1
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
2
|
Szachniewicz MM, Neustrup MA, van den Eeden SJF, van Meijgaarden KE, Franken KLMC, van Veen S, Koning RI, Limpens RWAL, Geluk A, Bouwstra JA, Ottenhoff THM. Evaluation of PLGA, lipid-PLGA hybrid nanoparticles, and cationic pH-sensitive liposomes as tuberculosis vaccine delivery systems in a Mycobacterium tuberculosis challenge mouse model - A comparison. Int J Pharm 2024; 666:124842. [PMID: 39424087 DOI: 10.1016/j.ijpharm.2024.124842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Tuberculosis (TB) continues to pose a global threat for millennia, currently affecting over 2 billion people and causing 10.6 million new cases and 1.3 million deaths annually. The only existing vaccine, Mycobacterium Bovis Bacillus Calmette-Guérin (BCG), provides highly variable and inadequate protection in adults and adolescents. This study explores newly developed subunit tuberculosis vaccines that use a multistage protein fusion antigen Ag85b-ESAT6-Rv2034 (AER). The protection efficacy, as well as in vivo induced immune responses, were compared for five vaccines: BCG; AER-CpG/MPLA mix; poly(D,L-lactic-co-glycolic acid) (PLGA); lipid-PLGA hybrid nanoparticles (NPs); and cationic pH-sensitive liposomes (the latter three delivering AER together with CpG and MPLA). All vaccines, except the AER-adjuvant mix, induced protection in Mycobacterium tuberculosis (Mtb)-challenged C57/Bl6 mice as indicated by a significant reduction in bacterial burden in lungs and spleens of the animals. Four AER-based vaccines significantly increased the number of circulating multifunctional CD4+ and CD8+ T-cells producing IL-2, IFNγ, and TNFα, exhibiting a central memory phenotype. Furthermore, AER-based vaccines induced an increase in CD69+ B-cell counts as well as high antigen-specific antibody titers. Unexpectedly, none of the observed immune responses were associated with the bacterial burden outcome, such that the mechanism responsible for the observed vaccine-induced protection of these vaccines remains unclear. These findings suggest the existence of non-classical protective mechanisms for Mtb infection, which could, once identified, provide interesting targets for novel vaccines.
Collapse
Affiliation(s)
- Mikołaj M Szachniewicz
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands.
| | - Malene A Neustrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Susan J F van den Eeden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Krista E van Meijgaarden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Kees L M C Franken
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Suzanne van Veen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Roman I Koning
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Ronald W A L Limpens
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Annemieke Geluk
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Joke A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Tom H M Ottenhoff
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
3
|
Freitas R, Ferreira E, Miranda A, Ferreira D, Relvas-Santos M, Castro F, Santos B, Gonçalves M, Quintas S, Peixoto A, Palmeira C, Silva AMN, Santos LL, Oliveira MJ, Sarmento B, Ferreira JA. Targeted and Self-Adjuvated Nanoglycovaccine Candidate for Cancer Immunotherapy. ACS NANO 2024; 18:10088-10103. [PMID: 38535625 DOI: 10.1021/acsnano.3c12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Advanced-stage solid primary tumors and metastases often express mucin 16 (MUC16), carrying immature glycans such as the Tn antigen, resulting in specific glycoproteoforms not found in healthy human tissues. This presents a valuable approach for designing targeted therapeutics, including cancer glycovaccines, which could potentially promote antigen recognition and foster the immune response to control disease spread and prevent relapse. In this study, we describe an adjuvant-free poly(lactic-co-glycolic acid) (PLGA)-based nanoglycoantigen delivery approach that outperforms conventional methods by eliminating the need for protein carriers while exhibiting targeted and adjuvant properties. To achieve this, we synthesized a library of MUC16-Tn glycoepitopes through single-pot enzymatic glycosylation, which were then stably engrafted onto the surface of PLGA nanoparticles, generating multivalent constructs that better represent cancer molecular heterogeneity. These glycoconstructs demonstrated affinity for Macrophage Galactose-type Lectin (MGL) receptor, known to be highly expressed by immature antigen-presenting cells, enabling precise targeting of immune cells. Moreover, the glycopeptide-grafted nanovaccine candidate displayed minimal cytotoxicity and induced the activation of dendritic cells in vitro, even in the absence of an adjuvant. In vivo, the formulated nanovaccine candidate was also nontoxic and elicited the production of IgG specifically targeting MUC16 and MUC16-Tn glycoproteoforms in cancer cells and tumors, offering potential for precise cancer targeting, including targeted immunotherapies.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Eduardo Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
| | - Andreia Miranda
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Flávia Castro
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Beatriz Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Martina Gonçalves
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Sofia Quintas
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- Immunology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Health School of University Fernando Pessoa, 4249-004 Porto, Portugal
| | - André M N Silva
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
- Department of Surgical Oncology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Maria José Oliveira
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- IUCS-CESPU, 4585-116 Gandra, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
| |
Collapse
|
4
|
Yilma AN, Sahu R, Subbarayan P, Villinger F, Coats MT, Singh SR, Dennis VA. PLGA-Chitosan Encapsulated IL-10 Nanoparticles Modulate Chlamydia Inflammation in Mice. Int J Nanomedicine 2024; 19:1287-1301. [PMID: 38348174 PMCID: PMC10860865 DOI: 10.2147/ijn.s432970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/12/2023] [Indexed: 02/15/2024] Open
Abstract
Introduction Interleukin-10 (IL-10) is a key anti-inflammatory mediator in protecting host from over-exuberant responses to pathogens and play important roles in wound healing, autoimmunity, cancer, and homeostasis. However, its application as a therapeutic agent for biomedical applications has been limited due to its short biological half-life. Therefore, it is important to prolong the half-life of IL-10 to replace the current therapeutic application, which relies on administering large and repeated dosages. Therefore, not a cost-effective approach. Thus, studies that aim to address this type of challenges are always in need. Methods Recombinant IL-10 was encapsulated in biodegradable nanoparticles (Poly-(Lactic-co-Glycolic Acid) and Chitosan)) by the double emulsion method and then characterized for size, surface charge, thermal stability, cytotoxicity, in vitro release, UV-visible spectroscopy, and Fourier Transform-Infrared Spectroscopy as well as evaluated for its anti-inflammatory effects. Bioactivity of encapsulated IL-10 was evaluated in vitro using J774A.1 macrophage cell-line and in vivo using BALB/c mice. Inflammatory cytokines (IL-6 and TNF-α) were quantified from culture supernatants using specific enzyme-linked immunosorbent assay (ELISA), and significance was analyzed using ANOVA. Results We obtained a high 96% encapsulation efficiency with smooth encapsulated IL-10 nanoparticles of ~100-150 nm size and release from nanoparticles as measurable to 22 days. Our result demonstrated that encapsulated IL-10 was biocompatible and functional by reducing the inflammatory responses induced by LPS in macrophages. Of significance, we also proved the functionality of encapsulated IL-10 by its capacity to reduce inflammation in BALB/c mice as provoked by Chlamydia trachomatis, an inflammatory sexually transmitted infectious bacterium. Discussion Collectively, our results show the successful IL-10 encapsulation, slow release to prolong its biological half-life and reduce inflammatory cytokines IL-6 and TNF production in vitro and in mice. Our results serve as proof of concept to further explore the therapeutic prospective of encapsulated IL-10 for biomedical applications, including inflammatory diseases.
Collapse
Affiliation(s)
- Abebayehu N Yilma
- Center for NanoBiotechnology Research (CNBR), Alabama State University, Montgomery, AL, USA
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Rajnish Sahu
- Center for NanoBiotechnology Research (CNBR), Alabama State University, Montgomery, AL, USA
| | - Praseetha Subbarayan
- Center for NanoBiotechnology Research (CNBR), Alabama State University, Montgomery, AL, USA
| | - Francois Villinger
- Department of Biology, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Mamie T Coats
- Department of Clinical and Diagnostics Sciences, School of Health Professionals, The University at Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Shree R Singh
- Center for NanoBiotechnology Research (CNBR), Alabama State University, Montgomery, AL, USA
| | - Vida A Dennis
- Center for NanoBiotechnology Research (CNBR), Alabama State University, Montgomery, AL, USA
| |
Collapse
|
5
|
Matos AI, Peres C, Carreira B, Moura LIF, Acúrcio RC, Vogel T, Wegener E, Ribeiro F, Afonso MB, Santos FMF, Martínez‐Barriocanal Á, Arango D, Viana AS, Góis PMP, Silva LC, Rodrigues CMP, Graca L, Jordan R, Satchi‐Fainaro R, Florindo HF. Polyoxazoline-Based Nanovaccine Synergizes with Tumor-Associated Macrophage Targeting and Anti-PD-1 Immunotherapy against Solid Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300299. [PMID: 37434063 PMCID: PMC10477894 DOI: 10.1002/advs.202300299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Immune checkpoint blockade reaches remarkable clinical responses. However, even in the most favorable cases, half of these patients do not benefit from these therapies in the long term. It is hypothesized that the activation of host immunity by co-delivering peptide antigens, adjuvants, and regulators of the transforming growth factor (TGF)-β expression using a polyoxazoline (POx)-poly(lactic-co-glycolic) acid (PLGA) nanovaccine, while modulating the tumor-associated macrophages (TAM) function within the tumor microenvironment (TME) and blocking the anti-programmed cell death protein 1 (PD-1) can constitute an alternative approach for cancer immunotherapy. POx-Mannose (Man) nanovaccines generate antigen-specific T-cell responses that control tumor growth to a higher extent than poly(ethylene glycol) (PEG)-Man nanovaccines. This anti-tumor effect induced by the POx-Man nanovaccines is mediated by a CD8+ -T cell-dependent mechanism, in contrast to the PEG-Man nanovaccines. POx-Man nanovaccine combines with pexidartinib, a modulator of the TAM function, restricts the MC38 tumor growth, and synergizes with PD-1 blockade, controlling MC38 and CT26 tumor growth and survival. This data is further validated in the highly aggressive and poorly immunogenic B16F10 melanoma mouse model. Therefore, the synergistic anti-tumor effect induced by the combination of nanovaccines with the inhibition of both TAM- and PD-1-inducing immunosuppression, holds great potential for improving immunotherapy outcomes in solid cancer patients.
Collapse
Affiliation(s)
- Ana I. Matos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Carina Peres
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Barbara Carreira
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liane I. F. Moura
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Rita C. Acúrcio
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Theresa Vogel
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Erik Wegener
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Marta B. Afonso
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Fábio M. F. Santos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Águeda Martínez‐Barriocanal
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Ana S. Viana
- Centro de Química EstruturalDepartamento de Química e BioquímicaInstitute of Molecular SciencesFaculty of SciencesUniversidade de LisboaLisbon1749‐016Portugal
| | - Pedro M. P. Góis
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liana C. Silva
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Cecília M. P. Rodrigues
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Luis Graca
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Rainer Jordan
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineSagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Helena F. Florindo
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| |
Collapse
|
6
|
Danaeifar M, Negahdari B, Eslam HM, Zare H, Ghanaat M, Koushali SS, Malekshahi ZV. Polymeric nanoparticles for DNA vaccine-based cancer immunotherapy: a review. Biotechnol Lett 2023; 45:1053-1072. [PMID: 37335426 DOI: 10.1007/s10529-023-03383-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 06/21/2023]
Abstract
Cancer is one of the leading causes of death and mortality in the world. There is an essential need to develop new drugs or therapeutic approaches to manage treatment-resistant cancers. Cancer immunotherapy is a type of cancer treatment that uses the power of the body's immune system to prevent, control, and eliminate cancer. One of the materials used as a vaccine in immunotherapy is DNA. The application of polymeric nanoparticles as carriers for DNA vaccines could be an effective therapeutic approach to activate immune responses and increase antigen presentation efficiency. Various materials have been used as polymeric nanoparticles, including: chitosan, poly (lactic-co-glycolic acid), Polyethylenimine, dendrimers, polypeptides, and polyesters. Application of these polymer nanoparticles has several advantages, including increased vaccine delivery, enhanced antigen presentation, adjuvant effects, and more sustainable induction of the immune system. Besides many clinical trials and commercial products that were developed based on polymer nanoparticles, there is still a need for more comprehensive studies to increase the DNA vaccine efficiency in cancer immunotherapy using this type of carrier.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Houra Mobaleghol Eslam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Zare
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Momeneh Ghanaat
- Department of Microbiology, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | - Sekinehe Shokouhi Koushali
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Kou P, Levy ES, Nguyen AD, Zhang D, Chen S, Cui Y, Zhang X, Broccatelli F, Pizzano J, Cantley J, Bortolon E, Rousseau E, Berlin M, Dragovich P, Sethuraman V. Development of Liposome Systems for Enhancing the PK Properties of Bivalent PROTACs. Pharmaceutics 2023; 15:2098. [PMID: 37631312 PMCID: PMC10458015 DOI: 10.3390/pharmaceutics15082098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Proteolysis-Targeting Chimeras (PROTACs) are a promising new technology in drug development. They have rapidly evolved in recent years, with several of them in clinical trials. While most of these advances have been associated with monovalent protein degraders, bivalent PROTACs have also entered clinical trials, although progression to market has been limited. One of the reasons is the complex physicochemical properties of the heterobifunctional PROTACs. A promising strategy to improve pharmacokinetics of highly lipophilic compounds, such as PROTACs, is encapsulation in liposome systems. Here we describe liposome systems for intravenous administration to enhance the PK properties of two bivalent PROTAC molecules, by reducing clearance and increasing systemic coverage. We developed and characterized a PROTAC-in-cyclodextrin liposome system where the drug was retained in the liposome core. In PK studies at 1 mg/kg for GNE-01 the PROTAC-in-cyclodextrin liposome, compared to the solution formulation, showed a 80- and a 380-fold enhancement in AUC for mouse and rat studies, respectively. We further investigated the same PROTAC-in-cyclodextrin liposome system with the second PROTAC (GNE-02), where we monitored both lipid and drug concentrations in vivo. Similarly, in a mouse PK study of GEN-02, the PROTAC-in-cyclodextrin liposome system exhibited enhancement in plasma concentration of a 23× increase over the conventional solution formulation. Importantly, the lipid CL correlated with the drug CL. Additionally, we investigated a conventional liposome approach for GNE-02, where the PROTAC resides in the lipid bilayer. Here, a 5× increase in AUC was observed, compared to the conventional solution formulation, and the drug CL was faster than the lipid CL. These results indicate that the different liposome systems can be tailored to translate across multiple PROTAC systems to modulate and improve plasma concentrations. Optimization of the liposomes could further improve tumor concentration and improve the overall therapeutic index (TI). This delivery technology may be well suited to bring novel protein targeted PROTACs into clinics.
Collapse
Affiliation(s)
- Ponien Kou
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - Elizabeth S. Levy
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - An D. Nguyen
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| | - Donglu Zhang
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Shu Chen
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Yusi Cui
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Xing Zhang
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Fabio Broccatelli
- Drug Metabolism & Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (D.Z.); (S.C.); (Y.C.); (X.Z.); (F.B.)
| | - Jennifer Pizzano
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Jennifer Cantley
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Elizabeth Bortolon
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Emma Rousseau
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Michael Berlin
- Arvinas LLC, 5 Science Park, New Haven, CT 06511, USA; (J.P.); (J.C.); (E.B.); (E.R.); (M.B.)
| | - Peter Dragovich
- Medicinal Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Vijay Sethuraman
- Small Molecules Pharmaceutics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA; (P.K.); (E.S.L.); (A.D.N.)
| |
Collapse
|
8
|
Schunke J, Mailänder V, Landfester K, Fichter M. Delivery of Immunostimulatory Cargos in Nanocarriers Enhances Anti-Tumoral Nanovaccine Efficacy. Int J Mol Sci 2023; 24:12174. [PMID: 37569548 PMCID: PMC10419017 DOI: 10.3390/ijms241512174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Finding a long-term cure for tumor patients still represents a major challenge. Immunotherapies offer promising therapy options, since they are designed to specifically prime the immune system against the tumor and modulate the immunosuppressive tumor microenvironment. Using nucleic-acid-based vaccines or cellular vaccines often does not achieve sufficient activation of the immune system in clinical trials. Additionally, the rapid degradation of drugs and their non-specific uptake into tissues and cells as well as their severe side effects pose a challenge. The encapsulation of immunomodulatory molecules into nanocarriers provides the opportunity of protected cargo transport and targeted uptake by antigen-presenting cells. In addition, different immunomodulatory cargos can be co-delivered, which enables versatile stimulation of the immune system, enhances anti-tumor immune responses and improves the toxicity profile of conventional chemotherapeutic agents.
Collapse
Affiliation(s)
- Jenny Schunke
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | | | - Michael Fichter
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
9
|
Sohrab SS, Raj R, Nagar A, Hawthorne S, Paiva-Santos AC, Kamal MA, El-Daly MM, Azhar EI, Sharma A. Chronic Inflammation's Transformation to Cancer: A Nanotherapeutic Paradigm. Molecules 2023; 28:molecules28114413. [PMID: 37298889 DOI: 10.3390/molecules28114413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The body's normal immune response against any invading pathogen that causes infection in the body results in inflammation. The sudden transformation in inflammation leads to the rise of inflammatory diseases such as chronic inflammatory bowel disease, autoimmune disorders, and colorectal cancer (different types of cancer develop at the site of chronic infection and inflammation). Inflammation results in two ways: short-term inflammation i.e., non-specific, involves the action of various immune cells; the other results in long-term reactions lasting for months or years. It is specific and causes angiogenesis, fibrosis, tissue destruction, and cancer progression at the site of inflammation. Cancer progression relies on the interaction between the host microenvironment and tumor cells along with the inflammatory responses, fibroblast, and vascular cells. The two pathways that have been identified connecting inflammation and cancer are the extrinsic and intrinsic pathways. Both have their own specific role in linking inflammation to cancer, involving various transcription factors such as Nuclear factor kappa B, Activator of transcription, Single transducer, and Hypoxia-inducible factor, which in turn regulates the inflammatory responses via Soluble mediators cytokines (such as Interleukin-6, Hematopoietin-1/Erythropoietin, and tumor necrosis factor), chemokines (such as Cyclooxygenase-2, C-X-C Motif chemokines ligand-8, and IL-8), inflammatory cells, cellular components (such as suppressor cells derived from myeloid, tumor-associated macrophage, and acidophils), and promotes tumorigenesis. The treatment of these chronic inflammatory diseases is challenging and needs early detection and diagnosis. Nanotechnology is a booming field nowadays for its rapid action and easy penetration inside the infected destined cells. Nanoparticles are widely classified into different categories based on their different factors and properties such as size, shape, cytotoxicity, and others. Nanoparticles emerged as excellent with highly progressive medical inventions to cure diseases such as cancer, inflammatory diseases, and others. Nanoparticles have shown higher binding capacity with the biomolecules in inflammation reduction and lowers the oxidative stress inside tissue/cells. In this review, we have overall discussed inflammatory pathways that link inflammation to cancer, major inflammatory diseases, and the potent action of nanoparticles in chronic inflammation-related diseases.
Collapse
Affiliation(s)
- Sayed Sartaj Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Riya Raj
- Department of Biochemistry, Bangalore University, Banglore 560056, India
| | - Amka Nagar
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida 201310, India
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mohammad Amjad Kamal
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Enzymoics Inc., Hebersham, NSW 2770, Australia
- Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Mai M El-Daly
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ankur Sharma
- Strathclyde Institute of Pharmaceutical and Biomedical Sciences, University of Strathclyde, Glasgow G1 0RE, UK
| |
Collapse
|
10
|
Zhao Z, Ortega-Rivera OA, Chung YH, Simms A, Steinmetz NF. A co-formulated vaccine of irradiated cancer cells and cowpea mosaic virus improves ovarian cancer rejection. J Mater Chem B 2023. [PMID: 36861401 DOI: 10.1039/d2tb02355e] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Ovarian cancer ranks fifth in cancer deaths amongst women, and most patients are diagnosed with late-stage and disseminated diseases. Surgical debulking and chemotherapy remove most of the tumor burden and provide a short period of remission; however, most patients experience cancer relapse and eventually succumb to the disease. Therefore, there is an urgent need for the development of vaccines to prime anti-tumor immunity and prevent its recurrence. Here we developed vaccine formulations composed of a mixture of irradiated cancer cells (ICCs, providing the antigen) and cowpea mosaic virus (CPMV) adjuvants. More specifically we compared the efficacy of co-formulated vs. mixtures of ICCs and CPMV. Specifically, we compared co-formulations where the ICCs and CPMV are bonded through natural CPMV-cell interactions or chemical coupling vs. mixtures of PEGylated CPMV and ICCs, where PEGylation of CPMV prevents ICC interactions. Flow cytometry and confocal imaging provided insights into the composition of the vaccines and their efficacy was tested using a mouse model of disseminated ovarian cancer. 67% of the mice receiving the co-formulated CPMV-ICCs survived the initial tumor challenge, and 60% of the surviving mice rejected tumors in a re-challenge experiment. In stark contrast, simple mixtures of the ICCs and (PEGylated) CPMV adjuvants were ineffective. Overall, this study highlights the importance of the co-delivery of cancer antigens and adjuvants in ovarian cancer vaccine development.
Collapse
Affiliation(s)
- Zhongchao Zhao
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA. .,Center for Nano-ImmunoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Moores Cancer Center, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Oscar A Ortega-Rivera
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.
| | - Young Hun Chung
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Department of Bioengineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Andrea Simms
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA. .,Center for Nano-ImmunoEngineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Moores Cancer Center, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Department of Bioengineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Department of Radiology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Institute for Materials Discovery and Design, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.,Center for Engineering in Cancer, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| |
Collapse
|
11
|
Hashemi Goradel N, Nemati M, Bakhshandeh A, Arashkia A, Negahdari B. Nanovaccines for cancer immunotherapy: Focusing on complex formation between adjuvant and antigen. Int Immunopharmacol 2023; 117:109887. [PMID: 36841155 DOI: 10.1016/j.intimp.2023.109887] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 02/27/2023]
Abstract
As an interesting cancer immunotherapy approach, cancer vaccines have been developed to deliver tumor antigens and adjuvants to antigen-presenting cells (APCs). Although the safety and easy production shifted the vaccine designing platforms toward the subunit vaccines, their efficacy is limited due to inefficient vaccine delivery. Nanotechnology-based vaccines, called nanovaccines, address the delivery limitations through co-delivery of antigens and adjuvants into lymphoid organs and APCs and their intracellular release, leading to cross-presentation of antigens and induction of potent anti-tumor immune responses. Although the nanovaccines, either as encapsulating agents or biomimetic nanoparticles, exert the desired anti-tumor activities, there is evidence that the mixing formulation to form nanocomplexes between antigens and adjuvants based on the electrostatic interactions provokes high levels of immune responses owing to Ags' availability and faster release. Here, we summarized the various platforms for developing cancer vaccines and the advantages of using delivery systems. The cancer nanovaccines, including nanoparticle-based and biomimetic-based nanovaccines, are discussed in detail. Finally, we focused on the nanocomplexes formation between antigens and adjuvants as promising cancer nanovaccine platforms.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Bakhshandeh
- Department of Industrial Engineering and Management Systems, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Koyande NP, Srivastava R, Padmakumar A, Rengan AK. Advances in Nanotechnology for Cancer Immunoprevention and Immunotherapy: A Review. Vaccines (Basel) 2022; 10:1727. [PMID: 36298592 PMCID: PMC9610880 DOI: 10.3390/vaccines10101727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 01/24/2023] Open
Abstract
One of the most effective cancer therapies, cancer immunotherapy has produced outstanding outcomes in the field of cancer treatment. However, the cost is excessive, which limits its applicability. A smart way to address this issue would be to apply the knowledge gained through immunotherapy to develop strategies for the immunoprevention of cancer. The use of cancer vaccines is one of the most popular methods of immunoprevention. This paper reviews the technologies and processes that support the advantages of cancer immunoprevention over traditional cancer immunotherapies. Nanoparticle drug delivery systems and nanoparticle-based nano-vaccines have been employed in the past for cancer immunotherapy. This paper outlines numerous immunoprevention strategies and how nanotechnology can be applied in immunoprevention. To comprehend the non-clinical and clinical evaluation of these cancer vaccines through clinical studies is essential for acceptance of the vaccines.
Collapse
Affiliation(s)
| | | | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, India
| |
Collapse
|
13
|
Choudhary P, Khajavinia A, Mohammadi R, Ng SH, Bérubé N, Yalamati D, Haddadi A, Wilson HL. A Single-Dose Intramuscular Nanoparticle Vaccine With or Without Prior Intrauterine Priming Triggers Specific Uterine and Colostral Mucosal Antibodies and Systemic Immunity in Gilts but Not Passive Protection for Suckling Piglets. Front Vet Sci 2022; 9:931232. [PMID: 35990278 PMCID: PMC9383261 DOI: 10.3389/fvets.2022.931232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022] Open
Abstract
An effective single-dose vaccine that protects the dam and her suckling offspring against infectious disease would be widely beneficial to livestock animals. We assessed whether a single-dose intramuscular (i.m.) porcine epidemic diarrhea virus (PEDV) vaccine administered to the gilt 30 days post-breeding could generate mucosal and systemic immunity and sufficient colostral and mature milk antibodies to protect suckling piglets against infectious challenge. The vaccine was comprised of polymeric poly-(lactide-co-glycolide) (PGLA)-nanoparticle (NP) encapsulating recombinant PEDV spike protein 1 (PEDVS1) associated with ARC4 and ARC7 adjuvants, a muramyl dipeptide analog and a monophosphoryl lipid A (MPLA) analog, respectively (NP-PEDVS1). To establish whether prior mucosal exposure could augment the i.m. immune response and/or contribute to mucosal tolerance, gilts were immunized with the NP-PEDVS1 vaccine via the intrauterine route at breeding, followed by the i.m. vaccine 30 days later. Archived colostrum from gilts that were challenged with low-dose PEDV plus alum was used as positive reference samples for neutralizing antibodies and passive protection. On day 100 of gestation (70 days post i.m. immunization), both vaccinated groups showed significant PEDVS1-specific IgG and IgA in the serum, as well as in uterine tissue collected on the day of euthanasia. Anti-PEDVS1 colostral IgG antibody titers collected at farrowing were significantly higher relative to the negative control gilts indicating that the NP vaccine was effective in contributing to the colostral antibodies. The PEDVS1-specific colostral IgA and anti-PEDVS1 IgG and IgA antibodies in the mature milk collected 6 days after farrowing were low for both vaccinated groups. No statistical differences between the vaccinated groups were observed, suggesting that the i.u. priming vaccine did not induce mucosal tolerance. Piglets born to either group of vaccinated gilts did not receive sufficient neutralizing antibodies to protect them against infectious PEDV at 3 days of age. In summary, a single i.m. NP vaccine administered 30 days after breeding and a joint i.u./i.m. vaccine administered at breeding and 30 days post-breeding induced significant anti-PEDVS1 immunity in systemic and mucosal sites but did not provide passive protection in suckling offspring.
Collapse
Affiliation(s)
- Pooja Choudhary
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Amir Khajavinia
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ramin Mohammadi
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Nathalie Bérubé
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Azita Haddadi
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Heather L. Wilson
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Vaccinology and Immunotherapeutics Program at the School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Heather L. Wilson
| |
Collapse
|
14
|
Rostamizadeh L, Molavi O, Rashid M, Ramazani F, Baradaran B, Lavasanaifar A, Lai R. Recent advances in cancer immunotherapy: Modulation of tumor microenvironment by Toll-like receptor ligands. BIOIMPACTS : BI 2022; 12:261-290. [PMID: 35677663 PMCID: PMC9124882 DOI: 10.34172/bi.2022.23896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
![]()
Immunotherapy is considered a promising approach for cancer treatment. An important strategy for cancer immunotherapy is the use of cancer vaccines, which have been widely used for cancer treatment. Despite the great potential of cancer vaccines for cancer treatment, their therapeutic effects in clinical settings have been limited. The main reason behind the lack of significant therapeutic outcomes for cancer vaccines is believed to be the immunosuppressive tumor microenvironment (TME). The TME counteracts the therapeutic effects of immunotherapy and provides a favorable environment for tumor growth and progression. Therefore, overcoming the immunosuppressive TME can potentially augment the therapeutic effects of cancer immunotherapy in general and therapeutic cancer vaccines in particular. Among the strategies developed for overcoming immunosuppression in TME, the use of toll-like receptor (TLR) agonists has been suggested as a promising approach to reverse immunosuppression. In this paper, we will review the application of the four most widely studied TLR agonists including agonists of TLR3, 4, 7, and 9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Rostamizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramazani
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanaifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Canada
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
15
|
Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Semin Cancer Biol 2022; 86:624-644. [DOI: 10.1016/j.semcancer.2022.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
|
16
|
Development of Peptide-Based Vaccines for Cancer. JOURNAL OF ONCOLOGY 2022; 2022:9749363. [PMID: 35342400 PMCID: PMC8941562 DOI: 10.1155/2022/9749363] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Peptides cancer vaccines are designed based on the epitope peptides that can elicit humoral and cellular immune responses targeting tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs). In order to develop a clinically safe and more effective vaccine for the future, several issues need to be addressed, and these include the selection of optimal antigen targets, adjuvants, and immunization regimens. Another emerging approach involves the use of personalized peptide-based vaccines based on neoantigens to enhance antitumor response. Rationally designed combinatorial therapy is currently being investigated with chemotherapeutic drugs or immune checkpoint inhibitor therapies to improve the efficacy. This review discusses an overview of the development of peptide-based vaccines, the role of adjuvants, and the delivery systems for peptide vaccines as well as combinatorial therapy as potential anticancer strategies.
Collapse
|
17
|
Verma P, Biswas S, Yadav N, Khatri A, Siddiqui H, Panda JJ, Rawat BS, Tailor P, Chauhan VS. Delivery of a Cancer-Testis Antigen-Derived Peptide Using Conformationally Restricted Dipeptide-Based Self-Assembled Nanotubes. Mol Pharm 2021; 18:3832-3842. [PMID: 34499836 DOI: 10.1021/acs.molpharmaceut.1c00451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Use of tumor-associated antigens for cancer immunotherapy is limited due to their poor in vivo stability and low cellular uptake. Delivery of antigenic peptides using synthetic polymer-based nanostructures has been actively pursued but with limited success. Peptide-based nanostructures hold much promise as delivery vehicles due to their easy design and synthesis and inherent biocompatibility. Here, we report self-assembly of a dipeptide containing a non-natural amino acid, α,β-dehydrophenylalanine (ΔF), into nanotubes, which efficiently entrapped a MAGE-3-derived peptide (M3). M3 entrapped in F-ΔF nanotubes was more stable to a nonspecific protease treatment and both F-ΔF and F-ΔF-M3 showed no cellular toxicity for four cancerous and noncancerous cell lines used. F-ΔF-M3 showed significantly higher cellular uptake in RAW 267.4 macrophage cells compared to M3 alone and also induced in vitro maturation of dendritic cells (DCs). Immunization of mice with F-ΔF-M3 selected a higher number of IFN-γ secreting CD8+ T cells and CD4+ T compared to M3 alone. On day 21, a tumor growth inhibition ratio (TGI, %) of 41% was observed in a murine melanoma model. These results indicate that F-ΔF nanotubes are highly biocompatible, efficiently delivered M3 to generate cytotoxic T lymphocytes responses, and able to protect M3 from degradation under in vivo conditions. The F-ΔF dipeptide-based nanotubes may be considered as a good platform for further development as delivery agents.
Collapse
Affiliation(s)
- Priyanka Verma
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Saikat Biswas
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Nitin Yadav
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Anjali Khatri
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Hamda Siddiqui
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.,Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Jiban Jyoti Panda
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.,Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | | | | | | |
Collapse
|
18
|
Zhao Y, Bilal M, Qindeel M, Khan MI, Dhama K, Iqbal HMN. Nanotechnology-based immunotherapies to combat cancer metastasis. Mol Biol Rep 2021; 48:6563-6580. [PMID: 34424444 DOI: 10.1007/s11033-021-06660-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023]
Abstract
Emerging concepts in nanotechnology have gained particular attention for their clinical translation of immunotherapies of cancer, autoimmune and infectious diseases. Several nanoconstructs have been engineered with unique structural, physicochemical, and functional features as robust alternatives for conventional chemotherapies. Traditional cancer therapies like chemotherapy, radiotherapy, and ultimately surgery are the most widely practiced in biomedical settings. Biomaterials and nanotechnology have introduced vehicles for drug delivery and have revolutionized the concept of the modern immunotherapeutic paradigm. Various types of nanomaterials, such as nanoparticles and, more specifically, drug-loaded nanoparticles are becoming famous for drug delivery applications because of safety, patient compliance, and smart action. Such therapeutic modalities have acknowledged regulatory endorsement and are being used in twenty-first-century clinical settings. Considering the emerging concepts and landscaping potentialities, herein, we spotlight and discuss nanoparticle-based immunotherapies as a smart and sophisticated drug delivery approach to combat cancer metastasis. The introductory part of this manuscript discusses a broad overview of cancer immunotherapy to understand better the tumor microenvironment and nanotechnology-oriented immunomodulatory strategies to cope with advanced-stage cancers. Following that, most addressable problems allied with conventional immunotherapies are given in comparison to nanoparticle-based immunotherapies. The later half of this work comprehensively highlights the requisite delivery of various bioactive entities with particular cases and examples. Finally, this review also encompasses a comprehensive concluding overview and future standpoints to strengthen a successful clinical translation of nanoparticle-based immunotherapies as a smart and sophisticated drug delivery approach.
Collapse
Affiliation(s)
- Yuping Zhao
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Maimoona Qindeel
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Hamdard Institute of Pharmaceutical Sciences, Hamdard University Islamabad Campus, Islamabad, Pakistan
| | - Muhammad Imran Khan
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849, Monterrey, Mexico.
| |
Collapse
|
19
|
Mannosylated polylactic-co-glycolic acid (MN-PLGA) nanoparticles induce potent anti-tumor immunity in murine model of breast cancer. Biomed Pharmacother 2021; 142:111962. [PMID: 34358752 DOI: 10.1016/j.biopha.2021.111962] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/20/2022] Open
Abstract
Nanoparticle-based cancer immunotherapy is considered a novel and promising therapeutic strategy aimed at stimulating host immune responses against tumors. To this end, in the present study, mannan-decorated polylactic-co-glycolic acid (PLGA) nanoparticles containing tumor cell lysate (TCL) and poly riboinosinic polycytidylic acid (poly I:C) were used as antigen delivery systems to immunize breast tumor-bearing Balb/c mice. PLGA nanoparticles were fabricated employing a double emulsion solvent evaporation method. The formation of spherical and uniform nanoparticles (NPs) ranging 150-250 nm was detected by field emission scanning electron microscopy (FESEM) and dynamic light scattering (DLS). Four nanoformulation were used to treat mice and vaccination-induced immunological responses. Tumor regression and overall survival rate were evaluated in four experimental groups. Tumor cell lysate and poly I:C loaded mannan-decorated nanoparticles (TCL-Poly I:C) NP-MN caused a significant decrease in tumor growth and 2- to 3-fold improvement in survival times of the treated mice. The NPs with or without mannan decoration elicited stronger responses in terms of lymphocyte proliferation, delayed-type hypersensitivity and CD107a expression. Moreover, our data indicated that the production of IFN-γ and IL-2 increased while the production of IL-4 and IL-10 decreased in splenocytes culture supernatants. In the pathological evaluations, we found that necrosis and immune cells infiltration rate in the tumor tissue of the treated mice was elevated, while tumor cellularity and lung metastases significantly decreased in particular in the group that received (TCL-Poly I:C) NP-MN. Altogether, our findings suggested that the mannan-decorated PLGA NPs antigen delivery system had significant anti-tumor effects against the murine model of breast cancer and it could be considered as a step forward to human breast cancer immunotherapy.
Collapse
|
20
|
Ram Kumar PS, Rencilin CF, Sundar K. Emerging nanomaterials for cancer immunotherapy. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy is a unique approach to treat cancer that targets tumours besides triggering the immune cells. It attempts to harness the supremacy and specificity of immune cells for the regression of malignancy. The key strategy of immunotherapy is that it boosts the natural defence and manipulates the immune system at both cellular and molecular levels. Long-lasting anti-tumour response, reduced metastasis, and recurrence can be achieved with immunotherapy than conventional treatments. For example, targeting cytotoxic T-lymphocyte antigen-4 (CTLA4) by monoclonal antibody is reported as an effective strategy against cancer progression in vivo and chimeric antigen receptor (CAR) modified T-cells are known to express a stronger anti-tumour activity. CTLA4 and CAR are, therefore, beneficial in cancer immunotherapy; however, in clinical settings, both are expensive and cause adverse side effects. Nanomaterials have augmented advantages in cancer immunotherapy, besides their utility in effective delivery and diagnostics. In particular, materials based on lipids, polymers, and metals have been sought-after for delivery technologies. Moreover, the surface of nanomaterials can be engineered using ligands, antigens, and antibodies to target immune cells. In this sense, checkpoint inhibitors, cytokines, agonistic antibodies, surface receptors, and engineered T-cells are promising to regulate the immune system against tumours. Therefore, emerging nanomaterials that can be used for the treatment of cancer is the prime focus of this review. The correlation of mode of administration and biodistribution of various nanomaterials is reviewed here. Besides, the acute and chronic side effects and outcome of clinical trials in the context of cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Pandian Sureshbabu Ram Kumar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Clayton Fernando Rencilin
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Krishnan Sundar
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| |
Collapse
|
21
|
Khan M, Sherwani S, Khan S, Alouffi S, Alam M, Al-Motair K, Khan S. Insights into Multifunctional Nanoparticle-Based Drug Delivery Systems for Glioblastoma Treatment. Molecules 2021; 26:molecules26082262. [PMID: 33919694 PMCID: PMC8069805 DOI: 10.3390/molecules26082262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GB) is an aggressive cancer with high microvascular proliferation, resulting in accelerated invasion and diffused infiltration into the surrounding brain tissues with very low survival rates. Treatment options are often multimodal, such as surgical resection with concurrent radiotherapy and chemotherapy. The development of resistance of tumor cells to radiation in the areas of hypoxia decreases the efficiency of such treatments. Additionally, the difficulty of ensuring drugs effectively cross the natural blood-brain barrier (BBB) substantially reduces treatment efficiency. These conditions concomitantly limit the efficacy of standard chemotherapeutic agents available for GB. Indeed, there is an urgent need of a multifunctional drug vehicle system that has potential to transport anticancer drugs efficiently to the target and can successfully cross the BBB. In this review, we summarize some nanoparticle (NP)-based therapeutics attached to GB cells with antigens and membrane receptors for site-directed drug targeting. Such multicore drug delivery systems are potentially biodegradable, site-directed, nontoxic to normal cells and offer long-lasting therapeutic effects against brain cancer. These models could have better therapeutic potential for GB as well as efficient drug delivery reaching the tumor milieu. The goal of this article is to provide key considerations and a better understanding of the development of nanotherapeutics with good targetability and better tolerability in the fight against GB.
Collapse
Affiliation(s)
- Mohd Khan
- Department of Chemistry, College of Sciences, University of Ha’il, Ha’il 2440, Saudi Arabia
- Molecular Diagnostic and Personalised Therapeutics Unit, University of Ha’il, Ha’il 2440, Saudi Arabia; (S.A.); (K.A.-M.)
- Correspondence: or
| | - Subuhi Sherwani
- Department of Biology, College of Sciences, University of Ha’il, Ha’il 2440, Saudi Arabia; (S.S.); (M.A.)
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Ha’il, Ha’il 2440, Saudi Arabia;
| | - Sultan Alouffi
- Molecular Diagnostic and Personalised Therapeutics Unit, University of Ha’il, Ha’il 2440, Saudi Arabia; (S.A.); (K.A.-M.)
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Ha’il, Ha’il 2440, Saudi Arabia
| | - Mohammad Alam
- Department of Biology, College of Sciences, University of Ha’il, Ha’il 2440, Saudi Arabia; (S.S.); (M.A.)
| | - Khalid Al-Motair
- Molecular Diagnostic and Personalised Therapeutics Unit, University of Ha’il, Ha’il 2440, Saudi Arabia; (S.A.); (K.A.-M.)
| | - Shahper Khan
- Interdisciplinary Nanotechnology Centre, Aligarh Muslim University, Aligarh 202002, U.P., India;
| |
Collapse
|
22
|
Gao Y, Zhao Q, Xiao M, Huang X, Wu X. A versatile photothermal vaccine based on acid-responsive glyco-nanoplatform for synergistic therapy of cancer. Biomaterials 2021; 273:120792. [PMID: 33872856 DOI: 10.1016/j.biomaterials.2021.120792] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/24/2021] [Accepted: 03/29/2021] [Indexed: 01/10/2023]
Abstract
The race is on for therapeutic agents that stop cancer. An effective vaccine offers a safe and promising approach for cancer immunotherapy. However, substantial barriers to immunotherapy in cancer vaccines include the low immunogenicity of cancer antigens and the immunosuppression commonly present in solid tumors, resulting in significant challenges for developing a clinically effective cancer vaccine. Here, the state of the art of synergistic therapy, which includes the photothermal effect combined with immunotherapy, was investigated to target tumors. For the first time, indocyanine green (ICG, referred to as I), imiquimod (R837, referred to as R) and a foreign cytotoxic T lymphocyte antigen peptide (CTL-Ap, referred to as Ap) with the sequence of SIINFEKL from ovalbumin (OVA) were encapsulated by acetalated dextran (AcDEX) to form nanoparticles (NPs) averaging 92 nm in diameter as an immunogen. Administration of the resulting multifunctional vaccine I-R-Ap-AcDEX NPs enhanced antitumor cytotoxic T lymphocyte (CTL) immunotherapy. On the one hand, subcutaneous immunization of the NPs allows foreign Ap to enter the major histocompatibility complex class I (MHC-I) cross-presentation pathway of antigen-presenting cells, thereby presenting Ap and eliciting high levels of Ap-specific CTLs. On the other hand, intratumor/intravenous injections of the NPs allow foreign Ap to enter tumor cells and present Ap through the MHC-I cross-presentation pathway. Ap-specific CTLs can kill Ap-presented tumor cells. Furthermore, the NPs generated near-infrared laser triggered the photothermal killing of tumor cells. To our knowledge, this is the first report of AcDEX NPs in antitumor photothermal therapy. Strikingly, systemic administration of the I-R-Ap-AcDEX NPs combined with near-infrared laser irradiation allowed for complete protection to mice from the tumors when applied to two non-OVA tumor models. This quite impressive result displays the great promise of synergistic therapy by the vaccine I-R-Ap-AcDEX NPs, an approach that harnesses the photothermal effect to boost antitumor immunotherapy.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China
| | - Qingyu Zhao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China
| | - Min Xiao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, United States
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China; Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
23
|
Volovat SR, Negru S, Stolniceanu CR, Volovat C, Lungulescu C, Scripcariu D, Cobzeanu BM, Stefanescu C, Grigorescu C, Augustin I, Lupascu Ursulescu C, Volovat CC. Nanomedicine to modulate immunotherapy in cutaneous melanoma (Review). Exp Ther Med 2021; 21:535. [PMID: 33815608 PMCID: PMC8014970 DOI: 10.3892/etm.2021.9967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shifted the paradigm in cancer treatment in recent years. Immune checkpoint blockage (ICB), the active cancer vaccination and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer represent the main developments, achieving a surprising increased survival in patients included in clinical trials. In spite of these results, the current state-of-the-art immunotherapy has its limitations in efficacy. The existence of an interdisciplinary interface involving current knowledge in biology, immunology, bioengineering and materials science represents important progress in increasing the effectiveness of immunotherapy in cancer. Cutaneous melanoma remains a difficult cancer to treat, in which immunotherapy is a major therapeutic option. In fact, enhancing immunotherapy is possible using sophisticated biomedical nanotechnology platforms of organic or inorganic materials or engineering various immune cells to enhance the immune system. In addition, biological devices have developed, changing the approach to and treatment results in melanoma. In this review, we present different modalities to modulate the immune system, as well as opportunities and challenges in melanoma treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Serban Negru
- Department of Medical Oncology, 'Victor Babes' University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Dragos Scripcariu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Mihail Cobzeanu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Grigorescu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Corina Lupascu Ursulescu
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Radiology, 'Sf. Spiridon' Emergency Clinic Hospital, 700111 Iasi, Romania
| |
Collapse
|
24
|
Levy ES, Chang R, Zamecnik CR, Dhariwala MO, Fong L, Desai TA. Multi-Immune Agonist Nanoparticle Therapy Stimulates Type I Interferons to Activate Antigen-Presenting Cells and Induce Antigen-Specific Antitumor Immunity. Mol Pharm 2021; 18:1014-1025. [PMID: 33541072 DOI: 10.1021/acs.molpharmaceut.0c00984] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer immunity is mediated by a delicate orchestration between the innate and adaptive immune system both systemically and within the tumor microenvironment. Although several adaptive immunity molecular targets have been proven clinically efficacious, stand-alone innate immunity targeting agents have not been successful in the clinic. Here, we report a nanoparticle optimized for systemic administration that combines immune agonists for TLR9, STING, and RIG-I with a melanoma-specific peptide to induce antitumor immunity. These immune agonistic nanoparticles (iaNPs) significantly enhance the activation of antigen-presenting cells to orchestrate the development and response of melanoma-sensitized T-cells. iaNP treatment not only suppressed tumor growth in an orthotopic solid tumor model, but also significantly reduced tumor burden in a metastatic animal model. This combination biomaterial-based approach to coordinate innate and adaptive anticancer immunity provides further insights into the benefits of stimulating multiple activation pathways to promote tumor regression, while also offering an important platform to effectively and safely deliver combination immunotherapies for cancer.
Collapse
Affiliation(s)
- Elizabeth S Levy
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| | - Ryan Chang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States.,Department of Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Colin R Zamecnik
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| | - Miqdad O Dhariwala
- Department of Dermatology, University of California San Francisco, San Francisco, California 94143, United Stats
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California 94143, United States.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, California 94143, United States
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
25
|
Doxorubicin-Loaded PLGA Nanoparticles for Cancer Therapy: Molecular Weight Effect of PLGA in Doxorubicin Release for Controlling Immunogenic Cell Death. Pharmaceutics 2020; 12:pharmaceutics12121165. [PMID: 33260446 PMCID: PMC7759870 DOI: 10.3390/pharmaceutics12121165] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
Direct local delivery of immunogenic cell death (ICD) inducers to a tumor site is an attractive approach for leading ICD effectively, due to enabling the concentrated delivery of ICD inducers to the tumor site. Herein, we prepared doxorubicin (DOX)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) using different molecular weight PLGA (7000 g/mol and 12,000 g/mol), showing different drug release kinetics. The different release kinetics of DOX might differently stimulate a tumor cell-specific immune response by releasing damage-associated molecular patterns (DAMPs), resulting in showing a different antitumor response in the living body. DOX-PLGA7K NPs showed faster DOX release kinetics than DOX-PLGA12K NPs in the physiological condition. DOX-PLGA7K NPs and DOX-PLGA12K NPs were successfully taken up by the CT-26 tumor cells, subsequently showing different DOX localization times at the nucleus. Released DOX successfully lead to cytotoxicity and HMGB1 release in vitro. Although the DOX-PLGA7K NPs and DOX-PLGA12K NPs showed different sustained DOX release kinetics in vitro, tumor growth of the CT-26 tumor was similarly inhibited for 28 days post-direct tumor injection. Furthermore, the immunological memory effect was successfully established by the ICD-based tumor-specific immune responses, including DC maturation and tumor infiltration of cytotoxic T lymphocytes (CTLs). We expect that the controlled release of ICD-inducible chemotherapeutic agents, using different types of nanomedicines, can provide potential in precision cancer immunotherapy by controlling the tumor-specific immune responses, thus improving the therapeutic efficacy.
Collapse
|
26
|
Haddadi A, Chaffey A, Ng SH, Yalamati D, Wilson HL. Combination of Innate Immune Modulators as Vaccine Adjuvants in Mice. Vaccines (Basel) 2020; 8:E569. [PMID: 33019524 PMCID: PMC7712867 DOI: 10.3390/vaccines8040569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
The development of new, effective, and safe vaccines necessarily requires the identification of new adjuvant(s) to enhance the potency and longevity of antigen-specific immune responses. In the present study, we compare the antibody-mediated and cell-mediated immune (CMI) responses within groups of mice vaccinated subcutaneously with ovalbumin (OVA; as an experimental antigen) plus polyphosphazene (an innate immune modulator), Polyinosinic:polycytidylic acid (poly-I:C; (an RNA mimetic) and glycopeptide ARC5 (which is a Toll-like receptor (TLR), TLR2 ligand and PAM3CSK4 analogue) formulated together in a soluble vaccine. We also investigated the effect of a polymeric nanoparticle of ARC4 and ARC7 (which are a novel muramyl dipeptide analogue and a monophosophoryl lipid A (MPLA) analogue, respectively) plus OVA +/- ARC5 as a subcutaneous vaccine in mice. OVA+ARC4/ARC7 nanoparticle +/- ARC5 triggered a robust and balanced Th1/Th2-type humoral response with significant anti-OVA IgA in serum, and significant interferon (IFN)-γ and interleukin (IL)-17 production in splenocytes after 35 days relative to the controls. Formulation of OVA with ARC4/ARC7 nanoparticles should be investigated for inducing protective immunity against infectious pathogens in mice and other species.
Collapse
Affiliation(s)
- Azita Haddadi
- Division of Pharmacy, College of Pharmacy & Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Alyssa Chaffey
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (A.C.); (S.H.N.)
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (A.C.); (S.H.N.)
| | | | - Heather L. Wilson
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (A.C.); (S.H.N.)
| |
Collapse
|
27
|
Jafari S, Molavi O, Kahroba H, Hejazi MS, Maleki-Dizaji N, Barghi S, Kiaie SH, Jadidi-Niaragh F. Clinical application of immune checkpoints in targeted immunotherapy of prostate cancer. Cell Mol Life Sci 2020; 77:3693-3710. [PMID: 32006051 PMCID: PMC11104895 DOI: 10.1007/s00018-020-03459-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/27/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
Immunotherapy is considered as an effective method for cancer treatment owing to the induction of specific and long-lasting anti-cancer effects. Immunotherapeutic strategies have shown significant success in human malignancies, particularly in prostate cancer (PCa), a major global health issue regarding its high metastatic rates. In fact, the first cancer vaccine approved by FDA was Provenge, which has been successfully used for treatment of PCa. Despite the remarkable success of cancer immunotherapy in PCa, many of the developed immunotherapy methods show poor therapeutic outcomes. Immunosuppression in tumor microenvironment (TME) induced by non-functional T cells (CD4+ and CD8+), tolerogenic dendritic cells (DCs), and regulatory T cells, has been reported to be the main obstacle to the effectiveness of anti-tumor immune responses induced by an immunotherapy method. The present review particularly focuses on the latest findings of the immune checkpoints (ICPs), including CTLA-4, PD-1, PD-L1, LAG-3, OX40, B7-H3, 4-1BB, VISTA, TIM-3, and ICOS; these checkpoints are able to have immune modulatory effects on the TME of PCa. This paper further discusses different approaches in ICPs targeting therapy and summarizes the latest advances in the clinical application of ICP-targeted therapy as monotherapy or in combination with other cancer therapy modalities in PCa.
Collapse
Affiliation(s)
- Sevda Jafari
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran.
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Science, Tabriz, Iran.
| | - Houman Kahroba
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Science, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Saied Hejazi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Science, Tabriz, Iran
| | - Nasrin Maleki-Dizaji
- Department of Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siamak Barghi
- Department of Medical Laboratory Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Seyed Hossein Kiaie
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Feng Z, Yi X, Hajavi J. New and old adjuvants in allergen-specific immunotherapy: With a focus on nanoparticles. J Cell Physiol 2020; 236:863-876. [PMID: 32657468 DOI: 10.1002/jcp.29941] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/01/2020] [Indexed: 12/19/2022]
Abstract
Allergic diseases have remarkably increased in recent years. Nowadays, efforts for curing and management of these disorders are an important concern worldwide. Allergen-specific immunotherapy (ASIT) has recently gained more attention as a means for the management of allergic diseases. Adjuvants or helper agents are materials applied for better stimulating and shifting of protective responses, and these belong to an extremely diverse collection of complexes. The main function of adjuvants includes acting as depot foundations, transferring vehicles, and immunostimulators. Immunostimulatory adjuvants have gained increasing attention for ASIT. In this regard, the present study provides a review of old and new adjuvants used in allergen immunotherapy.
Collapse
Affiliation(s)
- Zhongtao Feng
- Department of Clinical Laboratory, Jining No.1 People's Hospital, Jining, China
| | - Xin Yi
- Department of Clinical Laboratory, Jining No.1 People's Hospital, Jining, China
| | - Jafar Hajavi
- Department of Basic Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
29
|
Liu C, Zhang T, Chen L, Chen Y. The choice of anti-tumor strategies based on micromolecules or drug loading function of biomaterials. Cancer Lett 2020; 487:45-52. [PMID: 32474154 DOI: 10.1016/j.canlet.2020.05.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/08/2020] [Accepted: 05/15/2020] [Indexed: 01/22/2023]
Abstract
With advances in modern medicine, diverse tumor therapies have been developed. However, because of a lack of effective methods, the delivery of drugs or micromolecules in the human body has many limitations. Biomaterials are natural or synthetic functional materials that are prone to contact or interact with living systems. Therefore, the application of biomaterials provides innovative anti-tumor strategies, especially in tumor targeting, chemotherapy sensitization, tumor immunotherapy. The combination of biomaterials and drugs provides a promising strategy to overcome the biological barriers of drug delivery. Nanomaterials can target specific tumor sites to enhance the efficiency of tumor therapies and decrease the toxicity of drug through passive targeting, active targeting and direct targeting. Additionally, biomaterials can be used to enhance the sensitivity of tumor cells to chemotherapy drugs. Furthermore, modifiable biomaterials can induce effective anti-tumor immune response. Currently, the developmental trend of biomaterial for drug delivery is motivated by the combination and diversification of different therapies. With interdisciplinary development, a variety of anti-tumor strategies will emerge in an endless stream to bring great hope for tumor therapy. In this review, we will discuss the anti-tumor strategies based on nanoparticles and injectable scaffolds.
Collapse
Affiliation(s)
- Chengyi Liu
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China; Department of Urology, Lu'an Affiliated Hospital of Anhui Medical University, 237000, Anhui, China
| | - Tianke Zhang
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China; Department of Anorectal Surgery, Tianjin Union Medical Center, 300121, Tianjin, China
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, Tianjin, China
| | - Yue Chen
- Department of Urology, The Second Hospital of TianJin Medical University, TianJin Institute of Urology, Tianjin, 300211, China.
| |
Collapse
|
30
|
Alshamsan A, Binkhathlan Z, Kalam MA, Qamar W, Kfouri H, Alghonaim M, Lavasanifar A. Mitigation of Tacrolimus-Associated Nephrotoxicity by PLGA Nanoparticulate Delivery Following Multiple Dosing to Mice while Maintaining its Immunosuppressive Activity. Sci Rep 2020; 10:6675. [PMID: 32317681 PMCID: PMC7174389 DOI: 10.1038/s41598-020-63767-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to assess the ability of PLGA nanoparticles (NPs) to reduce the tacrolimus (TAC)-associated nephrotoxicity following multiple dose administration. The mean diameter of prepared NPs was in the range of 227 to 263 nm with an 8.32% drug loading (w/w). Moreover, in vitro release profile of TAC-loaded NPs showed a sustained release of the drug with only less than 30% release within 12 days. Flow cytometry as well as fluorescence microscopy results confirmed the uptake of FITC-labelled PLGA NPs by dendritic cells. The ex vivo study showed that TAC-loaded NPs caused a significant suppression of the proliferation of CD4+ and CD8+ cells, which was comparable to the control formulation (Prograf). In vivo immunosuppressive activity as well as the kidney function were assessed following drug administration to mice. The animals received TAC subcutaneously at a daily dose of 1 mg/kg for 30 days delivered as the control formulation (Prograf) or TAC-loaded NPs. The results revealed significantly lower drug-associated toxicity with an activity comparable to Prograf for TAC-loaded PLGA NPs. These findings show a potential for PLGA NPs in reducing the nephrotoxicity of TAC while preserving the immunosuppressive activity.
Collapse
Affiliation(s)
- Aws Alshamsan
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia. .,Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
| | - Ziyad Binkhathlan
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Mohd Abul Kalam
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Wajhul Qamar
- Central Laboratory, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hala Kfouri
- Department of Pathology, College of Medicine, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammed Alghonaim
- King Salman Bin Abdulaziz Chair for Kidney Disease, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.,Department of Chemical and Material Engineering, University of Alberta, Edmonton, Alberta, T6G 2V4, Canada
| |
Collapse
|
31
|
Wu M, Mei T, Lin C, Wang Y, Chen J, Le W, Sun M, Xu J, Dai H, Zhang Y, Xue C, Liu Z, Chen B. Melanoma Cell Membrane Biomimetic Versatile CuS Nanoprobes for Homologous Targeting Photoacoustic Imaging and Photothermal Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:16031-16039. [PMID: 32186357 DOI: 10.1021/acsami.9b23177] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Modulating the surface properties of nanoparticles (NPs) is an important approach to accomplish immune escape, prolonged the blood retention time, and enhance the ability of targeted drug delivery. The camouflage of cancer cell membrane onto nanoparticles has been proved to be an ideal approach to enhance active targeting ability of NPs. Herein, we isolated the membrane of melanoma cells to coat doxorubicin (DOX) and indocyanine green (ICG)-loaded hollow copper sulfide NPs (ID-HCuSNP@B16F10) for targeted photothermal therapy, photoacoustic imaging, and chemotherapy. A remarkable in vitro anticancer effect after irradiation and homologous targeting can be observed in B16F10 cells after the treatment of ID-HCuSNP@B16F10. Moreover, ID-HCuSNP@B16F10 exhibits excellent photothermal effect in melanoma animal models and achieves a high tumor ablation rate. This biomimetic system can realize high drug loading efficiency, enhanced targeting ability, and ideal antitumor efficiency.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Tianxiao Mei
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Chenyu Lin
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jingyao Chen
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenjun Le
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Mengyan Sun
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jianguo Xu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Haiying Dai
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yifan Zhang
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhongmin Liu
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Bingdi Chen
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
32
|
Chen Q, Bao Y, Burner D, Kaushal S, Zhang Y, Mendoza T, Bouvet M, Ozkan C, Minev B, Ma W. Tumor growth inhibition by mSTEAP peptide nanovaccine inducing augmented CD8 + T cell immune responses. Drug Deliv Transl Res 2020; 9:1095-1105. [PMID: 31228097 DOI: 10.1007/s13346-019-00652-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Poly(lactic-co-glycolic) acid (PLGA) has been successfully used in drug delivery and biomaterial applications, but very little attention has been directed towards the potential in vivo effects of peptide-loaded PLGA nanoparticles (NPs), specifically the potency of intravenous (IV) STEAP peptide-loaded PLGA-NP (nanovaccine) dosing and whether STEAP-specific CD8+ T cells directly play a key role in tumor inhibition. To address these concerns, syngeneic prostate cancer mouse models were established and treated with either mSTEAP peptide emulsified in incomplete Freund's adjuvant (IFA) via subcutaneous (SC) injection or mSTEAP peptide nanovaccine containing the same amount of peptide via IV or SC injection. Meanwhile, mice were treated with either CD8b mAb followed by nanovaccine treatment, free mSTEAP peptide, or empty PLGA-NPs. Immune responses in these mice were examined using cytotoxicity assays at 14 days after treatment. Tumor size and survival in various treatment groups were measured and monitored. The results demonstrated that mSTEAP peptide nanovaccine resulted in tumor inhibition by eliciting a significantly stronger CD8+ T cell immune response when compared with the controls. Moreover, the survival periods of mice treated with mSTEAP nanovaccine were significantly longer than those of mice treated with mSTEAP peptide emulsified in IFA or the treatment controls. Additionally, it was observed that the peptide nanovaccine was mainly distributed in the mouse liver and lungs after IV injection. These findings suggest that the peptide nanovaccine is a promising immunotherapeutic approach and offers a new opportunity for prostate cancer therapies.
Collapse
Affiliation(s)
- Qiuqiang Chen
- Key Laboratory for Translational Medicine, The First Affiliated Hospital of Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
- Department of Clinical Medicine, Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
| | - Ying Bao
- Key Laboratory for Translational Medicine, The First Affiliated Hospital of Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China
| | - Danielle Burner
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sharmeela Kaushal
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yu Zhang
- Materials Science and Engineering Program, Department of Mechanical Engineering, University of California Riverside, Riverside, CA, 92521, USA
- Mechanical and Automotive Engineering, School of Engineering, RMIT University, Melbourne, Victoria, 3083, Australia
| | - Theresa Mendoza
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Michael Bouvet
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cengiz Ozkan
- Materials Science and Engineering Program, Department of Mechanical Engineering, University of California Riverside, Riverside, CA, 92521, USA
| | - Boris Minev
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
- Calidi Biotherapeutics, San Diego, CA, 92121, USA.
| | - Wenxue Ma
- Department of Clinical Medicine, Huzhou University School of Medicine, Huzhou, 313000, Zhejiang, China.
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
33
|
Hashemi V, Farhadi S, Ghasemi Chaleshtari M, Seashore-Ludlow B, Masjedi A, Hojjat-Farsangi M, Namdar A, Ajjoolabady A, Mohammadi H, Ghalamfarsa G, Jadidi-Niaragh F. Nanomedicine for improvement of dendritic cell-based cancer immunotherapy. Int Immunopharmacol 2020; 83:106446. [PMID: 32244048 DOI: 10.1016/j.intimp.2020.106446] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has shown impressive outcomes, including the development of the first FDA-approved anti-cancer vaccine. However, the clinical application of DC-based cancer immunotherapy is associated with various challenges. Promising novel tools for the administration of cancer vaccines has emerged from recent developments in nanoscale biomaterials. One current strategy to enhance targeted drug delivery, while minimizing drug-related toxicities, is the use of nanoparticles (NPs). These can be utilized for antigen delivery into DCs, which have been shown to provide potent T cell-stimulating effects. Therefore, NP delivery represents one promising approach for creating an effective and stable immune response without toxic side effects. The current review surveys cancer immunotherapy with particular attention toward NP-based delivery methods that target DCs.
Collapse
Affiliation(s)
- Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Brinton Seashore-Ludlow
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Ali Masjedi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Amir Ajjoolabady
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
Emerging Prospects for Nanoparticle-Enabled Cancer Immunotherapy. J Immunol Res 2020; 2020:9624532. [PMID: 32377541 PMCID: PMC7199570 DOI: 10.1155/2020/9624532] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022] Open
Abstract
One of the standards for cancer treatment is cancer immunotherapy which treats both primary and metastasized tumors. Although cancer immunotherapeutics show better outcomes as compared with conventional approaches of cancer treatment, the currently used cancer immunotherapeutics have limited application in delivering cancer antigens to immune cells. Conversely, in solid tumors, tumor microenvironment suppresses the immune system leading to the evasion of anticancer immunity. Some promising attempts have been made to overcome these drawbacks by using different approaches, for instance, the use of biomaterial-based nanoparticles. Accordingly, various studies involving the application of nanoparticles in cancer immunotherapy have been discussed in this review article. This review not only describes the modes of cancer immunotherapy to reveal the importance of nanoparticles in this modality but also narrates nanoparticle-mediated delivery of cancer antigens and therapeutic supplements. Moreover, the impact of nanoparticles on the immunosuppressive behavior of tumor environment has been discussed. The last part of this review deals with cancer immunotherapy using a combination of traditional interventional oncology approach and image-guided local immunotherapy against cancer. According to recent studies, cancer therapy can potentially be improved through nanoparticle-based immunotherapy. In addition, drawbacks associated with the currently used cancer immunotherapeutics can be fixed by using nanoparticles.
Collapse
|
35
|
Pedersbæk D, Jønsson K, Madsen DV, Weller S, Bohn AB, Andresen TL, Simonsen JB. A quantitativeex vivostudy of the interactions between reconstituted high-density lipoproteins and human leukocytes. RSC Adv 2020; 10:3884-3894. [PMID: 35492676 PMCID: PMC9048990 DOI: 10.1039/c9ra08203d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/11/2020] [Indexed: 12/12/2022] Open
Abstract
Knowledge of the interactions between nanoparticles and immune cells is required for optimal design of nanoparticle-based drug delivery systems, either when aiming to avoid phagocytic clearance of the nanoparticles or promote an immune response by delivering therapeutic agents to specific immune cells. Several studies have suggested that reconstituted high-density lipoproteins (rHDL) are attractive drug delivery vehicles. However, detailed studies of rHDL interactions with circulating leukocytes are limited. Here, we evaluated the association of discoidal rHDL with leukocytes in human whole blood (HWB) using quantitative approaches. We found that while the rHDL of various lipid compositions associated preferentially with monocytes, the degree of association depended on the lipid composition. However, consistent with the long circulation half-life of rHDL, we show that only a minor fraction of the rHDL associated with the leukocytes. Furthermore, we used three-dimensional fluorescence microscopy and imaging flow cytometry to evaluate the possible internalization of rHDL cargo into the cells, and we show increased internalization of rHDL cargo in monocytes relative to granulocytes. The preferential rHDL association with monocytes and the internalization of rHDL cargo could possibly be mediated by the scavenger receptor class B type 1 (SR-BI), which we show is expressed to a higher extent on monocytes than on the other major leukocyte populations. Our work implies that drug-loaded rHDL can deliver its cargo to monocytes in circulation, which could lead to some off-target effects when using rHDL for systemic drug delivery, or it could pave the way for novel immunotherapeutic treatments aiming to target the monocytes. We used novel quantitative methods to study the interactions between reconstituted high-density lipoproteins (rHDL) and human leukocytes – showing that rHDL cargo are preferentially taken up by monocytes.![]()
Collapse
Affiliation(s)
- Dennis Pedersbæk
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Katrine Jønsson
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Ditte V. Madsen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Sven Weller
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Anja B. Bohn
- Department of Biomedicine
- Aarhus University
- 8000 Aarhus
- Denmark
| | - Thomas L. Andresen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Jens B. Simonsen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| |
Collapse
|
36
|
Artificially cloaked viral nanovaccine for cancer immunotherapy. Nat Commun 2019; 10:5747. [PMID: 31848338 PMCID: PMC6917704 DOI: 10.1038/s41467-019-13744-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022] Open
Abstract
Virus-based cancer vaccines are nowadays considered an interesting approach in the field of cancer immunotherapy, despite the observation that the majority of the immune responses they elicit are against the virus and not against the tumor. In contrast, targeting tumor associated antigens is effective, however the identification of these antigens remains challenging. Here, we describe ExtraCRAd, a multi-vaccination strategy focused on an oncolytic virus artificially wrapped with tumor cancer membranes carrying tumor antigens. We demonstrate that ExtraCRAd displays increased infectivity and oncolytic effect in vitro and in vivo. We show that this nanoparticle platform controls the growth of aggressive melanoma and lung tumors in vivo both in preventive and therapeutic setting, creating a highly specific anti-cancer immune response. In conclusion, ExtraCRAd might serve as the next generation of personalized cancer vaccines with enhanced features over standard vaccination regimens, representing an alternative way to target cancer. Cancer therapy using oncolytic virus has shown pre-clinical and clinical efficacy. Here, the authors report ExtraCRAd, an oncolytic virus cloaked with tumour cell membrane and report its therapeutic effects in vitro and in vivo in multiple mouse tumour models.
Collapse
|
37
|
Kim CG, Kye YC, Yun CH. The Role of Nanovaccine in Cross-Presentation of Antigen-Presenting Cells for the Activation of CD8 + T Cell Responses. Pharmaceutics 2019; 11:E612. [PMID: 31731667 PMCID: PMC6920862 DOI: 10.3390/pharmaceutics11110612] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 01/30/2023] Open
Abstract
Explosive growth in nanotechnology has merged with vaccine development in the battle against diseases caused by bacterial or viral infections and malignant tumors. Due to physicochemical characteristics including size, viscosity, density and electrostatic properties, nanomaterials have been applied to various vaccination strategies. Nanovaccines, as they are called, have been the subject of many studies, including review papers from a material science point of view, although a mode of action based on a biological and immunological understanding has yet to emerge. In this review, we discuss nanovaccines in terms of CD8+ T cell responses, which are essential for antiviral and anticancer therapies. We focus mainly on the role and mechanism, with particular attention to the functional aspects, of nanovaccines in inducing cross-presentation, an unconventional type of antigen-presentation that activates CD8+ T cells upon administration of exogenous antigens, in dendritic cells followed by activation of antigen-specific CD8+ T cell responses. Two major intracellular mechanisms that nanovaccines harness for cross-presentation are described; one is endosomal swelling and rupture, and the other is membrane fusion. Both processes eventually allow exogenous vaccine antigens to be exported from phagosomes to the cytosol followed by loading on major histocompatibility complex class I, triggering clonal expansion of CD8+ T cells. Advancement of nanotechnology with an enhanced understanding of how nanovaccines work will contribute to the design of more effective and safer nanovaccines.
Collapse
Affiliation(s)
- Cheol Gyun Kim
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
| | - Yoon-Chul Kye
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (C.G.K.); (Y.-C.K.)
- Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, Korea
| |
Collapse
|
38
|
Dhakal S, Renukaradhya GJ. Nanoparticle-based vaccine development and evaluation against viral infections in pigs. Vet Res 2019; 50:90. [PMID: 31694705 PMCID: PMC6833244 DOI: 10.1186/s13567-019-0712-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 10/20/2019] [Indexed: 11/10/2022] Open
Abstract
Virus infections possess persistent health challenges in swine industry leading to severe economic losses worldwide. The economic burden caused by virus infections such as Porcine Reproductive and Respiratory Syndrome Virus, Swine influenza virus, Porcine Epidemic Diarrhea Virus, Porcine Circovirus 2, Foot and Mouth Disease Virus and many others are associated with severe morbidity, mortality, loss of production, trade restrictions and investments in control and prevention practices. Pigs can also have a role in zoonotic transmission of some viral infections to humans. Inactivated and modified-live virus vaccines are available against porcine viral infections with variable efficacy under field conditions. Thus, improvements over existing vaccines are necessary to: (1) Increase the breadth of protection against evolving viral strains and subtypes; (2) Control of emerging and re-emerging viruses; (3) Eradicate viruses localized in different geographic areas; and (4) Differentiate infected from vaccinated animals to improve disease control programs. Nanoparticles (NPs) generated from virus-like particles, biodegradable and biocompatible polymers and liposomes offer many advantages as vaccine delivery platform due to their unique physicochemical properties. NPs help in efficient antigen internalization and processing by antigen presenting cells and activate them to elicit innate and adaptive immunity. Some of the NPs-based vaccines could be delivered through both parenteral and mucosal routes to trigger efficient mucosal and systemic immune responses and could be used to target specific immune cells such as mucosal microfold (M) cells and dendritic cells (DCs). In conclusion, NPs-based vaccines can serve as novel candidate vaccines against several porcine viral infections with the potential to enhance the broader protective efficacy under field conditions. This review highlights the recent developments in NPs-based vaccines against porcine viral pathogens and how the NPs-based vaccine delivery system induces innate and adaptive immune responses resulting in varied level of protective efficacy.
Collapse
Affiliation(s)
- Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691 USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Gourapura J. Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691 USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
39
|
Kordalivand N, Tondini E, Lau CYJ, Vermonden T, Mastrobattista E, Hennink WE, Ossendorp F, Nostrum CFV. Cationic synthetic long peptides-loaded nanogels: An efficient therapeutic vaccine formulation for induction of T-cell responses. J Control Release 2019; 315:114-125. [PMID: 31672626 DOI: 10.1016/j.jconrel.2019.10.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/18/2022]
Abstract
Recent studies have shown a high potency of protein-based vaccines for cell-mediated cancer immunotherapy. However, due to their poor cellular uptake, efficient immune responses with soluble protein antigens are often not observed. As a result of superior cellular uptake, nanogels loaded with antigenic peptides were investigated in this study as carrier systems for cancer immunotherapy. Different synthetic long peptides (SLPs) containing the CTL and CD4+ T-helper (Help) epitopes were synthesized and covalently conjugated via disulfide bonds to the polymeric network of cationic dextran nanogels. Cationic nanogels with a size of 210 nm, positive zeta potential (+24 mV) and high peptide loading content (15%) showed triggered release of the loaded peptides under reducing conditions. An in vitro study demonstrated the capability of cationic nanogels to maturate dendritic cells (DCs). Importantly, covalently SLP-loaded nanogels adjuvanted with poly(I:C) showed superior CD8+ T cell responses compared to soluble peptides and nanogel formulations with physically loaded peptides both in vitro and in vivo. In conclusion, covalently SLPs-loaded cationic nanogels are a promising system to provoke immune responses for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Neda Kordalivand
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Chun Yin Jerry Lau
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
40
|
Gastrodin, a traditional Chinese medicine monomer compound, can be used as adjuvant to enhance the immunogenicity of melanoma vaccines. Int Immunopharmacol 2019; 74:105699. [DOI: 10.1016/j.intimp.2019.105699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/26/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
|
41
|
Shae D, Baljon JJ, Wehbe M, Becker KW, Sheehy TL, Wilson JT. At the bench: Engineering the next generation of cancer vaccines. J Leukoc Biol 2019; 108:1435-1453. [PMID: 31430398 DOI: 10.1002/jlb.5bt0119-016r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/29/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer vaccines hold promise as an immunotherapeutic modality based on their potential to generate tumor antigen-specific T cell responses and long-lived antitumor responses capable of combating metastatic disease and recurrence. However, cancer vaccines have historically failed to deliver significant therapeutic benefit in the clinic, which we maintain is due in part to drug delivery challenges that have limited vaccine immunogenicity and efficacy. In this review, we examine some of the known and putative failure mechanisms of common first-generation clinical cancer vaccines, and describe how the rational design of materials engineered for vaccine delivery and immunomodulation can address these shortcomings. First, we outline vaccine design principles for augmenting cellular immunity to tumor antigens and describe how well-engineered materials can improve vaccine efficacy, highlighting recent innovations in vaccine delivery technology that are primed for integration into neoantigen vaccine development pipelines. We also discuss the importance of sequencing, timing, and kinetics in mounting effective immune responses to cancer vaccines, and highlight examples of materials that potentiate antitumor immunity through spatiotemporal control of immunomodulation. Furthermore, we describe several engineering strategies for improving outcomes of in situ cancer vaccines, which leverage local, intratumoral delivery to stimulate systemic immunity. Finally, we highlight recent innovations leveraging nanotechnology for increasing the immunogenicity of the tumor microenvironment (TME), which is critical to enhancing tumor infiltration and function of T cells elicited in response to cancer vaccines. These immunoengineering strategies and tools complement ongoing advances in cancer vaccines as they reemerge as an important component of the immunotherapeutic armamentarium.
Collapse
Affiliation(s)
- Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jessalyn J Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Kyle W Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Taylor L Sheehy
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - John Tanner Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
42
|
Chen M, Chen M, He J. Cancer cell membrane cloaking nanoparticles for targeted co-delivery of doxorubicin and PD-L1 siRNA. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1635-1641. [PMID: 31027450 DOI: 10.1080/21691401.2019.1608219] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nanoparticles coated with cell membranes have been garnering growing attention due to their homologous binding capability of membrane molecules and consequent self-recognition by their source cells. In the present study, we report on the construction of doxorubicin and PD-L1 siRNA-loaded PLGA nanoparticles and their biological functionalization by cancer cell-derived membrane cloaking. The resulting cancer cell membrane-coated nanoparticles (CCMNPs) presented a core-shell nanostructure with highly specific self-recognition affinity to the homotypic cells, which can be attributed to the transference of cell adhesion molecules with homotypic binding properties. These findings facilitate the application of this bioinspired strategy for effective delivery of siRNA and precise tumour therapy.
Collapse
Affiliation(s)
- Mushi Chen
- a Xiangya Hospital , Central South University , Changsha , P.R. China
| | - Ming Chen
- b Nanhai District People's Hospital , Foshan , P.R. China
| | - Jiantai He
- a Xiangya Hospital , Central South University , Changsha , P.R. China
| |
Collapse
|
43
|
Strategies for Targeting Cancer Immunotherapy Through Modulation of the Tumor Microenvironment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00113-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
TLR4 Agonist Monophosphoryl Lipid A Alleviated Radiation-Induced Intestinal Injury. J Immunol Res 2019; 2019:2121095. [PMID: 31275998 PMCID: PMC6589195 DOI: 10.1155/2019/2121095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/03/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
The small intestine is one of the most sensitive organs to irradiation injury, and the development of high effective radioprotectants especially with low toxicity for intestinal radiation sickness is urgently needed. Monophosphoryl lipid A (MPLA) was found to be radioprotective in our previous study, while its effect against the intestinal radiation injury remained unknown. In the present study, we firstly determined the intestinal apoptosis after irradiation injury according to the TUNEL assay. Subsequently, we adopted the immunofluorescence technique to assess the expression levels of different biomarkers including Ki67, γ-H2AX, and defensin 1 in vivo. Additionally, the inflammatory cytokines were detected by RT-PCR. Our data indicated that MPLA could protect the intestine from ionizing radiation (IR) damage through activating TLR4 signal pathway and regulating the inflammatory cytokines. This research shed new light on the protective effect of the novel TLR4 agonist MPLA against intestine detriment induced by IR.
Collapse
|
45
|
|
46
|
Wakabayashi R, Kono H, Kozaka S, Tahara Y, Kamiya N, Goto M. Transcutaneous Codelivery of Tumor Antigen and Resiquimod in Solid-in-Oil Nanodispersions Promotes Antitumor Immunity. ACS Biomater Sci Eng 2019; 5:2297-2306. [DOI: 10.1021/acsbiomaterials.9b00260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Koerner J, Horvath D, Groettrup M. Harnessing Dendritic Cells for Poly (D,L-lactide- co-glycolide) Microspheres (PLGA MS)-Mediated Anti-tumor Therapy. Front Immunol 2019; 10:707. [PMID: 31024545 PMCID: PMC6460768 DOI: 10.3389/fimmu.2019.00707] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
With emerging success in fighting off cancer, chronic infections, and autoimmune diseases, immunotherapy has become a promising therapeutic approach compared to conventional therapies such as surgery, chemotherapy, radiation therapy, or immunosuppressive medication. Despite the advancement of monoclonal antibody therapy against immune checkpoints, the development of safe and efficient cancer vaccine formulations still remains a pressing medical need. Anti-tumor immunotherapy requires the induction of antigen-specific CD8+ cytotoxic T lymphocyte (CTL) responses which recognize and specifically destroy tumor cells. Due to the crucial role of dendritic cells (DCs) in initiating anti-tumor immunity, targeting tumor antigens to DCs has become auspicious in modern vaccine research. Over the last two decades, micron- or nanometer-sized particulate delivery systems encapsulating tumor antigens and immunostimulatory molecules into biodegradable polymers have shown great promise for the induction of potent, specific and long-lasting anti-tumor responses in vivo. Enhanced vaccine efficiency of the polymeric micro/nanoparticles has been attributed to controlled and continuous release of encapsulated antigens, efficient targeting of antigen presenting cells (APCs) such as DCs and subsequent induction of CTL immunity. Poly (D, L-lactide-co-glycolide) (PLGA), as one of these polymers, has been extensively studied for the design and development of particulate antigen delivery systems in cancer therapy. This review provides an overview of the current state of research on the application of PLGA microspheres (PLGA MS) as anti-tumor cancer vaccines in activating and potentiating immune responses attempting to highlight their potential in the development of cancer therapeutics.
Collapse
Affiliation(s)
- Julia Koerner
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Dennis Horvath
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| |
Collapse
|
48
|
Hassan HAFM, Diebold SS, Smyth LA, Walters AA, Lombardi G, Al-Jamal KT. Application of carbon nanotubes in cancer vaccines: Achievements, challenges and chances. J Control Release 2019; 297:79-90. [PMID: 30659906 DOI: 10.1016/j.jconrel.2019.01.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Tumour-specific, immuno-based therapeutic interventions can be considered as safe and effective approaches for cancer therapy. Exploitation of nano-vaccinology to intensify the cancer vaccine potency may overcome the need for administration of high vaccine doses or additional adjuvants and therefore could be a more efficient approach. Carbon nanotube (CNT) can be described as carbon sheet(s) rolled up into a cylinder that is nanometers wide and nanometers to micrometers long. Stemming from the observed capacities of CNTs to enter various types of cells via diversified mechanisms utilising energy-dependent and/or passive routes of cell uptake, the use of CNTs for the delivery of therapeutic agents has drawn increasing interests over the last decade. Here we review the previous studies that demonstrated the possible benefits of these cylindrical nano-vectors as cancer vaccine delivery systems as well as the obstacles their clinical application is facing.
Collapse
Affiliation(s)
- Hatem A F M Hassan
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Sandra S Diebold
- Biotherapeutics Division, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Lesley A Smyth
- School of Health, Sport and Biosciences, University of East London, Stratford Campus, Water Lane, London E15 4LZ, United Kingdom
| | - Adam A Walters
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom
| | - Giovanna Lombardi
- School of Immunology and Microbial Sciences, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, United Kingdom.
| |
Collapse
|
49
|
Affiliation(s)
- Jenny Lou
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| | - Li Zhang
- Toronto General Hospital Research InstituteUniversity Health Network Toronto M5G 2C4 Canada
- Department of ImmunologyUniversity of Toronto Toronto M5S 1A8 Canada
- Department of Laboratory Medicine and PathobiologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Gang Zheng
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| |
Collapse
|
50
|
Exploiting PLGA-Based Biocompatible Nanoparticles for Next-Generation Tolerogenic Vaccines against Autoimmune Disease. Int J Mol Sci 2019; 20:ijms20010204. [PMID: 30626016 PMCID: PMC6337481 DOI: 10.3390/ijms20010204] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 12/01/2022] Open
Abstract
Tolerogenic vaccines are aimed at inhibiting antigen-specific immune responses. Antigen-loaded nanoparticles (NPs) have been recently emerged as ideal tools for tolerogenic vaccination because their composition, size, and capability of loading immunomodulatory molecules can be readily exploited to induce peripheral tolerance. Among polymeric NPs, poly(lactic-co-glycolic acid) (PLGA) NPs have the advantage of currently holding approval for several applications in drug delivery, diagnostics, and other clinical uses by the Food and Drug Administration (FDA). PLGA-NPs are non-toxic and display excellent biocompatibility and biodegradability properties. Moreover, surface functionalization may improve their interaction with biological materials, thereby optimizing targeting and performance. PLGA-NPs are the most extensively studied in pre-clinical model in the field of tolerogenic vaccination. Thus, this review describes their potential applications in the treatment of autoimmune diseases.
Collapse
|