Nanoparticles in Cancer Immunotherapy

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Immunology".

Deadline for manuscript submissions: closed (30 November 2020) | Viewed by 79348

Special Issue Editor


E-Mail Website
Guest Editor
Department of Dermatology, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany
Interests: nanoparticle-mediated immunotherapy; dendritic cells; beta-2 integrins; cutaneous oncology

Special Issue Information

Dear Colleagues,

For some time now, nanomedicine has entered the mainstream of biomedical research, with immunotherapy as one of its major focus areas. Since the immune system is evolutionarily trained to respond to nano-sized objects (e.g., bacteria, viruses), these structures are well recognized by cells of the innate and adaptive immune system. Effective immunomodulation almost always requires the concurrent and coordinated delivery of several distinct signals to individual immune cells: in addition to the respective antigen to be immunized or tolerized against, costimulatory or tolerizing signals as well as signals that selectively address certain immune cell types or alter the nature of an immune response all need to be co-delivered. For this, nanoparticles are ideally suited and are thus being explored as molecular vaccines or immunomodulatory agents in many laboratories all over the world.

To modulate cancer-specific immune responses, immunotherapeutics primarily address specific immune cells within the blood or lymphatic organs, or directly within the tumor microenvironment, with the aim to either stimulate cancer-specific immune-effector cells or to inactivate tumor-promoting immunoregulatory cells. The development of vaccines that aim to activate cancer-specific T and/or B cells is one major experimental approach, but tools to address innate immune cells or to deliver immunotherapeutics directly to tumor may revolutionize cancer therapy.

This Special Issue of Cells will comprise a selection of original research papers and reviews focusing on novel approaches for nanomedicine-based cancer immunotherapy. Manuscripts on novel nanomaterials that selectively target certain immune cell populations or are able to carry peptide-, DNA- or RNA-encoded antigens, immune adjuvants, cytokines, and immunomodulators are within the focus of this Special Issue, with a preference for translational studies or studies using experimental animal models. Also within the scope of the Special Issue are studies that address determinants of cell type- or organ-selective uptake of nanoparticles, such as the serum protein corona or receptor-mediated nanoparticle uptake mechanisms. Targeted delivery of immunotherapeutics to solid tumors or nanoparticle-based imaging of cancer-specific immune responses are also welcome topics.

Prof. Dr. Stephan Grabbe
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer immunotherapy
  • nanomedicine
  • nanoparticles
  • cancer vaccines
  • tumor microenvironment

Published Papers (14 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

12 pages, 2462 KiB  
Article
Unraveling the In Vivo Protein Corona
by Johanna Simon, Gabor Kuhn, Michael Fichter, Stephan Gehring, Katharina Landfester and Volker Mailänder
Cells 2021, 10(1), 132; https://doi.org/10.3390/cells10010132 - 12 Jan 2021
Cited by 27 | Viewed by 3478
Abstract
Understanding the behavior of nanoparticles upon contact with a physiological environment is of urgent need in order to improve their properties for a successful therapeutic application. Most commonly, the interaction of nanoparticles with plasma proteins are studied under in vitro conditions. However, this [...] Read more.
Understanding the behavior of nanoparticles upon contact with a physiological environment is of urgent need in order to improve their properties for a successful therapeutic application. Most commonly, the interaction of nanoparticles with plasma proteins are studied under in vitro conditions. However, this has been shown to not reflect the complex situation after in vivo administration. Therefore, here we focused on the investigation of magnetic nanoparticles with blood proteins under in vivo conditions. Importantly, we observed a radically different proteome in vivo in comparison to the in vitro situation underlining the significance of in vivo protein corona studies. Next to this, we found that the in vivo corona profile does not significantly change over time. To mimic the in vivo situation, we established an approach, which we termed “ex vivo” as it uses whole blood freshly prepared from an animal. Overall, we present a comprehensive analysis focusing on the interaction between nanoparticles and blood proteins under in vivo conditions and how to mimic this situation with our ex vivo approach. This knowledge is needed to characterize the true biological identity of nanoparticles. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

14 pages, 6085 KiB  
Article
Transient Multivalent Nanobody Targeting to CD206-Expressing Cells via PH-Degradable Nanogels
by Maximilian Scherger, Evangelia Bolli, Ana Rita Pombo Antunes, Sana Arnouk, Judith Stickdorn, Alexandra Van Driessche, Hansjörg Schild, Stephan Grabbe, Bruno G. De Geest, Jo A. Van Ginderachter and Lutz Nuhn
Cells 2020, 9(10), 2222; https://doi.org/10.3390/cells9102222 - 01 Oct 2020
Cited by 11 | Viewed by 3513
Abstract
To target nanomedicines to specific cells, especially of the immune system, nanobodies can be considered as an attractive tool, as they lack the Fc part as compared to traditional antibodies and, thus, prevent unfavorable Fc-receptor mediated mistargeting. For that purpose, we have site-specifically [...] Read more.
To target nanomedicines to specific cells, especially of the immune system, nanobodies can be considered as an attractive tool, as they lack the Fc part as compared to traditional antibodies and, thus, prevent unfavorable Fc-receptor mediated mistargeting. For that purpose, we have site-specifically conjugated CD206/MMR-targeting nanobodies to three types of dye-labeled nanogel derivatives: non-degradable nanogels, acid-degradable nanogels (with ketal crosslinks), and single polymer chains (also obtained after nanogel degradation). All of them can be obtained from the same reactive ester precursor block copolymer. After incubation with naïve or MMR-expressing Chinese hamster ovary (CHO) cells, a nanobody mediated targeting and uptake could be confirmed for the nanobody-modified nanocarriers. Thereby, the intact nanogels that display nanobodies on their surface in a multivalent way showed a much stronger binding and uptake compared to the soluble polymers. Based on their acidic pH-responsive degradation potential, ketal crosslinked nanogels are capable of mediating a transient targeting that gets diminished upon unfolding into single polymer chains after endosomal acidification. Such control over particle integrity and targeting performance can be considered as highly attractive for safe and controllable immunodrug delivery purposes. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

13 pages, 2665 KiB  
Article
Stability of Alkyl Chain-Mediated Lipid Anchoring in Liposomal Membranes
by Lukas Gleue, Jonathan Schupp, Niklas Zimmer, Eyleen Becker, Holger Frey, Andrea Tuettenberg and Mark Helm
Cells 2020, 9(10), 2213; https://doi.org/10.3390/cells9102213 - 29 Sep 2020
Cited by 11 | Viewed by 3513
Abstract
Lipid exchange among biological membranes, lipoprotein particles, micelles, and liposomes is an important yet underrated phenomenon with repercussions throughout the life sciences. The premature loss of lipid molecules from liposomal formulations severely impacts therapeutic applications of the latter and thus limits the type [...] Read more.
Lipid exchange among biological membranes, lipoprotein particles, micelles, and liposomes is an important yet underrated phenomenon with repercussions throughout the life sciences. The premature loss of lipid molecules from liposomal formulations severely impacts therapeutic applications of the latter and thus limits the type of lipids and lipid conjugates available for fine-tuning liposomal properties. While cholesterol derivatives, with their irregular lipophilic surface shape, are known to readily undergo lipid exchange and interconvert, e.g., with serum, the situation is unclear for lipids with regular, linear-shaped alkyl chains. This study compares the propensity of fluorescence-labeled lipid conjugates of systematically varied lengths to migrate from liposomal particles consisting mainly of egg phosphatidyl choline 3 (EPC3) and cholesterol into biomembranes. We show that dialkyl glyceryl lipids with chains of 18–20 methylene units are inherently stable in liposomal membranes. In contrast, C16 lipids show some lipid exchange, albeit significantly less than comparable cholesterol conjugates. Remarkably, the C18 chain length, which confers noticeable anchor stability, corresponds to the typical chain length in biological membranes. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

17 pages, 3558 KiB  
Article
Increased Uptake of Silica Nanoparticles in Inflamed Macrophages but Not upon Co-Exposure to Micron-Sized Particles
by Eva Susnik, Patricia Taladriz-Blanco, Barbara Drasler, Sandor Balog, Alke Petri-Fink and Barbara Rothen-Rutishauser
Cells 2020, 9(9), 2099; https://doi.org/10.3390/cells9092099 - 15 Sep 2020
Cited by 12 | Viewed by 3704
Abstract
Silica nanoparticles (NPs) are widely used in various industrial and biomedical applications. Little is known about the cellular uptake of co-exposed silica particles, as can be expected in our daily life. In addition, an inflamed microenvironment might affect a NP’s uptake and a [...] Read more.
Silica nanoparticles (NPs) are widely used in various industrial and biomedical applications. Little is known about the cellular uptake of co-exposed silica particles, as can be expected in our daily life. In addition, an inflamed microenvironment might affect a NP’s uptake and a cell’s physiological response. Herein, prestimulated mouse J774A.1 macrophages with bacterial lipopolysaccharide were post-exposed to micron- and nanosized silica particles, either alone or together, i.e., simultaneously or sequentially, for different time points. The results indicated a morphological change and increased expression of tumor necrosis factor alpha in lipopolysaccharide prestimulated cells, suggesting a M1-polarization phenotype. Confocal laser scanning microscopy revealed the intracellular accumulation and uptake of both particle types for all exposure conditions. A flow cytometry analysis showed an increased particle uptake in lipopolysaccharide prestimulated macrophages. However, no differences were observed in particle uptakes between single- and co-exposure conditions. We did not observe any colocalization between the two silica (SiO2) particles. However, there was a positive colocalization between lysosomes and nanosized silica but only a few colocalized events with micro-sized silica particles. This suggests differential intracellular localizations of silica particles in macrophages and a possible activation of distinct endocytic pathways. The results demonstrate that the cellular uptake of NPs is modulated in inflamed macrophages but not in the presence of micron-sized particles. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Graphical abstract

13 pages, 1654 KiB  
Article
Multivalency Beats Complexity: A Study on the Cell Uptake of Carbohydrate Functionalized Nanocarriers to Dendritic Cells
by Matthias Krumb, Marie-Luise Frey, Jens Langhanki, Robert Forster, Danuta Kowalczyk, Volker Mailänder, Katharina Landfester and Till Opatz
Cells 2020, 9(9), 2087; https://doi.org/10.3390/cells9092087 - 12 Sep 2020
Viewed by 2692
Abstract
Herein, we report the synthesis of carbohydrate and glycodendron structures for dendritic cell targeting, which were subsequently bound to hydroxyethyl starch (HES) nanocapsules prepared by the inverse miniemulsion technique. The uptake of the carbohydrate-functionalized HES nanocapsules into immature human dendritic cells (hDCs) revealed [...] Read more.
Herein, we report the synthesis of carbohydrate and glycodendron structures for dendritic cell targeting, which were subsequently bound to hydroxyethyl starch (HES) nanocapsules prepared by the inverse miniemulsion technique. The uptake of the carbohydrate-functionalized HES nanocapsules into immature human dendritic cells (hDCs) revealed a strong dependence on the used carbohydrate. A multivalent mannose-terminated dendron was found to be far superior in uptake compared to the structurally more complex oligosaccharides used. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

12 pages, 2866 KiB  
Article
Cellular Uptake of siRNA-Loaded Nanocarriers to Knockdown PD-L1: Strategies to Improve T-cell Functions
by Raweewan Thiramanas, Mengyi Li, Shuai Jiang, Katharina Landfester and Volker Mailänder
Cells 2020, 9(9), 2043; https://doi.org/10.3390/cells9092043 - 07 Sep 2020
Cited by 8 | Viewed by 3004
Abstract
T-cells are a type of lymphocyte (a subtype of white blood cells) that play a central role in cell-mediated immunity. Currently, adoptive T-cell immunotherapy is being developed to destroy cancer cells. In this therapy, T-cells are harvested from a patient’s blood. After several [...] Read more.
T-cells are a type of lymphocyte (a subtype of white blood cells) that play a central role in cell-mediated immunity. Currently, adoptive T-cell immunotherapy is being developed to destroy cancer cells. In this therapy, T-cells are harvested from a patient’s blood. After several weeks of growth in culture, tumor-specific T-cells can be reinfused into the same cancer patient. This technique has proved highly efficient in cancer treatment. However, there are several biological processes that can suppress the anti-cancer responses of T-cells, leading to a loss of their functionality and a reduction of their viability. Therefore, strategies are needed to improve T-cell survival and their functions. Here, a small interfering RNA (siRNA)-loaded nanocarrier was used to knockdown PD-L1, one of the most important proteins causing a loss in the functionality of T-cells. The biocompatibility and the cellular uptake of siRNA-loaded silica nanocapsules (SiNCs) were investigated in CD8+ T-cells. Then, the PD-L1 expression at protein and at mRNA levels of the treated cells were evaluated. Furthermore, the effect of the PD-L1 knockdown was observed in terms of cell proliferation and the expression of specific biomarkers CD25, CD69 and CD71, which are indicators of T-cell functions. The results suggest that this siRNA-loaded nanocarrier showed a significant potential in the delivery of siRNA into T-cells. This in turn resulted in enhanced T-cell survival by decreasing the expression of the inhibitory protein PD-L1. Such nanocarriers could, therefore, be applied in adoptive T-cell immunotherapy for the treatment of cancer. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Graphical abstract

19 pages, 3496 KiB  
Article
Hybrid Biopolymer and Lipid Nanoparticles with Improved Transfection Efficacy for mRNA
by Christian D. Siewert, Heinrich Haas, Vera Cornet, Sara S. Nogueira, Thomas Nawroth, Lukas Uebbing, Antje Ziller, Jozef Al-Gousous, Aurel Radulescu, Martin A. Schroer, Clement E. Blanchet, Dmitri I. Svergun, Markus P. Radsak, Ugur Sahin and Peter Langguth
Cells 2020, 9(9), 2034; https://doi.org/10.3390/cells9092034 - 05 Sep 2020
Cited by 54 | Viewed by 11164
Abstract
Hybrid nanoparticles from lipidic and polymeric components were assembled to serve as vehicles for the transfection of messenger RNA (mRNA) using different portions of the cationic lipid DOTAP (1,2-Dioleoyl-3-trimethylammonium-propane) and the cationic biopolymer protamine as model systems. Two different sequential assembly approaches in [...] Read more.
Hybrid nanoparticles from lipidic and polymeric components were assembled to serve as vehicles for the transfection of messenger RNA (mRNA) using different portions of the cationic lipid DOTAP (1,2-Dioleoyl-3-trimethylammonium-propane) and the cationic biopolymer protamine as model systems. Two different sequential assembly approaches in comparison with a direct single-step protocol were applied, and molecular organization in correlation with biological activity of the resulting nanoparticle systems was investigated. Differences in the structure of the nanoparticles were revealed by thorough physicochemical characterization including small angle neutron scattering (SANS), small angle X-ray scattering (SAXS), and cryogenic transmission electron microscopy (cryo-TEM). All hybrid systems, combining lipid and polymer, displayed significantly increased transfection in comparison to lipid/mRNA and polymer/mRNA particles alone. For the hybrid nanoparticles, characteristic differences regarding the internal organization, release characteristics, and activity were determined depending on the assembly route. The systems with the highest transfection efficacy were characterized by a heterogenous internal organization, accompanied by facilitated release. Such a system could be best obtained by the single step protocol, starting with a lipid and polymer mixture for nanoparticle formation. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

14 pages, 2664 KiB  
Article
In Vivo siRNA Delivery to Immunosuppressive Liver Macrophages by α-Mannosyl-Functionalized Cationic Nanohydrogel Particles
by Leonard Kaps, Nadine Leber, Adrian Klefenz, Niklas Choteschovsky, Rudolf Zentel, Lutz Nuhn and Detlef Schuppan
Cells 2020, 9(8), 1905; https://doi.org/10.3390/cells9081905 - 15 Aug 2020
Cited by 34 | Viewed by 4488
Abstract
Macrophages are the front soldiers of the innate immune system and are vital for immune defense, tumor surveillance, and tissue homeostasis. In chronic diseases, including cancer and liver fibrosis, macrophages can be forced into an immunosuppressive and profibrotic M2 phenotype. M2-type macrophages overexpress [...] Read more.
Macrophages are the front soldiers of the innate immune system and are vital for immune defense, tumor surveillance, and tissue homeostasis. In chronic diseases, including cancer and liver fibrosis, macrophages can be forced into an immunosuppressive and profibrotic M2 phenotype. M2-type macrophages overexpress the mannose receptor CD206. Targeting these cells via CD206 and macrophage repolarization towards an immune stimulating and antifibrotic M1 phenotype through RNA interference represents an appealing therapeutic approach. We designed nanohydrogel particles equipped with mannose residues on the surface (ManNP) that delivered siRNA more efficiently to M2 polarized macrophages compared to their untargeted counterparts (NonNP) in vitro. The ManNP were then assessed for their in vivo targeting potential in mice with experimental liver fibrosis that is characterized by increased profibrotic (and immunosuppressive) M2-type macrophages. Double-labelled siRNA-loaded ManNP carrying two different near infrared labels for siRNA and ManNP showed good biocompatibility and robust uptake in fibrotic livers as assessed by in vivo near infrared imaging. siRNA–ManNP were highly colocalized with CD206+ M2-type macrophages on a cellular level, while untargeted NP (NonNP) showed little colocalization and were non-specifically taken up by other liver cells. ManNP did not induce hepatic inflammation or kidney dysfunction, as demonstrated by serological analysis. In conclusion, α-mannosyl-functionalized ManNP direct NP towards M2-type macrophages in diseased livers and prevent unspecific uptake in non-target cells. ManNP are promising vehicles for siRNA and other drugs for immunomodulatory treatment of liver fibrosis and liver cancer. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Graphical abstract

Review

Jump to: Research

18 pages, 1272 KiB  
Review
Nano-Enhanced Cancer Immunotherapy: Immunology Encounters Nanotechnology
by Ernesto Bockamp, Sebastian Rosigkeit, Dominik Siegl and Detlef Schuppan
Cells 2020, 9(9), 2102; https://doi.org/10.3390/cells9092102 - 15 Sep 2020
Cited by 54 | Viewed by 7948
Abstract
Cancer immunotherapy utilizes the immune system to fight cancer and has already moved from the laboratory to clinical application. However, and despite excellent therapeutic outcomes in some hematological and solid cancers, the regular clinical use of cancer immunotherapies reveals major limitations. These include [...] Read more.
Cancer immunotherapy utilizes the immune system to fight cancer and has already moved from the laboratory to clinical application. However, and despite excellent therapeutic outcomes in some hematological and solid cancers, the regular clinical use of cancer immunotherapies reveals major limitations. These include the lack of effective immune therapy options for some cancer types, unresponsiveness to treatment by many patients, evolving therapy resistance, the inaccessible and immunosuppressive nature of the tumor microenvironment (TME), and the risk of potentially life-threatening immune toxicities. Given the potential of nanotechnology to deliver, enhance, and fine-tune cancer immunotherapeutic agents, the combination of cancer immunotherapy with nanotechnology can overcome some of these limitations. In this review, we summarize innovative reports and novel strategies that successfully combine nanotechnology and cancer immunotherapy. We also provide insight into how nanoparticular combination therapies can be used to improve therapy responsiveness, to reduce unwanted toxicity, and to overcome adverse effects of the TME. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

13 pages, 2858 KiB  
Review
Targeted Activation of T Cells with IL-2-Coupled Nanoparticles
by Verena K. Raker, Christian Becker, Katharina Landfester and Kerstin Steinbrink
Cells 2020, 9(9), 2063; https://doi.org/10.3390/cells9092063 - 09 Sep 2020
Cited by 13 | Viewed by 5958
Abstract
Interleukin-2 (IL-2) is a T cell growth factor particularly required in regulatory T cell maintenance and memory T cell responses. High-dose IL-2 treatment was the first FDA-approved immunotherapy for cancer, while low-dose IL-2 administration has shown promise in allograft rejection and autoimmune and [...] Read more.
Interleukin-2 (IL-2) is a T cell growth factor particularly required in regulatory T cell maintenance and memory T cell responses. High-dose IL-2 treatment was the first FDA-approved immunotherapy for cancer, while low-dose IL-2 administration has shown promise in allograft rejection and autoimmune and inflammatory diseases. However, its pleiotropic nature and the existence of IL-2 receptors with different binding affinity limit its therapeutic application. For an improved clinical applicability of the cytokine, a targeted receptor assignment must, therefore, be achieved. Nanoparticles allow controlling the location and dose of immunomodulating compounds and to specifically address specific receptors through targeted drug binding. In this review article we discuss the IL-2 biology and current clinical application with regard to nanoparticle-based IL-2-mediated manipulation of T cell responses in autoimmunity, chronic inflammation, and cancer. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

55 pages, 2383 KiB  
Review
Nucleic Acid-Based Approaches for Tumor Therapy
by Simone Hager, Frederic Julien Fittler, Ernst Wagner and Matthias Bros
Cells 2020, 9(9), 2061; https://doi.org/10.3390/cells9092061 - 09 Sep 2020
Cited by 41 | Viewed by 7119
Abstract
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients’ anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based [...] Read more.
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients’ anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based drugs aimed to initiate and to enhance anti-tumor responses have come of age. This review intends to provide a comprehensive overview of the current state of the therapeutic use of nucleic acids for cancer treatment on various levels, comprising (i) mRNA and DNA-based vaccines to be expressed by antigen presenting cells evoking sustained anti-tumor T cell responses, (ii) molecular adjuvants, (iii) strategies to inhibit/reprogram tumor-induced regulatory immune cells e.g., by RNA interference (RNAi), (iv) genetically tailored T cells and natural killer cells to directly recognize tumor antigens, and (v) killing of tumor cells, and reprograming of constituents of the tumor microenvironment by gene transfer and RNAi. Aside from further improvements of individual nucleic acid-based drugs, the major perspective for successful cancer therapy will be combination treatments employing conventional regimens as well as immunotherapeutics like checkpoint inhibitors and nucleic acid-based drugs, each acting on several levels to adequately counter-act tumor immune evasion. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

23 pages, 1521 KiB  
Review
Targeting Cancer Associated Fibroblasts in Liver Fibrosis and Liver Cancer Using Nanocarriers
by Leonard Kaps and Detlef Schuppan
Cells 2020, 9(9), 2027; https://doi.org/10.3390/cells9092027 - 03 Sep 2020
Cited by 80 | Viewed by 11990
Abstract
Cancer associated fibroblasts (CAF) and the extracellular matrix (ECM) produced by them have been recognized as key players in cancer biology and emerged as important targets for cancer treatment and drug discovery. Apart from their presence in stroma rich tumors, such as biliary, [...] Read more.
Cancer associated fibroblasts (CAF) and the extracellular matrix (ECM) produced by them have been recognized as key players in cancer biology and emerged as important targets for cancer treatment and drug discovery. Apart from their presence in stroma rich tumors, such as biliary, pancreatic and subtypes of hepatocellular cancer (HCC), both CAF and certain ECM components are also present in cancers without an overt intra-tumoral desmoplastic reaction. They support cancer development, growth, metastasis and resistance to chemo- or checkpoint inhibitor therapy by a multitude of mechanisms, including angiogenesis, ECM remodeling and active immunosuppression by secretion of tumor promoting and immune suppressive cytokines, chemokines and growth factors. CAF resemble activated hepatic stellate cells (HSC)/myofibroblasts, expressing α-smooth muscle actin and especially fibroblast activation protein (FAP). Apart from FAP, CAF also upregulate other functional cell surface proteins like platelet-derived growth factor receptor β (PDGFRβ) or the insulin-like growth factor receptor II (IGFRII). Notably, if formulated with adequate size and zeta potential, injected nanoparticles home preferentially to the liver. Several nanoparticular formulations were tested successfully to deliver dugs to activated HSC/myofibroblasts. Thus, surface modified nanocarriers with a cyclic peptide binding to the PDGFRβ or with mannose-6-phosphate binding to the IGFRII, effectively directed drug delivery to activated HSC/CAF in vivo. Even unguided nanohydrogel particles and lipoplexes loaded with siRNA demonstrated a high in vivo uptake and functional siRNA delivery in activated HSC, indicating that liver CAF/HSC are also addressed specifically by well-devised nanocarriers with optimized physicochemical properties. Therefore, CAF have become an attractive target for the development of stroma-based cancer therapies, especially in the liver. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Graphical abstract

17 pages, 1679 KiB  
Review
Novel Opportunities for Cathepsin S Inhibitors in Cancer Immunotherapy by Nanocarrier-Mediated Delivery
by Natalie Fuchs, Mergim Meta, Detlef Schuppan, Lutz Nuhn and Tanja Schirmeister
Cells 2020, 9(9), 2021; https://doi.org/10.3390/cells9092021 - 02 Sep 2020
Cited by 24 | Viewed by 5330
Abstract
Cathepsin S (CatS) is a secreted cysteine protease that cleaves certain extracellular matrix proteins, regulates antigen presentation in antigen-presenting cells (APC), and promotes M2-type macrophage and dendritic cell polarization. CatS is overexpressed in many solid cancers, and overall, it appears to promote an [...] Read more.
Cathepsin S (CatS) is a secreted cysteine protease that cleaves certain extracellular matrix proteins, regulates antigen presentation in antigen-presenting cells (APC), and promotes M2-type macrophage and dendritic cell polarization. CatS is overexpressed in many solid cancers, and overall, it appears to promote an immune-suppressive and tumor-promoting microenvironment. While most data suggest that CatS inhibition or knockdown promotes anti-cancer immunity, cell-specific inhibition, especially in myeloid cells, appears to be important for therapeutic efficacy. This makes the design of CatS selective inhibitors and their targeting to tumor-associated M2-type macrophages (TAM) and DC an attractive therapeutic strategy compared to the use of non-selective immunosuppressive compounds or untargeted approaches. The selective inhibition of CatS can be achieved through optimized small molecule inhibitors that show good pharmacokinetic profiles and are orally bioavailable. The targeting of these inhibitors to TAM is now more feasible using nanocarriers that are functionalized for a directed delivery. This review discusses the role of CatS in the immunological tumor microenvironment and upcoming possibilities for a nanocarrier-mediated delivery of potent and selective CatS inhibitors to TAM and related APC to promote anti-tumor immunity. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

25 pages, 1571 KiB  
Review
Role of Liver-Mediated Tolerance in Nanoparticle-Based Tumor Therapy
by Maximiliano L. Cacicedo, Carolina Medina-Montano, Leonard Kaps, Cinja Kappel, Stephan Gehring and Matthias Bros
Cells 2020, 9(9), 1985; https://doi.org/10.3390/cells9091985 - 28 Aug 2020
Cited by 8 | Viewed by 4308
Abstract
In the last decades, the use of nanocarriers for immunotherapeutic purposes has gained a lot of attention, especially in the field of tumor therapy. However, most types of nanocarriers accumulate strongly in the liver after systemic application. Due to the default tolerance-promoting role [...] Read more.
In the last decades, the use of nanocarriers for immunotherapeutic purposes has gained a lot of attention, especially in the field of tumor therapy. However, most types of nanocarriers accumulate strongly in the liver after systemic application. Due to the default tolerance-promoting role of liver non-parenchymal cells (NPCs), Kupffer cells (KCs), liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs), their potential role on the immunological outcome of systemic nano-vaccination approaches for therapy of tumors in the liver and in other organs needs to be considered. Concerning immunological functions, KCs have been the focus until now, but recent studies have elucidated an important role of LSECs and HSCs as well. Therefore, this review aims to summarize current knowledge on the employment of nanocarriers for immunotherapeutic therapy of liver diseases and the overall role of liver NPCs in the context of nano-vaccination approaches. With regard to the latter, we discuss strategies on how to address liver NPCs, aiming to exploit and modulate their immunological properties, and alternatively how to avoid unwanted engagement of nano-vaccines by liver NPCs for tumor therapy. Full article
(This article belongs to the Special Issue Nanoparticles in Cancer Immunotherapy)
Show Figures

Figure 1

Back to TopTop