Tumor target amplification: Implications for nano drug delivery systems

&NA; Tumor cells overexpress surface markers which are absent from normal cells. These tumor‐restricted antigenic signatures are a fundamental basis for distinguishing on‐target from off‐target cells for ligand‐directed targeting of cancer cells. Unfortunately, tumor heterogeneity impedes the establishment of a solid expression pattern for a given target marker, leading to drastic changes in quality (availability) and quantity (number) of the target. Consequently, a subset of cancer cells remains untargeted during the course of treatment, which subsequently promotes drug‐resistance and cancer relapse. Since target inefficiency is only problematic for cancer treatment and not for treatment of other pathological conditions such as viral/bacterial infections, target amplification or the generation of novel targets is key to providing eligible antigenic markers for effective targeted therapy. This review summarizes the limitations of current ligand‐directed targeting strategies and provides a comprehensive overview of tumor target amplification strategies, including self‐amplifying systems, dual targeting, artificial markers and peptide modification. We also discuss the therapeutic and diagnostic potential of these approaches, the underlying mechanism(s) and established methodologies, mostly in the context of different nanodelivery systems, to facilitate more effective ligand‐directed cancer cell monitoring and targeting. Graphical abstract Figure. No Caption available.

[1]  Xi Chen,et al.  Rational, modular adaptation of enzyme-free DNA circuits to multiple detection methods , 2011, Nucleic acids research.

[2]  S. Kizilel,et al.  Targeting cancer cells via tumor-homing peptide CREKA functional PEG nanoparticles. , 2016, Colloids and surfaces. B, Biointerfaces.

[3]  Erkki Ruoslahti,et al.  Tumor-Penetrating iRGD Peptide Inhibits Metastasis , 2014, Molecular Cancer Therapeutics.

[4]  D. Cheng,et al.  Novel DNA Polymer for Amplification Pretargeting. , 2015, ACS medicinal chemistry letters.

[5]  Jie Chao,et al.  Multivalent capture and detection of cancer cells with DNA nanostructured biosensors and multibranched hybridization chain reaction amplification. , 2014, Analytical chemistry.

[6]  K. Gustafsson,et al.  Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[7]  C. Bertozzi,et al.  A Strategy for the Selective Imaging of Glycans Using Caged Metabolic Precursors , 2010, Journal of the American Chemical Society.

[8]  R. Kontermann,et al.  Dual targeting strategies with bispecific antibodies , 2012, mAbs.

[9]  Tamer Refaat,et al.  Passive targeting of nanoparticles to cancer: A comprehensive review of the literature. , 2014, Molecular and clinical oncology.

[10]  N. Zarghami,et al.  Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation , 2017, Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine.

[11]  F. Malavasi,et al.  Retinoic acid-induced CD38 antigen as a target for immunotoxin-mediated killing of leukemia cells. , 2004, Molecular cancer therapeutics.

[12]  Shenghua Zhang,et al.  RGD and NGR modified TRAIL protein exhibited potent anti-metastasis effects on TRAIL-insensitive cancer cells in vitro and in vivo , 2017, Amino Acids.

[13]  P. Opolon,et al.  High vaccination efficiency of low-affinity epitopes in antitumor immunotherapy. , 2004, The Journal of clinical investigation.

[14]  Hyesung Jeon,et al.  Cellular uptake mechanism and intracellular fate of hydrophobically modified glycol chitosan nanoparticles. , 2009, Journal of controlled release : official journal of the Controlled Release Society.

[15]  Ruth Nussinov,et al.  Nanoparticle-induced vascular blockade in human prostate cancer. , 2010, Blood.

[16]  W. Arap,et al.  The neovasculature homing motif NGR: more than meets the eye. , 2008, Blood.

[17]  J Aaron,et al.  Directional conjugation of antibodies to nanoparticles for synthesis of multiplexed optical contrast agents with both delivery and targeting moieties , 2008, Nature Protocols.

[18]  Xin-guo Jiang,et al.  Precise glioma targeting of and penetration by aptamer and peptide dual-functioned nanoparticles. , 2012, Biomaterials.

[19]  Herren Wu,et al.  Insights into the molecular basis of a bispecific antibody's target selectivity , 2015, mAbs.

[20]  Eva Harth,et al.  Targeted nanoparticles that deliver a sustained, specific release of Paclitaxel to irradiated tumors. , 2010, Cancer research.

[21]  A. Pini,et al.  Cancer selectivity of tetrabranched neurotensin peptides is generated by simultaneous binding to sulfated glycosaminoglycans and protein receptors. , 2013, Journal of medicinal chemistry.

[22]  Emily Berry Two Birds , 2015 .

[23]  Youli Zu,et al.  Aptamers and their applications in nanomedicine. , 2015, Small.

[24]  Xiao-shi Zhang,et al.  Targeted therapy: resistance and re-sensitization , 2015, Chinese journal of cancer.

[25]  C. Swanton,et al.  Tumor Evolution as a Therapeutic Target. , 2017, Cancer discovery.

[26]  C. Ahn,et al.  Nano-sized metabolic precursors for heterogeneous tumor-targeting strategy using bioorthogonal click chemistry in vivo. , 2017, Biomaterials.

[27]  Weihong Tan,et al.  Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics , 2013, Proceedings of the National Academy of Sciences.

[28]  J W Smith,et al.  Integrin activation controls metastasis in human breast cancer. , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[29]  H. Abken,et al.  IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. , 2011, Cancer research.

[30]  R. Bellamkonda,et al.  A dual-ligand approach for enhancing targeting selectivity of therapeutic nanocarriers. , 2006, Journal of controlled release : official journal of the Controlled Release Society.

[31]  J. Pawelek,et al.  Tumor-targeted Salmonella as a novel anticancer vector. , 1997, Cancer research.

[32]  M. Czuczman,et al.  A Quantitative Exploration of Surface Antigen Expression in Common B-Cell Malignancies Using Flow Cytometry , 2006, Immunological investigations.

[33]  Jaime Conceição,et al.  Nanotechnological carriers for cancer chemotherapy: the state of the art. , 2015, Colloids and surfaces. B, Biointerfaces.

[34]  A. Roque,et al.  Antibody-conjugated nanoparticles for therapeutic applications. , 2012, Current medicinal chemistry.

[35]  S. Menichetti,et al.  Tumor-selective peptide-carrier delivery of Paclitaxel increases in vivo activity of the drug , 2015, Scientific Reports.

[36]  S. Bhatia,et al.  Drug-induced amplification of nanoparticle targeting to tumors. , 2014, Nano today.

[37]  S. Rabbani,et al.  Binding and internalization of NGR-peptide-targeted liposomal doxorubicin (TVT-DOX) in CD13-expressing cells and its antitumor effects , 2007, Anti-cancer drugs.

[38]  Alessandro Gori,et al.  NGR-tagged nano-gold: A new CD13-selective carrier for cytokine delivery to tumors , 2016, Nano Research.

[39]  H. Ju,et al.  Cascade signal amplification strategy for subattomolar protein detection by rolling circle amplification and quantum dots tagging. , 2010, Analytical chemistry.

[40]  F. Lemonnier,et al.  HER-2/neu and hTERT Cryptic Epitopes as Novel Targets for Broad Spectrum Tumor Immunotherapy1 , 2002, The Journal of Immunology.

[41]  Tsuyoshi Murata,et al.  {m , 1934, ACML.

[42]  W. Gmeiner,et al.  Nanotechnology for cancer treatment , 2014, Nanotechnology reviews.

[43]  Yang Yang,et al.  Dual-modified liposomes with a two-photon-sensitive cell penetrating peptide and NGR ligand for siRNA targeting delivery. , 2015, Biomaterials.

[44]  S. Prabha,et al.  Glycoengineered mesenchymal stem cells as an enabling platform for two-step targeting of solid tumors. , 2016, Biomaterials.

[45]  Erkki Ruoslahti,et al.  Nanocrystal targeting in vivo , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[46]  R. Reilly,et al.  Dual-Receptor–Targeted Radioimmunotherapy of Human Breast Cancer Xenografts in Athymic Mice Coexpressing HER2 and EGFR Using 177Lu- or 111In-Labeled Bispecific Radioimmunoconjugates , 2016, The Journal of Nuclear Medicine.

[47]  Erik C. Dreaden,et al.  Antiandrogen gold nanoparticles dual-target and overcome treatment resistance in hormone-insensitive prostate cancer cells. , 2012, Bioconjugate chemistry.

[48]  So Jin Lee,et al.  Chemical tumor-targeting of nanoparticles based on metabolic glycoengineering and click chemistry. , 2014, ACS nano.

[49]  Kwangmeyung Kim,et al.  Artificial Chemical Reporter Targeting Strategy Using Bioorthogonal Click Reaction for Improving Active-Targeting Efficiency of Tumor. , 2017, Molecular pharmaceutics.

[50]  Xin-guo Jiang,et al.  Study and evaluation of mechanisms of dual targeting drug delivery system with tumor microenvironment assays compared with normal assays. , 2014, Acta biomaterialia.

[51]  G. Heppner Tumor heterogeneity. , 1984, Cancer research.

[52]  Francesco M Veronese,et al.  PEGylation, successful approach to drug delivery. , 2005, Drug discovery today.

[53]  Jianbin Tang,et al.  A Tumor‐Specific Cascade Amplification Drug Release Nanoparticle for Overcoming Multidrug Resistance in Cancers , 2017, Advanced materials.

[54]  박소현,et al.  물리적(Self-Assembled) 수화젤 , 2006 .

[55]  Shaker A Mousa,et al.  Biosensors: the new wave in cancer diagnosis. , 2010, Nanotechnology, science and applications.

[56]  J. Byrd,et al.  Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas. , 2012, Blood.

[57]  R. Alemany,et al.  iRGD tumor-penetrating peptide-modified oncolytic adenovirus shows enhanced tumor transduction, intratumoral dissemination and antitumor efficacy , 2014, Gene Therapy.

[58]  L. Vermeulen,et al.  Cancer heterogeneity—a multifaceted view , 2013, EMBO reports.

[59]  D. Levy,et al.  STAT3 regulated ARF expression suppresses prostate cancer metastasis , 2015, Nature Communications.

[60]  Yuexian Liu,et al.  Anti-tumor activity of paclitaxel through dual-targeting carrier of cyclic RGD and transferrin conjugated hyperbranched copolymer nanoparticles. , 2012, Biomaterials.

[61]  Angelo Corti,et al.  Tumor vasculature targeting through NGR peptide-based drug delivery systems. , 2011, Current pharmaceutical biotechnology.

[62]  Cuichen Wu,et al.  Engineering a cell-surface aptamer circuit for targeted and amplified photodynamic cancer therapy. , 2013, ACS nano.

[63]  Hansoo Park,et al.  Self-Assembled Nanoconstructs Modified with Amplified Aptamers Inhibited Tumor Growth and Retinal Vascular Hyperpermeability via Vascular Endothelial Growth Factor Capturing. , 2017, Molecular pharmaceutics.

[64]  Meng Yang,et al.  The role of the intravascular microenvironment in spontaneous metastasis development , 2010, International journal of cancer.

[65]  Clemens Decristoforo,et al.  Tumor targeting and imaging with dual-peptide conjugated multifunctional liposomal nanoparticles , 2013, International journal of nanomedicine.

[66]  G. Sauter,et al.  Heterogeneity of amplification of HER2, EGFR, CCND1 and MYC in gastric cancer , 2015, BMC Gastroenterology.

[67]  A. Pini,et al.  Coupling to a cancer-selective heparan-sulfate-targeted branched peptide can by-pass breast cancer cell resistance to methotrexate , 2017, Oncotarget.

[68]  Sai Bi,et al.  DNA Polymerase-Directed Hairpin Assembly for Targeted Drug Delivery and Amplified Biosensing. , 2016, ACS applied materials & interfaces.

[69]  S. Weiss,et al.  Tumour‐targeting bacteria‐based cancer therapies for increased specificity and improved outcome , 2017, Microbial biotechnology.

[70]  Kwangmeyung Kim,et al.  Bioorthogonal copper-free click chemistry in vivo for tumor-targeted delivery of nanoparticles. , 2012, Angewandte Chemie.

[71]  Zhirong Zhang,et al.  Two birds, one stone: dual targeting of the cancer cell surface and subcellular mitochondria by the galectin-3-binding peptide G3-C12 , 2017, Acta Pharmacologica Sinica.

[72]  Qiang Zhang,et al.  The antitumor activity of tumor-homing peptide-modified thermosensitive liposomes containing doxorubicin on MCF-7/ADR: in vitro and in vivo , 2015, International journal of nanomedicine.

[73]  P. Hersey,et al.  Impediments to successful immunotherapy. , 1999, Pharmacology & therapeutics.

[74]  M. E. Kenney,et al.  Dual Receptor-Targeted Theranostic Nanoparticles for Localized Delivery and Activation of Photodynamic Therapy Drug in Glioblastomas. , 2015, Molecular pharmaceutics.

[75]  J. Byrd,et al.  Targeted drug delivery and cross-linking induced apoptosis with anti-CD37 based dual-ligand immunoliposomes in B chronic lymphocytic leukemia cells. , 2013, Biomaterials.

[76]  Feng Li,et al.  Ultrasensitive electrochemical detection of nucleic acid by coupling an autonomous cascade target replication and enzyme/gold nanoparticle-based post-amplification. , 2016, Biosensors & bioelectronics.

[77]  S. Lipkowitz,et al.  Engineered Multivalency Enhances Affibody-Based HER3 Inhibition and Downregulation in Cancer Cells. , 2017, Molecular pharmaceutics.

[78]  Dalong Ni,et al.  Magnesium silicide nanoparticles as a deoxygenation agent for cancer starvation therapy. , 2017, Nature nanotechnology.

[79]  Qingfeng Xiao,et al.  Dual-targeting upconversion nanoprobes across the blood-brain barrier for magnetic resonance/fluorescence imaging of intracranial glioblastoma. , 2014, ACS nano.

[80]  Carolyn R. Bertozzi,et al.  Chemical remodelling of cell surfaces in living animals , 2004, Nature.

[81]  Shijie Cao,et al.  Angiopep-2 and activatable cell-penetrating peptide dual-functionalized nanoparticles for systemic glioma-targeting delivery. , 2014, Molecular pharmaceutics.

[82]  Fabao Gao,et al.  Synergistic dual-ligand doxorubicin liposomes improve targeting and therapeutic efficacy of brain glioma in animals. , 2014, Molecular pharmaceutics.

[83]  C. Harding,et al.  Efficient metabolic engineering of GM3 on tumor cells by N-phenylacetyl-D-mannosamine. , 2006, Biochemistry.

[84]  Ji-Ho Park,et al.  Cooperative nanomaterial system to sensitize, target, and treat tumors , 2009, Proceedings of the National Academy of Sciences.

[85]  D. Curiel,et al.  Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor , 2010, Journal of ovarian research.

[86]  Wang Xing-sheng,et al.  Overview of Prostate-specific Membrane Antigen , 2010 .

[87]  Sai Bi,et al.  Initiator-catalyzed self-assembly of duplex-looped DNA hairpin motif based on strand displacement reaction for logic operations and amplified biosensing. , 2016, Biosensors & bioelectronics.

[88]  B. Cornelissen,et al.  111In-labeled immunoconjugates (ICs) bispecific for the epidermal growth factor receptor (EGFR) and cyclin-dependent kinase inhibitor, p27Kip1. , 2009, Cancer biotherapy & radiopharmaceuticals.

[89]  D. Rossi,et al.  Cell-Penetrating Peptides: From Basic Research to Clinics. , 2017, Trends in pharmacological sciences.

[90]  Kai Jiang,et al.  Shaping bio-inspired nanotechnologies to target thrombosis for dual optical-magnetic resonance imaging. , 2015, Journal of materials chemistry. B.

[91]  W. Biernat,et al.  Tumor Heterogeneity at Protein Level as an Independent Prognostic Factor in Endometrial Cancer1 , 2014, Translational oncology.

[92]  Chunning Yang,et al.  Enhanced tumor-targeting selectivity by modulating bispecific antibody binding affinity and format valence , 2017, Scientific Reports.

[93]  Zhongwu Guo,et al.  Efficient glycoengineering of GM3 on melanoma cell and monoclonal antibody-mediated selective killing of the glycoengineered cancer cell. , 2007, Bioorganic & medicinal chemistry.

[94]  B. Overell,et al.  Fibrin Binding , 2012, Drugs.

[95]  Samantha A. Meenach,et al.  Synthesis and characterization of CREKA-conjugated iron oxide nanoparticles for hyperthermia applications. , 2014, Acta biomaterialia.

[96]  A. Fernández-Medarde,et al.  Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. , 2015, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V.

[97]  Chiara Brignole,et al.  Targeting liposomal chemotherapy via both tumor cell-specific and tumor vasculature-specific ligands potentiates therapeutic efficacy. , 2006, Cancer research.

[98]  R. Majeti,et al.  A bispecific antibody targeting CD47 and CD20 selectively binds and eliminates dual antigen expressing lymphoma cells , 2015, mAbs.

[99]  Nuria Sanvicens,et al.  Multifunctional nanoparticles--properties and prospects for their use in human medicine. , 2008, Trends in biotechnology.

[100]  Dai Fukumura,et al.  Reengineering the Tumor Microenvironment to Alleviate Hypoxia and Overcome Cancer Heterogeneity. , 2016, Cold Spring Harbor perspectives in medicine.

[101]  Sanjeev Palta,et al.  Overview of the coagulation system , 2014, Indian journal of anaesthesia.

[102]  Fulvio Magni,et al.  Enhancement of tumor necrosis factor α antitumor immunotherapeutic properties by targeted delivery to aminopeptidase N (CD13) , 2000, Nature Biotechnology.

[103]  Jin Hai Zheng,et al.  Targeted Cancer Therapy Using Engineered Salmonella typhimurium , 2016, Chonnam medical journal.

[104]  Xuesi Chen,et al.  Selective in vivo metabolic cell-labeling-mediated cancer targeting. , 2017, Nature chemical biology.

[105]  Yu Nie,et al.  Dual-targeted polyplexes: one step towards a synthetic virus for cancer gene therapy. , 2011, Journal of controlled release : official journal of the Controlled Release Society.

[106]  A. Kabakov,et al.  Induction of Hsp70 in tumor cells treated with inhibitors of the Hsp90 activity: A predictive marker and promising target for radiosensitization , 2017, PloS one.

[107]  Michael J Sailor,et al.  Biomimetic amplification of nanoparticle homing to tumors , 2007, Proceedings of the National Academy of Sciences.

[108]  S. Hober,et al.  Engineering of Bispecific Affinity Proteins with High Affinity for ERBB2 and Adaptable Binding to Albumin , 2014, PloS one.

[109]  G. Morelli,et al.  Liposomes derivatized with multimeric copies of KCCYSL peptide as targeting agents for HER-2-overexpressing tumor cells , 2017, International journal of nanomedicine.

[110]  Christopher E. Nelson,et al.  Dual MMP7-Proximity-Activated and Folate Receptor-Targeted Nanoparticles for siRNA Delivery , 2014, Biomacromolecules.

[111]  Robert K Prud'homme,et al.  Multifunctional nanoparticles for imaging, delivery and targeting in cancer therapy , 2009, Expert opinion on drug delivery.

[112]  R. Salgia,et al.  Tumor Heterogeneity and Therapeutic Resistance. , 2016, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.

[113]  T. Iwamoto,et al.  Branched amphiphilic peptide capsules: cellular uptake and retention of encapsulated solutes. , 2014, Biochimica et biophysica acta.

[114]  R. Gomis,et al.  Tumor cell dormancy , 2017, Molecular oncology.

[115]  Ling Ye,et al.  The relative length of dual-target conjugated on iron oxide nanoparticles plays a role in brain glioma targeting , 2017 .

[116]  Chao Zhang,et al.  Endosomal pH-Responsive Polymer-Based Dual-Ligand-Modified Micellar Nanoparticles for Tumor Targeted Delivery and Facilitated Intracellular Release of Paclitaxel , 2015, Pharmaceutical Research.

[117]  Kwangmeyung Kim,et al.  Molecular imaging based on metabolic glycoengineering and bioorthogonal click chemistry. , 2017, Biomaterials.

[118]  Xin-guo Jiang,et al.  RGD and interleukin-13 peptide functionalized nanoparticles for enhanced glioblastoma cells and neovasculature dual targeting delivery and elevated tumor penetration. , 2014, Molecular pharmaceutics.

[119]  Laurie J. Gay,et al.  Contribution of platelets to tumour metastasis , 2011, Nature Reviews Cancer.

[120]  Enze Jiang,et al.  Dormant Cells: The Original Cause of Tumor Recurrence and Metastasis , 2015, Cell Biochemistry and Biophysics.

[121]  B. Felding-Habermann,et al.  Activation of tumor cell integrin αvβ3 controls angiogenesis and metastatic growth in the brain , 2009, Proceedings of the National Academy of Sciences.

[122]  M. Aubert,et al.  Glycoengineering of alphaGal xenoantigen on recombinant peptide bearing the J28 pancreatic oncofetal glycotope. , 2007, Glycobiology.

[123]  Quanyin Hu,et al.  Co-administration of dual-targeting nanoparticles with penetration enhancement peptide for antiglioblastoma therapy. , 2014, Molecular pharmaceutics.

[124]  I. Kwon,et al.  In situ dose amplification by apoptosis-targeted drug delivery. , 2011, Journal of controlled release : official journal of the Controlled Release Society.

[125]  W. Schiemann,et al.  Spatiotemporal Targeting of a Dual-Ligand Nanoparticle to Cancer Metastasis. , 2015, ACS nano.

[126]  J. Tomich,et al.  A review of solute encapsulating nanoparticles used as delivery systems with emphasis on branched amphipathic peptide capsules. , 2016, Archives of biochemistry and biophysics.

[127]  R. Reiter,et al.  Melatonin in regulation of inflammatory pathways in rheumatoid arthritis and osteoarthritis: involvement of circadian clock genes , 2018, British journal of pharmacology.

[128]  Neetu Singh,et al.  Nanoparticles that communicate in vivo to amplify tumour targeting. , 2011, Nature materials.

[129]  Ali Jahanban-Esfahlan,et al.  Combination of nanotechnology with vascular targeting agents for effective cancer therapy , 2018, Journal of cellular physiology.

[130]  Yu Cheng,et al.  Fibrin-binding, peptide amphiphile micelles for targeting glioblastoma☆ , 2013, Biomaterials.

[131]  S. Menichetti,et al.  Design and In vitro Evaluation of Branched Peptide Conjugates: Turning Nonspecific Cytotoxic Drugs into Tumor‐Selective Agents , 2010, ChemMedChem.

[132]  John-William Sidhom,et al.  Dual Targeting Nanoparticle Stimulates the Immune System To Inhibit Tumor Growth. , 2017, ACS Nano.

[133]  H. Koeppen,et al.  Identification and immunotherapeutic targeting of antigens induced by chemotherapy , 2006, Nature Biotechnology.

[134]  X. Luo,et al.  Tumor amplified protein expression therapy: Salmonella as a tumor-selective protein delivery vector. , 2001, Oncology research.

[135]  Chad A Mirkin,et al.  Aptamer nano-flares for molecular detection in living cells. , 2009, Nano letters.

[136]  J. Park,et al.  Hypoxia-responsive nanocarriers for cancer imaging and therapy: recent approaches and future perspectives. , 2016, Chemical communications.

[137]  R. Szoszkiewicz,et al.  Gene delivery and immunomodulatory effects of plasmid DNA associated with Branched Amphiphilic Peptide Capsules. , 2016, Journal of controlled release : official journal of the Controlled Release Society.

[138]  D. Hallahan,et al.  Tumor-targeted delivery of liposome-encapsulated doxorubicin by use of a peptide that selectively binds to irradiated tumors. , 2011, Journal of controlled release : official journal of the Controlled Release Society.

[139]  J. Winer,et al.  Synergistic Induction of Tumor Antigens by Wnt-1 Signaling and Retinoic Acid Revealed by Gene Expression Profiling* , 2002, The Journal of Biological Chemistry.

[140]  Jiehua Zhou,et al.  Aptamers as targeted therapeutics: current potential and challenges , 2017, Nature Reviews Drug Discovery.

[141]  Yanglong Hou,et al.  Multifunctional Nanoparticles for Multimodal Molecular Imaging , 2011 .

[142]  Wei Shi,et al.  Fibrin-targeting peptide CREKA-conjugated multi-walled carbon nanotubes for self-amplified photothermal therapy of tumor. , 2016, Biomaterials.

[143]  T. Efferth,et al.  Tumor Heterogeneity, Single-Cell Sequencing, and Drug Resistance , 2016, Pharmaceuticals.

[144]  Cheever,et al.  Human HER‐2/neu protein immunization circumvents tolerance to rat neu: a vaccine strategy for ‘self’ tumour antigens , 1998, Immunology.

[145]  E. Chavakis,et al.  Novel aspects in the regulation of the leukocyte adhesion cascade , 2009, Thrombosis and Haemostasis.

[146]  F. Veronese,et al.  The Impact of PEGylation on Biological Therapies , 2012, BioDrugs.

[147]  F. Doyle,et al.  Quantity and accessibility for specific targeting of receptors in tumours , 2014, Scientific Reports.

[148]  D. Artemov,et al.  Bioorthogonal two-component drug delivery in HER2(+) breast cancer mouse models , 2014, Scientific Reports.

[149]  A. Tutt,et al.  Selectin ligand sialyl-Lewis x antigen drives metastasis of hormone-dependent breast cancers. , 2011, Cancer research.

[150]  E. Zhukovsky,et al.  Trispecific antibodies for CD16A-directed NK cell engagement and dual-targeting of tumor cells , 2017, Protein engineering, design & selection : PEDS.

[151]  Ülo Langel,et al.  Design of a Tumor-Homing Cell-Penetrating Peptide , 2008 .

[152]  N. Zarghami,et al.  Tumor vascular infarction: prospects and challenges , 2017, International Journal of Hematology.

[153]  H. Levitsky,et al.  Reciprocal Changes in Tumor Antigenicity and Antigen-specific T Cell Function during Tumor Progression , 2004, The Journal of experimental medicine.

[154]  K. Brindle,et al.  Imaging sialylated tumor cell glycans in vivo , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[155]  X. Luo,et al.  Salmonella-based tumor-targeted cancer therapy: tumor amplified protein expression therapy (TAPET) for diagnostic imaging. , 2001, Journal of controlled release : official journal of the Controlled Release Society.

[156]  Leaf Huang,et al.  Nanoparticles targeted with NGR motif deliver c-myc siRNA and doxorubicin for anticancer therapy. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[157]  Davoud Farajzadeh,et al.  RGD delivery of truncated coagulase to tumor vasculature affords local thrombotic activity to induce infarction of tumors in mice , 2017, Scientific Reports.

[158]  A.V. Lakhin,et al.  Aptamers: Problems, Solutions and Prospects , 2013, Acta naturae.

[159]  M. de la Guardia,et al.  Modulating tumor hypoxia by nanomedicine for effective cancer therapy , 2018, Journal of cellular physiology.

[160]  Yuzhong Zhang,et al.  Ultrasensitive Multiplexed Immunoassay for Tumor Biomarkers Based on DNA Hybridization Chain Reaction Amplifying Signal. , 2016, ACS applied materials & interfaces.

[161]  M. Belting Glycosaminoglycans in cancer treatment. , 2014, Thrombosis research.

[162]  M. Rescigno,et al.  Salmonella engineered to express CD20-targeting antibodies and a drug-converting enzyme can eradicate human lymphomas. , 2013, Blood.

[163]  S. Lai,et al.  Pretargeting with bispecific fusion proteins facilitates delivery of nanoparticles to tumor cells with distinct surface antigens , 2017, Journal of controlled release : official journal of the Controlled Release Society.

[164]  S. Dave,et al.  HER2 overexpression elicits a proinflammatory IL-6 autocrine signaling loop that is critical for tumorigenesis. , 2011, Cancer research.

[165]  A. Üren,et al.  YK-4-279 effectively antagonizes EWS-FLI1 induced leukemia in a transgenic mouse model , 2015, Oncotarget.

[166]  A. Pini,et al.  Target‐Selective Drug Delivery through Liposomes Labeled with Oligobranched Neurotensin Peptides , 2011, ChemMedChem.

[167]  J. Penelle,et al.  Multivalent cationic pseudopeptide polyplexes as a tool for cancer therapy , 2017, Oncotarget.

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

[169]  H. Ghandehari,et al.  Synergistic enhancement of cancer therapy using a combination of heat shock protein targeted HPMA copolymer-drug conjugates and gold nanorod induced hyperthermia. , 2013, Journal of controlled release : official journal of the Controlled Release Society.

[170]  M. Walter,et al.  Upregulation of Key Molecules for Targeted Imaging and Therapy , 2016, The Journal of Nuclear Medicine.

[171]  Erkki Ruoslahti,et al.  Targeting of albumin-embedded paclitaxel nanoparticles to tumors. , 2009, Nanomedicine : nanotechnology, biology, and medicine.

[172]  Erkki Ruoslahti,et al.  De novo design of a tumor-penetrating peptide. , 2013, Cancer research.

[173]  Mie Kristensen,et al.  Applications and Challenges for Use of Cell-Penetrating Peptides as Delivery Vectors for Peptide and Protein Cargos , 2016, International journal of molecular sciences.