1
|
Srinivasarao DA, Shah S, Famta P, Vambhurkar G, Jain N, Pindiprolu SKSS, Sharma A, Kumar R, Padhy HP, Kumari M, Madan J, Srivastava S. Unravelling the role of tumor microenvironment responsive nanobiomaterials in spatiotemporal controlled drug delivery for lung cancer therapy. Drug Deliv Transl Res 2025; 15:407-435. [PMID: 39037533 DOI: 10.1007/s13346-024-01673-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Design and development of efficient drug delivery technologies that impart site-specificity is the need of the hour for the effective treatment of lung cancer. The emergence of materials science and nanotechnology partially helped drug delivery scientists to achieve this objective. Various stimuli-responsive materials that undergo degradation at the pathological tumor microenvironment (TME) have been developed and explored for drug delivery applications using nanotechnological approaches. Nanoparticles (NPs), owing to their small size and high surface area to volume ratio, demonstrated enhanced cellular internalization, permeation, and retention at the tumor site. Such passive accumulation of stimuli-responsive materials helped to achieve spatiotemporally controlled and targeted drug delivery within the tumors. In this review, we discussed various stimuli-physical (interstitial pressure, temperature, and stiffness), chemical (pH, hypoxia, oxidative stress, and redox state), and biological (receptor expression, efflux transporters, immune cells, and their receptors or ligands)-that are characteristic to the TME. We mentioned an array of biomaterials-based nanoparticulate delivery systems that respond to these stimuli and control drug release at the TME. Further, we discussed nanoparticle-based combinatorial drug delivery strategies. Finally, we presented our perspectives on challenges related to scale-up, clinical translation, and regulatory approvals.
Collapse
Affiliation(s)
- Dadi A Srinivasarao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, 533 437, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, 533 003, Andhra Pradesh, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Meenu Kumari
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
2
|
Pednekar K, Minnee J, de Vries IJM, Prakash J. Targeted nanomedicine for reprogramming the tumor innate immune system: From bench to bedside. Eur J Pharm Biopharm 2024; 204:114510. [PMID: 39307440 DOI: 10.1016/j.ejpb.2024.114510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Tumor-associated innate immune cells such as tumor-associated macrophages, neutrophils, dendritic cells play a crucial role in tumor progression, angiogenesis and metastasis. These cells also control the efficacy of chemotherapy and immunotherapy by inducing drug resistance and immunosuppression, leading to therapeutic failures. Therefore, targeting the tumor-associated innate immune cells has gained high attention for the development of effective cancer therapy. Nanomedicine based strategies to target these cells are highly relevant and can be used to reprogram these cells. In this review, we discuss the fundamental roles of the tumor-associated innate immune cells in the tumor microenvironment and different strategies to modulate them. Then, nanomedicine-based strategies to target different tumor innate immune cells are explained in detail. While the clinical development of the targeted nanomedicine remains a great challenge in practice, we have provided our perspectives on various factors such as pharmaceutical aspects, preclinical testing and biological aspects which are crucial to consider before translating these targeting strategies to clinics.
Collapse
Affiliation(s)
- Kunal Pednekar
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Julia Minnee
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jai Prakash
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
3
|
Zachová K, Bartheldyová E, Hubatka F, Křupka M, Odehnalová N, Turánek Knötigová P, Vaškovicová N, Sloupenská K, Hromádka R, Paulovičová E, Effenberg R, Ledvina M, Raška M, Turánek J. The immunogenicity of p24 protein from HIV-1 virus is strongly supported and modulated by coupling with liposomes and mannan. Carbohydr Polym 2024; 332:121844. [PMID: 38431385 DOI: 10.1016/j.carbpol.2024.121844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/31/2023] [Accepted: 01/18/2024] [Indexed: 03/05/2024]
Abstract
Anti-viral and anti-tumor vaccines aim to induce cytotoxic CD8+ T cells (CTL) and antibodies. Conserved protein antigens, such as p24 from human immunodeficiency virus, represent promising component for elicitation CTLs, nevertheless with suboptimal immunogenicity, if formulated as recombinant protein. To enhance immunogenicity and CTL response, recombinant proteins may be targeted to dendritic cells (DC) for cross presentation on MHCI, where mannose receptor and/or other lectin receptors could play an important role. Here, we constructed liposomal carrier-based vaccine composed of recombinant p24 antigen bound by metallochelating linkage onto surface of nanoliposomes with surface mannans coupled by aminooxy ligation. Generated mannosylated proteonanoliposomes were analyzed by dynamic light scattering, isothermal titration, and electron microscopy. Using murine DC line MutuDC and murine bone marrow derived DC (BMDC) we evaluated their immunogenicity and immunomodulatory activity. We show that p24 mannosylated proteonanoliposomes activate DC for enhanced MHCI, MHCII and CD40, CD80, and CD86 surface expression both on MutuDC and BMDC. p24 mannosylated liposomes were internalized by MutuDC with p24 intracellular localization within 1 to 3 h. The combination of metallochelating and aminooxy ligation could be used simultaneously to generate nanoliposomal adjuvanted recombinant protein-based vaccines versatile for combination of recombinant antigens relevant for antibody and CTL elicitation.
Collapse
Affiliation(s)
- K Zachová
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - E Bartheldyová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - F Hubatka
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - M Křupka
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - N Odehnalová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - P Turánek Knötigová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - N Vaškovicová
- Faculty of Medicine, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - K Sloupenská
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - R Hromádka
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - E Paulovičová
- Center for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia
| | - R Effenberg
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, Prague, Czech Republic
| | - M Ledvina
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, Prague, Czech Republic
| | - M Raška
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic.
| | - J Turánek
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic; C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic; Institute of Clinical Immunology & Allergology, Charles University Prague and University Hospital, Hradec Kralove, Sokolská 581, Hradec Kralove, Czech Republic.
| |
Collapse
|
4
|
Paurević M, Šrajer Gajdošik M, Ribić R. Mannose Ligands for Mannose Receptor Targeting. Int J Mol Sci 2024; 25:1370. [PMID: 38338648 PMCID: PMC10855088 DOI: 10.3390/ijms25031370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The mannose receptor (MR, CD 206) is an endocytic receptor primarily expressed by macrophages and dendritic cells, which plays a critical role in both endocytosis and antigen processing and presentation. MR carbohydrate recognition domains (CRDs) exhibit a high binding affinity for branched and linear oligosaccharides. Furthermore, multivalent mannose presentation on the various templates like peptides, proteins, polymers, micelles, and dendrimers was proven to be a valuable approach for the selective and efficient delivery of various therapeutically active agents to MR. This review provides a detailed account of the most relevant and recent aspects of the synthesis and application of mannosylated bioactive formulations for MR-mediated delivery in treatments of cancer and other infectious diseases. It further highlights recent findings related to the necessary structural features of the mannose-containing ligands for successful binding to the MR.
Collapse
Affiliation(s)
- Marija Paurević
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia; (M.P.); (M.Š.G.)
| | - Martina Šrajer Gajdošik
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia; (M.P.); (M.Š.G.)
| | - Rosana Ribić
- Department of Nursing, University Center Varaždin, University North, Jurja Križanića 31b, HR-42000 Varaždin, Croatia
| |
Collapse
|
5
|
Pundkar C, Antony F, Kang X, Mishra A, Babu RJ, Chen P, Li F, Suryawanshi A. Targeting Wnt/β-catenin signaling using XAV939 nanoparticles in tumor microenvironment-conditioned macrophages promote immunogenicity. Heliyon 2023; 9:e16688. [PMID: 37313143 PMCID: PMC10258387 DOI: 10.1016/j.heliyon.2023.e16688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
The aberrant activation of Wnt/β-catenin signaling in tumor cells and immune cells in the tumor microenvironment (TME) promotes malignant transformation, metastasis, immune evasion, and resistance to cancer treatments. The increased Wnt ligand expression in TME activates β-catenin signaling in antigen (Ag)-presenting cells (APCs) and regulates anti-tumor immunity. Previously, we showed that activation of Wnt/β-catenin signaling in dendritic cells (DCs) promotes induction of regulatory T cell responses over anti-tumor CD4+ and CD8+ effector T cell responses and promotes tumor progression. In addition to DCs, tumor-associated macrophages (TAMs) also serve as APCs and regulate anti-tumor immunity. However, the role of β-catenin activation and its effect on TAM immunogenicity in TME is largely undefined. In this study, we investigated whether inhibiting β-catenin in TME-conditioned macrophages promotes immunogenicity. Using nanoparticle formulation of XAV939 (XAV-Np), a tankyrase inhibitor that promotes β-catenin degradation, we performed in vitro macrophage co-culture assays with melanoma cells (MC) or melanoma cell supernatants (MCS) to investigate the effect on macrophage immunogenicity. We show that XAV-Np-treatment of macrophages conditioned with MC or MCS significantly upregulates the cell surface expression of CD80 and CD86 and suppresses the expression of PD-L1 and CD206 compared to MC or MCS-conditioned macrophages treated with control nanoparticle (Con-Np). Further, XAV-Np-treated macrophages conditioned with MC or MCS significantly increased IL-6 and TNF-α production, with reduced IL-10 production compared to Con-Np-treated macrophages. Moreover, the co-culture of MC and XAV-Np-treated macrophages with T cells resulted in increased CD8+ T cell proliferation compared to Con-Np-treated macrophages. These data suggest that targeted β-catenin inhibition in TAMs represents a promising therapeutic approach to promote anti-tumor immunity.
Collapse
Affiliation(s)
- Chetan Pundkar
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Ferrin Antony
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Pengyu Chen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Feng Li
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
6
|
De Mel J, Hossain M, Shofolawe-Bakare O, Mohammad SA, Rasmussen E, Milloy K, Shields M, Roth EW, Arora K, Cueto R, Tang SC, Wilson JT, Smith AE, Werfel TA. Dual-Responsive Glycopolymers for Intracellular Codelivery of Antigen and Lipophilic Adjuvants. Mol Pharm 2022; 19:4705-4716. [PMID: 36374992 PMCID: PMC10013197 DOI: 10.1021/acs.molpharmaceut.2c00750] [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] [Indexed: 11/16/2022]
Abstract
Traditional approaches to vaccines use whole organisms to trigger an immune response, but they do not typically generate robust cellular-mediated immunity and have various safety risks. Subunit vaccines composed of proteins and/or peptides represent an attractive and safe alternative to whole organism vaccines, but they are poorly immunogenic. Though there are biological reasons for the poor immunogenicity of proteins and peptides, one other key to their relative lack of immunogenicity could be attributed to the poor pharmacokinetic properties of exogenously delivered proteins and peptides. For instance, peptides often aggregate at the site of injection and are not stable in biological fluids, proteins and peptides are rapidly cleared from circulation, and both have poor cellular internalization and endosomal escape. Herein, we developed a delivery system to address the lack of protein immunogenicity by overcoming delivery barriers as well as codelivering immune-stimulating adjuvants. The glycopolymeric nanoparticles (glycoNPs) are composed of a dual-stimuli-responsive block glycopolymer, poly[2-(diisopropylamino)ethyl methacrylate]-b-poly[(pyridyl disulfide ethyl methacrylate)-co-(methacrylamidoglucopyranose)] (p[DPA-b-(PDSMA-co-MAG)]). This polymer facilitates protein conjugation and cytosolic release, the pH-responsive release of lipophilic adjuvants, and pH-dependent membrane disruption to ensure cytosolic delivery of antigens. We synthesized p[DPA-b-(PDSMA-co-MAG)] by reversible addition-fragmentation chain transfer (RAFT) polymerization, followed by the formation and physicochemical characterization of glycoNPs using the p[DPA-b-(PDSMA-co-MAG)] building blocks. These glycoNPs conjugated the model antigen ovalbumin (OVA) and released OVA in response to elevated glutathione levels. Moreover, the glycoNPs displayed pH-dependent drug release of the model hydrophobic drug Nile Red while also exhibiting pH-responsive endosomolytic behavior as indicated by a red blood cell hemolysis assay. GlycoNPs coloaded with OVA and the toll-like receptor 7/8 (TLR-7/8) agonist Resiquimod (R848) activated DC 2.4 dendritic cells (DCs) significantly more than free OVA and R848 and led to robust antigen presentation of the OVA epitope SIINFEKL on major histocompatibility complex I (MHC-I). In sum, the dual-stimuli-responsive glycopolymer introduced here overcomes major protein and peptide delivery barriers and could vastly improve the immunogenicity of protein-based vaccines.
Collapse
Affiliation(s)
- Judith De Mel
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Mehjabeen Hossain
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Oluwaseyi Shofolawe-Bakare
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Sk Arif Mohammad
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Emily Rasmussen
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Khadeeja Milloy
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Micaela Shields
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Eric W Roth
- Northwestern University Atomic and Nanoscale Characterization Experimental Center, Evanston, Illinois, 60208, United States
| | - Karan Arora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Rafael Cueto
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Adam E Smith
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Thomas A Werfel
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| |
Collapse
|
7
|
Mamun AA, Uddin MS, Perveen A, Jha NK, Alghamdi BS, Jeandet P, Zhang HJ, Ashraf GM. Inflammation-targeted nanomedicine against brain cancer: From design strategies to future developments. Semin Cancer Biol 2022; 86:101-116. [PMID: 36084815 DOI: 10.1016/j.semcancer.2022.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 08/08/2022] [Accepted: 08/21/2022] [Indexed: 02/07/2023]
Abstract
Brain cancer is an aggressive type of cancer with poor prognosis. While the immune system protects against cancer in the early stages, the tumor exploits the healing arm of inflammatory reactions to accelerate its growth and spread. Various immune cells penetrate the developing tumor region, establishing a pro-inflammatory tumor milieu. Additionally, tumor cells may release chemokines and cytokines to attract immune cells and promote cancer growth. Inflammation and its associated mechanisms in the progression of cancer have been extensively studied in the majority of solid tumors, especially brain tumors. However, treatment of the malignant brain cancer is hindered by several obstacles, such as the blood-brain barrier, transportation inside the brain interstitium, inflammatory mediators that promote tumor growth and invasiveness, complications in administering therapies to tumor cells specifically, the highly invasive nature of gliomas, and the resistance to drugs. To resolve these obstacles, nanomedicine could be a potential strategy that has facilitated advancements in diagnosing and treating brain cancer. Due to the numerous benefits provided by their small size and other features, nanoparticles have been a prominent focus of research in the drug-delivery field. The purpose of this article is to discuss the role of inflammatory mediators and signaling pathways in brain cancer as well as the recent advances in understanding the nano-carrier approaches for enhancing drug delivery to the brain in the treatment of brain cancer.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences, Uttaranchal University, Dehradun 248007, India
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Philippe Jeandet
- University of Reims Champagne-Ardenne, Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, PO Box 1039, 51687 Reims Cedex 2, France
| | - Hong-Jie Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
8
|
Yamada Y, Sato Y, Nakamura T, Harashima H. Innovative cancer nanomedicine based on immunology, gene editing, intracellular trafficking control. J Control Release 2022; 348:357-369. [PMID: 35623492 DOI: 10.1016/j.jconrel.2022.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
The recent rapid progress in the area of drug delivery systems (DDS) has opened a new era in medicine with a strong linkage to understanding the molecular mechanisms associated with cancer survival. In this review, we summarize new cancer strategies that have recently been developed based on our DDS technology. Cancer immunotherapy will be improved based on the concept of the cancer immunity cycle, which focuses on dynamic interactions between various types of cancer and immune cells in our body. The new technology of genome editing will also be discussed with reference to how these new DDS technologies can be used to introduce therapeutic cargoes into our body. Lastly, a new organelle, mitochondria will be the focus of creating a new cancer treatment strategy by a MITO-Porter which can deliver macromolecules directly to mitochondria of cancer cells via a membrane fusion approach and the impact of controlled intracellular trafficking will be discussed.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Japan Science and Technology Agency (JST) Fusion Oriented REsearch for disruptive Science and Technology (FOREST) Program, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
9
|
Li X, Omonova Tuychi Qizi C, Mohamed Khamis A, Zhang C, Su Z. Nanotechnology for Enhanced Cytoplasmic and Organelle Delivery of Bioactive Molecules to Immune Cells. Pharm Res 2022; 39:1065-1083. [PMID: 35661086 DOI: 10.1007/s11095-022-03284-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Immune cells stand as a critical component of the immune system to maintain the internal environment homeostasis. The dysfunction of immune cells can result in various life-threatening diseases, including refractory infection, diabetes, cardiovascular disease, and cancer. Therefore, strategies to standardize or even enhance the function of immune cells are critical. Recently, nanotechnology has been highly researched and extensively applied for enhancing the cytoplasmic delivery of bioactive molecules to immune cells, providing efficient approaches to correct in vivo and in vitro dysfunction of immune cells. This review focuses on the technologies and challenges involved in improving endo-lysosomal escape, cytoplasmic release and organelle targeted delivery of different bioactive molecules in immune cells. Furthermore, it will elaborate on the broader vision of applying nanotechnology for treating immune cell-related diseases and constructing immune therapies and cytopharmaceuticals as potential treatments for diseases.
Collapse
Affiliation(s)
- Xiaoyu Li
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Charos Omonova Tuychi Qizi
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Amari Mohamed Khamis
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Glass EB, Hoover AA, Bullock KK, Madden MZ, Reinfeld BI, Harris W, Parker D, Hufnagel DH, Crispens MA, Khabele D, Rathmell WK, Rathmell JC, Wilson AJ, Giorgio TD, Yull FE. Stimulating TAM-mediated anti-tumor immunity with mannose-decorated nanoparticles in ovarian cancer. BMC Cancer 2022; 22:497. [PMID: 35513776 PMCID: PMC9074180 DOI: 10.1186/s12885-022-09612-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/21/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Current cancer immunotherapies have made tremendous impacts but generally lack high response rates, especially in ovarian cancer. New therapies are needed to provide increased benefits. One understudied approach is to target the large population of immunosuppressive tumor-associated macrophages (TAMs). Using inducible transgenic mice, we recently reported that upregulating nuclear factor-kappaB (NF-κB) signaling in TAMs promotes the M1, anti-tumor phenotype and limits ovarian cancer progression. We also developed a mannose-decorated polymeric nanoparticle system (MnNPs) to preferentially deliver siRNA payloads to M2, pro-tumor macrophages in vitro. In this study, we tested a translational strategy to repolarize ovarian TAMs via MnNPs loaded with siRNA targeting the inhibitor of NF-κB alpha (IκBα) using mouse models of ovarian cancer. METHODS We evaluated treatment with MnNPs loaded with IκBα siRNA (IκBα-MnNPs) or scrambled siRNA in syngeneic ovarian cancer models. ID8 tumors in C57Bl/6 mice were used to evaluate consecutive-day treatment of late-stage disease while TBR5 tumors in FVB mice were used to evaluate repetitive treatments in a faster-developing disease model. MnNPs were evaluated for biodistribution and therapeutic efficacy in both models. RESULTS Stimulation of NF-κB activity and repolarization to an M1 phenotype via IκBα-MnNP treatment was confirmed using cultured luciferase-reporter macrophages. Delivery of MnNPs with fluorescent payloads (Cy5-MnNPs) to macrophages in the solid tumors and ascites was confirmed in both tumor models. A three consecutive-day treatment of IκBα-MnNPs in the ID8 model validated a shift towards M1 macrophage polarization in vivo. A clear therapeutic effect was observed with biweekly treatments over 2-3 weeks in the TBR5 model where significantly reduced tumor burden was accompanied by changes in immune cell composition, indicative of reduced immunosuppressive tumor microenvironment. No evidence of toxicity associated with MnNP treatment was observed in either model. CONCLUSIONS In mouse models of ovarian cancer, MnNPs were preferentially associated with macrophages in ascites fluid and solid tumors. Evidence of macrophage repolarization, increased inflammatory cues, and reduced tumor burden in IκBα-MnNP-treated mice indicate beneficial outcomes in models of established disease. We have provided evidence of a targeted, TAM-directed approach to increase anti-tumor immunity in ovarian cancer with strong translational potential for future clinical studies.
Collapse
Affiliation(s)
- Evan B Glass
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alyssa A Hoover
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Kennady K Bullock
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Matthew Z Madden
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bradley I Reinfeld
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Whitney Harris
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Dominique Parker
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Marta A Crispens
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew J Wilson
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
11
|
Musick M, Yu X. Manipulation of the tumor immuno-microenvironment via TAM-targeted expression of transcription factors. Immunol Res 2022; 70:432-440. [PMID: 35486115 DOI: 10.1007/s12026-022-09277-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
Abstract
An immunosuppressive tumor microenvironment (TME) leads to cancer growth, metastasis, and therapeutic resistance. Immunomodulatory immunotherapy aims to skew the immunosuppressive TME back to an immune active state. Tumor-associated macrophages (TAMs) are a critical component of the TME that are actively involved in tumor-specific inflammation and immunosuppression. TAMs exhibit a diverse range of phenotypes and functions, from pro-tumor to anti-tumor. The plasticity of TAMs makes them a promising target for immunotherapy, and TAM-targeted therapies via different strategies have shown great potential. This review discusses current TAM-specific delivery targets and genes of interest for TAM-reprogramming. As phagocytic cells, TAMs have several receptors that have been used to increase TAM-targeted in vivo delivery. Furthermore, a promising approach for reprogramming TAMs is to activate or suppress specific transcription factors in the signal transducers and activators of transcription (STAT) and interferon regulatory factor (IRF) families. Altering TAM transcription factor expression results in a potent shift in cytokine expression and overall TAM function potentially tipping the balance from an immunosuppressive to an immune active TME.
Collapse
Affiliation(s)
- Maggie Musick
- Department of Biological Sciences, Clemson University, 132 Long Hall, SC, 29631, Clemson, USA.
| | - Xianzhong Yu
- Department of Biological Sciences, Clemson University, 132 Long Hall, SC, 29631, Clemson, USA
| |
Collapse
|
12
|
Emerging Nanotherapeutic Approaches to Overcome Drug Resistance in Cancers with Update on Clinical Trials. Pharmaceutics 2022; 14:pharmaceutics14040866. [PMID: 35456698 PMCID: PMC9028322 DOI: 10.3390/pharmaceutics14040866] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
A key issue with modern cancer treatments is the emergence of resistance to conventional chemotherapy and molecularly targeted medicines. Cancer nanotherapeutics were created in order to overcome the inherent limitations of traditional chemotherapeutics. Over the last few decades, cancer nanotherapeutics provided unparalleled opportunities to understand and overcome drug resistance through clinical assessment of rationally designed nanoparticulate delivery systems. In this context, various design strategies such as passive targeting, active targeting, nano-drug, and multimodal nano-drug combination therapy provided effective cancer treatment. Even though cancer nanotherapy has made great technological progress, tumor biology complexity and heterogeneity and a lack of comprehensive knowledge of nano-bio interactions remain important roadblocks to future clinical translation and commercialization. The current developments and advancements in cancer nanotherapeutics employing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are discussed in this article. There is also a review of various nanotherapeutics-based approaches to cancer therapy, including targeting strategies for the tumor microenvironment and its components, advanced delivery systems for specific targeting of cancer stem cells (CSC), as well as exosomes for delivery strategies, and an update on clinical trials. Finally, challenges and the future perspective of the cancer nanotherapeutics to reverse cancer drug resistance are discussed.
Collapse
|
13
|
|
14
|
Kiani MH, Ali S, Qadry A, Arshad R, Aslam A, Shahnaz G. Polyethylene imine conjugated supramolecular stereocomplexed nanomicelles for intracellular delivery of rifampicin against Mycobacterium bovis. Colloids Surf B Biointerfaces 2021; 206:111976. [PMID: 34280682 DOI: 10.1016/j.colsurfb.2021.111976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/28/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022]
Abstract
The main objective of this study was to investigate polyethylene imine (PEI) based stereocomplexed nanomiceles for intracellular delivery of rifampicin against Mycobacterium bovis (M. bovis) and their in vitro-in vivo evaluation. The formation of Rifampicin (Rif) loaded isotactic (PEI-g-PLLA and PEI-g-PDLA) and stereocomplexed nanomicelles (StM) of PEI conjugated poly l- and poly d-lactic acid via self-assembly was thoroughly explored. Synthesis of polymer graft was confirmed via FTIR and NMR. A 2-fold reduction in CMC of StM was observed along with decreased particle size in comparison to isotactic nanomicelles. In vitro, StM exhibited a higher encapsulation efficiency and 84 % of drug release in 48 h. at pH 5 with minimal initial burst release in comparison to isotactic nanomicelles. Minimum inhibitory concentration (MIC) of StM was found to be four folds lower in contrast to isotactic nanomicelles. Ex vivo studies exhibited a better uptake of StM and minimum cytotoxicity in murine alveolar macrophages. Following oral administration in mice, drug loaded StM exhibited highest distribution in macrophage rich organs, longer half-life, AUC, AUMC and MRT in comparison to isotactic nanomicelles indicating maximum bioavailability and efficacy of StM. In vivo antimycobacterial activity also demonstrated a higher reduction (2.38fold) in M. bovis CFU at reduced dosing frequency by drug loaded StM in comparison to control group. Thus, StM can be regarded as a simple, stable, efficient, and biocompatible carrier system for delivery of rifampicin to intracellular M. bovis with added advantage of reduced dosing frequency and improved patient compliance.
Collapse
Affiliation(s)
- Maria Hassan Kiani
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sajjad Ali
- Veterinary Research Institute (VRI), Lahore, Pakistan.
| | - Ayesha Qadry
- Veterinary Research Institute (VRI), Lahore, Pakistan.
| | - Rabia Arshad
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Asma Aslam
- Department of Food Science and Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan.
| | - Gul Shahnaz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
15
|
Colonic macrophage-targeted curcumin nanoparticles alleviate DSS-induced colitis in mice through the NF-kappa B pathway. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Andrade RGD, Reis B, Costas B, Lima SAC, Reis S. Modulation of Macrophages M1/M2 Polarization Using Carbohydrate-Functionalized Polymeric Nanoparticles. Polymers (Basel) 2020; 13:polym13010088. [PMID: 33379389 PMCID: PMC7796279 DOI: 10.3390/polym13010088] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Exploiting surface endocytosis receptors using carbohydrate-conjugated nanocarriers brings outstanding approaches to an efficient delivery towards a specific target. Macrophages are cells of innate immunity found throughout the body. Plasticity of macrophages is evidenced by alterations in phenotypic polarization in response to stimuli, and is associated with changes in effector molecules, receptor expression, and cytokine profile. M1-polarized macrophages are involved in pro-inflammatory responses while M2 macrophages are capable of anti-inflammatory response and tissue repair. Modulation of macrophages’ activation state is an effective approach for several disease therapies, mediated by carbohydrate-coated nanocarriers. In this review, polymeric nanocarriers targeting macrophages are described in terms of production methods and conjugation strategies, highlighting the role of mannose receptor in the polarization of macrophages, and targeting approaches for infectious diseases, cancer immunotherapy, and prevention. Translation of this nanomedicine approach still requires further elucidation of the interaction mechanism between nanocarriers and macrophages towards clinical applications.
Collapse
Affiliation(s)
- Raquel G. D. Andrade
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - Bruno Reis
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; (B.R.); (B.C.); (S.R.)
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS-UP), Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal
| | - Benjamin Costas
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; (B.R.); (B.C.); (S.R.)
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS-UP), Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal
| | - Sofia A. Costa Lima
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
- Correspondence:
| | - Salette Reis
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; (B.R.); (B.C.); (S.R.)
| |
Collapse
|
17
|
Zhao YD, Muhetaerjiang M, An HW, Fang X, Zhao Y, Wang H. Nanomedicine enables spatiotemporally regulating macrophage-based cancer immunotherapy. Biomaterials 2020; 268:120552. [PMID: 33307365 DOI: 10.1016/j.biomaterials.2020.120552] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy, leveraging the host's coordinated immune system to fight against tumor has been clinically validated. However, the modest response owing to the multiple ways of tumor immune evasion is one of the challenges in cancer immunotherapy. Tumor associated macrophages (TAMs), as a major component of the leukocytes infiltrating in all tumors, play crucial roles in driving cancer initiation, progress and metastasis via multiple mechanisms such as mediating chronic inflammation, promoting angiogenesis, taming protective immune responses, and supporting migration and intravasation. TAMs targeted therapeutics have achieved remarkable successes in clinical trials mostly through the use of small-molecule agents and antibodies. However, efforts for further application have met with challenges of limited efficacy and safety. Nanomaterials can provide versatile approaches to realize the superior spatiotemporal control over immunomodulation to amplify immune responses, ultimately enhancing the therapeutic benefits and reducing toxicity. Here, the potential drugs used in TAM-centered cancer treatment in clinic are summarized and the recent advances of TAMs targeted nanomedicines in this filed are highlighted. More importantly, we focus on how nanomedicine can exert their advantages in spatial and temporal control of immunomodulation.
Collapse
Affiliation(s)
- Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China; School of Pharmacy, Shanxi Medical University, Shanxi, 030009, PR China
| | - Mamuti Muhetaerjiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Xiaohong Fang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
18
|
Mittelheisser V, Banerjee M, Pivot X, Charbonnière LJ, Goetz J, Detappe A. Leveraging Immunotherapy with Nanomedicine. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vincent Mittelheisser
- Centre Paul Strauss Strasbourg 67000 France
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Mainak Banerjee
- Centre Paul Strauss Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
| | - Loïc J. Charbonnière
- Université de Strasbourg Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Jacky Goetz
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Alexandre Detappe
- Centre Paul Strauss Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| |
Collapse
|
19
|
Rushworth JL, Montgomery KS, Cao B, Brown R, Dibb NJ, Nilsson SK, Chiefari J, Fuchter MJ. Glycosylated Nanoparticles Derived from RAFT Polymerization for Effective Drug Delivery to Macrophages. ACS APPLIED BIO MATERIALS 2020; 3:5775-5786. [PMID: 35021808 DOI: 10.1021/acsabm.0c00529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The functional group tolerance and simplicity of reversible addition fragmentation chain transfer (RAFT) polymerization enable its use in the preparation of a wide range of functional polymer architectures for a variety of applications, including drug delivery. Given the role of tumor-associated macrophages (TAMs) in cancer and their dependence on the tyrosine kinase receptor FMS (CSF-1R), the key aim of this work was to achieve effective delivery of an FMS inhibitor to cells using a polymer delivery system. Such a system has the potential to exploit biological features specific to macrophages and therefore provide enhanced selectivity. Building on our prior work, we have prepared RAFT polymers based on a poly(butyl methacrylate-co-methacrylic acid) diblock, which were extended with a hydrophilic block, a cross-linker, and a mannose-based monomer scaffold, exploiting the abundance of macrophage mannose receptors (MMRs, CD206) on the surface of macrophages. We demonstrate that the prepared polymers can be assembled into nanoparticles and are successfully internalized into macrophages, in part, via the MMR (CD206). Finally, we showcase the developed nanoparticles in the delivery of an FMS inhibitor to cells, resulting in inhibition of the FMS receptor. As such, this study lays the groundwork for further drug-delivery studies aimed at specifically targeting TAMs with molecularly targeted therapeutics.
Collapse
Affiliation(s)
- James L Rushworth
- Molecular Sciences Research Hub, Department of Chemistry, Imperial College London, White City Campus, London W12 0BZ, U.K.,CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia
| | - Katherine S Montgomery
- Molecular Sciences Research Hub, Department of Chemistry, Imperial College London, White City Campus, London W12 0BZ, U.K.,CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia
| | - Benjamin Cao
- CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Robert Brown
- Department of Surgery & Cancer, Imperial College London, London SW7 2AZ, U.K
| | - Nick J Dibb
- Department of Surgery & Cancer, Imperial College London, London SW7 2AZ, U.K
| | - Susan K Nilsson
- CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - John Chiefari
- CSIRO Manufacturing, Bag 10, Clayton South MDC, Victoria 3169, Australia
| | - Matthew J Fuchter
- Molecular Sciences Research Hub, Department of Chemistry, Imperial College London, White City Campus, London W12 0BZ, U.K
| |
Collapse
|
20
|
Shobaki N, Sato Y, Suzuki Y, Okabe N, Harashima H. Manipulating the function of tumor-associated macrophages by siRNA-loaded lipid nanoparticles for cancer immunotherapy. J Control Release 2020; 325:235-248. [DOI: 10.1016/j.jconrel.2020.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 01/04/2023]
|
21
|
Shoaib Sarwar H, Varikuti S, Farhan Sohail M, Sarwar M, Akhtar S, Satoskar AR, Shahnaz G. Oral delivery and enhanced efficacy of antimonal drug through macrophage-guided multifunctional nanocargoes against visceral Leishmaniasis. Eur J Pharm Biopharm 2020; 152:307-317. [PMID: 32485227 DOI: 10.1016/j.ejpb.2020.05.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/25/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
The present study aimed on the site specific delivery and enhanced in-vivo efficacy of antimonial drugs against the visceral leishmaniasis via macrophage targeted mannose anchored thiomer based nanoparticles. Mannose anchored thiolated nanoformulation [M-(CS-g-PEI)-TGA] was developed and evaluated in terms particle size, zeta-potential and entrapment efficacy. The TEM and EDX analysis was carried out to evaluate the morphology and successful entrapment of antimonial drug. Mucodhesion, permeation enhancement, oral pharmacokinetics, and in-vivo anti-leishmanial activity were carried out. The M-(CS-g-PEI)-TGA were found to be spherical having particle size of 287 ± 20 nm. Ex-vivo permeation indicated a 7.39-fold enhanced permeation of Meglumine Antimoniate with M-(CS-g-PEI)-TGA across Caco-2 cells compared to the Glucantime. Evaluation of in-vitro reduction in the parasitic burden via flow cytometric analysis indicated a 5.7-fold lower IC50 for M-(CS-g-PEI)-TGA compared to Glucantime. A 6.1-fold improvement in the oral bioavailability and 5.2-fold reduced parasitic burden in the L. donovani infected BALB/c mice model was observed with M-(CS-g-PEI)-TGA compared to Glucantime. The results encouraged the concept of M-(CS-g-PEI)-TGA nanoformulations as a promising strategy for oral therapy against visceral leishmaniasis.
Collapse
Affiliation(s)
- Hafiz Shoaib Sarwar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Lahore Campus, Lahore, Pakistan; Department of Pathology, Ohio State University Medical Center, Columbus, OH 43201, United States
| | - Sanjay Varikuti
- Department of Pathology, Ohio State University Medical Center, Columbus, OH 43201, United States
| | - Muhammad Farhan Sohail
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan; Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Muhammad Sarwar
- Department of Statistics, Government Postgraduate College, Sahiwal, Pakistan
| | - Sohail Akhtar
- Department of Entomology, University College of Agriculture & Environmental Sciences, The Islamia University, Bahawalpur, Pakistan
| | - Abhay R Satoskar
- Department of Pathology, Ohio State University Medical Center, Columbus, OH 43201, United States.
| | - Gul Shahnaz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
22
|
Gaspar N, Zambito G, Löwik CMWG, Mezzanotte L. Active Nano-targeting of Macrophages. Curr Pharm Des 2020; 25:1951-1961. [PMID: 31291874 DOI: 10.2174/1381612825666190710114108] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Macrophages play a role in almost every disease such as cancer, infections, injuries, metabolic and inflammatory diseases and are becoming an attractive therapeutic target. However, understanding macrophage diversity, tissue distribution and plasticity will help in defining precise targeting strategies and effective therapies. Active targeting of macrophages using nanoparticles for therapeutic purposes is still at its infancy but holds promises since macrophages have shown high specific uptake of nanoparticles. Here, we highlight recent progress in active nanotechnology-based systems gaining pivotal roles to target diverse macrophage subsets in diseased tissues.
Collapse
Affiliation(s)
- Natasa Gaspar
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Percuros B.V., Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands
| | - Giorgia Zambito
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Medres-Medical Research gmbh, Cologne, Germany
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Oncology, Lausanne University Hospital (CHUV), UNIL, Switzerland
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
23
|
Effect of particle size on the cellular uptake and anti-inflammatory activity of oral nanotherapeutics. Colloids Surf B Biointerfaces 2020; 187:110880. [PMID: 32098717 DOI: 10.1016/j.colsurfb.2020.110880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/18/2022]
Abstract
To elucidate the impacts of particle size on the cell internalization and anti-inflammatory activity of oral nanotherapeutics, curcumin (CUR)-loaded polymeric nanoparticles (NPs) with different particle sizes were fabricated. The obtained NPs with particle sizes (185-884 nm) and negative zeta potentials (approximately -25 mV) had desirable CUR loading amounts (5.1-6.1 %) and high CUR encapsulation efficiency (73.2-89.6 %). In vitro cellular uptake assays revealed that the cell internalization efficiencies of NPs were increased with the increase of their particle sizes, and NPs (900) showed the highest phagocytosis efficiency in macrophages among all the tested NPs. Importantly, NPs (900) exhibited significantly stronger capability to downregulate the production of the main pro-inflammatory cytokines from macrophages when they were compared with NPs (200) and NPs (500). Further animal studies suggested that oral administration of hydrogel (chitosan and alginate)-embedding NPs (900) could efficiently accumulate in colitis tissue in a manner that was comparable to that of NPs (200) and NPs (500) and could achieve the best treatment efficacy against ulcerative colitis (UC). Collectively, these findings can serve as a guideline for the rational design of nanotherapeutics with desirable accumulation profiles in colitis tissue, maximized cellular uptake efficiency in macrophages, and good therapeutic outcomes against UC.
Collapse
|
24
|
Zins K, Abraham D. Cancer Immunotherapy: Targeting Tumor-Associated Macrophages by Gene Silencing. Methods Mol Biol 2020; 2115:289-325. [PMID: 32006408 DOI: 10.1007/978-1-0716-0290-4_17] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages (TAMs) are representing a major leukocyte population in solid tumors. Macrophages are very heterogeneous and plastic cells and can acquire distinct functional phenotypes ranging from antitumorigenic to immunosuppressive tumor-promoting M2-like TAMs, depending on the local tissue microenvironment (TME). TAMs express cytokines, chemokines, growth factors, and extracellular matrix (ECM) modifying factors, and the cross talk with the TME regulates pathways involved in the recruitment, polarization, and metabolism of TAMs during tumor progression. Due to their crucial role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer with therapeutic agents that promote phagocytosis or suppress survival, proliferation, trafficking, or polarization of TAMs may prove to be beneficial in cancer therapy. In this chapter, we will discuss TAM biology and current strategies for the targeting of TAMs using small interfering RNA (siRNA)-based drugs. In the past few years, advances in the field of nanomedicine pave the way for the development of siRNA-based drugs as an additional class of personalized cancer immuno-nanomedicines. Fundamental challenges associated with this group of therapeutics include the development process, delivery system, and clinical translation for siRNA-based drugs.
Collapse
Affiliation(s)
- Karin Zins
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
He W, Kapate N, Shields CW, Mitragotri S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv Drug Deliv Rev 2019; 165-166:15-40. [PMID: 31816357 DOI: 10.1016/j.addr.2019.12.001] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play a key role in defending against foreign pathogens, healing wounds, and regulating tissue homeostasis. Driving this versatility is their phenotypic plasticity, which enables macrophages to respond to subtle cues in tightly coordinated ways. However, when this coordination is disrupted, macrophages can aid the progression of numerous diseases, including cancer, cardiovascular disease, and autoimmune disease. The central link between these disorders is aberrant macrophage polarization, which misguides their functional programs, secretory products, and regulation of the surrounding tissue microenvironment. As a result of their important and deterministic roles in both health and disease, macrophages have gained considerable attention as targets for drug delivery. Here, we discuss the role of macrophages in the initiation and progression of various inflammatory diseases, summarize the leading drugs used to regulate macrophages, and review drug delivery systems designed to target macrophages. We emphasize strategies that are approved for clinical use or are poised for clinical investigation. Finally, we provide a prospectus of the future of macrophage-targeted drug delivery systems.
Collapse
Affiliation(s)
- Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Ke X, Howard GP, Tang H, Cheng B, Saung MT, Santos JL, Mao HQ. Physical and chemical profiles of nanoparticles for lymphatic targeting. Adv Drug Deliv Rev 2019; 151-152:72-93. [PMID: 31626825 DOI: 10.1016/j.addr.2019.09.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/03/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Nanoparticles (NPs) have been gaining prominence as delivery vehicles for modulating immune responses to improve treatments against cancer and autoimmune diseases, enhancing tissue regeneration capacity, and potentiating vaccination efficacy. Various engineering approaches have been extensively explored to control the NP physical and chemical properties including particle size, shape, surface charge, hydrophobicity, rigidity and surface targeting ligands to modulate immune responses. This review examines a specific set of physical and chemical characteristics of NPs that enable efficient delivery targeted to secondary lymphoid tissues, specifically the lymph nodes and immune cells. A critical analysis of the structure-property-function relationship will facilitate further efforts to engineer new NPs with unique functionalities, identify novel utilities, and improve the clinical translation of NP formulations for immunotherapy.
Collapse
|
27
|
Glass EB, Masjedi S, Dudzinski SO, Wilson AJ, Duvall CL, Yull FE, Giorgio TD. Optimizing Mannose "Click" Conjugation to Polymeric Nanoparticles for Targeted siRNA Delivery to Human and Murine Macrophages. ACS OMEGA 2019; 4:16756-16767. [PMID: 31646220 PMCID: PMC6796989 DOI: 10.1021/acsomega.9b01465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/19/2019] [Indexed: 06/09/2023]
Abstract
"Smart", dual pH-responsive, and endosomolytic polymeric nanoparticles have demonstrated great potential for localized drug delivery, especially for siRNA delivery to the cytoplasm of cells. However, targeted delivery to a specific cell phenotype requires an additional level of functionality. Copper-catalyzed azide-alkyne cycloaddition (CuAAC) is a highly selective bioconjugation reaction that can be performed in conjunction with other polymerization techniques without adversely affecting reaction kinetics, but there exists some concern for residual copper causing cytotoxicity. To alleviate these concerns, we evaluated conjugation efficiency, residual copper content, and cell viability in relation to copper catalyst concentration. Our results demonstrated an optimal range for minimizing cytotoxicity while maintaining high levels of conjugation efficiency, and these conditions produced polymers with increased targeting to M2-polarized macrophages, as well as successful delivery of therapeutic siRNA that reprogrammed the macrophages to a proinflammatory phenotype.
Collapse
Affiliation(s)
- Evan B Glass
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, Tennessee 37232, United States
| | - Shirin Masjedi
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, Tennessee 37232, United States
| | - Stephanie O Dudzinski
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, Tennessee 37232, United States
| | - Andrew J Wilson
- Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, Tennessee 37232, United States
| | - Fiona E Yull
- Department of Pharmacology and Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, Tennessee 37232, United States
- Department of Pharmacology and Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
28
|
Corradetti B, Pisano S, Conlan RS, Ferrari M. Nanotechnology and Immunotherapy in Ovarian Cancer: Tracing New Landscapes. J Pharmacol Exp Ther 2019; 370:636-646. [PMID: 30737357 PMCID: PMC6806629 DOI: 10.1124/jpet.118.254979] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer (OC) is the seventh most common cancer in women worldwide. Standard therapeutic treatments involve debulking surgery combined with platinum-based chemotherapies. Of the patients with advanced-stage cancer who initially respond to current treatments, 50%-75% relapse. Immunotherapy-based approaches aimed at boosting antitumor immunity have recently emerged as promising tools to challenge tumor progression. Treatments with inhibitors of immune checkpoint molecules have shown impressive results in other types of tumors. However, only 15% of checkpoint inhibitors evaluated have proven successful in OC due to the immunosuppressive environment of the tumor and the transport barriers. This limits the efficacy of the existing immunotherapies. Nanotechnology-based delivery systems hold the potential to overcome such limitations. Various nanoformulations including polymeric, liposomes, and lipid-polymer hybrid nanoparticles have already been proposed to improve the biodistribution and targeting capabilities of drugs against tumor-associated immune cells, including dendritic cells and macrophages. In this review, we examine the impact of immunotherapeutic approaches that are currently under consideration for the treatment of OC. In this review, we also provide a comprehensive analysis of the existing nanoparticle-based synthetic strategies and their limitations and advantages over standard treatments. Furthermore, we discuss how the strength of the combination of nanotechnology with immunotherapy may help to overcome the current therapeutic limitations associated with their individual application and unravel a new paradigm in the treatment of this malignancy.
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Simone Pisano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Robert Steven Conlan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas (B.C., S.P., R.S.C., M.F.); Swansea University Medical School, Singleton Park, Swansea, United Kingdom (B.C., S.P., R.S.C.); and Department of Medicine, Weill Cornell Medical College, New York, New York (M.F.)
| |
Collapse
|
29
|
Zhou X, Liu Y, Huang Y, Ma Y, Lv J, Xiao B. Mucus-penetrating polymeric nanoparticles for oral delivery of curcumin to inflamed colon tissue. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.04.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Zang X, Zhang X, Hu H, Qiao M, Zhao X, Deng Y, Chen D. Targeted Delivery of Zoledronate to Tumor-Associated Macrophages for Cancer Immunotherapy. Mol Pharm 2019; 16:2249-2258. [PMID: 30969779 DOI: 10.1021/acs.molpharmaceut.9b00261] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor-associated macrophages (TAMs) are recruited from circulatory monocytes by tumor-derived factors, which differentiate into macrophages residing in the tumor microenvironment. TAMs play critical roles in promoting angiogenesis, invasion, metastasis and immune escape, and the direct depletion of TAMs is a promising strategy for tumor immunotherapy. In this study, we developed lipid-coated calcium zoledronate nanoparticles (CaZol@pMNPs) containing conjugated mannose, which were sterically shielded with an extracellular pH-sensitive material. The NPs specifically targeted TAMs and induced their apoptosis in vitro and in vivo. In a S180 tumor-bearing mouse model, CaZol@pMNPs effectively depleted TAMs, markedly decreased angiogenesis, reduced immune suppression, and eventually restrained tumor growth without eliciting systemic effects. The collective data indicate the potential of the direct depletion of TAMs using CaZol@pMNPs for cancer immunotherapy.
Collapse
Affiliation(s)
- Xinlong Zang
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Xiaoxu Zhang
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Haiyang Hu
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Mingxi Qiao
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Xiuli Zhao
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Yihui Deng
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| | - Dawei Chen
- School of Pharmacy , Shenyang Pharmaceutical University , Wenhua Road No. 103 , Shenyang 110016 , PR China
| |
Collapse
|
31
|
Cho Y, Milane L, Amiji MM. Genetic and epigenetic strategies for advancing ovarian cancer immunotherapy. Expert Opin Biol Ther 2019; 19:547-560. [DOI: 10.1080/14712598.2019.1602605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Youngwoo Cho
- School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Lara Milane
- Department of Pharmaceutical Science, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Mansoor M. Amiji
- Department of Pharmaceutical Science, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
32
|
Oral administration of colitis tissue-accumulating porous nanoparticles for ulcerative colitis therapy. Int J Pharm 2019; 557:135-144. [DOI: 10.1016/j.ijpharm.2018.12.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/16/2018] [Accepted: 12/21/2018] [Indexed: 01/25/2023]
|
33
|
Song Y, Tang C, Yin C. Combination antitumor immunotherapy with VEGF and PIGF siRNA via systemic delivery of multi-functionalized nanoparticles to tumor-associated macrophages and breast cancer cells. Biomaterials 2018; 185:117-132. [PMID: 30241030 DOI: 10.1016/j.biomaterials.2018.09.017] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/06/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022]
Abstract
Given that vascular endothelial growth factor (VEGF) and placental growth factor (PIGF), over-expressed in breast cancer cells and M2-like tumor-associated macrophages (M2-TAMs) within tumor microenvironment (TME), work synergistically and independently in mediating tumor progression and immunosuppression, combinatorial immune-based approaches targeting them are expected to be a potent therapeutic modality for patients. Here, polyethylene glycol (PEG) and mannose doubly modified trimethyl chitosan (PEG = MT) along with citraconic anhydride grafted poly (allylamine hydrochloride) (PC)-based nanoparticles (NPs) (PEG = MT/PC NPs) with dual pH-responsiveness were developed to deliver VEGF siRNA (siVEGF)/PIGF siRNA (siPIGF) to both M2-TAMs and breast cancer cells for antitumor immunotherapy. With prolonged blood circulation and intelligent pH-sensitivity, PEG = MT/PC NPs were highly accumulated in tumor tissues and then internalized in M2-TAMs and breast cancer cells via mannose-mediated active targeting and passive targeting, respectively. With the charge-reversal of PC, PEG = MT/PC NPs presented effective endosomal/lysosomal escape and intracellular siRNA release, resulting in efficient gene silencing. Due to the synergism between siVEGF and siPIGF in anti-proliferation of tumor cells and reversal of the TME from pro-oncogenic to anti-tumoral, PEG = MT/PC/siVEGF/siPIGF NPs (PEG = MT/PC/siV-P NPs) exerted robust suppression of breast tumor growth and lung metastasis. This combination strategy may provide a promising alternative for breast cancer therapy.
Collapse
Affiliation(s)
- Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
34
|
Vakili‐Ghartavol R, Mombeiny R, Salmaninejad A, Sorkhabadi SMR, Faridi‐Majidi R, Jaafari MR, Mirzaei H. Tumor‐associated macrophages and epithelial–mesenchymal transition in cancer: Nanotechnology comes into view. J Cell Physiol 2018; 233:9223-9236. [DOI: 10.1002/jcp.27027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Roghayyeh Vakili‐Ghartavol
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Reza Mombeiny
- Cellular and Molecular Research Center, Iran University of Medical Sciences Tehran Iran
| | - Arash Salmaninejad
- Drug Applied Research Center, Student Research Committee, Tabriz University of Medical Science Tabriz Iran
- Department of Medical Genetics Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Rezayat Sorkhabadi
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences Tehran Iran
- Department of Toxicology–Pharmacology Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS) Tehran Iran
| | - Reza Faridi‐Majidi
- Department of Medical Nanotechnology School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Nanotechnology School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Hamed Mirzaei
- Department of Biomaterials Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
35
|
Targeting Macrophages as a Potential Therapeutic Intervention: Impact on Inflammatory Diseases and Cancer. Int J Mol Sci 2018; 19:ijms19071953. [PMID: 29973487 PMCID: PMC6073303 DOI: 10.3390/ijms19071953] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/28/2018] [Accepted: 06/30/2018] [Indexed: 12/11/2022] Open
Abstract
Macrophages, cells belonging to the innate immune system, present a high plasticity grade, being able to change their phenotype in response to environmental stimuli. They play central roles during development, homeostatic tissue processes, tissue repair, and immunity. Furthermore, it is recognized that macrophages are involved in chronic inflammation and that they play central roles in inflammatory diseases and cancer. Due to their large involvement in the pathogenesis of several types of human diseases, macrophages are considered to be relevant therapeutic targets. Nanotechnology-based systems have attracted a lot of attention in this field, gaining a pivotal role as useful moieties to target macrophages in diseased tissues. Among the different approaches that can target macrophages, the most radical is represented by their depletion, commonly obtained by means of clodronate-containing liposomal formulations and/or depleting antibodies. These strategies have produced encouraging results in experimental mouse models. In this review, we focus on macrophage targeting, based on the results so far obtained in preclinical models of inflammatory diseases and cancer. Pros and cons of these therapeutic interventions will be highlighted.
Collapse
|
36
|
Harrison EB, Azam SH, Pecot CV. Targeting Accessories to the Crime: Nanoparticle Nucleic Acid Delivery to the Tumor Microenvironment. Front Pharmacol 2018; 9:307. [PMID: 29670528 PMCID: PMC5893903 DOI: 10.3389/fphar.2018.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
Nucleic acid delivery for cancer holds extraordinary promise. Increasing expression of tumor suppressor genes or inhibition of oncogenes in cancer cells has important therapeutic potential. However, several barriers impair progress in cancer gene delivery. These include effective delivery to cancer cells and relevant intracellular compartments. Although viral gene delivery can be effective, it has the disadvantages of being immuno-stimulatory, potentially mutagenic and lacking temporal control. Various nanoparticle (NP) platforms have been developed to overcome nucleic acid delivery hurdles, but several challenges still exist. One such challenge has been the accumulation of NPs in non-cancer cells within the tumor microenvironment (TME) as well as the circulation. While uptake by these cancer-associated cells is considered to be an off-target effect in some contexts, several strategies have now emerged to utilize NP-mediated gene delivery to intentionally alter the TME. For example, the similarity of NPs in shape and size to pathogens promotes uptake by antigen presenting cells, which can be used to increase immune stimulation and promote tumor killing by T-lymphocytes. In the era of immunotherapy, boosting the ability of the immune system to eliminate cancer cells has proven to be an exciting new area in cancer nanotechnology. Given the importance of cancer-associated cells in tumor growth and metastasis, targeting these cells in the TME opens up new therapeutic applications for NPs. This review will cover evidence for non-cancer cell accumulation of NPs in animal models and patients, summarize characteristics that promote NP delivery to different cell types, and describe several therapeutic strategies for gene modification within the TME.
Collapse
Affiliation(s)
- Emily B. Harrison
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Salma H. Azam
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
37
|
Shi X, Shiao SL. The role of macrophage phenotype in regulating the response to radiation therapy. Transl Res 2018; 191:64-80. [PMID: 29175267 PMCID: PMC6018060 DOI: 10.1016/j.trsl.2017.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/23/2017] [Accepted: 11/11/2017] [Indexed: 12/14/2022]
Abstract
Increasing experimental and clinical evidence has revealed a critical role for myeloid cells in the development and progression of cancer. The ability of monocytes and macrophages to regulate inflammation allows them to manipulate the tumor microenvironment to support the growth and development of malignant cells. Recent studies have shown that macrophages can exist in several functional states depending on the microenvironment they encounter in the tissue. These functional phenotypes influence not only the genesis and propagation of tumors, but also the efficacy of cancer therapies, particularly radiation. Early classification of the macrophage phenotypes, or "polarization states," identified 2 major states, M1 and M2, that have cytotoxic and wound repair capacity, respectively. In the context of tumors, classically activated or M1 macrophages driven by interferon-gamma support antitumor immunity while alternatively activated or M2 macrophages generated in part from interleukin-4 exposure hinder antitumor immunity by suppressing cytotoxic responses against a tumor. In this review, we discuss the role that the functional phenotype of a macrophage population plays in tumor development. We will then focus specifically on how macrophages and myeloid cells regulate the tumor response to radiation therapy.
Collapse
Affiliation(s)
- Xiaoshan Shi
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Stephen L Shiao
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA.
| |
Collapse
|
38
|
Wu M, Zhao H, Li M, Yue Y, Xiong S, Xu W. Intranasal Vaccination with Mannosylated Chitosan Formulated DNA Vaccine Enables Robust IgA and Cellular Response Induction in the Lungs of Mice and Improves Protection against Pulmonary Mycobacterial Challenge. Front Cell Infect Microbiol 2017; 7:445. [PMID: 29085809 PMCID: PMC5650621 DOI: 10.3389/fcimb.2017.00445] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 09/29/2017] [Indexed: 12/11/2022] Open
Abstract
Induction of specific humoral and cellular immunity in the lung airways is proposed to be critical for vaccine protection against Mycobacterium tuberculosis (M. tb). To facilitate airway delivery and antigen targeting to the antigen presenting cells in the alveoli, we employed mannosylated chitosan (MCS) to formulate a multi-T-epitope DNA vaccine, pPES, as an intranasal TB vaccine. MCS-DNA nanoparticles appeared spherical with the average particle sizes as 400 nm. HSP65-specific bronchoalveolar lavage fluid SIgA level was significantly elevated by 4 doses of MCS-pPES intranasal immunization as compared to chitosan (CS)-DNA and BCG vaccine. I.n. immunization with MCS-DNA induced a modest peptide-specific Th1(IFN-γ, TNF-α, and IL-2) response in the spleen, while a potent poly-functional CD4+ T response that largely produced TNF-α and IFN-γ, as well as IL-2 in the lung, qualitatively better than that induced by CS-DNA and BCG vaccination. Such response by i.n. immunization with MCS-DNA provided improved protection in the lung against airway Mycobacterial bovis BCG challenge over i.n. CS-DNA and DNA, that is comparable to protection achieved by s.c. BCG vaccination. This enhanced protection was correlated with much greater accessibility of DNA particles to the alveolar macrophages in the lung mediated by man-chitosan. Thus, man-chitosan TB vaccine represents a promising vaccine platform capable of eliciting robust multi-functional T response in the lung mucus and achieving enhanced mucosal immune protection against pulmonary TB.
Collapse
Affiliation(s)
- Manli Wu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Haoxin Zhao
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yan Yue
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Sidong Xiong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
39
|
Andón FT, Digifico E, Maeda A, Erreni M, Mantovani A, Alonso MJ, Allavena P. Targeting tumor associated macrophages: The new challenge for nanomedicine. Semin Immunol 2017; 34:103-113. [PMID: 28941641 DOI: 10.1016/j.smim.2017.09.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022]
Abstract
The engineering of new nanomedicines with ability to target and kill or re-educate Tumor Associated Macrophages (TAMs) stands up as a promising strategy to induce the effective switching of the tumor-promoting immune suppressive microenvironment, characteristic of tumors rich in macrophages, to one that kills tumor cells, is anti-angiogenic and promotes adaptive immune responses. Alternatively, the loading of monocytes/macrophages in blood circulation with nanomedicines, may be used to profit from the high infiltration ability of myeloid cells and to allow the drug release in the bulk of the tumor. In addition, the development of TAM-targeted imaging nanostructures, can be used to study the macrophage content in solid tumors and, hence, for a better diagnosis and prognosis of cancer disease. The major challenges for the effective targeting of TAM with nanomedicines and their application in the clinic have already been identified. These challenges are associated to the undesirable clearance of nanomedicines by, the mononuclear phagocyte system (macrophages) in competing organs (liver, lung or spleen), upon their intravenous injection; and also to the difficult penetration of nanomedicines across solid tumors due to the abnormal vasculature and the excessive extracellular matrix present in stromal tumors. In this review we describe the recent nanotechnology-base strategies that have been developed to target macrophages in tumors.
Collapse
Affiliation(s)
- Fernando Torres Andón
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), University of Santiago de Compostela, 15706 Campus Vida, Santiago de Compostela, Spain.
| | - Elisabeth Digifico
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Akihiro Maeda
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Marco Erreni
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Alberto Mantovani
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy; Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - María José Alonso
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), University of Santiago de Compostela, 15706 Campus Vida, Santiago de Compostela, Spain; Pharmacy & Pharmaceutical Technology Department, School of Pharmacy, University of Santiago de Compostela, 15705 Campus Vida, Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Paola Allavena
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| |
Collapse
|
40
|
Killing Is Not Enough: How Apoptosis Hijacks Tumor-Associated Macrophages to Promote Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 930:205-39. [PMID: 27558823 DOI: 10.1007/978-3-319-39406-0_9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Macrophages are a group of heterogeneous cells of the innate immune system that are crucial to the initiation, progression, and resolution of inflammation. Moreover, they control tissue homeostasis in healthy tissue and command a broad sensory arsenal to detect disturbances in tissue integrity. Macrophages possess a remarkable functional plasticity to respond to irregularities and to initiate programs that allow overcoming them in order to return back to normal. Thus, macrophages kill malignant or transformed cells, rearrange extracellular matrix, take up and recycle cellular as well as molecular debris, initiate cellular growth cascades, and favor directed migration of cells. As an example, apoptotic death of bystander cells is sensed by macrophages, initiating functional responses that support all hallmarks of cancer. In this chapter, we describe how tumor cell apoptosis hijacks tumor-associated macrophages to promote tumor growth. We propose that tumor therapy should not only kill malignant cells but also target the interaction of the host with apoptotic cancer cells, as this might be efficient to limit the protumor action of apoptotic cells and boost the antitumor potential of macrophages. Leaving the apoptotic cell/macrophage interaction untouched might also limit the benefit of conventional tumor cell apoptosis-focused therapy since surviving tumor cells might receive overwhelming support by the wound healing response that apoptotic tumor cells will trigger in local macrophages, thereby enhancing tumor recurrence.
Collapse
|
41
|
Sawa-Wejksza K, Kandefer-Szerszeń M. Tumor-Associated Macrophages as Target for Antitumor Therapy. Arch Immunol Ther Exp (Warsz) 2017; 66:97-111. [PMID: 28660349 PMCID: PMC5851686 DOI: 10.1007/s00005-017-0480-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
It is well known that the microenvironment of solid tumors is rich in inflammatory cells that influence tumor growth and development. Macrophages, called tumor-associated macrophages (TAMs), are the most abundant immune cell population present in tumor tissue. Several studies have demonstrated that the density of TAMs is associated with a poor prognosis and positively correlates with tumor growth. Several studies have proved that TAMs may activate and protect tumor stem cells, stimulate their proliferation as well as promote angiogenesis and metastasis. Furthermore, TAMs-derived cytokines and other proteins, such as CCL-17, CCL-22, TGF-β, IL-10, arginase 1, and galectin-3, make a significant contribution to immunosuppression. Since TAMs influence various aspects of cancer progression, there are many attempts to use them as a target for immunotherapy. The numerous studies have shown that the primary tumor growth and the number of metastatic sites can be significantly decreased by decreasing the population of macrophages in tumor tissue, for example, by blocking recruitment of monocytes or eliminating TAMs already present in the tumor tissue. Moreover, there are attempts at reprogramming TAMs into proinflammatory M1 macrophages or neutralizing the protumoral products of TAMs. Another approach uses TAMs for anticancer drug delivery into the tumor environment. In this review, we would like to summarize the clinical and preclinical trials that were focused on macrophages as a target for anticancer therapies.
Collapse
Affiliation(s)
- Katarzyna Sawa-Wejksza
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Martyna Kandefer-Szerszeń
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
42
|
Exploring the Potential of Nanotherapeutics in Targeting Tumor Microenvironment for Cancer Therapy. Pharmacol Res 2017; 126:109-122. [PMID: 28511988 DOI: 10.1016/j.phrs.2017.05.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/18/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022]
Abstract
Advanced research in the field of cancer biology clearly demonstrated the key role of tumor microenvironment (TME) in cancer development and metastasis particularly in solid tumors. Components of TME, being non-neoplastic in nature provide supportive and permissive conditions for the growth of cancer cells. Hence it is important to modify TME in cancer therapy and this would be achieved by better understanding of TME morphological features and functioning of stromal components. Nanotechnology based drug delivery offers various advantages such as prolonged circulation time, delivery of cargo at desired site, improved bioavailability, reduced toxicity etc. over conventional chemotherapeutics. Abnormal characteristic features of TME play a paradoxical role in nanoparticulate drug delivery. Leaky vasculature, acidic and hypoxic conditions of TME helps in the accumulation of tailored nanoparticles whereas high interstitial pressure and dense stroma restrict the extravasation, homogenous distribution of nanocarriers in TME. This review mainly discusses the potential of nanotherapeutics in targeting TME by briefly discussing stromal components, therapeutic opportunities and barriers offered by TME for nanoparticulate drug delivery. Updated information on TME remodeling strategies for improved drug delivery and specific targeting of individual stromal components are also outlined.
Collapse
|
43
|
Binnemars-Postma K, Storm G, Prakash J. Nanomedicine Strategies to Target Tumor-Associated Macrophages. Int J Mol Sci 2017; 18:E979. [PMID: 28471401 PMCID: PMC5454892 DOI: 10.3390/ijms18050979] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
In recent years, the influence of the tumor microenvironment (TME) on cancer progression has been better understood. Macrophages, one of the most important cell types in the TME, exist in different subtypes, each of which has a different function. While classically activated M1 macrophages are involved in inflammatory and malignant processes, activated M2 macrophages are more involved in the wound-healing processes occurring in tumors. Tumor-associated macrophages (TAM) display M2 macrophage characteristics and support tumor growth and metastasis by matrix remodeling, neo-angiogenesis, and suppressing local immunity. Due to their detrimental role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer may prove to be beneficial in the treatment of cancer. Due to the plastic nature of macrophages, their activities may be altered to inhibit tumor growth. In this review, we will discuss the therapeutic options for the modulation and targeting of TAM. Different therapeutic strategies to deplete, inhibit recruitment of, or re-educate TAM will be discussed. Current strategies for the targeting of TAM using nanomedicine are reviewed. Passive targeting using different nanoparticle systems is described. Since TAM display a number of upregulated surface proteins compared to non-TAM, specific targeting using targeting ligands coupled to nanoparticles is discussed in detail.
Collapse
Affiliation(s)
- Karin Binnemars-Postma
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| | - Gert Storm
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
- Department of Pharmaceutics, Utrecht University, 3584CS Utrecht, The Netherlands.
| | - Jai Prakash
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| |
Collapse
|
44
|
Dosekova E, Filip J, Bertok T, Both P, Kasak P, Tkac J. Nanotechnology in Glycomics: Applications in Diagnostics, Therapy, Imaging, and Separation Processes. Med Res Rev 2017; 37:514-626. [PMID: 27859448 PMCID: PMC5659385 DOI: 10.1002/med.21420] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
This review comprehensively covers the most recent achievements (from 2013) in the successful integration of nanomaterials in the field of glycomics. The first part of the paper addresses the beneficial properties of nanomaterials for the construction of biosensors, bioanalytical devices, and protocols for the detection of various analytes, including viruses and whole cells, together with their key characteristics. The second part of the review focuses on the application of nanomaterials integrated with glycans for various biomedical applications, that is, vaccines against viral and bacterial infections and cancer cells, as therapeutic agents, for in vivo imaging and nuclear magnetic resonance imaging, and for selective drug delivery. The final part of the review describes various ways in which glycan enrichment can be effectively done using nanomaterials, molecularly imprinted polymers with polymer thickness controlled at the nanoscale, with a subsequent analysis of glycans by mass spectrometry. A short section describing an active glycoprofiling by microengines (microrockets) is covered as well.
Collapse
Affiliation(s)
- Erika Dosekova
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Jaroslav Filip
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Tomas Bertok
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Peter Both
- School of Chemistry, Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DNUK
| | - Peter Kasak
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Jan Tkac
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| |
Collapse
|
45
|
Siegler EL, Kim YJ, Wang P. Nanomedicine targeting the tumor microenvironment: Therapeutic strategies to inhibit angiogenesis, remodel matrix, and modulate immune responses. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2016.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Ortega RA, Barham W, Sharman K, Tikhomirov O, Giorgio TD, Yull FE. Manipulating the NF-κB pathway in macrophages using mannosylated, siRNA-delivering nanoparticles can induce immunostimulatory and tumor cytotoxic functions. Int J Nanomedicine 2016; 11:2163-77. [PMID: 27274241 PMCID: PMC4876941 DOI: 10.2147/ijn.s93483] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are critically important in the context of solid tumor progression. Counterintuitively, these host immune cells can often support tumor cells along the path from primary tumor to metastatic colonization and growth. Thus, the ability to transform protumor TAMs into antitumor, immune-reactive macrophages would have significant therapeutic potential. However, in order to achieve these effects, two major hurdles would need to be overcome: development of a methodology to specifically target macrophages and increased knowledge of the optimal targets for cell-signaling modulation. This study addresses both of these obstacles and furthers the development of a therapeutic agent based on this strategy. Using ex vivo macrophages in culture, the efficacy of mannosylated nanoparticles to deliver small interfering RNA specifically to TAMs and modify signaling pathways is characterized. Then, selective small interfering RNA delivery is tested for the ability to inhibit gene targets within the canonical or alternative nuclear factor-kappaB pathways and result in antitumor phenotypes. Results confirm that the mannosylated nanoparticle approach can be used to modulate signaling within macrophages. We also identify appropriate gene targets in critical regulatory pathways. These findings represent an important advance toward the development of a novel cancer therapy that would minimize side effects because of the targeted nature of the intervention and that has rapid translational potential.
Collapse
Affiliation(s)
- Ryan A Ortega
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Whitney Barham
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Kavya Sharman
- Department of Neuroscience, Vanderbilt University, Nashville, TN, USA
| | - Oleg Tikhomirov
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Yull
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
47
|
Cella C, Martello F, Ghisletti S, Lenardi C, Milani P, Argentiere S. Amine-modified poly(vinyl alcohol) as a novel surfactant to modulate size and surface charge of poly(lactide-co-glycolide) nanoparticles. POLYM INT 2016. [DOI: 10.1002/pi.5122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Claudia Cella
- Fondazione Filarete; Viale Ortles 22/4 20139 Milan Italy
- CIMAINA, Dipartimento di Fisica; Università degli Studi di Milano; Via Celoria 16 20133 Milan Italy
- SEMM, European School of Molecular Medicine; Campus IFOM-IEO, Via Adamello 16 20139 Milan Italy
| | | | - Serena Ghisletti
- Department of Experimental Oncology; European Institute of Oncology (IEO); IFOM-IEO Campus, Via Adamello 16 20139 Milan Italy
| | - Cristina Lenardi
- CIMAINA, Dipartimento di Fisica; Università degli Studi di Milano; Via Celoria 16 20133 Milan Italy
| | - Paolo Milani
- CIMAINA, Dipartimento di Fisica; Università degli Studi di Milano; Via Celoria 16 20133 Milan Italy
| | | |
Collapse
|
48
|
Minardi S, Corradetti B, Taraballi F, Sandri M, Martinez JO, Powell ST, Tampieri A, Weiner BK, Tasciotti E. Biomimetic Concealing of PLGA Microspheres in a 3D Scaffold to Prevent Macrophage Uptake. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1479-1488. [PMID: 26797709 DOI: 10.1002/smll.201503484] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Indexed: 06/05/2023]
Abstract
Scaffolds functionalized with delivery systems for the release of growth factors is a robust strategy to enhance tissue regeneration. However, after implantation, macrophages infiltrate the scaffold, eventually initiating the degradation and clearance of the delivery systems. Herein, it is hypothesized that fully embedding the poly(d,l-lactide-co-glycolide acid) microspheres (MS) in a highly structured collagen-based scaffold (concealing) can prevent their detection, preserving the integrity of the payload. Confocal laser microscopy reveals that non-embedded MS are easily internalized; when concealed, J774 and bone marrow-derived macrophages (BMDM) cannot detect them. This is further demonstrated by flow cytometry, as a tenfold decrease is found in the number of MS engulfed by the cells, suggesting that collagen can cloak the MS. This correlates with the amount of nitric oxide and tumor necrosis factor-α produced by J774 and BMDM in response to the concealed MS, comparable to that found for non-functionalized collagen scaffolds. Finally, the release kinetics of a reporter protein is preserved in the presence of macrophages, only when MS are concealed. The data provide detailed strategies for fabricating three dimensional (3D) biomimetic scaffolds able to conceal delivery systems and preserve the therapeutic molecules for release.
Collapse
Affiliation(s)
- Silvia Minardi
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Bruna Corradetti
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131, Ancona, Italy
| | - Francesca Taraballi
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Monica Sandri
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Jonathan O Martinez
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Sebastian T Powell
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Anna Tampieri
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Bradley K Weiner
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Department of Orthopedic Surgery, Houston Methodist Hospital, 6550 Fannin St., Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| |
Collapse
|
49
|
Pei Y, Yeo Y. Drug delivery to macrophages: Challenges and opportunities. J Control Release 2015; 240:202-211. [PMID: 26686082 DOI: 10.1016/j.jconrel.2015.12.014] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/05/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Macrophages are prevalent in the body and have roles in almost every aspect of human biology. They have often been considered a subject to avoid during drug delivery. However, with recent understanding of their diverse functions in diseases, macrophages have gained increasing interest as important therapeutic targets. To develop drug carriers to macrophages, it is important to understand their biological roles and requirements for efficient targeting. This review provides an overview of representative carriers and various approaches to address challenges in drug delivery to macrophages such as biodistribution, cellular uptake, intracellular trafficking, and drug release.
Collapse
Affiliation(s)
- Yihua Pei
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
50
|
Wilson AJ, Saskowski J, Barham W, Khabele D, Yull F. Microenvironmental effects limit efficacy of thymoquinone treatment in a mouse model of ovarian cancer. Mol Cancer 2015; 14:192. [PMID: 26552746 PMCID: PMC4640396 DOI: 10.1186/s12943-015-0463-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/23/2015] [Indexed: 01/01/2023] Open
Abstract
Background Ovarian cancer is the most lethal gynecologic malignancy, with limited treatment options for chemoresistant disease. An important link between inflammation and peritoneal spread of ovarian cancer is NF-κB signaling. Thymoquinone (TQ) exerts multiple anti-tumorigenic cellular effects, including NF-κB inhibition. We aimed to investigate the therapeutic potential of TQ in an established murine syngeneic model of ovarian cancer. Methods ID8-NGL mouse ovarian cancer cells stably expressing an NF-κB reporter transgene were injected intra-peritoneally into C57BL/6 mice, and mice were treated with TQ or vehicle for 10 or 30 days. TQ was combined with the macrophage depleting drug, liposomal clodronate, in selected experiments. Effects on peritoneal tumor burden were measured by volume of ascites, number of peritoneal implants and mesenteric tumor mass. NF-κB reporter activity and markers of proliferation and apoptosis were measured in tumors and in confirmatory in vitro experiments. Protein or mRNA expression of M1 (anti-tumor) and M2 (pro-tumor) macrophage markers, and soluble cytokine profiles, were examined from harvested ascites fluid, peritoneal lavages and/or tumor sections. 2-tailed Mann–Whitney tests were used for measuring differences between groups in in vivo experiments. Results Consistent with its effects in vitro, TQ reduced proliferation and increased apoptosis in ID8-NGL tumors after 10 and 30 day treatment. Prolonged TQ treatment did not significantly alter tumor number or mass compared to vehicle, but rather exerted an overall deleterious effect by stimulating ascites formation. Increased ascites was accompanied by elevated NF-κB activity in tumors and macrophages, increased pro-tumor M2 macrophages and expression of pro-tumorigenic soluble factors such as VEGF in ascites fluid, and increased tumor infiltration of M2 macrophages. In contrast, a 10 day exposure to TQ produced no ascites, and reduced tumor NF-κB activity, M2 macrophages and soluble VEGF levels. Peritoneal macrophage depletion by clodronate significantly reduced tumor burden. However, TQ-stimulated ascites was further enhanced by co-treatment with clodronate, with macrophages present overwhelmingly of the M2 phenotype. Conclusions Our findings show that pro-tumorigenic microenvironmental effects limited the efficacy of TQ in a syngeneic mouse model of ovarian cancer, and provide caution regarding its potential use in clinical trials in ovarian cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0463-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew J Wilson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, B1100 Medical Center North, Nashville, TN, 37232, USA.
| | - Jeanette Saskowski
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Whitney Barham
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fiona Yull
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|