Multiple antigen-engineered DC vaccines with or without IFNα to promote antitumor immunity in melanoma

Lisa H Butterfield, Lazar Vujanovic, Patricia M Santos, Deena M Maurer, Andrea Gambotto, Joel Lohr, Chunlei Li, Jacob Waldman, Uma Chandran, Yan Lin, Huang Lin, Hussein A Tawbi, Ahmad A Tarhini, John M Kirkwood, Lisa H Butterfield, Lazar Vujanovic, Patricia M Santos, Deena M Maurer, Andrea Gambotto, Joel Lohr, Chunlei Li, Jacob Waldman, Uma Chandran, Yan Lin, Huang Lin, Hussein A Tawbi, Ahmad A Tarhini, John M Kirkwood

Abstract

Background: Cancer vaccines are designed to promote systemic antitumor immunity and tumor eradication. Cancer vaccination may be more efficacious in combination with additional interventions that may build on or amplify their effects.

Methods: Based on our previous clinical and in vitro studies, we designed an antigen-engineered DC vaccine trial to promote a polyclonal CD8+ and CD4+ T cell response against three shared melanoma antigens. The 35 vaccine recipients were then randomized to receive one month of high-dose IFNα or observation.

Results: The resulting clinical outcomes were 2 partial responses, 8 stable disease and 14 progressive disease among patients with measurable disease using RECIST 1.1, and, of 11 surgically treated patients with no evidence of disease (NED), 4 remain NED at a median follow-up of 3 years. The majority of vaccinated patients showed an increase in vaccine antigen-specific CD8+ and CD4+ T cell responses. The addition of IFNα did not appear to improve immune or clinical responses in this trial. Examination of the DC vaccine profiles showed that IL-12p70 secretion did not correlate with immune or clinical responses. In depth immune biomarker studies support the importance of circulating Treg and MDSC for development of antigen-specific T cell responses, and of circulating CD8+ and CD4+ T cell subsets in clinical responses.

Conclusions: DC vaccines are a safe and reliable platform for promoting antitumor immunity. This combination with one month of high dose IFNα did not improve outcomes. Immune biomarker analysis in the blood identified several predictive and prognostic biomarkers for further analysis, including MDSC.

Trial registration: NCT01622933 .

Keywords: Cancer vaccine; Immune biomarkers; Melanoma; Shared antigens; Tumor immunity.

Conflict of interest statement

Consent for publication

All authors confirm that they have no relevant conflicts of interest to report.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Kaplan-Meier plot of OS and PFS. a and b. OS and PFS is shown for patients who were randomized after DC vaccines (n = 23) to observation (no boost, n = 12 randomized + 2 not receiving HDI) or those randomized to HDI (n = 11 randomized, 9 receiving IFN). OS: IFN vs. OBS p = 0.54 (ns). PFS: IFN vs. OBS p = 0.43 (ns). Not shown are those who progressed early, before randomization (n = 12), who were the statistically significant different clinical group vs. those randomized (OS p = 0.0001, PFS p < 0.0001)
Fig. 2
Fig. 2
IFNγ ELISPOT assay for purified CD8+ and CD4+ T cells. A direct ELISPOT was performed to determine the frequency of T cells specific to full length antigens expressed in autologous DC and previously characterized peptides (n = 28 patients). Assay controls (no antigen, PMA + ionomycin) are also shown. The circle symbols denote trial arm, and the responses of the two PR patients are also noted (filled triangle)
Fig. 3
Fig. 3
Whole blood T and NK cell phenotyping. a-f The percentages and absolute counts for CD3+CD4+ and CD3+CD8+ T cells expressing naïve/effector/memory markers (CD45RA, CCR7) (a-c) or trafficking markers (CXCR3, CCR6) (d-f) are shown in melanoma patients (n = 35 patients) in comparison to HD controls (n = 35) (a, b) or by trial arm (d, e). Box plots for significant correlations with clinical response are shown (c, f, j). g-i NK cell subset phenotyping for NKG2D expression levels as shown for HD and patients
Fig. 4
Fig. 4
Circulating suppressor cell frequencies. The frequencies of (a) Treg and subsets of myeloid (b, c) MDSC are shown (n = 35 patients). The left side of each panel shows the difference between baseline and d = 43 post DC vaccines. The right side of each panel shows the change between d43 and d89 for each trial arm. Dotted lines represent median HD values (n = 35). Two examples of significant correlations between MDSC frequencies and patient development of vaccine antigen-specific T cell responses are shown (d)

References

    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2012;61:1019–1031. doi: 10.1007/s00262-011-1172-6.
    1. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782.
    1. Klebanoff CA, Acquavella N, Yu Z, Restifo NP. Therapeutic cancer vaccines: are we there yet? Immunol Rev. 2011;239:27–44. doi: 10.1111/j.1600-065X.2010.00979.x.
    1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–915. doi: 10.1038/nm1100.
    1. Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–2127. doi: 10.1056/NEJMoa1012863.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi: 10.1056/NEJMoa1001294.
    1. Gross S, Erdmann M, Haendle I, Voland S, Berger T, Schultz E, et al. Twelve-year survival and immune correlates in dendritic cell-vaccinated melanoma patients. JCI Insight. 2017;2:e91438. doi: 10.1172/jci.insight.91438.
    1. Lehmann PV, Forsthuber T, Miller A, Sercarz EE. Spreading of T-cell autoimmunity to cryptic determinants of an autoantigen. Nature. 1992;358:155–157. doi: 10.1038/358155a0.
    1. Tian J, Gregori S, Adorini L, Kaufman DL. The frequency of high avidity T cells determines the hierarchy of determinant spreading. J Immunol. 2001;166:7144–7150. doi: 10.4049/jimmunol.166.12.7144.
    1. Kaufman DL, Clare-Salzler M, Tian J, Forsthuber T, Ting GS, Robinson P, et al. Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature. 1993;366:69–72. doi: 10.1038/366069a0.
    1. Lehmann PV, Sercarz EE, Forsthuber T, Dayan CM, Gammon G. Determinant spreading and the dynamics of the autoimmune T-cell repertoire. Immunol Today. 1993;14:203–208. doi: 10.1016/0167-5699(93)90163-F.
    1. Banchereau J, Palucka AK, Dhodapkar M, Burkeholder S, Taquet N, Rolland A, et al. Immune and clinical responses in patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–6458.
    1. Butterfield LH, Ribas A, Dissette VB, Amarnani SN, Vu HT, Oseguera D, et al. Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res. 2003;9:998–1008.
    1. Ribas A, Glaspy JA, Lee Y, Dissette VB, Seja E, Vu HT, et al. Role of dendritic cell phenotype, determinant spreading, and negative costimulatory blockade in dendritic cell-based melanoma immunotherapy. J Immunother. 2004;27:354–367. doi: 10.1097/00002371-200409000-00004.
    1. Butterfield LH, Comin-Anduix B, Vujanovic L, Lee Y, Dissette VB, Yang JQ, et al. Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J Immunother. 2008;31:294–309. doi: 10.1097/CJI.0b013e31816a8910.
    1. Wierecky J, Muller MR, Wirths S, Halder-Oehler E, Dorfel D, Schmidt SM, et al. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res. 2006;66:5910–5918. doi: 10.1158/0008-5472.CAN-05-3905.
    1. Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, et al. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol. 2002;20:2624–2632. doi: 10.1200/JCO.2002.06.171.
    1. Ranieri E, Kierstead LS, Zarour H, Kirkwood JM, Lotze MT, Whiteside T, et al. Dendritic cell/peptide cancer vaccines: clinical responsiveness and epitope spreading. Immunol Investig. 2000;29:121–125. doi: 10.3109/08820130009062294.
    1. Gulley JL, Madan RA, Tsang KY, Jochems C, Marte JL, Farsaci B, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2:133–141. doi: 10.1158/2326-6066.CIR-13-0108.
    1. Blalock LT, Landsberg J, Messmer M, Shi J, Pardee AD, Haskell R, et al. Human dendritic cells adenovirally-engineered to express three defined tumor antigens promote broad adaptive and innate immunity. Oncoimmunology. 2012;1:287–357. doi: 10.4161/onci.18628.
    1. Arthur JF, Butterfield LH, Roth MD, Bui LA, Kiertscher SM, Lau R, et al. A comparison of gene transfer methods in human dendritic cells. Cancer Gene Ther. 1997;4:17–25.
    1. Vujanovic L, Whiteside TL, Potter DM, Chu J, Ferrone S, Butterfield LH. Regulation of antigen presentation machinery in human dendritic cells by recombinant adenovirus. Cancer Immunol Immunother. 2009;58:121–133. doi: 10.1007/s00262-008-0533-2.
    1. Jonuleit H, Giesecke-Tuettenberg A, Tüting T, Thurner-Schuler B, Stuge TB, Paragnik L, Kandemir A, Lee PP, Schuler G, Knop J, Enk AH. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer. 2001;93(2):243–251. doi: 10.1002/ijc.1323.
    1. Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001;193(2):233–238. doi: 10.1084/jem.193.2.233.
    1. Schumacher L, Ribas A, Dissette VB, McBride WH, Mukherji B, Economou JS, et al. Human dendritic cell maturation by adenovirus transduction enhances tumor antigen-specific T-cell responses. J Immunother. 2004;27:191–200. doi: 10.1097/00002371-200405000-00003.
    1. Nordlund JJ, Kirkwood JM, Forget BM, Milton G, Albert DM, Lerner AB. Vitiligo in patients with metastatic melanoma: a good prognostic sign. J Am Acad Dermatol. 1983;9:689–696. doi: 10.1016/S0190-9622(83)70182-9.
    1. Gogas H, Ioannovich J, Dafni U, Stavropoulou-Giokas C, Frangia K, Tsoutsos D, et al. Prognostic significance of autoimmunity during treatment of melanoma with interferon. N Engl J Med. 2006;354:709–718. doi: 10.1056/NEJMoa053007.
    1. Krauze MT, Tarhini A, Gogas H, Kirkwood JM. Prognostic significance of autoimmunity during treatment of melanoma with interferon. Semin Immunopathol. 2011;33:385–391. doi: 10.1007/s00281-011-0247-y.
    1. Moretta A. Natural killer cells and dendritic cells: rendezvous in abused tissues. Nat Rev Immunol. 2002;2(12):957–964. doi: 10.1038/nri956.
    1. Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, et al. Alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64:5934–5937. doi: 10.1158/0008-5472.CAN-04-1261.
    1. Curtsinger JM, Valenzuela JO, Agarwal P, Lins D, Mescher MF. Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation. J Immunol. 2005;174:4465–4469. doi: 10.4049/jimmunol.174.8.4465.
    1. Mohty M, Vialle-Castellano A, Nunes JA, Isnardon D, Olive D, Gaugler B. IFN-alpha skews monocyte differentiation into toll-like receptor 7-expressing dendritic cells with potent functional activities. J Immunol. 2003;171:3385–3393. doi: 10.4049/jimmunol.171.7.3385.
    1. Lapenta C, Santini SM, Spada M, Donati S, Urbani F, Accapezzato D, et al. IFN-alpha-conditioned dendritic cells are highly efficient in inducing cross-priming CD8(+) T cells against exogenous viral antigens. Eur J Immunol. 2006;36:2046–2060. doi: 10.1002/eji.200535579.
    1. Le Bon A, Etchart N, Rossmann C, Ashton M, Hou S, Gewert D, et al. Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat Immunol. 2003;4:1009–1015. doi: 10.1038/ni978.
    1. Butterfield LH, Gooding W, Whiteside TL. Development of a potency assay for human dendritic cells: IL-12p70 production. J Immunother. 2008;31:89–100. doi: 10.1097/CJI.0b013e318158fce0.
    1. Balch CM, Buzaid AC, Atkins MB, Cascinelli N, Coit DG, Fleming ID, et al. A new American joint committee on Cancer staging system for cutaneous melanoma. Cancer. 2000;88:1484–1491. doi: 10.1002/(SICI)1097-0142(20000315)88:6<1484::AID-CNCR29>;2-D.
    1. Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N, Coit DG, et al. New TNM melanoma staging system: linking biology and natural history to clinical outcomes. Semin Surg Oncol. 2003;21:43–52. doi: 10.1002/ssu.10020.
    1. Balch CM, Soong SJ, Atkins MB, Buzaid AC, Cascinelli N, Coit DG, et al. An evidence-based staging system for cutaneous melanoma. CA Cancer J Clin. 2004;54:131–149. doi: 10.3322/canjclin.54.3.131.
    1. Bedikian AY, Millward M, Pehamberger H, Conry R, Gore M, Trefzer U, et al. Bcl-2 antisense (oblimersen sodium) plus dacarbazine in patients with advanced melanoma: the Oblimersen melanoma study group. J Clin Oncol. 2006;24:4738–4745. doi: 10.1200/JCO.2006.06.0483.
    1. Tarhini AA, Cherian J, Moschos SJ, Tawbi HA, Shuai Y, Gooding WE, et al. Safety and efficacy of combination immunotherapy with interferon alfa-2b and tremelimumab in patients with stage IV melanoma. J Clin Oncol. 2012;30:322–328. doi: 10.1200/JCO.2011.37.5394.
    1. Yurkovetsky ZR, Kirkwood JM, Edington HD, Marrangoni AM, Velikokhatnaya L, Winans MT, et al. Multiplex analysis of serum cytokines in melanoma patients treated with interferon-alpha2b. Clin Cancer Res. 2007;13:2422–2428. doi: 10.1158/1078-0432.CCR-06-1805.
    1. Butterfield LH. Dendritic cells in cancer immunotherapy clinical trials: are we making progress? Front Immunol. 2013;4:454. doi: 10.3389/fimmu.2013.00454.
    1. Butterfield LH. Dendritic cell vaccines: release assays and potency assays. Telegraft. 2015;22:1–3.
    1. Butterfield LH. Cancer vaccines. BMJ. 2015;350:h988. doi: 10.1136/bmj.h988.
    1. Naveh HP, Vujanovic L, Butterfield LH. Cellular immunity induced by a recombinant adenovirus- human dendritic cell vaccine for melanoma. J Immunother Cancer. 2013;1:19. doi: 10.1186/2051-1426-1-19.
    1. Kuss I, Hathaway B, Ferris RL, Gooding W, Whiteside TL. Decreased absolute counts of T lymphocyte subsets and their relation to disease in squamous cell carcinoma of the head and neck. Clin Cancer Res. 2004;10:3755–3762. doi: 10.1158/1078-0432.CCR-04-0054.
    1. Mullins IM, Slingluff CL, Lee JK, Garbee CF, Shu J, Anderson SG, et al. CXC chemokine receptor 3 expression by activated CD8+ T cells is associated with survival in melanoma patients with stage III disease. Cancer Res. 2004;64:7697–7701. doi: 10.1158/0008-5472.CAN-04-2059.
    1. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18:1254–1261. doi: 10.1038/nm.2883.
    1. Retseck J, VanderWeele R, Lin HM, Lin Y, Butterfield LH, Tarhini AA. Phenotypic and functional testing of circulating regulatory T cells in advanced melanoma patients treated with neoadjuvant ipilimumab. J Immunother Cancer. 2016;4:38. doi: 10.1186/s40425-016-0141-1.
    1. Tarhini AA, Butterfield LH, Shuai Y, Gooding WE, Kalinski P, Kirkwood JM. Differing patterns of circulating regulatory T cells and myeloid-derived suppressor cells in metastatic melanoma patients receiving anti-CTLA4 antibody and interferon-alpha or TLR-9 agonist and GM-CSF with peptide vaccination. J Immunother. 2012;35:702–710. doi: 10.1097/CJI.0b013e318272569b.
    1. Tarhini AA, Edington H, Butterfield LH, Lin Y, Shuai Y, Tawbi H, et al. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One. 2014;9:e87705. doi: 10.1371/journal.pone.0087705.
    1. Zhou J, Mahoney KM, Giobbie-Hurder A, Zhao F, Lee S, Liao X, et al. Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade. Cancer Immunol Res. 2017;5:480–492. doi: 10.1158/2326-6066.CIR-16-0329.
    1. Tawbi HA, Burgess M, Bolejack V, Van Tine BA, Schuetze SM, Hu J, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18:1493–1501. doi: 10.1016/S1470-2045(17)30624-1.
    1. Astsaturov I, Petrella T, Bagriacik EU, de Benedette M, Uger R, Lumber G, et al. Amplification of virus-induced antimelanoma T-cell reactivity by high-dose interferon-alpha2b: implications for cancer vaccines. Clin Cancer Res. 2003;9:4347–4355.
    1. Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–336. doi: 10.1200/JCO.2010.30.7744.
    1. Mehrotra S, Chhabra A, Chakraborty A, Chattopadhyay S, Slowik M, Stevens R, et al. Antigen presentation by MART-1 adenovirus-transduced interleukin-10-polarized human monocyte-derived dendritic cells. Immunology. 2004;113:472–481. doi: 10.1111/j.1365-2567.2004.01978.x.
    1. He Y, Zhang J, Mi Z, Robbins P, Falo LD., Jr Immunization with lentiviral vector-transduced dendritic cells induces strong and long-lasting T cell responses and therapeutic immunity. J Immunol. 2005;174:3808–3817. doi: 10.4049/jimmunol.174.6.3808.
    1. Metharom P, Ellem KA, Wei MQ. Gene transfer to dendritic cells induced a protective immunity against melanoma. Cell Mol Immunol. 2005;2:281–288.
    1. Nakamura M, Iwahashi M, Nakamori M, Ueda K, Ojima T, Naka T, et al. Dendritic cells transduced with tumor-associated antigen gene elicit potent therapeutic antitumor immunity: comparison with immunodominant peptide-pulsed DCs. Oncology. 2005;68:163–170. doi: 10.1159/000086770.
    1. Yuan J, Latouche JB, Reagan JL, Heller G, Riviere I, Sadelain M, et al. Langerhans cells derived from genetically modified human CD34+ hemopoietic progenitors are more potent than peptide-pulsed Langerhans cells for inducing antigen-specific CD8+ cytolytic T lymphocyte responses. J Immunol. 2005;174:758–766. doi: 10.4049/jimmunol.174.2.758.
    1. Vujanovic L, Ballard W, Thorne SH, Vujanovic NL, Butterfield LH. Adenovirus-engineered human dendritic cells induce natural killer cell chemotaxis via CXCL8/IL-8 and CXCL10/IP-10. Oncoimmunology. 2012;1:448–457. doi: 10.4161/onci.19788.
    1. Vujanovic L, Szymkowski DE, Alber S, Watkins SC, Vujanovic NL, Butterfield LH. Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15. Blood. 2010;116:575–583. doi: 10.1182/blood-2009-08-240325.
    1. Ribas A, Butterfield LH, McBride WH, Dissette VB, Koh A, Vollmer CM, et al. Characterization of antitumor immunization to a defined melanoma antigen using genetically engineered murine dendritic cells. Cancer Gene Ther. 1999;6:523–536. doi: 10.1038/sj.cgt.7700076.
    1. Ribas A, Butterfield LH, McBride WH, Jilani SM, Bui LA, Vollmer CM, et al. Genetic immunization for the melanoma antigen MART-1/Melan-a using recombinant adenovirus-transduced murine dendritic cells. Cancer Res. 1997;57:2865–2869.
    1. Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS, et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med. 2006;12:1160–1166. doi: 10.1038/nm1475.
    1. Romero P, Zippelius A, Kurth I, Pittet MJ, Touvrey C, Iancu EM, et al. Four functionally distinct populations of human effector-memory CD8+ T lymphocytes. J Immunol. 2007;178:4112–4119. doi: 10.4049/jimmunol.178.7.4112.
    1. Wistuba-Hamprecht K, Martens A, Heubach F, Romano E, Geukes Foppen M, Yuan J, et al. Peripheral CD8 effector-memory type 1 T-cells correlate with outcome in ipilimumab-treated stage IV melanoma patients. Eur J Cancer. 2017;73:61–70. doi: 10.1016/j.ejca.2016.12.011.
    1. Sade-Feldman M, Kanterman J, Klieger Y, Ish-Shalom E, Olga M, Saragovi A, et al. Clinical significance of circulating CD33+CD11b+HLA-DR- myeloid cells in patients with stage IV melanoma treated with Ipilimumab. Clin Cancer Res. 2016;22:5661–5672. doi: 10.1158/1078-0432.CCR-15-3104.
    1. Kimura T, McKolanis JR, Dzubinski LA, Islam K, Potter DM, Salazar AM, et al. MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study. Cancer Prev Res (Phila) 2013;6:18–26. doi: 10.1158/1940-6207.CAPR-12-0275.
    1. Vujanovic L, Chuckran C, Lin Y, Ding F, Sander CA, Santos PM, Lohr J, Mashadi-Hossein A, Warren S, White A, Huang A, Kirkwood JM, Butterfield LH. CD56dim CD16- Natural Killer Cell Profiling in Melanoma Patients Receiving a Cancer Vaccine and Interferon-α. Front Immunol. 2019;10:14. eCollection 2019. PMID: 30761123. 10.3389/fimmu.2019.00014

Source: PubMed

3
Abonnieren