Skip to main content

Recent Advances in Tumor Targeting Approaches

  • Chapter
  • First Online:
Targeted Drug Delivery : Concepts and Design

Abstract

Tremendous technological advancement has been observed in the past few decades to combat ever increasing mortality rate in cancer therapy. It is now widely accepted that this reduction in mortality can be addressed effectively by materializing the concept of magic bullet, i.e., localizing the drug in concern at the site of action, thereby sparing normal tissues from unwanted toxicities and drastically improving the therapeutic efficacy. The present chapter inculcates such recent advances in the tumor targeting approaches. Basics of the peculiar tumor microenvironment and its discrimination from normal tissues have been primarily covered in brief to provide a background for the better understanding of the design and development of molecular and physicochemical targets. Concomitantly, barriers offered by altered tumor microenvironment to the drug delivery of anticancer drugs have been covered. The subsequent sections covers the conventional strategies for tumor targeting which essentially comprises passive targeting, active targeting, and physical targeting followed by recent advances in the tumor targeting approaches from clinical perspectives. These include deeper insights on molecular targeted therapies, tumor angiogenesis , cancer immunotherapy, and drug delivery of multiple drug resistance tumors. In a nutshell, with the advent of the molecularly targeted therapies, targeting tumor specific surface antigens and intracellular processes and components is a rapidly shifting paradigm of cancer therapy and unexceptional results have been observed till date by appropriately blending it with nanotechnology based approaches.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

17 AAG:

17-Allylamino-17-Demethoxygeldanamycin

17-DMAG:

17-dimethylaminoethylamine-17-demethoxy-geldanamycin

ADCC:

Antibody dependent cellular cytotoxicity

ADEPT :

Antibody directed enzyme prodrug therapy

ANA:

Monoclonal antinuclear autoantibody

ASM:

Acid sphingomyelinase

ATP:

Adenosine triphosphate

BCS:

Biopharmaceutical classification system

BET:

Bromodomain and extra-terminal

BP:

Binding protein

BRCA:

Breast cancer gene

CDC:

Complement-activation dependent cytotoxicity

CDKs:

Cyclin dependent kinases

CNS:

Central nervous system

CTLA:

Cytotoxic T-lymphocyte antigen

dgRTA:

Deglycosylated ricin A chain

DMXAA:

5,6-Dimethylxanthenone-4-acetic acid

DNA:

Deoxy ribose nucleic acid

DOTA:

d-Tyr-d-Lys(HSG)-d-Glu-d-Lys(HSG)-NH2

EBRT:

External-beam radiation therapy

ECM:

Extracellular matrix

EPCs:

Endothelial precursor cells

EZH2:

Enhancer of Zeste homolog 2

FAK:

Focal adhesion kinase

FDG:

2-Deoxy-2-(18F) fluoro-d-glucose

GMCSF:

Granulocyte-macrophage colony-stimulating factor

GRP78:

78 kDa glucose-regulated protein

H3K4me3/2:

Trimethylation and dimethylation of histone H3 at lysine 4

HAMA:

Human anti-mouse antibody

HARA:

Human anti-ricin antibody

hCG:

Human chorionic gonadotrophin

HDAC:

Histone deacetylase

HER2:

Human epidermal growth factor receptor 2

HIF:

Hypoxia inducible factor

HOP:

HSP90 organizing protein

HPMA:

N-(2-hydroxy propyl) methacrylamide

HSP:

Heat shock proteins

IFP:

Interstitial fluid pressure

IL:

Interleukin

IMP-288:

1,4,7,10-Tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid

JAG1:

Jagged 1 protein

LECs:

Lymphatic endothelial cells

LLC:

Lewis lung carcinoma

LTTs:

Ligand-targeted therapeutics

mAbs:

Monoclonal antibody

MAPK:

Mitogen-activated protein kinase

MCT:

Monocarboxylate transporters

MMPs:

Matrix metalloproteinases

MPS:

Macrophagocytosis systems

mTOR:

Mechanistic target of rapamycin

Myc:

Myelocytomatosis oncogene

p14ARF :

Alternate reading frame

PI-3:

Phosphoinositide 3-kinase

PDEPT:

Polymer directed enzyme prodrug therapy

PE:

Phosphatidyl ethanolamine

PKB:

Protein kinase B

PSMC2:

26S protease regulatory subunit 7 gene

pRb:

Retinoblastoma tumor suppressor protein

RAIT:

Radioimmunotherapy

Rb:

Retinoblastoma

RBC:

Red blood cell

RES:

Reticuloendothelial system

SAR:

Structure–activity relationship

STA:

3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole

TCMC:

2-(4-isothiocyanotobenzyl)-1, 4, 7, 10-tetraaza-1, 4, 7, 10-tetra-(2-carbamonyl methyl)-cyclododecane

TNF:

Tumor necrosis factors

TP53/p53:

Tumor protein P53

VEGF :

Vascular endothelial growth factor

vSMCs:

Vascular smooth muscle cells

WHO:

World Health Organization

References

  1. International Agency for Research on Cancer (IARC), World Heatlth Organization (WHO) (2012)Cancer fact sheets. In: Globocan 2012: estimated cancer incidence, mortalitity and prevalence worldwide in 2012. International Agency for Research on Cancer (IARC), World Heatlth Organization (WHO), Lyon, France

    Google Scholar 

  2. Lippman SM, Hong WK (2002) Cancer prevention science and practice. Cancer Res 62:5119–5125, PMID: 12234971

    CAS  PubMed  Google Scholar 

  3. Ghose T (2002) The current status of tumor targeting. In: Page M (ed) Tumor targeting in cancer therapy. Springer Science, New York, NY, pp 3–78

    Google Scholar 

  4. Poste G, Kirsh R (1983) Site–specific (targeted) drug delivery in cancer therapy. Nat Biotechnol 1:869–878. doi:10.1038/nbt1283-869

    CAS  Google Scholar 

  5. Parveen S, Sahoo SK (2008) Polymeric nanoparticles for cancer therapy. J Drug Target 16:108–123. doi:10.1080/10611860701794353

    CAS  PubMed  Google Scholar 

  6. Dewhirst MW, Kimura H, Rehmus SW, Braun RD, Papahadjopoulos D, Hong K, Secomb TW (1996) Microvascular studies on the origins of perfusion-limited hypoxia. Br J Cancer 27(Suppl):S247–S251, PMID 8763890

    CAS  Google Scholar 

  7. Cairns R, Papandreou I, Denko N (2006) Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment. Mol Cancer Res 4:61–70. doi:10.1158/1541-7786.MCR-06-0002

    CAS  PubMed  Google Scholar 

  8. Heldin CH, Rubin K, Pietras K, Ostman A (2004) High interstitial fluid pressure – an obstacle in cancer therapy. Nat Rev Cancer 4:806–813. doi:10.1038/nrc1456

    CAS  PubMed  Google Scholar 

  9. van den Berg AP, Wike-Hooley JL, van den Berg-Blok AE, van der Zee J, Reinhold HS (1982) Tumour pH in human mammary carcinoma. Eur J Cancer Clin Oncol 18:457–462. doi:10.1016/0277-5379(82)90114-6

    PubMed  Google Scholar 

  10. Tannock IF, Rotin D (1989) Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res 49:4373–4384, PMID: 2545340

    CAS  PubMed  Google Scholar 

  11. Weinhouse S (1976) The Warburg hypothesis fifty years later. Z Krebsforsch Klin Onkol Cancer Res Clin Oncol 87:115–126, PMID: 136820

    CAS  PubMed  Google Scholar 

  12. Yamagata M, Hasuda K, Stamato T, Tannock IF (1998) The contribution of lactic acid to acidification of tumours: studies of variant cells lacking lactate dehydrogenase. Br J Cancer 77:1726–1731. doi:10.1038/bjc.1998.289

    CAS  PubMed Central  PubMed  Google Scholar 

  13. Izumi H, Torigoe T, Ishiguchi H, Uramoto H, Yoshida Y, Tanabe M, Ise T, Murakami T, Yoshida T, Nomoto M, Kohno K (2003) Cellular pH regulators: potentially promising molecular targets for cancer chemotherapy. Cancer Treat Rev 29:541–549. doi:10.1016/S0305-7372(03)00106-3

    CAS  PubMed  Google Scholar 

  14. Griffiths JR, McIntyre DJ, Howe FA, Stubbs M (2001) Why are cancers acidic? A carrier-mediated diffusion model for H + transport in the interstitial fluid. Novartis Found Symp 240:46–62, discussion 62–47, 152–153; PMID: 11727936

    CAS  PubMed  Google Scholar 

  15. Narang AS, Varia S (2011) Role of tumor vascular architecture in drug delivery. Adv Drug Deliv Rev 63:640–658. doi:10.1016/j.addr.2011.04.002

    CAS  PubMed  Google Scholar 

  16. Thanki K, Gangwal RP, Sangamwar AT, Jain S (2013) Oral delivery of anticancer drugs: challenges and opportunities. J Control Release 170:15–40. doi:10.1016/j.jconrel.2013.04.020

    CAS  PubMed  Google Scholar 

  17. Weissig V, D’Souza GG (2010) Organelle-specific pharmaceutical nanotechnology. Wiley, Hoboken, NJ

    Google Scholar 

  18. Matsumura Y, Maeda H (1986) A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 46:6387–6392, PMID: 2946403

    CAS  PubMed  Google Scholar 

  19. Hobbs SK, Monsky WL, Yuan F, Roberts WG, Griffith L, Torchilin VP, Jain RK (1998) Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci U S A 95:4607–4612, PMID: 9539785

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Padera TP, Stoll BR, Tooredman JB, Capen D, di Tomaso E, Jain RK (2004) Pathology: cancer cells compress intratumour vessels. Nature 427:695. doi:10.1038/427695a

    CAS  PubMed  Google Scholar 

  21. Rabanel JM, Aoun V, Elkin I, Mokhtar M, Hildgen P (2012) Drug-loaded nanocarriers: passive targeting and crossing of biological barriers. Curr Med Chem 19:3070–3102. doi:10.2174/092986712800784702

    CAS  PubMed  Google Scholar 

  22. Nag OK, Awasthi V (2013) Surface engineering of liposomes for stealth behavior. Pharmaceutics 5:542–569. doi:10.3390/pharmaceutics5040542

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Salmaso S, Caliceti P (2013) Stealth properties to improve therapeutic efficacy of drug nanocarriers. J Drug Deliv 2013:374252. doi:10.1155/2013/374252

    PubMed Central  PubMed  Google Scholar 

  24. Allen TM, Hansen C, Rutledge J (1989) Liposomes with prolonged circulation times: factors affecting uptake by reticuloendothelial and other tissues. Biochim Biophys Acta 981:27–35. doi:10.1016/0005-2736(89)90078-3

    CAS  PubMed  Google Scholar 

  25. Gabizon A, Papahadjopoulos D (1988) Liposome formulations with prolonged circulation time in blood and enhanced uptake by tumors. Proc Natl Acad Sci USA 85:6949–6953, PMID: 3413128

    CAS  PubMed Central  PubMed  Google Scholar 

  26. Immordino ML, Dosio F, Cattel L (2006) Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine 1:297–315, PMID: 17717971

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Tirosh O, Barenholz Y, Katzhendler J, Priev A (1998) Hydration of polyethylene glycol-grafted liposomes. Biophys J 74:1371–1379. doi:10.1016/S0006-3495(98)77849-X

    CAS  PubMed Central  PubMed  Google Scholar 

  28. Lehtonen JY, Kinnunen PK (1995) Poly(ethylene glycol)-induced and temperature-dependent phase separation in fluid binary phospholipid membranes. Biophys J 68:525–535. doi:10.1016/S0006-3495(95)80214-6

    CAS  PubMed Central  PubMed  Google Scholar 

  29. Szebeni J (2005) Complement activation-related pseudoallergy: a new class of drug-induced acute immune toxicity. Toxicology 216:106–121. doi:10.1016/j.tox.2005.07.023

    CAS  PubMed  Google Scholar 

  30. Yang Q, Ma Y, Zhao Y, She Z, Wang L, Li J, Wang C, Deng Y (2013) Accelerated drug release and clearance of PEGylated epirubicin liposomes following repeated injections: a new challenge for sequential low-dose chemotherapy. Int J Nanomedicine 8:1257–1268. doi:10.2147/IJN.S41701

    PubMed Central  PubMed  Google Scholar 

  31. Herold DA, Keil K, Bruns DE (1989) Oxidation of polyethylene glycols by alcohol dehydrogenase. Biochem Pharmacol 38:73–76. doi:10.1016/0006-2952(89)90151-2

    CAS  PubMed  Google Scholar 

  32. Veronese FM, Pasut G (2005) PEGylation, successful approach to drug delivery. Drug Discov Today 10:1451–1458. doi:10.1016/S1359-6446(05)03575-0

    CAS  PubMed  Google Scholar 

  33. Allen TM (2002) Ligand-targeted therapeutics in anticancer therapy. Nat Rev Cancer 2:750–763. doi:10.1038/nrc903

    CAS  PubMed  Google Scholar 

  34. Gradishar WJ (2006) Albumin-bound paclitaxel: a next-generation taxane. Expert Opin Pharmacother 7:1041–1053. doi:10.1517/14656566.7.8.1041

    CAS  PubMed  Google Scholar 

  35. John TA, Vogel SM, Tiruppathi C, Malik AB, Minshall RD (2003) Quantitative analysis of albumin uptake and transport in the rat microvessel endothelial monolayer. Am J Physiol Lung Cell Mol Physiol 284:L187–196. doi:10.1152/ajplung.00152.2002

    CAS  PubMed  Google Scholar 

  36. Minshall RD, Tiruppathi C, Vogel SM, Malik AB (2002) Vesicle formation and trafficking in endothelial cells and regulation of endothelial barrier function. Histochem Cell Biol 117:105–112. doi:10.1007/s00418-001-0367-x

    CAS  PubMed  Google Scholar 

  37. Kumagai AK, Eisenberg JB, Pardridge WM (1987) Absorptive-mediated endocytosis of cationized albumin and a beta-endorphin-cationized albumin chimeric peptide by isolated brain capillaries. Model system of blood-brain barrier transport. J Biol Chem 262:15214–15219, PMID: 2959663

    CAS  PubMed  Google Scholar 

  38. Desai N, Trieu V, Yao Z, Louie L, Ci S, Yang A, Tao C, De T, Beals B, Dykes D, Noker P, Yao R, Labao E, Hawkins M, Soon-Shiong P (2006) Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res 12:1317–1324. doi:10.1158/1078-0432.CCR-05-1634

    CAS  PubMed  Google Scholar 

  39. Das M, Mohanty C, Sahoo SK (2009) Ligand-based targeted therapy for cancer tissue. Expert Opin Drug Deliv 6:285–304. doi:10.1517/17425240902780166

    CAS  PubMed  Google Scholar 

  40. Vasir JK, Labhasetwar V (2005) Targeted drug delivery in cancer therapy. Technol Cancer Res Treat 4:363–374, PMID: 16029056

    CAS  PubMed  Google Scholar 

  41. Jain S, Mathur R, Das M, Swarnakar NK, Mishra AK (2011) Synthesis, pharmacoscintigraphic evaluation and antitumor efficacy of methotrexate-loaded, folate-conjugated, stealth albumin nanoparticles. Nanomedicine (Lond) 6:1733–1754. doi:10.2217/nnm.11.53

    CAS  Google Scholar 

  42. Das M, Bandyopadhyay D, Mishra D, Datir S, Dhak P, Jain S, Maiti TK, Basak A, Pramanik P (2011) “Clickable”, trifunctional magnetite nanoparticles and their chemoselective biofunctionalization. Bioconjug Chem 22:1181–1193. doi:10.1021/bc2000484

    CAS  PubMed  Google Scholar 

  43. Das M, Datir SR, Singh RP, Jain S (2013) Augmented anticancer activity of a targeted, intracellularly activatable, theranostic nanomedicine based on fluorescent and radiolabeled, methotrexate-folic acid-multiwalled carbon nanotube conjugate. Mol Pharm 10:2543–2557. doi:10.1021/mp300701e

    CAS  PubMed  Google Scholar 

  44. Lu YJ, Wei KC, Ma CC, Yang SY, Chen JP (2012) Dual targeted delivery of doxorubicin to cancer cells using folate-conjugated magnetic multi-walled carbon nanotubes. Colloids Surf B: Biointerfaces 89:1–9. doi:10.1016/j.colsurfb.2011.08.001

    CAS  PubMed  Google Scholar 

  45. Li L, Yang Q, Zhou Z, Zhong J, Huang Y (2014) Doxorubicin-loaded, charge reversible, folate modified HPMA copolymer conjugates for active cancer cell targeting. Biomaterials 35:5171–5187. doi:10.1016/j.biomaterials.2014.03.027

    CAS  PubMed  Google Scholar 

  46. Corbin IR, Ng KK, Ding L, Jurisicova A, Zheng G (2013) Near-infrared fluorescent imaging of metastatic ovarian cancer using folate receptor-targeted high-density lipoprotein nanocarriers. Nanomedicine 8:875–890. doi:10.2217/nnm.12.137

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Hong G, Yuan R, Liang B, Shen J, Yang X, Shuai X (2008) Folate-functionalized polymeric micelle as hepatic carcinoma-targeted, MRI-ultrasensitive delivery system of antitumor drugs. Biomed Microdevices 10:693–700. doi:10.1007/s10544-008-9180-9

    CAS  PubMed  Google Scholar 

  48. Tyagi N, Ghosh PC (2011) Folate receptor mediated targeted delivery of ricin entrapped into sterically stabilized liposomes to human epidermoid carcinoma (KB) cells: effect of monensin intercalated into folate-tagged liposomes. Eur J Pharm Sci 43:343–353. doi:10.1016/j.ejps.2011.05.010

    CAS  PubMed  Google Scholar 

  49. Lale SV, GA R, Aravind A, Kumar DS, Koul V (2014) AS1411 Aptamer and folic acid functionalized pH-responsive ATRP fabricated pPEGMA-PCL-pPEGMA polymeric nanoparticles for targeted drug delivery in cancer therapy. Biomacromolecules 15:1737–1752. doi:10.1021/bm5001263

    CAS  PubMed  Google Scholar 

  50. El-Gogary RI, Rubio N, Wang JT, Al-Jamal WT, Bourgognon M, Kafa H, Naeem M, Klippstein R, Abbate V, Leroux F, Bals S, Van Tendeloo G, Kamel AO, Awad GA, Mortada ND, Al-Jamal KT (2014) Polyethylene glycol conjugated polymeric nanocapsules for targeted delivery of quercetin to folate-expressing cancer cells in vitro and in vivo. ACS Nano 8:1384–1401. doi:10.1021/nn405155b

    CAS  PubMed  Google Scholar 

  51. Tavassolian F, Kamalinia G, Rouhani H, Amini M, Ostad SN, Khoshayand MR, Atyabi F, Tehrani MR, Dinarvand R (2014) Targeted poly (L-gamma-glutamyl glutamine) nanoparticles of docetaxel against folate over-expressed breast cancer cells. Int J Pharm 467:123–138. doi:10.1016/j.ijpharm.2014.03.033

    CAS  PubMed  Google Scholar 

  52. Nair BP, Vaikkath D, Nair PD (2014) Polyhedral oligomeric silsesquioxane-F68 hybrid vesicles for folate receptor targeted anti-cancer drug delivery. Langmuir 30:340–347. doi:10.1021/la4036997

    CAS  PubMed  Google Scholar 

  53. Chen YC, Chiang CF, Chen LF, Liang PC, Hsieh WY, Lin WL (2014) Polymersomes conjugated with des-octanoyl ghrelin and folate as a BBB-penetrating cancer cell-targeting delivery system. Biomaterials 35:4066–4081. doi:10.1016/j.biomaterials.2014.01.042

    CAS  PubMed  Google Scholar 

  54. Dowlati A, Loo M, Bury T, Fillet G, Beguin Y (1997) Soluble and cell-associated transferrin receptor in lung cancer. Br J Cancer 75:1802–1806. doi:10.1038/bjc.1997.307

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Gosk S, Vermehren C, Storm G, Moos T (2004) Targeting anti-transferrin receptor antibody (OX26) and OX26-conjugated liposomes to brain capillary endothelial cells using in situ perfusion. J Cereb Blood Flow Metab 24:1193–1204. doi:10.1097/01.WCB.0000135592.28823.47

    CAS  PubMed  Google Scholar 

  56. Sahoo SK, Labhasetwar V (2005) Enhanced antiproliferative activity of transferrin-conjugated paclitaxel-loaded nanoparticles is mediated via sustained intracellular drug retention. Mol Pharm 2:373–383. doi:10.1021/mp050032z

    CAS  PubMed  Google Scholar 

  57. Sahoo SK, Ma W, Labhasetwar V (2004) Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer. Int J Cancer 112:335–340. doi:10.1002/ijc.20405

    CAS  PubMed  Google Scholar 

  58. Ferris DP, Lu J, Gothard C, Yanes R, Thomas CR, Olsen JC, Stoddart JF, Tamanoi F, Zink JI (2011) Synthesis of biomolecule-modified mesoporous silica nanoparticles for targeted hydrophobic drug delivery to cancer cells. Small 7:1816–1826. doi:10.1002/smll.201002300

    CAS  PubMed Central  PubMed  Google Scholar 

  59. Kim TH, Jo YG, Jiang HH, Lim SM, Youn YS, Lee S, Chen X, Byun Y, Lee KC (2012) PEG-transferrin conjugated TRAIL (TNF-related apoptosis-inducing ligand) for therapeutic tumor targeting. J Control Release 162:422–428. doi:10.1016/j.jconrel.2012.07.021

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Gaspar MM, Radomska A, Gobbo OL, Bakowsky U, Radomski MW, Ehrhardt C (2012) Targeted delivery of transferrin-conjugated liposomes to an orthotopic model of lung cancer in nude rats. J Aerosol Med Pulm Drug Deliv 25:310–318. doi:10.1089/jamp.2011.0928

    CAS  PubMed  Google Scholar 

  61. Liu G, Shen H, Mao J, Zhang L, Jiang Z, Sun T, Lan Q, Zhang Z (2013) Transferrin modified graphene oxide for glioma-targeted drug delivery: in vitro and in vivo evaluations. ACS Appl Mater Interfaces 5:6909–6914. doi:10.1021/am402128s

    CAS  PubMed  Google Scholar 

  62. Neves S, Faneca H, Bertin S, Konopka K, Duzgunes N, Pierrefite-Carle V, Simoes S, Pedroso de Lima MC (2009) Transferrin lipoplex-mediated suicide gene therapy of oral squamous cell carcinoma in an immunocompetent murine model and mechanisms involved in the antitumoral response. Cancer Gene Ther 16:91–101. doi:10.1038/cgt.2008.60

    CAS  PubMed  Google Scholar 

  63. Vaidya B, Vyas SP (2012) Transferrin coupled vesicular system for intracellular drug delivery for the treatment of cancer: development and characterization. J Drug Target 20:372–380. doi:10.3109/1061186X.2012.662687

    CAS  PubMed  Google Scholar 

  64. Abouzeid AH, Patel NR, Sarisozen C, Torchilin VP (2014) Transferrin-targeted polymeric micelles co-loaded with curcumin and paclitaxel: efficient killing of paclitaxel-resistant cancer cells. Pharm Res 1-8. doi: 10.1007/s11095-013-1295-x

  65. Yhee JY, Lee SJ, Lee S, Song S, Min HS, Kang SW, Son S, Jeong SY, Kwon IC, Kim SH, Kim K (2013) Tumor-targeting transferrin nanoparticles for systemic polymerized siRNA delivery in tumor-bearing mice. Bioconjug Chem 24:1850–1860. doi:10.1021/bc400226b

    CAS  PubMed  Google Scholar 

  66. Huang Y, He L, Liu W, Fan C, Zheng W, Wong Y-S, Chen T (2013) Selective cellular uptake and induction of apoptosis of cancer-targeted selenium nanoparticles. Biomaterials 34:7106–7116. doi:10.1016/j.biomaterials.2013.04.067

    CAS  PubMed  Google Scholar 

  67. Bies C, Lehr CM, Woodley JF (2004) Lectin-mediated drug targeting: history and applications. Adv Drug Deliv Rev 56:425–435. doi:10.1016/j.addr.2003.10.030

    CAS  PubMed  Google Scholar 

  68. Baenziger JU, Maynard Y (1980) Human hepatic lectin. Physiochemical properties and specificity. J Biol Chem 255:4607–4613, PMID: 7372599

    CAS  PubMed  Google Scholar 

  69. Duncan R (2003) The dawning era of polymer therapeutics. Nat Rev Drug Discov 2:347–360. doi:10.1038/nrd1088

    CAS  PubMed  Google Scholar 

  70. Yu HY, Lin JH (2000) Intracellular delivery of membrane-impermeable hydrophilic molecules to a hepatoblastoma cell line by asialoglycoprotein-labeled liposomes. J Formos Med Assoc 99:936–941, PMID: 11155748

    CAS  PubMed  Google Scholar 

  71. Dubey PK, Mishra V, Jain S, Mahor S, Vyas SP (2004) Liposomes modified with cyclic RGD peptide for tumor targeting. J Drug Target 12:257–264. doi:10.1080/10611860410001728040

    CAS  PubMed  Google Scholar 

  72. Aina OH, Sroka TC, Chen ML, Lam KS (2002) Therapeutic cancer targeting peptides. Biopolymers 66:184–199. doi:10.1002/bip.10257

    CAS  PubMed  Google Scholar 

  73. Torchilin VP (2000) Drug targeting. Eur J Pharm Sci 11(Suppl 2):S81–91. doi:10.1016/S0928-0987(00)00166-4

    CAS  PubMed  Google Scholar 

  74. Husseini GA, Diaz de la Rosa MA, Gabuji T, Zeng Y, Christensen DA, Pitt WG (2007) Release of doxorubicin from unstabilized and stabilized micelles under the action of ultrasound. J Nanosci Nanotechnol 7:1028–1033. doi:10.1166/jnn.2007.218

    CAS  PubMed Central  PubMed  Google Scholar 

  75. Knights CD, Pestell RG (2008) The cell cycle: therapeutic targeting of cell cycle regulatory components and effector pathways in cancer. In: Kaufman HL, Wadler S, Antman K (eds) Molecular targeting in oncology. Humana, Totowa, NJ

    Google Scholar 

  76. Liu MC, Marshall JL, Pestell RG (2004) Novel strategies in cancer therapeutics: targeting enzymes involved in cell cycle regulation and cellular proliferation. Curr Cancer Drug Targets 4:403–424. doi:10.2174/1568009043332907

    CAS  PubMed  Google Scholar 

  77. Chin K, de Solorzano CO, Knowles D, Jones A, Chou W, Rodriguez EG, Kuo WL, Ljung BM, Chew K, Myambo K, Miranda M, Krig S, Garbe J, Stampfer M, Yaswen P, Gray JW, Lockett SJ (2004) In situ analyses of genome instability in breast cancer. Nat Genet 36:984–988. doi:10.1038/ng1409

    CAS  PubMed  Google Scholar 

  78. Deshpande A, Sicinski P, Hinds PW (2005) Cyclins and cdks in development and cancer: a perspective. Oncogene 24:2909–2915. doi:10.1038/sj.onc.1208618

    CAS  PubMed  Google Scholar 

  79. Hedenfalk I, Duggan D, Chen Y, Radmacher M, Bittner M, Simon R, Meltzer P, Gusterson B, Esteller M, Kallioniemi OP, Wilfond B, Borg A, Trent J, Raffeld M, Yakhini Z, Ben-Dor A, Dougherty E, Kononen J, Bubendorf L, Fehrle W, Pittaluga S, Gruvberger S, Loman N, Johannsson O, Olsson H, Sauter G (2001) Gene-expression profiles in hereditary breast cancer. N Engl J Med 344:539–548. doi:10.1056/NEJM200102223440801

    CAS  PubMed  Google Scholar 

  80. Dobbelstein M, Moll U (2014) Targeting tumour-supportive cellular machineries in anticancer drug development. Nat Rev Drug Discov 13:179–196. doi:10.1038/nrd4201

    CAS  PubMed  Google Scholar 

  81. Armstrong GT, Liu W, Leisenring W, Yasui Y, Hammond S, Bhatia S, Neglia JP, Stovall M, Srivastava D, Robison LL (2011) Occurrence of multiple subsequent neoplasms in long-term survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol 29:3056–3064. doi:10.1200/JCO.2011.34.6585

    PubMed Central  PubMed  Google Scholar 

  82. Travis LB, Fossa SD, Schonfeld SJ, McMaster ML, Lynch CF, Storm H, Hall P, Holowaty E, Andersen A, Pukkala E, Andersson M, Kaijser M, Gospodarowicz M, Joensuu T, Cohen RJ, Boice JD Jr, Dores GM, Gilbert ES (2005) Second cancers among 40,576 testicular cancer patients: focus on long-term survivors. J Natl Cancer Inst 97:1354–1365. doi:10.1093/jnci/dji278

    PubMed  Google Scholar 

  83. De Koning P, Neijt JP, Jennekens FG, Gispen WH (1987) Evaluation of cis-diamminedichloroplatinum (II) (cisplatin) neurotoxicity in rats. Toxicol Appl Pharmacol 89:81–87. doi:10.1016/0041-008X(87)90178-5

    PubMed  Google Scholar 

  84. Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, Ramalingam SS, Doetsch PW (2013) Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One 8:e81162. doi:10.1371/journal.pone.0081162

    PubMed Central  PubMed  Google Scholar 

  85. Luo J, Solimini NL, Elledge SJ (2009) Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136:823–837. doi:10.1016/j.cell.2009.02.024

    CAS  PubMed Central  PubMed  Google Scholar 

  86. Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, Edkins S, O’Meara S, Vastrik I, Schmidt EE, Avis T, Barthorpe S, Bhamra G, Buck G, Choudhury B, Clements J, Cole J, Dicks E, Forbes S, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jenkinson A, Jones D, Menzies A, Mironenko T, Perry J, Raine K, Richardson D, Shepherd R, Small A, Tofts C, Varian J, Webb T, West S, Widaa S, Yates A, Cahill DP, Louis DN, Goldstraw P, Nicholson AG, Brasseur F, Looijenga L, Weber BL, Chiew YE, DeFazio A, Greaves MF, Green AR, Campbell P, Birney E, Easton DF, Chenevix-Trench G, Tan MH, Khoo SK, Teh BT, Yuen ST, Leung SY, Wooster R, Futreal PA, Stratton MR (2007) Patterns of somatic mutation in human cancer genomes. Nature 446:153–158. doi:10.1038/nature05610

    CAS  PubMed Central  PubMed  Google Scholar 

  87. Capdeville R, Buchdunger E, Zimmermann J, Matter A (2002) Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 1:493–502. doi:10.1038/nrd839

    CAS  PubMed  Google Scholar 

  88. Ashwell S, Zabludoff S (2008) DNA damage detection and repair pathways–recent advances with inhibitors of checkpoint kinases in cancer therapy. Clin Cancer Res 14:4032–4037. doi:10.1158/1078-0432.CCR-07-5138

    CAS  PubMed  Google Scholar 

  89. Guertin DA, Sabatini DM (2007) Defining the role of mTOR in cancer. Cancer Cell 12:9–22. doi:10.1016/j.ccr.2007.05.008

    CAS  PubMed  Google Scholar 

  90. Taipale M, Jarosz DF, Lindquist S (2010) HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 11:515–528. doi:10.1038/nrm2918

    CAS  PubMed  Google Scholar 

  91. Stadtman ER (2001) Protein oxidation in aging and age-related diseases. Ann N Y Acad Sci 928:22–38. doi:10.1111/j.1749-6632.2001.tb05632.x

    CAS  PubMed  Google Scholar 

  92. Soga S, Akinaga S, Shiotsu Y (2013) Hsp90 inhibitors as anti-cancer agents, from basic discoveries to clinical development. Curr Pharm Des 19:366–376. doi:10.2174/1381612811306030366

    CAS  PubMed  Google Scholar 

  93. Shen M, Ping Dou Q (2013) Proteasome inhibition as a novel strategy for cancer treatment. In: Johnson DE (ed) Cell death signaling in cancer biology and treatment. Springer Sciences, New York, NY, pp 303–329

    Google Scholar 

  94. Groll M, Berkers CR, Ploegh HL, Ovaa H (2006) Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome. Structure 14:451–456. doi:10.1016/j.str.2005.11.019

    CAS  PubMed  Google Scholar 

  95. D’Arcy P, Brnjic S, Olofsson MH, Fryknas M, Lindsten K, De Cesare M, Perego P, Sadeghi B, Hassan M, Larsson R, Linder S (2011) Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat Med 17:1636–1640. doi:10.1038/nm.2536

    PubMed  Google Scholar 

  96. Lee BH, Lee MJ, Park S, Oh DC, Elsasser S, Chen PC, Gartner C, Dimova N, Hanna J, Gygi SP, Wilson SM, King RW, Finley D (2010) Enhancement of proteasome activity by a small-molecule inhibitor of USP14. Nature 467:179–184. doi:10.1038/nature09299

    CAS  PubMed Central  PubMed  Google Scholar 

  97. Ceccarelli DF, Tang X, Pelletier B, Orlicky S, Xie W, Plantevin V, Neculai D, Chou YC, Ogunjimi A, Al-Hakim A, Varelas X, Koszela J, Wasney GA, Vedadi M, Dhe-Paganon S, Cox S, Xu S, Lopez-Girona A, Mercurio F, Wrana J, Durocher D, Meloche S, Webb DR, Tyers M, Sicheri F (2011) An allosteric inhibitor of the human Cdc34 ubiquitin-conjugating enzyme. Cell 145:1075–1087. doi:10.1016/j.cell.2011.05.039

    CAS  PubMed  Google Scholar 

  98. Soucy TA, Smith PG, Milhollen MA, Berger AJ, Gavin JM, Adhikari S, Brownell JE, Burke KE, Cardin DP, Critchley S, Cullis CA, Doucette A, Garnsey JJ, Gaulin JL, Gershman RE, Lublinsky AR, McDonald A, Mizutani H, Narayanan U, Olhava EJ, Peluso S, Rezaei M, Sintchak MD, Talreja T, Thomas MP, Traore T, Vyskocil S, Weatherhead GS, Yu J, Zhang J, Dick LR, Claiborne CF, Rolfe M, Bolen JB, Langston SP (2009) An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature 458:732–736. doi:10.1038/nature07884

    CAS  PubMed  Google Scholar 

  99. Deshpande AJ, Bradner J, Armstrong SA (2012) Chromatin modifications as therapeutic targets in MLL-rearranged leukemia. Trends Immunol 33:563–570. doi:10.1016/j.it.2012.06.002

    CAS  PubMed Central  PubMed  Google Scholar 

  100. Chi P, Allis CD, Wang GG (2010) Covalent histone modifications–miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 10:457–469. doi:10.1038/nrc2876

    CAS  PubMed Central  PubMed  Google Scholar 

  101. Kubota Y (2012) Tumor angiogenesis and anti-angiogenic therapy. Keio J Med 61:47–56. doi:10.2302/kjm.61.47

    CAS  PubMed  Google Scholar 

  102. Risau W (1997) Mechanisms of angiogenesis. Nature 386:671–674. doi:10.1038/386671a0

    CAS  PubMed  Google Scholar 

  103. Bikfalvi A, Bicknell R (2002) Recent advances in angiogenesis, anti-angiogenesis and vascular targeting. Trends Pharmacol Sci 23:576–582. doi:10.1016/S0165-6147(02)02109-0

    CAS  PubMed  Google Scholar 

  104. Chung AS, Ferrara N (2011) Developmental and pathological angiogenesis. Annu Rev Cell Dev Biol 27:563–584. doi:10.1146/annurev-cellbio-092910-154002

    CAS  PubMed  Google Scholar 

  105. Dayan F, Mazure NM, Brahimi-Horn MC, Pouyssegur J (2008) A dialogue between the hypoxia-inducible factor and the tumor microenvironment. Cancer Microenviron 1:53–68. doi:10.1007/s12307-008-0006-3

    PubMed Central  PubMed  Google Scholar 

  106. Nagy JA, Dvorak AM, Dvorak HF (2007) VEGF-A and the induction of pathological angiogenesis. Annu Rev Pathol 2:251–275. doi:10.1146/annurev.pathol.2.010506.134925

    CAS  PubMed  Google Scholar 

  107. Hendrix MJ, Seftor EA, Hess AR, Seftor RE (2003) Vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma. Nat Rev Cancer 3:411–421. doi:10.1038/nrc1092

    CAS  PubMed  Google Scholar 

  108. Adams RH, Alitalo K (2007) Molecular regulation of angiogenesis and lymphangiogenesis. Nat Rev Mol Cell Biol 8:464–478. doi:10.1038/nrm2183

    CAS  PubMed  Google Scholar 

  109. Shojaei F (2012) Anti-angiogenesis therapy in cancer: current challenges and future perspectives. Cancer Lett 320:130–137. doi:10.1016/j.canlet.2012.03.008

    CAS  PubMed  Google Scholar 

  110. Deng T, Zhang L, Liu XJ, Xu JM, Bai YX, Wang Y, Han Y, Li YH, Ba Y (2013) Bevacizumab plus irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) as the second-line therapy for patients with metastatic colorectal cancer, a multicenter study. Med Oncol 30:752. doi:10.1007/s12032-013-0752-z

    PubMed  Google Scholar 

  111. Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausova J, Macarulla T, Ruff P, van Hazel GA, Moiseyenko V, Ferry D, McKendrick J, Polikoff J, Tellier A, Castan R, Allegra C (2012) Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol 30:3499–3506. doi:10.1200/JCO.2012.42.8201

    PubMed  Google Scholar 

  112. Smith NR, Baker D, Farren M, Pommier A, Swann R, Wang X, Mistry S, McDaid K, Kendrew J, Womack C, Wedge SR, Barry ST (2013) Tumor stromal architecture can define the intrinsic tumor response to VEGF-targeted therapy. Clin Cancer Res 19:6943–6956. doi:10.1158/1078-0432.CCR-13-1637

    CAS  PubMed  Google Scholar 

  113. Vasudev NS, Reynolds AR (2014) Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. doi:10.1007/s10456-014-9420-y

    PubMed Central  PubMed  Google Scholar 

  114. Henkin J, Volpert OV (2011) Therapies using anti-angiogenic peptide mimetics of thrombospondin-1. Expert Opin Ther Targets 15:1369–1386. doi:10.1517/14728222.2011.640319

    CAS  PubMed  Google Scholar 

  115. Browder T, Butterfield CE, Kraling BM, Shi B, Marshall B, O’Reilly MS, Folkman J (2000) Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res 60:1878–1886, PMID: 10766175

    CAS  PubMed  Google Scholar 

  116. Damber JE, Vallbo C, Albertsson P, Lennernas B, Norrby K (2006) The anti-tumour effect of low-dose continuous chemotherapy may partly be mediated by thrombospondin. Cancer Chemother Pharmacol 58:354–360. doi:10.1007/s00280-005-0163-8

    CAS  PubMed  Google Scholar 

  117. Yap R, Veliceasa D, Emmenegger U, Kerbel RS, McKay LM, Henkin J, Volpert OV (2005) Metronomic low-dose chemotherapy boosts CD95-dependent antiangiogenic effect of the thrombospondin peptide ABT-510: a complementation antiangiogenic strategy. Clin Cancer Res 11:6678–6685. doi:10.1158/1078-0432.CCR-05-0621

    CAS  PubMed  Google Scholar 

  118. Streit M, Stephen AE, Hawighorst T, Matsuda K, Lange-Asschenfeldt B, Brown LF, Vacanti JP, Detmar M (2002) Systemic inhibition of tumor growth and angiogenesis by thrombospondin-2 using cell-based antiangiogenic gene therapy. Cancer Res 62:2004–2012, PMID: 11929817

    CAS  PubMed  Google Scholar 

  119. Butler GS, Overall CM (2009) Proteomic identification of multitasking proteins in unexpected locations complicates drug targeting. Nat Rev Drug Discov 8:935–948. doi:10.1038/nrd2945

    CAS  PubMed  Google Scholar 

  120. Hadler-Olsen E, Winberg JO, Uhlin-Hansen L (2013) Matrix metalloproteinases in cancer: their value as diagnostic and prognostic markers and therapeutic targets. Tumour Biol 34:2041–2051. doi:10.1007/s13277-013-0842-8

    CAS  PubMed  Google Scholar 

  121. Cooney MM, van Heeckeren W, Bhakta S, Ortiz J, Remick SC (2006) Drug insight: vascular disrupting agents and angiogenesis–novel approaches for drug delivery. Nat Clin Pract Oncol 3:682–692. doi:10.1038/ncponc0663

    CAS  PubMed  Google Scholar 

  122. Dark GG, Hill SA, Prise VE, Tozer GM, Pettit GR, Chaplin DJ (1997) Combretastatin A-4, an agent that displays potent and selective toxicity toward tumor vasculature. Cancer Res 57:1829–1834, PMID: 9157969

    CAS  PubMed  Google Scholar 

  123. Zhou S, Kestell P, Baguley BC, Paxton JW (2002) 5,6-dimethylxanthenone-4-acetic acid (DMXAA): a new biological response modifier for cancer therapy. Invest New Drugs 20:281–295. doi:10.1023/A:1016215015530

    CAS  PubMed  Google Scholar 

  124. Gajewski TF, Schreiber H, Fu YX (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14:1014–1022. doi:10.1038/ni.2703

    CAS  PubMed Central  PubMed  Google Scholar 

  125. Engels B, Engelhard VH, Sidney J, Sette A, Binder DC, Liu RB, Kranz DM, Meredith SC, Rowley DA, Schreiber H (2013) Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell 23:516–526. doi:10.1016/j.ccr.2013.03.018

    CAS  PubMed Central  PubMed  Google Scholar 

  126. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E, Samuels Y, Rosenberg SA (2013) Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 19:747–752. doi:10.1038/nm.3161

    CAS  PubMed Central  PubMed  Google Scholar 

  127. Goldenberg DM (1993) Monoclonal antibodies in cancer detection and therapy. Am J Med 94:297–312. doi:10.1016/0002-9343(93)90062-T

    CAS  PubMed  Google Scholar 

  128. Carter P (2001) Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 1:118–129. doi:10.1038/35101072

    CAS  PubMed  Google Scholar 

  129. Monoclonal antibody therapy (2014). http://en.wikipedia.org/wiki/Monoclonal_antibody_therapy. Accessed Mar 2014.

  130. Sharkey RM, Goldenberg DM (2011) Cancer radioimmunotherapy. Immunotherapy 3:349–370. doi:10.2217/imt.10.114

    PubMed Central  PubMed  Google Scholar 

  131. Pressman D, Korngold L (1953) The in vivo localization of anti-Wagner-osteogenic-sarcoma antibodies. Cancer 6:619–623, PMID: 13042784

    CAS  PubMed  Google Scholar 

  132. DeNardo SJ, DeNardo GL, O’Grady LF, Macey DJ, Mills SL, Epstein AL, Peng JS, McGahan JP (1987) Treatment of a patient with B cell lymphoma by I-131 LYM-1 monoclonal antibodies. Int J Biol Markers 2:49–53, PMID: 3501448

    CAS  PubMed  Google Scholar 

  133. Press OW, Eary JF, Badger CC, Martin PJ, Appelbaum FR, Levy R, Miller R, Brown S, Nelp WB, Krohn KA et al (1989) Treatment of refractory non-Hodgkin’s lymphoma with radiolabeled MB-1 (anti-CD37) antibody. J Clin Oncol 7:1027–1038, PMID: 2666588

    CAS  PubMed  Google Scholar 

  134. Kaminski MS, Zasadny KR, Francis IR, Milik AW, Ross CW, Moon SD, Crawford SM, Burgess JM, Petry NA, Butchko GM et al (1993) Radioimmunotherapy of B-cell lymphoma with [131I]anti-B1 (anti-CD20) antibody. N Engl J Med 329:459–465. doi:10.1056/NEJM199308123290703

    CAS  PubMed  Google Scholar 

  135. Goldenberg DM (2002) Targeted therapy of cancer with radiolabeled antibodies. J Nucl Med 43:693–713, PMID: 11994535

    CAS  PubMed  Google Scholar 

  136. Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L (2013) Trial watch: anticancer radioimmunotherapy. Oncoimmunology 2:e25595. doi:10.4161/onci.25595

    PubMed Central  PubMed  Google Scholar 

  137. www.clinicaltrials.gov. Accessed in Mar 2014

  138. Almqvist Y, Orlova A, Sjöström A, Jensen HJ, Lundqvist H, Sundin A, Tolmachev V (2005) In vitro characterization of 211At-labeled antibody A33-a potential therapeutic agent against metastatic colorectal carcinoma. Cancer Biother Radiopharm 20:514–523. doi:10.1089/cbr.2005.20.514

    CAS  PubMed  Google Scholar 

  139. Pickhard A, Piontek G, Seidl C, Kopping S, Blechert B, Misslbeck M, Brockhoff G, Bruchertseifer F, Morgenstern A, Essler M (2014) (2)(1)(3)Bi-anti-EGFR radioimmunoconjugates and X-ray irradiation trigger different cell death pathways in squamous cell carcinoma cells. Nucl Med Biol 41:68–76. doi:10.1016/j.nucmedbio.2013.09.010

    CAS  PubMed  Google Scholar 

  140. Qu C, Li Y, Song Y, Rizvi S, Raja C, Zhang D, Samra J, Smith R, Perkins A, Apostolidis C (2004) MUC1 expression in primary and metastatic pancreatic cancer cells for in vitro treatment by 213Bi-C595 radioimmunoconjugate. Br J Cancer 91:2086–2093. doi:10.1038/sj.bjc.6602232

    CAS  PubMed Central  PubMed  Google Scholar 

  141. Zhang DY, Li Y, Rizvi SMA, Qu C, Kearsley J, Allen BJ (2005) Cytotoxicity of breast cancer cells overexpressing HER2/neu by 213Bi-Herceptin radioimmunoconjugate. Cancer Lett 218:181–190. doi:10.1016/j.canlet.2004.07.050

    CAS  PubMed  Google Scholar 

  142. Li Y, Song E, Abbas Rizvi SM, Power CA, Beretov J, Raja C, Cozzi PJ, Morgenstern A, Apostolidis C, Allen BJ, Russell PJ (2009) Inhibition of micrometastatic prostate cancer cell spread in animal models by 213Bilabeled multiple targeted alpha radioimmunoconjugates. Clin Cancer Res 15:865–875. doi:10.1158/1078-0432.CCR-08-1203

    CAS  PubMed  Google Scholar 

  143. Pavlinkova G, Booth BJ, Batra SK, Colcher D (1999) Radioimmunotherapy of human colon cancer xenografts using a dimeric single-chain Fv antibody construct. Clin Cancer Res 5:2613–2619, PMID: 10499640

    CAS  PubMed  Google Scholar 

  144. Hu M, Chen P, Wang J, Scollard DA, Vallis KA, Reilly RM (2007) 123I-labeled HIV-1 tat peptide radioimmunoconjugates are imported into the nucleus of human breast cancer cells and functionally interact in vitro and in vivo with the cyclin-dependent kinase inhibitor, p21WAF-1/Cip-1. Eur J Nucl Med Mol Imaging 34:368–377. doi:10.1007/s00259-006-0189-0

    CAS  PubMed  Google Scholar 

  145. Brouwers AH, van Eerd JE, Frielink C, Oosterwijk E, Oyen WJ, Corstens FH, Boerman OC (2004) Optimization of radioimmunotherapy of renal cell carcinoma: labeling of monoclonal antibody cG250 with 131I, 90Y, 177Lu, or 186Re. J Nucl Med 45:327–337, PMID: 14960657

    CAS  PubMed  Google Scholar 

  146. Sharkey RM, Blumenthal RD, Behr TM, Wong GY, Haywood L, Forman D, Griffiths GL, Goldenberg DM (1997) Selection of radioimmunoconjugates for the therapy of well-established or micrometastatic colon carcinoma. Int J Cancer 72:477–485. doi:10.1002/(SICI)1097-0215(19970729)72:3<477::AID-IJC16>3.0.CO;2-9

    CAS  PubMed  Google Scholar 

  147. Cornelissen B, Darbar S, Kersemans V, Allen D, Falzone N, Barbeau J, Smart S, Vallis KA (2012) Amplification of DNA damage by a gammaH2AX-targeted radiopharmaceutical. Nucl Med Biol 39:1142–1151. doi:10.1016/j.nucmedbio.2012.06.001

    CAS  PubMed  Google Scholar 

  148. Chinn PC, Morena RA, Santoro DA, Kazules T, Kashmiri SV, Schlom J, Hanna N, Braslawsky G (2006) Pharmacokinetics and tumor localization of 111In-labeled HuCC49ΔCH2 in BALB/c mice and athymic murine colon carcinoma xenograft. Cancer Biother Radiopharm 21:106–116. doi:10.1089/cbr.2006.21.106

    CAS  PubMed  Google Scholar 

  149. Cornelissen B, McLarty K, Kersemans V, Scollard DA, Reilly RM (2008) Properties of [(111)In]-labeled HIV-1 tat peptide radioimmunoconjugates in tumor-bearing mice following intravenous or intratumoral injection. Nucl Med Biol 35:101–110. doi:10.1016/j.nucmedbio.2007.09.007

    CAS  PubMed  Google Scholar 

  150. Liu P, Boyle AJ, Lu Y, Reilly RM, Winnik MA (2012) Biotinylated polyacrylamide-based metal-chelating polymers and their influence on antigen recognition following conjugation to a trastuzumab Fab fragment. Biomacromolecules 13:2831–2842. doi:10.1021/bm300843u

    CAS  PubMed  Google Scholar 

  151. Sandstrom K, Nestor M, Ekberg T, Engstrom M, Anniko M, Lundqvist H (2008) Targeting CD44v6 expressed in head and neck squamous cell carcinoma: preclinical characterization of an 111In-labeled monoclonal antibody. Tumour Biol 29:137–144. doi:10.1159/000143399

    PubMed  Google Scholar 

  152. Price EW, Zeglis BM, Cawthray JF, Ramogida CF, Ramos N, Lewis JS, Adam MJ, Orvig C (2013) H(4)octapa-trastuzumab: versatile acyclic chelate system for 111In and 177Lu imaging and therapy. J Am Chem Soc 135:12707–12721. doi:10.1021/ja4049493

    CAS  PubMed Central  PubMed  Google Scholar 

  153. Tempero M, Leichner P, Baranowska-Kortylewicz J, Harrison K, Augustine S, Schlom J, Anderson J, Wisecarver J, Colcher D (2000) High-dose therapy with 90Yttrium-labeled monoclonal antibody CC49: a phase I trial. Clin Cancer Res 6:3095–3102, PMID: 10955789

    CAS  PubMed  Google Scholar 

  154. Fischer E, Grunberg J, Cohrs S, Hohn A, Waldner-Knogler K, Jeger S, Zimmermann K, Novak-Hofer I, Schibli R (2012) L1-CAM-targeted antibody therapy and (177)Lu-radioimmunotherapy of disseminated ovarian cancer. Int J Cancer 130:2715–2721. doi:10.1002/ijc.26321

    CAS  PubMed  Google Scholar 

  155. Nilsson R, Eriksson SE, Sjogren HO, Tennvall J (2011) Different toxicity profiles for drug- versus radionuclide-conjugated BR96 monoclonal antibodies in a syngeneic rat colon carcinoma model. Acta Oncol 50:711–718. doi:10.3109/0284186X.2010.547215

    CAS  PubMed  Google Scholar 

  156. Schneider N, Lobaugh M, Tan Z, Sandwall P, Chen P, Glover S, Cui L, Murry M, Dong Z, Torgue J (2013) Biodistribution of 212Pb conjugated trastuzumab in mice. J Radioanal Nucl Chem 296:75–81. doi:10.1007/s10967-012-2243-7

    CAS  Google Scholar 

  157. Mohsin H, Jia F, Sivaguru G, Hudson MJ, Shelton TD, Hoffman TJ, Cutler CS, Ketring AR, Athey PS, Simón J (2006) Radiolanthanide-labeled monoclonal antibody CC49 for radioimmunotherapy of cancer: biological comparison of DOTA conjugates and 149Pm, 166Ho, and 177Lu. Bioconjug Chem 17:485–492. doi:10.1021/bc0502356

    CAS  PubMed  Google Scholar 

  158. Stroomer JW, Roos JC, Sproll M, Quak JJ, Heider KH, Wilhelm BJ, Castelijns JA, Meyer R, Kwakkelstein MO, Snow GB, Adolf GR, van Dongen GA (2000) Safety and biodistribution of 99mTechnetium-labeled anti-CD44v6 monoclonal antibody BIWA 1 in head and neck cancer patients. Clin Cancer Res 6:3046–3055, PMID: 10955783

    CAS  PubMed  Google Scholar 

  159. Dahle J, Borrebaek J, Jonasdottir TJ, Hjelmerud AK, Melhus KB, Bruland OS, Press OW, Larsen RH (2007) Targeted cancer therapy with a novel low-dose rate alpha-emitting radioimmunoconjugate. Blood 110:2049–2056. doi:10.1182/blood-2007-01-066803

    CAS  PubMed  Google Scholar 

  160. Heyerdahl H, Abbas N, Brevik EM, Mollatt C, Dahle J (2012) Fractionated therapy of HER2-expressing breast and ovarian cancer xenografts in mice with targeted alpha emitting 227Th-DOTA-p-benzyl-trastuzumab. PLoS One 7:e42345. doi:10.1371/journal.pone.0042345

    CAS  PubMed Central  PubMed  Google Scholar 

  161. Maraveyas A, Snook D, Hird V, Kosmas C, Meares CF, Lambert HE, Epenetos AA (1994) Pharmacokinetics and toxicity of an yttrium-90-CITC-DTPA-HMFG1 radioimmunoconjugate for intraperitoneal radioimmunotherapy of ovarian cancer. Cancer 73:1067–1075, PMID: 8306249

    CAS  PubMed  Google Scholar 

  162. Jia F, Liu X, Li L, Mallapragada S, Narasimhan B, Wang Q (2013) Multifunctional nanoparticles for targeted delivery of immune activating and cancer therapeutic agents. J Control Release 172:1020–1034. doi:10.1016/j.jconrel.2013.10.012

    CAS  PubMed  Google Scholar 

  163. Kitson SL, Cuccurullo V, Moody TS, Mansi L (2013) Radionuclide antibody-conjugates, a targeted therapy towards cancer. Curr Radiopharm 6:57–71. doi:10.2174/1874471011306020001

    CAS  PubMed  Google Scholar 

  164. Zhang L, Chen H, Wang L, Liu T, Yeh J, Lu G, Yang L, Mao H (2010) Delivery of therapeutic radioisotopes using nanoparticle platforms: potential benefit in systemic radiation therapy. Nanotechnol Sci Appl 3:159–170. doi:10.2147/NSA.S7462

    CAS  PubMed Central  PubMed  Google Scholar 

  165. Barrett T, Ravizzini G, Choyke PL, Kobayashi H (2009) Dendrimers in medical nanotechnology. IEEE Eng Med Biol Mag 28:12–22. doi:10.1109/MEMB.2008.931012

    PubMed Central  PubMed  Google Scholar 

  166. Veiseh O, Kievit FM, Ellenbogen RG, Zhang M (2011) Cancer cell invasion: treatment and monitoring opportunities in nanomedicine. Adv Drug Deliv Rev 63:582–596. doi:10.1016/j.addr.2011.01.010

    CAS  PubMed Central  PubMed  Google Scholar 

  167. Li L, Wartchow CA, Danthi SN, Shen Z, Dechene N, Pease J, Choi HS, Doede T, Chu P, Ning S, Lee DY, Bednarski MD, Knox SJ (2004) A novel antiangiogenesis therapy using an integrin antagonist or anti-Flk-1 antibody coated 90Y-labeled nanoparticles. Int J Radiat Oncol Biol Phys 58:1215–1227. doi:10.1016/j.ijrobp.2003.10.057

    CAS  PubMed  Google Scholar 

  168. Belhaj-Tayeb H, Briane D, Vergote J, Kothan S, Leger G, Bendada SE, Tofighi M, Tamgac F, Cao A, Moretti JL (2003) In vitro and in vivo study of 99mTc-MIBI encapsulated in PEG-liposomes: a promising radiotracer for tumour imaging. Eur J Nucl Med Mol Imaging 30:502–509. doi:10.1007/s00259-002-1038-4

    CAS  PubMed  Google Scholar 

  169. Kobayashi H, Wu C, Kim MK, Paik CH, Carrasquillo JA, Brechbiel MW (1999) Evaluation of the in vivo biodistribution of indium-111 and yttrium-88 labeled dendrimer-1B4M-DTPA and its conjugation with anti-Tac monoclonal antibody. Bioconjug Chem 10:103–111. doi:10.1021/bc980091d

    CAS  PubMed  Google Scholar 

  170. Torchilin VP (2002) PEG based micelles as carriers of contrast agents for different imaging modalities. Adv Drug Deliv Rev 54:235–252. doi:10.1016/S0169-409X(02)00019-4

    CAS  PubMed  Google Scholar 

  171. Hallahan D, Geng L, Qu S, Scarfone C, Giorgio T, Donnelly E, Gao X, Clanton J (2003) Integrin-mediated targeting of drug delivery to irradiated tumor blood vessels. Cancer Cell 3:63–74. doi:10.1016/S1535-6108(02)00238-6

    CAS  PubMed  Google Scholar 

  172. Hu G, Lijowski M, Zhang H, Partlow KC, Caruthers SD, Kiefer G, Gulyas G, Athey P, Scott MJ, Wickline SA, Lanza GM (2007) Imaging of Vx-2 rabbit tumors with alpha(nu)beta3-integrin-targeted 111In nanoparticles. Int J Cancer 120:1951–1957. doi:10.1002/ijc.22581

    CAS  PubMed  Google Scholar 

  173. Chang YJ, Chang CH, Yu CY, Chang TJ, Chen LC, Chen MH, Lee TW, Ting G (2010) Therapeutic efficacy and microSPECT/CT imaging of 188Re-DXR-liposome in a C26 murine colon carcinoma solid tumor model. Nucl Med Biol 37:95–104. doi:10.1016/j.nucmedbio.2009.08.006

    CAS  PubMed  Google Scholar 

  174. Vicente S, Goins BA, Sanchez A, Alonso MJ, Phillips WT (2014) Biodistribution and lymph node retention of polysaccharide-based immunostimulating nanocapsules. Vaccine 32:1685–1692. doi:10.1016/j.vaccine.2014.01.059

    CAS  PubMed  Google Scholar 

  175. Chen J, Wu H, Han D, Xie C (2006) Using anti-VEGF McAb and magnetic nanoparticles as double-targeting vector for the radioimmunotherapy of liver cancer. Cancer Lett 231:169–175. doi:10.1016/j.canlet.2005.01.024

    CAS  PubMed  Google Scholar 

  176. Wu H, Wang J, Wang Z, Fisher DR, Lin Y (2008) Apoferritin-templated yttrium phosphate nanoparticle conjugates for radioimmunotherapy of cancers. J Nanosci Nanotechnol 8:2316–2322, PMID: 18572643

    CAS  PubMed  Google Scholar 

  177. Woodward JD, Kennel SJ, Mirzadeh S, Dai S, Wall JS, Richey T, Avenell J, Rondinone AJ (2007) In vivo SPECT/CT imaging and biodistribution using radioactive Cd125mTe/ZnS nanoparticles. Nanotechnology 18:175103. doi:10.1088/0957-4484/18/17/175103

    Google Scholar 

  178. Liu Z, Cai W, He L, Nakayama N, Chen K, Sun X, Chen X, Dai H (2007) In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nat Nanotechnol 2:47–52. doi:10.1038/nnano.2006.170

    CAS  PubMed  Google Scholar 

  179. Bandekar A, Zhu C, Jindal R, Bruchertseifer F, Morgenstern A, Sofou S (2014) Anti-prostate-specific membrane antigen liposomes loaded with 225Ac for potential targeted antivascular alpha-particle therapy of cancer. J Nucl Med 55:107–114. doi:10.2967/jnumed.113.125476

    CAS  PubMed  Google Scholar 

  180. Wayne AS, Fitzgerald DJ, Kreitman RJ, Pastan I (2014) Immunotoxins for leukemia. Blood. doi:10.1182/blood-2014-01-492256

    Google Scholar 

  181. Antignani A, Fitzgerald D (2013) Immunotoxins: the role of the toxin. Toxins (Basel) 5:1486–1502. doi:10.3390/toxins5081486

    CAS  Google Scholar 

  182. Olsen E, Duvic M, Frankel A, Kim Y, Martin A, Vonderheid E, Jegasothy B, Wood G, Gordon M, Heald P, Oseroff A, Pinter-Brown L, Bowen G, Kuzel T, Fivenson D, Foss F, Glode M, Molina A, Knobler E, Stewart S, Cooper K, Stevens S, Craig F, Reuben J, Bacha P, Nichols J (2001) Pivotal phase III trial of two dose levels of denileukin diftitox for the treatment of cutaneous T-cell lymphoma. J Clin Oncol 19:376–388, PMID: 11208829

    CAS  PubMed  Google Scholar 

  183. Jansen FK, Bourrie B, Casellas P, Dussossoy D, Gros O, Vic P, Vidal H, Gros P (1988) Toxin selection and modification: utilization of the A chain of ricin. Cancer Treat Res 37:97–111. doi:10.1007/978-1-4613-1083-9_7

    CAS  PubMed  Google Scholar 

  184. Hudson TH, Neville DM Jr (1988) Enhancement of immunotoxin action: manipulation of the cellular routing of proteins. Cancer Treat Res 37:371–389. doi:10.1007/978-1-4613-1083-9_20

    CAS  PubMed  Google Scholar 

  185. Casellas P, Jansen FK (1988) Immunotoxin enhancers. Cancer Treat Res 37:351–369. doi:10.1007/978-1-4613-1083-9_19

    CAS  PubMed  Google Scholar 

  186. Kantarjian H, Thomas D, Wayne AS, O’Brien S (2012) Monoclonal antibody-based therapies: a new dawn in the treatment of acute lymphoblastic leukemia. J Clin Oncol 30:3876–3883. doi:10.1200/JCO.2012.41.6768

    CAS  PubMed Central  PubMed  Google Scholar 

  187. Messmann RA, Vitetta ES, Headlee D, Senderowicz AM, Figg WD, Schindler J, Michiel DF, Creekmore S, Steinberg SM, Kohler D, Jaffe ES, Stetler-Stevenson M, Chen H, Ghetie V, Sausville EA (2000) A phase I study of combination therapy with immunotoxins IgG-HD37-deglycosylated ricin A chain (dgA) and IgG-RFB4-dgA (Combotox) in patients with refractory CD19(+), CD22(+) B cell lymphoma. Clin Cancer Res 6:1302–1313, PMID: 10778955

    CAS  PubMed  Google Scholar 

  188. Grossbard ML, Multani PS, Freedman AS, O’Day S, Gribben JG, Rhuda C, Neuberg D, Nadler LM (1999) A Phase II study of adjuvant therapy with anti-B4-blocked ricin after autologous bone marrow transplantation for patients with relapsed B-cell non-Hodgkin’s lymphoma. Clin Cancer Res 5:2392–2398, PMID: 10499609

    CAS  PubMed  Google Scholar 

  189. Vitetta ES (2000) Immunotoxins and vascular leak syndrome. Cancer J 6(Suppl 3):S218–224, PMID: 10874491

    PubMed  Google Scholar 

  190. Siegall CB, Liggitt D, Chace D, Mixan B, Sugai J, Davidson T, Steinitz M (1997) Characterization of vascular leak syndrome induced by the toxin component of Pseudomonas exotoxin-based immunotoxins and its potential inhibition with nonsteroidal anti-inflammatory drugs. Clin Cancer Res 3:339–345, PMID: 9815690

    CAS  PubMed  Google Scholar 

  191. Hu RG, Zhai QW, He WJ, Mei L, Liu WY (2002) Bioactivities of ricin retained and its immunoreactivity to anti-ricin polyclonal antibodies alleviated through pegylation. Int J Biochem Cell Biol 34:396–402. doi:10.1016/S1357-2725(01)00128-5

    CAS  PubMed  Google Scholar 

  192. Oraki Kohshour M, Mirzaie S, Zeinali M, Amin M, Said Hakhamaneshi M, Jalaili A, Mosaveri N, Jamalan M (2014) Ablation of breast cancer cells using trastuzumab-functionalized multi-walled carbon nanotubes and trastuzumab-diphtheria toxin conjugate. Chem Biol Drug Des 83:259–265. doi:10.1111/cbdd.12244

    CAS  PubMed  Google Scholar 

  193. Ramakrishnan S, Olson TA, Bautch VL, Mohanraj D (1996) Vascular endothelial growth factor-toxin conjugate specifically inhibits KDR/flk-1-positive endothelial cell proliferation in vitro and angiogenesis in vivo. Cancer Res 56:1324–1330, PMID: 8640821

    CAS  PubMed  Google Scholar 

  194. Selbo PK, Kaalhus O, Sivam G, Berg K (2001) 5-Aminolevulinic acid-based photochemical internalization of the immunotoxin MOC31-gelonin generates synergistic cytotoxic effects in vitro. Photochem Photobiol 74:303–310. doi:10.1562/0031-8655(2001)0740303AABPIO2.0.CO2

    CAS  PubMed  Google Scholar 

  195. Schiffer S, Letzian S, Jost E, Mladenov R, Hristodorov D, Huhn M, Fischer R, Barth S, Thepen T (2013) Granzyme M as a novel effector molecule for human cytolytic fusion proteins: CD64-specific cytotoxicity of Gm-H22 (scFv) against leukemic cells. Cancer Lett 341:178–185. doi:10.1016/j.canlet.2013.08.005

    CAS  PubMed  Google Scholar 

  196. Bergelt S, Frost S, Lilie H (2009) Listeriolysin O as cytotoxic component of an immunotoxin. Protein Sci 18:1210–1220. doi:10.1002/pro.130

    CAS  PubMed Central  PubMed  Google Scholar 

  197. Hassan R, Viner JL, Wang QC, Margulies I, Kreitman RJ, Pastan I (2000) Anti-tumor activity of K1-LysPE38QQR, an immunotoxin targeting mesothelin, a cell-surface antigen overexpressed in ovarian cancer and malignant mesothelioma. J Immunother 23:473–479. doi:10.1097/00002371-200007000-00011

    CAS  PubMed  Google Scholar 

  198. Bruell D, Stocker M, Huhn M, Redding N, Kupper M, Schumacher P, Paetz A, Bruns CJ, Haisma HJ, Fischer R, Finnern R, Barth S (2003) The recombinant anti-EGF receptor immunotoxin 425(scFv)-ETA′ suppresses growth of a highly metastatic pancreatic carcinoma cell line. Int J Oncol 23:1179–1186, PMID: 12964002

    CAS  PubMed  Google Scholar 

  199. Park JH, Kwon HW, Chung HK, Kim IH, Ahn K, Choi EJ, Pastan I, Choe M (2001) A divalent recombinant immunotoxin formed by a disulfide bond between the extension peptide chains. Mol Cells 12:398–402, PMID: 11804341

    CAS  PubMed  Google Scholar 

  200. Beers R, Chowdhury P, Bigner D, Pastan I (2000) Immunotoxins with increased activity against epidermal growth factor receptor vIII-expressing cells produced by antibody phage display. Clin Cancer Res 6:2835–2843, PMID: 10914732

    CAS  PubMed  Google Scholar 

  201. Salvatore G, Beers R, Margulies I, Kreitman RJ, Pastan I (2002) Improved cytotoxic activity toward cell lines and fresh leukemia cells of a mutant anti-CD22 immunotoxin obtained by antibody phage display. Clin Cancer Res 8:995–1002, PMID: 11948105

    CAS  PubMed  Google Scholar 

  202. Akamatsu Y, Murphy JC, Nolan KF, Thomas P, Kreitman RJ, Leung SO, Junghans RP (1998) A single-chain immunotoxin against carcinoembryonic antigen that suppresses growth of colorectal carcinoma cells. Clin Cancer Res 4:2825–2832, PMID: 9829749

    CAS  PubMed  Google Scholar 

  203. Liu W, Onda M, Kim C, Xiang L, Weldon JE, Lee B, Pastan I (2012) A recombinant immunotoxin engineered for increased stability by adding a disulfide bond has decreased immunogenicity. Protein Eng Des Sel 25:1–6. doi:10.1093/protein/gzr053

    CAS  PubMed Central  PubMed  Google Scholar 

  204. Wang L, Liu B, Schmidt M, Lu Y, Wels W, Fan Z (2001) Antitumor effect of an HER2-specific antibody-toxin fusion protein on human prostate cancer cells. Prostate 47:21–28. doi:10.1002/pros.1043

    CAS  PubMed  Google Scholar 

  205. Gadadhar S, Karande AA (2013) Abrin immunotoxin: targeted cytotoxicity and intracellular trafficking pathway. PLoS One 8:e58304. doi:10.1371/journal.pone.0058304

    CAS  PubMed Central  PubMed  Google Scholar 

  206. Martinez-Torrecuadrada JL, Cheung LH, Lopez-Serra P, Barderas R, Canamero M, Ferreiro S, Rosenblum MG, Casal JI (2008) Antitumor activity of fibroblast growth factor receptor 3-specific immunotoxins in a xenograft mouse model of bladder carcinoma is mediated by apoptosis. Mol Cancer Ther 7:862–873. doi:10.1158/1535-7163.MCT-07-0394

    CAS  PubMed  Google Scholar 

  207. Fracasso G, Bellisola G, Cingarlini S, Castelletti D, Prayer-Galetti T, Pagano F, Tridente G, Colombatti M (2002) Anti-tumor effects of toxins targeted to the prostate specific membrane antigen. Prostate 53:9–23. doi:10.1002/pros.10117

    CAS  PubMed  Google Scholar 

  208. Gilabert-Oriol R, Thakur M, von Mallinckrodt B, Hug T, Wiesner B, Eichhorst J, Melzig MF, Fuchs H, Weng A (2013) Modified trastuzumab and cetuximab mediate efficient toxin delivery while retaining antibody-dependent cell-mediated cytotoxicity in target cells. Mol Pharm 10:4347–4357. doi:10.1021/mp400444q

    CAS  PubMed  Google Scholar 

  209. Wei BR, Ghetie MA, Vitetta ES (2000) The combined use of an immunotoxin and a radioimmunoconjugate to treat disseminated human B-cell lymphoma in immunodeficient mice. Clin Cancer Res 6:631–642, PMID: 10690549

    CAS  PubMed  Google Scholar 

  210. Gillies SD, Young D, Lo KM, Roberts S (1993) Biological activity and in vivo clearance of antitumor antibody/cytokine fusion proteins. Bioconjug Chem 4:230–235. doi:10.1021/bc00021a008

    CAS  PubMed  Google Scholar 

  211. Schrama D, Reisfeld RA, Becker JC (2006) Antibody targeted drugs as cancer therapeutics. Nat Rev Drug Discov 5:147–159. doi:10.1038/nrd1957

    CAS  PubMed  Google Scholar 

  212. Manusama ER, Nooijen PT, Ten Hagen TL, Van Der Veen AH, De Vries MW, De Wilt JH, Van Ijken MG, Marquet RL, Eggermont AM (1998) Tumor necrosis factor-alpha in isolated perfusion systems in the treatment of cancer: the Rotterdam preclinical-clinical program. Semin Surg Oncol 14:232–237. doi:10.1002/(SICI)1098-2388(199804/05)14:3<232::AID-SSU7>3.0.CO;2-9

    CAS  PubMed  Google Scholar 

  213. Cohen J (1995) IL-12 deaths: explanation and a puzzle. Science 270:908. doi:10.1126/science.270.5238.908a

    CAS  PubMed  Google Scholar 

  214. Mattijssen V, De Mulder PH, De Graeff A, Hupperets PS, Joosten F, Ruiter DJ, Bier H, Palmer PA, Van den Broek P (1994) Intratumoral PEG-interleukin-2 therapy in patients with locoregionally recurrent head and neck squamous-cell carcinoma. Ann Oncol 5:957–960, PMID 7696170

    CAS  PubMed  Google Scholar 

  215. Lienard D, Ewalenko P, Delmotte JJ, Renard N, Lejeune FJ (1992) High-dose recombinant tumor necrosis factor alpha in combination with interferon gamma and melphalan in isolation perfusion of the limbs for melanoma and sarcoma. J Clin Oncol 10:52–60, PMID: 1727926

    CAS  PubMed  Google Scholar 

  216. Atkins MB, Kunkel L, Sznol M, Rosenberg SA (2000) High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am 6(Suppl 1):S11–S14, PMID: 10685652

    PubMed  Google Scholar 

  217. Becker JC, Pancook JD, Gillies SD, Furukawa K, Reisfeld RA (1996) T cell-mediated eradication of murine metastatic melanoma induced by targeted interleukin 2 therapy. J Exp Med 183:2361–2366, PMID: 8642346

    CAS  PubMed  Google Scholar 

  218. Halin C, Rondini S, Nilsson F, Berndt A, Kosmehl H, Zardi L, Neri D (2002) Enhancement of the antitumor activity of interleukin-12 by targeted delivery to neovasculature. Nat Biotechnol 20:264–269. doi:10.1038/nbt0302-264

    CAS  PubMed  Google Scholar 

  219. List T, Neri D (2013) Immunocytokines: a review of molecules in clinical development for cancer therapy. Clin Pharmacol 5:29–45. doi:10.2147/CPAA.S49231

    PubMed Central  PubMed  Google Scholar 

  220. Schrama D, Xiang R, Eggert AO, Andersen MH, Pedersen Ls LO, Kampgen E, Schumacher TN, Reisfeld RR, Becker JC (2004) Shift from systemic to site-specific memory by tumor-targeted IL-2. J Immunol 172:5843–5850, PMID: 15128763

    CAS  PubMed  Google Scholar 

  221. Schliemann C, Palumbo A, Zuberbuhler K, Villa A, Kaspar M, Trachsel E, Klapper W, Menssen HD, Neri D (2009) Complete eradication of human B-cell lymphoma xenografts using rituximab in combination with the immunocytokine L19-IL2. Blood 113:2275–2283. doi:10.1182/blood-2008-05-160747

    CAS  PubMed  Google Scholar 

  222. Schwager K, Hemmerle T, Aebischer D, Neri D (2013) The immunocytokine L19-IL2 eradicates cancer when used in combination with CTLA-4 blockade or with L19-TNF. J Invest Dermatol 133:751–758. doi:10.1038/jid.2012.376

    CAS  PubMed  Google Scholar 

  223. Bauer J, Namineni S, Reisinger F, Zoller J, Yuan D, Heikenwalder M (2012) Lymphotoxin, NF-kB, and cancer: the dark side of cytokines. Dig Dis 30:453–468. doi:10.1159/000341690

    PubMed  Google Scholar 

  224. Huang X, Ye D, Thorpe PE (2011) Enhancing the potency of a whole-cell breast cancer vaccine in mice with an antibody-IL-2 immunocytokine that targets exposed phosphatidylserine. Vaccine 29:4785–4793. doi:10.1016/j.vaccine.2011.04.082

    CAS  PubMed  Google Scholar 

  225. Ziebarth AJ, Felder MA, Harter J, Connor JP (2012) Uterine leiomyosarcoma diffusely express disialoganglioside GD2 and bind the therapeutic immunocytokine 14.18-IL2: implications for immunotherapy. Cancer Immunol Immunother 61:1149–1153

    CAS  PubMed  Google Scholar 

  226. Moschetta M, Pretto F, Berndt A, Galler K, Richter P, Bassi A, Oliva P, Micotti E, Valbusa G, Schwager K, Kaspar M, Trachsel E, Kosmehl H, Bani MR, Neri D, Giavazzi R (2012) Paclitaxel enhances therapeutic efficacy of the F8-IL2 immunocytokine to EDA-fibronectin-positive metastatic human melanoma xenografts. Cancer Res 72:1814–1824. doi:10.1158/0008-5472.CAN-11-1919

    CAS  PubMed  Google Scholar 

  227. Pedretti M, Verpelli C, Marlind J, Bertani G, Sala C, Neri D, Bello L (2010) Combination of temozolomide with immunocytokine F16-IL2 for the treatment of glioblastoma. Br J Cancer 103:827–836. doi:10.1038/sj.bjc.6605832

    CAS  PubMed Central  PubMed  Google Scholar 

  228. Frey K, Schliemann C, Schwager K, Giavazzi R, Johannsen M, Neri D (2010) The immunocytokine F8-IL2 improves the therapeutic performance of sunitinib in a mouse model of renal cell carcinoma. J Urol 184:2540–2548. doi:10.1016/j.juro.2010.07.030

    CAS  PubMed  Google Scholar 

  229. Hank JA, Gan J, Ryu H, Ostendorf A, Stauder MC, Sternberg A, Albertini M, Lo KM, Gillies SD, Eickhoff J, Sondel PM (2009) Immunogenicity of the hu14.18-IL2 immunocytokine molecule in adults with melanoma and children with neuroblastoma. Clin Cancer Res 15:5923–5930

    CAS  PubMed Central  PubMed  Google Scholar 

  230. Holden SA, Lan Y, Pardo AM, Wesolowski JS, Gillies SD (2001) Augmentation of antitumor activity of an antibody-interleukin 2 immunocytokine with chemotherapeutic agents. Clin Cancer Res 7:2862–2869, PMID: 11555604

    CAS  PubMed  Google Scholar 

  231. Johnson EE, Yamane BH, Buhtoiarov IN, Lum HD, Rakhmilevich AL, Mahvi DM, Gillies SD, Sondel PM (2009) Radiofrequency ablation combined with KS-IL2 immunocytokine (EMD 273066) results in an enhanced antitumor effect against murine colon adenocarcinoma. Clin Cancer Res 15:4875–4884. doi:10.1158/1078-0432.CCR-09-0110

    CAS  PubMed Central  PubMed  Google Scholar 

  232. Pasche N, Woytschak J, Wulhfard S, Villa A, Frey K, Neri D (2011) Cloning and characterization of novel tumor-targeting immunocytokines based on murine IL7. J Biotechnol 154:84–92. doi:10.1016/j.jbiotec.2011.04.003

    CAS  PubMed  Google Scholar 

  233. Fallon J, Tighe R, Kradjian G, Guzman W, Bernhardt A, Neuteboom B, Lan Y, Sabzevari H, Schlom J, Greiner JW (2014) The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget 5:1869–1884, PMID: 24681847

    PubMed Central  PubMed  Google Scholar 

  234. Kim H, Gao W, Ho M (2013) Novel immunocytokine IL12-SS1 (Fv) inhibits mesothelioma tumor growth in nude mice. PLoS One 8:e81919. doi:10.1371/journal.pone.0081919

    PubMed Central  PubMed  Google Scholar 

  235. Gillessen S, Gnad-Vogt US, Gallerani E, Beck J, Sessa C, Omlin A, Mattiacci MR, Liedert B, Kramer D, Laurent J, Speiser DE, Stupp R (2013) A phase I dose-escalation study of the immunocytokine EMD 521873 (Selectikine) in patients with advanced solid tumours. Eur J Cancer 49:35–44. doi:10.1016/j.ejca.2012.07.015

    CAS  PubMed  Google Scholar 

  236. Kaspar M, Trachsel E, Neri D (2007) The antibody-mediated targeted delivery of interleukin-15 and GM-CSF to the tumor neovasculature inhibits tumor growth and metastasis. Cancer Res 67:4940–4948. doi:10.1158/0008-5472.CAN-07-0283

    CAS  PubMed  Google Scholar 

  237. Liu D, Chang CH, Rossi EA, Cardillo TM, Goldenberg DM (2013) Interferon-lambda1 linked to a stabilized dimer of Fab potently enhances both antitumor and antiviral activities in targeted cells. PLoS One 8:e63940. doi:10.1371/journal.pone.0063940

    CAS  PubMed Central  PubMed  Google Scholar 

  238. Chen P, Nogusa S, Thapa RJ, Shaller C, Simmons H, Peri S, Adams GP, Balachandran S (2013) Anti-CD70 immunocytokines for exploitation of interferon-gamma-induced RIP1-dependent necrosis in renal cell carcinoma. PLoS One 8:e61446. doi:10.1371/journal.pone.0061446

    CAS  PubMed Central  PubMed  Google Scholar 

  239. Lyu MA, Kurzrock R, Rosenblum MG (2008) The immunocytokine scFv23/TNF targeting HER-2/neu induces synergistic cytotoxic effects with 5-fluorouracil in TNF-resistant pancreatic cancer cell lines. Biochem Pharmacol 75:836–846. doi:10.1016/j.bcp.2007.10.013

    CAS  PubMed  Google Scholar 

  240. Bagshawe KD (2006) Antibody-directed enzyme prodrug therapy (ADEPT) for cancer. Expert Rev Anticancer Ther 6:1421–1431. doi:10.1586/14737140.6.10.1421

    CAS  PubMed  Google Scholar 

  241. Cheng TL, Wei SL, Chen BM, Chern JW, Wu MF, Liu PW, Roffler SR (1999) Bystander killing of tumour cells by antibody-targeted enzymatic activation of a glucuronide prodrug. Br J Cancer 79:1378–1385. doi:10.1038/sj.bjc.6690221

    CAS  PubMed Central  PubMed  Google Scholar 

  242. Sharma SK (2013) Antibody-directed enzyme prodrug therapy (ADEPT). In: Schmidt SR (ed) Fusion proteins technologies for biopharmaceuticals. Wiley, New Jersey, pp 354–363

    Google Scholar 

  243. Begent RH, Bagshawe KD, Green AJ, Searle F (1987) The clinical value of imaging with antibody to human chorionic gonadotrophin in the detection of residual choriocarcinoma. Br J Cancer 55:657–660. doi:10.1038/bjc.1987.134

    CAS  PubMed Central  PubMed  Google Scholar 

  244. Bagshawe KD (2012) Antibody-directed enzyme prodrug therapy (ADEPT) – basic principles and its practice so far. In: Kratz F, Senter P, Steinhagen H (eds) Drug delivery in oncology. Wiley, Germany, pp 169–186

    Google Scholar 

  245. Baker M, Carr F (2010) Pre-clinical considerations in the assessment of immunogenicity for protein therapeutics. Curr Drug Saf 5:308–313. doi:10.2174/157488610792246000

    CAS  PubMed  Google Scholar 

  246. Holgate RG, Baker MP (2009) Circumventing immunogenicity in the development of therapeutic antibodies. IDrugs 12:233–237, PMID: 19350467

    CAS  PubMed  Google Scholar 

  247. Smith GK, Banks S, Blumenkopf TA, Cory M, Humphreys J, Laethem RM, Miller J, Moxham CP, Mullin R, Ray PH, Walton LM, Wolfe LA 3rd (1997) Toward antibody-directed enzyme prodrug therapy with the T268G mutant of human carboxypeptidase A1 and novel in vivo stable prodrugs of methotrexate. J Biol Chem 272:15804–15816, PMID: 9188478

    CAS  PubMed  Google Scholar 

  248. Syrigos KN, Rowlinson-Busza G, Epenetos AA (1998) In vitro cytotoxicity following specific activation of amygdalin by beta-glucosidase conjugated to a bladder cancer-associated monoclonal antibody. Int J Cancer 78:712–719. doi:10.1002/(SICI)1097-0215(19981209)78:6<712::AID-IJC8>3.0.CO;2-D

    CAS  PubMed  Google Scholar 

  249. Chen KC, Wu SY, Leu YL, Prijovich ZM, Chen BM, Wang HE, Cheng TL, Roffler SR (2011) A humanized immunoenzyme with enhanced activity for glucuronide prodrug activation in the tumor microenvironment. Bioconjug Chem 22:938–948. doi:10.1021/bc1005784

    CAS  PubMed  Google Scholar 

  250. Alderson RF, Toki BE, Roberge M, Geng W, Basler J, Chin R, Liu A, Ueda R, Hodges D, Escandon E (2006) Characterization of a CC49-based single-chain fragment-β-lactamase fusion protein for antibody-directed enzyme prodrug therapy (ADEPT). Bioconjug Chem 17:410–418. doi:10.1021/bc0503521

    CAS  PubMed  Google Scholar 

  251. Wang H, Shi PJ, Wu MF, Li N, Zhou XL, Fan FY (2010) Construction, expression and functional characterization of the beta-lactamase with alphav integrin ligands. Protein Pept Lett 17:1562–1565. doi:10.2174/0929866511009011562

    CAS  PubMed  Google Scholar 

  252. Hao XK, Liu JY, Yue QH, Wu GJ, Bai YJ, Yin Y (2006) In vitro and in vivo prodrug therapy of prostate cancer using anti-gamma-Sm-scFv/hCPA fusion protein. Prostate 66:858–866. doi:10.1002/pros.20402

    CAS  PubMed  Google Scholar 

  253. Deckert PM, Bornmann WG, Ritter G, Williams C Jr, Franke J, Keilholz U, Thiel E, Old LJ, Bertino JR, Welt S (2004) Specific tumour localisation of a huA33 antibody–carboxypeptidase A conjugate and activation of methotrexate-phenylalanine. Int J Oncol 24:1289–1295, PMID: 15067353

    CAS  PubMed  Google Scholar 

  254. Sharma SK, Bagshawe KD, Burke PJ, Boden JA, Rogers GT, Springer CJ, Melton RG, Sherwood RF (1994) Galactosylated antibodies and antibody-enzyme conjugates in antibody-directed enzyme prodrug therapy. Cancer 73:1114–1120, PMID: 8306255

    CAS  PubMed  Google Scholar 

  255. Webley SD, Francis RJ, Pedley RB, Sharma SK, Begent RH, Hartley JA, Hochhauser D (2001) Measurement of the critical DNA lesions produced by antibody-directed enzyme prodrug therapy (ADEPT) in vitro, in vivo and in clinical material. Br J Cancer 84:1671–1676. doi:10.1054/bjoc.2001.1843

    CAS  PubMed Central  PubMed  Google Scholar 

  256. Pedley RB, Sharma SK, Boxer GM, Boden R, Stribbling SM, Davies L, Springer CJ, Begent RH (1999) Enhancement of antibody-directed enzyme prodrug therapy in colorectal xenografts by an antivascular agent. Cancer Res 59:3998–4003, PMID: 10463598

    CAS  PubMed  Google Scholar 

  257. Mayer A, Sharma SK, Tolner B, Minton NP, Purdy D, Amlot P, Tharakan G, Begent RH, Chester KA (2004) Modifying an immunogenic epitope on a therapeutic protein: a step towards an improved system for antibody-directed enzyme prodrug therapy (ADEPT). Br J Cancer 90:2402–2410. doi:10.1038/sj.bjc.6601888

    CAS  PubMed Central  PubMed  Google Scholar 

  258. Park JI, Cao L, Platt VM, Huang Z, Stull RA, Dy EE, Sperinde JJ, Yokoyama JS, Szoka FC (2009) Antitumor therapy mediated by 5-fluorocytosine and a recombinant fusion protein containing TSG-6 hyaluronan binding domain and yeast cytosine deaminase. Mol Pharm 6:801–812. doi:10.1021/mp800013c

    CAS  PubMed Central  PubMed  Google Scholar 

  259. Coelho V, Dernedde J, Petrausch U, Panjideh H, Fuchs H, Menzel C, Dubel S, Keilholz U, Thiel E, Deckert PM (2007) Design, construction, and in vitro analysis of A33scFv::CDy, a recombinant fusion protein for antibody-directed enzyme prodrug therapy in colon cancer. Int J Oncol 31:951–957, PMID: 17786329

    CAS  PubMed  Google Scholar 

  260. Biela BH, Khawli LA, Hu P, Epstein AL (2003) Chimeric TNT-3/human beta-glucuronidase fusion proteins for antibody-directed enzyme prodrug therapy (ADEPT). Cancer Biother Radiopharm 18:339–353. doi:10.1089/108497803322285099

    CAS  PubMed  Google Scholar 

  261. Florent JC, Dong X, Gaudel G, Mitaku S, Monneret C, Gesson JP, Jacquesy JC, Mondon M, Renoux B, Andrianomenjanahary S, Michel S, Koch M, Tillequin F, Gerken M, Czech J, Straub R, Bosslet K (1998) Prodrugs of anthracyclines for use in antibody-directed enzyme prodrug therapy. J Med Chem 41:3572–3581. doi:10.1021/jm970589l

    CAS  PubMed  Google Scholar 

  262. Haisma HJ, Sernee MF, Hooijberg E, Brakenhoff RH, Vd Meulen-Muileman IH, Pinedo HM, Boven E (1998) Construction and characterization of a fusion protein of single-chain anti-CD20 antibody and human beta-glucuronidase for antibody-directed enzyme prodrug therapy. Blood 92:184–190, PMID 9639515

    CAS  PubMed  Google Scholar 

  263. Afshar S, Asai T, Morrison SL (2009) Humanized ADEPT comprised of an engineered human purine nucleoside phosphorylase and a tumor targeting peptide for treatment of cancer. Mol Cancer Ther 8:185–193. doi:10.1158/1535-7163.MCT-08-0652

    CAS  PubMed Central  PubMed  Google Scholar 

  264. Heinis C, Alessi P, Neri D (2004) Engineering a thermostable human prolyl endopeptidase for antibody-directed enzyme prodrug therapy. Biochemistry 43:6293–6303. doi:10.1021/bi0361160

    CAS  PubMed  Google Scholar 

  265. Knox RJ, Friedlos F, Boland MP (1993) The bioactivation of CB 1954 and its use as a prodrug in antibody-directed enzyme prodrug therapy (ADEPT). Cancer Metastasis Rev 12:195–212. doi:10.1007/bf00689810

    CAS  PubMed  Google Scholar 

  266. Lu JY, Lowe DA, Kennedy MD, Low PS (1999) Folate-targeted enzyme prodrug cancer therapy utilizing penicillin-V amidase and a doxorubicin prodrug. J Drug Target 7:43–53. doi:10.3109/10611869909085491

    CAS  PubMed  Google Scholar 

  267. Padiolleau-Lefevre S, Naya RB, Shahsavarian MA, Friboulet A, Avalle B (2014) Catalytic antibodies and their applications in biotechnology: state of the art. Biotechnol Lett 36(7):1369–79. doi:10.1007/s10529-014-1503-8

    CAS  PubMed  Google Scholar 

  268. Abraham S, Guo F, Li LS, Rader C, Liu C, Barbas CF 3rd, Lerner RA, Sinha SC (2007) Synthesis of the next-generation therapeutic antibodies that combine cell targeting and antibody-catalyzed prodrug activation. Proc Natl Acad Sci USA 104:5584–5589. doi:10.1073/pnas.0700223104

    CAS  PubMed Central  PubMed  Google Scholar 

  269. Zawilska JB, Wojcieszak J, Olejniczak AB (2013) Prodrugs: a challenge for the drug development. Pharmacol Rep 65:1–14. doi:10.1016/S1734-1140(13)70959-9

    CAS  PubMed  Google Scholar 

  270. Jain S, Kumar D, Swarnakar NK, Thanki K (2012) Polyelectrolyte stabilized multilayered liposomes for oral delivery of paclitaxel. Biomaterials 33:6758–6768. doi:10.1016/j.biomaterials.2012.05.026

    CAS  PubMed  Google Scholar 

  271. Sapra P, Shor B (2013) Monoclonal antibody-based therapies in cancer: advances and challenges. Pharmacol Ther 138:452–469. doi:10.1016/j.pharmthera.2013.03.004

    CAS  PubMed  Google Scholar 

  272. Iden DL, Allen TM (2001) In vitro and in vivo comparison of immunoliposomes made by conventional coupling techniques with those made by a new post-insertion approach. Biochim Biophys Acta 1513:207–216. doi:10.1016/S0005-2736(01)00357-1

    CAS  PubMed  Google Scholar 

  273. Ishida T, Iden DL, Allen TM (1999) A combinatorial approach to producing sterically stabilized (stealth) immunoliposomal drugs. FEBS Lett 460:129–133. doi:10.1016/S0014-5793(99)01320-4

    CAS  PubMed  Google Scholar 

  274. Sofou S, Sgouros G (2008) Antibody-targeted liposomes in cancer therapy and imaging. Expert Opin Drug Deliv 5:189–204. doi:10.1517/17425247.5.2.189

    CAS  PubMed  Google Scholar 

  275. Sapra P, Allen TM (2002) Internalizing antibodies are necessary for improved therapeutic efficacy of antibody-targeted liposomal drugs. Cancer Res 62:7190–7194, PMID: 12499256

    CAS  PubMed  Google Scholar 

  276. Mastrobattista E, Koning GA, van Bloois L, Filipe AC, Jiskoot W, Storm G (2002) Functional characterization of an endosome-disruptive peptide and its application in cytosolic delivery of immunoliposome-entrapped proteins. J Biol Chem 277:27135–27143. doi:10.1074/jbc.M200429200

    CAS  PubMed  Google Scholar 

  277. Torchilin VP (2005) Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov 4:145–160. doi:10.1038/nrd1632

    CAS  PubMed  Google Scholar 

  278. Gao J, Chen H, Yu Y, Song J, Song H, Su X, Li W, Tong X, Qian W, Wang H (2013) Inhibition of hepatocellular carcinoma growth using immunoliposomes for co-delivery of adriamycin and ribonucleotide reductase M2 siRNA. Biomaterials 34:10084–10098. doi:10.1016/j.biomaterials.2013.08.088

    CAS  PubMed  Google Scholar 

  279. Gao J, Liu W, Xia Y, Li W, Sun J, Chen H, Li B, Zhang D, Qian W, Meng Y (2011) The promotion of siRNA delivery to breast cancer overexpressing epidermal growth factor receptor through anti-EGFR antibody conjugation by immunoliposomes. Biomaterials 32:3459–3470. doi:10.1016/j.biomaterials.2011.01.034

    CAS  PubMed  Google Scholar 

  280. Deng L, Zhang Y, Ma L, Jing X, Ke X, Lian J, Zhao Q, Yan B, Zhang J, Yao J, Chen J (2013) Comparison of anti-EGFR-Fab′ conjugated immunoliposomes modified with two different conjugation linkers for siRNa delivery in SMMC-7721 cells. Int J Nanomedicine 8:3271–3283. doi:10.2147/IJN.S47597

    PubMed Central  PubMed  Google Scholar 

  281. Drummond DC, Noble CO, Guo Z, Hayes ME, Connolly-Ingram C, Gabriel BS, Hann B, Liu B, Park JW, Hong K, Benz CC, Marks JD, Kirpotin DB (2010) Development of a highly stable and targetable nanoliposomal formulation of topotecan. J Control Release 141:13–21. doi:10.1016/j.jconrel.2009.08.006

    CAS  PubMed  Google Scholar 

  282. Nishikawa K, Asai T, Shigematsu H, Shimizu K, Kato H, Asano Y, Takashima S, Mekada E, Oku N, Minamino T (2012) Development of anti-HB-EGF immunoliposomes for the treatment of breast cancer. J Control Release 160:274–280. doi:10.1016/j.jconrel.2011.10.010

    CAS  PubMed  Google Scholar 

  283. Apte A, Koren E, Koshkaryev A, Torchilin VP (2014) Doxorubicin in TAT peptide-modified multifunctional immunoliposomes demonstrates increased activity against both drug-sensitive and drug-resistant ovarian cancer models. Cancer Biol Ther 15:69–80. doi:10.4161/cbt.26609

    CAS  PubMed  Google Scholar 

  284. Wicki A, Rochlitz C, Orleth A, Ritschard R, Albrecht I, Herrmann R, Christofori G, Mamot C (2012) Targeting tumor-associated endothelial cells: anti-VEGFR2 immunoliposomes mediate tumor vessel disruption and inhibit tumor growth. Clin Cancer Res 18:454–464. doi:10.1158/1078-0432.CCR-11-1102

    CAS  PubMed  Google Scholar 

  285. Padhye SS, Guin S, Yao HP, Zhou YQ, Zhang R, Wang MH (2011) Sustained expression of the RON receptor tyrosine kinase by pancreatic cancer stem cells as a potential targeting moiety for antibody-directed chemotherapeutics. Mol Pharm 8:2310–2319. doi:10.1021/mp200193u

    CAS  PubMed  Google Scholar 

  286. Guin S, Ma Q, Padhye S, Zhou YQ, Yao HP, Wang MH (2011) Targeting acute hypoxic cancer cells by doxorubicin-immunoliposomes directed by monoclonal antibodies specific to RON receptor tyrosine kinase. Cancer Chemother Pharmacol 67:1073–1083. doi:10.1007/s00280-010-1408-8

    CAS  PubMed  Google Scholar 

  287. Mortensen JH, Jeppesen M, Pilgaard L, Agger R, Duroux M, Zachar V, Moos T (2013) Targeted antiepidermal growth factor receptor (cetuximab) immunoliposomes enhance cellular uptake in vitro and exhibit increased accumulation in an intracranial model of glioblastoma multiforme. J Drug Deliv 2013:209205. doi:10.1155/2013/209205

    PubMed Central  PubMed  Google Scholar 

  288. Lehtinen J, Raki M, Bergstrom KA, Uutela P, Lehtinen K, Hiltunen A, Pikkarainen J, Liang H, Pitkanen S, Maatta AM, Ketola RA, Yliperttula M, Wirth T, Urtti A (2012) Pre-targeting and direct immunotargeting of liposomal drug carriers to ovarian carcinoma. PLoS One 7:e41410. doi:10.1371/journal.pone.0041410

    CAS  PubMed Central  PubMed  Google Scholar 

  289. Hantel C, Lewrick F, Schneider S, Zwermann O, Perren A, Reincke M, Suss R, Beuschlein F (2010) Anti insulin-like growth factor I receptor immunoliposomes: a single formulation combining two anticancer treatments with enhanced therapeutic efficiency. J Clin Endocrinol Metab 95:943–952. doi:10.1210/jc.2009-1980

    CAS  PubMed  Google Scholar 

  290. Koren E, Apte A, Jani A, Torchilin VP (2012) Multifunctional PEGylated 2C5-immunoliposomes containing pH-sensitive bonds and TAT peptide for enhanced tumor cell internalization and cytotoxicity. J Control Release 160:264–273. doi:10.1016/j.jconrel.2011.12.002

    CAS  PubMed Central  PubMed  Google Scholar 

  291. Kullberg M, Mann K, Anchordoquy TJ (2012) Targeting Her-2+ breast cancer cells with bleomycin immunoliposomes linked to LLO. Mol Pharm 9:2000–2008. doi:10.1021/mp300049n

    CAS  PubMed  Google Scholar 

  292. Catania A, Barrajon-Catalan E, Nicolosi S, Cicirata F, Micol V (2013) Immunoliposome encapsulation increases cytotoxic activity and selectivity of curcumin and resveratrol against HER2 overexpressing human breast cancer cells. Breast Cancer Res Treat 141:55–65. doi:10.1007/s10549-013-2667-y

    CAS  PubMed  Google Scholar 

  293. Yamamoto Y, Yoshida M, Sato M, Sato K, Kikuchi S, Sugishita H, Kuwabara J, Matsuno Y, Kojima Y, Morimoto M, Horiuchi A, Watanabe Y (2011) Feasibility of tailored, selective and effective anticancer chemotherapy by direct injection of docetaxel-loaded immunoliposomes into Her2/neu positive gastric tumor xenografts. Int J Oncol 38:33–39, PMID: 21109923

    CAS  PubMed  Google Scholar 

  294. Srivastava A, O’Connor IB, Pandit A, Gerard Wall J (2014) Polymer-antibody fragment conjugates for biomedical applications. Prog Polym Sci 39:308–329. doi:10.1016/j.progpolymsci.2013.09.003

    CAS  Google Scholar 

  295. Hagemeyer CE, von Zur MC, von Elverfeldt D, Peter K (2009) Single-chain antibodies as diagnostic tools and therapeutic agents. Thromb Haemost 101:1012–1019, PMID: 19492141

    CAS  PubMed  Google Scholar 

  296. Hu X, Spada S, White S, Hudson S, Magner E, Wall JG (2006) Adsorption and activity of a domoic acid binding antibody fragment on mesoporous silicates. J Phys Chem B 110:18703–18709. doi:10.1021/jp062423e

    CAS  PubMed  Google Scholar 

  297. Albrecht H, Denardo GL, Denardo SJ (2006) Monospecific bivalent scFv-SH: effects of linker length and location of an engineered cysteine on production, antigen binding activity and free SH accessibility. J Immunol Methods 310:100–116. doi:10.1016/j.jim.2005.12.012

    CAS  PubMed  Google Scholar 

  298. Weisser NE, Hall JC (2009) Applications of single-chain variable fragment antibodies in therapeutics and diagnostics. Biotechnol Adv 27:502–520. doi:10.1016/j.biotechadv.2009.04.004

    CAS  PubMed  Google Scholar 

  299. Yang K, Basu A, Wang M, Chintala R, Hsieh MC, Liu S, Hua J, Zhang Z, Zhou J, Li M, Phyu H, Petti G, Mendez M, Janjua H, Peng P, Longley C, Borowski V, Mehlig M, Filpula D (2003) Tailoring structure-function and pharmacokinetic properties of single-chain Fv proteins by site-specific PEGylation. Protein Eng 16:761–770. doi:10.1093/protein/gzg093

    CAS  PubMed  Google Scholar 

  300. Kim SH, Lee YS, Hwang SY, Bae GW, Nho K, Kang SW, Kwak YG, Moon CS, Han YS, Kim TY, Kho WG (2007) Effects of PEGylated scFv antibodies against Plasmodium vivax duffy binding protein on the biological activity and stability in vitro. J Microbiol Biotechnol 17:1670–1674, PMID: 18156783

    CAS  PubMed  Google Scholar 

  301. Kitamura K, Takahashi T, Takashina K, Yamaguchi T, Noguchi A, Tsurumi H, Toyokuni T, Hakomori S (1990) Polyethylene glycol modification of the monoclonal antibody A7 enhances its tumor localization. Biochem Biophys Res Commun 171:1387–1394. doi:10.1016/0006-291X(90)90839-F

    CAS  PubMed  Google Scholar 

  302. Delgado C, Pedley RB, Herraez A, Boden R, Boden JA, Keep PA, Chester KA, Fisher D, Begent RH, Francis GE (1996) Enhanced tumour specificity of an anti-carcinoembryonic antigen Fab′ fragment by poly(ethylene glycol) (PEG) modification. Br J Cancer 73:175–182. doi:10.1038/bjc.1996.32

    CAS  PubMed Central  PubMed  Google Scholar 

  303. Casey J, Pedley R, King D, Boden R, Chapman A, Yarranton G, Begent R (1999) Improved tumour targeting of di-Fab′ fragments modified with polyethylene glycol. Tumor Target 4:235–244, http://discovery.ucl.ac.uk/333098/

    CAS  Google Scholar 

  304. Li L, Yazaki PJ, Anderson AL, Crow D, Colcher D, Wu AM, Williams LE, Wong JY, Raubitschek A, Shively JE (2006) Improved biodistribution and radioimmunoimaging with poly(ethylene glycol)-DOTA-conjugated anti-CEA diabody. Bioconjug Chem 17:68–76. doi:10.1021/bc0502614

    CAS  PubMed  Google Scholar 

  305. Lu ZR, Kopeckova P, Kopecek J (1999) Polymerizable Fab′ antibody fragments for targeting of anticancer drugs. Nat Biotechnol 17:1101–1104. doi:10.1038/15085

    CAS  PubMed  Google Scholar 

  306. Seymour LW, Ferry DR, Anderson D, Hesslewood S, Julyan PJ, Poyner R, Doran J, Young AM, Burtles S, Kerr DJ, Cancer Research Campaign Phase IIICTc (2002) Hepatic drug targeting: phase I evaluation of polymer-bound doxorubicin. J Clin Oncol 20:1668–1676, PMID: 11896118

    CAS  PubMed  Google Scholar 

  307. Hovorka O, St’astny M, Etrych T, Subr V, Strohalm J, Ulbrich K, Rihova B (2002) Differences in the intracellular fate of free and polymer-bound doxorubicin. J Control Release 80:101–117. doi:10.1016/S0168-3659(02)00016-0

    CAS  PubMed  Google Scholar 

  308. Satchi R, Connors TA, Duncan R (2001) PDEPT: polymer-directed enzyme prodrug therapy. I. HPMA copolymer-cathepsin B and PK1 as a model combination. Br J Cancer 85:1070–1076

    CAS  PubMed Central  PubMed  Google Scholar 

  309. Etrych T, Strohalm J, Kovar L, Kabesova M, Rihova B, Ulbrich K (2009) HPMA copolymer conjugates with reduced anti-CD20 antibody for cell-specific drug targeting. I. Synthesis and in vitro evaluation of binding efficacy and cytostatic activity. J Control Release 140:18–26

    CAS  PubMed  Google Scholar 

  310. Gao J, Xia Y, Chen H, Yu Y, Song J, Li W, Qian W, Wang H, Dai J, Guo Y (2014) Polymer-lipid hybrid nanoparticles conjugated with anti-EGF receptor antibody for targeted drug delivery to hepatocellular carcinoma. Nanomedicine (Lond) 9:279–293. doi:10.2217/nnm.13.20

    CAS  Google Scholar 

  311. Khaw BA, Gada KS, Patil V, Panwar R, Mandapati S, Hatefi A, Majewski S, Weisenberger A (2014) Bispecific antibody complex pre-targeting and targeted delivery of polymer drug conjugates for imaging and therapy in dual human mammary cancer xenografts : targeted polymer drug conjugates for cancer diagnosis and therapy. Eur J Nucl Med Mol Imaging 41(8):1603–1616. doi:10.1007/s00259-014-2738-2

    CAS  PubMed  Google Scholar 

  312. Liu J, Kopeckova P, Buhler P, Wolf P, Pan H, Bauer H, Elsasser-Beile U, Kopecek J (2009) Biorecognition and subcellular trafficking of HPMA copolymer-anti-PSMA antibody conjugates by prostate cancer cells. Mol Pharm 6:959–970. doi:10.1021/mp8002682

    CAS  PubMed Central  PubMed  Google Scholar 

  313. Satchi-Fainaro R, Wrasidlo W, Lode HN, Shabat D (2002) Synthesis and characterization of a catalytic antibody-HPMA copolymer-conjugate as a tool for tumor selective prodrug activation. Bioorg Med Chem 10:3023–3029. doi:10.1016/s0968-0896(02)00156-6

    CAS  PubMed  Google Scholar 

  314. Berguig GY, Convertine AJ, Shi J, Palanca-Wessels MC, Duvall CL, Pun SH, Press OW, Stayton PS (2012) Intracellular delivery and trafficking dynamics of a lymphoma-targeting antibody-polymer conjugate. Mol Pharm 9:3506–3514. doi:10.1021/mp300338s

    CAS  PubMed Central  PubMed  Google Scholar 

  315. Tappertzhofen K, Metz VV, Hubo M, Barz M, Postina R, Jonuleit H, Zentel R (2013) Synthesis of maleimide-functionalyzed HPMA-copolymers and in vitro characterization of the aRAGE- and human immunoglobulin (huIgG)-polymer conjugates. Macromol Biosci 13:203–214. doi:10.1002/mabi.201200344

    CAS  PubMed  Google Scholar 

  316. Rihova B, Kopeckova P, Strohalm J, Rossmann P, Vetvicka V, Kopecek J (1988) Antibody-directed affinity therapy applied to the immune system: in vivo effectiveness and limited toxicity of daunomycin conjugated to HPMA copolymers and targeting antibody. Clin Immunol Immunopathol 46:100–114. doi:10.1016/0090-1229(88)90010-4

    CAS  PubMed  Google Scholar 

  317. Merdan T, Callahan J, Petersen H, Kunath K, Bakowsky U, Kopeckova P, Kissel T, Kopecek J (2003) Pegylated polyethylenimine-Fab' antibody fragment conjugates for targeted gene delivery to human ovarian carcinoma cells. Bioconjug Chem 14:989–996. doi:10.1021/bc0340767

    CAS  PubMed  Google Scholar 

  318. Pechar M, Ulbrich K, Jelínková M, Říhová B (2003) Conjugates of antibody-targeted PEG multiblock polymers with doxorubicin in cancer therapy. Macromol Biosci 3:364–372. doi:10.1002/mabi.200350004

    CAS  Google Scholar 

  319. Patel NR, Pattni BS, Abouzeid AH, Torchilin VP (2013) Nanopreparations to overcome multidrug resistance in cancer. Adv Drug Deliv Rev 65:1748–1762. doi:10.1016/j.addr.2013.08.004

    CAS  PubMed  Google Scholar 

  320. Kirtane AR, Kalscheuer SM, Panyam J (2013) Exploiting nanotechnology to overcome tumor drug resistance: challenges and opportunities. Adv Drug Deliv Rev 65:1731–1747. doi:10.1016/j.addr.2013.09.001

    CAS  PubMed  Google Scholar 

  321. Yan Y, Bjornmalm M, Caruso F (2013) Particle carriers for combating multidrug-resistant cancer. ACS Nano 7:9512–9517. doi:10.1021/nn405632s

    CAS  PubMed  Google Scholar 

  322. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC (2014) Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 66C:2–25. doi:10.1016/j.addr.2013.11.009

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sanyog Jain .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Controlled Release Society

About this chapter

Cite this chapter

Thanki, K., Kushwah, V., Jain, S. (2015). Recent Advances in Tumor Targeting Approaches. In: Devarajan, P., Jain, S. (eds) Targeted Drug Delivery : Concepts and Design. Advances in Delivery Science and Technology. Springer, Cham. https://doi.org/10.1007/978-3-319-11355-5_2

Download citation

Publish with us

Policies and ethics