88 results on '"Bottazzi, A"'
Search Results
2. Vaccine value profile for Hookworm.
- Author
-
Puchner, Karl Philipp, Bottazzi, Maria Elena, Periago, Victoria, Grobusch, Martin, Maizels, Rick, McCarthy, James, Lee, Bruce, Gaspari, Erika, Diemert, David, and Hotez, Peter
- Subjects
- *
HOOKWORM disease , *HOOKWORMS , *MIDDLE-income countries , *LOW-income countries , *VACCINE development , *PARASITIC diseases - Abstract
Hookworm, a parasitic infection, retains a considerable burden of disease, affecting the most underprivileged segments of the general population in endemic countries and remains one of the leading causes of mild to severe anemia in Low and Middle Income Countries (LMICs), particularly in pregnancy and children under 5. Despite repeated large scale Preventive Chemotherapy (PC) interventions since more than 3 decades, there is broad consensus among scholars that elimination targets set in the newly launched NTD roadmap will require additional tools and interventions. Development of a vaccine could constitute a promising expansion of the existing arsenal against hookworm. Therefore, we have evaluated the biological and implementation feasibility of the vaccine development as well as the added value of such a novel tool. Based on pipeline landscaping and the current knowledge on key biological aspects of the pathogen and its interactions with the host, we found biological feasibility of development of a hookworm vaccine to be moderate. Also, our analysis on manufacturing and regulatory issues as well as potential uptake yielded moderate implementation feasibility. Modelling studies suggest a that introduction of a vaccine in parallel with ongoing integrated interventions (PC, WASH, shoe campaigns), could substantially reduce burden of disease in a cost – saving mode. Finally a set of actions are recommended that might impact positively the likelihood of timely development and introduction of a hookworm vaccine. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Neglected tropical disease vaccines: hookworm, leishmaniasis, and schistosomiasis
- Author
-
Hotez, Peter J., primary, Bottazzi, Maria Elena, additional, Kaye, Paul M., additional, Lee, Bruce Y., additional, and Puchner, Karl Philipp, additional
- Published
- 2023
- Full Text
- View/download PDF
4. Vaccine value profile for Hookworm
- Author
-
Puchner, Karl Philipp, primary, Bottazzi, Maria Elena, additional, Periago, Victoria, additional, Grobusch, Martin, additional, Maizels, Rick, additional, McCarthy, James, additional, Lee, Bruce, additional, Gaspari, Erika, additional, Diemert, David, additional, and Hotez, Peter, additional
- Published
- 2023
- Full Text
- View/download PDF
5. A simple fluorescence-based assay for quantification of the Toll-Like Receptor agonist E6020 in vaccine formulations
- Author
-
Pollet, Jeroen, Versteeg, Leroy, Rezende, Wanderson, Strych, Ulrich, Gusovsky, Fabian, Hotez, Peter J., and Bottazzi, Maria Elena
- Published
- 2017
- Full Text
- View/download PDF
6. Safety and immunogenicity of the Na-APR-1 hookworm vaccine in infection-naïve adults
- Author
-
David J, Diemert, Maria, Zumer, Doreen, Campbell, Shannon, Grahek, Guangzhao, Li, Jin, Peng, Maria, Elena Bottazzi, Peter, Hotez, and Jeffrey, Bethony
- Subjects
Adult ,Ancylostomatoidea ,Vaccines ,General Veterinary ,General Immunology and Microbiology ,Public Health, Environmental and Occupational Health ,Aluminum Hydroxide ,Antibodies, Neutralizing ,Hookworm Infections ,Immunogenicity, Vaccine ,Lipid A ,Infectious Diseases ,Adjuvants, Immunologic ,Immunoglobulin G ,Tobacco ,Animals ,Humans ,Molecular Medicine ,Peptide Hydrolases - Abstract
The Necator americanus hemoglobinase, aspartic protease-1 (Na-APR-1), facilitates the ability of adult hookworms to parasitize the intestine of their human hosts. A recombinant version of APR-1 protected laboratory animals against hookworm infection by inducing neutralizing antibodies that block the protein's enzymatic activity and thereby impair blood feeding. A catalytically inactive version of the wild-type hemoglobinase (Na-APR-1(M74)) was expressed by infiltrating Nicotiana benthamiana tobacco plants with an Agrobacterium tumefaciens strain engineered to express the vaccine antigen, which was adjuvanted with aluminum hydroxide adjuvant (Alhydrogel).An open-label dose-escalation Phase 1 clinical trial was conducted in 40 healthy, hookworm-naïve adult volunteers in the United States. Participants received 30 or 100 µg of recombinant Na-APR-1(M74) with Alhydrogel or with Alhydrogel co-administered with one of two doses (2.5 or 5.0 µg) of an aqueous formulation of Glucopyranosyl Lipid A (GLA-AF). Intramuscular injections of study vaccine were administered on days 0, 56, and 112.Na-APR-1(M74)/Alhydrogel was well-tolerated; the most frequent adverse events were mild or moderate injection site tenderness and pain, and mild or moderate nausea and headache. No serious adverse events or adverse events of special interest related to vaccination were observed. Significantly higher levels of antigen-specific IgG antibodies were induced in those who received 100 µg Na-APR-1(M74) than those who received 30 µg of antigen. Adding GLA-AF to Na-APR-1(M74)/Alhydrogel resulted in higher levels of IgG against Na-APR-1(M74) in both the 30 and 100 µg Na-APR-1(M74) groups in comparison to the non-GLA formulations at the same antigen dose.Vaccination of hookworm-naïve adults with recombinant Na-APR-1(M74) was well-tolerated, safe, and induced significant IgG responses against the vaccine antigen Na-APR-1(M74). Given these favorable results, clinical trials of this product were initiated in hookworm-endemic areas of Gabon and Brazil.
- Published
- 2022
- Full Text
- View/download PDF
7. Development of chimeric protein as a multivalent vaccine for human Kinetoplastid infections: Chagas disease and leishmaniasis
- Author
-
Clímaco, Marianna de Carvalho, primary, de Figueiredo, Luiza Almeida, additional, Lucas, Rayane Cristina, additional, Pinheiro, Guilherme Rafael Gomide, additional, Dias Magalhães, Luísa Mourão, additional, Oliveira, Ana Laura Grossi de, additional, Almeida, Raquel Martins, additional, Barbosa, Fernando Sérgio, additional, Castanheira Bartholomeu, Daniella, additional, Bueno, Lilian Lacerda, additional, Mendes, Tiago Antonio, additional, Zhan, Bin, additional, Jones, Kathryn Marie, additional, Hotez, Peter, additional, Bottazzi, Maria Elena, additional, Oliveira, Fabrício Marcus Silva, additional, and Fujiwara, Ricardo Toshio, additional
- Published
- 2023
- Full Text
- View/download PDF
8. Advancing a vaccine to prevent human schistosomiasis
- Author
-
Merrifield, Maureen, Hotez, Peter J., Beaumier, Coreen M., Gillespie, Portia, Strych, Ulrich, Hayward, Tara, and Bottazzi, Maria Elena
- Published
- 2016
- Full Text
- View/download PDF
9. Advancing a vaccine to prevent hookworm disease and anemia
- Author
-
Hotez, Peter J., Beaumier, Coreen M., Gillespie, Portia M., Strych, Ulrich, Hayward, Tara, and Bottazzi, Maria Elena
- Published
- 2016
- Full Text
- View/download PDF
10. Status of vaccine research and development of vaccines for Chagas disease
- Author
-
Beaumier, Coreen M., Gillespie, Portia M., Strych, Ulrich, Hayward, Tara, Hotez, Peter J., and Bottazzi, Maria Elena
- Published
- 2016
- Full Text
- View/download PDF
11. Status of vaccine research and development of vaccines for leishmaniasis
- Author
-
Gillespie, Portia M., Beaumier, Coreen M., Strych, Ulrich, Hayward, Tara, Hotez, Peter J., and Bottazzi, Maria Elena
- Published
- 2016
- Full Text
- View/download PDF
12. Modeling the economic and epidemiologic impact of hookworm vaccine and mass drug administration (MDA) in Brazil, a high transmission setting
- Author
-
Bartsch, Sarah M., Hotez, Peter J., Hertenstein, Daniel L., Diemert, David J., Zapf, Kristina M., Bottazzi, Maria Elena, Bethony, Jeffrey M., Brown, Shawn T., and Lee, Bruce Y.
- Published
- 2016
- Full Text
- View/download PDF
13. Expression, purification, immunogenicity, and protective efficacy of a recombinant Tc24 antigen as a vaccine against Trypanosoma cruzi infection in mice
- Author
-
Martinez-Campos, Viridiana, Martinez-Vega, Pedro, Ramirez-Sierra, Maria Jesus, Rosado-Vallado, Miguel, Seid, Christopher A., Hudspeth, Elissa M., Wei, Junfei, Liu, Zhuyun, Kwityn, Cliff, Hammond, Molly, Ortega-López, Jaime, Zhan, Bin, Hotez, Peter J., Bottazzi, Maria Elena, and Dumonteil, Eric
- Published
- 2015
- Full Text
- View/download PDF
14. Safety and immunogenicity of the Na-APR-1 hookworm vaccine in infection-naïve adults
- Author
-
Diemert, David J., primary, Zumer, Maria, additional, Campbell, Doreen, additional, Grahek, Shannon, additional, Li, Guangzhao, additional, Peng, Jin, additional, Elena Bottazzi, Maria, additional, Hotez, Peter, additional, and Bethony, Jeffrey, additional
- Published
- 2022
- Full Text
- View/download PDF
15. A novel multi-epitope recombinant protein elicits an antigen-specific CD8+ T cells response in Trypanosoma cruzi-infected mice
- Author
-
González-López, Cristina, primary, Chen, Wen-Hsiang, additional, Alfaro-Chacón, Andrea, additional, Villanueva-Lizama, Liliana E., additional, Rosado-Vallado, Miguel, additional, Ramirez-Sierra, Maria Jesús, additional, Teh-Poot, Christian F., additional, Pollet, Jeroen, additional, Asojo, Oluwatoyin, additional, Jones, Kathryn M., additional, Hotez, Peter J., additional, Elena Bottazzi, Maria, additional, and Cruz-Chan, Julio Vladimir, additional
- Published
- 2022
- Full Text
- View/download PDF
16. The Human Hookworm Vaccine
- Author
-
Hotez, Peter J., Diemert, David, Bacon, Kristina M., Beaumier, Coreen, Bethony, Jeffrey M., Bottazzi, Maria Elena, Brooker, Simon, Couto, Artur Roberto, da Silva Freire, Marcos, Homma, Akira, Lee, Bruce Y., Loukas, Alex, Loblack, Marva, Morel, Carlos Medicis, Oliveira, Rodrigo Correa, and Russell, Philip K.
- Published
- 2013
- Full Text
- View/download PDF
17. The potential economic value of a cutaneous leishmaniasis vaccine in seven endemic countries in the Americas
- Author
-
Bacon, Kristina M., Hotez, Peter J., Kruchten, Stephanie D., Kamhawi, Shaden, Bottazzi, Maria Elena, Valenzuela, Jesus G., and Lee, Bruce Y.
- Published
- 2013
- Full Text
- View/download PDF
18. Vaccination with chimeric protein induces protection in murine model against ascariasis
- Author
-
Maria Elena Bottazzi, Joseane Camilla de Castro, Bin Zhan, Denise Silva Nogueira, Fabrício Marcus Silva Oliveira, Daniella Castanheira Bartholomeu, Laila Viana de Almeida, Lilian Lacerda Bueno, João Luís Reis-Cunha, Peter J. Hotez, Ricardo Toshio Fujiwara, Luísa Mourão Dias Magalhães, and Mariana Santos Cardoso
- Subjects
Recombinant Fusion Proteins ,030231 tropical medicine ,Monophosphoryl Lipid A ,Epitope ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,030212 general & internal medicine ,Ascaris suum ,B cell ,Ascariasis ,Mice, Inbred BALB C ,General Veterinary ,General Immunology and Microbiology ,biology ,Ascaris ,Vaccination ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Fusion protein ,Virology ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Antigens, Helminth ,Molecular Medicine ,Female - Abstract
An estimated 400 million people are infected by parasites of the genus Ascaris and the existing control measures are inefficient. Vaccine development using B cell antigens is a promising strategy for increased protection against this parasite. The present study aimed at developing a chimeric protein capable of conferring protection against infection by Ascaris sp. For this purpose, we performed B-cell epitope predictions on previously described vaccine candidate proteins from Ascaris suum and the corresponding peptides were used to construct a chimeric protein. Female BALB / c mice were immunized subcutaneously in three doses at 10 day intervals with a vaccine formulation comprised of the chimeric protein together with monophosphoryl lipid A (MPLA). Control groups included protein alone, MPLA, or PBS. After challenge infection, animals vaccinated with chimeric protein plus MPLA showed a reduction of 73.54% of larval load in the lung compared to control group animals. Animals immunized with chimeric protein plus MPLA also display higher IgG response and a reduction in lung inflammation. Our study highlights how chimeric proteins containing more than one B cell epitope can enhance immune protection against helminthic infection and offer new approaches to the development of Ascaris vaccines.
- Published
- 2021
- Full Text
- View/download PDF
19. Receptor-binding domain recombinant protein on alum-CpG induces broad protection against SARS-CoV-2 variants of concern
- Author
-
Pollet, Jeroen, primary, Strych, Ulrich, additional, Chen, Wen-Hsiang, additional, Versteeg, Leroy, additional, Keegan, Brian, additional, Zhan, Bin, additional, Wei, Junfei, additional, Liu, Zhuyun, additional, Lee, Jungsoon, additional, Kundu, Rahki, additional, Adhikari, Rakesh, additional, Poveda, Cristina, additional, Jose Villar, Maria, additional, Rani Thimmiraju, Syamala, additional, Lopez, Brianna, additional, Gillespie, Portia M., additional, Ronca, Shannon, additional, Kimata, Jason T., additional, Reers, Martin, additional, Paradkar, Vikram, additional, Hotez, Peter J., additional, and Elena Bottazzi, Maria, additional
- Published
- 2022
- Full Text
- View/download PDF
20. Yeast-expressed SARS-CoV recombinant receptor-binding domain (RBD219-N1) formulated with aluminum hydroxide induces protective immunity and reduces immune enhancement
- Author
-
Ulrich Strych, Peter J. Hotez, Anurodh S. Agrawal, Jeroen Pollet, Lanying Du, Shibo Jiang, Wen-Hsiang Chen, Chien-Te K Tseng, Xinrong Tao, Bi Hung Peng, Abdullah Algaissi, Maria Elena Bottazzi, and Sara Lustigman
- Subjects
viruses ,medicine.medical_treatment ,coronavirus ,Aluminum Hydroxide ,Antibodies, Viral ,medicine.disease_cause ,law.invention ,Mice ,0302 clinical medicine ,law ,vaccine ,030212 general & internal medicine ,skin and connective tissue diseases ,Neutralizing antibody ,Coronavirus ,Mice, Inbred BALB C ,Vaccines, Synthetic ,biology ,Viral Vaccine ,Viral Load ,eosinophil infiltration ,Recombinant Proteins ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,Vaccines, Subunit ,Recombinant DNA ,Molecular Medicine ,Female ,Antibody ,Coronavirus Infections ,Viral load ,Adjuvant ,COVID-19 Vaccines ,Pneumonia, Viral ,030231 tropical medicine ,severe acute respiratory syndrome ,Article ,Virus ,Betacoronavirus ,03 medical and health sciences ,Immune system ,Protein Domains ,medicine ,Animals ,Humans ,Pandemics ,General Veterinary ,General Immunology and Microbiology ,SARS-CoV-2 ,fungi ,Public Health, Environmental and Occupational Health ,COVID-19 ,Viral Vaccines ,Antibodies, Neutralizing ,Virology ,respiratory tract diseases ,body regions ,biology.protein ,recombinant protein - Abstract
Highlights • A SARS-CoV recombinant protein-based receptor binding domain (RBD) vaccine on alum provides high neutralizing antibody titers eliciting 100% protection (survival) against homologous SARS CoV virus challenge. • A SARS-CoV RBD vaccine on alum prevents pulmonary cellular infiltrates upon virus challenge. • A SARS-CoV RBD vaccine on alum greatly reduces lung eosinophils compared to a vaccine comprised of the SARS-CoV S protein. • The SARS-CoV RBD vaccine on alum is being developed as a human vaccine., We developed a severe acute respiratory syndrome (SARS) subunit recombinant protein vaccine candidate based on a high-yielding, yeast- engineered, receptor-binding domain (RBD219-N1) of the SARS beta-coronavirus (SARS-CoV) spike (S) protein. When formulated with Alhydrogel®, RBD219-N1 induced high-level neutralizing antibodies against both pseudotyped virus and a clinical (mouse-adapted) isolate of SARS-CoV. Here, we report that mice immunized with RBD219-N1/Alhydrogel® were fully protected from lethal SARS-CoV challenge (0% mortality), compared to ∼ 30% mortality in mice immunized with the SARS S protein formulated with Alhydrogel®, and 100% mortality in negative controls. An RBD219-N1 formulation with Alhydrogel® was also superior to the S protein, unadjuvanted RBD, and AddaVax (MF59-like adjuvant)-formulated RBD in inducing specific antibodies and preventing cellular infiltrates in the lungs upon SARS-CoV challenge. Specifically, a formulation with a 1:25 ratio of RBD219-N1 to Alhydrogel® provided high neutralizing antibody titers, 100% protection with non-detectable viral loads with minimal or no eosinophilic pulmonary infiltrates. As a result, this vaccine formulation is under consideration for further development against SARS-CoV and potentially other emerging and re-emerging beta-CoVs such as SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
21. The potential economic value of a therapeutic Chagas disease vaccine for pregnant women to prevent congenital transmission
- Author
-
Jorge Abelardo Falcón-Lezama, Elizabeth A. Mitgang, Pierre Buekens, Ulrich Strych, Sheba Meymandi, Lindsey Asti, Maria Elena Bottazzi, Owen J. Stokes-Cawley, Patrick T. Wedlock, Sarah M. Bartsch, Peter J. Hotez, and Bruce Y. Lee
- Subjects
Chagas disease ,Pediatrics ,medicine.medical_specialty ,Cost-Benefit Analysis ,Uncertainty interval ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,030225 pediatrics ,medicine ,Humans ,Chagas Disease ,030212 general & internal medicine ,Mexico ,health care economics and organizations ,Vaccines ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Transmission (medicine) ,Vaccination ,Public Health, Environmental and Occupational Health ,Infant ,Diagnostic test ,medicine.disease ,Infant mortality ,Latin America ,Infectious Diseases ,Molecular Medicine ,Female ,Pregnant Women ,Congenital transmission ,business - Abstract
BACKGROUND: Currently, there are no solutions to prevent congenital transmission of Chagas disease during pregnancy, which affects 1–40% of pregnant women in Latin America and is associated with a 5% transmission risk. With therapeutic vaccines under development, now is the right time to determine the economic value of such a vaccine to prevent congenital transmission. METHODS: We developed a computational decision model that represented the clinical outcomes and diagnostic testing strategies for an infant born to a Chagas-positive woman in Mexico and evaluated the impact of vaccination. RESULTS: Compared to no vaccination, a 25% efficacious vaccine averted 125 [95% uncertainty interval (UI): 122–128] congenital cases, 1.9 (95% UI: 1.6–2.2) infant deaths, and 78 (95% UI: 66–91) DALYs per 10,000 infected pregnant women; a 50% efficacious vaccine averted 251 (95% UI: 248–254) cases, 3.8 (95% UI: 3.6–4.2) deaths, and 160 (95% UI: 148–171) DALYs; and a 75% efficacious vaccine averted 376 (95% UI: 374–378) cases, 5.8 (95% UI: 5.5–6.1) deaths, and 238 (95% UI: 227–249) DALYs. A 25% efficacious vaccine was cost-effective (incremental cost-effectiveness ratio
- Published
- 2020
- Full Text
- View/download PDF
22. A phase 1 study of the safety, reactogenicity, and immunogenicity of a Schistosoma mansoni vaccine with or without glucopyranosyl lipid A aqueous formulation (GLA-AF) in healthy adults from a non-endemic area
- Author
-
G.E. Potter, W. Jones, Maria Elena Bottazzi, H. M. El Sahly, Shital M. Patel, Jordan L. Plieskatt, Jeffrey M. Bethony, Wendy A. Keitel, Peter J. Hotez, J.K. Kennedy, Gregory A. Deye, Robert L. Atmar, and David Diemert
- Subjects
Male ,medicine.medical_treatment ,Gastroenterology ,Cohort Studies ,Immunogenicity, Vaccine ,0302 clinical medicine ,Glucosides ,Schistosomiasis ,Medicine ,030212 general & internal medicine ,Vaccines ,education.field_of_study ,biology ,Immunogenicity ,Schistosoma mansoni ,Middle Aged ,Healthy Volunteers ,Lipid A ,Infectious Diseases ,Cytokines ,Molecular Medicine ,Female ,Adjuvant ,Adult ,medicine.medical_specialty ,Adolescent ,030231 tropical medicine ,Population ,Antibodies, Helminth ,Placebo ,Article ,Young Adult ,03 medical and health sciences ,Adjuvants, Immunologic ,Double-Blind Method ,Internal medicine ,Animals ,Humans ,education ,Reactogenicity ,Dose-Response Relationship, Drug ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Public Health, Environmental and Occupational Health ,Tropical disease ,medicine.disease ,biology.organism_classification ,Antigens, Helminth ,Immunoglobulin G ,business - Abstract
BACKGROUND: Schistosomiasis caused by Schistosoma mansoni (Sm) is a chronic, debilitating and potentially deadly neglected tropical disease. The licensure of a vaccine to prevent schistosomiasis would represent a major breakthrough in public health. METHODS: The safety and immunogenicity of a candidate Sm vaccine were assessed in this phase I, double-blind, dose-escalation trial. Seventy-two healthy Sm-naïve 18–50 year olds were randomized to receive 3 doses~ 8 weeks apart of saline placebo, or 10 μg, 30 μg, or 100 μg of recombinant Sm-Tetraspanin-2 vaccine formulated on aluminum hydroxide adjuvant (Sm-TSP-2/Al) with or without 5 μg of glucopyranosyl lipid A aqueous formulation (GLA-AF). Clinical and serologic responses were assessed for 1 year after dose 3. RESULTS: Vaccines were safe and well-tolerated. The most common reactions were injection site tenderness and pain, and headache and fatigue. Tenderness and pain were more frequent in groups receiving vaccine with GLA-AF than placebo (p = 0.0036 and p = 0.0014, respectively). Injection site reactions among those given Sm-TSP-2/Al with GLA-AF lasted 1.22 and 1.33 days longer than those receiving Sm-TSP-2/Al without GLA-AF or placebo (p < 0.001 for both). Dose- and adjuvant-related increases in serum IgG against Sm-TSP-2 were observed. Peak IgG levels occurred 14 days after dose 3. Seroresponse frequencies were low among recipients of Sm-TSP-2/Al without GLA-AF, but higher among subjects receiving 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF. More seroresponses were observed among those given 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF compared to placebo (p = 0.023 and p < 0.001, respectively). Seroresponse frequencies were 0%, 30%, 50%, and 89%, respectively, among those given placebo, or 10 μg, 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF, suggesting a dose-response relationship for Sm-TSP-2/Al with GLA-AF (p = 0.0001). CONCLUSIONS: Sm-TSP-2/Al with or without GLA-AF was safe and well tolerated in a Sm-naïve population. A vaccine like the one under development may represent our best hope to eliminating this neglected tropical disease.
- Published
- 2019
- Full Text
- View/download PDF
23. Receptor-binding domain recombinant protein on alum-CpG induces broad protection against SARS-CoV-2 variants of concern
- Author
-
Rakhi Tyagi Kundu, Ulrich Strych, Peter J. Hotez, Wen-Hsiang Chen, Shannon E. Ronca, Vikram Paradkar, Maria Jose Villar, Jeroen Pollet, Martin Reers, Cristina Poveda, Rakesh Adhikari, Brian Keegan, Leroy Versteeg, Jason T. Kimata, Jungsoon Lee, Portia M. Gillespie, Bin Zhan, Junfei Wei, Zhuyun Liu, Maria Elena Bottazzi, and Brianna Lopez
- Subjects
COVID-19 Vaccines ,chemical and pharmacologic phenomena ,Booster dose ,Antibodies, Viral ,complex mixtures ,Article ,law.invention ,chemistry.chemical_compound ,Mice ,Immune system ,law ,Animals ,Humans ,Neutralizing antibody ,COVID-19 Serotherapy ,biology ,General Veterinary ,General Immunology and Microbiology ,Chemistry ,Alum ,SARS-CoV-2 ,Public Health, Environmental and Occupational Health ,Immunization, Passive ,COVID-19 ,Virology ,Antibodies, Neutralizing ,Recombinant Proteins ,Titer ,Infectious Diseases ,CpG site ,Spike Glycoprotein, Coronavirus ,biology.protein ,Recombinant DNA ,Molecular Medicine ,Alum Compounds ,Antibody - Abstract
We conducted preclinical studies in mice using a yeast-produced SARS-CoV-2 RBD219-N1C1 subunit vaccine candidate formulated with aluminum hydroxide (alum) and CpG deoxynucleotides. This vaccine formulation is similar to one that entered advanced phase 3 clinical development in India. We compared the immune response of mice vaccinated with RBD219-N1C1/alum to mice vaccinated with RBD219-N1C1/alum+CpG. We also evaluated mice immunized with RBD219-N1C1/alum+CpG and boosted with RBD219-N1C1/alum. Mice were immunized twice intramuscularly at a 21-day interval. Compared to two doses of the RBD219-N1C1/alum formulation, the RBD219-N1C1/alum+CpG vaccine induced a stronger and more balanced Th1/Th2 cellular immune response, with high levels of neutralizing antibodies against the original Wuhan isolate of SARS-CoV-2 as well as the B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.1 (Kappa) variants. Notably, the sera from mice that received two 7 µg doses of RBD219-N1C1/alum+CpG showed more than 18 times higher neutralizing antibody titers against B.1.351, than the WHO International Standard for anti-SARS-CoV-2 immunoglobulin NIBSC 20/136. Interestingly, a booster dose did not require the addition of CpG to induce this effect. The data reported here reinforces that the RBD219-N1C1/alum+CpG vaccine formulation is suitable for inducing broadly neutralizing antibodies against SARS-CoV-2 including three variants of concern, B.1.1.7 (Alpha), B.1.351 (Beta), and B.1.617.1 (Kappa).
- Published
- 2021
24. The parasite-derived rOv-ASP-1 is an effective antigen-sparing CD4 + T cell-dependent adjuvant for the trivalent inactivated influenza vaccine, and functions in the absence of MyD88 pathway
- Author
-
Sara Lustigman, Zhuyun Liu, Bin Zhan, Parakkal Jovvian George, Joseph Koussa, Junfei Wei, Scott E. Hensley, Wanyan Deng, Sonia Jain, Maria Elena Bottazzi, Jie Lu, Kaela Parkhouse, Hao Shen, and Jiu Jiang
- Subjects
0301 basic medicine ,Influenza vaccine ,medicine.medical_treatment ,T cell ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Medicine ,030212 general & internal medicine ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Public Health, Environmental and Occupational Health ,Vaccination ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Immunization ,Immunology ,biology.protein ,Molecular Medicine ,Antibody ,business ,Adjuvant - Abstract
Vaccination remains the most cost-effective biomedical approach for controlling influenza disease. In times of pandemics, however, these vaccines cannot be produced in sufficient quantities for worldwide use by the current manufacturing capacities and practices. What is needed is the development of adjuvanted vaccines capable of inducing an adequate or better immune response at a decreased antigen dose. Previously we showed that the protein adjuvant rOv-ASP-1 augments influenza-specific antibody titers and survival after virus challenge in both young adult and old-age mice when administered with the trivalent inactivated influenza vaccine (IIV3). In this study we show that a reduced amount of rOv-ASP-1, with 40-times less IIV3 can also induce protection. Apparently the potency of the rOv-ASP-1 adjuvanted IIV3 vaccine is independent of the IIV3-specific Th1/Th2 associated antibody responses, and independent of the presence of HAI antibodies. However, CD4+ T helper cells were indispensable for the protection. Further, rOv-ASP-1 with or without IIV3 elicited the increased level of various chemokines, which are known chemoattractant for immune cells, into the muscle 4 h after immunization, and significantly induced the recruitment of monocytes, macrophages and neutrophils into the muscles. The recruited monocytes had higher expression of the activation marker MHCII on their surface as well as CXCR3 and CCR2; receptors for IP-10 and MCP-1, respectively. These results show that the rOv-ASP-1 adjuvant allows substantial antigen sparing of IIV3 by stimulating at the site of injection the accumulation of chemokines and the recruitment of immune cells that can augment the activation of CD4+ T cell immune responses, essential for the production of antibody responses. Protection elicited by the rOv-ASP-1 adjuvanted IIV3 vaccine also appears to function in the absence of MyD88-signaling. Future studies will attempt to delineate the precise mechanisms by which the rOv-ASP-1 adjuvanted IIV3 vaccine works.
- Published
- 2018
- Full Text
- View/download PDF
25. Engineering a stable CHO cell line for the expression of a MERS-coronavirus vaccine antigen
- Author
-
Mun Peak Nyon, Shibo Jiang, Lanying Du, Bi Hung Peng, Jeroen Pollet, Wanbo Tai, Abdullah Algaissi, Kevin S. Naceanceno, Christopher A. Seid, Chien Te K. Tseng, Ulrich Strych, Maria Elena Bottazzi, Anurodh S. Agrawal, and Peter J. Hotez
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Protein subunit ,Genetic Vectors ,Gene Expression ,CHO Cells ,Article ,law.invention ,03 medical and health sciences ,Epitopes ,Mice ,Immune system ,Cricetulus ,Immunogenicity, Vaccine ,law ,medicine ,Animals ,Neutralizing antibody ,Antigens, Viral ,biology ,General Immunology and Microbiology ,General Veterinary ,Chemistry ,Chinese hamster ovary cell ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,Virology ,Recombinant Proteins ,3. Good health ,Immunoglobulin Fc Fragments ,030104 developmental biology ,Infectious Diseases ,Monoclonal ,biology.protein ,Recombinant DNA ,Middle East Respiratory Syndrome Coronavirus ,Molecular Medicine ,Antibody ,Coronavirus Infections ,Genetic Engineering ,Adjuvant ,Protein Processing, Post-Translational - Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) has infected at least 2040 patients and caused 712 deaths since its first appearance in 2012, yet neither pathogen-specific therapeutics nor approved vaccines are available. To address this need, we are developing a subunit recombinant protein vaccine comprising residues 377–588 of the MERS-CoV spike protein receptor-binding domain (RBD), which, when formulated with the AddaVax adjuvant, it induces a significant neutralizing antibody response and protection against MERS-CoV challenge in vaccinated animals. To prepare for the manufacture and first-in-human testing of the vaccine, we have developed a process to stably produce the recombinant MERS S377-588 protein in Chinese hamster ovary (CHO) cells. To accomplish this, we transfected an adherent dihydrofolate reductase-deficient CHO cell line (adCHO) with a plasmid encoding S377-588 fused with the human IgG Fc fragment (S377-588-Fc). We then demonstrated the interleukin-2 signal peptide-directed secretion of the recombinant protein into extracellular milieu. Using a gradually increasing methotrexate (MTX) concentration to 5 μM, we increased protein yield by a factor of 40. The adCHO-expressed S377-588-Fc recombinant protein demonstrated functionality and binding specificity identical to those of the protein from transiently transfected HEK293T cells. In addition, hCD26/dipeptidyl peptidase-4 (DPP4) transgenic mice vaccinated with AddaVax-adjuvanted S377-588-Fc could produce neutralizing antibodies against MERS-CoV and survived for at least 21 days after challenge with live MERS-CoV with no evidence of immunological toxicity or eosinophilic immune enhancement. To prepare for large scale-manufacture of the vaccine antigen, we have further developed a high-yield monoclonal suspension CHO cell line.
- Published
- 2018
- Full Text
- View/download PDF
26. Vaccination with irradiated Ancylostoma caninum third stage larvae induces a Th2 protective response in dogs
- Author
-
Fujiwara, Ricardo T., Loukas, Alex, Mendez, Susana, Williamson, Angela L., Bueno, Lilian L., Wang, Yan, Samuel, Andre, Zhan, Bin, Bottazzi, Maria Elena, Hotez, Peter J., and Bethony, Jeffrey M.
- Published
- 2006
- Full Text
- View/download PDF
27. Vaccination with chimeric protein induces protection in murine model against ascariasis
- Author
-
de Castro, Joseane C., primary, de Almeida, Laila V., additional, Cardoso, Mariana Santos, additional, Oliveira, Fabricio M. Silva, additional, Nogueira, Denise S., additional, Reis-Cunha, João Luis, additional, Magalhaes, Luisa M.D., additional, Zhan, Bin, additional, Bottazzi, Maria Elena, additional, Hotez, Peter J., additional, Bueno, Lilian L., additional, Bartholomeu, Daniella Castanheira, additional, and Fujiwara, Ricardo T., additional
- Published
- 2021
- Full Text
- View/download PDF
28. Yeast-expressed SARS-CoV recombinant receptor-binding domain (RBD219-N1) formulated with aluminum hydroxide induces protective immunity and reduces immune enhancement
- Author
-
Chen, Wen-Hsiang, primary, Tao, Xinrong, additional, Agrawal, Anurodh Shankar, additional, Algaissi, Abdullah, additional, Peng, Bi-Hung, additional, Pollet, Jeroen, additional, Strych, Ulrich, additional, Bottazzi, Maria Elena, additional, Hotez, Peter J., additional, Lustigman, Sara, additional, Du, Lanying, additional, Jiang, Shibo, additional, and Tseng, Chien-Te K., additional
- Published
- 2020
- Full Text
- View/download PDF
29. Safety and immunogenicity of a recombinant vaccine against Trypanosoma cruzi in Rhesus macaques
- Author
-
Dumonteil, Eric, primary, Herrera, Claudia, additional, Tu, Weihong, additional, Goff, Kelly, additional, Fahlberg, Marissa, additional, Haupt, Erin, additional, Kaur, Amitinder, additional, Marx, Preston A., additional, Ortega-Lopez, Jaime, additional, Hotez, Peter J., additional, and Bottazzi, Maria Elena, additional
- Published
- 2020
- Full Text
- View/download PDF
30. The potential economic value of a therapeutic Chagas disease vaccine for pregnant women to prevent congenital transmission
- Author
-
Bartsch, Sarah M., primary, Stokes-Cawley, Owen J., additional, Buekens, Pierre, additional, Asti, Lindsey, additional, Bottazzi, Maria Elena, additional, Strych, Ulrich, additional, Wedlock, Patrick T., additional, Mitgang, Elizabeth A., additional, Meymandi, Sheba, additional, Falcon-Lezama, Jorge Abelardo, additional, Hotez, Peter J., additional, and Lee, Bruce Y., additional
- Published
- 2020
- Full Text
- View/download PDF
31. Expression of the Necator americanus hookworm larval antigen Na-ASP-2 in Pichia pastoris and purification of the recombinant protein for use in human clinical trials
- Author
-
Goud, Gaddam Narsa, Bottazzi, Maria Elena, Zhan, Bin, Mendez, Susana, Deumic, Vehid, Plieskatt, Jordan, Liu, Sen, Wang, Yan, Bueno, Lilian, Fujiwara, Ricardo, Samuel, Andre, Ahn, So Yeong, Solanki, Maneesha, Asojo, Oluwatoyin A., Wang, Jin, Bethony, Jeffrey M., Loukas, Alex, Roy, Michael, and Hotez, Peter J.
- Published
- 2005
- Full Text
- View/download PDF
32. Advancing a vaccine to prevent hookworm disease and anemia
- Author
-
Coreen M. Beaumier, Portia M. Gillespie, Tara Hayward, Ulrich Strych, Maria Elena Bottazzi, and Peter J. Hotez
- Subjects
0301 basic medicine ,Hookworm ,Biomedical Research ,Necator americanus ,Anemia ,Neglected tropical disease ,Disease ,Hookworm Infections ,03 medical and health sciences ,Dogs ,Immunology and Microbiology(all) ,parasitic diseases ,medicine ,Animals ,Humans ,Hookworm infection ,Clinical Trials as Topic ,Vaccines ,Hookworm vaccine ,General Veterinary ,General Immunology and Microbiology ,biology ,Public Health, Environmental and Occupational Health ,Necator ,biology.organism_classification ,medicine.disease ,veterinary(all) ,Virology ,030104 developmental biology ,Infectious Diseases ,Immunization ,Neglected tropical diseases ,Molecular Medicine ,Vaccine ,medicine.drug - Abstract
A human hookworm vaccine is under development and in clinical trials in Africa and the Americas. The vaccine contains the Na-APR-1 and Na-GST-1 antigens. It elicits neutralizing antibodies that interfere with establishment of the adult hookworm in the gut and the ability of the parasite to feed on blood. The vaccine target product profile is focused on the immunization of children to prevent hookworm infection and anemia caused by Necator americanus. It is intended for use in low- and middle-income countries where hookworm is highly endemic and responsible for at least three million disability-adjusted life years. So far, the human hookworm vaccine is being developed in the non-profit sector through the Sabin Vaccine Institute Product Development Partnership (PDP), in collaboration with the HOOKVAC consortium of European and African partners. We envision the vaccine to be incorporated into health systems as part of an elimination strategy for hookworm infection and other neglected tropical diseases, and as a means to reduce global poverty and address the Sustainable Development Goals.
- Published
- 2016
- Full Text
- View/download PDF
33. Status of vaccine research and development of vaccines for Chagas disease
- Author
-
Tara Hayward, Maria Elena Bottazzi, Coreen M. Beaumier, Peter J. Hotez, Ulrich Strych, and Portia M. Gillespie
- Subjects
0301 basic medicine ,Vaccine research ,Chagas disease ,Protozoan Vaccines ,Biomedical Research ,General Veterinary ,General Immunology and Microbiology ,business.industry ,030231 tropical medicine ,Public Health, Environmental and Occupational Health ,medicine.disease ,Virology ,veterinary(all) ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Immunology and Microbiology(all) ,parasitic diseases ,Medicine ,Humans ,Molecular Medicine ,Chagas Disease ,business - Published
- 2016
- Full Text
- View/download PDF
34. Advancing a vaccine to prevent human schistosomiasis
- Author
-
Coreen M. Beaumier, Ulrich Strych, Maureen Merrifield, Portia M. Gillespie, Tara Hayward, Peter J. Hotez, and Maria Elena Bottazzi
- Subjects
Primates ,0301 basic medicine ,Biomedical Research ,Tetraspanins ,Neglected tropical disease ,Developing country ,Schistosomiasis ,03 medical and health sciences ,Immunology and Microbiology(all) ,parasitic diseases ,medicine ,Animals ,Humans ,Glutathione Transferase ,Schistosoma ,Schistosoma haematobium ,Antigens, Bacterial ,Clinical Trials as Topic ,Vaccines ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,biology ,Reverse vaccinology ,Public Health, Environmental and Occupational Health ,Helminth Proteins ,Schistosoma mansoni ,Fatty Acid Transport Proteins ,biology.organism_classification ,Omics ,medicine.disease ,veterinary(all) ,Praziquantel ,030104 developmental biology ,Infectious Diseases ,Antigens, Helminth ,Immunology ,Molecular Medicine ,Vaccine ,medicine.drug - Abstract
Several candidate human schistosomiasis vaccines are in different stages of preclinical and clinical development. The major targets are Schistosoma haematobium (urogenitial schistosomiasis) and Schistosoma mansoni (intestinal schistosomiasis) that account for 99% of the world's 252 million cases, with 90% of these cases in Africa. Two recombinant S. mansoni vaccines – Sm-TSP-2 and Sm-14 are in Phase 1 trials, while Smp80 (calpain) is undergoing testing in non-human primates. Sh28GST, also known as Bilhvax is in advanced clinical development for S. haematobium infection. The possibility remains that some of these vaccines may cross-react to target both schistosome species. These vaccines were selected on the basis of their protective immunity in preclinical challenge models, through human immune-epidemiological studies or both. They are being advanced through a combination of academic research institutions, non-profit vaccine product development partnerships, biotechnology companies, and developing country vaccine manufacturers. In addition, new schistosome candidate vaccines are being identified through bioinformatics, OMICs approaches, and moderate throughput screening, although the full potential of reverse vaccinology for schistosomiasis has not yet been realized. The target product profiles of these vaccines vary but many focus on vaccinating children, in some cases following mass treatment with praziquantel, also known as vaccine-linked chemotherapy. Several regulatory pathways have been proposed, some of which rely on World Health Organization prequalification.
- Published
- 2016
- Full Text
- View/download PDF
35. Economic value of a therapeutic Chagas vaccine for indeterminate and Chagasic cardiomyopathy patients
- Author
-
Sheba Meymandi, Bruce Y. Lee, Samuel L. Randall, Ulrich Strych, Lindsey Asti, Peter J. Hotez, Maria Elena Bottazzi, Jorge Abelardo Falcón-Lezama, and Sarah M. Bartsch
- Subjects
Chagas disease ,medicine.medical_specialty ,Cost-Benefit Analysis ,Trypanosoma cruzi ,030231 tropical medicine ,Cardiomyopathy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Chagas Disease ,030212 general & internal medicine ,Nifurtimox ,Adverse effect ,Vaccines ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Vaccination ,Public Health, Environmental and Occupational Health ,medicine.disease ,Regimen ,Infectious Diseases ,Benznidazole ,Nitroimidazoles ,Chronic Disease ,Disease Progression ,Molecular Medicine ,business ,Indeterminate ,Cardiomyopathies ,medicine.drug - Abstract
BACKGROUND Therapeutic vaccines to prevent Chagas disease progression to cardiomyopathy are under development because the only available medications (benznidazole and nifurtimox) are limited by their efficacy, long treatment course, and side effects. Better understanding the potential clinical and economic value of such vaccines can help guide development and implementation. METHODS We developed a computational Chagas Markov model to evaluate the clinical and economic value of a therapeutic vaccine given in conjunction with benznidazole in indeterminate and chronic Chagas patients. Scenarios explored the vaccine's impact on reducing drug treatment dosage, duration, and adverse events, and risk of disease progression. RESULTS When administering standard-of-care benznidazole to 1000 indeterminate patients, 148 discontinued treatment and 219 progressed to chronic disease, resulting in 119 Chagas-related deaths and 2293 DALYs, costing $18.9 million in lifetime societal costs. Compared to benznidazole-only, therapeutic vaccination administered with benznidazole (25-75% reduction in standard dose and duration), resulted in 37-111 more patients (of 1000) completing treatment, preventing 11-219 patients from progressing, 6-120 deaths, and 108-2229 DALYs (5-100% progression risk reduction), saving ≤$16,171 per patient. When vaccinating determinate Kuschnir class 1 Chagas patients, 10-197 fewer patients further progressed compared to benznidazole-only, averting 11-228 deaths and 144-3037 DALYs (5-100% progression risk reduction), saving ≤$34,059 per person. When vaccinating Kuschnir class 2 patients, 13-279 fewer progressed (279 with benznidazole-only), averting 13-692 deaths and 283-10,785 DALYs (5-100% progression risk reduction), saving ≤$89,759. Therapeutic vaccination was dominant (saved costs and provided health benefits) with ≥ 5% progression risk reduction, except when only reducing drug treatment regimen and adverse events, but remained cost-effective when costing
- Published
- 2019
36. A phase 1 study of the safety, reactogenicity, and immunogenicity of a Schistosoma mansoni vaccine with or without glucopyranosyl lipid A aqueous formulation (GLA-AF) in healthy adults from a non-endemic area
- Author
-
Keitel, W.A., primary, Potter, G.E., additional, Diemert, D., additional, Bethony, J., additional, El Sahly, H.M., additional, Kennedy, J.K., additional, Patel, S.M., additional, Plieskatt, J.L., additional, Jones, W., additional, Deye, G., additional, Bottazzi, M.E., additional, Hotez, P.J., additional, and Atmar, R.L., additional
- Published
- 2019
- Full Text
- View/download PDF
37. Economic value of a therapeutic Chagas vaccine for indeterminate and Chagasic cardiomyopathy patients
- Author
-
Bartsch, Sarah M., primary, Bottazzi, Maria Elena, additional, Asti, Lindsey, additional, Strych, Ulrich, additional, Meymandi, Sheba, additional, Falcón-Lezama, Jorge Abelardo, additional, Randall, Samuel, additional, Hotez, Peter J., additional, and Lee, Bruce Y., additional
- Published
- 2019
- Full Text
- View/download PDF
38. Expression, purification, immunogenicity, and protective efficacy of a recombinant Tc24 antigen as a vaccine against Trypanosoma cruzi infection in mice
- Author
-
Elissa M. Hudspeth, Christopher A. Seid, Molly Hammond, Peter J. Hotez, Viridiana Martinez-Campos, Miguel Rosado-Vallado, Cliff Kwityn, Eric Dumonteil, Maria Jesus Ramirez-Sierra, Zhuyun Liu, Junfei Wei, Bin Zhan, Maria Elena Bottazzi, Pedro Martínez-Vega, and Jaime Ortega-López
- Subjects
Protozoan Vaccines ,Chagas disease ,Trypanosoma cruzi ,medicine.medical_treatment ,Antibodies, Protozoan ,Gene Expression ,Antigens, Protozoan ,Parasitemia ,Parasite Load ,Pichia ,Interferon-gamma ,Immune system ,Adjuvants, Immunologic ,Antigen ,parasitic diseases ,Escherichia coli ,medicine ,Animals ,Chagas Disease ,Cloning, Molecular ,Mice, Inbred BALB C ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,biology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,Th1 Cells ,medicine.disease ,biology.organism_classification ,Survival Analysis ,Virology ,Recombinant Proteins ,Vaccination ,Disease Models, Animal ,Lipid A ,Infectious Diseases ,Immunology ,Leukocytes, Mononuclear ,Molecular Medicine ,Female ,Adjuvant ,Spleen - Abstract
The Tc24 calcium binding protein from the flagellar pocket of Trypanosoma cruzi is under evaluation as a candidate vaccine antigen against Chagas disease. Previously, a DNA vaccine encoding Tc24 was shown to be an effective vaccine (both as a preventive and therapeutic intervention) in mice and dogs, as evidenced by reductions in T. cruzi parasitemia and cardiac amastigotes, as well as reduced cardiac inflammation and increased host survival. Here we developed a suitable platform for the large scale production of recombinant Tc24 (rTc24) and show that when rTc24 is combined with a monophosphoryl-lipid A (MPLA) adjuvant, the formulated vaccine induces a Th1-biased immune response in mice, comprised of elevated IgG2a antibody levels and interferon-gamma levels from splenocytes, compared to controls. These immune responses also resulted in statistically significant decreased T. cruzi parasitemia and cardiac amastigotes, as well as increased survival following T. cruzi challenge infections, compared to controls. Partial protective efficacy was shown regardless of whether the antigen was expressed in Escherichia coli or in yeast (Pichia pastoris). While mouse vaccinations will require further modifications in order to optimize protective efficacy, such studies provide a basis for further evaluations of vaccines comprised of rTc24, together with alternative adjuvants and additional recombinant antigens.
- Published
- 2015
- Full Text
- View/download PDF
39. The parasite-derived rOv-ASP-1 is an effective antigen-sparing CD4
- Author
-
Sonia, Jain, Parakkal Jovvian, George, Wanyan, Deng, Joseph, Koussa, Kaela, Parkhouse, Scott E, Hensley, Jiu, Jiang, Jie, Lu, Zhuyun, Liu, Junfei, Wei, Bin, Zhan, Maria Elena, Bottazzi, Hao, Shen, and Sara, Lustigman
- Subjects
Mice, Knockout ,Aging ,Receptors, CXCR3 ,Neutrophils ,Receptors, CCR2 ,Macrophages ,Histocompatibility Antigens Class II ,Helminth Proteins ,T-Lymphocytes, Helper-Inducer ,Viral Load ,Antibodies, Viral ,Survival Analysis ,Monocytes ,Article ,Mice ,Adjuvants, Immunologic ,Gene Expression Regulation ,Orthomyxoviridae Infections ,Influenza Vaccines ,Antigens, Helminth ,Myeloid Differentiation Factor 88 ,Animals ,Female ,Immunization ,Muscle, Skeletal - Abstract
Vaccination remains the most cost-effective biomedical approach for controlling influenza disease. In times of pandemics, however, these vaccines cannot be produced in sufficient quantities for worldwide use by the current manufacturing capacities and practices. What is needed is the development of adjuvanted vaccines capable of inducing an adequate or better immune response at a decreased antigen dose. Previously we showed that the protein adjuvant rOv-ASP-1 augments influenza-specific antibody titers and survival after virus challenge in both young adult and old-age mice when administered with the trivalent inactivated influenza vaccine (IIV3). In this study we show that a reduced amount of rOv-ASP-1, with 40-times less IIV3 can also induce protection. Apparently the potency of the rOv-ASP-1 adjuvanted IIV3 vaccine is independent of the IIV3-specific Th1/Th2 associated antibody responses, and independent of the presence of HAI antibodies. However, CD4(+) T helper cells were indispensable for the protection. Further, rOv-ASP-1 with or without IIV3 elicited the increased level of various chemokines, which are known chemoattractant for immune cells, into the muscle 4 hours after immunization, and significantly induced the recruitment of monocytes, macrophages and neutrophils into the muscles. The recruited monocytes had higher expression of the activation marker MHCII on their surface as well as CXCR3 and CCR2; receptors for IP-10 and MCP-1, respectively. These results show that the rOv-ASP-1 adjuvant allows substantial antigen sparing of IIV3 by stimulating at the site of injection the accumulation of chemokines and the recruitment of immune cells that can augment the activation of CD4(+) T cell immune responses, essential for the production of antibody responses. Protection elicited by the rOv-ASP-1 adjuvanted IIV3 vaccine also appears to function in the absence of MyD88-signaling. Future studies will attempt to delineate the precise mechanisms by which the rOv-ASP-1 adjuvanted IIV3 vaccine works.
- Published
- 2017
40. Human anthelminthic vaccines: Rationale and challenges
- Author
-
Hotez, Peter J., Strych, Ulrich, Lustigman, Sara, and Bottazzi, Maria Elena
- Published
- 2016
- Full Text
- View/download PDF
41. The parasite-derived rOv-ASP-1 is an effective antigen-sparing CD4 + T cell-dependent adjuvant for the trivalent inactivated influenza vaccine, and functions in the absence of MyD88 pathway
- Author
-
Jain, Sonia, primary, George, Parakkal Jovvian, additional, Deng, Wanyan, additional, Koussa, Joseph, additional, Parkhouse, Kaela, additional, Hensley, Scott E., additional, Jiang, Jiu, additional, Lu, Jie, additional, Liu, Zhuyun, additional, Wei, Junfei, additional, Zhan, Bin, additional, Bottazzi, Maria Elena, additional, Shen, Hao, additional, and Lustigman, Sara, additional
- Published
- 2018
- Full Text
- View/download PDF
42. Engineering a stable CHO cell line for the expression of a MERS-coronavirus vaccine antigen
- Author
-
Nyon, Mun Peak, primary, Du, Lanying, additional, Tseng, Chien-Te Kent, additional, Seid, Christopher A., additional, Pollet, Jeroen, additional, Naceanceno, Kevin S., additional, Agrawal, Anurodh, additional, Algaissi, Abdullah, additional, Peng, Bi-Hung, additional, Tai, Wanbo, additional, Jiang, Shibo, additional, Bottazzi, Maria Elena, additional, Strych, Ulrich, additional, and Hotez, Peter J., additional
- Published
- 2018
- Full Text
- View/download PDF
43. The potential economic value of a cutaneous leishmaniasis vaccine in seven endemic countries in the Americas
- Author
-
Kristina M. Bacon, Shaden Kamhawi, Peter J. Hotez, Bruce Y. Lee, Maria Elena Bottazzi, Stephanie D. Kruchten, and Jesus G. Valenzuela
- Subjects
Veterinary medicine ,Latin Americans ,Endemic Diseases ,Population ,Leishmaniasis, Cutaneous ,Article ,Cutaneous leishmaniasis ,Environmental health ,parasitic diseases ,Humans ,Medicine ,Computer Simulation ,education ,Leishmaniasis Vaccines ,education.field_of_study ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Human migration ,Public Health, Environmental and Occupational Health ,Leishmaniasis ,medicine.disease ,Economic benefits ,Infectious Diseases ,Costs and Cost Analysis ,Neglected tropical diseases ,Molecular Medicine ,Americas ,business - Abstract
Cutaneous leishmaniasis (CL) and its associated complications, including mucocutaneous leishmaniasis (MCL) and diffuse CL (DCL) have emerged as important neglected tropical diseases in Latin America, especially in areas associated with human migration, conflict, and recent deforestation. Because of the limitations of current chemotherapeutic approaches to CL, MCL, and DCL, several prototype vaccines are in different states of product and clinical development. We constructed and utilized a Markov decision analytic computer model to evaluate the potential economic value of a preventative CL vaccine in seven countries in Latin America: Bolivia, Brazil, Colombia, Ecuador, Mexico, Peru, and Venezuela. The results indicated that even a vaccine with a relatively short duration of protection and modest efficacy could be recommended for use in targeted locations, as it could prevent a substantial number of cases at low-cost and potentially even result in cost savings. If the population in the seven countries were vaccinated using a vaccine that provides at least 10 years of protection, an estimated 41,000-144,784 CL cases could be averted, each at a cost less than the cost of current recommended treatments. Further, even a vaccine providing as little as five years duration of protection with as little as 50% efficacy remains cost-effective compared with chemotherapy; additional scenarios resembling epidemic settings such as the one that occurred in Chaparral, Colombia in 2004 demonstrate important economic benefits.
- Published
- 2013
- Full Text
- View/download PDF
44. A simple fluorescence-based assay for quantification of the Toll-Like Receptor agonist E6020 in vaccine formulations
- Author
-
Wanderson C. Rezende, Leroy Versteeg, Fabian Gusovsky, Maria Elena Bottazzi, Peter J. Hotez, Ulrich Strych, and Jeroen Pollet
- Subjects
Agonist ,medicine.drug_class ,02 engineering and technology ,Chemistry Techniques, Analytical ,Fluorescence ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Vaccine adjuvant ,Adjuvants, Immunologic ,medicine ,Humans ,Fluorometry ,030212 general & internal medicine ,Receptor ,Phospholipids ,Fluorescent Dyes ,Toll-like receptor ,Vaccines ,Chromatography ,General Veterinary ,General Immunology and Microbiology ,Staining and Labeling ,Toll-Like Receptors ,Public Health, Environmental and Occupational Health ,021001 nanoscience & nanotechnology ,Molecular biology ,PLGA ,Infectious Diseases ,chemistry ,Molecular Medicine ,0210 nano-technology - Abstract
Despite the generally accepted immunostimulatory effect of Toll-Like Receptor 4 (TLR4) agonists and their value as vaccine adjuvants, there remains a demand for fast and easy quantification assays for these TLR4 agonists in order to accelerate and improve vaccine formulation studies. A new medium-throughput method was developed for the quantification of the TLR4 agonist, E6020, independent of the formulation composition. The assay uses a fluorescent hydrazide (DCCH) to label the synthetic lipopolysaccharide (LPS) analog E6020 through its diketone groups. This novel, low-cost, and fluorescence based assay may obviate the need for traditional approaches that primarily rely on Fourier transform infrared spectroscopy (FTIR) or mass spectrometry. The experiments were performed in a wide diversity of vaccine formulations containing E6020 to assess method robustness and accuracy. The assay was also expanded to evaluate the loading efficiency of E6020 in poly(lactic-co-glycolic acid) (PLGA) micro-particles.
- Published
- 2016
45. Modeling the economic and epidemiologic impact of hookworm vaccine and mass drug administration (MDA) in Brazil, a high transmission setting
- Author
-
Peter J. Hotez, Daniel L. Hertenstein, Shawn T. Brown, Bruce Y. Lee, David Diemert, Maria Elena Bottazzi, Jeffrey M. Bethony, Sarah M. Bartsch, and Kristina M. Zapf
- Subjects
Adolescent ,Cost-Benefit Analysis ,030231 tropical medicine ,Booster dose ,Mass Vaccination ,Article ,law.invention ,03 medical and health sciences ,Hookworm Infections ,0302 clinical medicine ,law ,Immunology and Microbiology(all) ,Environmental health ,parasitic diseases ,Medicine ,Humans ,030212 general & internal medicine ,Mass drug administration ,Child ,Hookworm infection ,health care economics and organizations ,Anthelmintics ,Hookworm vaccine ,Vaccines ,General Veterinary ,General Immunology and Microbiology ,Cost–benefit analysis ,biology ,business.industry ,Public Health, Environmental and Occupational Health ,Infant, Newborn ,Infant ,biology.organism_classification ,veterinary(all) ,Vaccination ,Infectious Diseases ,Transmission (mechanics) ,Models, Economic ,Child, Preschool ,Immunology ,Molecular Medicine ,business ,Brazil ,medicine.drug - Abstract
Background Although mass drug administration (MDA) has helped reduce morbidity attributed to soil-transmitted helminth infections in children, its limitations for hookworm infection have motivated the development of a human hookworm vaccine to both improve morbidity control and ultimately help block hookworm transmission leading to elimination. However, the potential economic and epidemiologic impact of a preventive vaccine has not been fully evaluated. Methods We developed a dynamic compartment model coupled to a clinical and economics outcomes model representing both the human and hookworm populations in a high transmission region of Brazil. Experiments simulated different implementation scenarios of MDA and vaccination under varying circumstances. Results Considering only intervention costs, both annual MDA and vaccination were highly cost-effective (ICERs ≤$790/DALY averted) compared to no intervention, with vaccination resulting in lower incremental cost-effectiveness ratios (ICERs ≤$444/DALY averted). From the societal perspective, vaccination was economically dominant (i.e., less costly and more effective) versus annual MDA in all tested scenarios, except when vaccination was less efficacious (20% efficacy, 5 year duration) and MDA coverage was 75%. Increasing the vaccine's duration of protection and efficacy, and including a booster injection in adulthood all increased the benefits of vaccination (i.e., resulted in lower hookworm prevalence, averted more disability-adjusted life years, and saved more costs). Assuming its target product profile, a pediatric hookworm vaccine drastically decreased hookworm prevalence in children to 14.6% after 20 years, compared to 57.2% with no intervention and 54.1% with MDA. The addition of a booster in adulthood further reduced the overall prevalence from 68.0% to 36.0% and nearly eliminated hookworm infection in children. Conclusion Using a human hookworm vaccine would be cost-effective and in many cases economically dominant, providing both health benefits and cost-savings. It could become a key technology in effecting control and elimination efforts for hookworm globally.
- Published
- 2015
46. Human anthelminthic vaccines: Rationale and challenges
- Author
-
Sara Lustigman, Ulrich Strych, Peter J. Hotez, and Maria Elena Bottazzi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Helminthiasis ,Developing country ,Schistosomiasis ,Innovative financing ,03 medical and health sciences ,Hookworm Infections ,Environmental health ,medicine ,Humans ,Developing Countries ,Lymphatic filariasis ,Licensure ,Anthelmintics ,Vaccines ,General Veterinary ,General Immunology and Microbiology ,Poverty ,business.industry ,Public health ,Public Health, Environmental and Occupational Health ,medicine.disease ,030104 developmental biology ,Infectious Diseases ,Immunology ,Neglected tropical diseases ,Molecular Medicine ,business - Abstract
Helminth infections are the most common afflictions of humankind, affecting almost every single person living in profound poverty. Through mass drug administration (MDA) we have seen sharp declines in the global prevalence of some helminth infections, including lymphatic filariasis, onchocerciasis, and ascariasis. However, since 1990, there has been no appreciable decrease in the global prevalence of hookworm infection, schistosomiasis, or food-borne trematodiases. Through the activities of a non-profit product development partnerships and two research institutes, a total of five human anthelmintic vaccines for hookworm infection (two) and schistosomiasis (three) have advanced from discovery through manufacture and are now in Phase 1 clinical testing. At least three additional antigens, including two for onchocerciasis and one for schistosomiasis, are also advancing through preclinical development with the intention of moving into the clinic soon. These preventive human anthelmintic vaccines could be used as stand-alone technologies administered to infants as part of the Expanded Program on Immunization (EPI), or together with anthelmintic drugs in programs linked to MDA. Significant hurdles though could hinder the advancement of these vaccines into later-stage clinical and product development and licensure. They include the absence of a major pharma partner (and the resultant access to adjuvants and industrial scale manufacturing expertise), an uncharted roadmap for how to introduce anthelmintic vaccines into appropriate health systems, uncertain global access and regulatory strategies that might need to rely on developing country vaccine manufacturers and national regulatory authorities, and the lack of innovative financing schemes. However, the public health and economic benefits of introducing these vaccines could be massive and therefore deserve international attention and support.
- Published
- 2015
47. Status of vaccine research and development of vaccines for leishmaniasis
- Author
-
Coreen M. Beaumier, Ulrich Strych, Peter J. Hotez, Tara Hayward, Maria Elena Bottazzi, and Portia M. Gillespie
- Subjects
0301 basic medicine ,Biomedical Research ,Leishmaniasis, Cutaneous ,03 medical and health sciences ,Cutaneous leishmaniasis ,Immunology and Microbiology(all) ,parasitic diseases ,medicine ,Animals ,Humans ,Psychodidae ,Leishmaniasis ,Leishmaniasis Vaccines ,Clinical Trials as Topic ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Public Health, Environmental and Occupational Health ,Tropical disease ,biology.organism_classification ,medicine.disease ,Leishmania ,Virology ,veterinary(all) ,030104 developmental biology ,Visceral leishmaniasis ,Infectious Diseases ,Immunology ,Neglected tropical diseases ,Molecular Medicine ,Leishmaniasis, Visceral ,business - Abstract
A number of leishmaniasis vaccine candidates are at various stages of pre-clinical and clinical development. Leishmaniasis is a vector-borne neglected tropical disease (NTD) caused by a protozoan parasite of the genus Leishmania and transmitted to humans by the bite of a sand fly. Visceral leishmaniasis (VL, kala-azar) is a high mortality NTD found mostly in South Asia and East Africa, while cutaneous leishmaniasis (CL) is a disfiguring NTD highly endemic in the Middle East, Central Asia, North Africa, and the Americas. Estimates attribute 50,000 annual deaths and 3.3 million disability-adjusted life years to leishmaniasis. There are only a few approved drug treatments, no prophylactic drug and no vaccine. Ideally, an effective vaccine against leishmaniasis will elicit long-lasting immunity and protect broadly against VL and CL. Vaccines such as Leish-F1, F2 and F3, developed at IDRI and designed based on selected Leishmania antigen epitopes, have been in clinical trials. Other groups, including the Sabin Vaccine Institute in collaboration with the National Institutes of Health are investigating recombinant Leishmania antigens in combination with selected sand fly salivary gland antigens in order to augment host immunity. To date, both VL and CL vaccines have been shown to be cost-effective in economic modeling studies.
- Published
- 2015
48. Expression of the Necator americanus hookworm larval antigen Na-ASP-2 in Pichia pastoris and purification of the recombinant protein for use in human clinical trials
- Author
-
Oluwatoyin A. Asojo, Bin Zhan, Alex Loukas, Lilian Lacerda Bueno, Jordan L. Plieskatt, Maria Elena Bottazzi, Andre Samuel, Michael Roy, Jeffrey M. Bethony, Ricardo Toshio Fujiwara, Maneesha Solanki, Jin Wang, Sen Liu, Gaddam Goud, Susana Mendez, Vehid Deumic, Peter J. Hotez, Yan Wang, and So Yeong Ahn
- Subjects
Necator americanus ,Pichia ,Pichia pastoris ,law.invention ,Rats, Sprague-Dawley ,Antigen ,law ,medicine ,Animals ,Humans ,Potency ,Clinical Trials as Topic ,Vaccines, Synthetic ,Hookworm vaccine ,General Veterinary ,General Immunology and Microbiology ,biology ,Vaccination ,Public Health, Environmental and Occupational Health ,Antibody titer ,Helminth Proteins ,biology.organism_classification ,Molecular biology ,Recombinant Proteins ,Rats ,Infectious Diseases ,Larva ,Recombinant DNA ,biology.protein ,Molecular Medicine ,Antibody ,medicine.drug - Abstract
The ASP-2 protein secreted by infective larvae of the human hookworm, Necator americanus, is under development as a recombinant vaccine. Recombinant Na-ASP-2 was expressed in Pichia pastoris, and the purified protein was characterized. At the 60 L scale, the 21.3 kDa recombinant protein was produced at a yield of 0.4 g/L. When formulated with Alhydrogel and injected into rats to determine immunological potency, three 50 microg doses of the formulated recombinant protein elicited geometric mean antibody titers up to 1:234,881. Rat anti-Na-ASP-2 antibody recognized larval-derived ASP-2 and also inhibited larval migration through skin in vitro. The processes developed and tested for the high yield production of recombinant Na-ASP-2 provide a foundation for clinical vaccine development.
- Published
- 2005
- Full Text
- View/download PDF
49. Pentraxin 3, a non-redundant soluble pattern recognition receptor involved in innate immunity
- Author
-
Alberto Mantovani, Cecilia Garlanda, and Barbara Bottazzi
- Subjects
Innate immune system ,General Veterinary ,General Immunology and Microbiology ,biology ,Pentraxins ,Lipopolysaccharide ,C-reactive protein ,Public Health, Environmental and Occupational Health ,Pattern recognition receptor ,PTX3 ,Immunity, Innate ,Mice ,Serum Amyloid P-Component ,chemistry.chemical_compound ,C-Reactive Protein ,Infectious Diseases ,chemistry ,Immunology ,biology.protein ,Animals ,Humans ,Molecular Medicine ,Tumor necrosis factor alpha ,Serum amyloid P component - Abstract
Pentraxin 3 (PTX3) is the first long pentraxin identified. Long pentraxins consist of a C-terminal pentraxin domain, which has sequence similarity to C-reactive protein (CRP) and serum amyloid P (SAP) component (the classic short pentraxins), and of an unrelated N-terminal portion. PTX3 is made by diverse cell types, most prominently endothelial cells, macrophages and dendritic cells, in response to primary inflammatory signals (e.g. interleukin-1 (IL-1), tumour necrosis factor (TNF), lipopolysaccharide (LPS)). It binds diverse ligands, including microbial moieties, C1q and apoptotic cells. Evidence suggests that PTX3 plays a role in the regulation of innate resistance to pathogens, inflammatory reactions, possibly clearance of self-components and female fertility.
- Published
- 2003
- Full Text
- View/download PDF
50. The Human Hookworm Vaccine
- Author
-
Peter J, Hotez, David, Diemert, Kristina M, Bacon, Coreen, Beaumier, Jeffrey M, Bethony, Maria Elena, Bottazzi, Simon, Brooker, Artur Roberto, Couto, Marcos da Silva, Freire, Akira, Homma, Bruce Y, Lee, Alex, Loukas, Marva, Loblack, Carlos Medicis, Morel, Rodrigo Correa, Oliveira, and Philip K, Russell
- Subjects
Ancylostomatoidea ,Vaccines ,Hookworm ,Biomedical Research ,Public Sector ,Clinical Trials, Phase I as Topic ,Academies and Institutes ,Anemia ,Review ,Global Health ,Public-Private Sector Partnerships ,Recombinant Proteins ,Toll-Like Receptor 4 ,Hookworm Infections ,Technology Transfer ,Antigens, Helminth ,Helminth ,Animals ,Humans ,Na-GST-1 ,Na-APR-1 - Abstract
Highlights ► Human hookworm infection is a leading cause of iron deficiency anemia. ► An estimated 700 million people in developing countries are affected. ► The Sabin Vaccine Institute PDP is developing the vaccine in collaboration with FIOCRUZ. ► The vaccine comprises two recombinant protein antigens on alum and a TLR4 agonist. ► The partnership's plan is that the vaccine will be licensed by 2020., Hookworm infection is one of the world's most common neglected tropical diseases and a leading cause of iron deficiency anemia in low- and middle-income countries. A Human Hookworm Vaccine is currently being developed by the Sabin Vaccine Institute and is in phase 1 clinical testing. The candidate vaccine is comprised of two recombinant antigens known as Na-GST-1 and Na-APR-1, each of which is an important parasite enzyme required for hookworms to successfully utilize host blood as a source of energy. The recombinant proteins are formulated on Alhydrogel® and are being tested in combination with a synthetic Toll-like receptor 4 agonist. The aim of the vaccine is to induce anti-enzyme antibodies that will reduce both host blood loss and the number of hookworms attached to the gut. Transfer of the manufacturing technology to the Oswaldo Cruz Foundation (FIOCRUZ)/Bio-Manguinhos (a Brazilian public sector developing country vaccine manufacturer) is planned, with a clinical development plan that could lead to registration of the vaccine in Brazil. The vaccine would also need to be introduced in the poorest regions of Africa and Asia, where hookworm infection is highly endemic. Ultimately, the vaccine could become an essential tool for achieving hookworm control and elimination, a key target in the 2012 London Declaration on Neglected Tropical Diseases.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.