Future of nanotherapeutics: Targeting the cellular sub-organelles.

Many diseases originate from alterations at nanoscale levels. Precise drug delivery should be achieved not only at cell level, but also at organelle level to achieve maximum therapeutic responses as well as avoiding possible toxic side effects of the drugs. However, organelles and subcellular structures are natural barriers that hampering many therapeutics from taking effects. Nanodelivery vehicle is a favorable platform to navigate across physiological barriers and to achieve selective delivery of therapeutic and diagnostic agents to intracellular targets. In this review, we have highlighted recent innovations in organelle-targeted nanomaterials designed to treat a variety of currently challenging diseases. Targeting strategies of four main kinds of organelles: mitochondria, nucleus, lysosomes and endoplasmic reticulum are discussed in detail. This review will help to clarify the intracellular nanomaterial-organelle interactions, and understand the fundamentals of organelle-targeted drug delivery strategies, which is of vital importance for the design and successful biomedical applications of nanomaterials in therapeutic treatments. At the end of this review, challenge and perspectives of organelle-targeted nanotherapy are discussed.

[1]  M. Lahiri,et al.  Dual drug conjugated nanoparticle for simultaneous targeting of mitochondria and nucleus in cancer cells. , 2015, ACS applied materials & interfaces.

[2]  Yasuo Yoshioka,et al.  A strategy for efficient cross-presentation of CTL-epitope peptides leading to enhanced induction of in vivo tumor immunity. , 2007, Journal of controlled release : official journal of the Controlled Release Society.

[3]  V. Torchilin,et al.  Organelle-targeted nanocarriers: specific delivery of liposomal ceramide to mitochondria enhances its cytotoxicity in vitro and in vivo. , 2008, Nano letters.

[4]  Cuichen Wu,et al.  A Cell-Targeted, Size-Photocontrollable, Nuclear-Uptake Nanodrug Delivery System for Drug-Resistant Cancer Therapy , 2014, Nano letters.

[5]  Dar-Bin Shieh,et al.  Stabilizer-free poly(lactide-co-glycolide) nanoparticles for multimodal biomedical probes. , 2008, Biomaterials.

[6]  Y. Yoshioka,et al.  Induction of Endoplasmic Reticulum–Endosome Fusion for Antigen Cross-Presentation Induced by Poly (γ-Glutamic Acid) Nanoparticles , 2011, The Journal of Immunology.

[7]  D. Wirtz,et al.  Efficient active transport of gene nanocarriers to the cell nucleus , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[8]  Jinchao Zhang,et al.  Ultrasmall Gold Nanoparticles as Carriers for Nucleus-Based Gene Therapy Due to Size-Dependent Nuclear Entry , 2014, ACS nano.

[9]  D A Jans,et al.  Signals mediating nuclear targeting and their regulation: Application in drug delivery , 1998, Medicinal research reviews.

[10]  I. Komuro,et al.  Endoplasmic reticulum stress as a therapeutic target in cardiovascular disease. , 2010, Circulation research.

[11]  Y. Liu,et al.  Selective targeting of gold nanorods at the mitochondria of cancer cells: implications for cancer therapy. , 2011, Nano letters.

[12]  G. Pastorin,et al.  Enhanced cytotoxicity to cancer cells by mitochondria-targeting MWCNTs containing platinum(IV) prodrug of cisplatin. , 2014, Biomaterials.

[13]  Ge Lin,et al.  Rapid endosomal escape of prickly nanodiamonds: implications for gene delivery , 2015, Scientific Reports.

[14]  M. Akashi,et al.  Nanoparticles built by self-assembly of amphiphilic gamma-PGA can deliver antigens to antigen-presenting cells with high efficiency: a new tumor-vaccine carrier for eliciting effector T cells. , 2008, Vaccine.

[15]  M. Garcia-Parajo,et al.  pH-responsive polysaccharide-based polyelectrolyte complexes as nanocarriers for lysosomal delivery of therapeutic proteins. , 2011, Biomacromolecules.

[16]  R. Dwek,et al.  Uptake and trafficking of liposomes to the endoplasmic reticulum , 2010, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[17]  S. Muro,et al.  Altered Clathrin-Independent Endocytosis in Type A Niemann-Pick Disease Cells and Rescue by ICAM-1-Targeted Enzyme Delivery. , 2015, Molecular pharmaceutics.

[18]  M. El-Sayed,et al.  Nuclear targeting of gold nanoparticles in cancer cells induces DNA damage, causing cytokinesis arrest and apoptosis. , 2010, Journal of the American Chemical Society.

[19]  S. Dhar,et al.  The energy blocker inside the power house: mitochondria targeted delivery of 3-bromopyruvate , 2014, Chemical science.

[20]  L. Rajendran,et al.  Subcellular targeting strategies for drug design and delivery , 2010, Nature Reviews Drug Discovery.

[21]  Ya-ju Chang,et al.  Synergism through combination of chemotherapy and oxidative stress-induced autophagy in A549 lung cancer cells using redox-responsive nanohybrids: a new strategy for cancer therapy. , 2015, Biomaterials.

[22]  F. Scaglia,et al.  Mitochondrial cytopathies. , 2016, Cell calcium.

[23]  Huangxian Ju,et al.  Cell-specific and pH-activatable rubyrin-loaded nanoparticles for highly selective near-infrared photodynamic therapy against cancer. , 2013, Journal of the American Chemical Society.

[24]  Anthony H. Futerman,et al.  The cell biology of lysosomal storage disorders , 2004, Nature Reviews Molecular Cell Biology.

[25]  Samir Mitragotri,et al.  Control of endothelial targeting and intracellular delivery of therapeutic enzymes by modulating the size and shape of ICAM-1-targeted carriers. , 2008, Molecular therapy : the journal of the American Society of Gene Therapy.

[26]  S. Kelley,et al.  Targeted delivery of doxorubicin to mitochondria. , 2013, ACS chemical biology.

[27]  S. Cohen,et al.  ER stress potentiates insulin resistance through PERK-mediated FOXO phosphorylation. , 2013, Genes & development.

[28]  S. Dhar,et al.  Engineering of blended nanoparticle platform for delivery of mitochondria-acting therapeutics , 2012, Proceedings of the National Academy of Sciences.

[29]  Y. Shinohara,et al.  Quantitative studies on the nuclear transport of plasmid DNA and gene expression employing nonviral vectors. , 2001, Advanced drug delivery reviews.

[30]  Fang Zeng,et al.  Dual-targeting nanosystem for enhancing photodynamic therapy efficiency. , 2015, ACS applied materials & interfaces.

[31]  Priscille Brodin,et al.  A Truncated HIV-1 Tat Protein Basic Domain Rapidly Translocates through the Plasma Membrane and Accumulates in the Cell Nucleus* , 1997, The Journal of Biological Chemistry.

[32]  Virander S. Chauhan,et al.  Self-assembled nanoparticles based on modified cationic dipeptides and DNA: novel systems for gene delivery , 2013, Journal of Nanobiotechnology.

[33]  H. Harashima,et al.  Mitochondrial targeting functional peptides as potential devices for the mitochondrial delivery of a DF-MITO-Porter. , 2013, Mitochondrion.

[34]  V. Muzykantov,et al.  Lysosomal enzyme delivery by ICAM-1-targeted nanocarriers bypassing glycosylation- and clathrin-dependent endocytosis. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[35]  D. Stepensky,et al.  Intracellular targeting of PLGA nanoparticles encapsulating antigenic peptide to the endoplasmic reticulum of dendritic cells and its effect on antigen cross-presentation in vitro. , 2011, Molecular pharmaceutics.

[36]  Erik C. Dreaden,et al.  Detecting and destroying cancer cells in more than one way with noble metals and different confinement properties on the nanoscale. , 2012, Accounts of chemical research.

[37]  F. Rodríguez,et al.  Targeting plasmid‐encoded proteins to the antigen presentation pathways , 2004, Immunological reviews.

[38]  L. Du,et al.  Targeted surface-functionalized gold nanoclusters for mitochondrial imaging. , 2014, Biosensors & bioelectronics.

[39]  Qianjun He,et al.  MSN‐Mediated Sequential Vascular‐to‐Cell Nuclear‐Targeted Drug Delivery for Efficient Tumor Regression , 2014, Advanced materials.

[40]  Da Ma,et al.  Enhancing endosomal escape for nanoparticle mediated siRNA delivery. , 2014, Nanoscale.

[41]  Vladimir P Torchilin,et al.  Recent approaches to intracellular delivery of drugs and DNA and organelle targeting. , 2006, Annual review of biomedical engineering.

[42]  Yan-jun Zhong,et al.  Cathepsin B-cleavable doxorubicin prodrugs for targeted cancer therapy , 2012, International journal of oncology.

[43]  Erkki Ruoslahti,et al.  Targeting of drugs and nanoparticles to tumors , 2010, The Journal of cell biology.

[44]  Yu Chen,et al.  Nuclear-targeted drug delivery of TAT peptide-conjugated monodisperse mesoporous silica nanoparticles. , 2012, Journal of the American Chemical Society.

[45]  V. Lukyanenko Permeabilization of cell membrane for delivery of nano-objects to cellular sub-domains. , 2013, Methods in molecular biology.

[46]  H. Wootz,et al.  ER stress and neurodegenerative diseases , 2006, Cell Death and Differentiation.

[47]  R. Iacobazzi,et al.  In vitro targeting and imaging the translocator protein TSPO 18-kDa through G(4)-PAMAM-FITC labeled dendrimer. , 2013, Journal of controlled release : official journal of the Controlled Release Society.

[48]  Guankui Wang,et al.  KDEL peptide gold nanoconstructs: promising nanoplatforms for drug delivery. , 2013, Nanomedicine : nanotechnology, biology, and medicine.

[49]  D. Maysinger,et al.  Design and evaluation of multifunctional nanocarriers for selective delivery of coenzyme Q10 to mitochondria. , 2012, Biomacromolecules.

[50]  Samir Mitragotri,et al.  Multifunctional nanoparticles for drug delivery and molecular imaging. , 2013, Annual review of biomedical engineering.

[51]  R. Hill,et al.  High efficacy gold-KDEL peptide-siRNA nanoconstruct-mediated transfection in C2C12 myoblasts and myotubes. , 2014, Nanomedicine : nanotechnology, biology, and medicine.

[52]  Yuyang Sun,et al.  Functional role of TRP channels in modulating ER stress and Autophagy. , 2016, Cell calcium.

[53]  D. Stojanovski,et al.  Import of Nuclear‐Encoded Proteins into Mitochondria , 2003, Experimental physiology.

[54]  Elena Aznar,et al.  Enzyme-responsive intracellular controlled release using nanometric silica mesoporous supports capped with "saccharides". , 2010, ACS nano.

[55]  W. Lederer,et al.  Probing the outer mitochondrial membrane in cardiac mitochondria with nanoparticles. , 2007, Biophysical journal.

[56]  Masato Yasuhara,et al.  Quantum Dots Targeted to the Assigned Organelle in Living Cells , 2004, Microbiology and immunology.

[57]  Betty Y. S. Kim,et al.  Current concepts: Nanomedicine , 2010 .

[58]  Silvia Muro,et al.  Enhanced endothelial delivery and biochemical effects of α-galactosidase by ICAM-1-targeted nanocarriers for Fabry disease. , 2011, Journal of controlled release : official journal of the Controlled Release Society.

[59]  W. Mark Saltzman,et al.  Therapeutic siRNA: Principles, Challenges, and Strategies , 2012, The Yale journal of biology and medicine.

[60]  L. C. Moore,et al.  Nuclear envelope permeability , 1975, Nature.

[61]  Meng Zheng,et al.  pH-sensitive degradable chimaeric polymersomes for the intracellular release of doxorubicin hydrochloride. , 2012, Biomaterials.

[62]  V. Muzykantov,et al.  Polymer nanocarriers protecting active enzyme cargo against proteolysis. , 2005, Journal of controlled release : official journal of the Controlled Release Society.

[63]  Paul C. Wang,et al.  Single-walled carbon nanotubes alter cytochrome c electron transfer and modulate mitochondrial function. , 2012, ACS nano.

[64]  Youqing Shen,et al.  Nuclear drug delivery for cancer chemotherapy. , 2011, Journal of controlled release : official journal of the Controlled Release Society.

[65]  H. Harashima,et al.  Multifunctional envelope-type nano device (MEND) for organelle targeting via a stepwise membrane fusion process. , 2012, Methods in enzymology.

[66]  Teri W Odom,et al.  Direct observation of nanoparticle-cancer cell nucleus interactions. , 2012, ACS nano.

[67]  Tim Leshuk,et al.  Emerging nanomaterials for targeting subcellular organelles , 2011 .

[68]  F. Lim,et al.  General Considerations on the Biosafety of Virus-derived Vectors Used in Gene Therapy and Vaccination , 2013, Current gene therapy.

[69]  H. Harashima,et al.  Enhancement in selective mitochondrial association by direct modification of a mitochondrial targeting signal peptide on a liposomal based nanocarrier. , 2013, Mitochondrion.

[70]  Peng Huang,et al.  ROS stress in cancer cells and therapeutic implications. , 2004, Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy.

[71]  P. Couvreur,et al.  Self-assembled squalenoylated penicillin bioconjugates: an original approach for the treatment of intracellular infections. , 2012, ACS nano.

[72]  Sangeeta N. Bhatia,et al.  Intracellular Delivery of Quantum Dots for Live Cell Labeling and Organelle Tracking , 2004 .

[73]  S. Weinberg,et al.  Targeting mitochondria metabolism for cancer therapy. , 2015, Nature chemical biology.

[74]  Michal Juhas,et al.  Gold nanoparticles induce nuclear damage in breast cancer cells, which is further amplified by hyperthermia , 2014, Cellular and Molecular Life Sciences.

[75]  S. Kelley,et al.  Mitochondria‐Penetrating Peptides: Sequence Effects and Model Cargo Transport , 2009, Chembiochem : a European journal of chemical biology.

[76]  J. Nunnari,et al.  Mitochondrial form and function , 2014, Nature.

[77]  R. Dwek,et al.  Polyunsaturated liposomes are antiviral against hepatitis B and C viruses and HIV by decreasing cholesterol levels in infected cells , 2010, Proceedings of the National Academy of Sciences.

[78]  R. Dwek,et al.  Liposome-Mediated Delivery of Iminosugars Enhances Efficacy against Dengue Virus In Vivo , 2012, Antimicrobial Agents and Chemotherapy.

[79]  Y. Zhan,et al.  Targeted charge-reversal nanoparticles for nuclear drug delivery. , 2007, Angewandte Chemie.

[80]  Omid C Farokhzad,et al.  Insight into nanoparticle cellular uptake and intracellular targeting. , 2014, Journal of controlled release : official journal of the Controlled Release Society.

[81]  V. Torchilin,et al.  Intracellular Targets for DNA Delivery: Nuclei and Mitochondria , 2002, Somatic cell and molecular genetics.

[82]  Timothy J. Nelson,et al.  Nuclear Transport: Target for Therapy , 2007, Clinical pharmacology and therapeutics.

[83]  Xian‐Zheng Zhang,et al.  Dual-pH Sensitive Charge-Reversal Polypeptide Micelles for Tumor-Triggered Targeting Uptake and Nuclear Drug Delivery. , 2015, Small.

[84]  Dai Fukumura,et al.  Multistage nanoparticle delivery system for deep penetration into tumor tissue , 2011, Proceedings of the National Academy of Sciences.

[85]  E. Ruoslahti,et al.  Targeted nanoparticle enhanced proapoptotic peptide as potential therapy for glioblastoma , 2011, Proceedings of the National Academy of Sciences.

[86]  Katarina Kågedal,et al.  The lysosome: from waste bag to potential therapeutic target. , 2013, Journal of molecular cell biology.

[87]  M. Prato,et al.  Peptide-based carbon nanotubes for mitochondrial targeting. , 2013, Nanoscale.

[88]  I. Tabas,et al.  Role of endoplasmic reticulum stress in metabolic disease and other disorders. , 2012, Annual review of medicine.

[89]  Takenori Yamamoto,et al.  MITO-Porter: A liposome-based carrier system for delivery of macromolecules into mitochondria via membrane fusion. , 2008, Biochimica et biophysica acta.

[90]  Kirsten Sandvig,et al.  Endocytosis and intracellular transport of nanoparticles: Present knowledge and need for future studies , 2011 .

[91]  V. Weissig,et al.  Mitochondrial pharmaceutics. , 2004, Mitochondrion.

[92]  H. Padh,et al.  Organelle targeting: third level of drug targeting , 2013, Drug design, development and therapy.

[93]  S. Kelley,et al.  Mitochondria-penetrating peptides. , 2008, Chemistry & biology.

[94]  J. Feijen,et al.  Lysosomally cleavable peptide-containing polymersomes modified with anti-EGFR antibody for systemic cancer chemotherapy. , 2011, Biomaterials.

[95]  Jing Wang,et al.  Pegylated phospholipid micelles induce endoplasmic reticulum-dependent apoptosis of cancer cells but not normal cells. , 2012, ACS nano.

[96]  K. Raghu,et al.  A lysosome-targeted drug delivery system based on sorbitol backbone towards efficient cancer therapy. , 2014, Organic & biomolecular chemistry.

[97]  O. Inanami,et al.  Radiosensitization of tumor cells through endoplasmic reticulum stress induced by PEGylated nanogel containing gold nanoparticles. , 2014, Cancer letters.

[98]  Silvia Muro,et al.  Enhanced delivery of α-glucosidase for Pompe disease by ICAM-1-targeted nanocarriers: comparative performance of a strategy for three distinct lysosomal storage disorders. , 2012, Nanomedicine : nanotechnology, biology, and medicine.

[99]  S. Amigorena,et al.  Intracellular mechanisms of antigen cross presentation in dendritic cells. , 2010, Current opinion in immunology.

[100]  Liangping Zhou,et al.  Simultaneous nuclear imaging and intranuclear drug delivery by nuclear-targeted multifunctional upconversion nanoprobes. , 2012, Biomaterials.

[101]  Yaping Li,et al.  Large‐Pore Ultrasmall Mesoporous Organosilica Nanoparticles: Micelle/Precursor Co‐templating Assembly and Nuclear‐Targeted Gene Delivery , 2015, Advanced materials.

[102]  R. Kaufman,et al.  Endoplasmic reticulum stress and type 2 diabetes. , 2012, Annual review of biochemistry.