Cancer Nanomedicines in an Evolving Oncology Landscape

Trends in Pharmacological Sciences - Tập 41 - Trang 730-742 - 2020
Peng Guo1,2, Jing Huang1,2, Marsha A. Moses1,2
1Vascular Biology Program, Boston Children’s Hospital, Boston, MA, USA
2Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA

Tài liệu tham khảo

2019, Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2017: a systematic analysis for the Global Burden of Disease Study, JAMA Oncol., 5, 1749, 10.1001/jamaoncol.2019.2996 Shi, 2017, Cancer nanomedicine: progress, challenges and opportunities, Nat. Rev. Cancer, 17, 20, 10.1038/nrc.2016.108 Mitchell, 2017, Engineering and physical sciences in oncology: challenges and opportunities, Nat. Rev. Cancer, 17, 659, 10.1038/nrc.2017.83 Chen, 2017, Rethinking cancer nanotheranostics, Nat. Rev. Mater., 2, 10.1038/natrevmats.2017.24 Tran and Tran, 2019, Nanoconjugation and encapsulation strategies for improving drug delivery and therapeutic efficacy of poorly water-soluble drugs, Pharmaceutics, 11, 325, 10.3390/pharmaceutics11070325 Wang, 2017, External triggering and triggered targeting strategies for drug delivery, Nat. Rev. Mater., 2, 10.1038/natrevmats.2017.20 He, 2019, Survey of clinical translation of cancer nanomedicines – lessons learned from successes and failures, Acc. Chem. Res., 52, 2445, 10.1021/acs.accounts.9b00228 Wolfram, 2019, Clinical cancer nanomedicine, Nano Today, 25, 85, 10.1016/j.nantod.2019.02.005 Wan, 2020, Genome editing of mutant KRAS through supramolecular polymer-mediated delivery of Cas9 ribonucleoprotein for colorectal cancer therapy, J. Control. Release, 322, 236, 10.1016/j.jconrel.2020.03.015 Chiang, 2018, Combination of fucoidan-based magnetic nanoparticles and immunomodulators enhances tumour-localized immunotherapy, Nat. Nanotechnol., 13, 746, 10.1038/s41565-018-0146-7 Morad, 2019, Tumor-derived extracellular vesicles breach the intact blood–brain barrier via transcytosis, ACS Nano, 13, 13853, 10.1021/acsnano.9b04397 Kingston, 2019, Assessing micrometastases as a target for nanoparticles using 3D microscopy and machine learning, Proc. Natl. Acad. Sci. U. S. A., 116, 14937, 10.1073/pnas.1907646116 Zhou, 2017, Nonviral cancer gene therapy: delivery cascade and vector nanoproperty integration, Adv. Drug Deliv. Rev., 115, 115, 10.1016/j.addr.2017.07.021 Anguela, 2019, Entering the modern era of gene therapy, Annu. Rev. Med., 70, 273, 10.1146/annurev-med-012017-043332 Rui, 2019, Non-viral delivery to enable genome editing, Trends Biotechnol., 37, 281, 10.1016/j.tibtech.2018.08.010 Ginn, 2018, Gene therapy clinical trials worldwide to 2017: an update, J. Gene Med., 20, 10.1002/jgm.3015 Carboni, 2019, Synthetic vehicles for encapsulation and delivery of CRISPR/Cas9 gene editing machinery, Adv. Ther., 2 Kong, 2019, Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition, Sci. Transl. Med., 11, 10.1126/scitranslmed.aaw1565 Islam, 2018, Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA, Nat. Biomed. Eng., 2, 850, 10.1038/s41551-018-0284-0 Wang, 2018, Shape-controlled magnetic mesoporous silica nanoparticles for magnetically-mediated suicide gene therapy of hepatocellular carcinoma, Biomaterials, 154, 147, 10.1016/j.biomaterials.2017.10.047 Mullen, 1994, Tumors expressing the cytosine deaminase suicide gene can be eliminated in vivo with 5-fluorocytosine and induce protective immunity to wild type tumor, Cancer Res., 54, 1503 Rossignoli, 2019, Inducible caspase 9-mediated suicide gene for MSC-based cancer gene therapy, Cancer Gene Ther., 26, 11, 10.1038/s41417-018-0034-1 Erkan, 2017, Extracellular vesicle-mediated suicide mRNA/protein delivery inhibits glioblastoma tumor growth in vivo, Cancer Gene Ther., 24, 38, 10.1038/cgt.2016.78 Kowalski, 2019, Delivering the messenger: advances in technologies for therapeutic mRNA delivery, Mol. Ther., 27, 710, 10.1016/j.ymthe.2019.02.012 Hajj, 2017, Tools for translation: non-viral materials for therapeutic mRNA delivery, Nat. Rev. Mater., 2, 1, 10.1038/natrevmats.2017.56 Patel, 2019, Inhaled nanoformulated mRNA polyplexes for protein production in lung epithelium, Adv. Mater., 31 Kaczmarek, 2018, Optimization of a degradable polymer–lipid nanoparticle for potent systemic delivery of mRNA to the lung endothelium and immune cells, Nano Lett., 18, 6449, 10.1021/acs.nanolett.8b02917 Oberli, 2017, Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy, Nano Lett., 17, 1326, 10.1021/acs.nanolett.6b03329 Paunovska, 2019, Nanoparticles containing oxidized cholesterol deliver mRNA to the liver microenvironment at clinically relevant doses, Adv. Mater., 31 McKinlay, 2017, Charge-altering releasable transporters (CARTs) for the delivery and release of mRNA in living animals, Proc. Natl. Acad. Sci. U. S. A., 114, E448, 10.1073/pnas.1614193114 Haabeth, 2019, Local delivery of Ox40l, Cd80, and Cd86 mRNA kindles global anticancer immunity, Cancer Res., 79, 1624, 10.1158/0008-5472.CAN-18-2867 Yoshinaga, 2019, Bundling mRNA strands to prepare nano-assemblies with enhanced stability towards rnase for in vivo delivery, Angew. Chem. Int. Ed. Engl., 58, 11360, 10.1002/anie.201905203 Li, 2017, Structurally programmed assembly of translation initiation nanoplex for superior mRNA delivery, ACS Nano, 11, 2531, 10.1021/acsnano.6b08447 Li, 2017, Polyamine-mediated stoichiometric assembly of ribonucleoproteins for enhanced mRNA delivery, Angew. Chem. Int. Ed. Engl., 56, 13709, 10.1002/anie.201707466 Forterre, 2020, Extracellular vesicle-mediated in vitro transcribed mrna delivery for treatment of HER2+ breast cancer xenografts in mice by prodrug CB1954 without general toxicity, Mol. Cancer Ther., 19, 858, 10.1158/1535-7163.MCT-19-0928 Hoy, 2018, Patisiran: first global approval, Drugs, 78, 1625, 10.1007/s40265-018-0983-6 Zheng, 2018, Nanotechnology-nased strategies for siRNA brain delivery for disease therapy, Trends Biotechnol., 36, 562, 10.1016/j.tibtech.2018.01.006 Park, 2016, Pharmacokinetics and biodistribution of recently-developed siRNA nanomedicines, Adv. Drug Deliv. Rev., 104, 93, 10.1016/j.addr.2015.12.004 Kim, 2016, Recent progress in development of siRNA delivery vehicles for cancer therapy, Adv. Drug Deliv. Rev., 104, 61, 10.1016/j.addr.2016.06.011 Yin, 2016, Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo, Nat. Biotechnol., 34, 328, 10.1038/nbt.3471 Chen, 2019, A biodegradable nanocapsule delivers a Cas9 ribonucleoprotein complex for in vivo genome editing, Nat. Nanotechnol., 14, 974, 10.1038/s41565-019-0539-2 Liu, 2019, Fast and efficient CRISPR/Cas9 genome editing in vivo enabled by bioreducible lipid and messenger RNA nanoparticles, Adv. Mater., 31, 10.1002/adma.201902575 Xu, 2019, Development of 'CLAN' nanomedicine for nucleic acid therapeutics, Small, 15, 10.1002/smll.201900055 Guo, 2019, Therapeutic genome editing of triple-negative breast tumors using a noncationic and deformable nanolipogel, Proc. Natl. Acad. Sci. U. S. A., 116, 18295, 10.1073/pnas.1904697116 Wang, 2017, Genome editing for cancer therapy: delivery of Cas9 protein/sgRNA plasmid via a gold nanocluster/lipid core–shell nanocarrier, Adv. Sci., 4, 10.1002/advs.201700175 Mout, 2017, Direct cytosolic delivery of CRISPR/Cas9-ribonucleoprotein for efficient gene editing, ACS Nano, 11, 2452, 10.1021/acsnano.6b07600 Guo, 2018, Nanoparticle elasticity directs tumor uptake, Nat. Commun., 9, 130, 10.1038/s41467-017-02588-9 Yin, 2016, Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo, Nat. Biotechnol., 34, 328, 10.1038/nbt.3471 Maeder, 2019, Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10, Nat. Med., 25, 229, 10.1038/s41591-018-0327-9 De Ravin, 2017, CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease, Sci. Transl. Med., 9, 10.1126/scitranslmed.aah3480 Irvine, 2020, Enhancing cancer immunotherapy with nanomedicine, Nat. Rev. Immunol., 20, 321, 10.1038/s41577-019-0269-6 Martin, 2020, Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges, Nat. Rev. Clin. Oncol., 17, 251, 10.1038/s41571-019-0308-z Goldberg, 2019, Improving cancer immunotherapy through nanotechnology, Nat. Rev. Cancer, 19, 587, 10.1038/s41568-019-0186-9 Chauhan, 2019, Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy, Proc. Natl. Acad. Sci. U. S. A., 116, 10674, 10.1073/pnas.1819889116 Zhang, 2018, Nanoparticle anchoring targets immune agonists to tumors enabling anti-cancer immunity without systemic toxicity, Nat. Commun., 9 Schmid, 2017, T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity, Nat. Commun., 8, 1747, 10.1038/s41467-017-01830-8 Rodell, 2018, TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy, Nat. Biomed. Eng., 2, 578, 10.1038/s41551-018-0236-8 Chen, 2020, Intratumoral delivery of CCL25 enhances immunotherapy against triple-negative breast cancer by recruiting CCR9+ T cells, Sci. Adv., 6 Xu, 2020, Nano-puerarin regulates tumor microenvironment and facilitates chemo- and immunotherapy in murine triple negative breast cancer model, Biomaterials, 235, 10.1016/j.biomaterials.2020.119769 Chen, 2019, Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer, Proc. Natl. Acad. Sci. U. S. A., 116, 4558, 10.1073/pnas.1815515116 Billingsley, 2020, Ionizable lipid nanoparticle-mediated mRNA delivery for human CAR T cell engineering, Nano Lett., 20, 1578, 10.1021/acs.nanolett.9b04246 Olden, 2018, Cationic polymers for non-viral gene delivery to human T cells, J. Control. Release, 282, 140, 10.1016/j.jconrel.2018.02.043 Smith, 2017, In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers, Nat. Nanotechnol., 12, 813, 10.1038/nnano.2017.57 Tang, 2018, Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery, Nat. Biotechnol., 36, 707, 10.1038/nbt.4181 Zhu, 2017, Intertwining DNA–RNA nanocapsules loaded with tumor neoantigens as synergistic nanovaccines for cancer immunotherapy, Nat. Commun., 8, 10.1038/s41467-017-01386-7 Kuai, 2017, Designer vaccine nanodiscs for personalized cancer immunotherapy, Nat. Mater., 16, 489, 10.1038/nmat4822 Shae, 2019, Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy, Nat. Nanotechnol., 14, 269, 10.1038/s41565-018-0342-5 Min, 2017, Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy, Nat. Nanotechnol., 12, 877, 10.1038/nnano.2017.113 Chen, 2018, Clay nanoparticles elicit long-term immune responses by forming biodegradable depots for sustained antigen stimulation, Small, 14, 10.1002/smll.201704465 Kapadia, 2018, Extending antigen release from particulate vaccines results in enhanced antitumor immune response, J. Control. Release, 269, 393, 10.1016/j.jconrel.2017.11.020 Wiklander, 2019, Advances in therapeutic applications of extracellular vesicles, Sci. Transl. Med., 11, 10.1126/scitranslmed.aav8521 Xu, 2018, Extracellular vesicles in cancer – implications for future improvements in cancer care, Nat. Rev. Clin. Oncol., 15, 617, 10.1038/s41571-018-0036-9 Maas, 2017, Extracellular vesicles: unique intercellular delivery vehicles, Trends Cell Biol., 27, 172, 10.1016/j.tcb.2016.11.003 Pi, 2018, Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression, Nat. Nanotechnol., 13, 82, 10.1038/s41565-017-0012-z Yang, 2020, Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation, Nat. Biomed. Eng., 4, 69, 10.1038/s41551-019-0485-1 Nie, 2020, Use of lung-specific exosomes for miRNA-126 delivery in non-small cell lung cancer, Nanoscale, 12, 877, 10.1039/C9NR09011H Kim, 2017, Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting, J. Control. Release, 266, 8, 10.1016/j.jconrel.2017.09.013 Morishita, 2016, Exosome-based tumor antigens-adjuvant co-delivery utilizing genetically engineered tumor cell-derived exosomes with immunostimulatory CpG DNA, Biomaterials, 111, 55, 10.1016/j.biomaterials.2016.09.031 Garofalo, 2019, Extracellular vesicles enhance the targeted delivery of immunogenic oncolytic adenovirus and paclitaxel in immunocompetent mice, J. Control. Release, 294, 165, 10.1016/j.jconrel.2018.12.022 Hoshino, 2015, Tumour exosome integrins determine organotropic metastasis, Nature, 527, 329, 10.1038/nature15756 Morad, 2019, Brainwashed by extracellular vesicles: the role of extracellular vesicles in primary and metastatic brain tumour microenvironment, J. Extracell. Vesicles, 8, 10.1080/20013078.2019.1627164 Wang, 2019, Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation, Theranostics, 9, 1714, 10.7150/thno.30716 O’Brien, 2015, miR-134 in extracellular vesicles reduces triple-negative breast cancer aggression and increases drug sensitivity, Oncotarget, 6, 32774, 10.18632/oncotarget.5192 Watson, 2016, Efficient production and enhanced tumor delivery of engineered extracellular vesicles, Biomaterials, 105, 195, 10.1016/j.biomaterials.2016.07.003 Lunavat, 2016, RNAi delivery by exosome-mimetic nanovesicles – implications for targeting c-Myc in cancer, Biomaterials, 102, 231, 10.1016/j.biomaterials.2016.06.024 Yang, 2016, Functional exosome-mimic for deliv ery of siRNA to cancer: in vitro and in vivo evaluation, J. Control. Release, 243, 160, 10.1016/j.jconrel.2016.10.008 Kooijmans, 2013, Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles, J. Control. Release, 172, 229, 10.1016/j.jconrel.2013.08.014 Lamichhane, 2015, Exogenous DNA loading into extracellular vesicles via electroporation is size-dependent and enables limited gene delivery, Mol. Pharm., 12, 3650, 10.1021/acs.molpharmaceut.5b00364 Jeyaram, 2020, Enhanced loading of functional miRNA cargo via pH gradient modification of extracellular vesicles, Mol. Ther., 28, 975, 10.1016/j.ymthe.2019.12.007 Fuhrmann, 2015, Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins, J. Control. Release, 205, 35, 10.1016/j.jconrel.2014.11.029 Gao, 2018, Anchor peptide captures, targets, and loads exosomes of diverse origins for diagnostics and therapy, Sci. Transl. Med., 10, 10.1126/scitranslmed.aat0195 Piffoux, 2018, Modification of extracellular vesicles by fusion with liposomes for the design of personalized biogenic drug delivery systems, ACS Nano, 12, 6830, 10.1021/acsnano.8b02053 Reshke, 2020, Reduction of the therapeutic dose of silencing RNA by packaging it in extracellular vesicles via a pre-microRNA backbone, Nat. Biomed. Eng., 4, 52, 10.1038/s41551-019-0502-4 Sawada, 2020, Nanogel hybrid assembly for exosome intracellular delivery: effects on endocytosis and fusion by exosome surface polymer engineering, Biomater. Sci., 8, 619, 10.1039/C9BM01232J de Jong, 2019, Drug delivery with extracellular vesicles: from imagination to innovation, Acc. Chem. Res., 52, 1761, 10.1021/acs.accounts.9b00109 Smyth, 2015, Biodistribution and delivery efficiency of unmodified tumor-derived exosomes, J. Control. Release, 199, 145, 10.1016/j.jconrel.2014.12.013 Zhu, 2019, Embryonic stem cells-derived exosomes endowed with targeting properties as chemotherapeutics delivery vehicles for glioblastoma therapy, Adv. Sci., 6, 10.1002/advs.201801899 Kooijmans, 2016, PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time, J. Control. Release, 224, 77, 10.1016/j.jconrel.2016.01.009 Alvarez-Erviti, 2011, Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes, Nat. Biotechnol., 29, 341, 10.1038/nbt.1807 Kooijmans, 2016, Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting, J. Extracell. Vesicles, 5, 10.3402/jev.v5.31053 Royo, 2019, Modification of the glycosylation of extracellular vesicles alters their biodistribution in mice, Nanoscale, 11, 1531, 10.1039/C8NR03900C Topol, 2019, High-performance medicine: the convergence of human and artificial intelligence, Nat. Med., 25, 44, 10.1038/s41591-018-0300-7 McKinney, 2020, International evaluation of an AI system for breast cancer screening, Nature, 577, 89, 10.1038/s41586-019-1799-6 Yamankurt, 2019, Exploration of the nanomedicine-design space with high-throughput screening and machine learning, Nat. Biomed. Eng., 3, 318, 10.1038/s41551-019-0351-1 Li, 2019, Design of self-assembly dipeptide hydrogels and machine learning via their chemical features, Proc. Natl. Acad. Sci. U. S. A., 116, 11259, 10.1073/pnas.1903376116 Han, 2019, Predicting physical stability of solid dispersions by machine learning techniques, J. Control. Release, 311, 16, 10.1016/j.jconrel.2019.08.030 Shamay, 2018, Quantitative self-assembly prediction yields targeted nanomedicines, Nat. Mater., 17, 361, 10.1038/s41563-017-0007-z Price, 2019, An in vitro assay and artificial intelligence approach to determine rate constants of nanomaterial–cell interactions, Sci. Rep., 9 Large, 2019, Advances in receptor-mediated, tumor-targeted drug delivery, Adv. Ther., 2 Alshaer, 2018, Aptamer-guided nanomedicines for anticancer drug delivery, Adv. Drug Deliv. Rev., 134, 122, 10.1016/j.addr.2018.09.011 Guo, 2019, Dual complementary liposomes inhibit triple-negative breast tumor progression and metastasis, Sci. Adv., 5, 10.1126/sciadv.aav5010 Ho, 2019, Artificial intelligence in nanomedicine, Nanoscale Horiz., 4, 365, 10.1039/C8NH00233A Zhang, 2017, Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer, Nat. Commun., 8, 10.1038/s41467-017-01968-5 Chmielecki, 2011, Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling, Sci. Transl. Med., 3, 90ra59, 10.1126/scitranslmed.3002356 Enriquez-Navas, 2016, Exploiting evolutionary principles to prolong tumor control in preclinical models of breast cancer, Sci. Transl. Med., 8, 327ra24, 10.1126/scitranslmed.aad7842 Wang, 2015, Mechanism-independent pptimization of combinatorial nanodiamond and unmodified drug delivery using a phenotypically driven platform technology, ACS Nano, 9, 3332, 10.1021/acsnano.5b00638 von Maltzahn, 2011, Nanoparticles that communicate in vivo to amplify tumour targeting, Nat. Mater., 10, 545, 10.1038/nmat3049