1
|
Esen M, Fischer-Herr J, Gabor JJ, Gaile JM, Fleischmann WA, Smeenk GW, de Moraes RA, Bélard S, Calle CL, Woldearegai TG, Egger-Adam D, Haug V, Metz C, Reguzova A, Löffler MW, Balode B, Matthies LC, Ramharter M, Amann R, Kremsner PG. First-in-Human Phase I Trial to Assess the Safety and Immunogenicity of an Orf Virus-Based COVID-19 Vaccine Booster. Vaccines (Basel) 2024; 12:1288. [PMID: 39591190 PMCID: PMC11599021 DOI: 10.3390/vaccines12111288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The emergence of SARS-CoV-2 has necessitated the development of versatile vaccines capable of addressing evolving variants. Prime-2-CoV_Beta, a novel Orf virus-based COVID-19 vaccine, was developed to express the SARS-CoV-2 spike and nucleocapsid antigens. This first-in-human, phase I, dose-finding clinical trial was conducted to assess the safety, reactogenicity, and immunogenicity of Prime-2-CoV_Beta as a booster in healthy adults. From June 2022 to June 2023, 60 participants in Germany received varying doses of Prime-2-CoV_Beta. The study demonstrated a favorable safety profile, with no serious adverse events (AEs) reported. All AEs were mild (107) or moderate (10), with the most common symptoms being pain at the injection site, fatigue, and headache. Immunogenicity assessments revealed robust vaccine-induced antigen-specific immune responses. High doses notably elicited significant increases in antibodies against the spike and nucleocapsid proteins as well as neutralizing antibodies against SARS-CoV-2 and its variants. Additionally, the vaccine did not induce ORFV-neutralizing antibodies, indicating the potential for repeated administration. In conclusion, Prime-2-CoV_Beta was safe, well tolerated, and immunogenic, demonstrating potential as a broadly protective vaccine against SARS-CoV-2 and its variants. These promising results support further evaluation of higher doses and additional studies to confirm efficacy and long-term protection. This trial was registered at ClinicalTrials, NCT05389319.
Collapse
Affiliation(s)
- Meral Esen
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné BP 242, Gabon
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Johanna Fischer-Herr
- Center for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Bernhard Nocht Center for Clinical Trials (BNCCT), 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg-Lübeck-Borstel-Riems, Germany
| | - Julian Justin Gabor
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Klinikverbund-Suedwest, Germany
| | - Johanna Marika Gaile
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Klinikverbund-Suedwest, Germany
| | - Wim Alexander Fleischmann
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Center for Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Geerten Willem Smeenk
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- MediTÜV GmbH & Co. KG, Hannover, Standort Hagen, 44263 Dortmund, Germany
| | | | - Sabine Bélard
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Carlos Lamsfus Calle
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Tamirat Gebru Woldearegai
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Diane Egger-Adam
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Verena Haug
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Carina Metz
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Alena Reguzova
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Markus W. Löffler
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Baiba Balode
- Center for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Bernhard Nocht Center for Clinical Trials (BNCCT), 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg-Lübeck-Borstel-Riems, Germany
| | - Lars C. Matthies
- Center for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Bernhard Nocht Center for Clinical Trials (BNCCT), 20359 Hamburg, Germany
| | - Michael Ramharter
- Center for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Bernhard Nocht Center for Clinical Trials (BNCCT), 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg-Lübeck-Borstel-Riems, Germany
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Peter G. Kremsner
- Institute of Tropical Medicine, University Hospital Tübingen, 72074 Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné BP 242, Gabon
- German Center for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
2
|
Pagallies F, Labisch JJ, Wronska M, Pflanz K, Amann R. Efficient and scalable clarification of Orf virus from HEK suspension for vaccine development. Vaccine X 2024; 18:100474. [PMID: 38523620 PMCID: PMC10958475 DOI: 10.1016/j.jvacx.2024.100474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
The Orf virus (ORFV) is a promising vector platform for the generation of vaccines against infectious diseases and cancer, highlighted by its progression to clinical testing phases. One of the critical steps during GMP manufacturing is the clarification of crude harvest because of the enveloped nature and large size of ORFV. This study presents the first description of ORFV clarification process from a HEK suspension batch process. We examined various filter materials, membrane pore sizes, harvest timings, and nuclease treatments. Employing the Ambr® crossflow system for high-throughput, small-volume experiments, we identified polypropylene-based Sartopure® PP3 filters as ideal. These filters, used in two consecutive stages with reducing pore sizes, significantly enhanced ORFV recovery and addressed scalability challenges. Moreover, we demonstrated that the time of harvest and the use of a nuclease play a decisive role to increase ORFV yields. With these findings, we were able to establish an efficient and scalable clarification process of ORFV derived from a suspension production process, essential for advancing ORFV vaccine manufacturing.
Collapse
Affiliation(s)
- Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Jennifer J. Labisch
- Lab Essentials Applications Development, Sartorius, Otto-Brenner-Straße 20, 37079 Göttingen, Germany
| | - Malgorzata Wronska
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- PRiME Vector Technologies, Herrenberger Straße 24, 72070 Tübingen, Germany
| | - Karl Pflanz
- Lab Essentials Applications Development, Sartorius, Otto-Brenner-Straße 20, 37079 Göttingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- PRiME Vector Technologies, Herrenberger Straße 24, 72070 Tübingen, Germany
| |
Collapse
|
3
|
Metz C, Haug V, Müller M, Amann R. Pharmacokinetic and Environmental Risk Assessment of Prime-2-CoV, a Non-Replicating Orf Virus-Based Vaccine against SARS-CoV-2. Vaccines (Basel) 2024; 12:492. [PMID: 38793743 PMCID: PMC11126055 DOI: 10.3390/vaccines12050492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Viral vector vaccines represent a substantial advancement in immunization technology, offering numerous benefits over traditional vaccine modalities. The Orf virus (ORFV) strain D1701-VrV is a particularly promising candidate for vaccine development due to its distinctive attributes, such as a good safety profile, the ability to elicit both humoral and cellular immunity, and its favorable genetic and thermal stability. Despite ORFV's theoretical safety advantages, such as its narrow host range and limited systemic spread post-inoculation, a critical gap persists between these theoretical benefits and the empirical evidence regarding its in vivo safety profile. This discrepancy underscores the need for comprehensive preclinical validations to bridge this knowledge gap, especially considering ORFV's use in humans. Our research introduces Prime-2-CoV, an innovative ORFV-based vaccine candidate against COVID-19, designed to elicit a robust immune response by expressing SARS-CoV-2 Nucleocapsid and Spike proteins. Currently under clinical trials, Prime-2-CoV marks the inaugural application of ORFV in human subjects. Addressing the aforementioned safety concerns, our extensive preclinical evaluation, including an environmental risk assessment (ERA) and detailed pharmacokinetic studies in rats and immunocompromised NOG mice, demonstrates Prime-2-CoV's favorable pharmacokinetic profile, negligible environmental impact, and minimal ERA risks. These findings not only affirm the vaccine's safety and efficacy but also pioneer the use of ORFV-based therapeutics, highlighting its potential for wider therapeutic applications.
Collapse
Affiliation(s)
- Carina Metz
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Verena Haug
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Melanie Müller
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| |
Collapse
|
4
|
Yu Y, Cui Y, Song B. The cooperation between orf virus and Staphylococcus aureus leads to intractable lesions in skin infection. Front Cell Infect Microbiol 2024; 13:1213694. [PMID: 38259972 PMCID: PMC10800892 DOI: 10.3389/fcimb.2023.1213694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/31/2023] [Indexed: 01/24/2024] Open
Abstract
A large amount of evidence shows that different kinds of microorganisms can jointly cope with environmental pressures including cell hosts. For example, in many cases, it has been found that secondary or mixed infection of animals caused by ORFV (an epitheliophilic Parapoxvirus) and bacteria (such as Staphylococcus aureus or Streptococcus) shows a mutual aid mode that indirectly leads to the deterioration of the disease. However, the lack of research on the co-pathogenic mechanism, including how to hijack and destroy the cell host in the pathological microenvironment, has hindered the in-depth understanding of the pathogenic process and consequences of this complex infection and the development of clinical treatment methods. Here, we summarized the current strategies of trapping cell hosts together, based on the previously defined ORFV-Host (O-H) system. The opportunistic invasion of S. aureus destroyed the delicate dynamic balance of the O-H, thus aggravating tissue damage through bacterial products (mediated by Agr), even causing sepsis or inducing cytokine storms. In fact, the virus products from its adaptive regulatory system (VARS) weaken the immune attacks and block molecular pathways, so that S. aureus can settle there more smoothly, and the toxins can penetrate into local tissues more quickly. This paper focuses on the main challenges faced by cell hosts in dealing with mixed infection, which provides a starting point for us to deal with this disease in the future.
Collapse
Affiliation(s)
- Yongzhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yudong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Baifen Song
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Yao X, Jing T, Geng Q, Pang M, Zhao X, Li S, Chen D, Ma W. Dual analysis of wild-type and attenuated Orf virus and host cell transcriptomes revealed novel virus-host cell interactions. mSphere 2023; 8:e0039823. [PMID: 37982609 PMCID: PMC10732022 DOI: 10.1128/msphere.00398-23] [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: 07/16/2023] [Accepted: 10/10/2023] [Indexed: 11/21/2023] Open
Abstract
IMPORTANCE Currently, the only available commercial vaccines for Orf virus (ORFV) are live attenuated vaccines, which present a potential risk of reversion to virulence. Therefore, understanding the pathogenic mechanisms of different virulent strains of ORFV and host immune responses triggered by these viruses is crucial for developing new vaccines and interventions. In this study, we found that the attenuated strain downregulates the host innate immune response and antiviral activity. In addition, we noted that the wild-type strain can induce the immune response pattern centered on interferon-stimulated genes and interferon regulatory factor gene family. We predicted that STAT1 and STAT2 are the main transcription factors upstream of target gene promoters through gene regulatory networks and exert significant regulatory effects on co-expressed genes. Our study elucidated the complex interaction between ORFV strains and host cell immune responses, providing new insights into vaccine research for ORFV.
Collapse
Affiliation(s)
- Xiaoting Yao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Tian Jing
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qingru Geng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ming Pang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuanduo Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shaofei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dekun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Wentao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
6
|
Eilts F, Harsy YMJ, Lothert K, Pagallies F, Amann R, Wolff MW. An investigation of excipients for a stable Orf viral vector formulation. Virus Res 2023; 336:199213. [PMID: 37657509 PMCID: PMC10495626 DOI: 10.1016/j.virusres.2023.199213] [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: 06/12/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
The Orf virus (ORFV) is a promising candidate for vector vaccines as well as for immunomodulatory and oncolytic therapies. However, few publications are available on its infectivity degradation or on suitable additives for prolonging its viral stability. In this study, the non-supplemented ORFV itself showed a very high stability at storage temperatures up to 28 °C, with a linear titer loss of 0.10 log infectious particles per day at 4 °C over a period of five weeks. To prolong this inherent stability, thirty additives, i.e., detergents, sugars, proteins, salts, and buffers as well as amino acids, were tested for their time- and temperature-dependent influence on the ORFV infectivity. A stabilizing effect on the infectivity was identified for the addition of all tested proteins, i.e., gelatine, bovine serum albumin, and recombinant human serum albumin (rHSA), of several sugars, i.e., mannitol, galactose, sucrose, and trehalose, of amino acids, i.e., arginine and proline, of the detergent Pluronic F68, and of the salt Na2SO4. The infectivity preservation was especially pronounced for proteins in liquid and frozen formulations, sugars in frozen state, and arginine und Pluronic in liquid formulations at high storage temperatures (37 °C). The addition of 1% rHSA with and without 5% sucrose was evaluated as a very stable formulation with a high safety profile and economic validity at storage temperatures up to 28 °C. At increased temperatures, the supplementation with 200 mM arginine performed better than with rHSA. In summary, this comprehensive data provides different options for a stable ORFV formulation, considering temperature, storage time, economic aspects, and downstream processing integrity.
Collapse
Affiliation(s)
- Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, Giessen 35390, Germany
| | - Yasmina M J Harsy
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, Giessen 35390, Germany
| | - Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, Giessen 35390, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, Tuebingen 72076, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, Tuebingen 72076, Germany; PRiME Vector Technologies, Herrenberger Straße 24, Tuebingen 72070, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, Giessen 35390, Germany.
| |
Collapse
|
7
|
Shao Y, Wang P, Zheng Y, Cui H, Lou Z, Li S, Huang F, Wu C. A replicative recombinant HPV16 E7 expression virus upregulates CD36 in C33A cells. Front Microbiol 2023; 14:1259510. [PMID: 37795297 PMCID: PMC10545859 DOI: 10.3389/fmicb.2023.1259510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 08/08/2023] [Indexed: 10/06/2023] Open
Abstract
Objective In past decades, the role of high-risk HPV (HR-HPV) infection in cancer pathogenesis has been extensively studied. The viral E7 protein expressed in pre-malignant cells has been identified as an ideal target for immunological intervention. However, the cultivation of HPV in vitro remains a significant challenge, as well as the lack of methods for expressing the HPV E7 protein and generating replication-competent recombinant viral particles, which posed a major obstacle to further exploration of the function and carcinogenic mechanisms of the E7 oncoprotein. Therefore, it is imperative to investigate novel methodologies to construct replication-competent recombinant viral particles that express the HPV E7 protein to facilitate the study of its function. Methods We initiated the construction of recombinant viral particles by utilizing the ccdB-Kan forward/reverse screening system in conjunction with the Red/ExoCET recombinant system. We followed the infection of C33A cells with the obtained recombinant virus to enable the continuous expression of HPV16 E7. Afterwards, the total RNA was extracted and performed transcriptome sequencing using RNA-Seq technology to identify differentially expressed genes associated with HPV-induced oncogenicity. Results We successfully established replicative recombinant viral particles expressing HPV16 E7 stably and continuously. The C33A cells were infected with recombinant viral particles to achieve overexpression of the E7 protein. Subsequently, RNA-Seq analysis was conducted to assess the changes in host cell gene expression. The results revealed an upregulation of the CD36 gene, which is associated with the HPV-induced oncogenic pathways, including PI3K-Akt and p53 signaling pathway. qRT-PCR analysis further identified that the upregulation of the CD36 gene due to the expression of HPV16 E7. Conclusion The successful expression of HPV16 E7 in cells demonstrates that the replicated recombinant virus retains the replication and infection abilities of Ad4, while also upregulating the CD36 gene involved in the PI3K-Akt signaling and p53 pathways, thereby promoting cell proliferation. The outcome of this study provides a novel perspective and serves as a solid foundation for further exploration of HPV-related carcinogenesis and the development of replicative HPV recombinant vaccines capable of inducing protective immunity against HPV.
Collapse
Affiliation(s)
- Yunting Shao
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Peng Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yunji Zheng
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Hongtu Cui
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Zhangrong Lou
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Chengjun Wu
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
8
|
Eilts F, Labisch JJ, Orbay S, Harsy YMJ, Steger M, Pagallies F, Amann R, Pflanz K, Wolff MW. Stability studies for the identification of critical process parameters for a pharmaceutical production of the Orf virus. Vaccine 2023:S0264-410X(23)00722-3. [PMID: 37353451 DOI: 10.1016/j.vaccine.2023.06.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
A promising new vaccine platform is based on the Orf virus, a viral vector of the genus Parapoxvirus, which is currently being tested in phase I clinical trials. The application as a vaccine platform mandates a well-characterised, robust, and efficient production process. To identify critical process parameters in the production process affecting the virus' infectivity, the Orf virus was subjected to forced degradation studies, including thermal, pH, chemical, and mechanical stress conditions. The tests indicated a robust virus infectivity within a pH range of 5-7.4 and in the presence of the tested buffering substances (TRIS, HEPES, PBS). The ionic strength up to 0.5 M had no influence on the Orf virus' infectivity stability for NaCl and MgCl2, while NH4Cl destabilized significantly. Furthermore, short-term thermal stress of 2d up to 37 °C and repeated freeze-thaw cycles (20cycles) did not affect the virus' infectivity. The addition of recombinant human serum albumin was found to reduce virus inactivation. Last, the Orf virus showed a low shear sensitivity induced by peristaltic pumps and mixing, but was sensitive to ultrasonication. The isoelectric point of the applied Orf virus genotype D1707-V was determined at pH3.5. The broad picture of the Orf virus' infectivity stability against environmental parameters is an important contribution for the identification of critical process parameters for the production process, and supports the development of a stable pharmaceutical formulation. The work is specifically relevant for enveloped (large DNA) viruses, like the Orf virus and like most vectored vaccine approaches.
Collapse
Affiliation(s)
- Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Jennifer J Labisch
- Lab Essentials Applications Development, Sartorius Stedim Biotech GmbH, August-Spindler-Straße 11, 37079 Goettingen, Germany; Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Lower Saxony, Germany
| | - Sabri Orbay
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Yasmina M J Harsy
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Marleen Steger
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15/3.008, 72076 Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15/3.008, 72076 Tuebingen, Germany; Prime Vector Technologies, Herrenberger Straße 24, 72070 Tuebingen, Germany
| | - Karl Pflanz
- Lab Essentials Applications Development, Sartorius Stedim Biotech GmbH, August-Spindler-Straße 11, 37079 Goettingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany.
| |
Collapse
|
9
|
Lin J, Sun S, Zhao K, Gao F, Wang R, Li Q, Zhou Y, Zhang J, Li Y, Wang X, Du L, Wang S, Li Z, Lu H, Lan Y, Song D, Guo W, Chen Y, Gao F, Zhao Y, Fan R, Guan J, He W. Oncolytic Parapoxvirus induces Gasdermin E-mediated pyroptosis and activates antitumor immunity. Nat Commun 2023; 14:224. [PMID: 36641456 PMCID: PMC9840172 DOI: 10.1038/s41467-023-35917-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
The advantage of oncolytic viruses (OV) in cancer therapy is their dual effect of directly killing tumours while prompting anti-tumour immune response. Oncolytic parapoxvirus ovis (ORFV) and other OVs are thought to induce apoptosis, but apoptosis, being the immunogenically inert compared to other types of cell death, does not explain the highly inflamed microenvironment in OV-challenged tumors. Here we show that ORFV and its recombinant therapeutic derivatives are able to trigger tumor cell pyroptosis via Gasdermin E (GSDME). This effect is especially prominent in GSDME-low tumor cells, in which ORFV-challenge pre-stabilizes GSDME by decreasing its ubiquitination and subsequently initiates pyroptosis. Consistently, GSDME depletion reduces the proportion of intratumoral cytotoxic T lymphocytes, pyroptotic cell death and the success of tumor ORFV virotherapy. In vivo, the OV preferentially accumulates in the tumour upon systemic delivery and elicits pyroptotic tumor killing. Consequentially, ORFV sensitizes immunologically 'cold' tumors to checkpoint blockade. This study thus highlights the critical role of GSDME-mediated pyroptosis in oncolytic ORFV-based antitumor immunity and identifies combinatorial cancer therapy strategies.
Collapse
Affiliation(s)
- Jing Lin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shihui Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, 130062, Changchun, China
| | - Renling Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Qi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yanlong Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yue Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Xinyue Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Le Du
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shuai Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Zi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Huijun Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Wei Guo
- Department of Hematology, The first hospital of Jilin University, 130021, Changchun, China
| | - Yujia Chen
- Department of Gastrointestinal Surgery, The first hospital of Jilin University, 130021, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, 130017, Changchun, China
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183, Huddinge, Sweden
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| |
Collapse
|
10
|
Lim J, Frecot DI, Stubenrauch F, Iftner T, Simon C. Cottontail rabbit papillomavirus E6 proteins: Interaction with MAML1 and modulation of the Notch signaling pathway. Virology 2022; 576:52-60. [PMID: 36155393 DOI: 10.1016/j.virol.2022.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022]
Abstract
Animal models are necessary to study how cutaneous human papillomaviruses (HPVs) are associated with carcinogenesis. The cottontail rabbit papillomavirus (CRPV) induces papilloma in the -cutaneous skin of rabbits and serves as an established animal model for HPVlinked carcinogenesis where viral E6 proteins play crucial roles. Several studies have reported the dysregulation of the Notch signaling pathway by cutaneous beta HPV, bovine PV and mouse PV E6 via their association with Mastermind-like 1 protein (MAML1), thus interfering with cell proliferation and differentiation. However, the CRPV E6 gene encodes an elongated E6 protein (long E6, LE6) and an N-terminally truncated product (short E6, SE6) making it unique from other E6 proteins. Here, we describe the interaction between both CRPV E6 proteins and MAML1 and their ability to downregulate the Notch signaling pathway which could be a way CRPV infection induces carcinogenesis similar to beta HPV.
Collapse
Affiliation(s)
- JiaWen Lim
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tuebingen, Tuebingen, Germany
| | - Desiree Isabella Frecot
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tuebingen, Tuebingen, Germany
| | - Frank Stubenrauch
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tuebingen, Tuebingen, Germany
| | - Thomas Iftner
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tuebingen, Tuebingen, Germany.
| | - Claudia Simon
- Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital Tuebingen, Tuebingen, Germany.
| |
Collapse
|
11
|
Müller M, Reguzova A, Löffler MW, Amann R. Orf Virus-Based Vectors Preferentially Target Professional Antigen-Presenting Cells, Activate the STING Pathway and Induce Strong Antigen-Specific T Cell Responses. Front Immunol 2022; 13:873351. [PMID: 35615366 PMCID: PMC9124846 DOI: 10.3389/fimmu.2022.873351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background Orf virus (ORFV)-based vectors are attractive for vaccine development as they enable the induction of potent immune responses against specific transgenes. Nevertheless, the precise mechanisms of immune activation remain unknown. This study therefore aimed to characterize underlying mechanisms in human immune cells. Methods Peripheral blood mononuclear cells were infected with attenuated ORFV strain D1701-VrV and analyzed for ORFV infection and activation markers. ORFV entry in susceptible cells was examined using established pharmacological inhibitors. Using the THP1-Dual™ reporter cell line, activation of nuclear factor-κB and interferon regulatory factor pathways were simultaneously evaluated. Infection with an ORFV recombinant encoding immunogenic peptides (PepTrio-ORFV) was used to assess the induction of antigen-specific CD8+ T cells. Results ORFV was found to preferentially target professional antigen-presenting cells (APCs) in vitro, with ORFV uptake mediated primarily by macropinocytosis. ORFV-infected APCs exhibited an activated phenotype, required for subsequent lymphocyte activation. Reporter cells revealed that the stimulator of interferon genes pathway is a prerequisite for ORFV-mediated cellular activation. PepTrio-ORFV efficiently induced antigen-specific CD8+ T cell recall responses in a dose-dependent manner. Further, activation and expansion of naïve antigen-specific CD8+ T cells was observed in response. Discussion Our findings confirm that ORFV induces a strong antigen-specific immune response dependent on APC uptake and activation. These data support the notion that ORFV D1701-VrV is a promising vector for vaccine development and the design of innovative immunotherapeutic applications.
Collapse
Affiliation(s)
- Melanie Müller
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Alena Reguzova
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Markus W. Löffler
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tübingen, Tübingen, Germany
- *Correspondence: Ralf Amann, ; Markus W. Löffler,
| | - Ralf Amann
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- *Correspondence: Ralf Amann, ; Markus W. Löffler,
| |
Collapse
|
12
|
Reguzova A, Ghosh M, Müller M, Rziha HJ, Amann R. Orf Virus-Based Vaccine Vector D1701-V Induces Strong CD8+ T Cell Response against the Transgene but Not against ORFV-Derived Epitopes. Vaccines (Basel) 2020; 8:E295. [PMID: 32531997 PMCID: PMC7349966 DOI: 10.3390/vaccines8020295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 01/14/2023] Open
Abstract
The potency of viral vector-based vaccines depends on their ability to induce strong transgene-specific immune response without triggering anti-vector immunity. Previously, Orf virus (ORFV, Parapoxvirus) strain D1701-V was reported as a novel vector mediating protection against viral infections. The short-lived ORFV-specific immune response and the absence of virus neutralizing antibodies enables repeated immunizations and enhancement of humoral immune responses against the inserted antigens. However, only limited information exists about the D1701-V induced cellular immunity. In this study we employed major histocompatibility complex (MHC) ligandomics and immunogenicity analysis to identify ORFV-specific epitopes. Using liquid chromatography-tandem mass spectrometry we detected 36 ORFV-derived MHC I peptides, originating from various proteins. Stimulated splenocytes from ORFV-immunized mice did not exhibit specific CD8+ T cell responses against the tested peptides. In contrast, immunization with ovalbumin-expressing ORFV recombinant elicited strong SIINFEKL-specific CD8+ T lymphocyte response. In conclusion, our data indicate that cellular immunity to the ORFV vector is negligible, while strong CD8+ T cell response is induced against the inserted transgene. These results further emphasize the ORFV strain D1701-V as an attractive vector for vaccine development. Moreover, the presented experiments describe prerequisites for the selection of T cell epitopes exploitable for generation of ORFV-based vaccines by reverse genetics.
Collapse
Affiliation(s)
| | | | | | | | - Ralf Amann
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (A.R.); (M.G.); (M.M.); (H.-J.R.)
| |
Collapse
|