891
Views
28
CrossRef citations to date
0
Altmetric
Review Article

Nanomedicine and cancer immunotherapy – targeting immunosuppressive cells

&
Pages 656-671 | Received 20 May 2015, Accepted 13 Jul 2015, Published online: 09 Oct 2015

References

  • Trotta F, Leufkens HGM, Schellens JHM, et al. Evaluation of oncology drugs at the European Medicines Agency and US Food and Drug Administration: when differences have an impact on clinical practice. J Clin Oncol 2011;29:2266–72
  • Bertrand N, Wu J, Xu X, et al. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014;66:2–25
  • Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release 2014;200:138–57
  • Weissig V, Pettinger T, Murdock N. Nanopharmaceuticals (part 1): products on the market. Int J Nanomedicine 2014;9:4357–73
  • Jain S, Doshi AS, Iyer AK, Amiji MM. Multifunctional nanoparticles for targeting cancer and inflammatory diseases. J Drug Target 2013;21:888–903
  • Davis ME, Zuckerman JE, Choi CHJ, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010;464:1067–70
  • Hrkach J, Von Hoff D, Mukkaram Ali M, et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci Transl Med 2012;4:128ra39
  • Andón FT, Pazinatto DB, Fadeel B. Towards the clinic: intelligent design of nanomedicines for cancer treatment. Nanomedicine (Lond) 2012;7:1293
  • Fadeel B. Clear and present danger? Engineered nanoparticles and the immune system. Swiss Med Wkly 2012;142:w13609
  • Allavena P, Mantovani A. Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment. Clin Exp Immunol 2012;167:195–205
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012;12:253–68
  • Galdiero MR, Bonavita E, Barajon I, et al. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013;218:1402–10
  • Whiteside TL. What are regulatory T cells (Treg) regulating in cancer and why? Semin Cancer Biol 2012;22:327–34
  • Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science 2013;339:286–91
  • Zhang Q, Liu L, Gong C, et al. Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS One 2012;7:e50946
  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162–74
  • Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 2008;454:436–44
  • Colotta F, Allavena P, Sica A, et al. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009;30:1073–81
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74
  • Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013;342:1432–3
  • Mueller KL. Cancer immunology and immunotherapy. Realizing the promise. Introduction. Science 2015;348:54–5
  • Kirkwood JM, Butterfield LH, Tarhini AA, et al. Immunotherapy of cancer in 2012. CA Cancer J Clin 2012;62:309–35
  • Ito F, Chang AE. Cancer immunotherapy: current status and future directions. Surg Oncol Clin N Am 2013;22:765–83
  • Galluzzi L, Vacchelli E, Bravo-San Pedro J-M, et al. Classification of current anticancer immunotherapies. Oncotarget 2014;5:12472–508
  • Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res 2013;19:6286–95
  • Tonra JR, Corcoran E, Deevi DS, et al. Prioritization of EGFR/IGF-IR/VEGFR2 combination targeted therapies utilizing cancer models. Anticancer Res 2009;29:1999–2007
  • Bertrand N, Wu J, Xu X, et al. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014;66:2–25
  • Li W, Wei H, Li H, et al. Cancer nanoimmunotherapy using advanced pharmaceutical nanotechnology. Nanomedicine (Lond) 2014;9:2587–605
  • Galluzzi L, Vacchelli E, Eggermont A, et al. Trial watch: experimental toll-like receptor agonists for cancer therapy. Oncoimmunology 2012;1:699–716
  • Julien DC, Behnke S, Wang G, et al. Utilization of monoclonal antibody-targeted nanomaterials in the treatment of cancer. MAbs 2011;3:467–78
  • Bhattacharyya S, Bhattacharya R, Curley S, et al. Nanoconjugation modulates the trafficking and mechanism of antibody induced receptor endocytosis. Proc Natl Acad Sci USA 2010;107:14541–6
  • Owen SC, Patel N, Logie J, et al. Targeting HER2+ breast cancer cells: lysosomal accumulation of anti-HER2 antibodies is influenced by antibody binding site and conjugation to polymeric nanoparticles. J Control Release 2013;172:395–404
  • Canton I, Massignani M, Patikarnmonthon N, et al. Fully synthetic polymer vesicles for intracellular delivery of antibodies in live cells. FASEB J 2012;27:98–108
  • Lee Y, Ishii T, Kim HJ, et al. Efficient delivery of bioactive antibodies into the cytoplasm of living cells by charge-conversional polyion complex micelles. Angew Chem Int Ed 2010;49:2552–5
  • Berguig GY, Convertine AJ, Shi J, et al. Intracellular delivery and trafficking dynamics of a lymphoma-targeting antibody–polymer conjugate. Mol Pharm 2012;9:3506–14
  • Sievers EL, Senter PD. Antibody-drug conjugates in cancer therapy. Annu Rev Med Annu Rev 2013;64:15–29
  • Hansel TT, Kropshofer H, Singer T, et al. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010;9:325–38
  • Makkouk A, Weiner GJ. Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge. Cancer Res 2014;75:5–10
  • Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23
  • Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015;33:1974–82
  • Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet 2014;384:1109–17
  • Toporkiewicz M, Meissner J, Matusewicz L, et al. Toward a magic or imaginary bullet? Ligands for drug targeting to cancer cells: principles, hopes, and challenges. Int J Nanomedicine 2015;10:1399–414
  • Gao J, Feng S-S, Guo Y. Antibody engineering promotes nanomedicine for cancer treatment. Nanomedicine (Lond) 2010;5:1141–5
  • Zhao G, Rodriguez BL. Molecular targeting of liposomal nanoparticles to tumor microenvironment. Int J Nanomedicine 2013;8:61–71
  • de la Fuente M, Langer R, Alonso MJ. Nanotechnology approaches for cancer immunotherapy and immunomodulation. In: Alonso MJ, Garcia-Fuentes M, eds. Nano-Oncologicals: new targeting and delivery approaches. Cham: Springer International Publishing; 2014:215–42
  • Silva JM, Videira M, Gaspar R, et al. Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release 2013;168:179–99
  • Swartz MA, Hirosue S, Hubbell JA. Engineering approaches to immunotherapy. Sci Transl Med 2012;4:148rv9
  • De Pas T, Giovannini M, Rescigno M, et al. Vaccines in non-small cell lung cancer: rationale, combination strategies and update on clinical trials. Crit Rev Oncol Hematol 2012;83:432–43
  • Csaba N, Garcia-Fuentes M, Alonso MJ. Nanoparticles for nasal vaccination. Adv Drug Deliv Rev 2009;61:140–57
  • Zolnik BS, González-Fernández A, Sadrieh N, Dobrovolskaia MA. Nanoparticles and the immune system. Endocrinology 2010;151:458–65
  • González-Aramundiz JV, Lozano MV, Sousa-Herves A, et al. Polypeptides and polyaminoacids in drug delivery. Expert Opin Drug Deliv 2012;9:183–201
  • Prego C, Torres D, Fernandez-Megia E, et al. Chitosan-PEG nanocapsules as new carriers for oral peptide delivery. Effect of chitosan pegylation degree. J Control Release 2006;111:299–308
  • Garcia-Fuentes M, Alonso MJ. Chitosan-based drug nanocarriers: where do we stand? J Control Release 2012;161:496–504
  • Li X, Min M, Du N, et al. Chitin, chitosan, and glycated chitosan regulate immune responses: the novel adjuvants for cancer vaccine. Clin Dev Immunol 2013;2013:387023
  • Hanlon DJ, Aldo PB, Devine L, et al. Enhanced stimulation of anti-ovarian cancer CD8(+) T cells by dendritic cells loaded with nanoparticle encapsulated tumor antigen. Am J Reprod Immunol 2011;65:597–609
  • Prasad S, Cody V, Saucier-Sawyer JK, et al. Polymer nanoparticles containing tumor lysates as antigen delivery vehicles for dendritic cell-based antitumor immunotherapy. Nanomedicine 2011;7:1–10
  • Prasad S, Cody V, Saucier-Sawyer JK, et al. Optimization of stability, encapsulation, release, and cross-priming of tumor antigen-containing PLGA nanoparticles. Pharm Res 2012;29:2565–77
  • Ma W, Smith T, Bogin V, et al. Enhanced presentation of MHC class Ia, Ib and class II-restricted peptides encapsulated in biodegradable nanoparticles: a promising strategy for tumor immunotherapy. J Transl Med 2011;9:34
  • Tan G, Wang Z, Zhang X, et al. Induction of CTLs by DCs pulsed with K-ras mutant peptide on the surface of nanoparticles in the treatment of pancreatic cancer. Oncol Rep 2011;26:215–21
  • Hemmi H, Kaisho T, Takeuchi O, et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol 2002;3:196–200
  • Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 1976;193:1007–8
  • Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999;17:2105–16
  • Wang BX, Rahbar R, Fish EN. Interferon: current status and future prospects in cancer therapy. J Interferon Cytokine Res 2011;31:545–52
  • Sánchez A, Tobío M, González L, et al. Biodegradable micro- and nanoparticles as long-term delivery vehicles for interferon-alpha. Eur J Pharm Sci 2003;18:221–9
  • Zhang Z, Tongchusak S, Mizukami Y, et al. Induction of anti-tumor cytotoxic T cell responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials 2011;32:3666–78
  • Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 2004;4:71–8
  • Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 2002;196:254–65
  • Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol 2009;86:1065–73
  • Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature 2013;496:445–55
  • Shand FHW, Ueha S, Otsuji M, et al. Tracking of intertissue migration reveals the origins of tumor-infiltrating monocytes. Proc Natl Acad Sci USA 2014;111:7771–6
  • Bronte V, Murray PJ. Understanding local macrophage phenotypes in disease: modulating macrophage function to treat cancer. Nat Med 2015;21:117–19
  • Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012;122:787–95
  • Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11:889–96
  • Mantovani A, Vecchi A, Allavena P. Pharmacological modulation of monocytes and macrophages. Curr Opin Pharmacol 2014;17:38–44
  • Youn J-I, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 2008;181:5791–802
  • Sica A, Bronte V. Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest 2007;117:1155–66
  • Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol 2013;91:493–502
  • Sica A, Melillo G, Varesio L. Hypoxia: a double-edged sword of immunity. J Mol Med (Berl) 2011;89:657–65
  • Gregory AD, Houghton AM. Tumor-associated neutrophils: new targets for cancer therapy. Cancer Res 2011;71:2411–16
  • Houghton AM. The paradox of tumor-associated neutrophils: fueling tumor growth with cytotoxic substances. Cell Cycle 2010;9:1732–7
  • Yan C, Huo X, Wang S, et al. Stimulation of hepatocarcinogenesis by neutrophils upon induction of oncogenic kras expression in transgenic zebrafish. J Hepatol 2015;63:420–8
  • Nishikawa H, Sakaguchi S. Regulatory T cells in tumor immunity. Int J Cancer 2010;127:759–67
  • Roncarolo M-G, Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol 2007;7:585–98
  • Nishikawa H, Kato T, Tawara I, et al. Definition of target antigens for naturally occurring CD4(+) CD25(+) regulatory T cells. J Exp Med 2005;201:681–6
  • François V, Ottaviani S, Renkvist N, et al. The CD4(+) T-cell response of melanoma patients to a MAGE-A3 peptide vaccine involves potential regulatory T cells. Cancer Res 2009;69:4335–45
  • Danke NA, Koelle DM, Yee C, et al. Autoreactive T cells in healthy individuals. J Immunol 2004;172:5967–72
  • Nishikawa H, Jäger E, Ritter G, et al. CD4 + CD25 + regulatory T cells control the induction of antigen-specific CD4 + helper T cell responses in cancer patients. Blood 2005;106:1008–11
  • Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010;148:135–46
  • Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 1986;46:6387–92
  • Fang J, Nakamura H, Maeda H. The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Deliv Rev 2011;63:136–51
  • Chauhan VP, Stylianopoulos T, Martin JD, et al. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat Nanotechnol 2012;7:383–8
  • Andón FT, Pazinatto DB, Fadeel B. The importance of size: nanomedicine for delivery of anticancer drugs in solid tumors. Nanomedicine (Lond) 2012;7:1294–5
  • Maeda H. Vascular permeability in cancer and infection as related to macromolecular drug delivery, with emphasis on the EPR effect for tumor-selective drug targeting. Proc Jpn Acad Ser B Phys Biol Sci 2012;88:53–71
  • Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med 2013;19:1423–37
  • Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 2014;25:198–213
  • Mantovani A, Allavena P. The interaction of anticancer therapies with tumor-associated macrophages. J Exp Med 2015;212:435–45
  • Zamboni WC, Eiseman JL, Strychor S, et al. Tumor disposition of pegylated liposomal CKD-602 and the reticuloendothelial system in preclinical tumor models. J Liposome Res 2011;21:70–80
  • Caron WP, Song G, Kumar P, et al. Interpatient pharmacokinetic and pharmacodynamic variability of carrier-mediated anticancer agents. Clin Pharmacol Ther 2012;91:802–12
  • Cortez-Retamozo V, Etzrodt M, Newton A, et al. Origins of tumor-associated macrophages and neutrophils. Proc Natl Acad Sci USA 2012;109:2491–6
  • Leuschner F, Dutta P, Gorbatov R, et al. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat Biotechnol 2011;29:1005–10
  • Singh I, Swami R, Khan W, Sistla R. Lymphatic system: a prospective area for advanced targeting of particulate drug carriers. Expert Opin Drug Deliv 2014;11:211–29
  • Zhang X, Lu W. Recent advances in lymphatic targeted drug delivery system for tumor metastasis. Cancer Biol Med 2014;11:247–54
  • Swartz MA. Immunomodulatory roles of lymphatic vessels in cancer progression. Cancer Immunol Res 2014;2:701–7
  • Li T, Wang G, Tan Y, Wang H. Inhibition of lymphangiogenesis of endothelial progenitor cells with VEGFR-3 siRNA delivered with PEI-alginate nanoparticles. Int J Biol Sci 2014;10:160–70
  • Abellán-Pose R, Borrajo E, Rodríguez-Évora E, et al. New strategy to tackle lymphatic metastasizing tumors based on polyaminoacid nanocapsules. 5th Int Congr Nanotechnology, Biol Med Krems; 2014 Mar 26–28; Austria
  • Yan Z, Zhan C, Wen Z, et al. LyP-1-conjugated doxorubicin-loaded liposomes suppress lymphatic metastasis by inhibiting lymph node metastases and destroying tumor lymphatics. Nanotechnology 2011;22:415103
  • Luo G, Yu X, Jin C, et al. LyP-1-conjugated nanoparticles for targeting drug delivery to lymphatic metastatic tumors. Int J Pharm 2010;385:150–6
  • Yan Z, Wang F, Wen Z, et al. LyP-1-conjugated PEGylated liposomes: a carrier system for targeted therapy of lymphatic metastatic tumor. J Control Release 2012;157:118–25
  • Fogal V, Zhang L, Krajewski S, Ruoslahti E. Mitochondrial/cell-surface protein p32/gC1qR as a molecular target in tumor cells and tumor stroma. Cancer Res 2008;68:7210–18
  • Zhong Y, Meng F, Deng C, Zhong Z. Ligand-directed active tumor-targeting polymeric nanoparticles for cancer chemotherapy. Biomacromolecules 2014;15:1955–69
  • Andón FT, Fadeel B. Nanotoxicology: towards safety-by-design. In: Alonso MJ, Garcia-Fuentes M, eds. Nano-oncologicals: new targeting and delivery approaches. Cham: Springer International Publishing; 2014:391–424
  • Cabral H, Matsumoto Y, Mizuno K, et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat Nanotechnol 2011;6:815–23
  • Geng Y, Dalhaimer P, Cai S, et al. Shape effects of filaments versus spherical particles in flow and drug delivery. Nat Nanotechnol 2007;2:249–55
  • Han H-S, Martin JD, Lee J, et al. Spatial charge configuration regulates nanoparticle transport and binding behavior in vivo. Angew Chem Int Ed 2013;52:1414–19
  • Lieleg O, Baumgärtel RM, Bausch AR. Selective filtering of particles by the extracellular matrix: an electrostatic bandpass. Biophys J 2009;97:1569–77
  • Owen SC, Doak AK, Ganesh AN, et al. Colloidal drug formulations can explain “bell-shaped” concentration–response curves. ACS Chem Biol 2014;9:777–84
  • Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov 2010;9:615–27
  • Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev 2008;60:1615–26
  • Zhong Y, Meng F, Deng C, Zhong Z. Ligand-directed active tumor-targeting polymeric nanoparticles for cancer chemotherapy. Biomacromolecules 2014;15:1955–69
  • Acharya S, Dilnawaz F, Sahoo SK. Targeted epidermal growth factor receptor nanoparticle bioconjugates for breast cancer therapy. Biomaterials 2009;30:5737–50
  • Debotton N, Parnes M, Kadouche J, Benita S. Overcoming the formulation obstacles towards targeted chemotherapy: in vitro and in vivo evaluation of cytotoxic drug loaded immunonanoparticles. J Control Release 2008;127:219–30
  • Song H, He R, Wang K, et al. Anti-HIF-1alpha antibody-conjugated pluronic triblock copolymers encapsulated with Paclitaxel for tumor targeting therapy. Biomaterials 2010;31:2302–12
  • Gu F, Zhang L, Teply BA, et al. Precise engineering of targeted nanoparticles by using self-assembled biointegrated block copolymers. Proc Natl Acad Sci USA 2008;105:2586–91
  • Nasongkla N, Bey E, Ren J, et al. Multifunctional polymeric micelles as cancer-targeted, MRI-ultrasensitive drug delivery systems. Nano Lett 2006;6:2427–30
  • Zhu Z, Xie C, Liu Q, et al. The effect of hydrophilic chain length and iRGD on drug delivery from poly(ɛ-caprolactone)-poly(N-vinylpyrrolidone) nanoparticles. Biomaterials 2011;32:9525–35
  • Zhang Y, Zhang H, Wang X, et al. The eradication of breast cancer and cancer stem cells using octreotide modified paclitaxel active targeting micelles and salinomycin passive targeting micelles. Biomaterials 2012;33:679–91
  • Liu Z, Jiao Y, Wang Y, et al. Polysaccharides-based nanoparticles as drug delivery systems. Adv Drug Deliv Rev 2008;60:1650–62
  • Oh EJ, Park K, Kim KS, et al. Target specific and long-acting delivery of protein, peptide, and nucleotide therapeutics using hyaluronic acid derivatives. J Control Release 2010;141:2–12
  • Drabovich AP, Berezovski MV, Musheev MU, Krylov SN. Selection of smart small-molecule ligands: the proof of principle. Anal Chem 2009;81:490–4
  • Ugel S, Peranzoni E, Desantis G, et al. Immune tolerance to tumor antigens occurs in a specialized environment of the spleen. Cell Rep 2012;2:628–39
  • Movahedi K, Schoonooghe S, Laoui D, et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res 2012;72:4165–77
  • Locke LW, Mayo MW, Yoo AD, et al. PET imaging of tumor associated macrophages using mannose coated 64Cu liposomes. Biomaterials 2012;33:7785–93
  • Mitchem JB, Brennan DJ, Knolhoff BL, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res 2013;73:1128–41
  • Etzerodt A, Maniecki MB, Graversen JH, et al. Efficient intracellular drug-targeting of macrophages using stealth liposomes directed to the hemoglobin scavenger receptor CD163. J Control Release 2012;160:72–80
  • Huang Z, Zhang Z, Jiang Y, et al. Targeted delivery of oligonucleotides into tumor-associated macrophages for cancer immunotherapy. J Control Release 2012;158:286–92
  • Puig-Kröger A, Sierra-Filardi E, Domínguez-Soto A, et al. Folate receptor beta is expressed by tumor-associated macrophages and constitutes a marker for M2 anti-inflammatory/regulatory macrophages. Cancer Res 2009;69:9395–403
  • Hattori Y, Yamashita J, Sakaida C, et al. Evaluation of antitumor effect of zoledronic acid entrapped in folate-linked liposome for targeting to tumor-associated macrophages. J Liposome Res 2014. [Epub ahead of print]. doi: 10.3109/08982104.2014.954128
  • Turk MJ, Waters DJ, Low PS. Folate-conjugated liposomes preferentially target macrophages associated with ovarian carcinoma. Cancer Lett 2004;213:165–72
  • Luo Y, Zhou H, Krueger J, et al. Targeting tumor-associated macrophages as a novel strategy against breast cancer. J Clin Invest 2006;116:2132–41
  • Gomez-Cabrero A, Wrasidlo W, Reisfeld RA. IMD-0354 targets breast cancer stem cells: a novel approach for an adjuvant to chemotherapy to prevent multidrug resistance in a murine model. PLoS One 2013;8:e73607
  • Zhang X, Tian W, Cai X, et al. Hydrazinocurcumin Encapsuled nanoparticles “re-educate” tumor-associated macrophages and exhibit anti-tumor effects on breast cancer following STAT3 suppression. PLoS One 2013;8:e65896
  • McGreal EP, Miller JL, Gordon S. Ligand recognition by antigen-presenting cell C-type lectin receptors. Curr Opin Immunol 2005;17:18–24
  • Mantovani A, Sozzani S, Locati M, et al. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 2002;23:549–55
  • Kelly C, Jefferies C, Cryan S-A. Targeted liposomal drug delivery to monocytes and macrophages. J Drug Deliv 2011;2011:727241
  • Zhu S, Niu M, O’Mary H, Cui Z. Targeting of tumor-associated macrophages made possible by peg-sheddable, mannose-modified nanoparticles. Mol Pharm 2013;10:3525–30
  • Niu M, Naguib YW, Aldayel AM, et al. Biodistribution and in vivo activities of tumor-associated macrophage-targeting nanoparticles incorporated with doxorubicin. Mol Pharm 2014;11:4425–36
  • Ortega RA, Barham WJ, Kumar B, et al. Biocompatible mannosylated endosomal-escape nanoparticles enhance selective delivery of short nucleotide sequences to tumor associated macrophages. Nanoscale 2015;7:500–10
  • Sánchez-Martín D, Cuesta AM, Fogal V, et al. The multicompartmental p32/gClqR as a new target for antibody-based tumor targeting strategies. J Biol Chem 2011;286:5197–203
  • Sigalov AB. A novel ligand-independent peptide inhibitor of TREM-1 suppresses tumor growth in human lung cancer xenografts and prolongs survival of mice with lipopolysaccharide-induced septic shock. Int Immunopharmacol 2014;21:208–19
  • Liao D, Liu Z, Wrasidlo WJ, et al. Targeted therapeutic remodeling of the tumor microenvironment improves an HER-2 DNA vaccine and prevents recurrence in a murine breast cancer model. Cancer Res 2011;71:5688–96
  • Yokoi K, Godin B, Oborn CJ, et al. Porous silicon nanocarriers for dual targeting tumor associated endothelial cells and macrophages in stroma of orthotopic human pancreatic cancers. Cancer Lett 2013;334:319–27
  • Fridlender ZG, Sun J, Mishalian I, et al. Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils. PLoS One 2012;7:e31524
  • Ohkura N, Hamaguchi M, Sakaguchi S. FOXP3 + regulatory T cells: control of FOXP3 expression by pharmacological agents. Trends Pharmacol Sci 2011;32:158–66
  • Shevach EM. Application of IL-2 therapy to target T regulatory cell function. Trends Immunol 2012;33:626–32
  • Roth F, De La Fuente AC, Vella JL, et al. Aptamer-mediated blockade of IL4Rα triggers apoptosis of MDSCs and limits tumor progression. Cancer Res 2012;72:1373–83
  • Gessner A, Röllinghoff M. Biologic functions and signaling of the interleukin-4 receptor complexes. Immunobiology 2000;201:285–307
  • Movahedi K, Laoui D, Gysemans C, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res 2010;70:5728–39
  • Ries CH, Cannarile MA, Hoves S, et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014;25:846–59
  • Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013;23:249–62
  • Van Rooijen N. The liposome-mediated macrophage “suicide” technique. J Immunol Methods 1989;124:1–6
  • Gazzaniga S, Bravo AI, Guglielmotti A, et al. Targeting tumor-associated macrophages and inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J Invest Dermatol 2007;127:2031–41
  • Hiraoka K, Zenmyo M, Watari K, et al. Inhibition of bone and muscle metastases of lung cancer cells by a decrease in the number of monocytes/macrophages. Cancer Sci 2008;99:1595–602
  • Kimura YN, Watari K, Fotovati A, et al. Inflammatory stimuli from macrophages and cancer cells synergistically promote tumor growth and angiogenesis. Cancer Sci 2007;98:2009–18
  • Miselis NR, Wu ZJ, Van Rooijen N, Kane AB. Targeting tumor-associated macrophages in an orthotopic murine model of diffuse malignant mesothelioma. Mol Cancer Ther 2008;7:788–99
  • Zeisberger SM, Odermatt B, Marty C, et al. Clodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approach. Br J Cancer 2006;95:272–81
  • Sousa S, Auriola S, Mönkkönen J, Määttä J. Liposome encapsulated zoledronate favours M1-like behaviour in murine macrophages cultured with soluble factors from breast cancer cells. BMC Cancer 2015;15:4
  • Zhan X, Jia L, Niu Y, et al. Targeted depletion of tumour-associated macrophages by an alendronate–glucomannan conjugate for cancer immunotherapy. Biomaterials 2014;35:10046–57
  • Vinogradov S, Warren G, Wei X. Macrophages associated with tumors as potential targets and therapeutic intermediates. Nanomedicine (Lond) 2014;9:695–707
  • Colombo N, Peccatori F, Paganin C, et al. Anti-tumor and immunomodulatory activity of intraperitoneal IFN-gamma in ovarian carcinoma patients with minimal residual tumor after chemotherapy. Int J Cancer 1992;51:42–6
  • Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011;331:1612–16
  • Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 2013;19:1264–72
  • Ugel S, Delpozzo F, Desantis G, et al. Therapeutic targeting of myeloid-derived suppressor cells. Curr Opin Pharmacol 2009;9:470–81
  • Vasquez-Dunddel D, Pan F, Zeng Q, et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J Clin Invest 2013;123:1580–9
  • Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther 2010;18:1606–14
  • Vincent J, Mignot G, Chalmin F, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 2010;70:3052–61
  • Kodumudi KN, Woan K, Gilvary DL, et al. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res 2010;16:4583–94
  • Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1 + myeloid cells. J Immunol 2001;166:5398–406
  • Kusmartsev S, Gabrilovich DI. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukoc Biol 2003;74:186–96
  • Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 2006;203:2691–702
  • De Santo C, Serafini P, Marigo I, et al. Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc Natl Acad Sci USA 2005;102:4185–90
  • Molon B, Ugel S, Del Pozzo F, et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 2011;208:1949–62
  • Fernández A, Mesa C, Marigo I, et al. Inhibition of tumor-induced myeloid-derived suppressor cell function by a nanoparticulated adjuvant. J Immunol 2011;186:264–74
  • Estevez F, Carr A, Solorzano L, et al. Enhancement of the immune response to poorly immunogenic gangliosides after incorporation into very small size proteoliposomes (VSSP). Vaccine 1999;18:190–7
  • Ramírez BS, Pestana ES, Hidalgo GG, et al. Active antimetastatic immunotherapy in Lewis lung carcinoma with self EGFR extracellular domain protein in VSSP adjuvant. Int J Cancer 2006;119:2190–9
  • Torréns I, Mendoza O, Batte A, et al. Immunotherapy with CTL peptide and VSSP eradicated established human papillomavirus (HPV) type 16 E7-expressing tumors. Vaccine 2005;23:5768–74
  • Fernández A, Oliver L, Rábade-Chediak M, et al. Ganglioside containing nano-proteoliposome dampens myeloid-derived suppressor cells function and ability to cross-present tumor antigens: a new approach to recover CTL function on tumor bearing individuals. J Immunother Cancer 2013;1:P225
  • Thomas SN, Vokali E, Lund AW, et al. Targeting the tumor-draining lymph node with adjuvanted nanoparticles reshapes the anti-tumor immune response. Biomaterials 2014;35:814–24
  • Bekes EM, Schweighofer B, Kupriyanova TA, et al. Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation. Am J Pathol 2011;179:1455–70
  • Fridlender ZG, Sun J, Kim S, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009;16:183–94
  • Mishalian I, Bayuh R, Levy L, et al. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol Immunother 2013;62:1745–56
  • Rech AJ, Mick R, Martin S, et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med 2012;4:134ra62
  • Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol 2007;25:171–92
  • Klyushnenkova EN, Riabov VB, Kouiavskaia DV, et al. Breaking immune tolerance by targeting CD25 + regulatory T cells is essential for the anti-tumor effect of the CTLA-4 blockade in an HLA-DR transgenic mouse model of prostate cancer. Prostate 2014;74:1423–32
  • Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012;18:1254–61
  • Valzasina B, Guiducci C, Dislich H, et al. Triggering of OX40 (CD134) on CD4(+)CD25 + T cells blocks their inhibitory activity: a novel regulatory role for OX40 and its comparison with GITR. Blood 2005;105:2845–51
  • Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol 2014;27:1–7
  • Wing K, Onishi Y, Prieto-Martin P, et al. CTLA-4 control over Foxp3 + regulatory T cell function. Science 2008;322:271–5
  • Zhou Q, Xiao H, Liu Y, et al. Blockade of programmed death-1 pathway rescues the effector function of tumor-infiltrating T cells and enhances the antitumor efficacy of lentivector immunization. J Immunol 2010;185:5082–92
  • Movahedi K, Schoonooghe S, Laoui D, et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res 2012;72:4165–77
  • Zhu S, Niu M, O’Mary H, Cui Z. Targeting of tumor-associated macrophages made possible by PEG-sheddable, mannose-modified nanoparticles. Mol Pharm 2013;10:3525–30
  • Yokoi K, Godin B, Oborn CJ, et al. Porous silicon nanocarriers for dual targeting tumor associated endothelial cells and macrophages in stroma of orthotopic human pancreatic cancers. Cancer Lett 2013;334:319–27

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.