Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.114
Filter
1.
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
2.
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
3.
Front Immunol ; 15: 1411490, 2024.
Article in English | MEDLINE | ID: mdl-39139570

ABSTRACT

Immunotherapy has revolutionized cancer treatment by leveraging the immune system's innate capabilities to combat malignancies. Despite the promise of tumor antigens in stimulating anti-tumor immune responses, their clinical utility is hampered by limitations in eliciting robust and durable immune reactions, exacerbated by tumor heterogeneity and immune evasion mechanisms. Recent insights into the immunogenic properties of host homologous microbial antigens have sparked interest in their potential for augmenting anti-tumor immunity while minimizing off-target effects. This review explores the therapeutic potential of microbial antigen peptides in tumor immunotherapy, beginning with an overview of tumor antigens and their challenges in clinical translation. We further explore the intricate relationship between microorganisms and tumor development, elucidating the concept of molecular mimicry and its implications for immune recognition of tumor-associated antigens. Finally, we discuss methodologies for identifying and characterizing microbial antigen peptides, highlighting their immunogenicity and prospects for therapeutic application.


Subject(s)
Antigens, Bacterial , Antigens, Neoplasm , Immunotherapy , Neoplasms , Humans , Antigens, Neoplasm/immunology , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Animals , Antigens, Bacterial/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Molecular Mimicry/immunology
4.
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
5.
Nat Commun ; 15(1): 6870, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39127809

ABSTRACT

Current treatment outcome of patients with glioblastoma (GBM) remains poor. Following standard therapy, recurrence is universal with limited survival. Tumors from 173 GBM patients are analysed for somatic mutations to generate a personalized peptide vaccine targeting tumor-specific neoantigens. All patients were treated within the scope of an individual healing attempt. Among all vaccinated patients, including 70 treated prior to progression (primary) and 103 treated after progression (recurrent), the median overall survival from first diagnosis is 31.9 months (95% CI: 25.0-36.5). Adverse events are infrequent and are predominantly grade 1 or 2. A vaccine-induced immune response to at least one of the vaccinated peptides is detected in blood samples of 87 of 97 (90%) monitored patients. Vaccine-specific T-cell responses are durable in most patients. Significantly prolonged survival is observed for patients with multiple vaccine-induced T-cell responses (53 months) compared to those with no/low induced responses (27 months; P = 0.03). Altogether, our results highlight that the application of personalized neoantigen-targeting peptide vaccine is feasible and represents a promising potential treatment option for GBM patients.


Subject(s)
Brain Neoplasms , Cancer Vaccines , Glioblastoma , Precision Medicine , Vaccines, Subunit , Humans , Glioblastoma/immunology , Glioblastoma/therapy , Female , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Male , Middle Aged , Precision Medicine/methods , Aged , Adult , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , Treatment Outcome , Protein Subunit Vaccines
6.
Nat Commun ; 15(1): 6874, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39128904

ABSTRACT

Lymphoplasmacytic lymphoma (LPL) is an incurable low-grade lymphoma with no standard therapy. Nine asymptomatic patients treated with a first-in-human, neoantigen DNA vaccine experienced no dose limiting toxicities (primary endpoint, NCT01209871). All patients achieve stable disease or better, with one minor response, and median time to progression of 72+ months. Post-vaccine single-cell transcriptomics reveal dichotomous antitumor responses, with reduced tumor B-cells (tracked by unique B cell receptor) and their survival pathways, but no change in clonal plasma cells. Downregulation of human leukocyte antigen (HLA) class II molecules and paradoxical upregulation of insulin-like growth factor (IGF) by the latter suggest resistance mechanisms. Vaccine therapy activates and expands bone marrow T-cell clonotypes, and functional neoantigen-specific responses (secondary endpoint), but not co-inhibitory pathways or Treg, and reduces protumoral signaling by myeloid cells, suggesting favorable perturbation of the tumor immune microenvironment. Future strategies may require combinations of vaccines with agents targeting plasma cell subpopulations, or blockade of IGF-1 signaling or myeloid cell checkpoints.


Subject(s)
Cancer Vaccines , Waldenstrom Macroglobulinemia , Humans , Male , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Middle Aged , Female , Waldenstrom Macroglobulinemia/immunology , Waldenstrom Macroglobulinemia/therapy , Waldenstrom Macroglobulinemia/genetics , Aged , Tumor Microenvironment/immunology , Precision Medicine/methods , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology
10.
AAPS PharmSciTech ; 25(6): 168, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39044047

ABSTRACT

Immunotherapy is one of the most promising therapeutic approaches in the field of cancer treatment. As a tumor progresses, tumor cells employ an array of immune-regulatory mechanisms to suppress immune responses within the tumor microenvironment. Using our understanding of these mechanisms, cancer immunotherapy has been developed to enhance the immune system's effectiveness in treating cancer. Numerous cancer immunotherapies are currently in clinical use, yet many others are either in different stages of development or undergoing clinical studies. In this paper, we briefly discuss the features and current status of cancer immunotherapies. This includes the application of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell therapy, cytokine therapy, cancer vaccines, and gene therapy, all of which have gained significant recognition in clinical practice. Additionally, we discuss limitations that may hinder successful clinical utilization and promising strategies, such as combining immunotherapy with nanotechnology.


Subject(s)
Cancer Vaccines , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , Immunotherapy/trends , Tumor Microenvironment/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Animals , Genetic Therapy/methods , Genetic Therapy/trends , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Antibodies, Monoclonal/therapeutic use , Nanotechnology/methods , Cytokines/metabolism , Cytokines/immunology
11.
Hum Vaccin Immunother ; 20(1): 2379086, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39043175

ABSTRACT

Bladder cancer (BC) accounts for about 4% of all malignancies. Non-muscle-invasive BC, 75% of cases, is treated with transurethral resection and adjuvant intravesical instillation, while muscle-invasive BC warrants cisplatin-based perioperative chemotherapy. Although immune-checkpoint inhibitors, antibody drug conjugates and targeted agents have provided dramatic advances, metastatic BC remains a generally incurable disease and clinical trials continue to vigorously evaluate novel molecules. Cancer vaccines aim at activating the patient's immune system against tumor cells. Several means of delivering neoantigens have been developed, including peptides, antigen-presenting cells, virus, or nucleic acids. Various improvements are constantly being explored, such as adjuvants use and combination strategies. Nucleic acids-based vaccines are increasingly gaining attention in recent years, with promising results in other malignancies. However, despite the recent advantages, numerous obstacles persist. This review is aimed at describing the different types of cancer vaccines, their evaluations in UC patients and the more recent innovations in this field.


Subject(s)
Cancer Vaccines , Urinary Bladder Neoplasms , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/immunology , Immunotherapy/methods , Antigens, Neoplasm/immunology
12.
Expert Rev Clin Immunol ; 20(8): 839-847, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39007892

ABSTRACT

INTRODUCTION: Tertiary lymphoid structures (TLS) arise at chronic inflammatory sites where they function as miniature lymph nodes to generate immune responses, which can be beneficial or detrimental, in diseases as diverse as autoimmunity, chronic infections and cancer. A growing number of studies show that a TLS presence in tumors from cancer patients treated with immune checkpoint inhibitors is closely linked with improved clinical outcomes. TLS may foster the generation of specific anti-tumor immune responses and immunological memory that recognizes a patient's own tumor. Due to repeated rounds of chronic inflammation, some tumor-associated TLS may be immunologically inactive, with immune checkpoint inhibitors functioning to revitalize them through pathway activation. AREAS COVERED: This review summarizes work on TLS and how they mediate immune responses in human tumors. We also explore TLS as potential prognostic and predictive biomarkers for immunotherapy. EXPERT OPINION: The presence of TLS in human tumors has been linked with a better clinical prognosis, response to treatment(s) and overall survival. TLS provide a structured microenvironment for the activation, expansion and maturation of immune cells at the tumor site. These activities can enhance the efficacy of immunotherapeutic treatments such as checkpoint inhibitors and cancer vaccines by revitalizing local anti-tumor immunity.


Subject(s)
Immunotherapy , Neoplasms , Tertiary Lymphoid Structures , Tumor Microenvironment , Humans , Tertiary Lymphoid Structures/immunology , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Tumor Microenvironment/immunology , Animals , Immune Checkpoint Inhibitors/therapeutic use , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology
13.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38955422

ABSTRACT

BACKGROUND: Despite advances in checkpoint inhibitor (CPI) therapy for cancer treatment, many cancers remain resistant. Tumors deemed "cold" based on lack of T cell infiltration show reduced potential for CPI therapy. Cancer vaccines may overcome the inadequacy of existing T cells by inducing the needed antitumor T cell response to synergize with CPIs and overcome resistance. METHODS: CT26 and TC1 tumor cells were injected subcutaneously into mice. Mice were treated with combinations of CPIs alone or a cancer vaccine specific to the tumor antigen E7 present in TC1 cells. CPIs for the TC1 model were selected because of immunophenotyping TC1 tumors. Antitumor and protumor immunity, tumor size and survival, sequence and timing of vaccine and CPI administration, and efficacy of treatment in young and aged mice were probed. RESULTS: While "hot" CT26 tumors are treatable with combinations of second-generation CPIs alone or with anti-TGFß, "cold" TC1 tumor reduction requires the synergy of a tumor-antigen-specific vaccine in combination with two CPIs, anti-TIGIT and anti-PD-L1, predicted by tumor microenvironment (TME) characterization. The synergistic triple combination delays tumor growth better than any pairwise combination and improves survival in a CD8+T cell-dependent manner. Depletion of CD4+T cells improved the treatment response, and depleting regulatory T cells (Treg) revealed Tregs to be inhibiting the response as also predicted from TME analysis. We found the sequence of CPI and vaccine administration dictates the success of the treatment, and the triple combination administered concurrently induces the highest E7-specific T cell response. Contrary to young mice, in aged mice, the cancer vaccine alone is ineffective, requiring the CPIs to delay tumor growth. CONCLUSIONS: These findings show how pre-existing or vaccine-mediated de novo T cell responses can both be amplified by and facilitate synergistic CPIs and Treg depletion that together lead to greater survival, and how analysis of the TME can help rationally design combination therapies and precision medicine to enhance clinical response to CPI and cancer vaccine therapy.


Subject(s)
Cancer Vaccines , Immune Checkpoint Inhibitors , T-Lymphocytes, Regulatory , Tumor Microenvironment , Animals , Cancer Vaccines/pharmacology , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tumor Microenvironment/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Female , Cell Line, Tumor , Humans
14.
Theranostics ; 14(9): 3674-3692, 2024.
Article in English | MEDLINE | ID: mdl-38948057

ABSTRACT

Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2's structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.


Subject(s)
Antigens, Neoplasm , Cell Adhesion Molecules , Immunoconjugates , Molecular Targeted Therapy , Neoplasms , Humans , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/therapy , Immunotherapy/methods , Animals , Cancer Vaccines/therapeutic use
15.
Oncoimmunology ; 13(1): 2373526, 2024.
Article in English | MEDLINE | ID: mdl-38948931

ABSTRACT

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Subject(s)
Adjuvants, Immunologic , Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Prostatic Neoplasms , RNA, Messenger , Animals , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Antigens, Neoplasm/immunology , Mice , Dendritic Cells/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Humans , Mice, Inbred C57BL , Cell Line, Tumor , mRNA Vaccines , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immunotherapy/methods , Lymphocyte Activation/drug effects
16.
Egypt J Immunol ; 31(3): 123-130, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38995717

ABSTRACT

Cancer immunotherapy is a promising strategy in cancer management, including hepatocellular carcinoma (HCC). This experimental study aimed to evaluate interleukin-10 (IL-10) as a biomarker for monitoring the response of tumor-derived autophagosomes vaccine in inducing antitumor immunity in HCC induced mice. It was conducted on 56 BALB/c mice; divided into 20 normal and 36, cancer induced with human liver cancer cell line (HepG2) cells. The latter group was subdivided into a positive control group (n=6) and a treated group (n=30), that was subdivided into 3 subgroups: (A) treated with dendritic cells (DC) vaccine only, (B) treated with vaccine named Dribbles only, and (C) treated with DC plus Dribbles. Serum IL-10 was assessed after immunotherapy. The mean percentage of tumor volume reduction in mice vaccinated by DC plus Dribbles was significantly superior to DC and Dribbles groups (p= 0.013, and p= 0.043, respectively). There was a statistically significant difference in IL-10 levels between different immunotherapy groups (p= 0.0003). As the mean IL-10 level was 19.50 pg/ml for the positive control group, 13 pg/ml for Dribbles group, 10 pg/ml for DCs group and 3.50 pg/ml for DCs plus Dribbles group. We conclude that DC-Dribbles vaccine has a remarkable efficacy superior to either Dribbles alone or DC alone in the decline of HCC development and survival improvement. IL-10 is a predictive biomarker for response after immunotherapy.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Dendritic Cells , Immunotherapy , Interleukin-10 , Liver Neoplasms , Mice, Inbred BALB C , Animals , Interleukin-10/blood , Interleukin-10/immunology , Dendritic Cells/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Mice , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Immunotherapy/methods , Hep G2 Cells , Disease Models, Animal , Biomarkers, Tumor/blood , Biomarkers, Tumor/immunology
17.
ACS Nano ; 18(28): 18604-18621, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38952130

ABSTRACT

Chemo-immunotherapy has become a promising strategy for cancer treatment. However, the inability of the drugs to penetrate deeply into the tumor and form potent tumor vaccines in vivo severely restricts the antitumor effect of chemo-immunotherapy. In this work, an injectable sodium alginate platform is reported to promote penetration of the chemotherapeutic doxorubicin (DOX) and delivery of personalized tumor vaccines. The injectable multifunctional sodium alginate platform cross-links rapidly in the presence of physiological concentrations of Ca2+, forming a hydrogel that acts as a drug depot and releases loaded hyaluronidase (HAase), DOX, and micelles (IP-NPs) slowly and sustainedly. By degrading hyaluronic acid (HA) overexpressed in tumor tissue, HAase can make tumor tissue "loose" and favor other components to penetrate deeply. DOX induces potent immunogenic cell death (ICD) and produces tumor-associated antigens (TAAs), which could be effectively captured by polyethylenimine (PEI) coated IP-NPs micelles and form personalized tumor vaccines. The vaccines efficaciously facilitate the maturation of dendritic cells (DCs) and activation of T lymphocytes, thus producing long-term immune memory. Imiquimod (IMQ) loaded in the core could further activate the immune system and trigger a more robust antitumor immune effect. Hence, the research proposes a multifunctional drug delivery platform for the effective treatment of colorectal cancer.


Subject(s)
Alginates , Doxorubicin , Hydrogels , Immunotherapy , Nanoparticles , Alginates/chemistry , Hydrogels/chemistry , Animals , Nanoparticles/chemistry , Mice , Doxorubicin/chemistry , Doxorubicin/pharmacology , Humans , Cancer Vaccines/chemistry , Cancer Vaccines/administration & dosage , Hyaluronoglucosaminidase/metabolism , Micelles , Cell Line, Tumor
18.
ACS Nano ; 18(28): 18425-18443, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38975713

ABSTRACT

Tumor in situ vaccination (ISV) strategies have emerged in clinical trials as promising approaches, involving the release of tumor antigens through local radiotherapy and intratumorally adjuvant injections. However, the current fabrication strategy for achieving a sustainable immune response to ISV remains a pressing challenge. In this study, we present an empowered sustainable ISV method for antitumor therapy using 177Lu-labeled manganese-doped mesoporous hydroxyapatite (177Lu/Mn-HAP) microspheres. The ISV enables the sustained utilization of tumor antigens, leading to the activation of dendritic cells and polarization of macrophages toward the M1 subtype. Consequently, it facilitates the generation of potent CD8+ T-cell responses, enhancing the antitumor effects of internal radiation in both primary and distant tumors. Importantly, this approach achieves complete remission in all tumor-bearing mice and stimulates immune memory to prevent tumor recurrence. Our study highlights a universal and safe ISV strategy capable of inducing potent tumor-specific and sustainable immune response.


Subject(s)
Cancer Vaccines , Durapatite , Microspheres , Durapatite/chemistry , Animals , Mice , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , CD8-Positive T-Lymphocytes/immunology , Vaccination , Female , Mice, Inbred C57BL , Radioisotopes/chemistry , Cell Line, Tumor
19.
J Am Chem Soc ; 146(28): 19218-19228, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38955767

ABSTRACT

The messenger RNA (mRNA) vaccines hold great significance in contagion prevention and cancer immunotherapy. However, safely and effectively harnessing innate immunity to stimulate robust and durable adaptive immune protection is crucial, yet challenging. In this study, we synthesized a library of stimuli-responsive bivalent ionizable lipids (srBiv iLPs) with smart molecular blocks responsive to esterase, H2O2, cytochrome P450, alkaline phosphatase, nitroreductase, or glutathione (GSH), aiming to leverage physiological cues to trigger fast lipid degradation, promote mRNA translation, and induce robust antitumor immunity via reactive oxygen species (ROS)-mediated boosting. After subcutaneous immunization, esterase-responsive vaccine (eBiv-mVac) was rapidly internalized and transported into the draining lymph nodes. It then underwent fast decaging and self-immolative degradation in esterase-rich antigen-presenting cells, releasing sufficient mRNA for antigen translation and massive reactive quinone methides to elevate ROS levels. This resulted in broad activation of innate immunity to boost T cell response, prompting a large number of primed antigen-specific CD8+ T cells to circulate and infiltrate into tumors (>1000-fold versus unvaccinated control), thereby orchestrating innate and adaptive immunity to control tumor growth. Moreover, by further combining our vaccination strategy with immune checkpoint blockade, we demonstrated a synergism that significantly amplified the magnitude and function of antigen-specific CD8+ T cells. This, in turn, caused potent systemic antitumor efficacy and prolonged survival with high complete response rate in xenograft and metastasis models. Overall, our generalized stimuli-responsive mRNA delivery platform promises a paradigm shift in the design of potent vaccines for cancer immunotherapy, as well as effective and precise carriers for gene editing, protein replacement, and cell engineering.


Subject(s)
CD8-Positive T-Lymphocytes , Immunity, Innate , Reactive Oxygen Species , Reactive Oxygen Species/metabolism , Immunity, Innate/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , mRNA Vaccines/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , RNA, Messenger/immunology , RNA, Messenger/genetics , Humans , Mice, Inbred C57BL
20.
PLoS One ; 19(7): e0307204, 2024.
Article in English | MEDLINE | ID: mdl-39008481

ABSTRACT

Malignant pleural mesothelioma (MPM) is an aggressive cancer with a very poor prognosis. Recently, immune checkpoint inhibition (ICI) has taken center stage in the currently ongoing revolution that is changing standard-of-care treatment for several malignancies, including MPM. As multiple arguments and accumulating lines of evidence are in support of the existence of a therapeutic synergism between chemotherapy and immunotherapy, as well as between different classes of immunotherapeutics, we designed a multicenter, single-arm, phase I/II trial in which both programmed-death-ligand 1 (PD-L1) inhibition and dendritic cell (DC) vaccination are integrated in the first-line conventional platinum/pemetrexed-based treatment scheme for epithelioid MPM patients (Immuno-MESODEC, ClinicalTrials.gov identifier NCT05765084). Fifteen treatment-naïve patients with unresectable epithelioid subtype MPM will be treated with four 3-weekly (±3 days) chemo-immunotherapy cycles. Standard-of-care chemotherapy consisting of cisplatinum (75mg/m2) and pemetrexed (500mg/m2) will be supplemented with the anti-PD-L1 antibody atezolizumab (1200 mg) and autologous Wilms' tumor 1 mRNA-electroporated dendritic cell (WT1/DC) vaccination (8-10 x 106 cells/vaccination). Additional atezolizumab (1680 mg) doses and/or WT1/DC vaccinations (8-10 x 106 cells/vaccination) can be administered optionally following completion of the chemo-immunotherapy scheme. Follow-up of patients will last for up to 90 days after final atezolizumab administration and/or WT1/DC vaccination or 24 months after diagnosis, whichever occurs later. The trial's primary endpoints are safety and feasibility, secondary endpoints are clinical efficacy and immunogenicity. This phase I/II trial will evaluate whether addition of atezolizumab and WT1/DC vaccination to frontline standard-of-care chemotherapy for the treatment of epithelioid MPM is feasible and safe. If so, this novel combination strategy should be further investigated as a promising advanced treatment option for this hard-to-treat cancer.


Subject(s)
Antibodies, Monoclonal, Humanized , B7-H1 Antigen , Cancer Vaccines , Dendritic Cells , Mesothelioma, Malignant , Humans , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/immunology , Dendritic Cells/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Male , Female , WT1 Proteins/immunology , Pleural Neoplasms/immunology , Pleural Neoplasms/drug therapy , Pleural Neoplasms/therapy , Immunotherapy/methods , Middle Aged , Adult , Immune Checkpoint Inhibitors/therapeutic use , Aged , Vaccination , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pemetrexed/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mesothelioma/drug therapy , Mesothelioma/immunology , Mesothelioma/therapy , Cisplatin/therapeutic use , Cisplatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL