse
Affiliation(s)
- Franziska Gerwien
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Volha Skrahina
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Lydia Kasper
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| | - Sascha Brunke
- Department Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology– Hans Knoell Institute, 07745 Jena, Germany
| |
Collapse
|
17
|
Maldonado S, Fitzgerald-Bocarsly P. Antifungal Activity of Plasmacytoid Dendritic Cells and the Impact of Chronic HIV Infection. Front Immunol 2017; 8:1705. [PMID: 29255464 PMCID: PMC5723005 DOI: 10.3389/fimmu.2017.01705] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/20/2017] [Indexed: 01/10/2023] Open
Abstract
Due to the effectiveness of combined antiretroviral therapy, people living with HIV can control viral replication and live longer lifespans than ever. However, HIV-positive individuals still face challenges to their health and well-being, including dysregulation of the immune system resulting from years of chronic immune activation, as well as opportunistic infections from pathogenic fungi. This review focuses on one of the key players in HIV immunology, the plasmacytoid dendritic cell (pDC), which links the innate and adaptive immune response and is notable for being the body’s most potent producer of type-I interferons (IFNs). During chronic HIV infection, the pDC compartment is greatly dysregulated, experiencing a substantial depletion in number and compromise in function. This immune dysregulation may leave patients further susceptible to opportunistic infections. This is especially important when considering a new role for pDCs currently emerging in the literature: in addition to their role in antiviral immunity, recent studies suggest that pDCs also play an important role in antifungal immunity. Supporting this new role, pDCs express C-type lectin receptors including dectin-1, dectin-2, dectin-3, and mannose receptor, and toll-like receptors-4 and -9 that are involved in recognition, signaling, and response to a wide variety of fungal pathogens, including Aspergillus fumigatus, Cryptococcus neoformans, Candida albicans, and Pneumocystis jirovecii. Accordingly, pDCs have been demonstrated to recognize and respond to certain pathogenic fungi, measured via activation, cytokine production, and fungistatic activity in vitro, while in vivo mouse models indicated a strikingly vital role for pDCs in survival against pulmonary Aspergillus challenge. Here, we discuss the role of the pDC compartment and the dysregulation it undergoes during chronic HIV infection, as well as what is known so far about the role and mechanisms of pDC antifungal activity.
Collapse
Affiliation(s)
- Samuel Maldonado
- Rutgers School of Graduate Studies, Newark, NJ, United States.,Department of Pathology and Laboratory Medicine, New Jersey Medical School, Newark, NJ, United States
| | - Patricia Fitzgerald-Bocarsly
- Rutgers School of Graduate Studies, Newark, NJ, United States.,Department of Pathology and Laboratory Medicine, New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
18
|
Abstract
Safe and efficacious vaccines are arguably the most successful medical interventions of all time. Yet the ongoing discovery of new pathogens, along with emergence of antibiotic-resistant pathogens and a burgeoning population at risk of such infections, imposes unprecedented public health challenges. To meet these challenges, innovative strategies to discover and develop new or improved anti-infective vaccines are necessary. These approaches must intersect the most meaningful insights into protective immunity and advanced technologies with capabilities to deliver immunogens for optimal immune protection. This goal is considered through several recent advances in host-pathogen relationships, conceptual strides in vaccinology, and emerging technologies. Given a clear and growing risk of pandemic disease should the threat of infection go unmet, developing vaccines that optimize protective immunity against high-priority and antibiotic-resistant pathogens represents an urgent and unifying imperative.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90024.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509; .,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509.,Los Angeles Biomedical Research Institute, Torrance, California 90502
| | | |
Collapse
|
19
|
Sui X, Yan L, Jiang YY. The vaccines and antibodies associated with Als3p for treatment of Candida albicans infections. Vaccine 2017; 35:5786-5793. [DOI: 10.1016/j.vaccine.2017.08.082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/14/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
|
20
|
Wang H, Lee TJ, Fites SJ, Merkhofer R, Zarnowski R, Brandhorst T, Galles K, Klein B, Wüthrich M. Ligation of Dectin-2 with a novel microbial ligand promotes adjuvant activity for vaccination. PLoS Pathog 2017; 13:e1006568. [PMID: 28793349 PMCID: PMC5565193 DOI: 10.1371/journal.ppat.1006568] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/21/2017] [Accepted: 08/04/2017] [Indexed: 11/18/2022] Open
Abstract
The development of vaccines against fungi and other intracellular microbes is impeded in part by a lack of suitable adjuvants. While most current vaccines against infectious diseases preferentially induce production of antibodies, cellular immunity is essential for the resolution of fungal infections. Microbes such as fungi and Mycobacterium tuberculosis require Th17 and Th1 cells for resistance, and engage the C-type lectin receptors including Dectin-2. Herein, we discovered a novel Dectin-2 ligand, the glycoprotein Blastomyces Eng2 (Bl-Eng2). Bl-Eng2 triggers robust signaling in Dectin-2 reporter cells and induces IL-6 in human PBMC and BMDC from wild type but not Dectin-2-/- and Card9-/- mice. The addition of Bl-Eng2 to a pan-fungal subunit vaccine primed large numbers of Ag-specific Th17 and Th1 cells, augmented activation and killing of fungi by myeloid effector cells, and protected mice from lethal fungal challenge, revealing Bl-Eng2’s potency as a vaccine adjuvant. Thus, ligation of Dectin-2 by Bl-Eng-2 could be harnessed as a novel adjuvant strategy to protect against infectious diseases requiring cellular immunity. Despite several million new systemic fungal infections annually worldwide, there are no commercial vaccines available. The lack of appropriate adjuvants is one major impediment to developing safe and effective vaccines against infections with fungal pathogens. Current vaccines against infectious diseases preferentially induce protective antibodies, driven by adjuvants such as alum. While clonally-derived and adoptively transferred monoclonal antibodies may confer protection against fungi, the contribution of antibody to host defense is likely to be limited. Animal studies and clinical observations implicate cellular immunity as an essential component of the resolution of fungal infections. We found a promising adjuvant that augments cell mediated immune responses and vaccine-induced protection against fungal infection. We anticipate that our discovery will be a useful adjuvant for vaccination with non-replicating and safe subunit vaccines against many microbial pathogens that require protective cell mediated immune responses.
Collapse
Affiliation(s)
- Huafeng Wang
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Taek-Jin Lee
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Scott J. Fites
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Richard Merkhofer
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Robert Zarnowski
- Department of Medicine (Infectious Disease Division), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Tristan Brandhorst
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Kevin Galles
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Bruce Klein
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
| | - Marcel Wüthrich
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, United States of America
- * E-mail:
| |
Collapse
|
21
|
Carvalho A, Duarte-Oliveira C, Gonçalves SM, Campos A, Lacerda JF, Cunha C. Fungal Vaccines and Immunotherapeutics: Current Concepts and Future Challenges. CURRENT FUNGAL INFECTION REPORTS 2017. [DOI: 10.1007/s12281-017-0272-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
22
|
Ponzio TA, Sanders JW. The salivary gland as a target for enhancing immunization response. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2017; 3:4. [PMID: 28883974 PMCID: PMC5531011 DOI: 10.1186/s40794-017-0047-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/02/2022]
Abstract
Background An organism’s immune response to a vaccine is dependent on a number of factors, including the site of immunization. While muscle is the most common site for vaccine administration, other sites, including the salivary gland, are poised to confer stronger and broader immunoprotection. Findings Studies exploring the salivary gland as an immunization site have involved protein antigens, as well as live pathogens and DNA vaccines. While intraductal instillation of protein antigens into the salivary gland may result in a relatively transient increase in antibody production, DNA or attenuated pathogen vaccination appear to confer a lasting widespread mucosal immune response that includes robust salivary and enteric IgA, as well as high levels of circulating IgG. Furthermore, vaginal and lung antibodies are also seen. For enteric pathogens, a common class of pathogen encountered by travelers, this type of immune response provides for a level of redundant protection against foreign microbes with mucosal targets. Conclusion The strength of immune response conferred by salivary gland vaccination is generally stronger than that seen in response to the same vaccine at a comparison site. For example, where other routes fail, immunization of the salivary gland has been shown to confer protection in lethal challenge models of infectious pathogens. A host of vaccines currently under development suffer from immunogenicity challenges, adding to the widespread interest and search for novel routes and adjuvants. With its capability to facilitate a strong and broad immune response, the salivary gland warrants consideration as an immunization site, especially for vaccines with immunogenicity challenges, as well as vaccines that would benefit from combined systemic and mucosal immunity.
Collapse
Affiliation(s)
- Todd A Ponzio
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910 USA.,Wake Forest University School of Medicine, Winston-Salem, USA
| | - John W Sanders
- Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA.,Hefner Veterans Affairs Medical Center, Salisbury, NC UK
| |
Collapse
|
23
|
Abstract
Fungal organisms are ubiquitous in the environment. Pathogenic fungi, although relatively few in the whole gamut of microbial pathogens, are able to cause disease with varying degrees of severity in individuals with normal or impaired immunity. The disease state is an outcome of the fungal pathogen's interactions with the host immunity, and therefore, it stands to reason that deep/invasive fungal diseases be amenable to immunotherapy. Therefore, antifungal immunotherapy continues to be attractive as an adjunct to the currently available antifungal chemotherapy options for a number of reasons, including the fact that existing antifungal drugs, albeit largely effective, are not without limitations, and that morbidity and mortality associated with invasive mycoses are still unacceptably high. For several decades, intense basic research efforts have been directed at development of fungal immunotherapies. Nevertheless, this approach suffers from a severe bench-bedside disconnect owing to several reasons: the chemical and biological peculiarities of the fungal antigens, the complexities of host-pathogen interactions, an under-appreciation of the fungal disease landscape, the requirement of considerable financial investment to bring these therapies to clinical use, as well as practical problems associated with immunizations. In this general, non-exhaustive review, we summarize the features of ongoing research efforts directed towards devising safe and effective immunotherapeutic options for mycotic diseases, encompassing work on antifungal vaccines, adoptive cell transfers, cytokines, antimicrobial peptides (AMPs), monoclonal antibodies (mAbs), and other agents.
Collapse
Affiliation(s)
- Kausik Datta
- a Division of Infectious Diseases , Johns Hopkins University School of Medicine , Baltimore , MD , USA , and
| | - Mawieh Hamad
- b Department of Medical Laboratory Sciences and the Sharjah Institute for Medical Research , University of Sharjah , Sharjah , UAE
| |
Collapse
|
24
|
Johnson JS, Reeder DM, Lilley TM, Czirják GÁ, Voigt CC, McMichael JW, Meierhofer MB, Seery CW, Lumadue SS, Altmann AJ, Toro MO, Field KA. Antibodies to Pseudogymnoascus destructans are not sufficient for protection against white-nose syndrome. Ecol Evol 2015; 5:2203-14. [PMID: 26078857 PMCID: PMC4461422 DOI: 10.1002/ece3.1502] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/22/2015] [Indexed: 12/12/2022] Open
Abstract
White-nose syndrome (WNS) is a fungal disease caused by Pseudogymnoascus destructans (Pd) that affects bats during hibernation. Although millions of bats have died from WNS in North America, mass mortality has not been observed among European bats infected by the fungus, leading to the suggestion that bats in Europe are immune. We tested the hypothesis that an antibody-mediated immune response can provide protection against WNS by quantifying antibodies reactive to Pd in blood samples from seven species of free-ranging bats in North America and two free-ranging species in Europe. We also quantified antibodies in blood samples from little brown myotis (Myotis lucifugus) that were part of a captive colony that we injected with live Pd spores mixed with adjuvant, as well as individuals surviving a captive Pd infection trial. Seroprevalence of antibodies against Pd, as well as antibody titers, was greater among little brown myotis than among four other species of cave-hibernating bats in North America, including species with markedly lower WNS mortality rates. Among little brown myotis, the greatest titers occurred in populations occupying regions with longer histories of WNS, where bats lacked secondary symptoms of WNS. We detected antibodies cross-reactive with Pd among little brown myotis naïve to the fungus. We observed high titers among captive little brown myotis injected with Pd. We did not detect antibodies against Pd in Pd-infected European bats during winter, and titers during the active season were lower than among little brown myotis. These results show that antibody-mediated immunity cannot explain survival of European bats infected with Pd and that little brown myotis respond differently to Pd than species with higher WNS survival rates. Although it appears that some species of bats in North America may be developing resistance to WNS, an antibody-mediated immune response does not provide an explanation for these remnant populations.
Collapse
Affiliation(s)
- Joseph S Johnson
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| | - DeeAnn M Reeder
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| | | | - Gábor Á Czirják
- Leibniz Institute for Zoo and Wildlife Research Berlin, Germany
| | | | - James W McMichael
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| | | | | | - Shayne S Lumadue
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| | | | - Michael O Toro
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| | - Kenneth A Field
- Department of Biology, Bucknell University Lewisburg, Pennsylvania, 17837
| |
Collapse
|
25
|
Abstract
Candida albicans is the most common cause of hematogenously disseminated candidiasis, and this disease is particularly prevalent in immunocompromised patients. The mortality of invasive candidiasis remains 40% to 50% even with the proper treatment with current antifungal drugs. Recently, with the better understanding of host-fungus interactions, notable progress has been made in antifungal vaccine research. Most antifungal vaccines exert protection by inducing either (or both) B-cell and T-cell responses. Here we summarize the current available information on C. albicans vaccines, highlight the obstacles that researchers identified, and offer several suggestions.
Collapse
Affiliation(s)
- Xiao-juan Wang
- a Center for New Drug Research; School of Pharmacy ; Second Military Medical University ; 325 Guohe Road; Shanghai , P.R. China
| | | | | | | | | | | |
Collapse
|
26
|
Carneiro C, Correia A, Collins T, Vilanova M, Pais C, Gomes AC, Real Oliveira MEC, Sampaio P. DODAB:monoolein liposomes containing Candida albicans cell wall surface proteins: A novel adjuvant and delivery system. Eur J Pharm Biopharm 2015; 89:190-200. [DOI: 10.1016/j.ejpb.2014.11.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/28/2014] [Accepted: 11/29/2014] [Indexed: 11/26/2022]
|
27
|
Borghi M, Renga G, Puccetti M, Oikonomou V, Palmieri M, Galosi C, Bartoli A, Romani L. Antifungal Th Immunity: Growing up in Family. Front Immunol 2014; 5:506. [PMID: 25360137 PMCID: PMC4197763 DOI: 10.3389/fimmu.2014.00506] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 09/28/2014] [Indexed: 12/25/2022] Open
Abstract
Fungal diseases represent an important paradigm in immunology since they can result from either the lack of recognition or over-activation of the inflammatory response. Current understanding of the pathophysiology underlying fungal infections and diseases highlights the multiple cell populations and cell-signaling pathways involved in these conditions. A systems biology approach that integrates investigations of immunity at the systems-level is required to generate novel insights into this complexity and to decipher the dynamics of the host–fungus interaction. It is becoming clear that a three-way interaction between the host, microbiota, and fungi dictates the types of host–fungus relationship. Tryptophan metabolism helps support this interaction, being exploited by the mammalian host and commensals to increase fitness in response to fungi via resistance and tolerance mechanisms of antifungal immunity. The cellular and molecular mechanisms that provide immune homeostasis with the fungal biota and its possible rupture in fungal infections and diseases will be discussed within the expanding role of antifungal Th cell responses.
Collapse
Affiliation(s)
- Monica Borghi
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Giorgia Renga
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | | | - Vasileios Oikonomou
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Melissa Palmieri
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Claudia Galosi
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Andrea Bartoli
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| | - Luigina Romani
- Pathology Section, Department of Experimental Medicine, University of Perugia , Perugia , Italy
| |
Collapse
|
28
|
Moriyama B, Gordon LA, McCarthy M, Henning SA, Walsh TJ, Penzak SR. Emerging drugs and vaccines for candidemia. Mycoses 2014; 57:718-33. [PMID: 25294098 DOI: 10.1111/myc.12265] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 08/26/2014] [Accepted: 09/01/2014] [Indexed: 11/27/2022]
Abstract
Candidemia and other forms of invasive candidiasis are important causes of morbidity and mortality. The evolving challenge of antimicrobial resistance among fungal pathogens continues to highlight the need for potent, new antifungal agents. MEDLINE, EMBASE, Scopus and Web of Science searches (up to January 2014) of the English-language literature were performed with the keywords 'Candida' or 'Candidemia' or 'Candidiasis' and terms describing investigational drugs with activity against Candida spp. Conference abstracts and the bibliographies of pertinent articles were also reviewed for relevant reports. ClinicalTrials.gov was searched for relevant clinical trials. Currently available antifungal agents for the treatment of candidemia are summarised. Investigational antifungal agents with potential activity against Candida bloodstream infections and other forms of invasive candidiasis and vaccines for prevention of Candida infections are also reviewed as are selected antifungal agents no longer in development. Antifungal agents currently in clinical trials include isavuconazole, albaconazole, SCY-078, VT-1161 and T-2307. Further data are needed to determine the role of these compounds in the treatment of candidemia and other forms of invasive candidiasis. The progressive reduction in antimicrobial drug development may result in a decline in antifungal drug discovery. Still, there remains a critical need for new antifungal agents to treat and prevent invasive candidiasis and other life-threatening mycoses.
Collapse
Affiliation(s)
- Brad Moriyama
- Pharmacy Department, NIH Clinical Center, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
29
|
Luo S, Skerka C, Kurzai O, Zipfel PF. Complement and innate immune evasion strategies of the human pathogenic fungus Candida albicans. Mol Immunol 2013; 56:161-9. [DOI: 10.1016/j.molimm.2013.05.218] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 05/10/2013] [Indexed: 01/09/2023]
|
30
|
Development of vaccines for Candida albicans: fighting a skilled transformer. Nat Rev Microbiol 2013; 11:884-91. [DOI: 10.1038/nrmicro3156] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
31
|
Ibrahim AS, Luo G, Gebremariam T, Lee H, Schmidt CS, Hennessey JP, French SW, Yeaman MR, Filler SG, Edwards JE. NDV-3 protects mice from vulvovaginal candidiasis through T- and B-cell immune response. Vaccine 2013; 31:5549-56. [PMID: 24063977 DOI: 10.1016/j.vaccine.2013.09.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/28/2013] [Accepted: 09/10/2013] [Indexed: 11/19/2022]
Abstract
We have previously reported that vaccination with rAls3p-N protein of Candida albicans, formulated with alum adjuvant (also designated as NDV-3) protects immunocompetent mice from, lethal disseminated candidiasis and mucosal oropharyngeal candidiasis. NDV-3 vaccine was recently, tested in a Phase 1 clinical trial and found to be safe, well-tolerated, and induced robust humoral and, cellular immune responses with increased interferon (IFN)-gamma and interleukin (IL)-17 secretion. In preparation for a Phase 2 clinical trial against vulvovaginal candidiasis (VVC), we evaluated NDV-3, efficacy in a murine VVC model. Here, NDV-3 induced a strong immune response characterized by high, anti-rAls3p-N serum IgG and vaginal IgA titers. Furthermore, moderate doses of the vaccine (a range of 1-30μg given subcutaneously [SQ] or 0.3-10μg given intramuscularly [IM]) elicited a 10-1000 fold, decrease in vaginal fungal burden vs. control (mice injected with alum adjuvant alone) in both inbred, and outbred mice infected with different clinical C. albicans isolates. Additionally, NDV-3 required both, T and B lymphocytes for efficacy in reducing C. albicans tissue burden, which is followed by a reduction, in neutrophil influx to the affected site. Finally, anti-rAls3p-N antibodies enhanced the ex vivo killing, of C. albicans by neutrophils primed with IFN-gamma. These data indicate that NDV-3 protects mice, from VVC by a mechanism that involves the concerted priming of both humoral and adaptive immune, responses.
Collapse
Affiliation(s)
- Ashraf S Ibrahim
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California Los Angeles (UCLA) Medical Center and the St. John's Cardiovascular Research Center, Torrance, CA, United States; David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gamaletsou MN, Walsh TJ, Zaoutis T, Pagoni M, Kotsopoulou M, Voulgarelis M, Panayiotidis P, Vassilakopoulos T, Angelopoulou MK, Marangos M, Spyridonidis A, Kofteridis D, Pouli A, Sotiropoulos D, Matsouka P, Argyropoulou A, Perloretzou S, Leckerman K, Manaka A, Oikonomopoulos P, Daikos G, Petrikkos G, Sipsas NV. A prospective, cohort, multicentre study of candidaemia in hospitalized adult patients with haematological malignancies. Clin Microbiol Infect 2013; 20:O50-7. [PMID: 23889746 DOI: 10.1111/1469-0691.12312] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 03/22/2013] [Accepted: 06/19/2013] [Indexed: 01/06/2023]
Abstract
Invasive candidiasis is a life-threatening infection in patients with haematological malignancies. The objective of our study was to determine the incidence, microbiological characteristics and clinical outcome of candidaemia among hospitalized adult patients with haematological malignancies. This is a population-based, prospective, multicentre study of patients ≥ 18 years admitted to haematology and/or haematopoietic stem cell transplantation units of nine tertiary care Greek hospitals from January 2009 through to February 2012. Within this cohort, we conducted a nested case-control study to determine the risk factors for candidaemia. Stepwise logistic regression was used to identify independent predictors of 28-day mortality. Candidaemia was detected in 40 of 27,864 patients with haematological malignancies vs. 967 of 1,158,018 non-haematology patients for an incidence of 1.4 cases/1000 admissions vs. 0.83/1000 respectively (p <0.001). Candidaemia was caused predominantly (35/40, 87.5%) by non-Candida albicans species, particularly Candida parapsilosis (20/40, 50%). In vitro resistance to at least one antifungal agent was observed in 27% of Candida isolates. Twenty-one patients (53%) developed breakthrough candidaemia while receiving antifungal agents. Central venous catheters, hypogammaglobulinaemia and a high APACHE II score were independent risk factors for the development of candidaemia. Crude mortality at day 28 was greater in those with candidaemia than in control cases (18/40 (45%) vs. 9/80 (11%); p <0.0001). In conclusion, despite antifungal prophylaxis, candidaemia is a relatively frequent infection associated with high mortality caused by non-C. albicans spp., especially C. parapsilosis. Central venous catheters and hypogammaglobulinaemia are independent risk factors for candidaemia that provide potential targets for improving the outcome.
Collapse
Affiliation(s)
- M N Gamaletsou
- Laikon General Hospital and Medical School, National and Kapodistrian University of Athens, Athens, Greece; Weill Cornell Medical Center of Cornell University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kim OY, Hong BS, Park KS, Yoon YJ, Choi SJ, Lee WH, Roh TY, Lötvall J, Kim YK, Gho YS. Immunization with Escherichia coli outer membrane vesicles protects bacteria-induced lethality via Th1 and Th17 cell responses. THE JOURNAL OF IMMUNOLOGY 2013; 190:4092-102. [PMID: 23514742 DOI: 10.4049/jimmunol.1200742] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Outer membrane vesicles (OMVs), secreted from Gram-negative bacteria, are spherical nanometer-sized proteolipids enriched with outer membrane proteins. OMVs, also known as extracellular vesicles, have gained interests for use as nonliving complex vaccines and have been examined for immune-stimulating effects. However, the detailed mechanism on how OMVs elicit the vaccination effect has not been studied extensively. In this study, we investigated the immunological mechanism governing the protective immune response of OMV vaccines. Immunization with Escherichia coli-derived OMVs prevented bacteria-induced lethality and OMV-induced systemic inflammatory response syndrome. As verified by adoptive transfer and gene-knockout studies, the protective effect of OMV immunization was found to be primarily by the stimulation of T cell immunity rather than B cell immunity, especially by the OMV-Ag-specific production of IFN-γ and IL-17 from T cells. By testing the bacteria-killing ability of macrophages, we also demonstrated that IFN-γ and IL-17 production is the main factor promoting bacterial clearances. Our findings reveal that E. coli-derived OMV immunization effectively protects bacteria-induced lethality and OMV-induced systemic inflammatory response syndrome primarily via Th1 and Th17 cell responses. This study therefore provides a new perspective on the immunological detail regarding OMV vaccination.
Collapse
Affiliation(s)
- Oh Youn Kim
- Division of Molecular and Life Sciences, Department of Life Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duluc D, Gannevat J, Joo H, Ni L, Upchurch K, Boreham M, Carley M, Stecher J, Zurawski G, Oh S. Dendritic cells and vaccine design for sexually-transmitted diseases. Microb Pathog 2012. [PMID: 23201532 DOI: 10.1016/j.micpath.2012.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are major antigen presenting cells (APCs) that can initiate and control host immune responses toward either immunity or tolerance. These features of DCs, as immune orchestrators, are well characterized by their tissue localizations as well as by their subset-dependent functional specialties and plasticity. Thus, the level of protective immunity to invading microbial pathogens can be dependent on the subsets of DCs taking up microbial antigens and their functional plasticity in response to microbial products, host cellular components and the cytokine milieu in the microenvironment. Vaccines are the most efficient and cost-effective preventive medicine against infectious diseases. However, major challenges still remain for the diseases caused by sexually-transmitted pathogens, including HIV, HPV, HSV and Chlamydia. We surmise that the establishment of protective immunity in the female genital mucosa, the major entry and transfer site of these pathogens, will bring significant benefit for the protection against sexually-transmitted diseases. Recent progresses made in DC biology suggest that vaccines designed to target proper DC subsets may permit us to establish protective immunity in the female genital mucosa against sexually-transmitted pathogens.
Collapse
Affiliation(s)
- Dorothee Duluc
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
From memory to antifungal vaccine design. Trends Immunol 2012; 33:467-74. [DOI: 10.1016/j.it.2012.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/18/2012] [Accepted: 04/23/2012] [Indexed: 01/21/2023]
|
36
|
Carvalho A, Cunha C, Iannitti RG, Casagrande A, Bistoni F, Aversa F, Romani L. Host defense pathways against fungi: the basis for vaccines and immunotherapy. Front Microbiol 2012; 3:176. [PMID: 22590466 PMCID: PMC3349272 DOI: 10.3389/fmicb.2012.00176] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/21/2012] [Indexed: 12/16/2022] Open
Abstract
Fungal vaccines have long been a goal in the fields of immunology and microbiology to counter the high mortality and morbidity rates owing to fungal diseases, particularly in immunocompromised patients. However, the design of effective vaccination formulations for durable protection to the different fungi has lagged behind due to the important differences among fungi and their biology and our limited understanding of the complex host–pathogen interactions and immune responses. Overcoming these challenges is expected to contribute to improved vaccination strategies aimed at personalized efficacy across distinct target patient populations. This likely requires the integration of multifaceted approaches encompassing advanced immunology, systems biology, immunogenetics, and bioinformatics in the fields of fungal and host biology and their reciprocal interactions.
Collapse
Affiliation(s)
- Agostinho Carvalho
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
Cassone A, Casadevall A. Recent progress in vaccines against fungal diseases. Curr Opin Microbiol 2012; 15:427-33. [PMID: 22564747 DOI: 10.1016/j.mib.2012.04.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/03/2012] [Accepted: 04/10/2012] [Indexed: 12/20/2022]
Abstract
Diseases caused by fungi are increasingly impacting the health of the human population and now account for a large fraction of infectious disease complications in individuals with impaired immunity or breached tissue defenses. Antifungal therapy is often of limited effectiveness in these patients, resulting into treatment failures, chronic infections and unacceptable rates of mortality, morbidity and their associated costs. Consequently there is a real medical need for new treatments and preventive measures to combat fungal diseases and, toward this goal, safe and efficacious vaccines would constitute major progress. After decades of complacency and neglect of this critically important field of research, remarkable progress has been made in recent years. A number of highly immunogenic and protective vaccine formulations in preclinical setting have been developed, and at least two have undergone Phase 1 clinical trials as preventive and/or therapeutic tools against candidiasis.
Collapse
Affiliation(s)
- Antonio Cassone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
38
|
Bär E, Gladiator A, Bastidas S, Roschitzki B, Acha-Orbea H, Oxenius A, LeibundGut-Landmann S. A Novel Th Cell Epitope of Candida albicans Mediates Protection from Fungal Infection. THE JOURNAL OF IMMUNOLOGY 2012; 188:5636-43. [DOI: 10.4049/jimmunol.1200594] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
39
|
Luo G, Ibrahim AS, French SW, Edwards JE, Fu Y. Active and passive immunization with rHyr1p-N protects mice against hematogenously disseminated candidiasis. PLoS One 2011; 6:e25909. [PMID: 22028796 PMCID: PMC3189951 DOI: 10.1371/journal.pone.0025909] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/13/2011] [Indexed: 11/18/2022] Open
Abstract
We previously reported that Candida albicans cell surface protein Hyr1 encodes a phagocyte killing resistance factor and active vaccination with a recombinant N-terminus of Hyr1 protein (rHyr1p-N), significantly protects immunocompetent mice from disseminated candidiasis. Here we report the marked efficacy of rHyr1p-N vaccine on improving the survival and reducing the fungal burden of disseminated candidiasis in both immunocompetent and immunocompromised mice using the FDA-approved adjuvant, alum. Importantly, we also show that pooled rabbit anti-Hyr1p polyclonal antibodies raised against 8 different peptide regions of rHyr1p-N protected mice in a hematogenously disseminated candidiasis model, raising the possibility of developing a successful passive immunotherapy strategy to treat this disease. Our data suggest that the rabbit anti-Hyr1p antibodies directly neutralized the Hyr1p virulence function, rather than enhanced opsonophagocytosis for subsequent killing by neutrophil in vitro. Finally, the rHyr1p-N vaccine was protective against non-albicans Candida spp. These preclinical data demonstrate that rHyr1p-N is likely to be a novel target for developing both active and passive immunization strategies against Candida infections.
Collapse
Affiliation(s)
- Guanpingsheng Luo
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Ashraf S. Ibrahim
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Samuel W. French
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Pathology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - John E. Edwards
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Yue Fu
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
- David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Cutler JE, Corti M, Lambert P, Ferris M, Xin H. Horizontal transmission of Candida albicans and evidence of a vaccine response in mice colonized with the fungus. PLoS One 2011; 6:e22030. [PMID: 21818288 PMCID: PMC3139608 DOI: 10.1371/journal.pone.0022030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/13/2011] [Indexed: 11/27/2022] Open
Abstract
Disseminated candidiasis is the third leading nosocomial blood stream infection in the United States and is often fatal. We previously showed that disseminated candidiasis was preventable in normal mice by immunization with either a glycopeptide or a peptide synthetic vaccine, both of which were Candida albicans cell wall derived. A weakness of these studies is that, unlike humans, mice do not have a C. albicans GI flora and they lack Candida serum antibodies. We examined the influence of C. albicans GI tract colonization and serum antibodies on mouse vaccination responses to the peptide, Fba, derived from fructose bisphosphate aldolase which has cytosolic and cell wall distributions in the fungus. We evaluated the effect of live C. albicans in drinking water and antimicrobial agents on establishment of Candida colonization of the mouse GI tract. Body mass, C. albicans in feces, and fungal-specific serum antibodies were monitored longitudinally. Unexpectedly, C. albicans colonization occurred in mice that received only antibiotics in their drinking water, provided that the mice were housed in the same room as intentionally colonized mice. The fungal strain in unintentionally colonized mice appeared identical to the strain used for intentional GI-tract colonization. This is the first report of horizontal transmission and spontaneous C. albicans colonization in mice. Importantly, many Candida-colonized mice developed serum fungal-specific antibodies. Despite the GI-tract colonization and presence of serum antibodies, the animals made antibodies in response to the Fba immunogen. This mouse model has potential for elucidating C. albicans horizontal transmission and for exploring factors that induce host defense against disseminated candidiasis. Furthermore, a combined protracted GI-tract colonization with Candida and the possibility of serum antibody responses to the presence of the fungus makes this an attractive mouse model for testing the efficacy of vaccines designed to prevent human disseminated candidiasis.
Collapse
Affiliation(s)
- Jim E Cutler
- Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America.
| | | | | | | | | |
Collapse
|
41
|
Fidel PL, Cutler JE. Prospects for development of a vaccine to prevent and control vaginal candidiasis. Curr Infect Dis Rep 2011; 13:102-7. [PMID: 21308461 DOI: 10.1007/s11908-010-0143-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A vaccine against recurrent vulvovaginal candidiasis (RVVC) would benefit a large number of women who suffer from this debilitating syndrome. To date, several antigen formulations have been tested with modest results. In this article, we review the latest vaccine study reported in the literature. The candidate is a β-glucan conjugate administered with a human compatible adjuvant. Results in a mouse model of vaginitis were again modest for protection. However, the study included live animal imaging to quantify fungal burden; animals were challenged with a Candida strain carrying a gene encoding a glycophosphatidylinositol (GPI)-linked cell wall protein and luciferase. Fungal burden was expressed as photons following substrate administration. Protection appeared to be mediated by β-glucan antibodies. Although modest protection was observed, the imaging system was less variable than semi-quantitative plate counts of vaginal lavage fluid. Despite these advances in evaluating protection, a vaccine candidate against RVVC worthy of clinical testing remains elusive.
Collapse
Affiliation(s)
- Paul L Fidel
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA,
| | | |
Collapse
|
42
|
Abstract
Morbidity and mortality from invasive fungal infections remain unacceptably high despite availability of new antifungal agents, underscoring the need for more effective preventative strategies. Due to our enhanced understanding of the host defense and pathogenetic mechanisms that lead to invasive fungal infections, it should be feasible to develop vaccines targeting these infections. A common immunological theme across many vaccine candidates for invasive fungal infections has been the need to activate a cell-based, pro-inflammatory, Th1 or Th17 immune response to improve phagocytic killing of the fungi. Since neutralization of virulence factor functions has not been required for many active vaccines to function, the antigenic repertoire available for testing should not be limited to virulence factors. With expansion of our fundamental understanding of the immunology of fungal infections, the biggest barrier to development of fungal vaccines is the lack of available capital to translate discoveries made at the bench into biological agents used at the bedside. Continued education on the importance and feasibility of vaccination for such infections, combined with continued development of vaccine antigens and adjuvants, is necessary.
Collapse
Affiliation(s)
- Brad Spellberg
- Division of General Internal Medicine, Los Angeles Biomedical Research Institute, Harbor-University of California Los Angeles (UCLA) Medical Center 1124 West Carson Street, Liu Vaccine Center, Torrance, CA 90502 USA
| |
Collapse
|
43
|
Abstract
Fungal diseases represent an important paradigm in immunology, as they can result from either a lack of recognition by the immune system or overactivation of the inflammatory response. Research in this field is entering an exciting period of transition from studying the molecular and cellular bases of fungal virulence to determining the cellular and molecular mechanisms that maintain immune homeostasis with fungi. The fine line between these two research areas is central to our understanding of tissue homeostasis and its possible breakdown in fungal infections and diseases. Recent insights into immune responses to fungi suggest that functionally distinct mechanisms have evolved to achieve optimal host-fungus interactions in mammals.
Collapse
|
44
|
Liu M, Clemons KV, Bigos M, Medovarska I, Brummer E, Stevens DA. Immune responses induced by heat killed Saccharomyces cerevisiae: a vaccine against fungal infection. Vaccine 2011; 29:1745-53. [PMID: 21219976 DOI: 10.1016/j.vaccine.2010.12.119] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/04/2010] [Accepted: 12/23/2010] [Indexed: 01/30/2023]
Abstract
Heat-killed Saccharomyces cerevisiae (HKY) used as a vaccine protects mice against systemic aspergillosis and coccidioidomycosis. Little is known about the immune response induced by HKY vaccination, consequently our goal was to do an analysis of HKY-induced immune responses involved in protection. BALB/c mice were vaccinated subcutaneously 3 times with HKY, a protective reagent, and bronchoalveolar lavage fluid, spleen, lymph nodes, and serum collected 2-5 weeks later. Cultured spleen or lymph node cells were stimulated with HKY. Proliferation of HKY-stimulated spleen or lymph node cells was tested by Alamar Blue reduction and flow cytometry. Cytokines from lymphocyte supernatants and antibody to glycans in serum collected from HKY-vaccinated mice were measured by ELISA. The results show that HKY promoted spleen cell and lymph node cell proliferation from HKY-vaccinated mice but not from PBS-vaccinated control mice (all P<0.05). Cytokine measurement showed HKY significantly promoted IFNγ, IL-6 and IL-17A production by spleen cells and lymph node cells (all P<0.05 and P<0.01, respectively). Cytokine production by HKY-stimulated cells from PBS-vaccinated mice was lower than those from HKY-vaccinated (P<0.05). Cytokines in BAL from HKY-vaccinated were higher, 1.7-fold for IFNγ and 2.1-fold for TNFα, than in BAL from PBS-vaccinated. Flow cytometry of lymphocytes from HKY-vaccinated showed 52% of CD3(+) or 56% of CD8(+) cells exhibited cell division after stimulation with HKY, compared to non-stimulated controls (26 or 23%, respectively) or HKY-stimulated cells from PBS-vaccinated (31 or 34%). HKY also induced antibody against Saccharomyces glucan and mannan with titers 4- or 2-fold, respectively, above that in unvaccinated. Taken together, the results suggested that HKY vaccination induces significant and specific Th1 type cellular immune responses and antibodies to glucan and mannan.
Collapse
Affiliation(s)
- Min Liu
- California Institute for Medical Research, San Jose, CA, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Candida albicans is part of the normal human flora, and it grows on mucosal surfaces in healthy individuals. In susceptible hosts, this organism can cause both mucosal and hematogenously disseminated disease. For C. albicans to persist in the host and induce disease, it must be able to adhere to biotic and abiotic surfaces, invade host cells, and obtain iron. The C. albicans hypha-specific surface protein Als3 is a member of the agglutinin-like sequence (Als) family of proteins and is important in all of these processes. Functioning as an adhesin, Als3 mediates attachment to epithelial cells, endothelial cells, and extracellular matrix proteins. It also plays an important role in biofilm formation on prosthetic surfaces, both alone and in mixed infection with Streptococcus gordonii. Als3 is one of two known C. albicans invasins. It binds to host cell receptors such as E-cadherin and N-cadherin and thereby induces host cells to endocytose the organism. Als3 also binds to host cell ferritin and enables C. albicans to utilize this protein as a source of iron. Because of its multiple functions and its high expression level in vivo, Als3 is a promising target for vaccines that induce protective cell-mediated and antibody responses. This review will summarize the multiple functions of this interesting and multifunctional protein.
Collapse
|
46
|
An insight into the antifungal pipeline: selected new molecules and beyond. Nat Rev Drug Discov 2010; 9:719-27. [PMID: 20725094 DOI: 10.1038/nrd3074] [Citation(s) in RCA: 292] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Invasive fungal infections are increasing in incidence and are associated with substantial mortality. Improved diagnostics and the availability of new antifungals have revolutionized the field of medical mycology in the past decades. This Review focuses on recent developments in the antifungal pipeline, concentrating on promising candidates such as new azoles, polyenes and echinocandins, as well as agents such as nikkomycin Z and the sordarins. Developments in vaccines and antibody-based immunotherapy are also discussed. Few therapeutic products are currently in active development, and progression of therapeutic agents with fungus-specific mechanisms of action is of key importance.
Collapse
|
47
|
Baquir B, Lin L, Ibrahim AS, Fu Y, Avanesian V, Tu A, Edwards J, Spellberg B. Immunological reactivity of blood from healthy humans to the rAls3p-N vaccine protein. J Infect Dis 2010; 201:473-7. [PMID: 20039802 DOI: 10.1086/649901] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We determined reactivity of human blood to a vaccine based on the recombinant N-terminus of candidal Als3p (rAls3p-N) in preparation for future clinical trials. Healthy donor plasma had high immunoglobulin G titers (median, 1:51,200) and lower immunoglobulin A (median, 1:3,200) and immunoglobulin E (median, 1:128) titers to rAls3p-N by enzyme-linked immunosorbent assay. rAls3p-N stimulated interferon gamma (IFN-gamma) and interleukin (IL)-17, but not IL-4, from donor lymphocytes by enzyme-linked immunosorbent spot assay and IL-12 p70, IFN-gamma, IL-17, and IL-10 by cytometric bead array. Donors reacted to diverse immunodominant epitopes. Thus, facile humoral and cellular assays can monitor immune responses to the rAls3p-N vaccine in planned clinical trials.
Collapse
Affiliation(s)
- Beverlie Baquir
- Divisions of Infectious Diseases, Los Angeles Biomedical Research Institute, Harbor-University of California at Los Angeles (UCLA) Medical Center, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lin L, Ibrahim AS, Xu X, Farber JM, Avanesian V, Baquir B, Fu Y, French SW, Edwards JE, Spellberg B. Th1-Th17 cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog 2009; 5:e1000703. [PMID: 20041174 PMCID: PMC2792038 DOI: 10.1371/journal.ppat.1000703] [Citation(s) in RCA: 369] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 11/19/2009] [Indexed: 01/17/2023] Open
Abstract
We sought to define protective mechanisms of immunity to Staphylococcus aureus and Candida albicans bloodstream infections in mice immunized with the recombinant N-terminus of Als3p (rAls3p-N) vaccine plus aluminum hydroxide (Al(OH3) adjuvant, or adjuvant controls. Deficiency of IFN-γ but not IL-17A enhanced susceptibility of control mice to both infections. However, vaccine-induced protective immunity against both infections required CD4+ T-cell-derived IFN-γ and IL-17A, and functional phagocytic effectors. Vaccination primed Th1, Th17, and Th1/17 lymphocytes, which produced pro-inflammatory cytokines that enhanced phagocytic killing of both organisms. Vaccinated, infected mice had increased IFN-γ, IL-17, and KC, increased neutrophil influx, and decreased organism burden in tissues. In summary, rAls3p-N vaccination induced a Th1/Th17 response, resulting in recruitment and activation of phagocytes at sites of infection, and more effective clearance of S. aureus and C. albicans from tissues. Thus, vaccine-mediated adaptive immunity can protect against both infections by targeting microbes for destruction by innate effectors. The bacterium Staphylococcus aureus and the fungus Candida are the second and third leading cause of bloodstream infections in hospitalized patients. A vaccine to prevent such infections would be of enormous public health benefit. The leading hypothesis to explain why vaccines have not been successfully developed against these infections is that the microbes causing the infections are highly complex, and use multiple weapons (so-called “virulence factors”) to cause disease in humans. Therefore, a vaccine targeting either infection would have to neutralize many of these virulence factors at the same time. We have been developing a vaccine that simultaneously targets both types of infections. Our vaccine is based on a single virulence factor used by Candida, which has a similar shape to virulence factors used by S. aureus. In the current study, we report that our vaccine induces specialized cells in the immune system to more effectively call in reinforcements to kill the organisms. These data demonstrate that vaccines against both organisms can be developed even if they do not work by neutralizing multiple virulence factors, and therefore open the door to a far wider array of vaccine types against both infections.
Collapse
Affiliation(s)
- Lin Lin
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Ashraf S. Ibrahim
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Xin Xu
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua M. Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Valentina Avanesian
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Beverlie Baquir
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Yue Fu
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Samuel W. French
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- The Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - John E. Edwards
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Brad Spellberg
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- The Division of General Internal Medicine, Harbor-UCLA Medical Center, Torrance, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Kabir MA, Hussain MA. Human fungal pathogen Candida albicans in the postgenomic era: an overview. Expert Rev Anti Infect Ther 2009; 7:121-34. [PMID: 19622061 DOI: 10.1586/14787210.7.1.121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Candida albicans is an opportunistic human fungal pathogen and is responsible for candidiasis. Owing to the improvement in healthcare, the number of immunocompromised patients in hospitals has increased worldwide and these individuals are susceptible to infections caused by many pathogenic microbes, among which C. albicans is one of the major players. Currently, the complete genome sequence of this pathogen is available and the size of this was estimated to be of 16 Mb. Annotation of C. albicans genome revealed that there are 6114 open reading frames (ORFs), of which 774 are specific to C. albicans. This poses a challenge as well as an opportunity to the Candida community to understand the functions of the unknown genes, especially those specific to C. albicans. Efforts have been made by the Candida community to systematically delete the ORFs and assign the functions. This will, in turn, help in understanding the biology of C. albicans and its interactions with animals as well as humans, and better drugs can be developed to treat Candida infections. In this article, we review updates on the Candida biology in the context of the availability of the genome sequence, its functional analysis and anti-Candida therapy. Finally, in the light of present trends in Candida research and current challenges, various opportunities are identified and suggestions are made.
Collapse
Affiliation(s)
- M Anaul Kabir
- Department of Biotechnology, PA College of Engineering, Kairangala, Mangalore-574153, Karnataka, India.
| | | |
Collapse
|
50
|
Bozza S, Clavaud C, Giovannini G, Fontaine T, Beauvais A, Sarfati J, D'Angelo C, Perruccio K, Bonifazi P, Zagarella S, Moretti S, Bistoni F, Latgé JP, Romani L. Immune sensing of Aspergillus fumigatus proteins, glycolipids, and polysaccharides and the impact on Th immunity and vaccination. THE JOURNAL OF IMMUNOLOGY 2009; 183:2407-14. [PMID: 19625642 DOI: 10.4049/jimmunol.0900961] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ability of the fungus Aspergillus fumigatus to activate, suppress, or subvert host immune response during life cycle in vivo through dynamic changing of cell wall structure and secretion implicates discriminative immune sensing of distinct fungal components. In this study, we have comparatively assessed secreted- and membrane-anchored proteins, glycolipids, and polysaccharides for the ability to induce vaccine-dependent protection in transplanted mice and Th cytokine production by human-specific CD4(+) T cell clones. The results show that the different fungal components are endowed with the distinct capacity to activate Th cell responses in mice and humans, with secreted proteins inducing Th2 cell activation, membrane proteins Th1/Treg, glycolipids Th17, and polysaccharides mostly IL-10 production. Of interest, the side-by-side comparison revealed that at least three fungal components (a protease and two glycosylphosphatidylinositol-anchored proteins) retained their immunodominant Th1/Treg activating potential from mice to humans. This suggests that the broadness and specificity of human T cell repertoire against the fungus could be selectively exploited with defined immunoactive Aspergillus Ags.
Collapse
Affiliation(s)
- Silvia Bozza
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|