1
|
Ebrahimi A, Barati T, Mirzaei Z, Fattahi F, Mansoori Derakhshan S, Shekari Khaniani M. An overview on the interaction between non-coding RNAs and CTLA-4 gene in human diseases. Med Oncol 2024; 42:13. [PMID: 39585522 DOI: 10.1007/s12032-024-02552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024]
Abstract
Cytotoxic T lymphocyte antigen 4 (CTLA-4), in conjunction with PD-1 and CD28, plays a pivotal role in the modulation of T-cell activation. Specifically, CTLA-4 exerts its influence by impeding the generation of IL-2 and the proliferation of T cells. CTLA-4, being a receptor with a high affinity, engages in competitive binding with CD28 for the interaction with primary T-cell activator molecules, specifically CD80 and CD86. The appropriate functioning of T-cell activation is contingent upon maintaining a precise equilibrium between CTLA-4 and CD28. Consequently, any disruption in the expression of CTLA-4 significantly enhances the risk for a range of severe ailments, such as cancer, infectious diseases, allergies, and notably autoimmune diseases. The significance of epigenetic regulation of CTLA-4, particularly through non-coding RNAs (ncRNAs) such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), has considerable weight within this particular framework. To date, there have been associations shown between various abnormalities in the expression of ncRNAs that regulate CTLA-4 and clinicopathological characteristics. Nevertheless, it is evident that there is a lack of a comprehensive investigation. Hence, the present work was undertaken to summarize the existing research on the epigenetic control of CTLA-4, with a primary emphasis on elucidating the regulatory procedures, biological processes, and clinical applications in human diseases. The objective of this review is to acquire a thorough comprehension of the relationship between RNA/lncRNA/miRNA/mRNA (CTLA-4) and its role in the progression of diverse human disorders.
Collapse
Affiliation(s)
- Amir Ebrahimi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Tahereh Barati
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Zohreh Mirzaei
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Fatemeh Fattahi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Sima Mansoori Derakhshan
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Mahmoud Shekari Khaniani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran.
| |
Collapse
|
2
|
Yi XM, Lei YL, Li M, Zhong L, Li S. The monkeypox virus-host interplays. CELL INSIGHT 2024; 3:100185. [PMID: 39144256 PMCID: PMC11321328 DOI: 10.1016/j.cellin.2024.100185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Monkeypox virus (MPXV) is a DNA virus belonging to the Orthopoxvirus genus within the Poxviridae family which can cause a zoonotic infection. The unexpected non-endemic outbreak of mpox in 2022 is considered as a new global threat. It is imperative to take proactive measures, including enhancing our understanding of MPXV's biology and pathogenesis, and developing novel antiviral strategies. The host immune responses play critical roles in defensing against MPXV infection while the virus has also evolved multiple strategies for immune escape. This review summarizes the biological features, antiviral immunity, immune evasion mechanisms, pathogenicity, and prevention strategies for MPXV.
Collapse
Affiliation(s)
- Xue-Mei Yi
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ya-Li Lei
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Mi Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Li Zhong
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Shu Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
3
|
Fang D, Liu Y, Dou D, Su B. The unique immune evasion mechanisms of the mpox virus and their implication for developing new vaccines and immunotherapies. Virol Sin 2024; 39:709-718. [PMID: 39181538 DOI: 10.1016/j.virs.2024.08.008] [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: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Mpox is an infectious and contagious zoonotic disease caused by the mpox virus (MPXV), which belongs to the genus Orthopoxvirus. Since 2022, MPXV has posed a significant threat to global public health. The emergence of thousands of cases across the Western Hemisphere prompted the World Health Organization to declare an emergency. The extensive coevolutionary history of poxviruses with humans has enabled these viruses to develop sophisticated mechanisms to counter the human immune system. Specifically, MPXV employs unique immune evasion strategies against a wide range of immunological elements, presenting a considerable challenge for treatment, especially following the discontinuation of routine smallpox vaccination among the general population. In this review, we start by discussing the entry of the mpox virus and the onset of early infection, followed by an introduction to the mechanisms by which the mpox virus can evade the innate and adaptive immune responses. Two caspase-1 inhibitory proteins and a PKR escape-related protein have been identified as phylogenomic hubs involved in modulating the immune environment during the MPXV infection. With respect to adaptive immunity, mpox viruses exhibit unique and exceptional T-cell inhibition capabilities, thereby comprehensively remodeling the host immune environment. The viral envelope also poses challenges for the neutralizing effects of antibodies and the complement system. The unique immune evasion mechanisms employed by MPXV make novel multi-epitope and nucleic acid-based vaccines highly promising research directions worth investigating. Finally, we briefly discuss the impact of MPXV infection on immunosuppressed patients and the current status of MPXV vaccine development. This review may provide valuable information for the development of new immunological treatments for mpox.
Collapse
Affiliation(s)
- Dong Fang
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China
| | - Yan Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Dou Dou
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China
| | - Bin Su
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Central Laboratory, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
4
|
Chen L, Chao Y, Li W, Wu Z, Wang Q. Soluble immune checkpoint molecules in cancer risk, outcomes prediction, and therapeutic applications. Biomark Res 2024; 12:95. [PMID: 39218939 PMCID: PMC11368031 DOI: 10.1186/s40364-024-00647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy has emerged as a pivotal modality in cancer treatment, with immune checkpoint inhibitors effectively combating malignancies by impeding crucial pathways within the immune system and stimulating patients' immune responses. Soluble forms of immune checkpoints exhibit a remarkable diversity and can be readily tracked in circulation, holding immense potential as biomarkers for cancer treatment. An increasing number of studies focused on soluble immune checkpoints in cancer have emerged thanks to technological advancements. In this systematic review, we comprehensively summarized the recent studies on soluble immune checkpoints in human cancer risk prediction, outcome prediction, therapeutic applications, and potential molecular mechanisms, which demonstrated the promising future of soluble immune checkpoints in clinical applications. The clinical relevance of soluble immune checkpoints has been recognized in multiple cancers, yet the therapeutic applications and mechanisms remain obscure. Interpreting the impacts and mechanisms of soluble immune checkpoints could shed a light on the novel strategies of cancer screening, treatments, and outcome prediction.
Collapse
Affiliation(s)
- Lin Chen
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Zhejiang, PR China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuqing Chao
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Zhejiang, PR China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenjing Li
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Zhejiang, PR China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhixia Wu
- Department of Service and Purchase, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Qinchuan Wang
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Zhejiang, PR China.
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Azar F, Deforges J, Demeusoit C, Kleinpeter P, Remy C, Silvestre N, Foloppe J, Fend L, Spring-Giusti C, Quéméneur E, Marchand JB. TG6050, an oncolytic vaccinia virus encoding interleukin-12 and anti-CTLA-4 antibody, favors tumor regression via profound immune remodeling of the tumor microenvironment. J Immunother Cancer 2024; 12:e009302. [PMID: 39060022 DOI: 10.1136/jitc-2024-009302] [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] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND TG6050 was designed as an improved oncolytic vector, combining the intrinsic properties of vaccinia virus to selectively replicate in tumors with the tumor-restricted expression of recombinant immune effectors to modify the tumor immune phenotype. These properties might be of particular interest for "cold" tumors, either poorly infiltrated or infiltrated with anergic T cells. METHODS TG6050, an oncolytic vaccinia virus encodes single-chain human interleukin-12 (hIL-12) and full-length anti-cytotoxic T-lymphocyte-associated antigen-4 (@CTLA-4) monoclonal antibody. The relevant properties of TG6050 (replication, cytopathy, transgenes expression and functionality) were extensively characterized in vitro. The biodistribution and pharmacokinetics of the viral vector, @CTLA-4 and IL-12, as well as antitumoral activities (alone or combined with immune checkpoint inhibitors) were investigated in several "hot" (highly infiltrated) and "cold" (poorly infiltrated) syngeneic murine tumor models. The mechanism of action was deciphered by monitoring both systemic and intratumoral immune responses, and by tumor transcriptome analysis. The safety of TG6050 after repeated intravenous administrations was evaluated in cynomolgus monkeys, with a focus on the level of circulating IL-12. RESULTS Multiplication and propagation of TG6050 in tumor cells in vitro and in vivo were associated with local expression of functional IL-12 and @CTLA-4. This dual mechanism translated into a strong antitumoral activity in both "cold" and "hot" tumor models (B16F10, LLC1 or EMT6, CT26, respectively) that was further amplified when combined with anti-programmed cell death protein-1. Analysis of changes in the tumor microenvironment (TME) after treatment with TG6050 showed increases in interferon-gamma, of CD8+T cells, and of M1/M2 macrophages ratio, as well as a drastic decrease of regulatory T cells. These local modifications were observed alongside bolstering a systemic and specific antitumor adaptive immune response. In toxicology studies, TG6050 did not display any observable adverse effects in cynomolgus monkeys. CONCLUSIONS TG6050 effectively delivers functional IL-12 and @CTLA-4 into the tumor, resulting in strong antitumor activity. The shift towards an inflamed TME correlated with a boost in systemic antitumor T cells. The solid preclinical data and favorable benefit/risk ratio paved the way for the clinical evaluation of TG6050 in metastatic non-small cell lung cancer (NCT05788926 trial in progress).
Collapse
Affiliation(s)
- Fadi Azar
- Transgene SA, Illkirch-Graffenstaden, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Cao J, Lv G, Wei F. Engineering exosomes to reshape the immune microenvironment in breast cancer: Molecular insights and therapeutic opportunities. Clin Transl Med 2024; 14:e1645. [PMID: 38572668 PMCID: PMC10993163 DOI: 10.1002/ctm2.1645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/19/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Breast cancer remains a global health challenge, necessitating innovative therapeutic approaches. Immunomodulation and immunotherapy have emerged as promising strategies for breast cancer treatment. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. Through suitable modifications, engineered exosomes exhibit the capability to overcome the limitations associated with traditional therapeutic approaches. This ability opens up novel avenues for the development of more effective, personalized, and minimally invasive interventions. MAIN BODY In this comprehensive review, we explore the molecular insights and therapeutic potential of engineered exosomes in breast cancer. We discuss the strategies employed for exosome engineering and delve into their molecular mechanisms in reshaping the immune microenvironment of breast cancer. CONCLUSIONS By elucidating the contribution of engineered exosomes to breast cancer immunomodulation, this review underscores the transformative potential of this emerging field for improving breast cancer therapy. HIGHLIGHTS Surface modification of exosomes can improve the targeting specificity. The engineered exosome-loaded immunomodulatory cargo regulates the tumour immune microenvironment. Engineered exosomes are involved in the immune regulation of breast cancer.
Collapse
Affiliation(s)
- Jilong Cao
- Party Affairs and Administration Officethe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| | - Gang Lv
- Department of Thyroid and Breast SurgeryChaohu Hospital of Anhui Medical UniversityChaohuP. R. China
| | - Fang Wei
- Department of General Surgerythe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| |
Collapse
|
7
|
Umer BA, Noyce RS, Kieser Q, Favis NA, Shenouda MM, Rans KJ, Middleton J, Hitt MM, Evans DH. Oncolytic vaccinia virus immunotherapy antagonizes image-guided radiotherapy in mouse mammary tumor models. PLoS One 2024; 19:e0298437. [PMID: 38498459 PMCID: PMC10947714 DOI: 10.1371/journal.pone.0298437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/23/2024] [Indexed: 03/20/2024] Open
Abstract
Ionizing radiation (IR) and oncolytic viruses are both used to treat cancer, and the effectiveness of both agents depends upon stimulating an immune response against the tumor. In this study we tested whether combining image guided ionizing radiation (IG-IR) with an oncolytic vaccinia virus (VACV) could yield a better therapeutic response than either treatment alone. ΔF4LΔJ2R VACV grew well on irradiated human and mouse breast cancer cells, and the virus can be combined with 4 or 8 Gy of IR to kill cells in an additive or weakly synergistic manner. To test efficacy in vivo we used immune competent mice bearing orthotopic TUBO mammary tumors. IG-IR worked well with 10 Gy producing 80% complete responses, but this was halved when the tumors were treated with VACV starting 2 days after IG-IR. VACV monotherapy was ineffective in this model. The antagonism was time dependent as waiting for 21 days after IG-IR eliminated the inhibitory effect but without yielding any further benefits over IR alone. In irradiated tumors, VACV replication was also lower, suggesting that irradiation created an environment that did not support infection as well in vivo as in vitro. A study of how four different treatment regimens affected the immune composition of the tumor microenvironment showed that treating irradiated tumors with VACV altered the immunological profiles in tumors exposed to IR or VACV alone. We detected more PD-1 and PD-L1 expression in tumors exposed to IR+VACV but adding an αPD-1 antibody to the protocol did not change the way VACV interferes with IG-IR therapy. VACV encodes many immunosuppressive gene products that may interfere with the ability of radiotherapy to induce an effective anti-tumor immune response through the release of danger-associated molecular patterns. These data suggest that infecting irradiated tumors with VACV, too soon after exposure, may interfere in the innate and linked adaptive immune responses that are triggered by radiotherapy to achieve a beneficial impact.
Collapse
Affiliation(s)
- Brittany A. Umer
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Ryan S. Noyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute for Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Quinten Kieser
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Nicole A. Favis
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Mira M. Shenouda
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Kim J. Rans
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Jackie Middleton
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Mary M. Hitt
- Li Ka Shing Institute for Virology, University of Alberta, Edmonton, Alberta, Canada
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - David H. Evans
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute for Virology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Yang S, Wang Y, Yu F, Cheng R, Zhang Y, Zhou D, Ren X, Deng Z, Zhao H. Structural and functional insights into the modulation of T cell costimulation by monkeypox virus protein M2. Nat Commun 2023; 14:5186. [PMID: 37626059 PMCID: PMC10457294 DOI: 10.1038/s41467-023-40748-2] [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: 02/09/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
The rapid spread of monkeypox in multiple countries has resulted in a global public health threat and has caused international concerns since May 2022. Poxvirus encoded M2 protein is a member of the poxvirus immune evasion family and plays roles in host immunomodulation via the regulation of innate immune response mediated by the NF-κB pathway and adaptive immune response mediated by B7 ligands. However, the interaction of monkeypox virus (MPXV) M2 with B7 ligands and structural insight into poxviral M2 function have remained elusive. Here we reveal that MPXV M2, co-existing as a hexamer and a heptamer, recognizes human B7.1 and B7.2 (hB7.1/2) with high avidities. The binding of oligomeric MPXV M2 interrupts the interactions of hB7.1/2 with CD28 and CTLA4 and subverts T cell activation mediated by B7.1/2 costimulatory signals. Cryo-EM structures of M2 in complex with hB7.1/2 show that M2 binds to the shallow concave face of hB7.1/2 and displays sterically competition with CD28 and CTLA4 for the binding to hB7.1/2. Our findings provide structural mechanisms of poxviral M2 function and immune evasion deployed by poxviruses.
Collapse
Affiliation(s)
- Shangyu Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yong Wang
- Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feiyang Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Rao Cheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yiwei Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Dan Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xuanxiu Ren
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zengqin Deng
- Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China.
| | - Haiyan Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
9
|
Ling Q, Zheng B, Chen X, Ye S, Cheng Q. The employment of vaccinia virus for colorectal cancer treatment: A review of preclinical and clinical studies. Hum Vaccin Immunother 2022; 18:2143698. [PMID: 36369829 DOI: 10.1080/21645515.2022.2143698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading malignancies that causes death worldwide. Cancer vaccines and oncolytic immunotherapy bring new hope for patients with advanced CRC. The capability of vaccinia virus (VV) in carrying foreign genes as antigens or immunostimulatory factors has been demonstrated in animal models. VV of Wyeth, Western Reserve, Lister, Tian Tan, and Copenhagen strains have been engineered for the induction of antitumor response in multiple cancers. This paper summarized the preclinical and clinical application and development of VV serving as cancer vaccines and oncolytic vectors in CRC treatment. Additionally, the remaining challenges and future direction are also discussed.
Collapse
Affiliation(s)
- Qiaoyun Ling
- Department of Anorectal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Bichun Zheng
- Department of Anorectal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Xudong Chen
- Department of Anorectal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Shaoshun Ye
- Department of Anorectal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Quan Cheng
- Department of Anorectal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
10
|
Fang J, Chen R, Liu D. Talaromyces marneffei Can Capture CD86 Proteins of Macrophages in vitro. Infect Drug Resist 2022; 15:6801-6810. [DOI: 10.2147/idr.s389612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/12/2022] [Indexed: 11/26/2022] Open
|
11
|
Depierreux DM, Altenburg AF, Soday L, Fletcher-Etherington A, Antrobus R, Ferguson BJ, Weekes MP, Smith GL. Selective modulation of cell surface proteins during vaccinia infection: A resource for identifying viral immune evasion strategies. PLoS Pathog 2022; 18:e1010612. [PMID: 35727847 PMCID: PMC9307158 DOI: 10.1371/journal.ppat.1010612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/22/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy. Vaccinia virus (VACV) is the vaccine used to eradicate smallpox and an excellent model for studying host-pathogen interactions. Many VACV-mediated immune evasion strategies are known, however how immune cells recognise VACV-infected cells is incompletely understood because of the complexity of surface proteins regulating such interactions. Here, a systematic study of proteins on the cell surface at different times during infection with VACV is presented. This shows not only the precise nature and kinetics of appearance of VACV proteins, but also the selective alteration of cellular surface proteins. The latter thereby identified potential novel immune evasion strategies and host proteins regulating immune activation. Comprehensive comparisons with published datasets provided further insight into mechanisms used to regulate surface protein expression. Such comparisons also identified proteins that are targeted by both VACV and human cytomegalovirus (HCMV), and which are therefore likely to represent host proteins regulating immune recognition and activation. Collectively, this work provides a valuable resource for studying viral immune evasion mechanisms and novel host proteins critical in host immunity.
Collapse
Affiliation(s)
| | | | - Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| |
Collapse
|
12
|
Lin LCW, Croft SN, Croft NP, Wong YC, Smith SA, Tang SS, Purcell AW, Tscharke DC. Direct Priming of CD8 + T Cells Persists in the Face of Cowpox Virus Inhibitors of Antigen Presentation. J Virol 2021; 95:JVI.00186-21. [PMID: 33692206 PMCID: PMC8139650 DOI: 10.1128/jvi.00186-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/27/2021] [Indexed: 11/30/2022] Open
Abstract
Vaccinia virus (VACV) was the vaccine used to eradicate smallpox and is being repurposed as a vaccine vector. CD8+ T cells are key anti-viral mediators, but require priming to become effector or memory cells. Priming requires an interaction with dendritic cells that are either infected (direct priming), or that have acquired virus proteins but remain uninfected (cross priming). To investigate CD8+ T cell priming pathways for VACV, we engineered the virus to express CPXV12 and CPXV203, two inhibitors of antigen presentation encoded by cowpox virus. These intracellular proteins would be expected to block direct but not cross priming. The inhibitors had diverse impacts on the size of anti-VACV CD8+ T cell responses across epitopes and by different infection routes in mice, superficially suggesting variable use of direct and cross priming. However, when we then tested a form of antigen that requires direct priming, we found surprisingly that CD8+ T cell responses were not diminished by co-expression with CPXV12 and CPXV203. We then directly quantified the impact of CPXV12 and CPXV203 on viral antigen presentation using mass spectrometry, which revealed strong, but incomplete inhibition of antigen presentation by the CPXV proteins. Therefore, direct priming of CD8+ T cells by poxviruses is robust enough to withstand highly potent viral inhibitors of antigen presentation. This is a reminder of the limits of viral immune evasion and shows that viral inhibitors of antigen presentation cannot be assumed to dissect cleanly direct and cross priming of anti-viral CD8+ T cells.ImportanceCD8+ T cells are key to anti-viral immunity, so it is important to understand how they are activated. Many viruses have proteins that protect infected cells from T cell attack by interfering with the process that allows virus infection to be recognised by CD8+ T cells. It is thought that these proteins would also stop infected cells from activating T cells in the first place. However, we show here that this is not the case for two very powerful inhibitory proteins from cowpox virus. This demonstrates the flexibility and robustness of immune processes that turn on the immune responses required to fight infection.
Collapse
Affiliation(s)
- Leon C. W. Lin
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah N. Croft
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Nathan P. Croft
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yik Chun Wong
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Stewart A. Smith
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Swee-Seong Tang
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Anthony W. Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - David C. Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
13
|
Li X, Zheng J, Chen S, Meng FD, Ning J, Sun SL. Oleandrin, a cardiac glycoside, induces immunogenic cell death via the PERK/elF2α/ATF4/CHOP pathway in breast cancer. Cell Death Dis 2021; 12:314. [PMID: 33762577 PMCID: PMC7990929 DOI: 10.1038/s41419-021-03605-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/22/2022]
Abstract
Chemotherapeutic agents have been linked to immunogenic cell death (ICD) induction that is capable of augmenting anti-tumor immune surveillance. The cardiac glycoside oleandrin, which inhibits Na+/K+-ATPase pump (NKP), has been shown to suppress breast cancer growth via inducing apoptosis. In the present study, we showed that oleandrin treatment triggered breast cancer cell ICD by inducing calreticulin (CRT) exposure on cell surface and the release of high-mobility group protein B1 (HMGB1), heat shock protein 70/90 (HSP70/90), and adenosine triphosphate (ATP). The maturation and activation of dendritic cells (DCs) were increased by co-culturing with the oleandrin-treated cancer cells, which subsequently enhanced CD8+ T cell cytotoxicity. Murine breast cancer cell line EMT6 was engrafted into BALB/c mice, and tumor-bearing mice were administered with oleandrin intraperitoneally every day. Oleandrin inhibited tumor growth and increased tumor infiltrating lymphocytes including DCs and T cells. Furthermore, the differential mRNA expression incurred by oleandrin was investigated by mRNA sequencing and subsequently confirmed by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. Mechanistically, oleandrin induced endoplasmic reticulum (ER) stress-associated, caspase-independent ICD mainly through PERK/elF2α/ATF4/CHOP pathway. Pharmacological and genetic inhibition of protein kinase R-like ER kinase (PERK) suppressed oleandrin-triggered ICD. Taken together, our findings showed that oleandrin triggered ER stress and induced ICD-mediated immune destruction of breast cancer cells. Oleandrin combined with immune checkpoint inhibitors might improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Xiaoxi Li
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Jian Zheng
- Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shi Chen
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Fan-Dong Meng
- Molecular Oncology Laboratory of Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Jing Ning
- Department of General Medicine (VIP ward) & Department of Tumor Supportive and Palliative Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shu-Lan Sun
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| |
Collapse
|
14
|
Zhang Z, Dong L, Zhao C, Zheng P, Zhang X, Xu J. Vaccinia virus-based vector against infectious diseases and tumors. Hum Vaccin Immunother 2021; 17:1578-1585. [PMID: 33606578 DOI: 10.1080/21645515.2020.1840887] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vaccinia virus was used to prevent smallpox. After the World Health Organization declared smallpox extinct, vaccinia virus has been explored for the development of vaccines against a variety of infectious diseases. It also finds a new place in oncolytic therapy. Here we provide a brief review of the history, current status, and future prospect of vaccinia virus-based vaccine and oncolytic virus. New advancements, including a single vaccine targeting multiple viruses, strategies of arming vaccinia viruses to enhance anti-tumor activity, the promise and challenge of combining vaccinia-based virotherapy with immunotherapy, are discussed as special focus.
Collapse
Affiliation(s)
- Ziling Zhang
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lanlan Dong
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhao
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory for Infectious Disease Prevention and Control, China Centers for Disease Control and Prevention, Beijing, China
| | - Jianqing Xu
- Shanghai Public Health ClinicalCenter& Institutes of Biomedical Science, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory for Infectious Disease Prevention and Control, China Centers for Disease Control and Prevention, Beijing, China
| |
Collapse
|
15
|
Pelin A, Boulton S, Tamming LA, Bell JC, Singaravelu R. Engineering vaccinia virus as an immunotherapeutic battleship to overcome tumor heterogeneity. Expert Opin Biol Ther 2020; 20:1083-1097. [PMID: 32297534 DOI: 10.1080/14712598.2020.1757066] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Immunotherapy is a rapidly evolving area of cancer therapeutics aimed at driving a systemic immune response to fight cancer. Oncolytic viruses (OVs) are at the cutting-edge of innovation in the immunotherapy field. Successful OV platforms must be effective in reshaping the tumor microenvironment and controlling tumor burden, but also be highly specific to avoid off-target side effects. Large DNA viruses, like vaccinia virus (VACV), have a large coding capacity, enabling the encoding of multiple immunostimulatory transgenes to reshape the tumor immune microenvironment. VACV-based OVs have shown promising results in both pre-clinical and clinical studies, including safe and efficient intravenous delivery to metastatic tumors. AREA COVERED This review summarizes attenuation strategies to generate a recombinant VACV with optimal tumor selectivity and immunogenicity. In addition, we discuss immunomodulatory transgenes that have been introduced into VACV and summarize their effectiveness in controlling tumor burden. EXPERT OPINION VACV encodes several immunomodulatory genes which aid the virus in overcoming innate and adaptive immune responses. Strategic deletion of these virulence factors will enable an optimal balance between viral persistence and immunogenicity, robust tumor-specific expression of payloads and promotion of a systemic anti-cancer immune response. Rational selection of therapeutic transgenes will maximize the efficacy of OVs and their synergy in combinatorial immunotherapy schemes.
Collapse
Affiliation(s)
- Adrian Pelin
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Stephen Boulton
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Levi A Tamming
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Ragunath Singaravelu
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Liu X, Tang C, Song X, Cheng L, Liu Y, Ding F, Xia C, Xue L, Xiao J, Huang B. Clinical value of CTLA4-associated microRNAs combined with inflammatory factors in the diagnosis of non-small cell lung cancer. Ann Clin Biochem 2020; 57:151-161. [PMID: 31906699 DOI: 10.1177/0004563220901564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background The current study aimed to explore the value of cytotoxic T-lymphocyte-associated protein 4 (CTLA4)-associated microRNAs combined with inflammatory factors in the differential diagnosis of non-small cell lung cancer (NSCLC). Methods A retrospective study including 245 NSCLC patients and 245 healthy controls was conducted on a testing group. A regression formula for NSCLC prediction was established based on the testing group. Two validation groups from two centres were used to assess the novel logistic regression model including 144 NSCLC patients and 144 healthy controls recruited from the Wuchang Hospital Affiliated to Wuhan University of Science and Technology, and 128 NSCLC patients and 128 healthy controls recruited from the Zhongnan Hospital of Wuhan University. Results Predictive software and dual-luciferase reporter assays showed that miR-155-5p and miR-630 could target CTLA4 expression. The miR-155-5p and miR-630 concentrations in the NSCLC patients were significantly lower, and the neutrophil to lymphocyte ratio, hypersensitive C-reactive protein (hs-CRP), interleukin 6, cytokeratin-19-fragment (CYFRA21-1), squamous cell carcinoma antigen (SCCA) concentrations and the smoking rate were significantly higher than that in healthy controls ( P < 0.05). A logistic regression model that included smoking, neutrophil to lymphocyte ratio, hs-CRP, interleukin 6, CYFRA21-1, SCCA, miR-155-5p and miR-630 was performed. This model presented a high discriminating value (AUC: 0.830, sensitivity/specificity: 74.6%/89.7%) than any single indicator. In the validation groups, this model still showed a high discriminating value (AUC = 0.838 with the internal validation group; AUC = 0.851 with the external validation group). Conclusion The current model has potential significance for the non-invasive diagnosis for NSCLC.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Chunlian Tang
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xianda Song
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Lanfang Cheng
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Ying Liu
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Fan Ding
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Chun Xia
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Lian Xue
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Baorong Huang
- Department of Clinical Laboratory, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Cowpox virus encodes a protein that binds B7.1 and B7.2 and subverts T cell costimulation. Proc Natl Acad Sci U S A 2019; 116:21113-21119. [PMID: 31575740 DOI: 10.1073/pnas.1909414116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Costimulation is required for optimal T cell activation, yet it is unclear whether poxviruses dedicatedly subvert costimulation during infection. Here, we report that the secreted M2 protein encoded by cowpox virus (CPXV) specifically interacts with human and murine B7.1 (CD80) and B7.2 (CD86). We also show that M2 competes with CD28 and CTLA4 for binding to cell surface B7 ligands, with stronger efficacy against CD28. Functionally, recombinant M2 and culture supernatants from wild-type (WT) but not M2-deficient (∆M2) CPXV-infected cells can potently suppress B7 ligand-mediated T cell proliferation and interleukin-2 (IL-2) production. Furthermore, we observed increased antiviral CD4 and CD8 T cell responses in C57BL/6 mice challenged by ∆M2 CPXV compared with WT virus. These differences in immune responses to ∆M2 and WT CPXV were not observed in CD28-deficient mice. Taken together, our findings define a mechanism of viral sabotage of T cell activation that highlights the role of CD28 costimulation in host defense against poxvirus infections.
Collapse
|
18
|
Schönrich G, Raftery MJ. The PD-1/PD-L1 Axis and Virus Infections: A Delicate Balance. Front Cell Infect Microbiol 2019; 9:207. [PMID: 31263684 PMCID: PMC6584848 DOI: 10.3389/fcimb.2019.00207] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/27/2019] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death protein (PD-1) and its ligands play a fundamental role in the evasion of tumor cells from antitumor immunity. Less well appreciated is the fact that the PD-1/PD-L1 axis also regulates antiviral immune responses and is therefore modulated by a number of viruses. Upregulation of PD-1 and its ligands PD-L1 and PD-L2 is observed during acute virus infection and after infection with persistent viruses including important human pathogens such as human immunodeficiency virus (HIV), hepatitis C virus (HCV), and hepatitis B virus (HBV). Experimental evidence suggests that insufficient signaling through the PD-1 pathway promotes immunopathology during acute infection by exaggerating primary T cell responses. If chronic infection is established, however, high levels of PD-1 expression can have unfavorable immunological consequences. Exhaustion and suppression of antiviral immune responses can result in viral immune evasion. The role of the PD-1/PD-L1 axis during viral infections is further complicated by evidence that PD-L1 also mediates inflammatory effects in the acute phase of an immune response. In this review, we discuss the intricate interplay between viruses and the PD-1/PD-L1 axis.
Collapse
Affiliation(s)
- Günther Schönrich
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | | |
Collapse
|