Loading...

Table of Content

    30 April 2024, Volume 5 Issue 2
    Invited Review
    Applications of synthetic biology in developing microbial-vectored cancer vaccines
    Zibin TAN, Kang LIANG, Youhai CHEN
    2024, 5(2):  221-238.  doi:10.12211/2096-8280.2023-079
    Asbtract ( 331 )   HTML ( 35)   PDF (1910KB) ( 222 )  
    Figures and Tables | References | Related Articles | Metrics

    The development of cancer vaccines is confronted with significant challenges. Synthetic biology emerges as a potent tool for addressing these challenges, due to its ability to modify and engineer microbes capable of adapting to and colonizing on tumor tissues to change the immunosuppressive tumor microenvironments, augment antigen presentations, and stimulate both innate and adaptive immune responses against tumors in situ. This review comments on several pivotal applications of synthetic biology in engineering bacterial and viral vectored cancer vaccines. We start with discussion on methods to mitigate the pathogenicity of bacterial or viral vectors, including the removal, deactivation, or modification of their virulent genes. Furthermore, we address strategies for enhancing their tropism and fitness within tumor tissues, such as the alteration of their cellular entry proteins or the implementation of environmentally controlled gene expression systems. Approaches to minimize their systemic toxicity are also described. To fully harness the potential of tumor microenvironment modifications induced by microbial replication, we underscore studies employing synthetic biology methods, which involve the introduction of foreign genes into the microbial genomes, thereby enabling the production of agents like cytokines, chemokines, or monoclonal antibodies to enhance the recruitment and activation of innate and adaptive cells, promote immunogenic cell death, and augment the presentation of tumor-associated antigens. We also delve into the applications of synthetic biology for the introduction of tumor antigens to the vectors, discussing various loading methods, locations, and releasing mechanisms to generate an optimized tumor-specific immune response. At the end, we highlight substantial challenges that arise in the development of microbial vectored cancer vaccines, including safety considerations, intricate interactions between anti-vector and anti-tumor immunity, and the inherent complexity of tumor biology, and propose strategies for addressing these obstacles. In conclusion, this review emphasizes the crucial role of synthetic biology in the engineering of microbes, which is instrumental in advancing the development of cancer vaccines. {L-End}

    Progress with the application of synthetic biology in designing of cancer vaccines
    Chao FANG, Weiren HUANG
    2024, 5(2):  239-253.  doi:10.12211/2096-8280.2023-061
    Asbtract ( 256 )   HTML ( 25)   PDF (1693KB) ( 153 )  
    Figures and Tables | References | Related Articles | Metrics

    The central dogma of biology, which delineates the flow of genetic information from DNA to RNA to protein, along with the principles of cellular immunology, provides a foundational understanding for harnessing the power of synthetic biology to combat cancer. The application of synthetic biology in the design and production of novel tumor vaccines marks a pivotal advance in the field of cancer immunotherapy. This study delves into the cutting-edge development in the creation of therapeutic tumor vaccines, with a particular focus on two critical components: antigen selection and vaccine design. The request for more precise and effective tumor vaccines has garnered the attention of researchers globally. These vaccines are designed to target tumor-specific antigens or those related to tumor growth and survival pathways. Traditional approaches to antigen selection have typically involved targeting specific genes with tumors. However, the advent of high-throughput sequencing and mass spectrometry has revolutionized this process by enabling the screening of novel antigens, thereby enhancing the precision and immunogenicity of vaccines. In recent years, the landscape of tumor vaccines has been significantly broadened by the engineering of vaccines through various platforms. These include DNA-based vaccines, mRNA vaccines, viral or bacterial vector vaccines, and cell-based vaccines. These innovative approaches offer a stark contrast to traditional peptide vaccines, significantly amplifying the immune response against a variety of tumor types. The versatility of synthetic biology allows for the customization of vaccines to target a wide array of tumor antigens, thereby potentiating a more robust and targeted immune reaction. The progress made in synthetic biology is not only refining existing vaccine strategies but also accelerating the pace of experimental research in tumor vaccines. This rapid advancement holds the promise of continually improving the clinical therapeutic effects of these vaccines. As researchers continue to unravel the complexities of tumor immunology and synthetic biology techniques become more efficient, the intersection of these fields is expected to yield a new generation of tumor vaccines that are not only more effective but also safer and more accessible to patients. In conclusion, the integration of biological knowledge and technological innovation in synthetic biology is transforming the development of tumor vaccines. The focus on optimizing antigen selection and vaccine design is driving the creation of more potent and tailored immunotherapies. It is anticipated that synthetic biology will play an even greater role in enhancing the efficacy of tumor vaccines, offering cancer patients with hope in the ongoing battle against this devastating disease. {L-End}

    Strategies for the design and optimization of tumor neoantigen vaccines
    Huiyang TU, Weidong HAN, Bin ZHANG
    2024, 5(2):  254-266.  doi:10.12211/2096-8280.2023-060
    Asbtract ( 326 )   HTML ( 40)   PDF (1264KB) ( 273 )  
    References | Related Articles | Metrics

    With the research progress and clinical application of immune checkpoint inhibitors and chimeric antigen receptor T-cell therapies, immunotherapy has substantially changed the treating modalities for various tumors. Tumor neoantigen vaccines, as a promising immunotherapy method, aim to trigger a novel T cell response against neoantigens. Neoantigens, with their high specificity, can induce and expand the tumor-specific T cell receptor repertoire, which were discovered through the second-generation sequencing of DNA extracted from both the patient’s tumor and non-tumor tissue samples. The sequences and HLA types are then analyzed for alignment to pinpoint tumor-specific mutations. To validate the significance of these mutations, RNA sequencing data are integrated with the results. Subsequently, bioinformatics platforms are employed for the prediction and analysis of neoantigens encoded by mutated genes and HLA types, enabling the identification of potential immunogenic neoantigens. Finally, the immunogenicity of these neoantigens is assessed through techniques such as ELISPOT and tetramer assays. Tumor vaccines can be categorized as peptide-based, DNA-based, RNA-based, and DC-based products. Viruses, lipid nanoparticles, and nano delivery systems can activate antigen-presenting cells, enhancing their ability to recognize and present tumor-associated antigens, thus promoting the activation of CD8+ T cells. Neoantigen vaccines can be administered through various routes, including subcutaneous injection, intramuscular injection, intraperitoneal injection, intradermal injection, intravenous injection, or intralymphatic injection. Preliminary clinical studies have shown that neoantigen tumor vaccines have demonstrated evidence of strong tumor-specific immunogenicity and antitumor activity. In this review, we summarize in detail the source, prediction, and identification of tumor neoantigens, as well as the classification and immunization scheme of neoantigen vaccines. In addition, we highlight strategies for optimizing tumor neoantigen vaccines, including prediction algorithms, expressing multiple epitope structures, increasing immunogenicity, administration methods and delivery systems, and combining adjuvants and various treatments, providing new insights for the development of personalized immunotherapy. {L-End}

    New strategies for engineering influenza viruses and their applications
    Xiya GUO, Ji CHEN, Mingxin DONG
    2024, 5(2):  267-280.  doi:10.12211/2096-8280.2023-078
    Asbtract ( 39 )   HTML ( 5)   PDF (2451KB) ( 53 )  
    Figures and Tables | References | Related Articles | Metrics

    Influenza viruses are highly variable and transmissible, and their infections can cause infectious respiratory diseases, such as seasonal influenza outbreaks around the world, one of the most serious public health problems at present, which can be prevented by influenza vaccination. The genome sequences, protein structures and functions of influenza viruses, as well as their packaging mechanisms are relatively clear. they are also important models, which can be used for developing conditional control genetic elements and the construction of intelligent responsive viruses. With the development of reverse genetics and synthetic biology technology, influenza viruses that are genetically engineered can better control virus replication to improve the safety of vaccines, and induce strong immune responses in human being, which have attracted wide attention in tumor immunotherapy. Several studies using simple or modified influenza viruses for treating liver cancer, melanoma, or lung cancer have found breakthroughs. In this paper, three novel strategies for attenuating influenza viruses, namely, proteolytic targeted chimeric virus, conditionally replicating influenza-attenuated live virus and highly interferon-sensitive virus, are described. The oncolytic effects of influenza viruses encoding premature stop codon chimeric antigen peptide, influenza viruses recombining with PD-L1 or CTLA4 immune checkpoint and influenza viruses expressing GM-CSF with truncated NS1 fragment on melanoma and hepatocellular carcinoma are reviewed, respectively, which suggest that the influenza viruses can be used as a live attenuated vaccine and a potential carrier for oncolytic viruses, and future researchers can be focused on constructing influenza viruses with more innovative strategies and different viruses to build a live attenuated vaccine and oncolytic viruses, in order to obtain high safety and more clinical curative treatment, improving the life quality of the patients. {L-End}

    Advances in virus-vectored vaccines
    Busen WANG, Jinghan XU, Zhiqiang GAO, Lihua HOU
    2024, 5(2):  281-293.  doi:10.12211/2096-8280.2023-063
    Asbtract ( 45 )   HTML ( 6)   PDF (1378KB) ( 59 )  
    Figures and Tables | References | Related Articles | Metrics

    Recent outbreaks of infectious diseases, such as the middle east respiratory syndrome, Zika infection, Ebola hemorrhagic fever, and Coronavirus disease (COVID-19) pose significant challenges on the rapid development of efficacious vaccines. Virus-vectored vaccines, as an important new vaccine, can be administrated noninvasively through aerosol inhalation or oral administration, which could stimulate humoral, cellular, and mucosal immune responses without the need for adjuvants, showing good immunogenicity and safety in clinical trials or in emergency use. With the deeper understanding of the viral genome and structural proteins, synthetic biology has enabled the design and modification of viruses to produce recombinant viral vector-based vaccines with high titer, safety, and immunogenicity, and such research has significant implications for the vaccine development. This review highlights major strategies employed in the construction of virus-vectored vaccines, including the construction method of replication-competent or replication-defective viral vectors, and the development of viral vectors commonly used in producing the recombinant vaccines. Among these viral vectors, replication-deficient adenovirus-based vectors with gene deletion in the E1 and E3 regions are most mature for use. Currently, adenoviral vectors that have been used in the approved recombinant vaccines include Ad5, Ad26 and ChAdOx1. Vesicular stomatitis virus and flavivirus with small genomes are negative-sense and positive-sense single-stranded RNA viruses, respectively, which are easy to prepare and more suitable for being used in developing recombinant vaccines with small antigen proteins. Poxviruses and herpesviruses have large genomes for high packing capacity, but they are most difficult to be modified with synthetic biology methods. Different viral vectors need to be prepared using different strategies, and consequently vaccines developed with these vectors have different immune effects. The construction strategies of different viral vector vaccines introduced in this review will provide valuable theoretical reference for the research and development of novel viral vector vaccines. In the future, virus-vectored vaccines will be iteratively developed for higher safety, stronger protection, better compliance and lower production cost. {L-End}

    Applications of vector vaccines developed through T-cell immune responses in preventing and treating human diseases
    Shasha JIANG, Chen WANG, Ran LU, Fengjun LIU, Jun LI, Bin WANG
    2024, 5(2):  294-309.  doi:10.12211/2096-8280.2023-071
    Asbtract ( 55 )   HTML ( 4)   PDF (1419KB) ( 41 )  
    Figures and Tables | References | Related Articles | Metrics

    Human diseases, especially infectious diseases and cancers, pose unprecedented challenges to public health and the global economy, making the development of preventive and therapeutic vaccines a top priority for addressing these challenges. Among all vaccines, vector vaccines that activate T cell immune responses have significant advantages. This article reviews the immunological principles of vector vaccines, strategies for designing T cell vector vaccines, and their research advances. T cells, upon infection, can differentiate into various effector T cell subsets that play a crucial role in clearing pathogens. Research on the functions and mechanisms of effector T cells is essential for designing vaccines that can elicit T cell-mediated immunity. Currently, the development of vaccines for many viruses such as HIV and HCMV as well as cancers focuses on T cell-based vaccines. Various vectors, including viral vectors, bacterial vectors, and nucleic acid vectors, exhibit excellent performance on antigen delivery capability, immunogenicity, and protective efficacy. In addition, this article summarizes strategies for designing T-cell vector vaccines, including identifying appropriate antigen presentation pathways and vector delivery routes, ensuring biological safety, selecting suitable vaccine vectors, and evaluating the advantages and disadvantages of various vector vaccines. Notably, mRNA vaccines have played a crucial role in addressing the challenges posed by the COVID-19 pandemic. Technological advancements in vector vaccines are expected to accelerate the development of novel vaccines and enhance preparedness for emerging public health events. This review provides insights for the design of vector vaccines that are both safe and efficient. With advancements in vector vaccine technology and the progress of various interdisciplinary approaches, the next generation of vaccine development will continue to drive the evolution of vaccinology. {L-End}

    Development of mRNA vaccines in response to the Public Health Emergency of International Concern
    Qing YE, Chengfeng QIN
    2024, 5(2):  310-320.  doi:10.12211/2096-8280.2023-072
    Asbtract ( 44 )   HTML ( 5)   PDF (1288KB) ( 61 )  
    Figures and Tables | References | Related Articles | Metrics

    A Public Health Emergency of International Concern (PHEIC) is defined by the World Health Organization (WHO) as “an extraordinary event which is determined to constitute a public health risk to other states through the international spread of disease and potentially requires a coordinated international response”. To date, WHO has declared seven PHEIC events, including the H1N1 influenza, Ebola, poliomyelitis, Zika, COVID-19 and mpox. Vaccination remains as an effective method in preventing infectious diseases. The International Health Regulations (IHR) Emergency Committee's recommendations for preventing or reducing the international spread of disease and avoiding unnecessary interference with international traffic include an emphases on the development of diagnostics and therapeutics for diseases, as well as the vaccine development. The mRNA vaccine represents a platform technology for the development of next-generation vaccines, and possesses distinct advantages, such as a shortened development cycle, scalable and cost-effective production, as well as enhanced amplification capacity, highlighting its potential in rapid responding to emerging and re-emerging infectious diseases. In recent decades, the development of mRNA synthesis technology and nucleic acid delivery system has facilitated the rapid development of mRNA vaccines and their clinical applications. Here, we overview the development of mRNA vaccines in response to the past PHEICs, and discuss challenges and trends in this regard. Currently, COVID-19 mRNA vaccines have been authorized for human use, while multiple mRNA vaccines against influenza, Zika, mpox and Ebola have been evaluated in clinical or pre-clinical studies. Despite their proven efficacy, there is still room for further improvement of the mRNA vaccines. The mRNA design, optimization, delivery, formulation, manufacturing, storage, and transportation can be further improved by integrating synthetic biology, biochemistry, artificial intelligence, and other multidisciplinary technologies. Although the emergence of the next PHEIC cannot be predicted with certainty, we are optimistic that the mRNA vaccine technology will play a pivotal role in preventing pandemics in the future. {L-End}

    Synthetic biology promotes the development of bacterial vaccines
    Jinyong ZHANG, Jiang GU, Shan GUAN, Haibo LI, Hao ZENG, Quanming ZOU
    2024, 5(2):  321-337.  doi:10.12211/2096-8280.2023-070
    Asbtract ( 119 )   HTML ( 3)   PDF (1459KB) ( 97 )  
    Figures and Tables | References | Related Articles | Metrics

    In recent years, bacterial infections have emerged as the second leading cause of death globally, posing a serious threat to public health and demanding prioritized intervention from the healthcare community worldwide. While antibiotics have conventionally been used as the primary strategy to combat bacterial infections, their efficacy is increasingly compromised due to the emergence of drug-resistant bacteria, especially multi-drug-resistant and even pan-drug-resistant superbacteria. Vaccines are thus considered as one of the most scientific, economical, safe, and effective means to prevent infectious diseases and improve public health, which are estimated to save 2 to 3 million lives annually, and can serve as a critical tool in the battle against antimicrobial resistance. However, the complexity of bacterial structure and pathogenic mechanism has hindered the development of vaccines. Challenges include screening and rationally design of effective antigens, ensuring compatibility of various antigen combinations, establishing animal models for preclinical evaluation, and defining reliable endpoints for clinical efficacy assessment. As a result, only a small number of bacteria vaccines have been successfully developed so far, and none of them has been licensed to combat the most prevalent drug-resistant infections, such as Staphylococcus aureus, Acinetobacter baumannii, Pseudomonas aeruginosa and Klebsiella pneumoniae. Synthetic biology is a brand-new multidisciplinary focusing on repurposing natural biological systems and inventing innovative biological tools, technologies, devices, and systems for practical applications, and its concepts, principles and technologies have been extensively employed to facilitate vaccine development, including rational design, screening, and optimization of antigen, carrier, adjuvant and delivery system as well as the modulation of bacterial pathogenicity and immune responses. Herein, we outline the current status of the development of bacterial vaccines and the advancement of clinical trials for drug-resistant bacterial vaccines. Then, we summarize the application of synthetic biology technology in the development of major bacterial vaccines. Finally, we prospect the potential of synthetic biology in creating novel bacterial vaccines. Researchers have access to a greater variety of design possibilities for bacterial vaccines through synthetic biology. To maximize these benefits, we should employ synthetic biology and related technologies more efficiently in developing bacterial vaccines. Meanwhile, we should develop a scientific, reasonable, effective, and feasible management system, as well as regulatory measures, to expedite the development of efficient bacterial vaccines, therefore addressing the problem of antibiotic resistance to protect human health. {L-End}

    Applications of synthetic biology in developing polysaccharide conjugate vaccines
    Jingqin YE, Wenhua HUANG, Chao PAN, Li ZHU, Hengliang WANG
    2024, 5(2):  338-352.  doi:10.12211/2096-8280.2023-054
    Asbtract ( 505 )   HTML ( 53)   PDF (1354KB) ( 318 )  
    Figures and Tables | References | Related Articles | Metrics

    The precise design and synthesis of carbohydrates with important biological functions and more complex structures is a frontier in synthetic biology. Recently, a novel strategy named Protein Glycan Coupling Technology (PGCT) based on bacterial oligosaccharyltransferases has been developed and widely used in the biosynthesis of bacterial glycoconjugate vaccines, which are one of achievements in modern medicine due to their effectiveness in fighting against infectious diseases. Herein, progress in developing key components for manufacturing glycoconjugate vaccines, such as oligosaccharyltransferases (PglL, PglS, PglB, and TfmP), carrier proteins (CRM197, diphtheria toxoid, recombinant Pseudomonas aeruginosa exotoxin A, and nanoparticles), polysaccharide biosynthesis gene circuits, and glyco-engineered strains is reviewed. Meanwhile, producing glycoconjugate vaccines through fermentation presents advantages in good product quality control for safety and efficacy, low production cost, and environmental-friendly manufacturing. PGCT has potentials to overcome some limitations of chemical conjugation production processes, such as complex purification and high cost, for competitiveness with existing chemical conjugates. As an emerging technology, more technological innovations are needed for PGCT. In the future, the directed evolution of oligosaccharyltransferases, the application of protein nanoparticle carriers, the combination rearrangement of glycosyltransferases, and the optimization of engineered bacterial strains with better metabolic pathways are expected to further promote the biosynthesis of conjugate vaccines. The next few years will be an important and exciting time for PGCT, as recent technological advances are being applied to the development of novel glycoconjugates, and ongoing large-scale clinic trials on the efficacy of glycoconjugate vaccines will also demonstrate the feasibility of this technology, making the future of PGCT vaccinology promising. {L-End}

    Dawn of the rational design of nanoparticle vaccines aided by the advance of synthetic biology techniques
    Xuejing MA, Chang GUO, Zhaolin HUA, Baidong HOU
    2024, 5(2):  353-368.  doi:10.12211/2096-8280.2023-055
    Asbtract ( 343 )   HTML ( 29)   PDF (2047KB) ( 257 )  
    Figures and Tables | References | Related Articles | Metrics

    Nanoparticle vaccines have been established firmly as a cornerstone of modern immunization strategies, with a compelling history that trace their pioneering use in human being back to 1981. Within the past four decades, these vaccines have not only demonstrated their efficacy, but have also been developed as powerful tools in fighting against a range of infectious diseases, most notably hepatitis B virus (HBV) and human papillomavirus (HPV). Their success can be attributed to their exceptional immunogenicity and impeccable safety as well, making them invaluable in curbing the spread of viruses and safeguarding the health and well-being of human being. The global outbreaks of the COVID-19 pandemic, driven by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has made vaccination into the forefront of public health priorities. This unprecedent challenge has accelerated the progress of various vaccine technologies, with nanoparticle vaccines attracting considerable attention. However, due to their relatively empirical design approaches and complicated manufacturing processes, progress in the clinical trials of SARS-CoV-2 nanoparticle vaccines has not been highlighted particularly. Therefore, the imperative for developing nanoparticle vaccines is to figure out their rational design, requiring groundbreaking advancement in novel technologies and theories. In this endeavor, synthetic biotechnology has emerged as an indispensable tool, driving the technological innovations of the production of nanoparticle vaccines. This article begins with an overview of technological advancements in the development of nanoparticle vaccines, encompassing progress from self-assembled nanoparticles to assist-assembled nanoparticles, and ultimately to antigen-display on formed nanoparticles. Furthermore, discoveries in understanding the unique roles of nanoparticle vaccines in enhancing antigen immunogenicity are updated, particularly in the function of nanoparticles with novel antigen presentation pathways. Finally, a comprehensive summary of the clinical trials of nanoparticle vaccines on fighting the COVID-19 pandemic is presented. In conclusion, we firmly believe that nanoparticle vaccines, bolstered by the scaffolding of synthetic biotechnology, are poised to emerge as steadfast guardians in the global battle against emerging and highly infectious diseases, and ongoing progress in this regard not only holds great promise, but also has potentials to revolutionize contagious disease prevention and control on a global scale. {L-End}

    Applications of synthetic biology in the development of SARS-CoV-2 broad-spectrum vaccines
    Weifeng YUAN, Yongliang ZHAO, Zhixuan WU, Ke XU
    2024, 5(2):  369-384.  doi:10.12211/2096-8280.2023-088
    Asbtract ( 67 )   HTML ( 6)   PDF (1944KB) ( 32 )  
    Figures and Tables | References | Related Articles | Metrics

    Since the outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at the end of 2019, it has evolved into different lineages, including Alpha, Beta, Delta, and Omicron. The development of broad-spectrum vaccines has become a necessity for preventing the highly mutated respiratory viruses. Traditional vaccine antigens, originating from prototype strains, cannot cover rapid mutations with these viruses, leading to breakthrough infections. With the development of synthetic biology, new technologies such as multivalent coupling of antigens, reconstructed dominate antigen modules, engineering design of conserved epitopes, epitope display, and computation-guided reconstruction have enabled redesigning antigens to achieve stronger immunogenicity with broader spectrum. The technology of synthetic biology is also applicable in the vaccine production process, such as antigen expression in nanoparticles, viral vectors, nucleic acids, and subunits. This article reviews the applications of synthetic biology technology in developing broad-spectrum vaccines in recent years, particularly for the broad-spectrum SARS-CoV-2 vaccines, and summarizes how to display common antigens and cross-antigenic sites by the reverse vaccinology for the activation of broad-spectrum immune responses against different mutant strains, achieving broad-spectrum vaccine protection effects through “remaining constant in response to ever-changing”. The article also provides a comprehensive comparison of the strengths and limitations of different broad-spectrum vaccine design strategies and discusses challenges to applying synthetic biology in the development of vaccines, offering valuable insights for universal against highly mutation viruses. {L-End}

    Research Article
    Applications of the recombinant human collagen type Ⅲ-based trimerization motif in the design of vaccines to fight against SARS-CoV-2 and influenza virus
    Zezhong LIU, Jie ZHOU, Yun ZHU, Lu LU, Shibo JIANG
    2024, 5(2):  385-395.  doi:10.12211/2096-8280.2023-058
    Asbtract ( 384 )   HTML ( 27)   PDF (2261KB) ( 263 )  
    Figures and Tables | References | Related Articles | Metrics

    Glycoproteins with enveloped viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus, and human immunodeficiency virus (HIV), display a trimeric conformation. Different from the monomeric form, the trimeric proteins exhibit superior immunogenicity. Several trimerization motifs, such as Foldon derived from phage T4 fibritin, have been used to promote the formation of trimeric proteins with natural conformations. Although the Foldon-induced trimeric proteins are stable, their high immunogenicity limits applications in the development of vaccine antigens. In a previous study, we developed a recombinant human collagen type Ⅲ protein and determined its crystal structure, revealing a triple-helix conformation. However, the potential of this recombinant protein as a trimerization motif remained unknown. In this study, we demonstrated that the recombinant humanized type Ⅲ collagen (Rh3C) was able to act as a trimerization motif, facilitating the spontaneous trimer formation of the Rh3C-conjugated receptor-binding domain (RBD) within the spike (S) protein of SARS-CoV-2. This trimeric protein could induce a stronger SARS-CoV-2 RBD-specific IgG, IgG1, and IgG2a immune response, when compared with the monomeric RBD protein in the immunized mice. Notably, the Rh3C-RBD protein, when adjuvanted with the novel STING agonist CF501, also elicited significantly higher neutralizing antibody responses against both the pseudotyped SARS-CoV-2 (D614G) and its variant Omicron (BA.2.2) in the immunized mice. To showcase the broad applications of the Rh3C trimerization motif, we further demonstrated that the Rh3C-conjugated HA1 of the influenza virus could also elicit a stronger antibody response than free HA1. Considering the wide distribution of the Rh3C protein in human bodies, its use as a trimerization motif would not induce an immune response due to immune tolerance, thereby allowing the immune response to concentrate on targeted viral proteins. Therefore, this Rh3C-based trimerization motif holds great potential for the design and optimization of vaccines consisting of trimeric protein antigens. {L-End}