We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Dendritic cell-based vaccines: clinical applications in breast cancer

    Lucia Gelao

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Carmen Criscitiello

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Angela Esposito

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ,
    Michele De Laurentiis

    Department of Breast Oncology, National Cancer Institute “Fondazione Pascale”, Naples, Italy

    ,
    Luca Fumagalli

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ,
    Marzia Adelia Locatelli

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ,
    Ida Minchella

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    ,
    Michele Santangelo

    Department of Advanced Medical Biosciences, Operative Unit of General Surgery & Transplants, University of Naples Federico II, Naples, Italy

    ,
    Sabino De Placido

    Department of Endocrinology & Molecular and Clinical Oncology, University of Naples Federico II, Naples, Italy

    ,
    Aron Goldhirsch

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    &
    Giuseppe Curigliano

    * Author for correspondence:

    E-mail Address: giuseppe.curigliano@ieo.it

    Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy

    Published Online:https://doi.org/10.2217/imt.13.169

    Abstract: 

    Recent evidence suggests that the immune system is involved in the carcinogenesis process and the antitumor immune responses impact the clinical outcome, thus emphasizing the concept of cancer immune surveillance. In this context, dendritic cells (DCs) seem to play a crucial role, as they are the most potent antigen-presenting cells (APCs) and are able to stimulate naive T lymphocytes and to generate memory T lymphocytes. Immunotherapy with DC-based vaccines is a very attractive approach to treat cancer, offering the potential for high tumor-specific cytotoxicity. Although breast cancer (BC) is traditionally considered a poorly immunogenic tumor, increasing numbers of both preclinical and clinical studies demonstrate that vaccination with DCs is capable of inducing an antitumor-specific response, while being well tolerated and safe. However, clinical objective responses are still disappointing and many reasons may explain the difficulty of developing effective DC-based therapies for BC. In this review, we discuss the characteristics of DCs, and the major clinical indications for DC-based immunotherapy in BC with related drawbacks.

    Papers of special note have highlighted as:

    •of interest

    ••of considerable interest

    References

    • 1 Adam JK, Odhav B, Bhoola KD. Immune responses in cancer. Pharmacol. Ther. 99(1), 113–132 (2003).
    • 2 Andre F, Dieci MV, Dubsky P et al. Molecular pathways: involvement of immune pathways in the therapeutic response and outcome in breast cancer. Clin. Cancer Res. 19(1), 28–33 (2013).
    • 3 Yaguchi T, Sumimoto H, Kudo-Saito C. The mechanisms of cancer immunoescape and development of overcoming strategies. Int. J. Hematol. 93(3), 294–300 (2011).
    • 4 Zheng J, Liu Q, Yang J et al. Co-culture of apoptotic breast cancer cells with immature dendritic cells: a novel approach for DC-based vaccination in breast cancer. Braz. J. Med. Biol. Res. 45(6), 510–515 (2012).
    • 5 Penn I. Tumors of the immunocompromised patient. Annu. Rev. Med. 39, 63–73 (1998).
    • 6 Chen Q, Zhang XH, Massague J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell 20(4), 538–549 (2011).
    • 7 DeNardo DG, Barreto JB, Andreu P et al. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16(2), 91–102 (2009).
    • 8 Bidwell BN, Slaney CY, Withana NP et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18(8), 1224–1231 (2012).
    • 9 Mittendorf EA, Peoples GE, Singletary SE. Breast cancer vaccines: promise for the future or pipe dream? Cancer 110(8), 1677–1686 (2007).
    • 10 Topalian SL, Hodi FS, Brahmer JR et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366(26), 2443–2454 (2012). • The activity and safety of BMS-936558, an antibody that specifically blocks PD-1, was assessed in 296 patients with advanced melanoma, non-small-cell lung cancer, castration-resistant prostate cancer, renal cell cancer or colorectal cancer. Objective responses were recorded in approximately one in four to one in five patients with non-small-cell lung cancer, melanoma, or renal-cell cancer.
    • 11 Eggermont AM, Kroemer G, Zitvogel L. Immunotherapy and the concept of a clinical cure. Eur. J. Cancer 49(14), 2965–2967 (2013).
    • 12 Palucka K, Banchereau J. Human dendritic cell subsets in vaccination. Curr. Opin. Immunol. 25(3), 396–402 (2013).
    • 13 Criscitiello C, Curigliano G. Immunotherapeutics for breast cancer. Curr. Opin. Oncol. 25(6), 602–608 (2013). •• Broad summary of the literature illustrating the immune approaches that should be evaluated in the treatment of breast cancer (BC).
    • 14 Dunn GP, Bruce AT, Ikeda H et al. Cancer immunoediting: from immunosurveillance to tumour escape. Nat. Immunol. 3(11), 991–998 (2002).
    • 15 Curigliano G. Immunity and autoimmunity: revising the concepts of response to breast cancer. Breast 20(3), 71–74 (2011).
    • 16 Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature 411(6835), 380–384 (2001).
    • 17 Turnis ME, Rooney CM. Enhancement of dendritic cells as vaccines for cancer. Immunotherapy 2(6), 847–862 (2010). •• Detailed review explaining the use of dendritic cells (DCs) in immunotherapy.
    • 18 Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu. Rev. Immunol. 21, 685–711(2003).
    • 19 Berzofsky JA, Terabe M, Oh S et al. Progress on new vaccine strategies for the immunotherapy and prevention of cancer. J. Clin. Invest. 113(11), 1515–1525 (2004).
    • 20 Nencioni A, Grünebach F, Schmidt SM et al. The use of dendritic cells in cancer immunotherapy. Crit. Rev. Oncol. Hematol. 65(3), 191–199 (2008). • Includes a complete description of DC biology and methods of vaccination, as well as discussion about clinical application.
    • 21 Ma Y, Shurin GV, Peiyuan Z et al. Dendritic cells in the cancer microenvironment. J. Cancer 4(1), 36–44 (2013).
    • 22 Engelhardt JJ, Boldajipour B, Beemiller P et al. Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells. Cancer Cell 21(3), 402–417 (2012).
    • 23 Mu CY, Huang JA, Chen Y et al. High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med. Oncol. 28 (3), 682–688 (2011).
    • 24 Eisenbarth SC, Williams A, Colegio OR et al. NLRP10 is a NOD-like receptor essential to initiate adaptive immunity by dendritic cells. Nature 484(7395), 510–513 (2012).
    • 25 Satthaporn S, Robins A, Vassanasiri W et al. Dendritic cells are dysfunctional in patients with operable breast cancer. Cancer Immunol. Immunother. 53(6), 510–518 (2004) • Indicates the impaired immunological function of DCs of breast cancer patients compared with normal subjects.
    • 26 Banchereau J, Schuler-Thurner B, Palucka AK et al. Dendritic cells as vectors for therapy. Cell 106(3), 271–274 (2001).
    • 27 Lee AW, Truong T, Bickham K et al. A clinical grade cocktail of cytokines and PGE2 results in uniform maturation of human monocyte- derived dendritic cells: implications for immunotherapy. Vaccine 20 (4), 8–22 (2002).
    • 28 Ballestrero A, Boy D, Moran E et al. Immunotherapy with dendritic cells for cancer. Adv. Drug Deliv. Rev. 60(2), 173–183 (2008).
    • 29 Gogas H, Polyzos A, Kirkwood J. Immunotherapy for advanced melanoma: fulfilling the promise. Cancer Treat Rev. 39(8), 879–885 (2013).
    • 30 Khattak M, Fisher R, Turajlic S et al. Targeted therapy and immunotherapy in advanced melanoma: an evolving paradigm. Ther. Adv. Med. Oncol. 5(2), 105–118 (2013).
    • 31 Escudier B. Emerging immunotherapies for renal cell carcinoma. Ann. Oncol. 23(8), 35–40 (2012).
    • 32 Rini BI, Stein M, Shannon P et al. Phase 1 dose-escalation trial of tremelimumab plus sunitinib in patients with metastatic renal cell carcinoma. Cancer 117, 758–767 (2011).
    • 33 Pal SK, Hu A, Figlin RA. A new age for vaccine therapy in renal cell carcinoma. Cancer J. 19(4), 365–370 (2013).
    • 34 Cheever MA.. PROVENGE (sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin. Cancer Res. 17, 3520–3526 (2011). •• Sipuleucel-T (PROVENGE®, Dendreon) is the first therapeutic cancer vaccine to be approved by the US FDA.
    • 35 Lissoni P, Vigore L, Ferranti R et al. Circulating dendritic cells in early and advanced cancer patients: diminished percent in the metastatic disease. J. Biol. Regul. Homeost. Agents 13(4), 216–219 (1999).
    • 36 Fields RC, Shimizu K, Mulé JJ. Murine dendritic cells pulsed with whole tumor lysates mediate potent antitumor immune responses in vitro and in vivo. Proc. Natl Acad. Sci. USA 95(16), 9482–9487 (1998).
    • 37 Wright SE. Immunotherapy of breast cancer. Expert Opin. Biol. Ther. 12, 479–490 (2012).
    • 38 Gong J, Avigan D, Chen D et al. Activation of antitumor cytotoxic T lymphocytes by fusions of human DCs and breast carcinoma cells. Proc. Natl Acad. Sci. USA 97(6), 2715–2718 (2000).
    • 39 Neidhardt-berard EM, Berard F, Banchereau J et al. Dendritic cells loaded with killed breast cancer cells induce differentiation of tumor-specific cytotoxic T lymphocytes. Breast Cancer Res. 6(4), 322–328 (2004).
    • 40 Wang B, Zaidi N, He LZ et al. Targeting of the non-mutated tumor antigen HER2/neu to mature dendritic cells induces an integrated immune response that protects against breast cancer in mice. Breast Cancer Res. 14, R39 (2012).
    • 41 Wang L, Xie Y, Ahmed KA et al. Exosomal pMHC-I complex targets T cell-based vaccine to directly stimulate CTL responses leading to antitumor immunity in transgenic FVBneuN and HLA-A2/HER2 mice and eradicating trastuzumab-resistant tumor in athymic nude mice. Breast Cancer Res. Treat. 140(2), 273–284 (2013).
    • 42 Sakai Y, Morrison BJ, Burke JD et al. Vaccination by genetically modified dendritic cells expressing a truncated neu oncogene prevents development of breast cancer in transgenic mice. Cancer Res. 64(21), 8022–8028 (2004).
    • 43 Huang X, Ye D, Thorpe PE. Enhancing the potency of a whole-cell breast cancer vaccine in mice with an antibody-IL-2 immunocytokine that targets exposed phosphatidylserine. Vaccine 29(29–30), 4785–4793 (2011).
    • 44 Brossart P, Wirths S, Stuhler G et al. Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 96(9), 3102–2108 (2000).
    • 45 Avigan D, Vasir B, Gong J et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses. Clin. Cancer Res. 10(14), 4699–4708 (2004).
    • 46 Qi CJ, Ning YL, Han YS et al. Autologous dendritic cell vaccine for estrogen receptor (ER)/progestin receptor (PR) double-negative breast cancer. Cancer Immunol. Immunother. 61(9), 1415–1424 (2012).
    • 47 Baek S, Kim CS, Kim SB et al. Combination therapy of renal cell carcinoma or breast cancer patients with dendritic cell vaccine and IL-2: results from a Phase I/II trial. J. Transl. Med. 20, 9–178 (2011). •• First trial to evaluate the safety and efficacy of the combination of IL-2 and DC vaccine.
    • 48 www.clinicaltrials.gov
    • 49 Cui Y, Yang X, Zhu W et al. Immune response, clinical outcome and safety of dendritic cell vaccine in combination with cytokine-induced killer cell therapy in cancer patients. Oncol. Lett. 6(2), 537–541 (2013).
    • 50 Ren J, Di L, Song G et al. Selections of appropriate regimen of high-dose chemotherapy combined with adoptive cellular therapy with dendritic and cytokine-induced killer cells improved progression-free and overall survival in patients with metastatic breast cancer: reargument of such contentious therapeutic preferences. Clin. Transl. Oncol. 15(10), 780–788. (2013).
    • 51 Finn OJ, Forni G. Prophylactic cancer vaccines. Curr. Opin. Immunol. 14(2), 172–177 (2002).
    • 52 Koski GK, Koldovsky U, Xu S et al. A novel dendritic cell-based immunization approach for the induction of durable Th1-polarized anti-HER-2/neu responses in women with early breast cancer. J. Immunother. 35(1), 54–65 (2012).
    • 53 Czerniecki BJ, Koski GK, Koldovsky U et al . Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res. 67(4), 1842–1852 (2007).
    • 54 Sharma A, Koldovsky U, Xu S et al. HER-2 pulsed dendritic cell vaccine can eliminate HER-2 expression and impact ductal carcinoma in situ. Cancer 118(17), 4354–4362 (2012).
    • 55 Wolchok JD, Hoos A, O’Day S et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009).
    • 56 Galluzzi L, Senovilla L, Vacchelli E et al. Trial watch: dendritic cell-based interventions for cancer therapy. Oncoimmunology 1(7), 1111–1134 (2012).
    • 57 Strasser EF, Eckstein R. Optimization of leukocyte collection and monocyte isolation for dendritic cell culture. Transfus. Med. Rev. 24(2), 130–139 (2010).
    • 58 Dikov MM, Ohm JE, Ray N. Differential roles of vascular endothelial growth factor receptors 1 and 2 in dendritic cell differentiation. J. Immunol. 174, 215–222 (2005).
    • 59 Kawakami Y, Yaguchi T, Sumimoto H et al. Improvement of cancer immunotherapy by combining molecular targeted therapy. Front. Oncol. 3, 136 (2013).
    • 60 Tacken PJ, de Vries IJ, Torensma R et al. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 7, 790–802 (2007).
    • 61 Sancho D, Mourão-Sá D, Joffre OP et al. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J. Clin. Invest. 118, 2098–2110 (2008).
    • 62 Bonifaz L, Bonnyay D, Mahnke K et al. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 196, 1627–1638 (2002).
    • 63 Hawiger D, Inaba K, Dorsett Y et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194, 769–779 (2001).
    • 64 Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat. Rev. Immunol. 4 (10), 762–774 (2004).
    • 65 Zheng X, Koropatnick J, Chen D et al. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int. J. Cancer 132(4), 967–977 (2013).
    • 66 Criscitiello C. Tumor-associated antigens in breast cancer. Breast Care (Basel) 7(4), 262–266 (2012).
    • 67 Van Tendeloo VF, Ponsaerts P, Berneman ZN. mRNA based gene transfer as a tool for gene and cell therapy. Curr. Opin. Mol. Ther. 9, 423–431 (2007).
    • 68 Zhang B, Beeghly-Fadiel A, Long J, Genetic variants associated with breast-cancer risk: comprehensive research synopsis, meta-analysis, and epidemiological evidence. Lancet Oncol. 12(5), 477–488 (2011).
    • 69 Vonderheide RH, LoRusso PM, Khalil M. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin. Cancer Res. 16(13), 3485–3494 (2010).
    • 70 Hodi FS, Butler M, Oble DA. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc. Natl Acad. Sci. USA 105(8), 3005 (2008).
    • 71 Zitvogel L, Apetoh L, Ghiringhelli, F et al. Immunological aspects of cancer chemotherapy. Nat. Rev. Immunol. 8, 59–73 (2008).
    • 72 Ghiringhelli F, Apetoh L, Tesniere A et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta dependent adaptive immunity against tumors. Nat. Med. 15, 1170–1178 (2009).
    • 73 Stagg J, Andre F, Loi S. Immunomodulation via chemotherapy and targeted therapy: a new paradigm in breast cancer therapy? Breast Care (Basel) 7(4), 267–272 (2012). • Discusses the underlying mechanisms of how cytotoxic chemotherapy can stimulate an antitumor immune response and how combinations of traditional agents with new immunotherapeutic therapies may significantly advance our treatment of breast cancer.