Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Chem Sci ; 14(13): 3501-3513, 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37006677

ABSTRACT

The overexpression of aberrantly glycosylated tumor-associated mucin-1 (TA-MUC1) in human cancers makes it a major target for the development of anticancer vaccines derived from synthetic MUC1-(glyco)peptide antigens. However, glycopeptide-based subunit vaccines are weakly immunogenic, requiring adjuvants and/or additional immunopotentiating approaches to generate optimal immune responses. Among these strategies, unimolecular self-adjuvanting vaccine constructs that do not need coadministration of adjuvants or conjugation to carrier proteins emerge as a promising but still underexploited approach. Herein, we report the design, synthesis, immune-evaluation in mice, and NMR studies of new, self-adjuvanting and self-assembling vaccines based on our QS-21-derived minimal adjuvant platform covalently linked to TA-MUC1-(glyco)peptide antigens and a peptide helper T-cell epitope. We have developed a modular, chemoselective strategy that harnesses two distal attachment points on the saponin adjuvant to conjugate the respective components in unprotected form and high yields via orthogonal ligations. In mice, only tri-component candidates but not unconjugated or di-component combinations induced significant TA-MUC1-specific IgG antibodies able to recognize the TA-MUC1 on cancer cells. NMR studies revealed the formation of self-assembled aggregates, in which the more hydrophilic TA-MUC1 moiety gets exposed to the solvent, favoring B-cell recognition. While dilution of the di-component saponin-(Tn)MUC1 constructs resulted in partial aggregate disruption, this was not observed for the more stably-organized tri-component candidates. This higher structural stability in solution correlates with their increased immunogenicity and suggests a longer half-life of the construct in physiological media, which together with the enhanced antigen multivalent presentation enabled by the particulate self-assembly, points to this self-adjuvanting tri-component vaccine as a promising synthetic candidate for further development.

2.
Front Oncol ; 12: 956940, 2022.
Article in English | MEDLINE | ID: mdl-36059712

ABSTRACT

In vitro cell culture studies are common in the cancer research field, and reliable biomimetic 3D models are needed to ensure physiological relevance. In this manuscript, we hypothesized that decellularized xenograft tumors can serve as an optimal 3D substrate to generate a top-down approach for in vitro tumor modeling. Multiple tumor cell lines were xenografted and the formed solid tumors were recovered for their decellularization by several techniques and further characterization by histology and proteomics techniques. Selected decellularized tumor xenograft samples were seeded with the HCC1806 human triple-negative breast cancer (TNBC) basal-like subtype cell line, and cell behavior was compared among them and with other control 2D and 3D cell culture methods. A soft treatment using Freeze-EDTA-DNAse allows proper decellularization of xenografted tumor samples. Interestingly, proteomic data show that samples decellularized from TNBC basal-like subtype xenograft models had different extracellular matrix (ECM) compositions compared to the rest of the xenograft tumors tested. The in vitro recellularization of decellularized ECM (dECM) yields tumor-type-specific cell behavior in the TNBC context. Data show that dECM derived from xenograft tumors is a feasible substrate for reseeding purposes, thereby promoting tumor-type-specific cell behavior. These data serve as a proof-of-concept for further potential generation of patient-specific in vitro research models.

3.
ACS Appl Mater Interfaces ; 13(42): 49589-49601, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34643365

ABSTRACT

The incidence and mortality of cancer demand more innovative approaches and combination therapies to increase treatment efficacy and decrease off-target side effects. We describe a boron-rich nanoparticle composite with potential applications in both boron neutron capture therapy (BNCT) and photothermal therapy (PTT). Our strategy is based on gold nanorods (AuNRs) stabilized with polyethylene glycol and functionalized with the water-soluble complex cobalt bis(dicarbollide) ([3,3'-Co(1,2-C2B9H11)2]-), commonly known as COSAN. Radiolabeling with the positron emitter copper-64 (64Cu) enabled in vivo tracking using positron emission tomography imaging. 64Cu-labeled multifunctionalized AuNRs proved to be radiochemically stable and capable of being accumulated in the tumor after intravenous administration in a mouse xenograft model of gastrointestinal cancer. The resulting multifunctional AuNRs showed high biocompatibility and the capacity to induce local heating under external stimulation and trigger cell death in heterogeneous cancer spheroids as well as the capacity to decrease cell viability under neutron irradiation in cancer cells. These results position our nanoconjugates as suitable candidates for combined BNCT/PTT therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Boron Neutron Capture Therapy , Gold/pharmacology , Nanotubes/chemistry , Photothermal Therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Gold/administration & dosage , Gold/chemistry , Humans , Injections, Intravenous , Materials Testing , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Positron-Emission Tomography
4.
Chemistry ; 27(14): 4731-4737, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33236801

ABSTRACT

Adjuvants are key immunostimulatory components in vaccine formulations, which improve the immune response to the co-administered antigen. The saponin natural product QS-21 is one of the most promising immunoadjuvants in the development of vaccines against cancer and infectious diseases but suffers from limitations that have hampered its widespread human use. Previous structure-activity relationship studies have identified simplified saponin variants with truncated carbohydrate chains, but have not focused on the influence of the linear oligosaccharide domain of QS-21 in adjuvant activity. Herein, an expeditious 15-step synthesis of new linear trisaccharide variants of simplified QS-21-derived adjuvants is reported, in which the complex terminal xylose-rhamnose moiety has been replaced with commercially available, simpler lactose and cellobiose disaccharides in a ß-anomeric configuration. In vivo immunological evaluation of the synthetic saponins showed attenuated antibody responses, highlighting the negative impact of such carbohydrate modifications on adjuvant activity, which could be associated with higher saponin conformational flexibility.


Subject(s)
Adjuvants, Immunologic , Saponins , Disaccharides , Humans , Rhamnose , Xylose
5.
iScience ; 23(9): 101499, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32919370

ABSTRACT

Immunotherapy has yielded impressive results, but only for a minority of patients with cancer. Therefore, new approaches that potentiate immunotherapy are a pressing medical need. Ferroptosis is a newly described type of programmed cell death driven by iron-dependent phospholipid peroxidation via Fenton chemistry. Here, we developed iron oxide-loaded nanovaccines (IONVs), which, chemically programmed to integrate iron catalysis, drug delivery, and tracking exploiting the characteristics of the tumor microenvironment (TME), improves immunotherapy and activation of ferroptosis. The IONVs trigger danger signals and use molecular disassembly and reversible covalent bonds for targeted antigen delivery and improved immunostimulatory capacity and catalytic iron for targeting tumor cell ferroptosis. IONV- and antibody-mediated TME modulation interfaced with imaging was important toward achieving complete eradication of aggressive and established tumors, eliciting long-lived protective antitumor immunity with no toxicities. This work establishes the feasibility of using nanoparticle iron catalytic activity as a versatile and effective feature for enhancing immunotherapy.

6.
Immunol Rev ; 296(1): 169-190, 2020 07.
Article in English | MEDLINE | ID: mdl-32594569

ABSTRACT

Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.


Subject(s)
Adjuvants, Immunologic , Immunomodulation , Vaccination , Vaccines/immunology , Vaccines/therapeutic use , Animals , Antibody Formation/immunology , Autoimmunity , Disease Management , Humans , Immunity, Cellular , Immunity, Humoral , Molecular Targeted Therapy , Treatment Outcome , Vaccination/methods , Vaccines/administration & dosage
7.
Chem Sci ; 11(17): 4488-4498, 2020 Apr 08.
Article in English | MEDLINE | ID: mdl-34122907

ABSTRACT

Tumor associated carbohydrate antigens (TACAs), such as the Tn antigen, have emerged as key targets for the development of synthetic anticancer vaccines. However, the induction of potent and functional immune responses has been challenging and, in most cases, unsuccessful. Herein, we report the design, synthesis and immunological evaluation in mice of Tn-based vaccine candidates with multivalent presentation of the Tn antigen (up to 16 copies), both in its native serine-linked display (Tn-Ser) and as an oxime-linked Tn analogue (Tn-oxime). The high valent vaccine prototypes were synthesized through a late-stage convergent assembly (Tn-Ser construct) and a versatile divergent strategy (Tn-oxime analogue), using chemoselective click-type chemistry. The hexadecavalent Tn-oxime construct induced robust, Tn-specific humoral and CD4+/CD8+ cellular responses, with antibodies able to bind the Tn antigen on the MCF7 cancer cell surface. The superior synthetic accessibility and immunological properties of this fully-synthetic vaccine prototype makes it a compelling candidate for further advancement towards safe and effective synthetic anticancer vaccines.

8.
Chem Commun (Camb) ; 56(5): 719-722, 2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31833496

ABSTRACT

We report the design, synthesis, immunological evaluation, and conformational analysis of new saponin variants as promising vaccine adjuvants. These studies have provided expedient synthetic access to streamlined adjuvant-active saponins and yielded molecular-level insights into saponin conformation that correlated with their in vivo adjuvant activities.


Subject(s)
Adjuvants, Immunologic/pharmacology , Saponins/pharmacology , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/toxicity , Animals , Drug Design , Female , Immunity, Humoral/drug effects , Mice, Inbred C57BL , Molecular Conformation , Molecular Dynamics Simulation , Ovalbumin/immunology , Saponins/chemical synthesis , Saponins/toxicity , Structure-Activity Relationship
9.
Nanoscale ; 11(4): 1531-1537, 2019 Jan 23.
Article in English | MEDLINE | ID: mdl-30623961

ABSTRACT

Extracellular vesicles (EVs) are considered sophisticated vehicles for cell-to-cell communication, thanks to the possibility of handling a variable cargo in a shell with multiple types of decoders. Surface glycosylation of EVs is a method that could be used to control their interaction with different cells and, consequently, the biodistribution of the vesicles in the body. Herein, we produced EVs derived from mouse liver proliferative cells, and we treated them with neuraminidase, an enzyme that digests the terminal sialic acid residues from glycoproteins. Afterwards, we labeled the EVs directly with [124I]Na and injected them in mice intravenously or into the hock. The amount of radioactivity in major organs was measured at different time points after administration both in vivo using positron emission tomography and ex vivo (after animal sacrifice) using dissection and gamma counting. The results showed that intravenous injection leads to the rapid accumulation of EVs in the liver. Moreover, after some hours the distribution led to the presence of EVs in different organs including the brain. Glycosidase treatment induced an accumulation in the lungs, compared with the intact EVs. Furthermore, when the EVs were injected through the hock, the neuraminidase-treated vesicles distributed better at the axillary lymph nodes than the untreated EVs. This result shows that modification of the glycosylated complexes on the EV surface can affect the distribution of these vesicles, and specifically removing the sialic acid residues allows more EVs to reach and accumulate at the lungs.


Subject(s)
Extracellular Vesicles/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Extracellular Vesicles/chemistry , Glycosylation , Liver/diagnostic imaging , Liver/metabolism , Lung/diagnostic imaging , Lung/metabolism , Lymph Nodes/metabolism , Mice , Neuraminidase/metabolism , Positron-Emission Tomography , Sodium Iodide/chemistry , Sodium Iodide/metabolism , Tissue Distribution
10.
Small ; 15(4): e1803993, 2019 01.
Article in English | MEDLINE | ID: mdl-30569516

ABSTRACT

Despite the tremendous potential of Toll-like receptor 4 (TLR4) agonists in vaccines, their efficacy as monotherapy to treat cancer has been limited. Only some lipopolysaccharides (LPS) isolated from particular bacterial strains or structures like monophosphoryl lipid A (MPLA) derived from lipooligosaccharide (LOS), avoid toxic overactivation of innate immune responses while retaining adequate immunogenicity to act as adjuvants. Here, different LOS structures are incorporated into nanoparticle-filled phospholipid micelles for efficient vaccine delivery and more potent cancer immunotherapy. The structurally unique LOS of the plant pathogen Xcc is incorporated into phospholipid micelles encapsulating iron oxide nanoparticles, producing stable pathogen-mimicking nanostructures suitable for targeting antigen presenting cells in the lymph nodes. The antigen is conjugated via a hydrazone bond, enabling rapid, easy-to-monitor and high-yield antigen ligation at low concentrations. The protective effect of these constructs is investigated against a highly aggressive model for tumor immunotherapy. The results show that the nanovaccines lead to a higher-level antigen-specific cytotoxic T lymphocyte (CTL) effector and memory responses, which when combined with abrogation of the immunosuppressive programmed death-ligand 1 (PD-L1), provide 100% long-term protection against repeated tumor challenge. This nanovaccine platform in combination with checkpoint inhibition of PD-L1 represents a promising approach to improve the cancer immunotherapy of TLR4 agonists.


Subject(s)
Antigens/chemistry , B7-H1 Antigen/metabolism , Immunotherapy/methods , Magnetite Nanoparticles/chemistry , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Animals , Cell Line , Escherichia coli/metabolism , Ferric Compounds/chemistry , Flow Cytometry , Lipopolysaccharides/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neoplasms/metabolism , Neoplasms/therapy , Quantum Dots , T-Lymphocytes, Cytotoxic , Toll-Like Receptor 4/metabolism
11.
Biomaterials ; 170: 95-115, 2018 07.
Article in English | MEDLINE | ID: mdl-29656235

ABSTRACT

Encouraging results are emerging from systems that exploit Toll like receptor (TLR) signaling, nanotechnology, checkpoint inhibition and molecular imaging for cancer immunotherapy. A major remaining challenge is developing effective, durable and tumour-specific immune responses without systemic toxicity. Here, we report a simple and versatile system based on synergistic activation of immune responses and direct cancer cell killing by combined TLR ligation using polyIC as TLR3 and imiquimod (R837) as TLR7 agonist, in combination with the model antigen ovalbumin (OVA) and phospholipid micelles loaded with zinc-doped iron oxide magnetic nanoparticles (MNPs). The combination of TLR agonists triggered a strong innate immune response in the lymph nodes (LNs) without systemic release of pro-inflammatory cytokines. The vaccines showed excellent efficacy against aggressive B16-F10 melanoma cells expressing OVA, which was improved with immune checkpoint abrogation of the immunosuppressive programmed death-ligand 1 (PD-L1) at the level of the cancer cells. By magnetic resonance (MR) and nuclear imaging we could track the vaccine migration from the site of injection to LNs and tumour. Overall, we show this synergistic TLR agonists and their combination with MNPs and immune checkpoint blockade to have considerable potential for preclinical and clinical development of vaccines for cancer immunotherapy.


Subject(s)
Imiquimod/pharmacology , Immunotherapy , Magnetite Nanoparticles/chemistry , Nanotechnology , Neoplasms/immunology , Neoplasms/therapy , Poly I-C/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Cancer Vaccines/immunology , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Drug Synergism , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Imiquimod/therapeutic use , Immunity, Innate/drug effects , Immunization , Lymph Nodes/drug effects , Lymph Nodes/pathology , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice, Inbred C57BL , Neoplasms/diagnosis , Neoplasms/pathology , Phospholipids/chemistry , Poly I-C/therapeutic use , Polyethylene Glycols/chemistry
12.
Chem Sci ; 8(6): 4619-4625, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28626570

ABSTRACT

Encouraging developments demonstrate that few transition metal and organometallic catalysts can operate in a bioorthogonal fashion and promote non-natural chemistry in living systems by minimizing undesired side reactions with cellular components. These catalytic processes have potential for applications in medicinal chemistry and chemical biology. However, the stringent conditions of the cell environment severely limit the number of accessible metal catalysts and exogenous reactions. Herein, we report an unorthodox approach and a new type of bioorthogonal catalytic reaction, in which a metal complex is an unconventional substrate and an exogenous biological molecule acts as a catalyst. In this reaction, riboflavin photocatalytically converts a PtIV prodrug into cisplatin within the biological environment. Due to the catalytic activity of riboflavin, cisplatin-like apoptosis is induced in cancer cells under extremely low doses of light, potentially preventing systemic off-target reactions. Photocatalytic and bioorthogonal turnover of PtIV into PtII species is an attractive strategy to amplify the antineoplastic action of metal-based chemotherapeutics with spatio-temporal control.

13.
ACS Nano ; 10(1): 1602-18, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26678549

ABSTRACT

Development of vaccines to prevent and treat emerging new pathogens and re-emerging infections and cancer remains a major challenge. An attractive approach is to build the vaccine upon a biocompatible NP that simultaneously acts as accurate delivery vehicle and radiotracer for PET/SPECT imaging for ultrasensitive and quantitative in vivo imaging of NP delivery to target tissues/organs. Success in developing these nanovaccines will depend in part on having a "correct" NP size and accommodating and suitably displaying antigen and/or adjuvants (e.g., TLR agonists). Here we develop and evaluate a NP vaccine based on iron oxide-selective radio-gallium labeling suitable for SPECT((67)Ga)/PET((68)Ga) imaging and efficient delivery of antigen (OVA) and TLR 9 agonists (CpGs) using lipid-coated magnetite micelles. OVA, CpGs and rhodamine are easily accommodated in the hybrid micelles, and the average size of the construct can be controlled to be ca. 40 nm in diameter to target direct lymphatic delivery of the vaccine cargo to antigen presenting cells (APCs) in the lymph nodes (LNs). While the OVA/CpG-loaded construct showed effective delivery to endosomal TLR 9 in APCs, SPECT imaging demonstrated migration from the injection site to regional and nonregional LNs. In correlation with the imaging results, a range of in vitro and in vivo studies demonstrate that by using this microdosed nanosystem the cellular and humoral immune responses are greatly enhanced and provide protection against tumor challenge. These results suggest that these nanosystems have considerable potential for image-guided development of targeted vaccines that are more effective and limit toxicity.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Lymph Nodes/immunology , Melanoma, Experimental/prevention & control , Skin Neoplasms/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation , Antigens/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Dendritic Cells/pathology , Gallium Radioisotopes/administration & dosage , Gene Expression , Immunity, Cellular , Lymph Nodes/diagnostic imaging , Lymph Nodes/pathology , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Micelles , Oligodeoxyribonucleotides/administration & dosage , Ovalbumin/administration & dosage , Positron-Emission Tomography/methods , Rhodamines/administration & dosage , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
14.
Small ; 10(24): 5054-67, 2014 Dec 29.
Article in English | MEDLINE | ID: mdl-25123704

ABSTRACT

The success of nanoparticle-based therapies will depend in part on accurate delivery to target receptors and organs. There is, therefore, considerable potential in nanoparticles which achieve delivery of the right drug(s) using the right route of administration to the right location at the right time, monitoring the process by non-invasive molecular imaging. A challenge is harnessing immunotherapy via activation of Toll-like receptors (TLRs) for the development of vaccines against major infectious diseases and cancer. In immunotherapy, delivery of the vaccine components to lymph nodes (LNs) is essential for effective stimulation of the immune response. Although some promising advances have been made, delivering therapeutics to LNs remains challenging. It is here shown that iron-oxide nanoparticles can be engineered to combine in a single and small (<50 nm) nanocarrier complementary multimodal imaging features with the immunostimulatory activity of polyinosinic-polycytidylic acid (poly (I:C)). Whilst the fluorescence properties of the nanocarrier show effective delivery to endosomes and TLR3 in antigen presenting cells, MRI/SPECT imaging reveals effective delivery to LNs. Importantly, in vitro and in vivo studies show that, using this nanocarrier, the immunostimulatory activity of poly (I:C) is greatly enhanced. These nanocarriers have considerable potential for cancer diagnosis and the development of new targeted and programmable immunotherapies.


Subject(s)
Drug Carriers , Ferric Compounds/administration & dosage , Immune System/drug effects , Lymph Nodes/drug effects , Nanoparticles , RNA, Double-Stranded/administration & dosage , Animals , Cell Line , Immune System/immunology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...