Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819238

ABSTRACT

The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger natural killer (NK)-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is under-explored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles targeting SLAMF7 and Glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surface of solid tumors with SLAMF7, these nanovesicles directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD-1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific nanovesicles as an anticancer strategy with implications for designing next-generation targeted cancer therapies.

2.
Tissue Eng Regen Med ; 20(7): 1063-1077, 2023 12.
Article in English | MEDLINE | ID: mdl-37801227

ABSTRACT

BACKGROUND: Myocardial infarction (MI) leads to cardiomyocyte death, poor cardiac remodeling, and heart failure, making it a major cause of mortality and morbidity. To restore cardiac pumping function, induction of cardiomyocyte regeneration has become a focus of academic interest. The Hippo pathway is known to regulate cardiomyocyte proliferation and heart size, and its inactivation allows adult cardiomyocytes to re-enter the cell cycle. METHODS: In this study, we investigated whether exosomes from adipose-derived stem cells (ADSCs) could effectively transfer siRNA for the Hippo pathway regulator Salvador (SAV) into cardiomyocytes to induce cardiomyocyte regeneration in a mouse model of MI. RESULTS: Our results showed that exosomes loaded with SAV-siRNA effectively transferred siRNA into cardiomyocytes and induced cardiomyocyte re-entry into the cell cycle, while retaining the previously demonstrated therapeutic efficacy of ADSC-derived exosomes to improve post-infarction cardiac function through anti-fibrotic, pro-angiogenic, and other effects. CONCLUSIONS: Our findings suggest that siRNA delivery via ADSC-derived exosomes may be a promising approach for the treatment of MI.


Subject(s)
Exosomes , Myocardial Infarction , Mice , Animals , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Exosomes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocytes, Cardiac , Stem Cells
3.
ACS Nano ; 17(17): 16770-16786, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37624742

ABSTRACT

Paclitaxel (PTX)-based chemotherapy remains the main approach to treating lung cancer but systemic toxicity limits its use. As chimeric antigen receptor-T (CAR-T) cell-derived exosomes contain tumor-targeted CARs and cytotoxic granules (granzyme B and perforin), they are considered potential delivery vehicles for PTX. However, the low drug-loading capacity and hepatotropic properties of exosomes are obstacles to their application to extrahepatic cancer. Here, a hybrid nanovesicle named Lip-CExo@PTX was designed for immunochemotherapy of lung cancer by fusing exosomes derived from bispecific CAR-T cells targeting both mesothelin (MSLN) and programmed death ligand-1 (PD-L1) with lung-targeted liposomes. Due to the lung-targeting ability of the liposomes, over 95% of intravenously administered Lip-CExo@PTX accumulated in lung tissue. In addition, with the help of the anti-MSLN single-chain variable fragment (scFv), the PTX and cytotoxic granules inside Lip-CExo@PTX were further delivered into MSLN-positive tumors. Notably, the anti-PD-L1 scFv on Lip-CExo@PTX blocked PD-L1 on the tumors to avoid T cell exhaustion and promoted PTX-induced immunogenic cell death. Furthermore, Lip-CExo@PTX prolonged the survival time of tumor-bearing mice in a CT-26 metastatic lung cancer model. Therefore, Lip-CExo@PTX may deliver PTX to tumor cells through sequential targeted delivery and enhance the antitumor effects, providing a promising strategy for immunochemotherapy of lung cancer.


Subject(s)
Exosomes , Lung Neoplasms , Receptors, Chimeric Antigen , Animals , Mice , Liposomes , T-Lymphocytes , Lung Neoplasms/drug therapy , Paclitaxel
4.
Mol Cancer ; 22(1): 131, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563723

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) -T cell therapy is an efficient therapeutic strategy for specific hematologic malignancies. However, positive outcomes of this novel therapy in treating solid tumors are curtailed by the immunosuppressive tumor microenvironment (TME), wherein signaling of the checkpoint programmed death-1 (PD-1)/PD-L1 directly inhibits T-cell responses. Although checkpoint-targeted immunotherapy succeeds in increasing the number of T cells produced to control tumor growth, the desired effect is mitigated by the action of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) in the TME. Previous studies have confirmed that targeting triggering-receptor-expressed on myeloid cells 2 (TREM2) on TAMs and MDSCs enhances the outcomes of anti-PD-1 immunotherapy. METHODS: We constructed carcinoembryonic antigen (CEA)-specific CAR-T cells for colorectal cancer (CRC)-specific antigens with an autocrine PD-1-TREM2 single-chain variable fragment (scFv) to target the PD-1/PD-L1 pathway, MDSCs and TAMs. RESULTS: We found that the PD-1-TREM2-targeting scFv inhibited the activation of the PD-1/PD-L1 pathway. In addition, these secreted scFvs blocked the binding of ligands to TREM2 receptors present on MDSCs and TAMs, reduced the proportion of MDSCs and TAMs, and enhanced T-cell effector function, thereby mitigating immune resistance in the TME. PD-1-TREM2 scFv-secreting CAR-T cells resulted in highly effective elimination of tumors compared to that achieved with PD-1 scFv-secreting CAR-T therapy in a subcutaneous CRC mouse model. Moreover, the PD-1-TREM2 scFv secreted by CAR-T cells remained localized within tumors and exhibited an extended half-life. CONCLUSIONS: Together, these results indicate that PD-1-TREM2 scFv-secreting CAR-T cells have strong potential as an effective therapy for CRC.


Subject(s)
Colorectal Neoplasms , Immunotherapy, Adoptive , Single-Chain Antibodies , Animals , Mice , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/therapy , Colorectal Neoplasms/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , T-Lymphocytes , Tumor Microenvironment
5.
J Transl Med ; 21(1): 383, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37308954

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is a worldwide health threat with high annual morbidity and mortality. Chemotherapeutic drugs such as paclitaxel (PTX) have been widely applied clinically. However, systemic toxicity due to the non-specific circulation of PTX often leads to multi-organ damage, including to the liver and kidney. Thus, it is necessary to develop a novel strategy to enhance the targeted antitumor effects of PTX. METHODS: Here, we engineered exosomes derived from T cells expressing the chimeric antigen receptor (CAR-Exos), which targeted mesothelin (MSLN)-expressing Lewis lung cancer (MSLN-LLC) through the anti-MSLN single-chain variable fragment (scFv) of CAR-Exos. PTX was encapsulated into CAR-Exos (PTX@CAR-Exos) and administered via inhalation to an orthotopic lung cancer mouse model. RESULTS: Inhaled PTX@CAR-Exos accumulated within the tumor area, reduced tumor size, and prolonged survival with little toxicity. In addition, PTX@CAR-Exos reprogrammed the tumor microenvironment and reversed the immunosuppression, which was attributed to infiltrating CD8+ T cells and elevated IFN-γ and TNF-α levels. CONCLUSIONS: Our study provides a nanovesicle-based delivery platform to promote the efficacy of chemotherapeutic drugs with fewer side effects. This novel strategy may ameliorate the present obstacles to the clinical treatment of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Exosomes , Lung Neoplasms , Animals , Mice , Paclitaxel , CD8-Positive T-Lymphocytes , Tumor Microenvironment
6.
Mol Ther ; 31(8): 2376-2390, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37312452

ABSTRACT

Induced pluripotent stem cells (iPSCs) express a broad spectrum of tumor-associated antigens and exert prophylactic effects on various tumors. However, some problems remain, such as potential tumorigenicity, challenges in transport to the lymph nodes and spleen, and limited antitumor effects. Thus, designing a safe and effective iPSC-based tumor vaccine is necessary. We prepared iPSC-derived exosomes and incubated them with DCs (dendritic cells) for pulsing to explore their antitumor effects in murine melanoma models. The antitumor immune response induced by the DC vaccine pulsed with iPSC exosomes (DC + EXO) was assessed in vitro and in vivo. After DC + EXO vaccination, extracted spleen T cells effectively killed a variety of tumor cells (melanoma, lung cancer, breast cancer, and colorectal cancer) in vitro. In addition, DC + EXO vaccination significantly inhibited melanoma growth and lung metastasis in mouse models. Furthermore, DC + EXO vaccination induced long-term T cell responses and prevented melanoma rechallenge. Finally, biocompatibility studies showed that the DC vaccine did not significantly alter the viability of normal cells and mouse viscera. Hence, our research may provide a prospective strategy of a safe and effective iPSC-based tumor vaccine for clinical use.


Subject(s)
Cancer Vaccines , Exosomes , Induced Pluripotent Stem Cells , Lung Neoplasms , Melanoma , Mice , Animals , Mice, Inbred C57BL , Melanoma/therapy , Immunity, Cellular , Dendritic Cells
7.
Adv Mater ; 35(19): e2211138, 2023 May.
Article in English | MEDLINE | ID: mdl-36814099

ABSTRACT

Chimeric antigen receptor-T (CAR-T) cell therapy has shown remarkable success in eradicating hematologic malignancies; however, its efficacy in treating solid tumors has always been limited due to the presence of an immune-suppressive tumor microenvironment (TME). Here, genetically programmable cellular vesicles expressing high-affinity anti-programmed death-ligand 1 single chain variable fragment (anti-PD-L1 scFv) loaded with glutamine antagonist (D@aPD-L1 NVs) are developed to metabolically dismantle the immunosuppressive TME and enhance the efficiency of anti-mesothelin CAR-T cells in orthotopic lung cancer. As anti-PD-L1 scFv can specifically bind to the programmed death-ligand 1 (PD-L1) on tumor cells, D@aPD-L1 NVs enable the targeted delivery of glutamine antagonists to the tumor site and address the upregulation of PD-L1 on tumor cells, which prevents the premature exhaustion of CAR-T cells. More importantly, D@aPD-L1 NVs effectively reduce the number of immunosuppressive cells and promote the recruitment of inflammatory cells and the secretion of inflammatory cytokines in tumor tissues. These unique features of D@aPD-L1 NVs improve the infiltration and effector functions of CAR-T cells, which ultimately enhance the anti-tumor ability and long-term memory immunity of CAR-T cells. The findings support that D@aPD-L1 NVs act as a promising drug to strengthen the effectiveness of CAR-T cells against solid tumors.


Subject(s)
Receptors, Chimeric Antigen , Receptors, Chimeric Antigen/metabolism , Receptors, Antigen, T-Cell , T-Lymphocytes , Glutamine/metabolism , Cell Line, Tumor , Tumor Microenvironment
8.
Acta Biomater ; 157: 451-466, 2023 02.
Article in English | MEDLINE | ID: mdl-36442821

ABSTRACT

Immune checkpoint blockade therapy targeting programmed death-1 (PD-1) or its major ligand programmed death-ligand 1 (PD-L1) has achieved remarkable success in the treatment of several tumors, including colorectal cancer. However, the efficacy of PD-1/PD-L1 inhibitors is limited in some colorectal cancers within the immunosuppressive tumor microenvironment (such as when there is a lack of immune cell infiltration). Herein, anti-PD-L1 functionalized biomimetic polydopamine-modified gold nanostar nanoparticles (PDA/GNS@aPD-L1 NPs) were developed for synergistic anti-tumor treatment by combining PD-1/PD-L1 blockade with photothermal ablation. PDA/GNS@aPD-L1 NPs were prepared by encapsulating photothermal nanoparticles (polydopamine-modified gold nanostar, PDA-GNS) with cell membrane isolated from anti-PD-L1 single-chain variable fragment (scFv) over-expressing cells. In addition to disrupting PD-1/PD-L1 immunosuppressive signals, the anti-PD-L1 scFv on the membrane of PDA/GNS@aPD-L1 NPs was conducive to the accumulation of PDA-GNS at tumor sites. Importantly, the tumor photothermal ablation induced by PDA-GNS could reverse the immunosuppressive tumor microenvironment, thereby further improving the efficiency of PD-1/PD-L1 blockade therapy. In this study, the synthetized PDA/GNS@aPD-L1 NPs exhibited good biocompatibility, efficient photothermal conversion ability, and enhanced tumor-targeting ability. In vivo studies revealed that a PDA/GNS@aPD-L1 NP-based therapeutic strategy significantly inhibited tumor growth, and prolonged overall survival by further promoting the maturation of dendritic cells (DCs), increasing the infiltration of CD8+T cells, and decreasing the number of immunosuppressive cells (such as regulatory T cells and myeloid-derived suppressive cells). Collectively, the developed PDA/GNS@aPD-L1 NP-based therapeutic strategy combines PD-1/PD-L1 blockade with photothermal ablation, which could remodel the tumor microenvironment for effective clinical colorectal cancer therapy. STATEMENT OF SIGNIFICANCE: Immunosuppressive tumor microenvironment is the main challenge facing programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) blockade therapy. By encapsulating photothermal nanoparticles (polydopamine-modified gold nanostar, PDA-GNS) with cell membrane over-expressing anti-PD-L1 single-chain variable fragment, we constructed anti-PD-L1 functionalized biomimetic nanoparticles (PDA/GNS@aPD-L1 NPs). By specific binding to the PD-L1 present on tumor cells, PDA/GNS@aPD-L1 NPs could disrupt PD-1/PD-L1 immunosuppression signaling, and effectively deliver PDA-GNS targeting to tumor sites. Additionally, PDA-GNS-mediated local photothermal ablation of tumors promoted the release of tumor-associated antigens and thus activated anti-tumor immune responses. Meanwhile, hyperthermia facilitates immune cell infiltration by increasing tumor vascular permeability. Therefore, PDA/GNS@aPD-L1 NPs could sensitize tumors to PD-1/PD-L1 blockade therapy by remodeling the immunosuppressive tumor microenvironment, which provides a new strategy for tumor treatment.


Subject(s)
Colorectal Neoplasms , Nanoparticles , Single-Chain Antibodies , Humans , Programmed Cell Death 1 Receptor/metabolism , Biomimetics , Ligands , Immunotherapy , Colorectal Neoplasms/metabolism , Gold/pharmacology , Cell Line, Tumor , Tumor Microenvironment
9.
J Nanobiotechnology ; 19(1): 391, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34823562

ABSTRACT

BACKGROUND: Considering the threat of the COVID-19 pandemic, caused by SARS-CoV-2, there is an urgent need to develop effective treatments. At present, neutralizing antibodies and small-molecule drugs such as remdesivir, the most promising compound to treat this infection, have attracted considerable attention. However, some potential problems need to be concerned including viral resistance to antibody-mediated neutralization caused by selective pressure from a single antibody treatment, the unexpected antibody-dependent enhancement (ADE) effect, and the toxic effect of small-molecule drugs. RESULTS: Here, we constructed a type of programmed nanovesicle (NV) derived from bispecific CAR-T cells that express two single-chain fragment variables (scFv), named CR3022 and B38, to target SARS-CoV-2. Nanovesicles that express both CR3022 and B38 (CR3022/B38 NVs) have a stronger ability to neutralize Spike-pseudovirus infectivity than nanovesicles that express either CR3022 or B38 alone. Notably, the co-expression of CR3022 and B38, which target different epitopes of spike protein, could reduce the incidence of viral resistance. Moreover, the lack of Fc fragments on the surface of CR3022/B38 NVs could prevent ADE effects. Furthermore, the specific binding ability to SARS-CoV-2 spike protein and the drug loading capacity of CR3022/B38 NVs can facilitate targeted delivery of remdesiver to 293 T cells overexpressing spike protein. These results suggest that CR3022/B38 NVs have the potential ability to target antiviral drugs to the main site of viral infection, thereby enhancing the antiviral ability by inhibiting intracellular viral replication and reducing adverse drug reactions. CONCLUSIONS: In summary, we demonstrate that nanovesicles derived from CAR-T cells targeting the spike protein of SARS-COV-2 have the ability to neutralize Spike-pseudotyped virus and target antiviral drugs. This novel therapeutic approach may help to solve the dilemma faced by neutralizing antibodies and small-molecule drugs in the treatment of COVID-19.


Subject(s)
COVID-19/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antiviral Agents/therapeutic use , COVID-19/immunology , Humans , Models, Theoretical
10.
J Nanobiotechnology ; 19(1): 173, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34112203

ABSTRACT

BACKGROUND: The worldwide pandemic of COVID-19 remains a serious public health menace as the lack of efficacious treatments. Cytokine storm syndrome (CSS) characterized with elevated inflammation and multi-organs failure is closely correlated with the bad outcome of COVID-19. Hence, inhibit the process of CSS by controlling excessive inflammation is considered one of the most promising ways for COVID-19 treatment. RESULTS: Here, we developed a biomimetic nanocarrier based drug delivery system against COVID-19 via anti-inflammation and antiviral treatment simultaneously. Firstly, lopinavir (LPV) as model antiviral drug was loaded in the polymeric nanoparticles (PLGA-LPV NPs). Afterwards, macrophage membranes were coated on the PLGA-LPV NPs to constitute drugs loaded macrophage biomimetic nanocarriers (PLGA-LPV@M). In the study, PLGA-LPV@M could neutralize multiple proinflammatory cytokines and effectively suppress the activation of macrophages and neutrophils. Furthermore, the formation of NETs induced by COVID-19 patients serum could be reduced by PLGA-LPV@M as well. In a mouse model of coronavirus infection, PLGA-LPV@M exhibited significant targeted ability to inflammation sites, and superior therapeutic efficacy in inflammation alleviation and tissues viral loads reduction. CONCLUSION: Collectively, such macrophage biomimetic nanocarriers based drug delivery system showed favorable anti-inflammation and targeted antiviral effects, which may possess a comprehensive therapeutic value in COVID-19 treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Biomimetics , COVID-19 Drug Treatment , Cytokine Release Syndrome/prevention & control , Drug Carriers , Inflammation/prevention & control , Nanoparticles , SARS-CoV-2/drug effects , COVID-19/virology , Cytokine Release Syndrome/etiology , Humans , Inflammation/complications , SARS-CoV-2/isolation & purification
11.
ACS Appl Mater Interfaces ; 13(18): 20995-21006, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33930273

ABSTRACT

COVID-19 has been diffusely pandemic around the world, characterized by massive morbidity and mortality. One of the remarkable threats associated with mortality may be the uncontrolled inflammatory processes, which were induced by SARS-CoV-2 in infected patients. As there are no specific drugs, exploiting safe and effective treatment strategies is an instant requirement to dwindle viral damage and relieve extreme inflammation simultaneously. Here, highly biocompatible glycyrrhizic acid (GA) nanoparticles (GANPs) were synthesized based on GA. In vitro investigations revealed that GANPs inhibit the proliferation of the murine coronavirus MHV-A59 and reduce proinflammatory cytokine production caused by MHV-A59 or the N protein of SARS-CoV-2. In an MHV-A59-induced surrogate mouse model of COVID-19, GANPs specifically target areas with severe inflammation, such as the lungs, which appeared to improve the accumulation of GANPs and enhance the effectiveness of the treatment. Further, GANPs also exert antiviral and anti-inflammatory effects, relieving organ damage and conferring a significant survival advantage to infected mice. Such a novel therapeutic agent can be readily manufactured into feasible treatment for COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Glycyrrhizic Acid/therapeutic use , Inflammation/drug therapy , Nanoparticles/therapeutic use , Virus Diseases/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Antioxidants/therapeutic use , Antiviral Agents/chemistry , Coronavirus Nucleocapsid Proteins/pharmacology , Cytokines/metabolism , Female , Glycyrrhizic Acid/chemistry , Humans , Liver/pathology , Lung/pathology , Mice , Mice, Inbred BALB C , Murine hepatitis virus/drug effects , Nanoparticles/chemistry , Phosphoproteins/pharmacology , RAW 264.7 Cells , SARS-CoV-2/chemistry , THP-1 Cells , Viral Load/drug effects , Virus Diseases/pathology , Virus Replication/drug effects , COVID-19 Drug Treatment
12.
Anal Biochem ; 556: 112-118, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29966589

ABSTRACT

Dengue is the most prevalent mosquito-borne viral disease in tropical and subtropical regions worldwide. Since its clinical symptoms are non-specific and easily mistaken as other kinds of infection, laboratory diagnosis is required to confirm dengue infections. In this study, ten peptides (E1-E10) from the envelope protein of dengue virus (DENV) were first identified using bioinformatic tool. The screened peptides were then synthesized for the peptide-based chemiluminescence enzyme immunoassay (CLEIA). Two peptides, E1 and E7, were found as the best candidate antigen and therefore used as downstream application in the development of low-cost peptide-based anti-DENV immunoglobulin M antibodies (IgM) indirect CLEIA. 176 serum samples were used to study the presence of anti-DENV IgM antibodies to evaluate the diagnostic ability of IgM-CLEIA. Receiver operating characteristic curve (ROC) was used to estimate the diagnostic cut-off value. The sensitivity and the specificity reached 82.5% and 94.6% respectively when peptide E1 was used, but declined to 79.2% and 92.9% respectively when peptide E7 was used. Therefore, the combination of E1 and E7 was used to improve the sensitivity and the specificity to 85.0% and 96.4% respectively in 1.5 h assay time, providing a potentially practical use for the diagnosis of DENV infections in patients' serum.


Subject(s)
Antibodies, Viral/blood , Dengue Virus/chemistry , Dengue/blood , Immunoglobulin M/chemistry , Luminescent Measurements/methods , Peptides/chemistry , Viral Proteins/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...