Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 226(2): 319-323, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35262728

ABSTRACT

The protozoan pathogen Giardia lamblia is an important worldwide cause of diarrheal disease and malabsorption. Infection is managed with antimicrobials, although drug resistance and treatment failures are a clinical challenge. Prior infection provides significant protection, yet a human vaccine has not been realized. Individual antigens can elicit partial protection in experimental models, but protection is weaker than after prior infection. Here, we developed a multivalent nanovaccine by coating membranes derived from the parasite onto uniform and stable polymeric nanoparticles loaded with a mucosal adjuvant. Intranasal immunization with the nanovaccine induced adaptive immunity and effectively protected mice from G. lamblia infection.


Subject(s)
Giardia lamblia , Giardiasis , Nanoparticles , Parasites , Adjuvants, Immunologic , Animals , Giardiasis/parasitology , Giardiasis/prevention & control , Humans , Immunity, Mucosal , Mice
2.
Leukemia ; 36(4): 994-1005, 2022 04.
Article in English | MEDLINE | ID: mdl-34845316

ABSTRACT

Cancer vaccines are promising treatments to prevent relapse after chemotherapy in acute myeloid leukemia (AML) patients, particularly for those who cannot tolerate intensive consolidation therapies. Here, we report the development of an AML cell membrane-coated nanoparticle (AMCNP) vaccine platform, in which immune-stimulatory adjuvant-loaded nanoparticles are coated with leukemic cell membrane material. This AMCNP vaccination strategy stimulates leukemia-specific immune responses by co-delivering membrane-associated antigens along with adjuvants to antigen-presenting cells. To demonstrate that this AMCNP vaccine enhances leukemia-specific antigen presentation and T cell responses, we modified a murine AML cell line to express membrane-bound chicken ovalbumin as a model antigen. AMCNPs were efficiently acquired by antigen-presenting cells in vitro and in vivo and stimulated antigen cross-presentation. Vaccination with AMCNPs significantly enhanced antigen-specific T cell expansion and effector function compared with control vaccines. Prophylactic vaccination with AMCNPs enhanced cellular immunity and protected against AML challenge. Moreover, in an AML post-remission vaccination model, AMCNP vaccination significantly enhanced survival in comparison to vaccination with whole leukemia cell lysates. Collectively, AMCNPs retained AML-specific antigens, elicited enhanced antigen-specific immune responses, and provided therapeutic benefit against AML challenge.


Subject(s)
Cancer Vaccines , Leukemia, Myeloid, Acute , Nanoparticles , Animals , Antigen Presentation , Cell Membrane , Humans , Immunotherapy , Leukemia, Myeloid, Acute/drug therapy , Mice , Vaccination
3.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34272285

ABSTRACT

Programmed cell death protein-1 (PD-1) expressed on activated T cells inhibits T cell function and proliferation to prevent an excessive immune response, and disease can result if this delicate balance is shifted in either direction. Tumor cells often take advantage of this pathway by overexpressing the PD-1 ligand PD-L1 to evade destruction by the immune system. Alternatively, if there is a decrease in function of the PD-1 pathway, unchecked activation of the immune system and autoimmunity can result. Using a combination of computation and experiment, we designed a hyperstable 40-residue miniprotein, PD-MP1, that specifically binds murine and human PD-1 at the PD-L1 interface with a Kd of ∼100 nM. The apo crystal structure shows that the binder folds as designed with a backbone RMSD of 1.3 Što the design model. Trimerization of PD-MP1 resulted in a PD-1 agonist that strongly inhibits murine T cell activation. This small, hyperstable PD-1 binding protein was computationally designed with an all-beta interface, and the trimeric agonist could contribute to treatments for autoimmune and inflammatory diseases.


Subject(s)
B7-H1 Antigen/chemistry , Programmed Cell Death 1 Receptor/agonists , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , B7-H1 Antigen/chemical synthesis , B7-H1 Antigen/immunology , B7-H1 Antigen/pharmacology , Computational Biology , Drug Design , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
Adv Mater ; 32(30): e2001808, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32538494

ABSTRACT

The recent success of immunotherapies has highlighted the power of leveraging the immune system in the fight against cancer. In order for most immune-based therapies to succeed, T cell subsets with the correct tumor-targeting specificities must be mobilized. When such specificities are lacking, providing the immune system with tumor antigen material for processing and presentation is a common strategy for stimulating antigen-specific T cell populations. While straightforward in principle, experience has shown that manipulation of the antigen presentation process can be incredibly complex, necessitating sophisticated strategies that are difficult to translate. Herein, the design of a biomimetic nanoparticle platform is reported that can be used to directly stimulate T cells without the need for professional antigen-presenting cells. The nanoparticles are fabricated using a cell membrane coating derived from cancer cells engineered to express a co-stimulatory marker. Combined with the peptide epitopes naturally presented on the membrane surface, the final formulation contains the necessary signals to promote tumor antigen-specific immune responses, priming T cells that can be used to control tumor growth. The reported approach represents an emerging strategy that can be used to develop multiantigenic, personalized cancer immunotherapies.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Cell Membrane/metabolism , Engineering , Nanomedicine/methods , Nanoparticles/chemistry , Cell Line, Tumor , Humans , Immunotherapy
5.
Adv Mater ; 32(13): e1901255, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31206841

ABSTRACT

While traditional approaches for disease management in the era of modern medicine have saved countless lives and enhanced patient well-being, it is clear that there is significant room to improve upon the current status quo. For infectious diseases, the steady rise of antibiotic resistance has resulted in super pathogens that do not respond to most approved drugs. In the field of cancer treatment, the idea of a cure-all silver bullet has long been abandoned. As a result of the challenges facing current treatment and prevention paradigms in the clinic, there is an increasing push for personalized therapeutics, where plans for medical care are established on a patient-by-patient basis. Along these lines, vaccines, both against bacteria and tumors, are a clinical modality that could benefit significantly from personalization. Effective vaccination strategies could help to address many challenging disease conditions, but current vaccines are limited by factors such as a lack of potency and antigenic breadth. Recently, researchers have turned toward the use of biomimetic nanotechnology as a means of addressing these hurdles. Recent progress in the development of biomimetic nanovaccines for antibacterial and anticancer applications is discussed, with an emphasis on their potential for personalized medicine.


Subject(s)
Nanomedicine/methods , Precision Medicine/methods , Vaccination/methods , Animals , Bacterial Infections/prevention & control , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/therapeutic use , Biomimetic Materials/chemistry , Biomimetics/methods , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Drug Delivery Systems/methods , Humans , Nanoparticles/chemistry , Nanotechnology/methods , Neoplasms/prevention & control
6.
Adv Biosyst ; 3(1): e1800219, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31728404

ABSTRACT

It is currently understood that, in order for a tumor to successfully grow, it must evolve means of evading immune surveillance. In the past several decades, researchers have leveraged increases in our knowledge of tumor immunology to develop therapies capable of augmenting endogenous immunity and eliciting strong antitumor responses. In particular, the goal of anticancer vaccination is to train the immune system to properly utilize its own resources in the fight against cancer. Although attractive in principle, there are currently only limited examples of anticancer vaccines that have been successfully translated to the clinic. Recently, there has been a significant push towards the use of nanotechnology for designing vaccine candidates that exhibit enhanced potency and specificity. In this progress report, we discuss recent developments in the field of anticancer nanovaccines. By taking advantage of the flexibility offered by nanomedicine to purposefully program immune responses, this new generation of vaccines has the potential to address many of the hurdles facing traditional platforms. A specific emphasis is placed on the emergence of cell membrane-coated nanoparticles, a novel biomimetic platform that can be used to generate personalized nanovaccines that elicit strong, multi-antigenic antitumor responses.

7.
Nano Lett ; 19(7): 4760-4769, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31184899

ABSTRACT

Infections caused by multidrug-resistant Gram-negative bacteria have emerged as a major threat to public health worldwide. The high mortality and prevalence, along with the slow pace of new antibiotic discovery, highlight the necessity for new disease management paradigms. Here, we report on the development of a multiantigenic nanotoxoid vaccine based on macrophage membrane-coated nanoparticles for eliciting potent immunity against pathogenic Pseudomonas aeruginosa. The design of this biomimetic nanovaccine leverages the specific role of macrophages in clearing pathogens and their natural affinity for various virulence factors secreted by the bacteria. It is demonstrated that the macrophage nanotoxoid is able to display a wide range of P. aeruginosa antigens, and the safety of the formulation is confirmed both in vitro and in vivo. When used to vaccinate mice via different administration routes, the nanotoxoid is capable of eliciting strong humoral immune responses that translate into enhanced protection against live bacterial infection in a pneumonia model. Overall, the work presented here provides new insights into the design of safe, multiantigenic antivirulence vaccines using biomimetic nanotechnology and the application of these nanovaccines toward the prevention of difficult-to-treat Gram-negative infections.


Subject(s)
Bacterial Vaccines , Drug Resistance, Bacterial , Pseudomonas Infections , Pseudomonas aeruginosa/immunology , Toxoids , Vaccination , Virulence Factors/immunology , Animals , Bacterial Vaccines/immunology , Bacterial Vaccines/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/immunology , Immunity, Humoral/drug effects , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/pathogenicity , Toxoids/immunology , Toxoids/pharmacology
8.
Adv Mater ; 30(23): e1706759, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29582476

ABSTRACT

Nanoparticle-based therapeutic, prevention, and detection modalities have the potential to greatly impact how diseases are diagnosed and managed in the clinic. With the wide range of nanomaterials available, the rational design of nanocarriers on an application-specific basis has become increasingly commonplace. Here, a comprehensive overview is provided on an emerging platform: cell-membrane-coating nanotechnology. As a fundamental unit of biology, cells carry out a wide range of functions, including the remarkable ability to interface and interact with their surrounding environment. Instead of attempting to replicate such functions via synthetic techniques, researchers are now directly leveraging naturally derived cell membranes as a means of bestowing nanoparticles with enhanced biointerfacing capabilities. This top-down technique is facile, highly generalizable, and has the potential to greatly augment existing nanocarriers. Further, the introduction of a natural membrane substrate onto nanoparticles surfaces has enabled additional applications beyond those traditionally associated with nanomedicine. Despite its relative youth, there exists an impressive body of literature on cell membrane coating, which is covered here in detail. Overall, there is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology.


Subject(s)
Cell Membrane , Drug Delivery Systems , Nanomedicine , Nanoparticles , Nanotechnology
9.
ACS Nano ; 12(1): 109-116, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29216423

ABSTRACT

Cardiovascular disease represents one of the major causes of death across the global population. Atherosclerosis, one of its most common drivers, is characterized by the gradual buildup of arterial plaque over time, which can ultimately lead to life-threatening conditions. Given the impact of the disease on public health, there is a great need for effective and noninvasive imaging modalities that can provide valuable information on its biological underpinnings during development. Here, we leverage the role of platelets in atherogenesis to design nanocarriers capable of targeting multiple biological elements relevant to plaque development. Biomimetic nanoparticles are prepared by coating platelet membrane around a synthetic nanoparticulate core, the product of which is capable of interacting with activated endothelium, foam cells, and collagen. The effects are shown to be exclusive to platelet membrane-coated nanoparticles. These biomimetic nanocarriers are not only capable of efficiently localizing to well-developed atherosclerotic plaque, but can also target subclinical regions of arteries susceptible to plaque formation. Using a commonly employed magnetic resonance imaging contrast agent, live detection is demonstrated using an animal model of atherosclerosis. Ultimately, this strategy may be leveraged to better assess the development of atherosclerosis, offering additional information to help clinicians better manage the disease.


Subject(s)
Atherosclerosis/diagnostic imaging , Atherosclerosis/metabolism , Blood Platelets/metabolism , Cell Membrane/metabolism , Coated Materials, Biocompatible/metabolism , Nanoparticles/metabolism , Animals , Atherosclerosis/pathology , Biomimetic Materials/metabolism , Cell Line , Foam Cells/metabolism , Foam Cells/pathology , Human Umbilical Vein Endothelial Cells , Humans , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Nanotechnology/methods , Optical Imaging/methods , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
10.
Adv Mater ; 29(47)2017 Dec.
Article in English | MEDLINE | ID: mdl-29239517

ABSTRACT

Anticancer vaccines train the body's own immune system to recognize and eliminate malignant cells based on differential antigen expression. While conceptually attractive, clinical efficacy is lacking given several key challenges stemming from the similarities between cancerous and healthy tissue. Ideally, an effective vaccine formulation would deliver multiple tumor antigens in a fashion that potently stimulates endogenous immune responses against those antigens. Here, it is reported on the fabrication of a biomimetic, nanoparticulate anticancer vaccine that is capable of delivering autologously derived tumor antigen material together with a highly immunostimulatory adjuvant. The two major components, tumor antigens and adjuvant, are presented concurrently in a fashion that maximizes their ability to promote effective antigen presentation and activation of downstream immune processes. Ultimately, it is demonstrated that the formulation can elicit potent antitumor immune responses in vivo. When combined with additional immunotherapies such as checkpoint blockades, the nanovaccine demonstrates substantial therapeutic effect. Overall, the work represents the rational application of nanotechnology for immunoengineering and can provide a blueprint for the future development of personalized, autologous anticancer vaccines with broad applicability.


Subject(s)
Cell Membrane , Antigens, Neoplasm , Cancer Vaccines , Humans , Immunotherapy , Nanostructures , Neoplasms
11.
Adv Mater ; 29(33)2017 Sep.
Article in English | MEDLINE | ID: mdl-28656663

ABSTRACT

Antivirulence vaccination is a promising strategy for addressing bacterial infection that focuses on removing the harmful toxins produced by bacteria. However, a major challenge for creating vaccines against biological toxins is that the vaccine potency is often limited by lack of antigenic breadth, as most formulations have focused on single antigens, while most bacteria secrete a plethora of toxins. Here, a facile approach for generating multiantigenic nanotoxoids for use as vaccines against pathogenic bacteria by leveraging the natural affinity of virulence factors for cellular membranes is reported. Specifically, multiple virulent toxins from bacterial protein secretions are concurrently and naturally entrapped using a membrane-coated nanosponge construct. The resulting multivalent nanotoxoids are capable of delivering virulence factors together, are safe both in vitro and in vivo, and can elicit functional immunity capable of combating live bacterial infections in a mouse model. Despite containing the same bacterial antigens, the reported nanotoxoid formulation consistently outperforms a denatured protein preparation in all of the metrics studied, which underscores the utility of biomimetic nanoparticle-based neutralization and delivery. Overall this strategy helps to address major hurdles in the design of antivirulence vaccines, enabling increased antigenic breadth while maintaining safety.


Subject(s)
Bacterial Toxins/chemistry , Animals , Antigens , Disease Models, Animal , Mice , Nanoparticles , Vaccination
12.
Adv Mater ; 29(16)2017 Apr.
Article in English | MEDLINE | ID: mdl-28199033

ABSTRACT

Cell-membrane-coated nanoparticles have recently been studied extensively for their biological compatibility, retention of cellular properties, and adaptability to a variety of therapeutic and imaging applications. This class of nanoparticles, which has been fabricated with a variety of cell membrane coatings, including those derived from red blood cells (RBCs), platelets, white blood cells, cancer cells, and bacteria, exhibit properties that are characteristic of the source cell. In this study, a new type of biological coating is created by fusing membrane material from two different cells, providing a facile method for further enhancing nanoparticle functionality. As a proof of concept, the development of dual-membrane-coated nanoparticles from the fused RBC membrane and platelet membrane is demonstrated. The resulting particles, termed RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), are thoroughly characterized, and it is shown that they carry properties of both source cells. Further, the [RBC-P]NP platform exhibits long circulation and suitability for further in vivo exploration. The reported strategy opens the door for the creation of biocompatible, custom-tailored biomimetic nanoparticles with varying hybrid functionalities, which may be used to overcome the limitations of current nanoparticle-based therapeutic and imaging platforms.


Subject(s)
Blood Platelets , Erythrocytes , Biomimetics , Erythrocyte Membrane , Nanoparticles
13.
Bioconjug Chem ; 28(1): 23-32, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27798829

ABSTRACT

The cell membrane-coated nanoparticle is a biomimetic platform consisting of a nanoparticulate core coated with membrane derived from a cell, such as a red blood cell, platelet, or cancer cell. The cell membrane "disguise" allows the particles to be perceived by the body as the source cell by interacting with its surroundings using the translocated surface membrane components. The newly bestowed characteristics of the membrane-coated nanoparticle can be utilized for biological interfacing in the body, providing natural solutions to many biomedical issues. This Review will cover the interactions of these cell membrane-coated nanoparticles and their applications within three biomedical areas of interest, including (i) drug delivery, (ii) detoxification, and (iii) immune modulation.


Subject(s)
Cell Membrane , Coated Materials, Biocompatible , Nanoparticles , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/therapy , Bacteria/drug effects , Biomimetics , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Drug Carriers , Humans , Neoplasms/therapy , Rats , Wounds and Injuries/drug therapy
14.
Biomaterials ; 111: 116-123, 2016 12.
Article in English | MEDLINE | ID: mdl-27728811

ABSTRACT

Immune thrombocytopenia purpura (ITP) is characterized by the production of pathological autoantibodies that cause reduction in platelet counts. The disease can have serious medical consequences, leading to uncontrolled bleeding that can be fatal. Current widely used therapies for the treatment of ITP are non-specific and can, at times, result in complications that are more burdensome than the disease itself. In the present study, the use of platelet membrane-coated nanoparticles (PNPs) as a platform for the specific clearance of anti-platelet antibodies is explored. The nanoparticles, whose outer layer displays the full complement of native platelet surface proteins, act as decoys that strongly bind pathological anti-platelet antibodies in order to minimize disease burden. Here, we study the antibody binding properties of PNPs and assess the ability of the nanoparticles to neutralize antibody activity both in vitro and in vivo. Ultimately, we leverage the neutralization capacity of PNPs to therapeutically treat a murine model of antibody-induced thrombocytopenia and demonstrate considerable efficacy as shown in a bleeding time assay. PNPs represent a promising platform for the specific treatment of antibody-mediated immune thrombocytopenia by acting as an alternative target for anti-platelet antibodies, thus preserving circulating platelets with the potential of leaving broader immune function intact.


Subject(s)
Autoantibodies/immunology , Coated Materials, Biocompatible/administration & dosage , Nanoparticles/administration & dosage , Platelet Membrane Glycoproteins/administration & dosage , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Purpura, Thrombocytopenic, Idiopathic/immunology , Animals , Coated Materials, Biocompatible/chemistry , Male , Mice , Nanoparticles/chemistry , Platelet Membrane Glycoproteins/chemistry , Treatment Outcome
15.
Nanoscale ; 8(30): 14411-9, 2016 Aug 14.
Article in English | MEDLINE | ID: mdl-27411852

ABSTRACT

Lipid-polymer hybrid nanoparticles, consisting of a polymeric core coated by a layer of lipids, are a class of highly scalable, biodegradable nanocarriers that have shown great promise in drug delivery applications. Here, we demonstrate the facile synthesis of ultra-small, sub-25 nm lipid-polymer hybrid nanoparticles using an adapted nanoprecipitation approach and explore their utility for targeted delivery of a model chemotherapeutic. The fabrication process is first optimized to produce a monodisperse population of particles that are stable under physiological conditions. It is shown that these ultra-small hybrid nanoparticles can be functionalized with a targeting ligand on the surface and loaded with drug inside the polymeric matrix. Further, the in vivo fate of the nanoparticles after intravenous injection is characterized by examining the blood circulation and biodistribution. In a final proof-of-concept study, targeted ultra-small hybrid nanoparticles loaded with the cancer drug docetaxel are used to treat a mouse tumor model and demonstrate improved efficacy compared to a clinically available formulation of the drug. The ability to synthesize a significantly smaller version of the established lipid-polymer hybrid platform can ultimately enhance its applicability across a wider range of applications.


Subject(s)
Drug Delivery Systems , Lipids/chemistry , Nanoparticles , Neoplasms, Experimental/drug therapy , Animals , Cell Line, Tumor , Docetaxel , Female , Male , Mice , Mice, Nude , Polymers , Taxoids/administration & dosage , Tissue Distribution
16.
Adv Funct Mater ; 26(10): 1628-1635, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-27325913

ABSTRACT

With the rising threat of antibiotic-resistant bacteria, vaccination is becoming an increasingly important strategy to prevent and manage bacterial infections. Made from deactivated bacterial toxins, toxoid vaccines are widely used in the clinic as they help to combat the virulence mechanisms employed by different pathogens. Herein, the efficacy of a biomimetic nanoparticle-based anti-virulence vaccine is examined in a mouse model of methicillin-resistant Staphylococcus aureus (MRSA) skin infection. Vaccination with nanoparticle-detained staphylococcal α-hemolysin (Hla) effectively triggers the formation of germinal centers and induces high anti-Hla titers. Compared to mice vaccinated with control samples, those vaccinated with the nanoparticle toxoid show superior protective immunity against MRSA skin infection. The vaccination not only inhibits lesion formation at the site of bacterial challenge, but also reduces the invasiveness of MRSA, preventing dissemination into other organs. Overall, this biomimetic nanoparticle-based toxin detainment strategy is a promising method for the design of potent anti-virulence vaccines for managing bacterial infections.

17.
Nature ; 526(7571): 118-21, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26374997

ABSTRACT

Development of functional nanoparticles can be encumbered by unanticipated material properties and biological events, which can affect nanoparticle effectiveness in complex, physiologically relevant systems. Despite the advances in bottom-up nanoengineering and surface chemistry, reductionist functionalization approaches remain inadequate in replicating the complex interfaces present in nature and cannot avoid exposure of foreign materials. Here we report on the preparation of polymeric nanoparticles enclosed in the plasma membrane of human platelets, which are a unique population of cellular fragments that adhere to a variety of disease-relevant substrates. The resulting nanoparticles possess a right-side-out unilamellar membrane coating functionalized with immunomodulatory and adhesion antigens associated with platelets. Compared to uncoated particles, the platelet membrane-cloaked nanoparticles have reduced cellular uptake by macrophage-like cells and lack particle-induced complement activation in autologous human plasma. The cloaked nanoparticles also display platelet-mimicking properties such as selective adhesion to damaged human and rodent vasculatures as well as enhanced binding to platelet-adhering pathogens. In an experimental rat model of coronary restenosis and a mouse model of systemic bacterial infection, docetaxel and vancomycin, respectively, show enhanced therapeutic efficacy when delivered by the platelet-mimetic nanoparticles. The multifaceted biointerfacing enabled by the platelet membrane cloaking method provides a new approach in developing functional nanoparticles for disease-targeted delivery.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Blood Platelets/cytology , Cell Membrane/metabolism , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Platelet Adhesiveness , Animals , Anti-Bacterial Agents/pharmacokinetics , Blood Vessels/cytology , Blood Vessels/metabolism , Blood Vessels/pathology , Collagen/chemistry , Collagen/immunology , Complement Activation/immunology , Coronary Restenosis/blood , Coronary Restenosis/drug therapy , Coronary Restenosis/metabolism , Disease Models, Animal , Docetaxel , Humans , Macrophages/immunology , Male , Mice , Polymers/chemistry , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/blood , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/cytology , Staphylococcus aureus/metabolism , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Unilamellar Liposomes/chemistry , Vancomycin/administration & dosage , Vancomycin/pharmacokinetics
18.
Small ; 11(41): 5483-96, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26331993

ABSTRACT

Immunotherapeutic approaches for treating cancer overall have been receiving a considerable amount of interest due to the recent approval of several clinical formulations. Among the different modalities, anticancer vaccination acts by training the body to endogenously generate a response against tumor cells. However, despite the large amount of work that has gone into the development of such vaccines, the near absence of clinically approved formulations highlights the many challenges facing those working in the field. The generation of potent endogenous anticancer responses poses unique challenges due to the similarity between cancer cells and normal, healthy cells. As researchers continue to tackle the limited efficacy of vaccine formulations, fresh and novel approaches are being sought after to address many of the underlying problems. Here the application of nanoparticle technology towards the development of anticancer vaccines is discussed. Specifically, there is a focus on the benefits of using such strategies to manipulate antigen presenting cells (APCs), which are essential to the vaccination process, and how nanoparticle-based platforms can be rationally engineered to elicit appropriate downstream immune responses.


Subject(s)
Antigen-Presenting Cells/immunology , Immunotherapy, Adoptive/methods , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Neoplasms/immunology , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Humans , Nanocapsules/ultrastructure , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...