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

Targeting KIT on innate immune cells to enhance the antitumor activity of checkpoint inhibitors

    Maximilian Stahl

    Department of Internal Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, USA

    ,
    Richard Gedrich

    Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA

    ,
    Ronald Peck

    Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA

    ,
    Theresa LaVallee

    Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA

    &
    Joseph Paul Eder

    *Author for correspondence:

    E-mail Address: joseph.eder@yale.edu

    Department of Medical Oncology & Yale Cancer Center, Yale School of Medicine, 333 Cedar Street, WWW211, New Haven, CT 06520, USA

    Published Online:https://doi.org/10.2217/imt-2016-0040

    Innate immune cells such as mast cells and myeloid-derived suppressor cells are key components of the tumor microenvironment. Recent evidence indicates that levels of myeloid-derived suppressor cells in melanoma patients are associated with poor survival to checkpoint inhibitors. This suggests that targeting both the innate and adaptive suppressive components of the immune system will maximize clinical benefit and elicit more durable responses in cancer patients. Preclinical data suggest that targeting signaling by the receptor tyrosine kinase KIT, particularly on mast cells, may modulate innate immune cell numbers and activity in tumors. Here, we review data highlighting the importance of the KIT signaling in regulating antitumor immune responses and the potential benefit of combining selective KIT inhibitors with immune checkpoint inhibitors.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Lennartsson J, Ronnstrand L. The stem cell factor receptor/c-Kit as a drug target in cancer. Curr. Cancer Drug Targets 6(1), 65–75 (2006).
    • 2 Yuzawa S, Opatowsky Y, Zhang Z, Mandiyan V, Lax I, Schlessinger J. Structural basis for activation of the receptor tyrosine kinase KIT by stem cell factor. Cell 130(2), 323–334 (2007).
    • 3 Turin L, Acocella F, Stefanello D et al. Expression of c-kit proto-oncogene in canine mastocytoma: a kinetic study using real-time polymerase chain reaction. J. Vet. Diagn. Invest. 18(4), 343–349 (2006).
    • 4 Xu Z, Huo X, Tang C et al. Frequent KIT mutations in human gastrointestinal stromal tumors. Sci. Rep. 4, 5907 (2014).
    • 5 Slipicevic A, Herlyn M. KIT in melanoma: many shades of gray. J. Invest. Dermatol. 135(2), 337–338 (2015).
    • 6 Advani AS. C-kit as a target in the treatment of acute myelogenous leukemia. Curr. Hematol. Rep. 4(1), 51–58 (2005).
    • 7 Butt AQ, Mills KH. Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines. Oncogene 33(38), 4623–4631 (2014).
    • 8 Galli SJ, Tsai M. Mast cells in allergy and infection: versatile effector and regulatory cells in innate and adaptive immunity. Eur. J. Immunol. 40(7), 1843–1851 (2010).
    • 9 Oldford SA, Marshall JS. Mast cells as targets for immunotherapy of solid tumors. Mol. Immunol. 63(1), 113–124 (2015).
    • 10 Huang B, Lei Z, Zhang GM et al. SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment. Blood 112(4), 1269–1279 (2008).
    • 11 Yang Z, Zhang B, Li D et al. Mast cells mobilize myeloid-derived suppressor cells and Treg cells in tumor microenvironment via IL-17 pathway in murine hepatocarcinoma model. PLoS ONE 5(1), e8922 (2010).
    • 12 Pan PY, Wang GX, Yin B et al. Reversion of immune tolerance in advanced malignancy: modulation of myeloid-derived suppressor cell development by blockade of stem-cell factor function. Blood 111(1), 219–228 (2008). •• This important study demonstrated that the blockade of the stem cell factor (SCF) receptor (KIT)–SCF interaction by an anti-c-kit antibody prevents tumor-specific T-cell anergy, Treg development and tumor angiogenesis. Furthermore, it showed a synergistic therapeutic effect in prevention of myeloid-derived suppressor cell (MDSC) accumulation in conjunction with immune activation therapy in mice bearing large tumors.
    • 13 Starkey JR, Crowle PK, Taubenberger S. Mast-cell-deficient W/Wv mice exhibit a decreased rate of tumor angiogenesis. Int. J. Cancer 42(1), 48–52 (1988).
    • 14 Coussens LM, Raymond WW, Bergers G et al. Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev. 13(11), 1382–1397 (1999).
    • 15 Soucek L, Lawlor ER, Soto D, Shchors K, Swigart LB, Evan GI. Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors. Nat. Med. 13(10), 1211–1218 (2007).
    • 16 Wasiuk A, Dalton DK, Schpero WL, Stan RV, Conejo-Garcia JR, Noelle RJ. Mast cells impair the development of protective anti-tumor immunity. Cancer Immunol. Immunother. 61(12), 2273–2282 (2012). • The authors used the syngeneic MB49 tumor model in mast cell-deficient KIT mutant (c-KitWsh) mice to show that mast cells are required for immune suppression in tumors. c-KitWsh mice controlled tumor growth and exhibited enhanced survival.
    • 17 Saleem SJ, Martin RK, Morales JK et al. Cutting edge: mast cells critically augment myeloid-derived suppressor cell activity. J. Immunol. 189(2), 511–515 (2012). •• The authors used the syngeneic B16 melanoma model in mast cell-deficient KitWsh/Wsh mice to show that mast cells are required for enhanced lung metastasis following adoptive transfer of MDSCs from tumor bearing mice, suggesting interactions between mast cells and MDSCs in promoting tumor progression and metastasis.
    • 18 Danelli L, Frossi B, Gri G et al. Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment. Cancer Immunol. Res. 3(1), 85–95 (2015). •• The authors presented data suggesting direct interactions between mast cells and MDSCs in C26 tumors via CD40:CD40L and that mast cells augmented the ability of monocytic MDSCs to inhibit CD4+ T-cell proliferation in vitro, indicating mast cells can modulate the immunosuppressive effects of monocytic MDSCs.
    • 19 Tsai MJ, Chang WA, Huang MS, Kuo PL. Tumor microenvironment: a new treatment target for cancer. ISRN Biochem. 2014, 351959 (2014).
    • 20 Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14(10), 1014–1022 (2013).
    • 21 Galon J, Costes A, Sanchez-Cabo F et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313(5795), 1960–1964 (2006).
    • 22 Azimi F, Scolyer RA, Rumcheva P et al. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J. Clin. Oncol. 30(21), 2678–2683 (2012).
    • 23 Mahmoud SM, Paish EC, Powe DG et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J. Clin. Oncol. 29(15), 1949–1955 (2011).
    • 24 Galon J, Pages F, Marincola FM et al. Cancer classification using the Immunoscore: a worldwide task force. J. Transl. Med. 10, 205 (2012).
    • 25 Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J. Clin. Invest. 125(9), 3384–3391 (2015).
    • 26 Wang XF, Wang HS, Wang H et al. The role of indoleamine 2,3-dioxygenase (IDO) in immune tolerance: focus on macrophage polarization of THP-1 cells. Cell. Immunol. 289(1–2), 42–48 (2014).
    • 27 Byrne WL, Mills KH, Lederer JA, O'sullivan GC. Targeting regulatory T cells in cancer. Cancer Res. 71(22), 6915–6920 (2011).
    • 28 Topalian SL, Hodi FS, Brahmer JR et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366(26), 2443–2454 (2012).
    • 29 Larkin J, Hodi FS, Wolchok JD. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373(13), 1270–1271 (2015).
    • 30 Garon EB, Rizvi NA, Hui R et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372(21), 2018–2028 (2015).
    • 31 Powles T, Eder JP, Fine GD et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515(7528), 558–562 (2014).
    • 32 Motzer RJ, Escudier B, Mcdermott DF et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373(19), 1803–1813 (2015).
    • 33 Sonnenfeld A. Leukamische reaktiones bei carcinoma. Zeitschrift f Klin Med. 111, 108 (1929).
    • 34 Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 13(10), 739–752 (2013).
    • 35 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9(3), 162–174 (2009).
    • 36 Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J. Immunol. 181(8), 5791–5802 (2008).
    • 37 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. 172(2), 989–999 (2004).
    • 38 Vasquez-Dunddel D, Pan F, Zeng Q et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J. Clin. Invest. 123(4), 1580–1589 (2013).
    • 39 Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells – an overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 4(1), e954829 (2015).
    • 40 Kao J, Ko EC, Eisenstein S, Sikora AG, Fu S, Chen SH. Targeting immune suppressing myeloid-derived suppressor cells in oncology. Crit. Rev. Oncol. Hematol. 77(1), 12–19 (2011).
    • 41 Kusmartsev S, Cheng F, Yu B et al. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res. 63(15), 4441–4449 (2003).
    • 42 Talmadge JE, Hood KC, Zobel LC, Shafer LR, Coles M, Toth B. Chemoprevention by cyclooxygenase-2 inhibition reduces immature myeloid suppressor cell expansion. Int. Immunopharmacol. 7(2), 140–151 (2007).
    • 43 De Santo C, Serafini P, Marigo I et al. Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc. Natl Acad. Sci. USA 102(11), 4185–4190 (2005).
    • 44 Kusmartsev S, Eruslanov E, Kubler H et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: link to tumor-induced immune suppression in renal cell carcinoma. J. Immunol. 181(1), 346–353 (2008).
    • 45 Lennartsson J, Ronnstrand L. Stem cell factor receptor/c-Kit: from basic science to clinical implications. Physiol. Rev. 92(4), 1619–1649 (2012).
    • 46 Larmonier N, Janikashvili N, Lacasse CJ et al. Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL- tumors. J. Immunol. 181(10), 6955–6963 (2008).
    • 47 Ozao-Choy J, Ma G, Kao J et al. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res. 69(6), 2514–2522 (2009).
    • 48 Finke J, Ko J, Rini B, Rayman P, Ireland J, Cohen P. MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy. Int. Immunopharmacol. 11(7), 856–861 (2011).
    • 49 Balachandran VP, Cavnar MJ, Zeng S et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat. Med. 17(9), 1094–1100 (2011).
    • 50 Christiansson L, Soderlund S, Mangsbo S et al. The tyrosine kinase inhibitors imatinib and dasatinib reduce myeloid suppressor cells and release effector lymphocyte responses. Mol. Cancer Ther. 14(5), 1181–1191 (2015). •• This study demonstrated that both imatinib and dasatinib promote immune stimulation in the immunosuppressive CML tumor milieu by reduction of CD11b(+)CD14(-)CD33(+) myeloid-derived suppressor cells. It suggested the combination of TKIs with cancer immunotherapy.
    • 51 Santegoets SJ, Stam AG, Lougheed SM 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 2, 31 (2014).
    • 52 Chang DZ, Ma Y, Ji B et al. Mast cells in tumor microenvironment promotes the in vivo growth of pancreatic ductal adenocarcinoma. Clin. Cancer. Res. 17(22), 7015–7023 (2011).
    • 53 Groot Kormelink T, Abudukelimu A, Redegeld FA. Mast cells as target in cancer therapy. Curr. Pharm. Des. 15(16), 1868–1878 (2009).
    • 54 Von Mehren M. Beyond imatinib: second generation c-KIT inhibitors for the management of gastrointestinal stromal tumors. Clin. Colorectal Cancer 6(Suppl. 1), S30–S34 (2006).
    • 55 Kee D, Zalcberg JR. Current and emerging strategies for the management of imatinib-refractory advanced gastrointestinal stromal tumors. Ther. Adv. Med. Oncol. 4(5), 255–270 (2012).
    • 56 Reshetnyak AV, Nelson B, Shi X et al. Structural basis for KIT receptor tyrosine kinase inhibition by antibodies targeting the D4 membrane-proximal region. Proc. Natl Acad. Sci. USA 110(44), 17832–17837 (2013). •• Malignancies with somatic oncogenic mutations in the receptor tyrosine kinase KIT function can be treated with tyrosine kinase inhibitors but drug resistance followed by clinical progression of disease is frequent. This paper describes inhibitory KIT antibodies that bind to the membrane-proximal Ig-like D4 of KIT with significant overlap with an epitope in D4 that mediates homotypic interactions essential for KIT activation. By targeting the extracellular region of KIT mutants resistant to traditional tyrosine kinase inhibitors can be inhibited.
    • 57 Lebron MB, Brennan L, Damoci CB et al. A human monoclonal antibody targeting the stem cell factor receptor (c-Kit) blocks tumor cell signaling and inhibits tumor growth. Cancer Biol. Ther. 15(9), 1208–1218 (2014).
    • 58 London C, Rippy S, Gardner S et al. KTN0158, a humanized anti-KIT monoclonal antibody, demonstrates antitumor activity in dogs with mast cell tumors. Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics. Mol. Cancer Ther. 14(12 Suppl. 2), Abstract nr C167 (2015).
    • 59 Gedrich R, Seibel S, Lavallee L, Eder JP. Targeting KIT on innate immune cells enhances the antitumor activity of checkpoint inhibitors in vivo. J. Immunother. Cancer 3(Suppl. 2), O12 (2015). •• In this oral presentation, the authors showed that the combination of an antimouse KIT monoclonal antibody and anti-CTLA-4 had substantial antitumor effects in the Colon26 model, comparable to those observed for anti-CTLA-4 plus anti-PD1. Anti-KIT antibody treatment resulted in substantial decreases in monocytic MDSCs.
    • 60 Kitano S, Postow MA, Ziegler CG et al. Computational algorithm-driven evaluation of monocytic myeloid-derived suppressor cell frequency for prediction of clinical outcomes. Cancer Immunol. Res. 2(8), 812–821 (2014). • This manuscript showed that patients with a pretreatment monocytic MDSC frequency outside a preliminary definition of healthy donor range (<14.9%) were significantly more likely to achieve prolonged overall survival following treatment with ipilimumab. These observations suggest that MDSC frequency is a novel prognostic indicator of overall survival in patients with metastatic melanoma treated with ipilimumab.
    • 61 Martens A, Wistuba-Hamprecht K, Geukes Foppen MH et al. Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin. Cancer. Res. doi:10.1158/1078-0432.CCR-15-2412 (2016) (Epub ahead of print).
    • 62 Weber JS, Geoffrey Thomas G, Yu B et al. Survival, biomarker, and toxicity analysis of nivolumab (NIVO) in patients that progressed on ipilimumab (IPI). J. Clin. Oncol. 33, Abstract 9055 (2015).
    • 63 Le Tourneau C, Raymond E, Faivre S. Sunitinib: a novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Ther. Clin. Risk Manag. 3(2), 341–348 (2007).