1
|
Padhy A, Das P, Mahadik NS, Panda S, Anas M, Das S, Banerjee R, Sen Gupta S. Design and synthesis of a shikimoyl-functionalized cationic di-block copolypeptide for cancer cell specific gene transfection. J Mater Chem B 2024; 12:8952-8965. [PMID: 39171401 DOI: 10.1039/d4tb01233j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Targeted and efficient gene delivery systems hold tremendous potential for the improvement of cancer therapy by enabling appropriate modification of biological processes. Herein, we report the design and synthesis of a novel cationic di-block copolypeptide, incorporating homoarginine (HAG) and shikimoyl (LSA) functionalities (HDA-b-PHAGm-b-PLSAn), tailored for enhanced gene transfection specifically in cancer cells. The di-block copolypeptide was synthesized via sequential N-carboxyanhydride (NCA) ring-opening polymerization (ROP) techniques and its physicochemical properties were characterized, including molecular weight, dispersity, secondary conformation, size, morphology, and surface charge. In contrast to the cationic poly-L-homoarginine, we observed a significantly reduced cytotoxic effect of this di-block copolypeptide due to the inclusion of the shikimoyl glyco-polypeptide block, which also added selectivity in internalizing particular cells. This di-block copolypeptide was internalized via mannose-receptor-mediated endocytosis, which was investigated by competitive receptor blocking with mannan. We evaluated the transfection efficiency of the copolypeptide in HEK 293T (noncancerous cells), MDA-MB-231 (breast cancer cells), and RAW 264.7 (dendritic cells) and compared it with commonly employed transfection agents (Lipofectamine). Our findings demonstrate that the homoarginine and shikimoyl-functionalized cationic di-block copolypeptide exhibits potent gene transfection capabilities with minimal cytotoxic effects, particularly in cancer cells, while it is ineffective for normal cells, indicative of its potential as a promising platform for cancer cell-specific gene delivery systems. To evaluate this, we delivered an artificially designed miRNA-plasmid against Hsp90 (amiR-Hsp90) which upon successful transfection depleted the Hsp90 (a chaperone protein responsible for tumour growth) level specifically in cancerous cells and enforced apoptosis. This innovative approach offers a new avenue for the development of targeted therapeutics with an improved efficacy and safety profile in cancer treatment.
Collapse
Affiliation(s)
- Abinash Padhy
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Pritam Das
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Namita S Mahadik
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sidharth Panda
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Mahammad Anas
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700 032, India
| | - Sabyasachi Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Rajkumar Banerjee
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sayam Sen Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| |
Collapse
|
2
|
Mahalingam G, Rachamalla HK, Arjunan P, Karuppusamy KV, Periyasami Y, Mohan A, Subramaniyam K, M S, Rajendran V, Moorthy M, Varghese GM, Mohankumar KM, Thangavel S, Srivastava A, Marepally S. SMART-lipid nanoparticles enabled mRNA vaccine elicits cross-reactive humoral responses against the omicron sub-variants. Mol Ther 2024; 32:1284-1297. [PMID: 38414245 PMCID: PMC11081802 DOI: 10.1016/j.ymthe.2024.02.028] [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: 08/10/2023] [Revised: 12/19/2023] [Accepted: 02/23/2024] [Indexed: 02/29/2024] Open
Abstract
The continual emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has necessitated the development of broad cross-reactive vaccines. Recent findings suggest that enhanced antigen presentation could lead to cross-reactive humoral responses against the emerging variants. Toward enhancing the antigen presentation to dendritic cells (DCs), we developed a novel shikimoylated mannose receptor targeting lipid nanoparticle (SMART-LNP) system that could effectively deliver mRNAs into DCs. To improve the translation of mRNA, we developed spike domain-based trimeric S1 (TS1) mRNA with optimized codon sequence, base modification, and engineered 5' and 3' UTRs. In a mouse model, SMART-LNP-TS1 vaccine could elicit robust broad cross-reactive IgGs against Omicron sub-variants, and induced interferon-γ-producing T cells against SARS-CoV-2 virus compared with non-targeted LNP-TS1 vaccine. Further, T cells analysis revealed that SMART-LNP-TS1 vaccine induced long-lived memory T cell subsets, T helper 1 (Th1)-dominant and cytotoxic T cells immune responses against the SARS-CoV-2 virus. Importantly, SMART-LNP-TS1 vaccine produced strong Th1-predominant humoral and cellular immune responses. Overall, SMART-LNPs can be explored for precise antigenic mRNA delivery and robust immune responses. This platform technology can be explored further as a next-generation delivery system for mRNA-based immune therapies.
Collapse
Affiliation(s)
- Gokulnath Mahalingam
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Hari Krishnareddy Rachamalla
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Porkizhi Arjunan
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Karthik V Karuppusamy
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Yogapriya Periyasami
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Aruna Mohan
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Kanimozhi Subramaniyam
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Salma M
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Vigneshwar Rajendran
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Mahesh Moorthy
- Department of Clinical Virology, Christian Medical College and Hospital, Vellore, TN 632002, India
| | - George M Varghese
- Department of Infectious Diseases, Christian Medical College and Hospital, Vellore, TN 632002, India
| | - Kumarasamypet M Mohankumar
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Saravanabhavan Thangavel
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Alok Srivastava
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India
| | - Srujan Marepally
- Centre for Stem Cell Research (CSCR) (a unit of inStem, Bengaluru), CMC Campus, Vellore, TN 632002, India.
| |
Collapse
|
3
|
Barui S, Saha S, Venu Y, Moku GK, Chaudhuri A. In vivo targeting of a tumor-antigen encoded DNA vaccine to dendritic cells in combination with tumor-selective chemotherapy eradicates established mouse melanoma. Biomater Sci 2023; 11:6135-6148. [PMID: 37555308 DOI: 10.1039/d3bm00702b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Despite remarkable progress during the past decade, eradication of established tumors by targeted cancer therapy and cancer immunotherapy remains an uphill task. Herein, we report on a combination approach for eradicating established mouse melanoma. Our approach employs the use of tumor selective chemotherapy in combination with in vivo dendritic cell (DC) targeted DNA vaccination. Liposomes of a newly synthesized lipopeptide containing a previously reported tumor-targeting CGKRK-ligand covalently grafted in its polar head-group region were used for tumor selective delivery of cancer therapeutics. Liposomally co-loaded STAT3siRNA and WP1066 (a commercially available inhibitor of the JAK2/STAT3 pathway) were used as cancer therapeutics. In vivo targeting of a melanoma antigen (MART-1) encoded DNA vaccine (p-CMV-MART1) to dendritic cells was accomplished by complexing it with a previously reported mannose-receptor selective in vivo DC-targeting liposome. Liposomes of the CGKRK-lipopeptide containing encapsulated FITC-labeled siRNA, upon intravenous administration in B16F10 melanoma bearing mice, showed remarkably higher accumulation in tumors 24 h post i.v. treatment, compared to their degree of accumulation in other body tissues including the lungs, liver, kidneys, spleen and heart. Importantly, the findings in tumor growth inhibition studies revealed that only in vivo DC-targeted genetic immunization or only tumor-selective chemotherapy using the presently described systems failed to eradicate the established mouse melanoma. The presently described combination approach is expected to find future applications in combating various malignancies (with well-defined surface antigens).
Collapse
Affiliation(s)
- Sugata Barui
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad-500007, Telangana, India.
| | - Soumen Saha
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad-500007, Telangana, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201002, India
| | - Yakati Venu
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad-500007, Telangana, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201002, India
| | - Gopi Krishna Moku
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad-500007, Telangana, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201002, India
- Department of Physical Sciences, Kakatiya Institute of Technology and Science, Yerragattu Gutta, Warangal 506 015, Telangana, India
| | - Arabinda Chaudhuri
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad-500007, Telangana, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201002, India
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia-74126, West Bengal, India
| |
Collapse
|
4
|
Wang Q, Wang Z, Sun X, Jiang Q, Sun B, He Z, Zhang S, Luo C, Sun J. Lymph node-targeting nanovaccines for cancer immunotherapy. J Control Release 2022; 351:102-122. [PMID: 36115556 DOI: 10.1016/j.jconrel.2022.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/20/2022]
Abstract
Cancer immunotherapies such as tumor vaccines, chimeric antigen receptor T cells and immune checkpoint blockades, have attracted tremendous attention. Among them, tumor vaccines prime immune response by delivering antigens and adjuvants to the antigen presenting cells (APCs), thus enhancing antitumor immunotherapy. Despite tumor vaccines have made considerable achievements in tumor immunotherapy, it remains challenging to efficiently deliver tumor vaccines to activate the dendritic cells (DCs) in lymph nodes (LNs). Rational design of nanovaccines on the basis of biomedical nanotechnology has emerged as one of the most promising strategies for boosting the outcomes of cancer immunotherapy. In recent years, great efforts have been made in exploiting various nanocarrier-based LNs-targeting tumor nanovaccines. In view of the rapid advances in this field, we here aim to summarize the latest progression in LNs-targeting nanovaccines for cancer immunotherapy, with special attention to various nano-vehicles developed for LNs-targeting delivery of tumor vaccines, including lipid-based nanoparticles, polymeric nanocarriers, inorganic nanocarriers and biomimetic nanosystems. Moreover, the recent trends in nanovaccines-based combination cancer immunotherapy are provided. Finally, the rationality, advantages and challenges of LNs-targeting nanovaccines for clinical translation and application are spotlighted.
Collapse
Affiliation(s)
- Qiu Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhe Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinxin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
5
|
Mondal B, Mahadik NS, Banerjee R, Sen Gupta S. Design and Synthesis of Shikimoylated-Polypeptides for Nuclear Specific Internalization. ACS Macro Lett 2022; 11:289-295. [PMID: 35575367 DOI: 10.1021/acsmacrolett.1c00740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Targeted delivery of therapeutics such as small molecule drugs or nucleic acids exclusively to the nucleus of diseased mammalian cells poses a significant challenge. The development of targeting ligands that can specifically enter certain cancer cells via a specific receptor-mediated endocytosis and then traffic exclusively to the nucleus to deliver the cargo inside it can achieve this goal. We have developed an end-functionalized shikimoylated-polypeptide with pendant shikimoyl moieties that can enter mammalian cells via the mannose receptors and are then exclusively trafficked into the nucleus. The presence of the shikimoyl group in the polypeptide, which traffics it exclusively to the nucleus, contrasts with the mannosylated or galactosylated glycopolypeptides that are distributed all over the cytoplasm or the mannose-6-phosphate containing polypeptide that is exclusively trafficked to the lysosome. Using challenge experiments, we demonstrate that these polypeptides can enter both dendritic and cancer cells through mannose-receptors and subsequently enter the cell nucleus via the interaction with a nuclear pore complex (NPC) protein importin-α/β1. To the best of our knowledge, this represents the first example of a synthetic polyvalent glycopolypeptide mimic that performs the dual function of entering mammalian cells through specific receptors and subsequently traffics into the nucleus. The conjugation of these end-functionalized shikimoylated-polypeptides to other biological entities, such as recombinant anticancer drugs, DNA, RNA, and CRISPR-Cas9, may be a suitable alternative for delivery of these biological entities into cells affected by cancer and other genetic diseases.
Collapse
Affiliation(s)
- Basudeb Mondal
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur Campus, Nadia, West Bengal-741246, India
| | - Namita S. Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana-500007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad-201002, Uttar Pradesh, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana-500007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad-201002, Uttar Pradesh, India
| | - Sayam Sen Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur Campus, Nadia, West Bengal-741246, India
| |
Collapse
|
6
|
Mahadik N, Bhattacharya D, Padmanabhan A, Sakhare K, Narayan KP, Banerjee R. Targeting steroid hormone receptors for anti-cancer therapy-A review on small molecules and nanotherapeutic approaches. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1755. [PMID: 34541822 DOI: 10.1002/wnan.1755] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The steroid hormone receptors (SHRs) among nuclear hormone receptors (NHRs) are steroid ligand-dependent transcription factors that play important roles in the regulation of transcription of genes promoted via hormone responsive elements in our genome. Aberrant expression patterns and context-specific regulation of these receptors in cancer, have been routinely reported by multiple research groups. These gave an window of opportunity to target those receptors in the context of developing novel, targeted anticancer therapeutics. Besides the development of a plethora of SHR-targeting synthetic ligands and the availability of their natural, hormonal ligands, development of many SHR-targeted, anticancer nano-delivery systems and theranostics, especially based on small molecules, have been reported. It is intriguing to realize that these cytoplasmic receptors have become a hot target for cancer selective delivery. This is in spite of the fact that these receptors do not fall in the category of conventional, targetable cell surface bound or transmembrane receptors that enjoy over-expression status. Glucocorticoid receptor (GR) is one such exciting SHR that in spite of it being expressed ubiquitously in all cells, we discovered it to behave differently in cancer cells, thus making it a truly druggable target for treating cancer. This review selectively accumulates the knowledge generated in the field of SHR-targeting as a major focus for cancer treatment with various anticancer small molecules and nanotherapeutics on progesterone receptor, mineralocorticoid receptor, and androgen receptor while selectively emphasizing on GR and estrogen receptor. This review also briefly highlights lipid-modification strategy to convert ligands into SHR-targeted cancer nanotherapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Namita Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Akshaya Padmanabhan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
7
|
Ho W, Gao M, Li F, Li Z, Zhang X, Xu X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and mRNA Delivery. Adv Healthc Mater 2021; 10:e2001812. [PMID: 33458958 PMCID: PMC7995055 DOI: 10.1002/adhm.202001812] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/06/2020] [Indexed: 01/07/2023]
Abstract
Nucleic acid vaccines are a method of immunization aiming to elicit immune responses akin to live attenuated vaccines. In this method, DNA or messenger RNA (mRNA) sequences are delivered to the body to generate proteins, which mimic disease antigens to stimulate the immune response. Advantages of nucleic acid vaccines include stimulation of both cell-mediated and humoral immunity, ease of design, rapid adaptability to changing pathogen strains, and customizable multiantigen vaccines. To combat the SARS-CoV-2 pandemic, and many other diseases, nucleic acid vaccines appear to be a promising method. However, aid is needed in delivering the fragile DNA/mRNA payload. Many delivery strategies have been developed to elicit effective immune stimulation, yet no nucleic acid vaccine has been FDA-approved for human use. Nanoparticles (NPs) are one of the top candidates to mediate successful DNA/mRNA vaccine delivery due to their unique properties, including unlimited possibilities for formulations, protective capacity, simultaneous loading, and delivery potential of multiple DNA/mRNA vaccines. This review will summarize the many varieties of novel NP formulations for DNA and mRNA vaccine delivery as well as give the reader a brief synopsis of NP vaccine clinical trials. Finally, the future perspectives and challenges for NP-mediated nucleic acid vaccines will be explored.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Mingzhu Gao
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Fengqiao Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Zhongyu Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringNew Jersey Institute of Technology323 Dr Martin Luther King Jr BlvdNewarkNJ07102USA
| |
Collapse
|
8
|
Yazdani M, Jaafari MR, Verdi J, Alani B, Noureddini M, Badiee A. Ex vivo-generated dendritic cell-based vaccines in melanoma: the role of nanoparticulate delivery systems. Immunotherapy 2020; 12:333-349. [DOI: 10.2217/imt-2019-0173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Melanoma is a poor immunogenic cancer and many treatment strategies have been used to enhance specific or nonspecific immunity against it. Dendritic cell (DC)-based cancer vaccine is the most effective therapies that have been used so far. Meanwhile, the efficacy of DC-based immunotherapy relies on critical factors relating to DCs such as the state of maturation and proper delivery of antigens. In this regard, the use of nanoparticulate delivery systems for effective delivery of antigen to ex vivo-generated DC-based vaccines that also poses adjuvanticity would be an ideal approach. In this review article, we attempt to summarize the role of different types of nanoparticulate antigen delivery systems used in the development of ex vivo-generated DC-based vaccines against melanoma and describe their adjuvanticity in mediation of DC maturation, cytoplasmic presentation of antigens to MHC class I molecules, which led to potent antigen-specific immune responses. As were represented, cationic liposomes were the most used approach, which suggest its potential applicability as delivery systems for further experiments in combination with either adjuvants or monoclonal antibodies.
Collapse
Affiliation(s)
- Mona Yazdani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Mahdi Noureddini
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| |
Collapse
|
9
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
10
|
Nanotechnology in the arena of cancer immunotherapy. Arch Pharm Res 2020; 43:58-79. [DOI: 10.1007/s12272-020-01207-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
|
11
|
Gulla SK, Rao BR, Moku G, Jinka S, Nimmu NV, Khalid S, Patra CR, Chaudhuri A. In vivo targeting of DNA vaccines to dendritic cells using functionalized gold nanoparticles. Biomater Sci 2019; 7:773-788. [PMID: 30601510 DOI: 10.1039/c8bm01272e] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The clinical success of dendritic cell (DC)-based genetic immunization remains critically dependent on the availability of effective and safe nano-carriers for targeting antigen-encoded DNA vaccines to DCs, the most potent antigen-presenting cells in the human body in vivo. Recent studies revealed the efficacies of mannose receptor-mediated in vivo DC-targeted genetic immunization by liposomal DNA vaccine carriers containing both mannose-mimicking shikimoyl and transfection enhancing guanidinyl functionalities. However, to date, the efficacies of this approach have not been examined for metal-based nanoparticle DNA vaccine carriers. Herein, we report for the first time, the design, synthesis, physico-chemical characterization and bioactivities of gold nanoparticles covalently functionalized with a thiol ligand containing both shikimoyl and guanidinyl functionalities (Au-SGSH). We show that Au-SGSH nanoparticles can deliver DNA vaccines to mouse DCs under in vivo conditions. Subcutaneous administration of near infrared (NIR) dye-labeled Au-SGSH showed significant accumulation of the NIR dye in the DCs of the nearby lymph nodes compared to that for the non-targeting NIR-labeled Au-GSH nanoconjugate containing only a covalently tethered guanidinyl group, not the shikimoyl-functionality. Under prophylactic settings, in vivo immunization (s.c.) with the Au-SGSH-pCMV-MART1 nanoplex induced a long-lasting (180 days) immune response against murine melanoma. Notably, mannose receptor-mediated in vivo DC-targeted immunization (s.c.) with the Au-SGSH-MART1 nanoplex significantly inhibited established melanoma growth and increased the overall survivability of melanoma-bearing mice under therapeutic settings. The Au-SGSH nanoparticles reported herein have potential use for in vivo DC-targeted genetic immunization against cancer and infectious diseases.
Collapse
Affiliation(s)
- Suresh Kumar Gulla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Meka RR, Mukherjee S, Patra CR, Chaudhuri A. Shikimoyl-ligand decorated gold nanoparticles for use in ex vivo engineered dendritic cell based DNA vaccination. NANOSCALE 2019; 11:7931-7943. [PMID: 30964937 DOI: 10.1039/c8nr10293g] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Since mannose receptors (MRs) are expressed on the surfaces of dendritic cells (DCs), the most professional antigen presenting cells in our body, DNA vaccine carriers containing either covalently grafted mannosyl- or mannose-mimicking shikimoyl-ligands are being increasingly used in ex vivo DC-transfection based DNA vaccination. To this end, we have recently demonstrated that ex vivo immunization of mice with liposomes of shikimoylated cationic amphiphiles containing a 6-amino hexanoic acid spacer group in the head-group region in complexation with melanoma antigen (MART1) encoded DNA vaccine (pCMV-MART1) induces long lasting anti-melanoma immune responses (C. Voshavar, et al., J. Med. Chem., 2017, 60, 1605-1610). This finding prompted us to examine, in the present investigation, the efficacies of gold nanoparticles conjugated to the mannose-mimicking shikimoyl ligand (SL) via a 6-amino hexane thiol spacer (AuNPs-SL) for use in ex vivo DC-transfection based genetic immunization. Herein, we report on the design, synthesis, physico-chemical characterization and bioactivities of AuNPs-SL. Dynamic light scattering and transmission electron microscopy studies revealed the hydrodynamic diameters of theAuNPs-SL nanoconjugates to be within the range of 23-44 nm and their surface potentials within the range of 9-28 mV. MTT-assay showed the non-cytotoxic nature of AuNPs-SL and the findings in the electrophoretic gel retardation assays revealed strong DNA binding properties of the AuNPs-SL. Importantly, subcutaneous immunization of C57BL/6J mice with DCs ex vivo transfected with an electrostatic complex of AuNPs-SL & melanoma antigen (MART1) encoded DNA vaccine (p-CMV-MART1) induced a long lasting (100 days) anti-tumor immune response in immunized mice upon subsequent challenge with a lethal dose of melanoma. Notably, mice immunized with either autologous mbmDCs ex vivo pre-transfected with nanoplexes of shikimoylated AuNPs-SL & an irrelevant pCMV-SPORT-β-gal plasmid (without having encoded melanoma antigen) or untransfected DCs showed no lasting protection against subsequent tumor challenge. The presently described shikimoyl-decorated gold nanoparticles (AuNPs-SL) are expected to find future use in ex vivo DC-transfection based genetic immunization against cancer and other infectious diseases.
Collapse
Affiliation(s)
- Rakeshchandra R Meka
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| | | | | | | |
Collapse
|
13
|
Recent advances in applying nanotechnologies for cancer immunotherapy. J Control Release 2018; 288:239-263. [PMID: 30223043 DOI: 10.1016/j.jconrel.2018.09.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aimed at boosting cancer-specific immunoresponses to eradicate tumor cells has evolved as a new treatment modality. Nanoparticles incorporating antigens and immunomodulatory agents can activate immune cells and modulate the tumor microenvironment to enhance anti-tumor immunity. The nanotechnology approach has been demonstrated to be superior to standard formulations in in-vivo settings. In this article, we focus on recent advances made within the last 5 years in nanoparticle-based cancer immunotherapy, including peptide- and nucleic acid-based nanovaccines, nanomedicines containing an immunoadjuvant to activate anti-tumor immunity, nanoparticle delivery of immune checkpoint inhibitors and the combination of the above approaches. Encouraging results and new emerging nanotechnologies in drug delivery promise the continuous growth of this field and ultimately clinical translation of enhanced immunotherapy of cancer.
Collapse
|
14
|
Rangasami VK, Lohchania B, Voshavar C, Rachamalla HR, Banerjee R, Dhayani A, Thangavel S, Vemula PK, Marepally S. Exploring membrane permeability of Tomatidine to enhance lipid mediated nucleic acid transfections. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:327-334. [PMID: 29902420 DOI: 10.1016/j.bbamem.2018.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 05/05/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023]
Abstract
Intracellular delivery of nucleic acids is one of the critical steps in the transfections. Prior findings demonstrated various strategies including membrane fusion, endosomal escape for the efficient cytoplasmic delivery. In our continuing efforts to improve the nucleic acids transfections, we harnessed cell permeable properties of Tomatidine (T), a steroidal alkaloid abundantly found in green tomatoes for maximizing intracellular delivery of lipoplexes. We doped Tomatidine into liposomes of cationic lipid with amide linker (A) from our lipid library. Six liposomal formulations (AT) of Lipid A (1 mM) with varying concentrations of Tomatidine (0-1 mM) were prepared and evaluated for their transfection efficacies. Owing to its signature characteristic of cell membrane permeability, Tomatidine modulated endocytosis process, enhanced the intracellular delivery of the lipoplexes, and in turn increased the transfection efficacy of cationic liposomes. Our findings provide 'proof of concept' for enhancing transfections in gene delivery applications with Tomatidine in cationic liposomal formulations. These findings can be further applied in lipid mediated gene therapy and drug delivery applications.
Collapse
Affiliation(s)
- Vignesh K Rangasami
- Centre for Stem Cell Research, Christian Medical College Campus, Bagayam, Vellore 632002, India
| | - Brijesh Lohchania
- Centre for Stem Cell Research, Christian Medical College Campus, Bagayam, Vellore 632002, India
| | | | | | - Rajkumar Banerjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Ashish Dhayani
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), GKVK-post, Bellary Road, Bengaluru 560065, India; The School of Chemical and Biotechnology, SASTRA University, Thirumalaisamudram, Thanjavur-613401,Tamil Nadu, India
| | | | - Praveen K Vemula
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), GKVK-post, Bellary Road, Bengaluru 560065, India.
| | - Srujan Marepally
- Centre for Stem Cell Research, Christian Medical College Campus, Bagayam, Vellore 632002, India; Institute for Stem Cell Biology and Regenerative Medicine (inStem), GKVK-post, Bellary Road, Bengaluru 560065, India.
| |
Collapse
|
15
|
Muripiti V, Lohchania B, Marepally SK, Patri SV. Hepatocellular targeted α-tocopherol based pH sensitive galactosylated lipids: design, synthesis and transfection studies. MEDCHEMCOMM 2018; 9:264-274. [PMID: 30108920 PMCID: PMC6083795 DOI: 10.1039/c7md00503b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 12/05/2017] [Indexed: 11/21/2022]
Abstract
Receptor mediated gene delivery to the liver offers advantages in treating genetic disorders such as hemophilia and hereditary tyrosinemia type I (HTI). Prior findings demonstrated that tethering the d-galactose head group to cationic lipids directs genes to the liver via asialoglycoprotein receptors (ASGPRs). In our continued efforts to develop safer and efficient lipofectins, we demonstrated that cationic lipids bearing α-tocopherol, an antioxidant, as a hydrophobic domain could deliver genes efficiently with high safety profiles in multiple cell lines. Towards developing ASGPR targeted pH sensitive cationic lipids, we have designed a galactosylated cationic lipid (Toc-Gal) with α-tocopherol as the hydrophobic core covalently connected with a pH responsive triazole moiety and a non-targeting control lipid (Toc-OH) without the galactose head group. In this study, we present the design and synthesis of a pH sensitive galactosylated cationic lipid (Toc-Gal), its comparative transfection biology, cellular uptake studies, serum stability and cytotoxicity profiles in both ASGPR positive and negative liver cells, i.e. HepG2 and SK-Hep-1, respectively.
Collapse
Affiliation(s)
- Venkanna Muripiti
- National Institute of Technology , Warangal-506004 , Telangana , India .
| | | | | | - Srilakshmi V Patri
- National Institute of Technology , Warangal-506004 , Telangana , India .
| |
Collapse
|