Journal List > Chonnam Med J > v.46(1) > 1017944

Lee: The Clinical Impact of the Dendritic Cell-based Cancer Vaccine: the Role in the Inflammatory Tumor Micro-environment

Abstract

As a novel therapeutic module, the dendritic cell (DC) based cancer vaccine has been recognized with great hope in eliminating cancers, including minimal residual cells, without harming normal tissue. A key factor in initiating and operating the immune system against foreign bodies including tumor cells, the DC has been regarded as the next possible breakthrough in new cancer therapy. However, the results of more than 15 years of clinical studies with DC vaccine revealed the difficulties fulfilling this expectation. Evidence has disclosed that the DC activation required for proper tumor-specific effector CD4+ and CD8+ T cell stimulation is inhibited in the micro-environment of cancer. Studies have further reported that DC phenotypes in cancer tissue and draining lymph nodes are mostly immature, which results in regulatory immune responses. Also, the existence of myeloid derived suppressor cells (MDSCs) and tumor associated macrophages (TAMs) adversely affect both DC function and immune suppression in the cancer-environment. In this review, the impact of an inflammatory micro-environment induced by cancer on the effect of DC-based cancer immunotherapy and the possibility of a clinical efficacy improvement are discussed.

Introduction

The role of DC is at the center of the immune system by initiating, progressing and regulating the responses against pathogens including tumors. After the first successful clinical achievement in DC-based immunotherapy trials in follicular lymphoma and melanoma in the mid-1990s,1,2 the DC vaccine has been successfully used to treat patients with melanoma, lymphoma and renal cell carcinoma.3-7 However clinical expectations have not been fulfilled due to an overall clinical response rates of under 10~15%, the usual response rates observed in various types of immunotherapies.6-11 Although the clinical expectation has not been satisfied, the outcomes of many clinical trials with tumor antigen-loaded conventional DCs have provided proof that therapeutic immunity can be elicited.12-14 The clinical data has helped to establish a standard for properly activated DCs with appropriate form and doses of loading antigens. These activated DCs can migrate to the lymph nodes which then initiate and expand tumor-specific CD4+ and CD8+ T cell responses and later induce meaningful therapeutic responses in patients.
Several mechanisms are involved in unsatisfactory antitumor responses of DC vaccine in the clinic. Mechanisms include; the presence of keep leukocytes like MDSCs, TAMs with or without the presence of constitutive p-STAT3 signaling, immunoediting, abnormal tumor vasculature inhibiting effector T cell entry or tumor cell interaction with the stromal environment.15-19 On the other hand, in order to improve the DC vaccine clinical efficacy, it is critical to control the therapeutic DC quality and standardize the vaccine design and protocol. Looking at this very view, several investigators have analyzed DC vaccine problems in their publications.4,6,12,14,20-22 Thus, without further discussing about the DC vaccine quality, cancer or host side hindering factors and the possibility of improving antitumor immune-therapeutic efficacy will be discussed in this review.

DCs in cancer patients

DCs are lymphocytes in the immune system which control overall immunity by interacting with other immune cells, including T cell, B cell and NK cells.6,23-24 DCs themselves are a complicated system consisting of various anatomic localizations, subsets and functions that are correlated with one another. DCs control the immune system, not only in stimulatory but also in regulatory immunity as professional APC.22,24 In cancer tissues or cancer-draining lymph nodes, DCs are found as resting, non-activated and immature cells.25-29 Cancer-induced immunosuppressive milieu generally causes a decrease in the numbers of conventional myeloid DCs in patients.25 In rodent models, immature myeloid DCs promote the expansion of regulatory T cells (Treg) in tumor-draining lymph nodes, which are associated with tumor progression in a TGF-β dependent fashion. Immunosuppressive factors, mostly pro-inflammatory molecules from the cancer microenvironment, target endogenous DCs in patients resulting in dysfunction and impaired development of tumor-specific effector lymphocytes.30-31 Typical inflammatory mediators of tumor-induced DC dysfunctions include; IL-10, TGF-β, VEGF, IL-6 and prostanoids such as PGE2-6.32-35 These mediators are produced from either the cancer itself or the infiltrated host factors including MDSCs and TAMs. In this milieu, DCs are having trouble maturing, expressing the co-stimulatory molecules needed for T cell activation, and producing the cytokines needed to support tumor specific effector T cell activation and survival.36-39 Cancer-related malfunctions of DCs are noted in patients with ovarian, breast, melanoma, renal cell, prostate carcinoma,40-43 and in the blood of head & neck, lung and breast cancer patients.36,44 The major intracellular signaling pathway required for DC activation and final maturation in the immunosuppressive milieu of the cancer micro-environment is STAT3.45 Oncogene or cytokine-induced over-expression of the STAT3 protein in cancer cells up-regulates the expression of several immunosuppressive cytokines, including IL-10 and TGF-β, and suppresses Th1 cell immune responses.15,46-47 STAT3 expression from cancer cells leads to STAT3 production by a variety of leukocytes, including DCs. STAT3 expression in tumor-associated DCs causes reduced expression of co-stimulatory and MHC II molecules, and correlates with an accumulation of immature DCs, which may induce Treg,48 an inhibitor of effector T cell function. Anti-tumor effects of the STAT3 inhibitor, Cucurbitacin I was observed in mice, human cancer cell lines, and in vivo mouse tumor models.49-50 Although dysfunctional tumor-associated DCs may support immune suppression and promote oncogenesis, it may be possible to evoke therapeutic antitumor activity in these DCs by molecularly defined triggers of DC maturation, causing induction of tumor-specific effector T cells.

Inflammatory nature of tumor micro-environment

The development of about 15~20% of malignancies worldwide are known to be related to chronic inflammation, including esophageal, gastric, hepatic, pancreatic, and colorectal cancer.51 Inflammatory mediators produced by the cancer cell can create an inflammatory micro-environment and cause both leukocyte recruitment and angiogenesis.52-54 Also, these inflammatory milieus can help tumor cell survival, motility and chemotaxis. For example, breast cancer cells are known to produce the inflammatory chemokines CCL2 and CCL5, which are poorly expressed in normal breast cells. These chemokines recruit TAMS and inhibit potential antitumor effector T cells.55 In other words, the immunosuppressive tumor micro-environment is created by the inflammatory nature of tumors and an infiltration of assorted leukocytes, in particular MDSCs and TAMs. This infiltration leads to the suppression of the DC-induced effectors, CD4+ and CD8+ T cell responses and the induction of Treg.25

MDSCs in the tumor micro-environment

The mechanisms by which chronic inflammation promotes the onset and development of tumors are classified into non-immunological and immunological ways.56 The non-immune mechanisms include 1) the production of reactive oxygen species which cause DNA mutation, 2) the production of pro-angiogenic factors, like VEGF which promote tumor neo-vascularization, 3) the production of matrix metalloproteases which facilitate invasion and metastasis.57-59 The predominant immune mechanism is the disturbance of myelopoiesis and hemopoiesis, which causes a deficiency in APCs and in dysfunctional cell-mediated antitumor immunity. A key molecule in this deficiency is MDSC.25 In individuals with an established cancer, MDSCs are a major factor in preventing the efficacy of cancer vaccines that require an immune-competent host.60 In most cancer patients and experimental mice tumor settings, the accumulation of MDSCs in the blood, lymph nodes, bone marrow and tumor sites is observed. These cells are known to inhibit both adoptive and innate immunity. MDSC induction and recruitment into the tumor site is mediated by tumor-secreted and host-secreted factors, many of which are pro-inflammatory molecules. Thus it may be said that inflammation promotes the accumulation of MDSCs, which down-regulate immune surveillance and antitumor immunity, thereby facilitating tumor growth.56
Identification of MDSCs in cancer patients and experimental mice were analyzed by the activity in T cell suppression. In mice, MDSCs are characterized as Gr1+CD11b+ expressing cells. Gr1 includes Ly6C, a macrophage marker and Ly6G, a neutrophil marker. CD11b is the characteristic marker of macrohphage.56 In some subsets of MDSCs, several markers are ascribed, including the IL-4 and IL-13 receptor alpha chain (IL-4Ra),61,62 F4/80, a macrophage marker,61,63,64 c-fms (CD115),64 and CD80.65 Among the MDSCs, mononuclear cells are defined as "monocytic" CD11b+Ly6G+/-Ly6Chigh, whereas "granulocytic/neutrophil-like" multilobed nuclei possessing cells are characterized by CD11b+Ly6G+Ly6Clow.63,66,67 Immunosuppresive substances produced from MDSCs include arginase, inducible NO synthase, and/or ROS.68-72 Unlike mice, MDSC characterization in cancer patients is complicated but typically characterized by the phenotype CD11b+CD33+CD34+ CD14-HLA-DR- with various expressions of CD15 and other markers. Recent findings have identified CD14+ HLA-DR-/low as a new MDSC subtype in melanoma and hepatoma patients.73-76 It is known that different tumors induce different subtypes of MDSCs in cancer patients.73,74 Along with heterogeneity characterized by the surface phenotype, internal markers, morphology and suppressive substances in both mice and humans, MDSCs suppressed multiple immune effectors include; inhibition of CD4+ and CD8+ T cell functions,77-80 induction of Treg by secreting TGF-β, IL-10 or arginase,64 interaction with NKT cells to enhance tumor growth by suppressing antitumor immunity.81

Improvement of clinical efficacy of the DC vaccine

Considering the inflammatory tumor micro-environment and dysfunctional DCs with suppressed-immunity in cancer patients, it is not surprising to see recent reports indicating that the cancer vaccine induced tumor-specific T cells is not necessarily associated with the induction of functional cytotoxic T lymphocytes, but instead leading to the undesirable activation and expansion of regulatory T cells.14 Tumor antigen-induced immune responses are weak or ineffective, because unlike infectious pathogens, tumors do not induce the strong enough inflammatory responses for the optimal activation of DCs. Thus, the primary purpose of a cancer vaccine is to overcome this defect by educating DCs with a stronger antigenic signal and providing optimal conditions for the maturation into potent immune-stimulatory APCs.22 In the immunosuppressive milieu in cancer patients, sufficient numbers of properly activated tumor-specific Th1 cells and CTLs are not generated despite ample expression of tumor-associated antigens in cancers. The effects of therapeutic cancer vaccines, including DC based therapy, can be expected to enhance by combination with the methods that overcome the immune-suppression associated with cancer. Such therapies include; 1) administration of STAT3 inhibitors, 2) local or systemic treatment with molecularly defined triggers of DC activation, such as TLR ligands and CD40 agonistic antibodies 3) treatment with monoclonal antibodies that block inhibitory co-stimulation pathways, CTLA-4, and PD-1 4) antibodies that enhance the T cell effector function, including OX40.13 Another therapeutic vaccination protocol can combine improved DC vaccine with chemotherapy to exploit immunogenic chemotherapy regimens.82 Conclusively, correcting the immunological balance in the cancer micro-environment from suppression to a tumor-rejecting condition may be the key factor in succeeding with a DC vaccine clinical trial.

References

1. Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med. 1996. 2:52–58.
crossref
2. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998. 4:328–332.
crossref
3. Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005. 5:296–306.
crossref
4. Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell immunotherapy: mapping the way. Nat Med. 2004. 10:475–480.
crossref
5. Gilboa E. The promise of cancer vaccines. Nat Rev Cancer. 2004. 4:401–411.
crossref
6. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007. 449:419–426.
crossref
7. Nestle FO, Farkas A, Conrad C. Dendritic-cell-based therapeutic vaccination against cancer. Curr Opin Immunol. 2005. 17:163–169.
crossref
8. Pardoll DM. Cancer vaccines. Nat Med. 1998. 4:5 Suppl. 525–531.
crossref
9. Parmiani G, Pilla L, Castelli C, Rivoltini L. Vaccination of patients with solid tumours. Ann Oncol. 2003. 14:817–824.
crossref
10. Steinman RM. Dendritic cells and immune-based therapies. Exp Hematol. 1996. 24:859–862.
11. Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. Int J Cancer. 2001. 94:459–473.
crossref
12. Palucka AK, Ueno H, Fay J, Banchereau J. Dendritic cells: a critical player in cancer therapy? J Immunother. 2008. 31:793–805.
crossref
13. Melief C. Cancer immunotherapy by dendritic cells. Immunity. 2008. 29:372–383.
crossref
14. Kalinski P, Urban J, Narang R, Berk E, Wieckowski E, Muthuswamy R. Dendritic cell-based therapeutic cancer vaccines: what we have and what we need. Future Oncol. 2009. 5:379–390.
crossref
15. Kortylewski M, Yu H. Role of Stat3 in suppressing anti-tumor immunity. Curr Opin Immunol. 2008. 20:228–233.
crossref
16. Koebel C, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007. 450:903–907.
crossref
17. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006. 6:836–848.
crossref
18. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature. 2008. 453:410–414.
crossref
19. Hamzah J, Nelson D, et al. Vascular targeting of anti-CD40 antibodies and IL-2 into autochthonous tumors enhances immunotherapy in mice. J Clin Invest. 2008. 118:1691–1699.
crossref
20. Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P, Svane IM. Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother. 2009. 58:1–14.
crossref
21. Ballestrero A, Boy D, Moran E, Cirmena G, Brossart P, Nencioni A. Immunotherapy with dendritic cells for cancer. Adv Drug Deliv Rev. 2008. 60:173–183.
crossref
22. Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007. 117:1195–1203.
crossref
23. Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991. 9:271–296.
crossref
24. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998. 392:245–252.
crossref
25. Gabrilovich D. Mechanisms and functional significance of tumour induced dendritic-cell defects. Nat Rev Immunol. 2004. 4:941–952.
crossref
26. Schuurhuis D, Fu N, Ossendorp F, Melief CJ. Ins and outs of dendritic cells. Int Arch Allergy Immunol. 2006. 140:53–72.
crossref
27. Dhodapkar MV, Dhodapkar KM, Palucka AK. Interactions of tumor cells with dendritic cells: balancing immunity and tolerance. Cell Death Differ. 2008. 15:39–50.
crossref
28. Boonman ZF, van Mierlo GJ, Fransen MF, de Keizer RJ, Jager MJ, Melief CJ, et al. Maintenance of immune tolerance depends on normal tissue homeostasis. J Immunol. 2005. 175:4247–4254.
crossref
29. van Mierlo G, Boonman Z, Dumortier HM, den Boer AT, Fransen MF, Nouta J, et al. Activation of dendritic cells that cross-present tumor-derived antigen licenses CD8+ CTL to cause tumor eradication. J Immunol. 2004. 173:6753–6759.
crossref
30. Yang L, Carbone DP. Tumor host immune interactions and dendritic cell dysfunction. Adv Cancer Res. 2004. 92:13–27.
31. Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005. 83:451–461.
crossref
32. Uchida K, Schneider S, Yochim JM, Kuramochi H, Hayashi K, Takasaki K, et al. Intratumoral COX-2 gene expression is a predictive factor for colorectal cancer response to fluoropyrimidine-based chemotherapy. Clin Cancer Res. 2005. 11:3363–3368.
crossref
33. Williams C, Shattuck-Brandt RL, DuBois RN. The role of COX-2 in intestinal cancer. Ann N Y Acad Sci. 1999. 889:72–83.
crossref
34. Inaba T, Sano H, Kawahito Y, Hla T, Akita K, Toda M, et al. Induction of cyclooxygenase-2 in monocyte /macrophage by mucins secreted from colon cancer cells. Proc Natl Acad Sci USA. 2003. 100:2736–2741.
crossref
35. Soumaoro LT, Uetake H, Higuchi T, Takagi Y, Enomoto M, Sugihara K. Cyclooxygenase-2 expression: a significant prognostic indicator for patients with colorectal cancer. Clin Cancer Res. 2004. 10:8465–8471.
36. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol. 2001. 166:678–689.
crossref
37. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, et al. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res. 2000. 6:1755–1766.
38. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001. 166:5398–5306.
crossref
39. Young MR, Wright MA, Lozano Y, Prechel MM, Benefield J, Leonetti JP, et al. Increased recurrence and metastasis in patients whose primary head and neck squamous cell carcinomas secreted granulocyte-macrophage colony stimulating factor and contained CD34+ natural suppressor cells. Int J Cancer. 1997. 74:69–74.
crossref
40. Bell D, Chomarat P, Broyles D, Netto G, Harb GM, Lebecque S, et al. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med. 1999. 190:1417–1426.
crossref
41. Toriyama K, Wen DR, Paul E, Cochran AJ. Variations in the distribution, frequency, and phenotype of Langerhans cells during the evolution of malignant melanoma of the skin. J Invest Dermatol. 1993. 100:S269–S273.
crossref
42. Troy AJ, Summers KL, Davidson PJ, Atkinson CH, Hart DN. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin Cancer Res. 1998. 4:585–593.
crossref
43. Zou W, Machelon V, Coulomb-L'Hermin A, Borvak J, Nome F, Isaeva T, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001. 7:1339–1346.
crossref
44. Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, et al. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003. 89:1463–1472.
crossref
45. Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, et al. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J Immunol. 2004. 172:464–474.
crossref
46. Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004. 10:48–54.
crossref
47. Kortylewski M, Kujawski M, Wang T, Wei S, Zhang S, Pilon-Thomas S, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005. 11:1314–1321.
crossref
48. Ghiringhelli F, Puig PE, Roux S, Parcellier A, Schmitt E, Solary E, et al. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J Exp Med. 2005. 202:919–929.
crossref
49. Blaskovich MA, Sun J, Cantor A, Turkson J, Jove R, Sebti SM. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003. 63:1270–1279.
50. van Kester MS, Out-Luiting JJ, von dem Borne PA, Willemze R, Tensen CP, Vermeer MH. Cucurbitacin I inhibits Stat3 and induces apoptosis in Sezary cells. J Invest Dermatol. 2008. 128:1691–1695.
51. Mantovani A, Pierotti MA. Cancer and inflammation: a complex relationship. Cancer Lett. 2008. 267:180–181.
crossref
52. Sica A, Bronte V. Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest. 2007. 117:1155–1166.
crossref
53. Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumorinduced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008. 222:162–179.
crossref
54. Borrello MG, Degl'innocenti D, Pierotti M. Inflammation and cancer: the oncogene-driven connection. Cancer Lett. 2008. 267:262–270.
crossref
55. Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008. 267:271–285.
crossref
56. Ostrand-Rosenberg S, Sinha P. Myeoid-Derived suppressor cells: linking inflammation and cancer. J Immunol. 2009. 182:4499–4506.
crossref
57. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002. 420:860–867.
crossref
58. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008. 8:579–591.
crossref
59. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008. 13:23–35.
crossref
60. Marx J. Cancer immunology. Cancer's bulwark against immune attack: MDS cells. Science. 2008. 319:154–156.
61. Umemura N, Saio M, Suwa T, Kitoh Y, Bai J, Nonaka K, et al. Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J Leukocyte Biol. 2008. 83:1136–1144.
crossref
62. Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest. 2006. 116:2777–2790.
crossref
63. Rossner S, Voigtlander C, Wiethe C, Hänig J, Seifarth C, Lutz MB. Myeloid dendritic cell precursors generated from bone marrow suppress T cell responses via cell contact and nitric oxide production in vitro. Eur J Immunol. 2005. 35:3533–3544.
64. Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006. 66:1123–1131.
crossref
65. Yang R, Cai Z, Zhang Y, Yutzy WH 4th, Roby KF, Roden RB. CD80 in immune suppression by mouse ovarian carcinoma-associated Gr-1+ CD11b+ myeloid cells. Cancer Res. 2006. 66:6807–6815.
crossref
66. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumorbearing mice. J Immunol. 2008. 181:5791–5802.
crossref
67. Sawanobori Y, Ueha S, Kurachi M, Shimaoka T, Talmadge JE, Abe J, et al. Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice. Blood. 2008. 111:5457–5466.
crossref
68. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004. 172:989–999.
crossref
69. Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, et al. IL-4-induced arginase 1 suppresses allo-reactive T cells in tumor-bearing mice. J Immunol. 2003. 170:270–278.
crossref
70. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007. 67:4507–4513.
crossref
71. Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 2003. 24:302–306.
crossref
72. Kusmartsev S, Gabrilovich DI. Inhibition of myeloid cell differentiation in cancer: the role of reactive oxygen species. J Leukocyte Biol. 2003. 74:186–196.
crossref
73. Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol. 2007. 25:2546–2553.
crossref
74. Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008. 135:234–243.
crossref
75. Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, et al. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res. 2005. 65:3044–3048.
crossref
76. Srivastava MK, Bosch JJ, Thompson JA, Ksander BR, Edelman MJ, Ostrand-Rosenberg S. Lung cancer patients' CD4(+) T cells are activated in vitro by MHC II cell-based vaccines despite the presence of myeloid-derived suppressor cells. Cancer Immunol Immunother. 2008. 57:1493–1504.
crossref
77. Bronte V, Wang M, Overwijk WW, Surman DR, Pericle F, Rosenberg SA, et al. Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1 +/Gr-1+ cells. J Immunol. 1998. 161:5313–5320.
78. Bronte V, Apolloni E, Cabrelle A, Ronca R, Serafini P, Zamboni P, et al. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood. 2000. 96:3838–3846.
crossref
79. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001. 166:5398–5406.
crossref
80. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. 2002. 168:689–695.
crossref
81. Terabe M, Swann J, Ambrosino E, Sinha P, Takaku S, Hayakawa Y, et al. A non-classical non-Valpha 14Jalpha 18 CD1d-restricted (type II) NKT cell is sufficient for down-regulation of tumor immunosurveillance. J Exp Med. 2005. 202:1627–1633.
crossref
82. Baxevanis CN, Perez SA, Papamichail M. Combinatorial treatments including vaccines, chemotherapy and monoclonal antibodies for cancer therapy. Cancer Immunol Immunother. 2009. 58:317–324.
crossref
TOOLS
Similar articles