Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters











Publication year range
1.
Biomaterials ; 313: 122788, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39236628

ABSTRACT

Drug resistance is a significant challenge in cancer chemotherapy and is a primary factor contributing to poor recovery for cancer patients. Although drug-loaded nanoparticles have shown promise in overcoming chemotherapy resistance, they often carry a combination of drugs and require advanced design and manufacturing processes. Furthermore, they seldom approach chemotherapy-resistant tumors from an immunotherapy perspective. In this study, we developed a therapeutic nanovaccine composed solely of chemotherapy-induced resistant tumor antigens (CIRTAs) and the immune adjuvant Toll-like receptor (TLR) 7/8 agonist R848 (CIRTAs@R848). This nanovaccine does not require additional carriers and has a simple production process. It efficiently delivers antigens and immune stimulants to dendritic cells (DCs) simultaneously, promoting DCs maturation. CIRTAs@R848 demonstrated significant tumor suppression, particularly when used in combination with the immune checkpoint blockade (ICB) anti-PD-1 (αPD-1). The combined therapy increased the infiltration of T cells into the tumor while decreasing the proportion of regulatory T cells (Tregs) and modulating the tumor microenvironment, resulting in long-term immune memory. Overall, this study introduces an innovative strategy for treating chemotherapy-resistant tumors from a novel perspective, with potential applications in personalized immunotherapy and precision medicine.


Subject(s)
Cancer Vaccines , Deoxycytidine , Drug Resistance, Neoplasm , Gemcitabine , Immunotherapy , Nanoparticles , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/pharmacology , Animals , Immunotherapy/methods , Drug Resistance, Neoplasm/drug effects , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Nanoparticles/chemistry , Mice , Humans , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Female , Imidazoles/pharmacology , Imidazoles/therapeutic use , Tumor Microenvironment/drug effects , Antigens, Neoplasm/immunology , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , Nanovaccines
2.
Biomater Sci ; 12(19): 5115-5122, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39225616

ABSTRACT

Virus-like nanoparticle vaccines can efficiently activate the humoral immune response by cross-linking B cell receptors with their surface multivalent antigen arrays. This structurally dependent mechanism makes it crucial to regulate and optimize structural parameters to enhance the efficacy of nanoparticle vaccines. In this study, we prepared nanoparticle vaccines with different aspect ratios by chemically modifying antigen proteins onto the surfaces of poly(amino acid) nanoparticles of various shapes (spherical, ellipsoidal, and rod-like). This allowed us to investigate the impact of structural anisotropy on the humoral immune activation efficacy of nanoparticle vaccines. Furthermore, the end-group molecules of poly(amino acid) materials possess aggregation-induced emission (AIE) properties, which facilitate monitoring the dynamics of nano-assemblies within the body. Results showed that rod-like nanoparticle vaccines (RLNVax) with a higher aspect ratio (AR = 5) exhibited greater lymph node draining efficiency and could elicit more effective B cell activation compared to conventional isotropic spherical nanoparticle vaccines. In a murine subcutaneous immunization model using ovalbumin (OVA) as a model antigen, RLNVax elicited antigen-specific antibody titers that were about 64 times and 4.6 times higher than those induced by free antigen proteins and spherical nanoparticle vaccines, respectively. Additionally, when combined with an aluminum adjuvant, antibody titers elicited by RLNVax were further enhanced by 4-fold. These findings indicate that the anisotropic rod-like structure is advantageous for improving the humoral immune activation efficacy of nanoparticle vaccines, providing significant insights for the design and optimization of next-generation nanoparticle vaccines.


Subject(s)
Immunity, Humoral , Nanoparticles , Animals , Immunity, Humoral/drug effects , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Female , Ovalbumin/immunology , Ovalbumin/chemistry , Ovalbumin/administration & dosage , Mice, Inbred BALB C , B-Lymphocytes/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/chemistry , Vaccines, Virus-Like Particle/administration & dosage , Nanovaccines
3.
Nat Commun ; 15(1): 8121, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284814

ABSTRACT

Inducing high levels of antigen-specific CD8α+ T cells in the tumor is beneficial for cancer immunotherapy, but achieving this in a safe and effective manner remains challenging. Here, we have developed a designer liposomal nanovaccine containing a sonosensitizer (LNVS) to efficiently program T cell immunity in mice. Following intravenous injection, LNVS accumulates in the spleen in a protein corona and fluidity-dependent manner, leading to greater frequencies of antigen-specific CD8α+ T cells than soluble vaccines (the mixture of antigens and adjuvants). Meanwhile, some LNVS passively accumulates in the tumor, where it responds to ultrasound (US) to increase the levels of chemokines and adhesion molecules that are beneficial for recruiting CD8α+ T cells to the tumor. LNVS + US induces higher levels of intratumoral antitumor T cells than traditional sonodynamic therapy, regresses established mouse MC38 tumors and orthotopic cervical cancer, and protects cured mice from relapse. Our platform sheds light on the importance of tuning the fluidity and protein corona of naovaccines to program T cell immunity in mice and may inspire new strategies for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Cancer Vaccines , Immunotherapy , Liposomes , Mice, Inbred C57BL , Animals , Liposomes/chemistry , Mice , Female , Immunotherapy/methods , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/therapy , Humans , Nanovaccines
4.
Int J Nanomedicine ; 19: 9437-9458, 2024.
Article in English | MEDLINE | ID: mdl-39290859

ABSTRACT

Background: Tumor vaccines have achieved remarkable progress in treating patients with various tumors in clinical studies. Nevertheless, extensive research has also revealed that tumor vaccines are not up to expectations for the treatment of solid tumors due to their low immunogenicity. Therefore, there is an urgent need to design a tumor vaccine that can stimulate a broad anti-tumor immune response. Methods: In this work, we developed a nanovaccine (NP-TCL@APS), which includes nanoparticles loaded with colorectal cancer tumor cell lysates (TCL) and Astragalus polysaccharides (APS) into poly (lactic-co-glycolic acid) to induce a robust innate immune response. The NP-TCL@APS was identified by transmission electron microscopy and Malvern laser particle size analyzer. The killing and immune activation effects of NP-TCL@APS were evaluated in vitro. Finally, safety and anti-tumor efficacy were evaluated in the colorectal cancer tumor-bearing mouse model. Results: We found that NP-TCL@APS was preferentially uptaken by DC and further promoted the activation of DC in vitro. Additionally, nanoparticles codelivery of TCL and APS enhanced the antigen-specific CD8+ T cell response and suppressed the growth of tumors in mouse models with good biocompatibility. Conclusion: We successfully prepared a nanovaccine termed NP-TCL@APS, which can promote the maturation of DC and induce strong responses by T lymphocytes to exert anti-tumor effects. The strategy proposed here is promising for generating a tumor vaccine and can be extended to various types of cancers.


Subject(s)
Cancer Vaccines , Colorectal Neoplasms , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Polysaccharides , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/drug therapy , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polysaccharides/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Humans , Mice , Nanoparticles/chemistry , Cell Line, Tumor , Astragalus Plant/chemistry , Mice, Inbred BALB C , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Female , Nanovaccines
5.
J Control Release ; 374: 325-336, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39154934

ABSTRACT

mRNA-based vaccines symbolize a new paradigm shift in personalized medicine for the treatment of infectious and non-infectious diseases. However, the reactogenicity associated with the currently approved formulations limits their applicability in autoinflammatory disorders, such as tumour therapeutics. In this study, we present a delivery system showing controlled immunogenicity and minimal non-specific inflammation, allowing for selective delivery of mRNA to antigen presenting cells (APCs) within the medullary region of the lymph nodes. Our platform offers precise control over the trafficking of nanoparticles within the lymph nodes by optimizing stealth and targeting properties, as well as the subsequent opsonization process. By targeting specific cells, we observed a potent adaptive and humoral immune response, which holds promise for preventive and therapeutic anti-tumoral vaccines. Through spatial programming of nanoparticle distribution, we can promote robust immunization, thus improving and expanding the utilization of mRNA vaccines. This innovative approach signifies a remarkable step forward in the field of targeted nanomedicine.


Subject(s)
Lymph Nodes , Mice, Inbred C57BL , Nanoparticles , RNA, Messenger , Lymph Nodes/immunology , Animals , RNA, Messenger/administration & dosage , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Female , mRNA Vaccines , Antigen-Presenting Cells/immunology , Humans , Mice , Vaccines/administration & dosage , Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Nanovaccines
6.
J Nanobiotechnology ; 22(1): 513, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192264

ABSTRACT

The application of nanoscale scaffolds has become a promising strategy in vaccine design, with protein-based nanoparticles offering desirable avenues for the biocompatible and efficient delivery of antigens. Here, we presented a novel endogenous capsid-forming protein, activated-regulated cytoskeleton-associated protein (ARC), which could be engineered through the plug-and-play strategy (SpyCatcher3/SpyTag3) for multivalent display of antigens. Combined with the self-assembly capacity and flexible modularity of ARC, ARC-based vaccines elicited robust immune responses against Mpox or SARS-CoV-2, comparable to those induced by ferritin-based vaccines. Additionally, ARC-based nanoparticles functioned as immunostimulants, efficiently stimulating dendritic cells and facilitating germinal center responses. Even without adjuvants, ARC-based vaccines generated protective immune responses in a lethal challenge model. Hence, this study showed the feasibility of ARC as a novel protein-based nanocarrier for multivalent surface display of pathogenic antigens and demonstrated the potential of exploiting recombinant mammalian retrovirus-like protein as a delivery vehicle for bioactive molecules.


Subject(s)
COVID-19 Vaccines , COVID-19 , Nanoparticles , SARS-CoV-2 , Animals , Nanoparticles/chemistry , Mice , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , COVID-19 Vaccines/chemistry , Humans , Mice, Inbred BALB C , Capsid Proteins/chemistry , Capsid Proteins/immunology , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Female , Dendritic Cells/immunology , Nanovaccines , Nerve Tissue Proteins
7.
Cell Mol Life Sci ; 81(1): 372, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39196331

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a global health problem that substantially increases the risk of developing liver disease. The development of a novel strategy to induce anti-HB seroconversion and achieve a long-lasting immune response against chronic HBV infection remains challenging. Here, we found that chronic HBV infection affected the signaling pathway involved in STING-mediated induction of host immune responses in dendritic cells (DCs) and then generated a lymph node-targeted nanovaccine that co-delivered hepatitis B surface antigen (HBsAg) and cyclic diguanylate monophosphate (c-di-GMP) (named the PP-SG nanovaccine). The feasibility and efficiency of the PP-SG nanovaccine for CHB treatment were evaluated in HBV-carrier mice. Serum samples were analyzed for HBsAg, anti-HBs, HBV DNA, and alanine aminotransferase levels, and liver samples were evaluated for HBV DNA and RNA and HBcAg, accompanied by an analysis of HBV-specific cellular and humoral immune responses during PP-SG nanovaccine treatment. The PP-SG nanovaccine increased antigen phagocytosis and DC maturation, efficiently and safely eliminated HBV, achieved a long-lasting immune response against HBV reinjection, and disrupted chronic HBV infection-induced immune tolerance, as characterized by the generation and multifunctionality of HBV-specific CD8+ T and CD4+ T cells and the downregulation of immune checkpoint molecules. HBV-carrier mice immunized with the PP-SG nanovaccine achieved partial anti-HBs seroconversion. The PP-SG nanovaccine can induce sufficient and persistent viral suppression and achieve anti-HBs seroconversion, rendering it a promising vaccine candidate for clinical chronic hepatitis B therapy.


Subject(s)
Dendritic Cells , Hepatitis B Surface Antigens , Hepatitis B virus , Hepatitis B, Chronic , Lymph Nodes , Membrane Proteins , Mice, Inbred C57BL , Animals , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/drug therapy , Mice , Dendritic Cells/immunology , Hepatitis B virus/immunology , Hepatitis B Surface Antigens/immunology , Lymph Nodes/immunology , Lymph Nodes/drug effects , Membrane Proteins/immunology , Hepatitis B Vaccines/immunology , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Female , Humans , Nanoparticles/chemistry , Nanovaccines
8.
J Nanobiotechnology ; 22(1): 483, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138475

ABSTRACT

The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body's immune potential, providing an eutherapeutic strategy for the treatment of OC.


Subject(s)
Antigens, Neoplasm , B7-H1 Antigen , Cancer Vaccines , Nanoparticles , Ovarian Neoplasms , Female , Ovarian Neoplasms/drug therapy , Animals , Mice , Cancer Vaccines/therapeutic use , Nanoparticles/chemistry , Cell Line, Tumor , Antigens, Neoplasm/immunology , Humans , Dendritic Cells/drug effects , Peptides/chemistry , Peptides/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Immunotherapy/methods , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Immune Checkpoint Inhibitors/pharmacology , Nanovaccines
9.
Nano Lett ; 24(33): 10114-10123, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39109634

ABSTRACT

Personalized cancer vaccines targeting specific neoantigens have been envisioned as one of the most promising approaches in cancer immunotherapy. However, the physicochemical variability of the identified neoantigens limits their efficacy as well as vaccine manufacturing in a uniform format. Herein, we developed a uniform nanovaccine platform based on poly(2-oxazoline)s (POx) to chemically conjugate neoantigen peptides, regardless of their physicochemical properties. This vaccine system could self-assemble into nanoparticles with uniform size (around 50 nm) and improve antigen accumulation as well as infiltration in the lymph node to increase antigen presentation. In vivo vaccination using this system conjugated with three predicted peptide neoantigen peptides from the MC38 tumor cell line induced 100% robust CD8+ T cell responses and superior tumor clearance compared to free peptides. This POx-based vaccine carrier represents a generalizable approach to increase the availability and efficacy of screened neoantigen peptides for a personalized cancer vaccine.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Nanoparticles , Peptides , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Peptides/chemistry , Peptides/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Mice , Nanoparticles/chemistry , Humans , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , Oxazoles/chemistry , Polymers/chemistry , Immunotherapy/methods , Nanovaccines
10.
Front Immunol ; 15: 1423212, 2024.
Article in English | MEDLINE | ID: mdl-39136021

ABSTRACT

Background: Nanovaccine treatment is an exciting area of research in immunology and personalized medicine, holding great promise for enhancing immune responses and targeting specific diseases. Their small size allows efficient uptake by immune cells, leading to robust immune activation. They can incorporate immune-stimulating molecules to boost vaccine efficacy. Therefore, nanovaccine can be personalized to target tumor-specific antigens, activating the immune system against cancer cells. Currently, there have been ample evidence showing the effectiveness and potential of nanovaccine as a treatment for cancer. However, there was rare bibliometric analysis of nanovaccine for cancer. Here we performed a bibliometric and visual analysis of published studies related to nanovaccine treatment for cancer, providing the trend of future development of nanovaccine. Methods: We collected the literatures based on the Web of Science Core Collection SCI-Expanded database. The bibliometric analysis was performed via utilizing visualization analysis tools VOSviewer, Co-Occurrence (COOC), Citespace, Bibliometrix (R-Tool of R-Studio), and HitCite. Results: A total of 517 literatures were included in this study. China is the country with the most publications and the highest total local citation score (TLCS). The Chinese Academy of Sciences holds the largest research count in this field and the most prolific author is Deling Kong from Nankai University. The most prominent journal for publishing in this area is Biomaterials. The researches mainly focus on the therapeutic process of tumor nanovaccines, the particle composition and the application of nanovaccines, suggesting the potential hotspots and trends of nanovaccine. Conclusion: In this study, we summarized the characteristics and variation trends of publications involved in nanovaccine, and categorized the most influential countries, institutions, authors, journals, hotspots and trends regarding the nanovaccine for cancer. With the continuous development of nanomaterials and tumor immunotherapy, nanovaccine for cancer provides a research field of significant clinical value and potential application.


Subject(s)
Bibliometrics , Cancer Vaccines , Neoplasms , Humans , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Neoplasms/therapy , Neoplasms/immunology , Nanoparticles , Animals , Nanovaccines
11.
Drug Dev Res ; 85(5): e22244, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39138855

ABSTRACT

Nanovaccines have been designed to overcome the limitations associated with conventional vaccines. Effective delivery methods such as engineered carriers or smart nanoparticles (NPs) are critical requisites for inducing self-tolerance and optimizing vaccine immunogenicity with minimum side effects. NPs can be used as adjuvants, immunogens, or nanocarriers to develop nanovaccines for efficient antigen delivery. Multiloaded nanovaccines carrying multiple tumor antigens along with immunostimulants can effectively increase immunity against tumor cells. They can be biologically engineered to boost interactions with dendritic cells and to allow a gradual and constant antigen release. Modifying NPs surface properties, using high-density lipoprotein-mimicking nanodiscs, and developing nano-based artificial antigen-presenting cells such as dendritic cell-derived-exosomes are amongst the new developed technologies to enhance antigen-presentation and immune reactions against tumor cells. The present review provides an overview on the different perspectives, improvements, and barriers of successful clinical application of current cancer therapeutic and vaccination options. The immunomodulatory effects of different types of nanovaccines and the nanoparticles incorporated into their structure are described. The advantages of using nanovaccines to prevent and treat common illnesses such as AIDS, malaria, cancer and tuberculosis are discussed. Further, potential paths to develop optimal cancer vaccines are described. Given the immunosuppressive characteristics of both cancer cells and the tumor microenvironment, applying immunomodulators and immune checkpoint inhibitors in combination with other conventional anticancer therapies are necessary to boost the effectiveness of the immune response.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Immunotherapy , Nanoparticles , Neoplasms , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Nanoparticles/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/administration & dosage , Animals , Combined Modality Therapy , Drug Delivery Systems/methods , Nanovaccines
12.
ACS Nano ; 18(37): 25826-25840, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39196858

ABSTRACT

Engineering nanovaccines capable of targeting dendritic cells (DCs) is desperately required to maximize antigen cross-presentation to effector immune cells, elicit strong immune responses, and avoid adverse reactions. Here, we showed that glucose transporter 1 (Glut-1) on DCs is a reliable target for delivering antigens to DCs, and thus, a versatile antigen delivery strategy using glucosylated nanovaccines was developed for DC-targeted antigen delivery and tumor immunotherapy. The developed glucosylated ovalbumin-loaded nanovaccines highly accumulated in lymph nodes and efficiently engaged with Glut-1 on DCs to accelerate intracellular antigen delivery and promote DC maturation and antigen presentation, which elicited potent antitumor immunity to prevent and inhibit ovalbumin-expressing melanoma. Moreover, immunotherapeutic experiments in DC- and macrophage-depleted animal models confirmed that the glucosylated nanovaccines functioned mainly through DCs. In addition, the neoantigen-delivering glucosylated nanovaccines were further engineered to elicit tumor-specific immune responses against MC38 tumors. This study offers a DC-targeted antigen delivery strategy for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Immunotherapy , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/administration & dosage , Mice , Ovalbumin/immunology , Ovalbumin/chemistry , Nanoparticles/chemistry , Antigens, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Female , Antigen Presentation/immunology , Cell Line, Tumor , Humans , Nanovaccines
13.
ACS Nano ; 18(35): 24219-24235, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39172516

ABSTRACT

Fibrosarcoma, a malignant mesenchymal tumor, is characterized by aggressive invasiveness and a high recurrence rate, leading to poor prognosis. Anthracycline drugs, such as doxorubicin (DOX), represent the frontline chemotherapy for fibrosarcoma, but often exhibit suboptimal efficacy. Recently, exploiting the stimulator of interferon genes (STING)-mediated innate immunity has emerged as a hopeful strategy for cancer treatment. Integrating chemotherapy with immunomodulators in chemo-immunotherapy has shown potential for enhancing treatment outcomes. Herein, we introduce an advanced dendritic cell (DC) nanovaccine, cGAMP@PLGA@CRTM (GP@CRTM), combined with low-dose DOX to enhance fibrosarcoma chemo-immunotherapy. The nanovaccine consists of poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulating the STING agonist 2,3-cGAMP (cGAMP@PLGA, GP) as its core, and a calreticulin (CRT) high-expressing fibrosarcoma cell membrane (CRTM) as the shell. Exposing CRT on the vaccine surface aids in recruiting DCs and stimulating uptake, facilitating efficient simultaneous delivery of STING agonists and tumor antigens to DCs. This dual delivery method effectively activates the STING pathway in DCs, triggering sustained immune stimulation. Simultaneously, low-dose DOX reduces chemotherapy-related side effects, directly kills a subset of tumor cells, and increases tumor immunogenicity, thus further amplifying immune therapeutic performance. Hence, these findings demonstrate the potential of DC nanovaccine GP@CRTM as a booster for chemotherapy. Synergistically combining low-dose DOX with the DC nanovaccine emerges as a powerful chemo-immunotherapy strategy, optimizing systemic fibrosarcoma therapy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Doxorubicin , Fibrosarcoma , Nanoparticles , Nucleotides, Cyclic , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fibrosarcoma/immunology , Fibrosarcoma/therapy , Animals , Doxorubicin/pharmacology , Doxorubicin/chemistry , Mice , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/pharmacology , Nanoparticles/chemistry , Cancer Vaccines/immunology , Humans , Membrane Proteins/metabolism , Cell Line, Tumor , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Inbred C57BL , Immunotherapy , Calreticulin/metabolism , Nanovaccines
14.
Front Immunol ; 15: 1419634, 2024.
Article in English | MEDLINE | ID: mdl-39081325

ABSTRACT

Herpes zoster (HZ), also known as shingles, remains a significant global health issue and most commonly seen in elderly individuals with an early exposure history to varicella-zoster virus (VZV). Currently, the licensed vaccine Shingrix, which comprises a recombinant VZV glycoprotein E (gE) formulated with a potent adjuvant AS01B, is the most effective shingles vaccine on the market. However, undesired reactogenicity and increasing global demand causing vaccine shortage, prompting the development of novel shingles vaccines. Here, we developed novel vaccine candidates utilising multiple nanoparticle (NP) platforms to display the recombinant gE antigen, formulated in an MF59-biosimilar adjuvant. In naïve mice, all tested NP vaccines induced higher humoral and cellular immune responses than Shingrix, among which, the gEM candidate induced the highest cellular response. In live attenuated VZV (VZV LAV)-primed mouse and rhesus macaque models, the gEM candidate elicited superior cell-mediated immunity (CMI) over Shingrix. Collectively, we demonstrated that NP technology remains a suitable tool for developing shingles vaccine, and the reported gEM construct is a highly promising candidate in the next-generation shingles vaccine development.


Subject(s)
Herpes Zoster Vaccine , Herpesvirus 3, Human , Immunity, Cellular , Nanoparticles , Viral Envelope Proteins , Animals , Mice , Herpesvirus 3, Human/immunology , Viral Envelope Proteins/immunology , Herpes Zoster Vaccine/immunology , Herpes Zoster Vaccine/administration & dosage , Macaca mulatta , Herpes Zoster/prevention & control , Herpes Zoster/immunology , Female , Antibodies, Viral/immunology , Antibodies, Viral/blood , Adjuvants, Immunologic/administration & dosage , Humans , Antigens, Viral/immunology , Immunogenicity, Vaccine , Mice, Inbred BALB C , Nanovaccines
15.
Nat Commun ; 15(1): 5800, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987276

ABSTRACT

Enhancing influenza vaccine cross-protection is imperative to alleviate the significant public health burden of influenza. Heterologous sequential immunization may synergize diverse vaccine formulations and routes to improve vaccine potency and breadth. Here we investigate the effects of immunization strategies on the generation of cross-protective immune responses in female Balb/c mice, utilizing mRNA lipid nanoparticle (LNP) and protein-based PHC nanoparticle vaccines targeting influenza hemagglutinin. Our findings emphasize the crucial role of priming vaccination in shaping Th bias and immunodominance hierarchies. mRNA LNP prime favors Th1-leaning responses, while PHC prime elicits Th2-skewing responses. We demonstrate that cellular and mucosal immune responses are pivotal correlates of cross-protection against influenza. Notably, intranasal PHC immunization outperforms its intramuscular counterpart in inducing mucosal immunity and conferring cross-protection. Sequential mRNA LNP prime and intranasal PHC boost demonstrate optimal cross-protection against antigenically drifted and shifted influenza strains. Our study offers valuable insights into tailoring immunization strategies to optimize influenza vaccine effectiveness.


Subject(s)
Administration, Intranasal , Cross Protection , Influenza Vaccines , Mice, Inbred BALB C , Nanoparticles , Orthomyxoviridae Infections , Animals , Female , Humans , Mice , Antibodies, Viral/immunology , Cross Protection/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunity, Mucosal/immunology , Immunization/methods , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Lipids/chemistry , Liposomes , Nanoparticles/chemistry , Nanovaccines/administration & dosage , Nanovaccines/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccination/methods
16.
Vet Microbiol ; 296: 110198, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067145

ABSTRACT

Senecavirus A (SVA) is a causative agent that can cause vesicular disease in swine, which causes a great threat to the swine husbandry in the world. Therefore, it is necessary to develop a vaccine that can effectively prevent the spread of SVA. In this study, we developed a 24-polymeric nano-scaffold using ß-annulus peptide from tomato bushy effect virus (TBSV) by coupling this antigen to SVA B cell epitope VP121-26 and VP2 proteins via linkers, respectively. The SVA-based nanoparticle protein of the VP1(B)-ß-VP2 was expressed and purified by low-cost prokaryotic system to prepare a SVA nanoparticle vaccine. The immunological protective effect of SVA nanoparticle vaccine was evaluated in mouse and swine models, respectively. The results suggested that both mice and swine could induce high levels SVA neutralizing antibodies and IgG antibodies after two doses immunization. In addition, the swine challenge protection experiment showed that the protection rate of immune SVA nanoparticle vaccine and SVA inactivated vaccine both were 80 %, while the negative control had no protection effect. It demonstrated that SVA nanoparticle vaccine effectively prevented SVA infection in swine. In summary, the preparation of SVA vaccine by using ß-annulus peptide is a promising candidate vaccine for prevent SVA transmission, and provides a new idea for the development of novel SVA vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Nanovaccines , Picornaviridae Infections , Picornaviridae , Swine Diseases , Viral Vaccines , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Capsid Proteins/immunology , Mice, Inbred BALB C , Nanovaccines/administration & dosage , Nanovaccines/immunology , Picornaviridae/immunology , Picornaviridae Infections/veterinary , Picornaviridae Infections/prevention & control , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Viral Structural Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage
17.
J Control Release ; 373: 358-369, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009083

ABSTRACT

Cancer vaccines based on single-source (exogenous or endogenous) tumor-associated antigens (TAAs) are often challenged by the insufficient T cell response and the immunosuppressive tumor microenvironment (TME). Herein, a dual TAAs-boosted nanovaccine based on cancer cell (4T1) membrane-cloaked, CO-immobilized Prussian blue nanoparticles (4T1-PB-CO NPs) is developed and coupled with anti-interleukin (IL)-10 therapy to maximize the efficacy of antitumor immunotherapy. 4T1 cell membrane not only endows NPs with tumor targeting ability, but also serves as exogenous TAAs to trigger CD4+ T cell response and M1-phenotype polarization of tumor-associated macrophages. Under near-infrared light irradiation, 4T1-PB-CO NPs release CO to induce immunogenic cell death (ICD) of tumor cells, thus generating endogenous TAAs to activate CD8+ T cell response. Meanwhile, ICD triggers release of damage-associated molecular patterns, which can promote DC maturation to amplify the antitumor T cell response. When combined with anti-IL-10 that reverses the immunosuppressive TME, 4T1-PB-CO NPs efficiently suppress the primary tumors and produce an abscopal effect to inhibit distant tumors in a breast tumor-bearing mouse model. Such a two-pronged cancer vaccine represents a promising paradigm for robust antitumor immunotherapy.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Mice, Inbred BALB C , Nanoparticles , Tumor Microenvironment , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Female , Cell Line, Tumor , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Ferrocyanides/chemistry , Interleukin-10/immunology , Mice , Immunotherapy/methods , CD8-Positive T-Lymphocytes/immunology , Nanovaccines
18.
J Control Release ; 373: 568-582, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067792

ABSTRACT

Cancer vaccine is regarded as an effective immunotherapy approach mediated by dendritic cells (DCs) which are crucial for antigen presentation and the initiation of adaptive immune responses. However, lack of DC-targeting properties significantly hampers the efficacy of cancer vaccines. Here, by using the phage display technique, peptides targeting the endocytic receptor DEC-205 primarily found on cDC1s were initially screened. An optimized hydrolysis-resistant peptide, hr-8, was identified and conjugated to PLGA-loaded antigen (Ag) and CpG adjuvant nanoparticles, resulting in a DC-targeting nanovaccine. The nanovaccine hr-8-PLGA@Ag/CpG facilitates dendritic cell maturation and improves antigen cross-presentation. The nanovaccine can enhance the antitumor immune response mediated by CD8+ T cells by encapsulating the nanovaccine with either exogenous OVA protein antigen or endogenous gp100/E7 antigenic peptide. As a result, strong antitumor effects are observed in both anti-PD-1 responsive B16-OVA and anti-PD-1 non-responsive B16 and TC1 immunocompetent tumor models. In summary, this study presents the initial documentation of a nanovaccine that targets dendritic cells via the novel DEC-205 binding peptide. This approach offers a new method for developing cancer vaccines that can potentially improve the effectiveness of cancer immunotherapy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Immunotherapy , Lectins, C-Type , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Nanoparticles , Peptides , Receptors, Cell Surface , Dendritic Cells/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Animals , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Immunotherapy/methods , Nanoparticles/chemistry , Peptides/chemistry , Peptides/administration & dosage , Minor Histocompatibility Antigens/immunology , Cell Line, Tumor , Ovalbumin/immunology , Ovalbumin/administration & dosage , Female , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Mice , Antigens, CD/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , CD8-Positive T-Lymphocytes/immunology , Nanovaccines
19.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38862192

ABSTRACT

To begin to optimize the immunization routes for our reported PLGA-rMOMP nanovaccine [PLGA-encapsulated Chlamydia muridarum (Cm) recombinant major outer membrane protein (rMOMP)], we compared two prime-boost immunization strategies [subcutaneous (SC) and intramuscular (IM-p) prime routes followed by two SC-boosts)] to evaluate the nanovaccine-induced protective efficacy and immunogenicity in female BALB/c mice. Our results showed that mice immunized via the SC and IM-p routes were protected against a Cm genital challenge by a reduction in bacterial burden and with fewer bacteria in the SC mice. Protection of mice correlated with rMOMP-specific Th1 (IL-2 and IFN-γ) and not Th2 (IL-4, IL-9, and IL-13) cytokines, and CD4+ memory (CD44highCD62Lhigh) T-cells, especially in the SC mice. We also observed higher levels of IL-1α, IL-6, IL-17, CCL-2, and G-CSF in SC-immunized mice. Notably, an increase of cytokines/chemokines was seen after the challenge in the SC, IM-p, and control mice (rMOMP and PBS), suggesting a Cm stimulation. In parallel, rMOMP-specific Th1 (IgG2a and IgG2b) and Th2 (IgG1) serum, mucosal, serum avidity, and neutralizing antibodies were more elevated in SC than in IM-p mice. Overall, the homologous SC prime-boost immunization of mice induced enhanced cellular and antibody responses with better protection against a genital challenge compared to the heterologous IM-p.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Chlamydia Infections , Chlamydia muridarum , Cytokines , Mice, Inbred BALB C , Animals , Female , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Chlamydia muridarum/immunology , Cytokines/metabolism , Chlamydia Infections/prevention & control , Chlamydia Infections/immunology , Mice , Antibodies, Bacterial/blood , Injections, Intramuscular , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Immunization, Secondary , Disease Models, Animal , Immunogenicity, Vaccine , Injections, Subcutaneous , Nanoparticles/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/administration & dosage , Vaccine Efficacy , Th1 Cells/immunology , Nanovaccines
20.
Front Immunol ; 15: 1387811, 2024.
Article in English | MEDLINE | ID: mdl-38911870

ABSTRACT

The Nipah virus (NiV), a highly deadly bat-borne paramyxovirus, poses a substantial threat due to recurrent outbreaks in specific regions, causing severe respiratory and neurological diseases with high morbidity. Two distinct strains, NiV-Malaysia (NiV-M) and NiV-Bangladesh (NiV-B), contribute to outbreaks in different geographical areas. Currently, there are no commercially licensed vaccines or drugs available for prevention or treatment. In response to this urgent need for protection against NiV and related henipaviruses infections, we developed a novel homotypic virus-like nanoparticle (VLP) vaccine co-displaying NiV attachment glycoproteins (G) from both strains, utilizing the self-assembling properties of ferritin protein. In comparison to the NiV G subunit vaccine, our nanoparticle vaccine elicited significantly higher levels of neutralizing antibodies and provided complete protection against a lethal challenge with NiV infection in Syrian hamsters. Remarkably, the nanoparticle vaccine stimulated the production of antibodies that exhibited superior cross-reactivity to homologous or heterologous henipavirus. These findings underscore the potential utility of ferritin-based nanoparticle vaccines in providing both broad-spectrum and long-term protection against NiV and emerging zoonotic henipaviruses challenges.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Ferritins , Henipavirus Infections , Mesocricetus , Nanoparticles , Nipah Virus , Viral Vaccines , Animals , Nipah Virus/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Ferritins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Cricetinae , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Female , Humans , Nanovaccines
SELECTION OF CITATIONS
SEARCH DETAIL