Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.938
Filter
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.
Elife ; 132024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269893

ABSTRACT

Tumor neoantigen peptide vaccines hold potential for boosting cancer immunotherapy, yet efficiently co-delivering peptides and adjuvants to antigen-presenting cells in vivo remains challenging. Virus-like particle (VLP), which is a kind of multiprotein structure organized as virus, can deliver therapeutic substances into cells and stimulate immune response. However, the weak targeted delivery of VLP in vivo and its susceptibility to neutralization by antibodies hinder their clinical applications. Here, we first designed a novel protein carrier using the mammalian-derived capsid protein PEG10, which can self-assemble into endogenous VLP (eVLP) with high protein loading and transfection efficiency. Then, an engineered tumor vaccine, named ePAC, was developed by packaging genetically encoded neoantigen into eVLP with further modification of CpG-ODN on its surface to serve as an adjuvant and targeting unit to dendritic cells (DCs). Significantly, ePAC can efficiently target and transport neoantigens to DCs, and promote DCs maturation to induce neoantigen-specific T cells. Moreover, in mouse orthotopic liver cancer and humanized mouse tumor models, ePAC combined with anti-TIM-3 exhibited remarkable antitumor efficacy. Overall, these results support that ePAC could be safely utilized as cancer vaccines for antitumor therapy, showing significant potential for clinical translation.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Animals , Cancer Vaccines/immunology , Cancer Vaccines/genetics , Cancer Vaccines/administration & dosage , Mice , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Humans , Dendritic Cells/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Peptides/immunology , Female , Mice, Inbred C57BL , Cell Line, Tumor , Vaccination
3.
Sci Adv ; 10(38): eadn3002, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292792

ABSTRACT

In situ vaccine (ISV) is a versatile and personalized local immunotherapeutic strategy. However, the compromised viability and function of dendritic cells (DCs) in a tumor microenvironment (TME) largely limit the therapeutic efficacy. We designed a hybrid nanoparticle-based ISV, which accomplished superior cancer immunotherapy via simultaneously scavenging reactive oxygen species (ROS) and activating the stimulator of interferon genes (STING) pathway in DCs. This ISV was constructed by encapsulating a chemodrug, SN38, into diselenide bond-bridged organosilica nanoparticles, followed by coating with a Mn2+-based metal phenolic network. We show that this ISV can activate the STING pathway through Mn2+ and SN38 comediated signaling and simultaneously scavenge preexisting H2O2 in the TME and Mn2+-catalyzed •OH by leveraging the antioxidant property of diselenide and polyphenol. This ISV effectively activated DCs and protected them from oxidative damage, leading to remarkable downstream T cell activation and systemic antitumor immunity. This work highlights a nanoparticle design that manipulates DCs in the TME for improving the ISV.


Subject(s)
Cancer Vaccines , Dendritic Cells , Membrane Proteins , Nanoparticles , Reactive Oxygen Species , Tumor Microenvironment , Reactive Oxygen Species/metabolism , Animals , Nanoparticles/chemistry , Mice , Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor , Immunotherapy/methods , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry
4.
Sci Adv ; 10(38): eado6491, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292790

ABSTRACT

Neoantigens arising from somatic mutations are tumor specific and induce antitumor host T cell responses. However, their sequences are individual specific and need to be identified for each patient for therapeutic applications. Here, we present a proteogenomic approach for neoantigen identification, named Neoantigen Selection using a Surrogate Immunopeptidome (NESSIE). This approach uses an autologous wild-type immunopeptidome as a surrogate for the tumor immunopeptidome and allows human leukocyte antigen (HLA)-agnostic identification of both HLA class I (HLA-I) and HLA class II (HLA-II) neoantigens. We demonstrate the direct identification of highly immunogenic HLA-I and HLA-II neoantigens using NESSIE in patients with colorectal cancer and endometrial cancer. Fresh or frozen tumor samples are not required for analysis, making it applicable to many patients in clinical settings. We also demonstrate tumor prevention by vaccination with selected neoantigens in a preclinical mouse model. This approach may benefit personalized T cell-mediated immunotherapies.


Subject(s)
Antigens, Neoplasm , Histocompatibility Antigens Class II , Histocompatibility Antigens Class I , Humans , Antigens, Neoplasm/immunology , Animals , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Mice , Histocompatibility Antigens Class II/immunology , Female , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , Peptides/immunology , Endometrial Neoplasms/immunology , Immunotherapy/methods
5.
Clin Transl Med ; 14(9): e70020, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39275923

ABSTRACT

Significant strides have been made in identifying tumour-associated antigens over the past decade, revealing unique epitopes crucial for targeted cancer therapy. Among these, the New York esophageal squamous cell carcinoma (NY-ESO-1) protein, a cancer/testis antigen, stands out. This protein is presented on the cell surface by major histocompatibility complex class I molecules and exhibits restricted expression in germline cells and various cancers, marking it as an immune-privileged site. Remarkably, NY-ESO-1 serves a dual role as both a tumour-associated antigen and its own adjuvant, implying a potential function as a damage-associated molecular pattern. It elicits strong humoural immune responses, with specific antibody frequencies significantly correlating with disease progression. These characteristics make NY-ESO-1 an appealing candidate for developing effective and specific immunotherapy, particularly for advanced stages of disease. In this review, we provide a comprehensive overview of NY-ESO-1 as an immunogenic tumour antigen. We then explore the diverse strategies for targeting NY-ESO-1, including cancer vaccination with peptides, proteins, DNA, mRNA, bacterial vectors, viral vectors, dendritic cells and artificial adjuvant vector cells, while considering the benefits and drawbacks of each strategy. Additionally, we offer an in-depth analysis of adoptive T-cell therapies, highlighting innovative techniques such as next-generation NY-ESO-1 T-cell products and the integration with lymph node-targeted vaccines to address challenges and enhance therapeutic efficacy. Overall, this comprehensive review sheds light on the evolving landscape of NY-ESO-1 targeting and its potential implications for cancer treatment, opening avenues for future tailored directions in NY-ESO-1-specific immunotherapy. HIGHLIGHTS: Endogenous immune response: NY-ESO-1 exhibited high immunogenicity, activating endogenous dendritic cells, T cells and B cells. NY-ESO-1-based cancer vaccines: NY-ESO-1 vaccines using protein/peptide, RNA/DNA, microbial vectors and artificial adjuvant vector cells have shown promise in enhancing immune responses against tumours. NY-ESO-1-specific T-cell receptor-engineered cells: NY-ESO-1-targeted T cells, along with ongoing innovations in engineered natural killer cells and other cell therapies, have improved the efficacy of immunotherapy.


Subject(s)
Antigens, Neoplasm , Immunotherapy , Membrane Proteins , Neoplasms , Humans , Antigens, Neoplasm/immunology , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Membrane Proteins/immunology , Membrane Proteins/therapeutic use , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology
6.
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
7.
J Nanobiotechnology ; 22(1): 562, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39272180

ABSTRACT

BACKGROUND: Tumor neoantigen peptide-based vaccines, systemic immunotherapies that enhance antitumor immunity by activating and expanding antigen-specific T cells, have achieved remarkable results in the treatment of a variety of solid tumors. However, how to effectively deliver neoantigens to induce robust antitumor immune responses remains a major obstacle. RESULTS: Here, we developed a safe and effective neoantigen peptide delivery system (neoantigen-ferritin nanoparticles, neoantigen-FNs) that successfully achieved effective lymph node targeting and induced robust antitumor immune responses. The genetically engineered self-assembled particles neoantigen-FNs with a size of 12 nm were obtained by fusing a neoantigen with optimized ferritin, which rapidly drainage to and continuously accumulate in lymph nodes. The neoantigen-FNs vaccine induced a greater quantity and quality of antigen-specific CD8+ T cells and resulted in significant growth control of multiple tumors, dramatic inhibition of melanoma metastasis and regression of established tumors. In addition, no obvious toxic side effects were detected in the various models, indicating the high safety of optimized ferritin as a vaccine carrier. CONCLUSIONS: Homogeneous and safe neoantigen-FNs could be a very promising system for neoantigen peptide delivery because of their ability to efficiently drainage to lymph nodes and induce efficient antitumor immune responses.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Ferritins , Mice, Inbred C57BL , Nanoparticles , Animals , Ferritins/chemistry , Antigens, Neoplasm/immunology , Nanoparticles/chemistry , Cancer Vaccines/immunology , Mice , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , Female , Immunotherapy/methods , Neoplasm Metastasis , Humans , Lymph Nodes , Recombinant Proteins
9.
Nat Commun ; 15(1): 7664, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227567

ABSTRACT

The immunosuppressive tumor microenvironment (TME) remains a major obstacle to tumor control and causes suboptimal responses to immune checkpoint blockade (ICB) therapy. Thus, developing feasible therapeutic strategies that trigger inflammatory responses in the TME could improve the ICB efficacy. Mitochondria play an essential role in inflammation regulation and tumor immunogenicity induction. Herein, we report the discovery and characterization of a class of small molecules that can recapitulate aqueous self-assembly behavior, specifically target cellular organelles (e.g., mitochondria), and invigorate tumor cell immunogenicity. Mechanistically, this nanoassembly platform dynamically rewires mitochondria, induces endoplasmic reticulum stress, and causes apoptosis/paraptosis-associated immunogenic cell death. After treatment, stressed and dying tumor cells can act as prophylactic or therapeutic cancer vaccines. In preclinical mouse models of cancers with intrinsic or acquired resistance to PD-1 blockade, the local administration of nanoassemblies inflames the immunologically silent TME and synergizes with ICB therapy, generating potent antitumor immunity. This chemically programmed small-molecule immune enhancer acts distinctly from regular cytotoxic therapeutics and offers a promising strategy for synchronous and dynamic tailoring of innate immunity to achieve traceless cancer therapy and overcome immunosuppression in cancers.


Subject(s)
Mitochondria , Neoplasms , Tumor Microenvironment , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Humans , Cell Line, Tumor , Neoplasms/immunology , Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Apoptosis/drug effects , Female , Immunogenic Cell Death/drug effects , Mice, Inbred C57BL , Nanoparticles/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Immunotherapy/methods
10.
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
11.
Carbohydr Polym ; 346: 122637, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245502

ABSTRACT

Tumor vaccines have become a promising approach for cancer treatment by triggering antigen-specific responses against tumors. However, autophagy and immunosuppressive tumor microenvironment (TME) reduce antigen exposure and immunogenicity, which limit the effect of tumor vaccines. Here, we develop fucoidan (Fuc) based chlorin e6 (Ce6)-chloroquine (CQ) self-assembly hydrogels (CCFG) as in situ vaccines. Ce6 triggers immune response in situ by photodynamic therapy (PDT) induced immunogenic cell death (ICD) effect, which is further enhanced by macrophage polarization of Fuc and autophagy inhibition of CQ. In vivo studies show that CCFG effectively enhances antigen presentation under laser irradiation, which induces a powerful in situ vaccine effect and significantly inhibits tumor metastasis and recurrence. Our study provides a novel approach for enhancing tumor immunotherapy and inhibiting tumor recurrence and metastasis.


Subject(s)
Autophagy , Cancer Vaccines , Chlorophyllides , Chloroquine , Hydrogels , Immunotherapy , Macrophages , Photochemotherapy , Polysaccharides , Porphyrins , Animals , Polysaccharides/pharmacology , Polysaccharides/chemistry , Mice , Cancer Vaccines/pharmacology , Cancer Vaccines/immunology , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/therapeutic use , Autophagy/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Immunotherapy/methods , Photochemotherapy/methods , Macrophages/drug effects , Macrophages/immunology , Chloroquine/pharmacology , Mice, Inbred C57BL , Tumor Microenvironment/drug effects , RAW 264.7 Cells , Cell Line, Tumor , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Mice, Inbred BALB C , Female
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.
J Immunother Cancer ; 12(8)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39209449

ABSTRACT

BACKGROUND: Targeting kinases presents a potential strategy for treating solid tumors; however, the therapeutic potential of vaccines targeting kinases remains uncertain. METHODS: Adenovirus (Ad) vaccines encoding Aurora kinase A (AURKA) or cyclin-dependent kinase 7 (CDK7) were developed, and their therapeutic potentials were investigated by various methods including western blot, flow cytometry, cytotoxic T lymphocyte assay, and enzyme-linked immunospot (ELISpot), in mouse and humanized solid tumor models. RESULTS: Co-immunization with Ad-AURKA/CDK7 effectively prevented subcutaneous tumor growth in the Renca, RM-1, MC38, and Hepa1-6 tumor models. In therapeutic tumor models, Ad-AURKA/CDK7 treatment impeded tumor growth and increased immune cell infiltration. Administration of Ad-AURKA/CDK7 promoted the induction and maturation of dendritic cell subsets and augmented multifunctional CD8+ T-cell antitumor immunity. Furthermore, the vaccine induced a long-lasting antitumor effect by promoting the generation of memory CD8+ T cells. Tumor recovery on CD8+ T-cell depletion underscored the indispensable role of these cells in the observed therapeutic effects. The potent efficacy of the Ad-AURKA/CDK7 vaccine was consistently demonstrated in lung metastasis, orthotopic, and humanized tumor models by inducing multifunctional CD8+ T-cell antitumor immune responses. CONCLUSIONS: Our findings illustrate that the Ad-AURKA/CDK7 vaccine targeting dual kinases AURKA and CDK7 emerges as a promising and effective therapeutic approach for the treatment of solid tumors.


Subject(s)
Aurora Kinase A , Cancer Vaccines , Animals , Mice , Humans , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Adenoviridae , Cell Line, Tumor , Cyclin-Dependent Kinase-Activating Kinase , Female , Neoplasms/immunology , Neoplasms/therapy , Adenovirus Vaccines/immunology , Adenovirus Vaccines/therapeutic use , CD8-Positive T-Lymphocytes/immunology
14.
ACS Appl Mater Interfaces ; 16(35): 45917-45928, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39178210

ABSTRACT

Effective activation of an antigen-specific immune response hinges upon the intracellular delivery of cancer antigens to antigen-presenting cells (APCs), marking the initial stride in cancer vaccine development. Leveraging biomimetic topological morphology, we employed virus-like mesoporous silica nanoparticles (VMSNs) coloaded with antigens and toll-like receptor 9 (TLR9) agonists to craft a potent cancer vaccine. Our VMSNs could be efficiently internalized by APCs to a greater extent than their nonviral structured counterparts, thereby promoting the activation of APCs by upregulating the TLR9 pathway and cross-presenting ovalbumin (OVA) epitopes. In in vivo animal study, VMSN-based nanovaccines triggered substantial CD4+ and CD8+ lymphocyte populations in both lymph nodes and spleen while inducing the effector memory of adaptive T cells. Consequently, VMSN-based nanovaccines suppressed tumor progression and increased the survival rate of B16-OVA-bearing mice in both prophylactic and therapeutic studies. The combination of immune checkpoint blockade (ICB) with the VMSN-based nanovaccine has synergistic effects in significantly preventing tumor progression under therapeutic conditions. These findings highlight the potential of viral structure-mimicking mesoporous silica nanoparticles as promising candidates for antigen-delivering nanocarriers in vaccine development.


Subject(s)
Mice, Inbred C57BL , Nanoparticles , Silicon Dioxide , Silicon Dioxide/chemistry , Animals , Nanoparticles/chemistry , Mice , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , Ovalbumin/chemistry , Ovalbumin/immunology , Porosity , Adaptive Immunity/drug effects , Humans , Antigen-Presenting Cells/immunology , Neoplasms/immunology , Female , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/agonists , Antigens, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Cell Line, Tumor
15.
Front Immunol ; 15: 1404861, 2024.
Article in English | MEDLINE | ID: mdl-39192978

ABSTRACT

Background: Glioblastoma (GBM) is a poor prognosis grade 4 glioma. After surgical resection, the standard therapy consists of concurrent radiotherapy (RT) and temozolomide (TMZ) followed by TMZ alone. Our previous data on melanoma patients showed that Dendritic Cell vaccination (DCvax) could increase the amount of intratumoral-activated cytotoxic T lymphocytes. Methods: This is a single-arm, monocentric, phase II trial in two steps according to Simon's design. The trial aims to evaluate progression-free survival (PFS) at three months and the safety of a DCvax integrated with standard therapy in resected GBM patients. DCvax administration begins after completion of RT-CTwith weekly administrations for 4 weeks, then is alternated monthly with TMZ cycles. The primary endpoints are PFS at three months and safety. One of the secondary objectives is to evaluate the immune response both in vitro and in vivo (DTH skin test). Results: By December 2022, the first pre-planned step of the study was concluded with the enrollment, treatment and follow up of 9 evaluable patients. Two patients had progressed within three months after leukapheresis, but none had experienced DCvax-related G3-4 toxicities Five patients experienced a positive DTH test towards KLH and one of these also towards autologous tumor homogenate. The median PFS from leukapheresis was 11.3 months and 12.2 months from surgery. Conclusions: This combination therapy is well-tolerated, and the two endpoints required for the first step have been achieved. Therefore, the study will proceed to enroll the remaining 19 patients. (Eudract number: 2020-003755-15 https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-003755-15/IT).


Subject(s)
Brain Neoplasms , Cancer Vaccines , Dendritic Cells , Glioblastoma , Humans , Glioblastoma/therapy , Glioblastoma/immunology , Glioblastoma/mortality , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Dendritic Cells/immunology , Dendritic Cells/transplantation , Middle Aged , Female , Male , Adult , Aged , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Temozolomide/therapeutic use , Temozolomide/administration & dosage , Progression-Free Survival
16.
Hum Vaccin Immunother ; 20(1): 2395680, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39208856

ABSTRACT

We have previously reported two single-agent phase I trials, evaluating the dose or schedule, of a DNA vaccine (pTVG-HP) encoding prostatic acid phosphatase (PAP) administered with GM-CSF as the adjuvant. These were in patients with PSA-recurrent, radiographically non-metastatic, prostate cancer (PCa). We report here the long-term safety and overall survival of these patients. Specifically, 22 patients with non-metastatic, castration-sensitive PCa (nmCSPC) were treated with pTVG-HP, 100-1500 µg, administered over 12 weeks and followed for 15 y. 17 patients with non-metastatic castration-resistant PCa (nmCRPC) were treated with 100 µg pTVG-HP with different schedules of administration over 1 y and followed for 5 y. No adverse events were detected in long-term follow-up from either trial that were deemed possibly related to vaccination. Patients with nmCSPC had a median overall survival of 12.3 y, with 5/22 (23%) alive at 15 y. 8/22 (36%) died due to prostate cancer with a median survival of 11.0 y, and 9/22 (41%) died of other causes. Patients with nmCRPC had a median overall survival of 4.5 y, with 8/17 (47%) alive at 5 y. The presence of T-cells specific for the PAP target antigen was detectable in 6/10 (60%) individuals with nmCSPC, and 3/5 (60%) individuals with nmCRPC, many years after immunization. The detection of immune responses to the vaccine target years after immunization suggests durable immunity can be elicited in patients using a DNA vaccine encoding a tumor-associated antigen.Trial Registration: NCT00582140 and NCT00849121.


Subject(s)
Cancer Vaccines , Prostate-Specific Antigen , Prostatic Neoplasms , Vaccines, DNA , Humans , Male , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Prostate-Specific Antigen/immunology , Cancer Vaccines/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/administration & dosage , Aged , Follow-Up Studies , Prostatic Neoplasms/immunology , Middle Aged , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Treatment Outcome , Aged, 80 and over , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Neoplasm Recurrence, Local , Survival Analysis , Acid Phosphatase , Protein Tyrosine Phosphatases/immunology
17.
Int Immunopharmacol ; 141: 112956, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39168022

ABSTRACT

DNA vaccines are prospective for their efficient manufacturing process, but their immunogenicity is limited as they cannot efficiently induce CD8+ T cell responses. A promising approach is to induce cross-presentation by targeting antigens to DCs. Flt3L can expand the number of type 1 conventional DCs and thereby improve cross-presentation. In this study, we first constructed a DNA vaccine expressing soluble PD1 and found that the therapeutic effect of targeting DCs with only the sPD1 vaccine was limited. When combined the vaccine with Flt3L, the anti-tumor effect was significantly enhanced. Considering the complexity of tumors and that a single method may not be able to activate a large number of effective CD8+ T cells, we combined different drugs and the vaccine with Flt3L based on the characteristics of different tumors. In 4T1 model, we reduced Tregs through cyclophosphamide. In Panc02 model, we increased activated DCs by using aCD40. Both strategies triggered strong CD8+ T cell responses and significantly improved the therapeutic effect. Our study provides important support for the clinical exploration of DC-targeted DNA vaccines in combination with Flt3L.


Subject(s)
CD8-Positive T-Lymphocytes , Cancer Vaccines , Dendritic Cells , Membrane Proteins , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor , Vaccines, DNA , Vaccines, DNA/immunology , Animals , Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Cancer Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/immunology , Membrane Proteins/genetics , Female , Mice , Cell Line, Tumor , Humans , Mice, Inbred C57BL
18.
Life Sci ; 356: 123018, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39214286

ABSTRACT

GBM is the most common, aggressive, and intracranial primary brain tumor; it originates from the glial progenitor cells, has poor overall survival (OS), and has limited treatment options. In this decade, GBM immunotherapy is in trend and preferred over several conventional therapies, due to their better patient survival outcome. This review explores the clinical trials of several immunotherapeutic approaches (immune checkpoint blockers (ICBs), CAR T-cell therapy, cancer vaccines, and adoptive cell therapy) with their efficacy and safety. Despite significant progress, several challenges (viz., immunosuppressive microenvironment, heterogeneity, and blood-brain barrier (BBB)) were experienced that hamper their immunotherapeutic potential. Furthermore, these challenges were clinically studied to be resolved by multiple combinatorial approaches, discussed in the later part of the review. Thus, this review suggests the clinical use and potential of immunotherapy in GBM and provides the holistic recent knowledge and future perspectives.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunotherapy , Tumor Microenvironment , Humans , Immunotherapy/methods , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Glioblastoma/therapy , Glioblastoma/immunology , Tumor Microenvironment/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Immune Checkpoint Inhibitors/therapeutic use , Animals , Blood-Brain Barrier/immunology
19.
Anticancer Res ; 44(9): 3713-3724, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39197920

ABSTRACT

Cancer immunotherapy activates the host immune system against tumor cells and has the potential to lead to the development of innovative strategies for cancer treatment. Neoantigens are non-self-antigens produced by genetic mutations in tumor cells that induce a strong immune response against tumor cells without central immune tolerance. Along with advances in neoantigen analysis technology, the development of vaccines focusing on neoantigens is being accelerated. Whereas there are various platforms for neoantigen vaccines, combined immuno-therapies are being developed simultaneously with the clinical application of synthetic long peptides and mRNA and dendritic-cell (DC)-based vaccines. Personalized DC-based vaccines not only can load various antigens including neoantigens, but also have the potential to elicit a strong immune response in T cells as antigen-presenting cells. In this review, we describe the properties of neoantigens and the basic characteristics of DCs. We also discuss the clinical applications of neoantigen vaccines, focusing on personalized DC-based vaccines, as well as future research and development directions and challenges.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Neoplasms , Precision Medicine , Humans , Dendritic Cells/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Antigens, Neoplasm/immunology , Neoplasms/immunology , Neoplasms/therapy , Precision Medicine/methods , Immunotherapy/methods , Animals
20.
Hum Vaccin Immunother ; 20(1): 2392961, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39161160

ABSTRACT

Dendritic cells, which are crucial for inducing T-cell responses, are pivotal in current immunotherapy strategies aiming to replenish depleted T cells within the tumor microenvironment to combat tumors. Consequently, dendritic cell vaccine-based cancer therapies have garnered significant attention. Through bibliometric analysis, we examined research trends in this field. We searched the Web of Science core database and identified 16,476 articles on dendritic cell-based vaccines published from January 1, 2009, to December 30, 2023. The United States leads in this research domain, with Emory University being a prominent collaborator. The Journal of Immunology is the primary publication outlet, and Banchereau, J emerges as the most influential author. Recent hot keywords include nanoparticle, delivery, cancer vaccine, and clinical trial, indicating that cancer immunotherapy research, especially dendritic cell-based vaccines, is poised to become a future trend and hotspot.


Subject(s)
Bibliometrics , Cancer Vaccines , Dendritic Cells , Dendritic Cells/immunology , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL