Dual effects of a targeted small-molecule inhibitor (cabozantinib) on immune-mediated killing of tumor cells and immune tumor microenvironment permissiveness when combined with a cancer vaccine

Anna R Kwilas, Andressa Ardiani, Renee N Donahue, Dana T Aftab, James W Hodge, Anna R Kwilas, Andressa Ardiani, Renee N Donahue, Dana T Aftab, James W Hodge

Abstract

Background: Growing awareness of the complexity of carcinogenesis has made multimodal therapies for cancer increasingly compelling and relevant. In recent years, immunotherapy has gained acceptance as an active therapeutic approach to cancer treatment, even though cancer is widely considered an immunosuppressive disease. Combining immunotherapy with targeted agents that have immunomodulatory capabilities could significantly improve its efficacy.

Methods: We evaluated the ability of cabozantinib, a receptor tyrosine kinase inhibitor, to modulate the immune system in vivo as well as alter the phenotype of tumor cells in vitro in order to determine if this inhibitor could act synergistically with a cancer vaccine.

Results: Our studies indicated that cabozantinib altered the phenotype of MC38-CEA murine tumor cells, rendering them more sensitive to immune-mediated killing. Cabozantinib also altered the frequency of immune sub-populations in the periphery as well as in the tumor microenvironment, which generated a more permissive immune environment. When cabozantinib was combined with a poxviral-based cancer vaccine targeting a self-antigen, the combination significantly reduced the function of regulatory T cells and increased cytokine production from effector T cells in response to the antigen. These alterations to the immune landscape, along with direct modification of tumor cells, led to markedly improved antitumor efficacy.

Conclusions: These studies support the clinical combination of cabozantinib with immunotherapy for the treatment of cancer.

Figures

Figure 1
Figure 1
Cabozantinib inhibits the growth, alters the phenotype and increases the sensitivity of MC38-CEA cells to T cell-mediated killing. (A) MC38-CEA cells were treated with 2.5 μg/mL cabozantinib or vehicle for 1, 3, and 5 days then assayed for growth and viability. Inset panel: MET and VEGFR2 expression of MC38-CEA cells. (B) MC38-CEA cells were exposed to 2.5 μg/mL cabozantinib or vehicle for 24 h, then analyzed by flow cytometry for surface expression of CEA, MHC-I (H-2Kb, H-2Db), ICAM-1, Fas, and calreticulin. Percent positivity and mean fluorescence intensity, in parentheses, are shown. Values in bold denote an increase of >40% relative to vehicle-treated cells. (C, D) MC38-CEA cells treated with cabozantinib (black bars) or vehicle (open bars) or left untreated (gray bars) for 24 h were labeled with 111In, then coincubated with CTLs specific for CEA (C) or gp70 (D) for 18 h at a ratio of 30:1. Bars indicate mean ± SEM. from quadruplicate measurements. Statistical analyses were done by Student’s t test. * = P <0.01. Data are representative of 3 independent experiments.
Figure 2
Figure 2
Cabozantinib alters the immune-cell repertoire of C57BL/6 mice. (A) Serum concentration of cabozantinib in mice fed control diet or diet containing cabozantinib for 10 days (n =5/group). Dashed line indicates the plasma concentration achieved at the maximum tolerated dose in a phase 1 human clinical trial. (B) Number of viable cells/spleen in mice fed control diet or cabozantinib diet for 35 days. The frequency of (C) CD4+ T cells (CD3+ CD4+), (D) CD8+ T cells (CD3+ CD8+), (E) Tregs (CD3+CD4+CD25+FoxP3+), and (F) MDSCs (CD11b+GR-1+) in spleens of mice fed control diet or cabozantinib diet for 35 days was determined by flow cytometry. Error bars indicate mean ± SEM. Statistical analyses were done by Student’s t test. * = P <0.05. Data are representative of 4 independent experiments.
Figure 3
Figure 3
Combining cabozantinib with a cancer vaccine improves antigen-specific immune responses in CEA-Tg C57BL/6 mice. (A) Schema of combination therapy study (n =9). (B) To evaluate Treg function, Tregs (CD3+CD4+CD25+FoxP3+) were isolated from spleens and cocultured with CD4+ T cells from naïve mice, APCs (allogeneic splenocytes irradiated with 30 Gy), and soluble anti-CD3 for 72 h. Control wells containing Tregs, APCs, and anti-CD3 without CD4+ T cells were used to determine the background level of Treg proliferation. Control wells containing CD4+ T cells, APCs, and anti-CD3 without Tregs were used to determine the background level of CD4+ T-cell proliferation. Error bars indicate mean ± SEM. Statistical analyses were done by Student’s t test. * = P <0.01 relative to CD4+ T-cell proliferation in the presence of Tregs from untreated mice; n.s. indicates no significant difference between the proliferation of CD4+ T cells in the absence of Tregs or in the presence of Tregs from mice treated with combination therapy. (C, D) To evaluate CD8+ T-cell responses, harvested splenocytes were incubated with CEA peptide (1 μg/mL) for 7 days. Lymphocytes were then restimulated with fresh, irradiated, naïve splenocytes and 1 μg/mL of either CEA or HIV-gag peptide for 24 h. Supernatants were collected and analyzed for murine IFN-γ (C) and TNF-α (D) using a cytometric bead array. Nonspecific cytokine production in response to HIV-gag was subtracted from that induced by the CEA peptide. Error bars indicate mean ± SEM. Statistical analyses were done by Student’s t test. * = P <0.05 relative to control and single-agent treatments. Data are representative of 2 independent experiments.
Figure 4
Figure 4
Cabozantinib reduces tumor vascularity and improves T cell infiltration when combined with a cancer vaccine. (A) Schema of combination therapy study (n =2/group). (B) Representative histological staining for indicated markers (top) on tumor sections from mice given indicated treatments (left). Inset panels: isotype staining. (C–F) Quantification of (C) blood vessels/tumor section; (D) number of CD3+ T-cells/tumor section; (E) number of CD4+ T-cells /tumor section; and (F) number of CD8+ T-cells /tumor section. Inset panels: number of indicated cells/mm2 of tumor. Marker staining was identified and enumerated using Aperio ImageScope image analysis software. Error bars indicate mean ± SEM. Statistical analyses were done by Student’s t test. * = P <0.05 relative to control.
Figure 5
Figure 5
Cabozantinib alters the tumor infiltration of negative regulatory cell subsets. A portion of the tumors depicted in Figure 4 were homogenized into a single-cell suspension and analyzed by flow cytometry for the presence of (A) Tregs (CD3+CD4+CD25+FoxP3+), (B) MDSCs (CD11b+Gr-1+), and (C) TAMs (CD11b+Gr-1−). Statistical analyses were done by Kolmogorov-Smirnov test. * = P <0.01 upregulation relative to control. ** = P <0.01 downregulation relative to control.
Figure 6
Figure 6
The combination of cabozantinib and immunotherapy significantly reduced tumor growth in a T cell-dependent manner. (A) Schema of combination therapy study (n =10/group). Tumor growth in mice treated with (B) control, (C) vaccine, (D) cabozantinib, and (E) combination therapy, including the number of tumor-free mice at day 35. (F) To determine the role of T cells in the efficacy of combination therapy, we measured tumor growth in control mice with no depletion (open circles), mice receiving combination therapy with no depletion (closed circles), and mice receiving combination therapy in the absence of CD4+ T cells (open squares) or CD8+ T cells (open triangles) (n =8). Depletion antibodies were administered on days 1–3, 10, 17, 24, and 31 (arrows). Tumor dimensions were measured weekly. Error bars indicate mean ± SEM. Statistical analyses were done by Student’s t test. * = P <0.0001 relative to control.

References

    1. Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, Qian F, Chu F, Bentzien F, Cancilla B, Orf J, You A, Laird AD, Engst S, Lee L, Lesch J, Chou YC, Joly AH. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther. 2011;10:2298–2308. doi: 10.1158/1535-7163.MCT-11-0264.
    1. Viola D, Cappagli V, Elisei R. Cabozantinib (XL184) for the treatment of locally advanced or metastatic progressive medullary thyroid cancer. Future Oncol. 2013;9:1083–1092. doi: 10.2217/fon.13.128.
    1. Kurzrock R, Sherman SI, Ball DW, Forastiere AA, Cohen RB, Mehra R, Pfister DG, Cohen EE, Janisch L, Nauling F, Hong DS, Ng CS, Ye L, Gagel RF, Frye J, Muller T, Ratain MJ, Salgia R. Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J Clin Oncol. 2011;29:2660–2666. doi: 10.1200/JCO.2010.32.4145.
    1. Elisei R, Schlumberger MJ, Muller SP, Schoffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V, Kreissl MC, Niederle B, Cohen EE, Wirth LJ, Ali H, Hessel C, Yaron Y, Ball D, Nelkin B, Sherman SI. Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol. 2013;31:3639–3646. doi: 10.1200/JCO.2012.48.4659.
    1. Boccaccio C, Comoglio PM. Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer. 2006;6:637–645. doi: 10.1038/nrc1912.
    1. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–925. doi: 10.1038/nrm1261.
    1. Shojaei F, Lee JH, Simmons BH, Wong A, Esparza CO, Plumlee PA, Feng J, Stewart AE, Hu-Lowe DD, Christensen JG. HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Cancer Res. 2010;70:10090–10100. doi: 10.1158/0008-5472.CAN-10-0489.
    1. Kosaka T, Yamaki E, Mogi A, Kuwano H. Mechanisms of resistance to EGFR TKIs and development of a new generation of drugs in non-small-cell lung cancer. J Biomed Biotechnol. 2011;2011:165214. doi: 10.1155/2011/165214.
    1. Park S, Choi YL, Sung CO, An J, Seo J, Ahn MJ, Ahn JS, Park K, Shin YK, Erkin OC, Song K, Kim J, Shim YM, Han J. High MET copy number and MET overexpression: poor outcome in non-small cell lung cancer patients. Histol Histopathol. 2012;27:197–207.
    1. De Oliveira AT, Matos D, Logullo AF SRDAS, Neto RA, Filho AL, Saad SS. MET is highly expressed in advanced stages of colorectal cancer and indicates worse prognosis and mortality. Anticancer Res. 2009;29:4807–4811.
    1. Lengyel E, Prechtel D, Resau JH, Gauger K, Welk A, Lindemann K, Salanti G, Richter T, Knudsen B, Vande Woude GF, Harbeck N. C-Met overexpression in node-positive breast cancer identifies patients with poor clinical outcome independent of Her2/neu. Int J Cancer. 2005;113:678–682. doi: 10.1002/ijc.20598.
    1. Ahmed SI, Thomas AL, Steward WP. Vascular endothelial growth factor (VEGF) inhibition by small molecules. J Chemother. 2004;16(Suppl 4):59–63. doi: 10.1179/joc.2004.16.Supplement-1.59.
    1. Schenone S, Bondavalli F, Botta M. Antiangiogenic agents: an update on small molecule VEGFR inhibitors. Curr Med Chem. 2007;14:2495–2516. doi: 10.2174/092986707782023622.
    1. Lien S, Lowman HB. Therapeutic anti-VEGF antibodies. Handb Exp Pharmacol. 2008;181:131–150. doi: 10.1007/978-3-540-73259-4_6.
    1. Tejpar S, Prenen H, Mazzone M. Overcoming resistance to antiangiogenic therapies. Oncologist. 2012;17:1039–1050. doi: 10.1634/theoncologist.2012-0068.
    1. Smith DC, Smith MR, Sweeney C, Elfiky AA, Logothetis C, Corn PG, Vogelzang NJ, Small EJ, Harzstark AL, Gordon MS, Vaishampayan UN, Haas NB, Spira AI, Lara PN, Jr, Lin CC, Srinivas S, Sella A, Schoffski P, Scheffold C, Weitzman AL, Hussain M. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. J Clin Oncol. 2013;31:412–419. doi: 10.1200/JCO.2012.45.0494.
    1. Xiang Q, Chen W, Ren M, Wang J, Zhang H, Deng DY, Zhang L, Shang C, Chen Y. Cabozantinib suppresses tumor growth and metastasis in hepatocellular carcinoma by a dual blockade of VEGFR2 and MET. Clin Cancer Res. 2014;20:2959–2970. doi: 10.1158/1078-0432.CCR-13-2620.
    1. Choueiri TK, Kumar Pal S, McDermott DF, Morrissey S, Ferguson KC, Holland J, Kaelin WG, Jr, Dutcher JP. A phase I study of cabozantinib (XL184) in patients with renal cell cancer. Ann Oncol. 2014;25:1603–1608. doi: 10.1093/annonc/mdu184.
    1. Higano CS, Schellhammer PF, Small EJ, Burch PA, Nemunaitis J, Yuh L, Provost N, Frohlich MW. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670–3679. doi: 10.1002/cncr.24429.
    1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466.
    1. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, Weber JS, Joshua AM, Hwu WJ, Gangadhar TC, Patnaik A, Dronca R, Zarour H, Joseph RW, Boasberg P, Chmielowski B, Mateus C, Postow MA, Gergich K, Elassaiss-Schaap J, Li XN, Iannone R, Ebbinghaus SW, Kang SP, Daud A. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117. doi: 10.1016/S0140-6736(14)60958-2.
    1. Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol. 2014;192:5451–5458. doi: 10.4049/jimmunol.1490019.
    1. Tarhini AA, Gogas H, Kirkwood JM. IFN-alpha in the treatment of melanoma. J Immunol. 2012;189:3789–3793. doi: 10.4049/jimmunol.1290060.
    1. Arlen PM, Pazdur M, Skarupa L, Rauckhorst M, Gulley JL. A randomized phase II study of docetaxel alone or in combination with PANVAC-V (vaccinia) and PANVAC-F (fowlpox) in patients with metastatic breast cancer (NCI 05-C-0229) Clin Breast Cancer. 2006;7:176–179. doi: 10.3816/CBC.2006.n.032.
    1. Farsaci B, Kwilas A, Hodge JW. Design, development, and translation of poxvirus-based vaccines for cancer. In: Bot A, Obrocea M, Marincola F, editors. Cancer Vaccines: From Research to Clinical Practice. London, UK: Informa Healthcare; 2011. pp. 56–77.
    1. Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099–1105. doi: 10.1200/JCO.2009.25.0597.
    1. Hodge JW, Ardiani A, Farsaci B, Kwilas AR, Gameiro SR. The tipping point for combination therapy: cancer vaccines with radiation, chemotherapy, or targeted small molecule inhibitors. Semin Oncol. 2012;39:323–339. doi: 10.1053/j.seminoncol.2012.02.006.
    1. Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Res. 2006;66:5527–5536. doi: 10.1158/0008-5472.CAN-05-4128.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609.
    1. Ardiani A, Farsaci B, Rogers CJ, Protter A, Guo Z, King TH, Apelian D, Hodge JW. Combination therapy with a second-generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model. Clin Cancer Res. 2013;19:6205–6218. doi: 10.1158/1078-0432.CCR-13-1026.
    1. Chakraborty M, Abrams SI, Camphausen K, Liu K, Scott T, Coleman CN, Hodge JW. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol. 2003;170:6338–6347. doi: 10.4049/jimmunol.170.12.6338.
    1. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 2004;64:4328–4337. doi: 10.1158/0008-5472.CAN-04-0073.
    1. Gameiro SR, Caballero JA, Hodge JW. Defining the molecular signature of chemotherapy-mediated lung tumor phenotype modulation and increased susceptibility to T-cell killing. Cancer Biother Radiopharm. 2012;27:23–35. doi: 10.1089/cbr.2012.1203.
    1. Farsaci B, Higgins JP, Hodge JW. Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy. Int J Cancer. 2012;130:1948–1959. doi: 10.1002/ijc.26219.
    1. Farsaci B, Sabzevari H, Higgins JP, Di Bari MG, Takai S, Schlom J, Hodge JW. Effect of a small molecule BCL-2 inhibitor on immune function and use with a recombinant vaccine. Int J Cancer. 2010;127:1603–1613. doi: 10.1002/ijc.25177.
    1. Gameiro SR, Caballero JA, Higgins JP, Apelian D, Hodge JW. Exploitation of differential homeostatic proliferation of T-cell subsets following chemotherapy to enhance the efficacy of vaccine-mediated antitumor responses. Cancer Immunol Immunother. 2011;60:1227–1242. doi: 10.1007/s00262-011-1020-8.
    1. Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403–416.
    1. Gameiro SR, Higgins JP, Dreher MR, Woods DL, Reddy G, Wood BJ, Guha C, Hodge JW. Combination therapy with local radiofrequency ablation and systemic vaccine enhances antitumor immunity and mediates local and distal tumor regression. PLoS One. 2013;8:e70417. doi: 10.1371/journal.pone.0070417.
    1. You WK, Sennino B, Williamson CW, Falcon B, Hashizume H, Yao LC, Aftab DT, McDonald DM. VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res. 2011;71:4758–4768. doi: 10.1158/0008-5472.CAN-10-2527.
    1. Sennino B, Ishiguro-Oonuma T, Wei Y, Naylor RM, Williamson CW, Bhagwandin V, Tabruyn SP, You WK, Chapman HA, Christensen JG, Aftab DT, McDonald DM. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2012;2:270–287. doi: 10.1158/-11-0240.
    1. Coudriet GM, He J, Trucco M, Mars WM, Piganelli JD. Hepatocyte growth factor modulates interleukin-6 production in bone marrow derived macrophages: implications for inflammatory mediated diseases. PLoS One. 2010;5:e15384. doi: 10.1371/journal.pone.0015384.
    1. Okunishi K, Dohi M, Nakagome K, Tanaka R, Mizuno S, Matsumoto K, Miyazaki J, Nakamura T, Yamamoto K. A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J Immunol. 2005;175:4745–4753. doi: 10.4049/jimmunol.175.7.4745.
    1. Singhal E, Sen P. Hepatocyte growth factor-induced c-Src-phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin pathway inhibits dendritic cell activation by blocking IkappaB kinase activity. Int J Biochem Cell Biol. 2011;43:1134–1146. doi: 10.1016/j.biocel.2011.04.006.
    1. Apolo A, Tomita Y, Lee M, Lee S, Frosch A, Steinberg S, Gulley JL, Schlom J, Bottaro D, Trepel J. Effect of cabozantinib on immunosuppressive subsets in metastatic urothelial carcinoma. J Clin Oncol. 2014;32(5s):abstr 4501.
    1. Overwijk WW. Breaking tolerance in cancer immunotherapy: time to ACT. Curr Opin Immunol. 2005;17:187–194. doi: 10.1016/j.coi.2005.01.011.
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102.
    1. Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, Sargent DJ. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology. 2009;137:1270–1279. doi: 10.1053/j.gastro.2009.06.053.
    1. Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, Santosuosso M, Martin JD, Martin MR, Vianello F, Leblanc P, Munn LL, Huang P, Duda DG, Fukumura D, Jain RK, Poznansky MC. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A. 2012;109:17561–17566. doi: 10.1073/pnas.1215397109.
    1. Hodge JW, Grosenbach DW, Aarts WM, Poole DJ, Schlom J. Vaccine therapy of established tumors in the absence of autoimmunity. Clin Cancer Res. 2003;9:1837–1849.
    1. Clarke P, Mann J, Simpson JF, Rickard-Dickson K, Primus FJ. Mice transgenic for human carcinoembryonic antigen as a model for immunotherapy. Cancer Res. 1998;58:1469–1477.
    1. Robbins PF, Kantor JA, Salgaller M, Hand PH, Fernsten PD, Schlom J. Transduction and expression of the human carcinoembryonic antigen gene in a murine colon carcinoma cell line. Cancer Res. 1991;51:3657–3662.
    1. Hodge JW, Poole DJ, Aarts WM, Gomez Yafal A, Gritz L, Schlom J. Modified vaccinia virus ankara recombinants are as potent as vaccinia recombinants in diversified prime and boost vaccine regimens to elicit therapeutic antitumor responses. Cancer Res. 2003;63:7942–7949.
    1. Boehm AL, Higgins J, Franzusoff A, Schlom J, Hodge JW. Concurrent vaccination with two distinct vaccine platforms targeting the same antigen generates phenotypically and functionally distinct T-cell populations. Cancer Immunol Immunother. 2010;59:397–408. doi: 10.1007/s00262-009-0759-7.
    1. Kudo-Saito C, Schlom J, Hodge JW. Induction of an antigen cascade by diversified subcutaneous/intratumoral vaccination is associated with antitumor responses. Clin Cancer Res. 2005;11:2416–2426. doi: 10.1158/1078-0432.CCR-04-1380.

Source: PubMed

3
Abonnieren