SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4+ T Lymphocytes

Markus D Lacher, Gerhard Bauer, Brian Fury, Sanne Graeve, Emily L Fledderman, Tye D Petrie, Dane P Coleal-Bergum, Tia Hackett, Nicholas H Perotti, Ying Y Kong, William W Kwok, Joseph P Wagner, Charles L Wiseman, William V Williams, Markus D Lacher, Gerhard Bauer, Brian Fury, Sanne Graeve, Emily L Fledderman, Tye D Petrie, Dane P Coleal-Bergum, Tia Hackett, Nicholas H Perotti, Ying Y Kong, William W Kwok, Joseph P Wagner, Charles L Wiseman, William V Williams

Abstract

Targeted cancer immunotherapy with irradiated, granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43-59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.

Keywords: GVAX; SV-BR-1-GM; antigen-presenting cells; targeted immunotherapy; therapeutic cancer vaccine; whole-cell vaccine.

Figures

Figure 1
Figure 1
Development of SV-BR-1-GM. (A) The SV-BR-1-GM cell line was derived from SV-BR-1 breast cancer cells following stable transfection with CSF2 [encoding human granulocyte–macrophage colony-stimulating factor (GM-CSF)]. The SV-BR-1 cell line itself was established from a chest wall lesion of a metastatic breast cancer patient (16, 17). The depicted developmental stages of SV-BR-1-GM represent samples used for this study rather than provide a comprehensive overview of all lots generated thus far. From an original master cell bank (MCB), several “clinical product” (CP) lots for actual or potential clinical use were established. “RES Lot II” refers to a research sample type and “ECB” to an engineering cell bank. RNA for gene expression analysis was extracted from cells taken directly from cryogenic vials (“cryo”) or following a culturing step (“culture”). “CP Lot VIII” was studied both as a presumptive static culture (“1d”) and as an expanded culture (“3d”), whereby for the former, samples were harvested on the first day (t = 0), and for the latter, on the third day (t = 2, i.e., 2 days later) of a time-course assessing GM-CSF secretion (outlined in Figure S5 in Supplementary Presentation S1 in Supplementary Material). Although no culture expansion took place from seeding to t = 0 (“static culture”), cell numbers of cultures harvested at t = 2 were 1.7–3.2 times the seeding cell numbers (“expanded culture”). (B) Culture morphology of SV-BR-1-GM, as exemplified by 40× and 100× original magnifications of a culture derived from the SV-BR-1-GM Lot 11 bank following 24 h of serum starvation. Of note, SV-BR-1-GM cells may grow in monolayers but can also grow as minimally adherent, sphere-like, structures, especially when seeded at very low densities. (C,D) Quality control (QC). (C) Hierarchical clustering of SV-BR-1-GM samples based on their microarray gene expression profiles. Normalized gene expression levels of samples belonging to the same sample type were averaged (arithmetic mean) prior to clustering. (D) Only samples with a RINe value of at least 7.5 were used for this study. Note that the CP Lot V cryo sample clustered separately and did not pass the minimal variability QC metric (see Materials and Methods) and was thus excluded from additional analyses.
Figure 2
Figure 2
Microarray-based transcript levels of immunostimulatory factors expressed in SV-BR-1-GM cells. 111 genes with known immunostimulatory roles were identified in published reports (–83) (Data Sheet S3 in Supplementary Material) and their microarray-based RNA expression levels determined. The 22 genes shown presented with transcript levels >1.5 times the background cutoff value (median quantile-normalized “expression” level, see Materials and Methods) in each of the SV-BR-1-GM samples. “Relative Expression Values” refers to quantile-normalized mRNA levels. ILMN_… refer to the Illumina probe sequence identifiers (PROBE_ID) of the probes yielding the expression levels shown.
Figure 3
Figure 3
nCounter-based transcript levels of immunostimulatory factors expressed in irradiated and nonirradiated SV-BR-1-GM cells. SV-BR-1-GM cells from 4- and 24-h-old clinical formulations were seeded in 6-well plates and cultured in full medium. Cells were harvested after 1 day and 3 days of culturing then subjected to nCounter-based assessment of the transcript levels of a set of 24 immune-related genes (Immune Signature candidates). For the 24-h series, also nonirradiated cells resuspended in Lactated Ringer’s solution (LRS) and processed in parallel were included. In a separate experiment, transcript levels of the Immune Signature candidates were measured from nonirradiated human mammary epithelial cells (HMECs) and SV-BR-1-GM cells cultured in parallel. Shown are background-subtracted, normalized transcript levels of HLA-A, -B, and -DRB3, whereby the probes (CodeSets) were designed to distinguish between HLA-A*01:01(A) and HLA-A*24:02(B), HLA-B*35:08(C) and HLA-A*55:01(D), and HLA-DRB3*01:01(E) and HLA-DRB3*02:02(F); and of IL6(G) and KITLG(H). (I) Comparison of the expression levels of the 24 immune-related genes between SV-BR-1 and SV-BR-1-GM cells by the nCounter approach. Comparing the 4-h formulations cultured for 1 day, almost identical expression profiles were obtained. Note that the values of CSF2 (encoding GM-CSF) shown indicate background or minute transcript levels of endogenous CSF2 since the nCounter ProbeSet employed did not match in sequence with the exogenous CSF2 expressed by SV-BR-1-GM. For (A–H), values shown are arithmetic means of background-subtracted, normalized transcript levels from triplicate wells ± SDs or from three aliquots per formulation ± SDs for the d0 samples. d0 denotes cells obtained from the original 4- or 24-h-old formulations and lysed without culturing. GM refers to cultured, nonirradiated SV-BR-1-GM cells not subjected to the formulation process. For (I), values shown are arithmetic means of background-subtracted, normalized transcript levels from triplicate (SV-BR-1-GM) or duplicate (SV-BR-1) wells. Values for all genes of the Immune Signature candidates are shown in Data Sheet S3 in Supplementary Material.
Figure 4
Figure 4
Cytokines secretion by irradiated and nonirradiated SV-BR-1-GM cells. (A) SV-BR-1-GM cells from 4- and 24-h-old clinical formulations were seeded in 6-well plates and cultured in full medium. Culture supernatants (SNs) were harvested after 1 day and 3 days of culturing then assessed for cytokine release. Note the substantially reduced levels of granulocyte–macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-6 for the 24 h compared to the 4 h old formulation. Values shown are arithmetic means from triplicate wells ± SDs, expressed as pg cytokine/1 million viable cells (at time of seeding)/24 h. For KITLG, one of the 24 h, 3d wells was excluded as the obtained cytokine levels were substantially higher than those of the other two wells. For IL-8, ELISAs for the 4 and 24 h samples were conducted on different days. (B) Cytokine levels in the Lactated Ringer’s solution (LRS) fractions of the formulations from (A). For IL-10, data are only shown for the 24 h sample. Values shown are arithmetic means from technical duplicates, expressed as pg cytokine/1 million total cells. (C) Nonirradiated human mammary epithelial cells (HMECs) and SV-BR-1-GM cells were cultured in parallel then assessed for cytokine release 24 h after replacing the culture medium. Note that the IL-6 and IL-8 levels from the nonirradiated SV-BR-1-GM cells shown here are substantially lower than those from the irradiated SV-BR-1-GM cells shown in (A). Values shown are arithmetic means from triplicate wells ± SDs, expressed as pg cytokine/1 million viable cells/24 h, whereby cell viability was determined on the day of medium change (initiation of cytokine accumulation) from cells cultured in parallel wells.
Figure 5
Figure 5
HLA-DR expression on irradiated and nonirradiated SV-BR-1-GM cells. SV-BR-1-GM cells were stained with a FITC-conjugated mouse monoclonal anti-HLA-DR antibody (clone L243) recognizing an epitope from HLA-DRα only present when complexed with an HLA-DRβ chain. Shown are arithmetic means ± SDs of technical triplicates (SV-BR-1-GM) or duplicates (A549) from (i) the percentages of HLA-DR positive cells (left axis) and (ii) the per-cell HLA-DR signal intensities normalized to the geometric mean of the signal intensity by the A549 negative control and reference cell line (value of A549 = 1) (right axis) (86, 87) Non-IRR, non-irradiated; IRR, irradiated.
Figure 6
Figure 6
Anti-HLA-DRβ3 antibody staining of a tumor specimen from the strong clinical responder. To assess whether the strong clinical responder (16), referred to as subject A002 in this article, presented with tumor expression of HLA-DRβ3, paraffin-embedded A002 tumor material was stained with a rabbit polyclonal antibody raised against an N-terminal region of human HLA-DRβ3. As demonstrated, immunoreactivity was apparent.
Figure 7
Figure 7
Cancer/testis antigen (CTA) expression in SV-BR-1-GM cells. RNA expression levels of 279 confirmed or putative CTAs (Data Sheets S4 in Supplementary Material) were compared between SV-BR-1-GM, other established breast cancer cell lines, and several normal human breast cell types. Quantile-normalized and log2-transformed microarray-based RNA expression levels are displayed according to a global gradient color scheme. Red means a higher expression level than white, and white means a higher expression level than blue. Only CTAs with a maximum representative expression value among all samples >1.5 times the background cutoff value were included.
Figure 8
Figure 8
Cancer/testis antigen selectively expressed in SV-BR-1-GM cells. Quantile-normalized microarray-based RNA levels of PRAME(A), KIF2C(B), CEP55(C), and PBK(D) “Relative Expression Values” refers to quantile-normalized mRNA levels. Values shown are arithmetic means. Error bars indicate SDs, except for STROMAL, NCL, BASAL, ALDH neg, ALDH POS, ERBB3 neg. for which standard error of the mean (S.E.M.) values are shown.
Figure 9
Figure 9
SV-BR-1-GM cells act as antigen-presenting cells (APCs). SV-BR-1-GM cells were cultured and serum-starved for 24 h then coincubated with yellow fever virus (YFV) Envelope (Env) 43–59 peptides (35) known to bind to HLA-DR complexes with an HLA-DRB3*01:01-based β chain and a YFV-DRB3*01:01-specific CD4+ T cell clone (A). (A) T cell clone after staining with YFV Env p8/DRB3*01:01 tetramers, as assessed by flow cytometry. Almost all T cells are both YFV Env p8/DRB3 and CD4 positive. (B) After 72 h of coculturing, T cell activation was assessed by determining the levels of secreted interferon (IFN)-γ. Values shown are arithmetic means from technical triplicates ± SDs, normalized to the mean IFN-γ level obtained from the YFV peptide-treated non-DRB3 PBMC reference wells. Background IFN-γ levels obtained from T cells treated with peptides in the absence of APCs (SV-BR-1-GM or PBMCs) were subtracted. (C) IFN-γ levels without background subtraction and normalization of a part of the experiment represented by panel (B). Values shown are arithmetic means of the Europium emission values at 615 nm from technical triplicates ± SDs. (B,C) one-tailed Student’s t-tests were employed to assess significance, with * referring to 0.01 ≤ p < 0.05, ** to 0.001 ≤ p < 0.01, and *** to p < 0.001.

References

    1. Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood (2014) 123(17):2625–35.10.1182/blood-2013-11-492231
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med (2014) 371(16):1507–17.10.1056/NEJMoa1407222
    1. Kontermann RE, Brinkmann U. Bispecific antibodies. Drug Discov Today (2015) 20(7):838–47.10.1016/j.drudis.2015.02.008/28220
    1. Robbins PF, Kassim SH, Tran TL, Crystal JS, Morgan RA, Feldman SA, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res (2015) 21(5):1019–27.10.1158/1078-0432.CCR-14-2708
    1. Prickett TD, Crystal JS, Cohen CJ, Pasetto A, Parkhurst MR, Gartner JJ, et al. Durable complete response from metastatic melanoma after transfer of autologous T cells recognizing 10 mutated tumor antigens. Cancer Immunol Res (2016) 4(8):669–78.10.1158/2326-6066.CIR-15-0215
    1. Groeper C, Gambazzi F, Zajac P, Bubendorf L, Adamina M, Rosenthal R, et al. Cancer/testis antigen expression and specific cytotoxic T lymphocyte responses in non small cell lung cancer. Int J Cancer (2007) 120(2):337–43.10.1002/ijc.22309
    1. Chen G, Gupta R, Petrik S, Laiko M, Leatherman JM, Asquith JM, et al. A feasibility study of cyclophosphamide, trastuzumab, and an allogeneic GM-CSF-secreting breast tumor vaccine for HER2+ metastatic breast cancer. Cancer Immunol Res (2014) 2(10):949–61.10.1158/2326-6066.CIR-14-0058
    1. Creelan BC, Antonia S, Noyes D, Hunter TB, Simon GR, Bepler G, et al. Phase II trial of a GM-CSF-producing and CD40L-expressing bystander cell line combined with an allogeneic tumor cell-based vaccine for refractory lung adenocarcinoma. J Immunother (2013) 36(8):442–50.10.1097/CJI.0b013e3182a80237
    1. Gupta R, Emens LA. GM-CSF-secreting vaccines for solid tumors: moving forward. Discov Med (2010) 10(50):52–60.
    1. Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, et al. Safety and survival with GVAX pancreas prime and Listeria monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol (2015) 33(12):1325–33.10.1200/JCO.2014.57.4244
    1. Lipson EJ, Sharfman WH, Chen S, McMiller TL, Pritchard TS, Salas JT, et al. Safety and immunologic correlates of melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J Transl Med (2015) 13:214.10.1186/s12967-015-0572-3
    1. Lollini PL, Cavallo F, Nanni P, Quaglino E. The promise of preventive cancer vaccines. Vaccines (Basel) (2015) 3(2):467–89.10.3390/vaccines3020467
    1. Mittendorf EA, Clifton GT, Holmes JP, Schneble E, van Echo D, Ponniah S, et al. Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients. Ann Oncol (2014) 25(9):1735–42.10.1093/annonc/mdu211
    1. Santegoets SJ, Stam AG, Lougheed SM, Gall H, Jooss K, Sacks N, et al. Myeloid derived suppressor and dendritic cell subsets are related to clinical outcome in prostate cancer patients treated with prostate GVAX and ipilimumab. J Immunother Cancer (2014) 2:31.10.1186/s40425-014-0031-3
    1. Srivatsan S, Patel JM, Bozeman EN, Imasuen IE, He S, Daniels D, et al. Allogeneic tumor cell vaccines: the promise and limitations in clinical trials. Hum Vaccin Immunother (2014) 10(1):52–63.10.4161/hv.26568
    1. Wiseman CL, Kharazi A. Objective clinical regression of metastatic breast cancer in disparate sites after use of whole-cell vaccine genetically modified to release sargramostim. Breast J (2006) 12(5):475–80.10.1111/j.1075-122X.2006.00319.x
    1. Wiseman CL, Kharazi A. Phase I study with SV-BR-1 breast cancer cell line vaccine and GM-CSF: clinical experience in 14 patients. Open Breast Cancer J (2010) 2:4–11.10.2174/1876817201002010004
    1. Silvestri I, Cattarino S, Agliano AM, Collalti G, Sciarra A. Beyond the immune suppression: the immunotherapy in prostate cancer. Biomed Res Int (2015) 2015:794968.10.1155/2015/794968
    1. Keenan BP, Jaffee EM. Whole cell vaccines – past progress and future strategies. Semin Oncol (2012) 39(3):276–86.10.1053/j.seminoncol.2012.02.007
    1. De Remigis A, de Gruijl TD, Uram JN, Tzou SC, Iwama S, Talor MV, et al. Development of thyroglobulin antibodies after GVAX immunotherapy is associated with prolonged survival. Int J Cancer (2015) 136(1):127–37.10.1002/ijc.28973
    1. Soares KC, Rucki AA, Kim V, Foley K, Solt S, Wolfgang CL, et al. TGF-beta blockade depletes T regulatory cells from metastatic pancreatic tumors in a vaccine dependent manner. Oncotarget (2015) 6(40):43005–15.10.18632/oncotarget.5656
    1. Borrello IM, Levitsky HI, Stock W, Sher D, Qin L, DeAngelo DJ, et al. Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting cellular immunotherapy in combination with autologous stem cell transplantation (ASCT) as postremission therapy for acute myeloid leukemia (AML). Blood (2009) 114(9):1736–45.10.1182/blood-2009-02-205278
    1. Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol (2009) 27(35):5911–8.10.1200/JCO.2009.23.3494
    1. Ogawa T, Kusumoto M, Mizumoto K, Sato N, Tanaka M. Adenoviral GM-CSF gene transduction into breast cancer cells induced long-lasting antitumor immunity in mice. Breast Cancer (1999) 6(4):301–4.10.1007/BF02966444
    1. Le DT, Jaffee EM. Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res (2012) 72(14):3439–44.10.1158/0008-5472.CAN-11-3912
    1. Arico E, Belardelli F. Interferon-alpha as antiviral and antitumor vaccine adjuvants: mechanisms of action and response signature. J Interferon Cytokine Res (2012) 32(6):235–47.10.1089/jir.2011.0077
    1. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet (2006) 38(5):500–1.10.1038/ng0506-500
    1. Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res (2002) 30(1):207–10.10.1093/nar/30.1.207
    1. Jongeneel CV, Delorenzi M, Iseli C, Zhou D, Haudenschild CD, Khrebtukova I, et al. An atlas of human gene expression from massively parallel signature sequencing (MPSS). Genome Res (2005) 15(7):1007–14.10.1101/gr.4041005
    1. Lowe R, Overhoff MG, Ramagopalan SV, Garbe JC, Koh J, Stampfer MR, et al. The senescent methylome and its relationship with cancer, ageing and germline genetic variation in humans. Genome Biol (2015) 16:194.10.1186/s13059-015-0748-4
    1. Shehata M, Teschendorff A, Sharp G, Novcic N, Russell IA, Avril S, et al. Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland. Breast Cancer Res (2012) 14(5):R134.10.1186/bcr3334
    1. Dobrynin P, Matyunina E, Malov SV, Kozlov AP. The novelty of human cancer/testis antigen encoding genes in evolution. Int J Genomics (2013) 2013:105108.10.1155/2013/105108
    1. Chapman KB, Prendes MJ, Kidd JL, Sternberg H, West MD, Wagner J. Elevated expression of cancer/testis antigen FSIP1 in ER-positive breast tumors. Biomark Med (2013) 7(4):601–11.10.2217/bmm.13.58
    1. Almeida LG, Sakabe NJ, deOliveira AR, Silva MC, Mundstein AS, Cohen T, et al. CTdatabase: a knowledge-base of high-throughput and curated data on cancer-testis antigens. Nucleic Acids Res (2009) 37(Database issue):D816–9.10.1093/nar/gkn673
    1. James EA, LaFond RE, Gates TJ, Mai DT, Malhotra U, Kwok WW. Yellow fever vaccination elicits broad functional CD4+ T cell responses that recognize structural and nonstructural proteins. J Virol (2013) 87(23):12794–804.10.1128/JVI.01160-13
    1. Gragert L, Madbouly A, Freeman J, Maiers M. Six-locus high resolution HLA haplotype frequencies derived from mixed-resolution DNA typing for the entire US donor registry. Hum Immunol (2013) 74(10):1313–20.10.1016/j.humimm.2013.06.025
    1. Broughton SE, Hercus TR, Nero TL, Dottore M, McClure BJ, Dhagat U, et al. Conformational changes in the GM-CSF receptor suggest a molecular mechanism for affinity conversion and receptor signaling. Structure (2016) 24(8):1271–81.10.1016/j.str.2016.05.017
    1. Brown CB, Pihl CE, Kaushansky K. Mapping of human granulocyte-macrophage-colony-stimulating-factor domains interacting with the human granulocyte-macrophage-colony-stimulating-factor-receptor alpha-subunit. Eur J Biochem (1994) 225(3):873–80.10.1111/j.1432-1033.1994.0873b.x
    1. Hansen G, Hercus TR, McClure BJ, Stomski FC, Dottore M, Powell J, et al. The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation. Cell (2008) 134(3):496–507.10.1016/j.cell.2008.05.053
    1. Hu ZB, Ma W, Zaborski M, MacLeod R, Quentmeier H, Drexler HG. Establishment and characterization of two novel cytokine-responsive acute myeloid and monocytic leukemia cell lines, MUTZ-2 and MUTZ-3. Leukemia (1996) 10(6):1025–40.
    1. Arruda LB, Sim D, Chikhlikar PR, Maciel M, Jr, Akasaki K, August JT, et al. Dendritic cell-lysosomal-associated membrane protein (LAMP) and LAMP-1-HIV-1 gag chimeras have distinct cellular trafficking pathways and prime T and B cell responses to a diverse repertoire of epitopes. J Immunol (2006) 177(4):2265–75.10.4049/jimmunol.177.4.2265
    1. Bates JM, Flanagan K, Mo L, Ota N, Ding J, Ho S, et al. Dendritic cell CD83 homotypic interactions regulate inflammation and promote mucosal homeostasis. Mucosal Immunol (2015) 8(2):414–28.10.1038/mi.2014.79
    1. Caux C, Massacrier C, Vanbervliet B, Dubois B, Van Kooten C, Durand I, et al. Activation of human dendritic cells through CD40 cross-linking. J Exp Med (1994) 180(4):1263–72.10.1084/jem.180.4.1263
    1. Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel) (2011) 3(4):3856–93.10.3390/cancers3043856
    1. Leitner J. The Role of Costimulatory Pathways During the Activation of Human T Cells [Dissertation (Ph.D. Thesis)]. Austria: University of Vienna; (2011). Available from: (Accessed: April 5, 2018).
    1. Ohshima Y, Tanaka Y, Tozawa H, Takahashi Y, Maliszewski C, Delespesse G. Expression and function of OX40 ligand on human dendritic cells. J Immunol (1997) 159(8):3838–48.
    1. Rescigno M, Piguet V, Valzasina B, Lens S, Zubler R, French L, et al. Fas engagement induces the maturation of dendritic cells (DCs), the release of interleukin (IL)-1beta, and the production of interferon gamma in the absence of IL-12 during DC-T cell cognate interaction: a new role for Fas ligand in inflammatory responses. J Exp Med (2000) 192(11):1661–8.10.1084/jem.192.11.1661
    1. Wong BR, Josien R, Lee SY, Sauter B, Li HL, Steinman RM, et al. TRANCE (tumor necrosis factor [TNF]-related activation-induced cytokine), a new TNF family member predominantly expressed in T cells, is a dendritic cell-specific survival factor. J Exp Med (1997) 186(12):2075–80.10.1084/jem.186.12.2075
    1. Zou GM, Tam YK. Cytokines in the generation and maturation of dendritic cells: recent advances. Eur Cytokine Netw (2002) 13(2):186–99.
    1. Bleijs DA, de Waal-Malefyt R, Figdor CG, van Kooyk Y. Co-stimulation of T cells results in distinct IL-10 and TNF-alpha cytokine profiles dependent on binding to ICAM-1, ICAM-2 or ICAM-3. Eur J Immunol (1999) 29(7):2248–58.
    1. Damle NK, Aruffo A. Vascular cell adhesion molecule 1 induces T-cell antigen receptor-dependent activation of CD4+T lymphocytes. Proc Natl Acad Sci U S A (1991) 88(15):6403–7.10.1073/pnas.88.15.6403
    1. Dieu MC, Vanbervliet B, Vicari A, Bridon JM, Oldham E, Ait-Yahia S, et al. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J Exp Med (1998) 188(2):373–86.10.1084/jem.188.2.373
    1. Gokhale A, Kanthala S, Latendresse J, Taneja V, Satyanarayanajois S. Immunosuppression by co-stimulatory molecules: inhibition of CD2-CD48/CD58 interaction by peptides from CD2 to suppress progression of collagen-induced arthritis in mice. Chem Biol Drug Des (2013) 82(1):106–18.10.1111/cbdd.12138
    1. He S, Wang L, Wu Y, Li D, Zhang Y. CCL3 and CCL20-recruited dendritic cells modified by melanoma antigen gene-1 induce anti-tumor immunity against gastric cancer ex vivo and in vivo. J Exp Clin Cancer Res (2010) 29:37.10.1186/1756-9966-29-37
    1. Leitner J, Grabmeier-Pfistershammer K, Steinberger P. Receptors and ligands implicated in human T cell costimulatory processes. Immunol Lett (2010) 128(2):89–97.10.1016/j.imlet.2009.11.009
    1. Sutavani RV, Bradley RG, Ramage JM, Jackson AM, Durrant LG, Spendlove I. CD55 costimulation induces differentiation of a discrete T regulatory type 1 cell population with a stable phenotype. J Immunol (2013) 191(12):5895–903.10.4049/jimmunol.1301458
    1. Arango Duque G, Descoteaux A. Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol (2014) 5:491.10.3389/fimmu.2014.00491
    1. Goswami R, Kaplan MH. A brief history of IL-9. J Immunol (2011) 186(6):3283–8.10.4049/jimmunol.1003049
    1. Hirano N, Butler MO, Xia Z, Ansen S, von Bergwelt-Baildon MS, Neuberg D, et al. Engagement of CD83 ligand induces prolonged expansion of CD8+ T cells and preferential enrichment for antigen specificity. Blood (2006) 107(4):1528–36.10.1182/blood-2005-05-2073
    1. Imai T, Baba M, Nishimura M, Kakizaki M, Takagi S, Yoshie O. The T cell-directed CC chemokine TARC is a highly specific biological ligand for CC chemokine receptor 4. J Biol Chem (1997) 272(23):15036–42.10.1074/jbc.272.23.15036
    1. Luo H, Yu G, Wu Y, Wu J. EphB6 crosslinking results in costimulation of T cells. J Clin Invest (2002) 110(8):1141–50.10.1172/JCI15883
    1. Mandapathil M, Szczepanski M, Harasymczuk M, Ren J, Cheng D, Jackson EK, et al. CD26 expression and adenosine deaminase activity in regulatory T cells (Treg) and CD4(+) T effector cells in patients with head and neck squamous cell carcinoma. Oncoimmunology (2012) 1(5):659–69.10.4161/onci.20387
    1. Prazma CM, Tedder TF. Dendritic cell CD83: a therapeutic target or innocent bystander? Immunol Lett (2008) 115(1):1–8.10.1016/j.imlet.2007.10.001
    1. Rodriguez RM, Pitzalis C, Kingsley GH, Henderson E, Humphries MJ, Panayi GS. T lymphocyte adhesion to fibronectin (FN): a possible mechanism for T cell accumulation in the rheumatoid joint. Clin Exp Immunol (1992) 89(3):439–45.10.1111/j.1365-2249.1992.tb06977.x
    1. Shimizu Y, van Seventer GA, Horgan KJ, Shaw S. Costimulation of proliferative responses of resting CD4+ T cells by the interaction of VLA-4 and VLA-5 with fibronectin or VLA-6 with laminin. J Immunol (1990) 145(1):59–67.
    1. Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta (2014) 1843(11):2563–82.10.1016/j.bbamcr.2014.05.014
    1. Gibbert K, Schlaak JF, Yang D, Dittmer U. IFN-alpha subtypes: distinct biological activities in anti-viral therapy. Br J Pharmacol (2013) 168(5):1048–58.10.1111/bph.12010
    1. Oft M. IL-10: master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res (2014) 2(3):194–9.10.1158/2326-6066.CIR-13-0214
    1. Geijtenbeek TB, Torensma R, van Vliet SJ, van Duijnhoven GC, Adema GJ, van Kooyk Y, et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell (2000) 100(5):575–85.10.1016/S0092-8674(00)80693-5
    1. Le Y, Zhou Y, Iribarren P, Wang J. Chemokines and chemokine receptors: their manifold roles in homeostasis and disease. Cell Mol Immunol (2004) 1(2):95–104.
    1. Lebre MC, Burwell T, Vieira PL, Lora J, Coyle AJ, Kapsenberg ML, et al. Differential expression of inflammatory chemokines by Th1- and Th2-cell promoting dendritic cells: a role for different mature dendritic cell populations in attracting appropriate effector cells to peripheral sites of inflammation. Immunol Cell Biol (2005) 83(5):525–35.10.1111/j.1440-1711.2005.01365.x
    1. Ray P, Krishnamoorthy N, Ray A. Emerging functions of c-kit and its ligand stem cell factor in dendritic cells: regulators of T cell differentiation. Cell Cycle (2008) 7(18):2826–32.10.4161/cc.7.18.6752
    1. Dorak MT, Lawson T, Machulla HK, Mills KI, Burnett AK. Increased heterozygosity for MHC class II lineages in newborn males. Genes Immun (2002) 3(5):263–9.10.1038/sj.gene.6363862
    1. Foroni I, Couto AR, Bettencourt BF, Santos M, Lima M, Bruges-Armas J. HLA-E, HLA-F and HLA-G — the non-classical side of the MHC cluster. In: Xi Y, editor. HLA and Associated Important Diseases. (Chap. 3), (2014). Distributed Under Terms of Creative Commons Attribution License.10.5772/57507
    1. Fortin JS, Cloutier M, Thibodeau J. Exposing the specific roles of the invariant chain isoforms in shaping the MHC class II peptidome. Front Immunol (2013) 4:443.10.3389/fimmu.2013.00443
    1. Shiina T, Hosomichi K, Inoko H, Kulski JK. The HLA genomic loci map: expression, interaction, diversity and disease. J Hum Genet (2009) 54(1):15–39.10.1038/jhg.2008.5
    1. Yang J, Yi Q. Killing tumor cells through their surface beta(2)-microglobulin or major histocompatibility complex class I molecules. Cancer (2010) 116(7):1638–45.10.1002/cncr.24953
    1. Ezinne CC, Yoshimitsu M, White Y, Arima N. HTLV-1 specific CD8+ T cell function augmented by blockade of 2B4/CD48 interaction in HTLV-1 infection. PLoS One (2014) 9(2):e87631.10.1371/journal.pone.0087631
    1. Ma DY, Clark EA. The role of CD40 and CD154/CD40L in dendritic cells. Semin Immunol (2009) 21(5):265–72.10.1016/j.smim.2009.05.010
    1. Ohnuma K, Uchiyama M, Yamochi T, Nishibashi K, Hosono O, Takahashi N, et al. Caveolin-1 triggers T-cell activation via CD26 in association with CARMA1. J Biol Chem (2007) 282(13):10117–31.10.1074/jbc.M609157200
    1. Gessani S, Conti L, Del Corno M, Belardelli F. Type I interferons as regulators of human antigen presenting cell functions. Toxins (Basel) (2014) 6(6):1696–723.10.3390/toxins6061696
    1. Hillyer P, Raviv N, Gold DM, Dougherty D, Liu J, Johnson TR, et al. Subtypes of type I IFN differentially enhance cytokine expression by suboptimally stimulated CD4(+) T cells. Eur J Immunol (2013) 43(12):3197–208.10.1002/eji.201243288
    1. Lee N, You S, Shin MS, Lee WW, Kang KS, Kim SH, et al. IL-6 receptor alpha defines effector memory CD8+ T cells producing Th2 cytokines and expanding in asthma. Am J Respir Crit Care Med (2014) 190(12):1383–94.10.1164/rccm.201403-0601OC
    1. Guce AI, Mortimer SE, Yoon T, Painter CA, Jiang W, Mellins ED, et al. HLA-DO acts as a substrate mimic to inhibit HLA-DM by a competitive mechanism. Nat Struct Mol Biol (2013) 20(1):90–8.10.1038/nsmb.2460
    1. Edgecombe AD. HLA Class II Expression on Breast Cancer Cells. Masters thesis, Faculty of Medicine, Memorial University of Newfoundland, Canada: (2002). Available from: (Accessed: April 5, 2018).
    1. Jiang Y, Arase N, Kohyama M, Hirayasu K, Suenaga T, Jin H, et al. Transport of misfolded endoplasmic reticulum proteins to the cell surface by MHC class II molecules. Int Immunol (2013) 25(4):235–46.10.1093/intimm/dxs155
    1. Corbiere V, Dirix V, Norrenberg S, Cappello M, Remmelink M, Mascart F. Phenotypic characteristics of human type II alveolar epithelial cells suitable for antigen presentation to T lymphocytes. Respir Res (2011) 12:15.10.1186/1465-9921-12-15
    1. Suri A, Jagadish N, Saini S, Gupta N. Targeting cancer testis antigens for biomarkers and immunotherapy in colorectal cancer: current status and challenges. World J Gastrointest Oncol (2015) 7(12):492–502.10.4251/wjgo.v7.i12.492
    1. Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol (2011) 5(2):164–82.10.1016/j.molonc.2011.02.001
    1. Gjerstorff MF, Andersen MH, Ditzel HJ. Oncogenic cancer/testis antigens: prime candidates for immunotherapy. Oncotarget (2015) 6(18):15772–87.10.18632/oncotarget.4694
    1. LaVoy EC, Bollard CM, Hanley PJ, Blaney JW, O’Connor DP, Bosch JA, et al. A single bout of dynamic exercise enhances the expansion of MAGE-A4 and PRAME-specific cytotoxic T-cells from healthy adults. Exerc Immunol Rev (2015) 21:144–53.
    1. Marchini C, Kalogris C, Garulli C, Pietrella L, Gabrielli F, Curcio C, et al. Tailoring DNA vaccines: designing strategies against HER2-positive cancers. Front Oncol (2013) 3:122.10.3389/fonc.2013.00122
    1. Wu TC, Huang AY, Jaffee EM, Levitsky HI, Pardoll DM. A reassessment of the role of B7-1 expression in tumor rejection. J Exp Med (1995) 182(5):1415–21.10.1084/jem.182.5.1415
    1. Bonaccorsi I, Pezzino G, Morandi B, Ferlazzo G. Drag cells in immunity (plasmacytoid DCs dress up as cancer cells). Oncoimmunology (2014) 3:e27897.10.4161/onci.28184
    1. Nakayama M. Antigen presentation by MHC-dressed cells. Front Immunol (2015) 5:672.10.3389/fimmu.2014.00672
    1. Smyth LA, Harker N, Turnbull W, El-Doueik H, Klavinskis L, Kioussis D, et al. The relative efficiency of acquisition of MHC:peptide complexes and cross-presentation depends on dendritic cell type. J Immunol (2008) 181(5):3212–20.10.4049/jimmunol.181.5.3212
    1. Huang AY, Bruce AT, Pardoll DM, Levitsky HI. Does B7-1 expression confer antigen-presenting cell capacity to tumors in vivo? J Exp Med (1996) 183(3):769–76.10.1084/jem.183.3.769
    1. Huang AY, Golumbek P, Ahmadzadeh M, Jaffee E, Pardoll D, Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science (1994) 264(5161):961–5.10.1126/science.7513904
    1. Sharma A, Bode B, Wenger RH, Lehmann K, Sartori AA, Moch H, et al. gamma-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo. PLoS One (2011) 6(11):e28217.10.1371/journal.pone.0028217
    1. Safi S, Beckhove P, Warth A, Benner A, Roeder F, Rieken S, et al. A randomized phase II study of radiation induced immune boost in operable non-small cell lung cancer (RadImmune trial). BMC Cancer (2015) 15:988.10.1186/s12885-015-2006-2
    1. Finkelstein SE, Salenius S, Mantz CA, Shore ND, Fernandez EB, Shulman J, et al. Combining immunotherapy and radiation for prostate cancer. Clin Genitourin Cancer (2015) 13(1):1–9.10.1016/j.clgc.2014.09.001
    1. Reynders K, Illidge T, Siva S, Chang JY, De Ruysscher D. The abscopal effect of local radiotherapy: using immunotherapy to make a rare event clinically relevant. Cancer Treat Rev (2015) 41(6):503–10.10.1016/j.ctrv.2015.03.011
    1. Makkouk A, Weiner GJ. Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge. Cancer Res (2015) 75(1):5–10.10.1158/0008-5472.CAN-14-2538
    1. Hongisto V, Aure MR, Makela R, Sahlberg KK. The HER2 amplicon includes several genes required for the growth and survival of HER2 positive breast cancer cells – a data description. Genom Data (2014) 2:249–53.10.1016/j.gdata.2014.06.025
    1. Sahlberg KK, Hongisto V, Edgren H, Makela R, Hellstrom K, Due EU, et al. The HER2 amplicon includes several genes required for the growth and survival of HER2 positive breast cancer cells. Mol Oncol (2013) 7(3):392–401.10.1016/j.molonc.2012.10.012
    1. Evans EE, Henn AD, Jonason A, Paris MJ, Schiffhauer LM, Borrello MA, et al. C35 (C17orf37) is a novel tumor biomarker abundantly expressed in breast cancer. Mol Cancer Ther (2006) 5(11):2919–30.10.1158/1535-7163.MCT-06-0389
    1. Katz E, Dubois-Marshall S, Sims AH, Faratian D, Li J, Smith ES, et al. A gene on the HER2 amplicon, C35, is an oncogene in breast cancer whose actions are prevented by inhibition of Syk. Br J Cancer (2010) 103(3):401–10.10.1038/sj.bjc.6605763
    1. Kpetemey M, Chaudhary P, Van Treuren T, Vishwanatha JK. MIEN1 drives breast tumor cell migration by regulating cytoskeletal-focal adhesion dynamics. Oncotarget (2016) 7(34):54913–24.10.18632/oncotarget.10798
    1. Knaus A, Awaya T, Helbig I, Afawi Z, Pendziwiat M, Abu-Rachma J, et al. Rare noncoding mutations extend the mutational spectrum in the PGAP3 subtype of hyperphosphatasia with mental retardation syndrome. Hum Mutat (2016) 37(8):737–44.10.1002/humu.23006
    1. Vassilev B, Sihto H, Li S, Holtta-Vuori M, Ilola J, Lundin J, et al. Elevated levels of StAR-related lipid transfer protein 3 alter cholesterol balance and adhesiveness of breast cancer cells: potential mechanisms contributing to progression of HER2-positive breast cancers. Am J Pathol (2015) 185(4):987–1000.10.1016/j.ajpath.2014.12.018

Source: PubMed

3
Subscribe