Nanovaccines for remodeling the suppressive tumor microenvironment: New horizons in cancer immunotherapy
Kai Shi, Matthew Haynes, Leaf Huang
Nanovaccines for remodeling the suppressive tumor microenvironment: New horizons in cancer immunotherapy
Despite limited successes in clinical development, therapeutic cancer vaccines have experienced resurgence in recent years due to an enhanced emphasis upon co-mitigating factors underlying immune response. Specifically, reversing the immune-suppressive effects of the tumor microenvironment, mediated by a variety of cellular and molecular signaling mechanisms, has become fundamental toward enhancing therapeutic efficacy. Therein, our lab has implemented various nano-vaccines based on the lipid-coated calcium phosphate platform for combined immunotherapy, in which antigenic, epitope-associated peptides as well as immune-suppression inhibitors can be co-delivered, often functioning through the same formulation. In probing the mechanism of action of such systems in vitro andin vivo, an improved effect synergy can be elucidated, inspiring future preclinical efforts and hope for clinical success.
vaccine / nanoparticle / tumor / immunotherapy / microenvironment
[1] |
Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nature Reviews. Cancer, 2012, 12(4): 237–251
CrossRef
Google scholar
|
[2] |
Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature, 2011, 480(7378): 480–489
CrossRef
Google scholar
|
[3] |
Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nature Reviews. Cancer, 2012, 12(4): 265–277
CrossRef
Google scholar
|
[4] |
Jeanbart L, Swartz M A. Engineering opportunities in cancer immunotherapy. Proceedings of the National Academy of Sciences of the United States of America, 2015, 112(47): 14467–14472
CrossRef
Google scholar
|
[5] |
Anguille S, Smits E L, Lion E, van Tendeloo V F, Berneman Z N. Clinical use of dendritic cells for cancer therapy. Lancet Oncology, 2014, 15(7): e257–e267
CrossRef
Google scholar
|
[6] |
Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, Thatcher N, Wagstaff J, Zielinski C, Faulkner I,
CrossRef
Google scholar
|
[7] |
Bloy N, Pol J, Aranda F, Eggermont A, Cremer I, Fridman W H, Fucikova J, Galon J, Tartour E, Spisek R,
CrossRef
Google scholar
|
[8] |
Lesterhuis W J, de Vries I J, Adema G J, Punt C J. Dendritic cell-based vaccines in cancer immunotherapy: An update on clinical and immunological results. Annals of Oncology, 2004, 15(Suppl 4): 145–151
|
[9] |
Smith J D, Morton L D, Ulery B D. Nanoparticles as synthetic vaccines. Current Opinion in Biotechnology, 2015, 34: 217–224
CrossRef
Google scholar
|
[10] |
Yang L, Li W, Kirberger M, Liao W, Ren J. Design of nanomaterial based systems for novel vaccine development. Biomaterials Science, 2016, 4(5): 785–802
CrossRef
Google scholar
|
[11] |
Schwendener R A. Liposomes as vaccine delivery systems: A review of the recent advances. Therapeutic Advances in Vaccines, 2014, 2(6): 159–182
CrossRef
Google scholar
|
[12] |
Silva J M, Videira M, Gaspar R, Preat V, Florindo H F. Immune system targeting by biodegradable nanoparticles for cancer vaccines. Journal of Controlled Release, 2013, 168(2): 179–199
CrossRef
Google scholar
|
[13] |
Quail D F, Joyce J A. Microenvironmental regulation of tumor progression and metastasis. Nature Medicine, 2013, 19(11): 1423–1437
CrossRef
Google scholar
|
[14] |
Motz G T, Coukos G. Deciphering and reversing tumor immune suppression. Immunity, 2013, 39(1): 61–73
CrossRef
Google scholar
|
[15] |
Munn D H, Bronte V. Immune suppressive mechanisms in the tumor microenvironment. Current Opinion in Immunology, 2016, 39: 1–6
CrossRef
Google scholar
|
[16] |
Gajewski T F, Schreiber H, Fu Y X. Innate and adaptive immune cells in the tumor microenvironment. Nature Immunology, 2013, 14(10): 1014–1022
CrossRef
Google scholar
|
[17] |
Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nature Reviews. Cancer, 2005, 5(4): 263–274
CrossRef
Google scholar
|
[18] |
Rabinovich G A, Gabrilovich D, Sotomayor E M. Immunosuppressive strategies that are mediated by tumor cells. Annual Review of Immunology, 2007, 25(1): 267–296
CrossRef
Google scholar
|
[19] |
Shiao S L, Ganesan A P, Rugo H S, Coussens L M. Immune microenvironments in solid tumors: New targets for therapy. Genes & Development, 2011, 25(24): 2559–2572
CrossRef
Google scholar
|
[20] |
McAllister S S, Weinberg R A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nature Cell Biology, 2014, 16(8): 717–727
CrossRef
Google scholar
|
[21] |
Pandiyan P, Zheng L, Ishihara S, Reed J, Lenardo M J. CD4+CD25+Foxp3+ regulatory T-cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nature Immunology, 2007, 8(12): 1353–1362
CrossRef
Google scholar
|
[22] |
Chakraborty N G, Chattopadhyay S, Mehrotra S, Chhabra A, Mukherji B. Regulatory T-cell response and tumor vaccine-induced cytotoxic T lymphocytes in human melanoma. Human Immunology, 2004, 65(8): 794–802
CrossRef
Google scholar
|
[23] |
Huang Y H, Zozulya A L, Weidenfeller C, Schwab N, Wiendl H. T cell suppression by naturally occurring HLA-G-expressing regulatory CD4+ T cells is IL-10-dependent and reversible. Journal of Leukocyte Biology, 2009, 86(2): 273–281
CrossRef
Google scholar
|
[24] |
Huang B, Pan P Y, Li Q, Sato A I, Levy D E, Bromberg J, Divino C M, Chen S H. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell energy in tumor-bearing host. Cancer Research, 2006, 66(2): 1123–1131
CrossRef
Google scholar
|
[25] |
Zea A H, Rodriguez P C, Atkins M B, Hernandez C, Signoretti S, Zabaleta J, McDermott D, Quiceno D, Youmans A, O’Neill A,
|
[26] |
Gabrilovich D I, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Reviews. Immunology, 2009, 9(3): 162–174
CrossRef
Google scholar
|
[27] |
Wu A A, Drake V, Huang H S, Chiu S, Zheng L. Reprogramming the tumor microenvironment: Tumor-induced immunosuppressive factors paralyze T-cells. OncoImmunology, 2015, 4(7): e1016700
CrossRef
Google scholar
|
[28] |
Qin Z, Noffz G, Mohaupt M, Blankenstein T. Interleukin-10 prevents dendritic cell accumulation and vaccination with granulocyte-macrophage colony-stimulating factor gene-modified tumor cells. Journal of Immunology (Baltimore, Md.: 1950), 1997, 159(2): 770–776
|
[29] |
Peng Y, Laouar Y, Li M O, Green E A, Flavell R A. TGF-beta regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T-cells responsible for protection against diabetes. Proceedings of the National Academy of Sciences of the United States of America, 2004, 101(13): 4572–4577
CrossRef
Google scholar
|
[30] |
Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Research, 2006, 66(11): 5527–5536
CrossRef
Google scholar
|
[31] |
Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, Boon T, van den Eynde B J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nature Medicine, 2003, 9(10): 1269–1274
CrossRef
Google scholar
|
[32] |
Munn D H, Sharma M D, Lee J R, Jhaver K G, Johnson T S, Keskin D B, Marshall B, Chandler P, Antonia S J, Burgess R,
CrossRef
Google scholar
|
[33] |
Rohrig U F, Majjigapu S R, Vogel P, Zoete V, Michielin O. Challenges in the discovery of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. Journal of Medicinal Chemistry, 2015, 58(24): 9421–9437
CrossRef
Google scholar
|
[34] |
Saito T, Yokosuka T, Hashimoto-Tane A. Dynamic regulation of T-cell activation and co-stimulation through TCR-microclusters. FEBS Letters, 2010, 584(24): 4865–4871
CrossRef
Google scholar
|
[35] |
Walker L S, Sansom D M. Confusing signals: Recent progress in CTLA-4 biology. Trends in Immunology, 2015, 36(2): 63–70
CrossRef
Google scholar
|
[36] |
Dong H, Strome S E, Salomao D R, Tamura H, Hirano F, Flies D B, Roche P C, Lu J, Zhu G, Tamada K,
|
[37] |
Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nature Medicine, 1999, 5(12): 1365–1369
CrossRef
Google scholar
|
[38] |
Dong H, Chen L. B7-H1 pathway and its role in the evasion of tumor immunity. Journal of Molecular Medicine, 2003, 81(5): 281–287
CrossRef
Google scholar
|
[39] |
Curiel T J, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson K L, Daniel B, Zimmermann M C,
CrossRef
Google scholar
|
[40] |
Webster R M. The immune checkpoint inhibitors: Where are we now? Nature Reviews. Drug Discovery, 2014, 13(12): 883–884
CrossRef
Google scholar
|
[41] |
Juliano R L, Carver K. Cellular uptake and intracellular trafficking of oligonucleotides. Advanced Drug Delivery Reviews, 2015, 87: 35–45
CrossRef
Google scholar
|
[42] |
Midoux P, Pichon C. Lipid-based mRNA vaccine delivery systems. Expert Review of Vaccines, 2015, 14(2): 221–234
CrossRef
Google scholar
|
[43] |
Marceau F, Bawolak M T, Lodge R, Bouthillier J, Gagne-Henley A, Gaudreault R C, Morissette G. Cation trapping by cellular acidic compartments: Beyond the concept of lysosomotropic drugs. Toxicology and Applied Pharmacology, 2012, 259(1): 1–12
CrossRef
Google scholar
|
[44] |
Mansourian M, Badiee A, Jalali S A, Shariat S, Yazdani M, Amin M, Jaafari M R. Effective induction of anti-tumor immunity using p5 HER-2/neu derived peptide encapsulated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunology Letters, 2014, 162(1): 87–93
CrossRef
Google scholar
|
[45] |
Horiuchi Y, Takagi A, Uchida T, Akatsuka T. Targeting cryptic epitope with modified antigen coupled to the surface of liposomes induces strong antitumor CD8 T-cell immune responses in vivo. Oncology Reports, 2015, 34(6): 2827–2836
|
[46] |
Kojima N, Biao L, Nakayama T, Ishii M, Ikehara Y, Tsujimura K. Oligomannose-coated liposomes as a therapeutic antigen-delivery and an adjuvant vehicle for induction of in vivo tumor immunity. Journal of Controlled Release, 2008, 129(1): 26–32
CrossRef
Google scholar
|
[47] |
Ikehara Y, Shiuchi N, Kabata-Ikehara S, Nakanishi H, Yokoyama N, Takagi H, Nagata T, Koide Y, Kuzushima K, Takahashi T,
CrossRef
Google scholar
|
[48] |
Choi D H, Kim K S, Yang S H, Chung D H, Song B, Sprent J, Cho J H, Sung Y C. Dendritic cell internalization of alpha-galactosylceramide from CD8 T-cells induces potent antitumor CD8 T-cell responses. Cancer Research, 2011, 71(24): 7442–7451
CrossRef
Google scholar
|
[49] |
Neumann S, Young K, Compton B, Anderson R, Painter G, Hook S. Synthetic TRP2 long-peptide and alpha-galactosylceramide formulated into cationic liposomes elicit CD8+ T-cell responses and prevent tumour progression. Vaccine, 2015, 33(43): 5838–5844
CrossRef
Google scholar
|
[50] |
Xu Z, Ramishetti S, Tseng Y C, Guo S, Wang Y, Huang L. Multifunctional nanoparticles co-delivering Trp2 peptide and CpG adjuvant induce potent cytotoxic T-lymphocyte response against melanoma and its lung metastasis. Journal of Controlled Release, 2013, 172(1): 259–265
CrossRef
Google scholar
|
[51] |
Xu Z, Wang Y, Zhang L, Huang L. Nanoparticle-delivered transforming growth factor-beta siRNA enhances vaccination against advanced melanoma by modifying tumor microenvironment. ACS Nano, 2014, 8(4): 3636–3645
CrossRef
Google scholar
|
[52] |
Vangasseri D P, Han S J, Huang L. Lipid-protamine-DNA-mediated antigen delivery. Current Drug Delivery, 2005, 2(4): 401–406
CrossRef
Google scholar
|
[53] |
Dileo J, Banerjee R, Whitmore M, Nayak J V, Falo L D Jr, Huang L. Lipid-protamine-DNA-mediated antigen delivery to antigen-presenting cells results in enhanced anti-tumor immune responses. Molecular Therapy, 2003, 7(5): 640–648
CrossRef
Google scholar
|
[54] |
Miura N, Shaheen S M, Akita H, Nakamura T, Harashima H. A KALA-modified lipid nanoparticle containing CpG-free plasmid DNA as a potential DNA vaccine carrier for antigen presentation and as an immune-stimulative adjuvant. Nucleic Acids Research, 2015, 43(3): 1317–1331
CrossRef
Google scholar
|
[55] |
Shaheen S M, Akita H, Nakamura T, Takayama S, Futaki S, Yamashita A, Katoono R, Yui N, Harashima H. KALA-modified multi-layered nanoparticles as gene carriers for MHC class-I mediated antigen presentation for a DNA vaccine. Biomaterials, 2011, 32(26): 6342–6350
CrossRef
Google scholar
|
[56] |
Zhuang X, Wu T, Zhao Y, Hu X, Bao Y, Guo Y, Song Q, Li G, Tan S, Zhang Z. Lipid-enveloped zinc phosphate hybrid nanoparticles for codelivery of H-2K(b) and H-2D(b)-restricted antigenic peptides and monophosphoryl lipid A to induce antitumor immunity against melanoma. Journal of Controlled Release, 2016, 228: 26–37
CrossRef
Google scholar
|
[57] |
Ruiz-de-Angulo A, Zabaleta A, Gomez-Vallejo V, Llop J, Mareque-Rivas J C. Microdosed Lipid-coated (67)Ga-magnetite enhances antigen-specific immunity by image tracked delivery of antigen and CpG to lymph nodes. ACS Nano, 2016, 10(1): 1602–1618
CrossRef
Google scholar
|
[58] |
Zhang Z, Tongchusak S, Mizukami Y, Kang Y J, Ioji T, Touma M, Reinhold B, Keskin D B, Reinherz E L, Sasada T. Induction of anti-tumor cytotoxic T-cell responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials, 2011, 32(14): 3666–3678
CrossRef
Google scholar
|
[59] |
Ma W, Chen M, Kaushal S, McElroy M, Zhang Y, Ozkan C, Bouvet M, Kruse C, Grotjahn D, Ichim T,
CrossRef
Google scholar
|
[60] |
Heo M B, Lim Y T. Programmed nanoparticles for combined immunomodulation, antigen presentation and tracking of immunotherapeutic cells. Biomaterials, 2014, 35(1): 590–600
CrossRef
Google scholar
|
[61] |
Guo Y, Wang D, Song Q, Wu T, Zhuang X, Bao Y, Kong M, Qi Y, Tan S, Zhang Z. Erythrocyte membrane-enveloped polymeric nanoparticles as nanovaccine for induction of antitumor immunity against melanoma. ACS Nano, 2015, 9(7): 6918–6933
CrossRef
Google scholar
|
[62] |
Silva J M, Zupancic E, Vandermeulen G, Oliveira V G, Salgado A, Videira M, Gaspar M, Graca L, Preat V, Florindo H F. In vivo delivery of peptides and Toll-like receptor ligands by mannose-functionalized polymeric nanoparticles induces prophylactic and therapeutic anti-tumor immune responses in a melanoma model. Journal of Controlled Release, 2015, 198: 91–103
CrossRef
Google scholar
|
[63] |
Cui L, Osada K, Imaizumi A, Kataoka K, Nakano K. Feasibility of a subcutaneously administered block/homo-mixed polyplex micelle as a carrier for DNA vaccination in a mouse tumor model. Journal of Controlled Release, 2015, 206: 220–231
CrossRef
Google scholar
|
[64] |
Furugaki K, Cui L, Kunisawa Y, Osada K, Shinkai K, Tanaka M, Kataoka K, Nakano K. Intraperitoneal administration of a tumor-associated antigen SART3, CD40L, and GM-CSF gene-loaded polyplex micelle elicits a vaccine effect in mouse tumor models. PLoS One, 2014, 9(7): e101854
CrossRef
Google scholar
|
[65] |
Luo Z, Wang C, Yi H, Li P, Pan H, Liu L, Cai L, Ma Y. Nanovaccine loaded with poly I:C and STAT3 siRNA robustly elicits anti-tumor immune responses through modulating tumor-associated dendritic cells in vivo. Biomaterials, 2015, 38: 50–60
CrossRef
Google scholar
|
[66] |
Luo Z, Li P, Deng J, Gao N, Zhang Y, Pan H, Liu L, Wang C, Cai L, Ma Y. Cationic polypeptide micelle-based antigen delivery system: A simple and robust adjuvant to improve vaccine efficacy. Journal of Controlled Release, 2013, 170(2): 259–267
CrossRef
Google scholar
|
[67] |
Li J, Chen Y C, Tseng Y C, Mozumdar S, Huang L. Biodegradable calcium phosphate nanoparticle with lipid coating for systemic siRNA delivery. Journal of Controlled Release, 2010, 142(3): 416–421
CrossRef
Google scholar
|
[68] |
Li J, Yang Y, Huang L. Calcium phosphate nanoparticles with an asymmetric lipid bilayer coating for siRNA delivery to the tumor. Journal of Controlled Release, 2012, 158(1): 108–114
CrossRef
Google scholar
|
[69] |
Haynes M T, Huang L. Lipid-coated calcium phosphate nanoparticles for nonviral gene therapy. Advances in Genetics Incorporating Molecular Genetic Medicine, 2014, 88: 205–229
|
[70] |
Curran M A, Montalvo W, Yagita H, Allison J P. PD-1 and CTLA-4 combination blockade expands infiltrating T-cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proceedings of the National Academy of Sciences of the United States of America, 2010, 107(9): 4275–4280
CrossRef
Google scholar
|
[71] |
Fridlender Z G, Buchlis G, Kapoor V, Cheng G, Sun J, Singhal S, Crisanti M C, Wang L C, Heitjan D, Snyder L A, et al. CCL2 blockade augments cancer immunotherapy. Cancer Research, 2010, 70(1): 109–118
CrossRef
Google scholar
|
[72] |
Kusmartsev S, Cheng F, Yu B, Nefedova Y, Sotomayor E, Lush R, Gabrilovich D. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Research, 2003, 63(15): 4441–4449
|
[73] |
Zeng J, Cai S, Yi Y, He Y, Wang Z, Jiang G, Li X, Du J. Prevention of spontaneous tumor development in a ret transgenic mouse model by ret peptide vaccination with indoleamine 2,3-dioxygenase inhibitor 1-methyl tryptophan. Cancer Research, 2009, 69(9): 3963–3970
CrossRef
Google scholar
|
[74] |
Liao D, Liu Z, Wrasidlo W J, Luo Y, Nguyen G, Chen T, Xiang R, Reisfeld R A. Targeted therapeutic remodeling of the tumor microenvironment improves an HER-2 DNA vaccine and prevents recurrence in a murine breast cancer model. Cancer Research, 2011, 71(17): 5688–5696
CrossRef
Google scholar
|
[75] |
Chono S, Li S D, Conwell C C, Huang L. An efficient and low immunostimulatory nanoparticle formulation for systemic siRNA delivery to the tumor. Journal of Controlled Release, 2008, 131(1): 64–69
CrossRef
Google scholar
|
[76] |
Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nature Reviews. Immunology, 2007, 7(1): 41–51
CrossRef
Google scholar
|
[77] |
Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nature Reviews. Cancer, 2014, 14(11): 736–746
CrossRef
Google scholar
|
[78] |
Furtek S L, Backos D S, Matheson C J, Reigan P. Strategies and approaches of targeting STAT3 for cancer treatment. ACS Chemical Biology, 2016, 11(2): 308–318
CrossRef
Google scholar
|
[79] |
Al Zaid Siddiquee K, Turkson J. STAT3 as a target for inducing apoptosis in solid and hematological tumors. Cell Research, 2008, 18(2): 254–267
CrossRef
Google scholar
|
[80] |
Singh M, Ramos I, Asafu-Adjei D, Quispe-Tintaya W, Chandra D, Jahangir A, Zang X, Aggarwal B B, Gravekamp C. Curcumin improves the therapeutic efficacy of Listeria(at)-Mage-b vaccine in correlation with improved T-cell responses in blood of a triple-negative breast cancer model 4T1. Cancer Medicine, 2013, 2(4): 571–582
CrossRef
Google scholar
|
[81] |
Molavi O, Ma Z, Hamdy S, Lai R, Lavasanifar A, Samuel J. Synergistic antitumor effects of CpG oligodeoxynucleotide and STAT3 inhibitory agent JSI-124 in a mouse melanoma tumor model. Immunology and Cell Biology, 2008, 86(6): 506–514
CrossRef
Google scholar
|
[82] |
Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nature Reviews. Cancer, 2009, 9(11): 798–809
CrossRef
Google scholar
|
[83] |
Lu Y, Miao L, Wang Y, Xu Z, Zhao Y, Shen Y, Xiang G, Huang L. Curcumin micelles remodel tumor microenvironment and enhance vaccine activity in an advanced melanoma model. Molecular Therapy, 2016, 24(2): 364–374
CrossRef
Google scholar
|
[84] |
Nagaraj S, Youn J I, Weber H, Iclozan C, Lu L, Cotter M J, Meyer C, Becerra C R, Fishman M, Antonia S,
CrossRef
Google scholar
|
[85] |
Zhao Y, Huo M, Xu Z, Wang Y, Huang L. Nanoparticle delivery of CDDO-Me remodels the tumor microenvironment and enhances vaccine therapy for melanoma. Biomaterials, 2015, 68: 54–66
CrossRef
Google scholar
|
/
〈 | 〉 |