Tivozanib mediated inhibition of c-Kit/SCF signaling on Tregs and MDSCs and reversal of tumor induced immune suppression correlates with survival of HCC patients

Suresh Gopi Kalathil, Katy Wang, Alan Hutson, Renuka Iyer, Yasmin Thanavala, Suresh Gopi Kalathil, Katy Wang, Alan Hutson, Renuka Iyer, Yasmin Thanavala

Abstract

The immune modulatory effect of tivozanib, a tyrosine kinase inhibitor, and the underlying immune mechanisms impacting survival of HCC patients have not been investigated. Pre-clinical studies have shown that tivozanib reduces Tregs and MDSCs accumulation through inhibition of c-Kit/SCF axis. We rationalized that c-Kit/SCF axis antagonism by tivozanib may reverse tumor-induced immune suppression in HCC patients. The frequency of circulating Tregs, MDSCs, CTLA-4+Tregs, PD-1+T cells, c-Kit+pERK-2+Tregs, and c-Kit+pERK-2+MDSCs were quantified in HCC patients at baseline and two time points during tivozanib treatment. We report for the first time that reduction in Tregs after tivozanib treatment and increased levels of baseline CD4+PD-1+T cells correlated with significant improvement in overall survival (OS) of the patients and these signatures may be potential biomarkers of prognostic significance. This immune modulation resulted from tivozanib-mediated blockade of c-Kit/SCF signaling, impacting ERK2 phosphorylation on Tregs and MDSCs. Low pre-treatment CD4+T cells: Treg ratio and reduction in the frequencies of Foxp3+c-Kit+pERK+Tregs after tivozanib treatment correlated significantly with progression free survival. In a comparative analysis of tivozanib vs sorafenib treatment in HCC patients, we demonstrate that decrease in the baseline numbers or frequencies of Foxp3+Tregs, MDSCs and exhausted T cells was significantly greater following tivozanib treatment. Additionally, greater increase in CD4+T cell: Treg ratio after tivozanib treatment was associated with significant improvement in OS compared to sorafenib treatment, highlighting the greater efficacy of tivozanib. These insights may help identify patients who likely would benefit from c-Kit/SCF antagonism and inform efforts to improve the efficacy of tivozanib in combination with immunotherapy.

Trial registration: ClinicalTrials.gov NCT01835223.

Keywords: HCC; MDSC; Tivozanib; Tregs; c-Kit/SCF; survival.

© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.

Figures

Figure 1.
Figure 1.
Reduction in Tregs and dynamics of ERK phosphorylation on c-Kit+Tregs during tivozanib therapy (A) Representative histogram offset showing frequency of CD4+Foxp3+ Tregs measured at pre, 28–35d and 160d of tivozanib treatment. (B) Frequency and (C) absolute numbers of CD4+Foxp3+ Tregs pre vs 28–35d (D) Representative histogram offset showing frequency of Foxp3+c-Kit+ Tregs measured at pre, 28–35d and 160d of tivozanib treatment. (E) Frequency and (F) absolute numbers of Foxp3+c-Kit+ Tregs pre vs 28–35d (G) Representative histogram offset showing frequency of c-Kit+pPERK+ Tregs measured at pre, 28–35d and 160d of tivozanib treatment. (H) Frequency and (I) absolute numbers of c-Kit+pPERK+ Tregs pre vs 28–35d (J) Ratio of CD4+CD127+ T cells to CD4+Foxp3+ T cells (CD4+CD127+ T cells/CD4+Foxp3+ T cells) pre vs 28–35d. (K) Ratio of CD8+CD127+ T cells to CD4+Foxp3+ T cells pre vs 28–35d. Each symbol represents an individual HCC patient. Frequencies of Tregs and T effector cells were calculated based on CD3+CD4+ T/CD3+CD8+ T cell population. **** P < 0.0001, *** P < 0. 001, ** P < 0.01, * P < 0.05, paired t-test, Pre vs 28–35d n = 17.
Figure 2.
Figure 2.
Reduction in MDSC and dynamics of ERK phosphorylation on c-Kit+ MDSCs during tivozanib therapy. (A) Representative histogram offset showing frequency of CD11b+CD33+ MDSCs measured at pre, 28–35d and 160d of tivozanib treatment. (B) Frequency and (C) absolute numbers of MDSCs pre vs 28–35. (D) Representative histogram offset showing frequency of c-Kit+ MDSCs measured at pre, 28–35d and 160d of tivozanib treatment. (E) Frequency and (F) absolute numbers of c-Kit+ MDSCs pre vs 28–35. (G) Representative histogram offset showing frequency of c-Kit+pERK+ MDSCs measured at pre, 28–35d and 160d of tivozanib treatment. (H) Frequency and (I) absolute numbers of c-Kit+pERK+ MDSCs pre vs 28–35. Each symbol represents an individual HCC patient. Frequencies of MDSCs were calculated based on CD14−HLA-DR− population **** P < 0.0001, *** P < 0. 001, ** P < 0.01, * P < 0.05, paired t-test, n = 17.
Figure 3.
Figure 3.
Reduction in immune checkpoint receptors during and after tivozanib therapy. (A) Representative histogram offset showing frequency of Foxp3+CTLA-4+ Tregs measured at pre, 28–35d and 160d of tivozanib treatment. (B) Frequency and (C) absolute numbers of Foxp3+CTLA-4+ Tregs pre vs 28–35d. (D) Representative histogram offset showing frequency of CD4+PD-1+ T cells measured at pre, 28–35d and 160d of tivozanib treatment. (E) Frequency of CD4+PD-1+ T cells pre vs 28–35d (F) Absolute number of CD4+ PD-1+ T cells pre vs 28–35d (G) Representative histogram offset showing frequency of CD8+PD-1+ T cells measured at pre, 28–35d and 160d of tivozanib treatment. (H) Frequency of CD8+PD-1+ T cells pre vs 28–35d (I) Absolute number of CD8+PD-1+ T cells pre vs 28–35d. Each symbol represents an individual HCC patient. Frequencies of CTLA-4+Tregs and PD-1+ T cells were calculated based on CD3+CD4+ T cell/CD3+CD8+ T cell population and. **** P < 0.0001, *** P < 0. 001, ** P < 0.01, * P < 0.05, paired t-test, n = 17.
Figure 4.
Figure 4.
Kaplan-Meier plots showing the predictive immune correlates of responses to tivozanib treatment in HCC patient. Association between immunophenotypic signatures and OS or PFS of patients was calculated as described in methods. (A) OS of the patients and increased frequencies of baseline CD4+PD-1+ T cells (HR = 0.92, 95% CI: 0.9–1.0, P = 0.02). (B) Reduction in the frequencies of CD4+Foxp3+ Tregs (post- pre changes) (median reduction: −2.5, range: −7.6 to −0.8), quantified after tivozanib therapy and OS of the patients (HR = 1.6, 95% CI: 1.0–2.3, P = 0.03). (C) Low baseline ratio of CD4+T effector cells to Foxp3+ Tregs and OS of the patients (HR = 1.2, 95% CI: 1.0–1.5, P = 0.046) as well as (D) Low baseline ratio of CD4+T effector cells to Foxp3+ Tregs and PFS of the patients at 24 weeks (HR = 1.4, 95% CI: 1.0–1.9, P = 0.03). (E). Decrease in the frequencies of Foxp3+c-Kit+pERK+Tregs (post-pre changes) and PFS of the patients at 24 weeks (HR = 1.2, 95% CI: 1.0–1.5, P = 0.03).
Figure 5.
Figure 5.
Differential effect of tivozanib vs sorafenib treatment on immune suppressive cell subsets in HCC patients and impact of post-treatment changes in the ratio of CD4+T effector cell: Foxp3+ Tregs on overall survival of HCC patients after tivozanib vs sorafenib treatment. Box plots represent mean/median percentage changes from baseline number or frequency of different immune cell subsets after 28–25 days of sorafenib vs. tivozanib treatment (A) Foxp3+ Tregs (B) Foxp3+CTLA-4+ Tregs (C) CD4+PD-1+ T cells (D) CD8+PD-1+ T cells (E) % CD11b+CD33+ MDSC (F) # CD11b+CD33+ MDSC (G) Kaplan-Meir plots showing the association of greater percentage change or increase in the ratio of CD4+T cells: Tregs from baseline and survival probability of HCC patients after sorafenib or tivozanib treatment. P values are shown inside the respective plots; sorafenib n = 49, tivozanib n = 17.

References

    1. El-Serag HB, Kanwal F.. Epidemiology of hepatocellular carcinoma in the United States: where are we? Where do we go? Hepatology. 2014;60(5):1767–11. doi:10.1002/hep.27222.
    1. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, Gores G. Hepatocellular carcinoma. Nat Rev Dis Primers. 2016;2:16018.
    1. Regad T. Targeting RTK Signaling Pathways in Cancer. Cancers (Basel). 2015;7(3):1758–1784. doi:10.3390/cancers7030860.
    1. Du Z, Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17(1):58. doi:10.1186/s12943-018-0782-4.
    1. Momeny M, Moghaddaskho F, Gortany NK, Yousefi H, Sabourinejad Z, Zarrinrad G, Mirshahvaladi S, Eyvani H, Barghi F, Ahmadinia L, et al... Blockade of vascular endothelial growth factor receptors by tivozanib has potential anti-tumour effects on human glioblastoma cells. Sci Rep. 2017;7(1):44075. doi:10.1038/srep44075.
    1. Hepgur M, Sadeghi S, Dorff TB, Quinn DI. Tivozanib in the treatment of renal cell carcinoma. Biologics. 2013;7:139–148.
    1. Motzer RJ, Nosov D, Eisen T, Bondarenko I, Lesovoy V, Lipatov O, Tomczak P, Lyulko O, Alyasova A, Harza M, et al.. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol. 2013;31(30):3791–3799. doi:10.1200/JCO.2012.47.4940.
    1. Momeny M, Sabourinejad Z, Zarrinrad G, Moghaddaskho F, Eyvani H, Yousefi H, Mirshahvaladi S, Poursani EM, Barghi F, Poursheikhani A, et al.. Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci Rep. 2017;7(1):45954. doi:10.1038/srep45954.
    1. Benson AB, Kiss I, Bridgewater J, Eskens FALM, Sasse C, Vossen S, Chen J, Van Sant C, Ball HA, Keating A, et al.. BATON-CRC: A Phase II Randomized Trial Comparing Tivozanib Plus mFOLFOX6 with Bevacizumab Plus mFOLFOX6 in Stage IV Metastatic Colorectal Cancer. Clin Cancer Res. 2016;22(20):5058–5067. doi:10.1158/1078-0432.CCR-15-3117.
    1. Jamil MO, Hathaway A, Mehta A. Tivozanib: status of development. Curr Oncol Rep. 2015;17(6):24. doi:10.1007/s11912-015-0451-3.
    1. Escudier B, Porta C, Eisen T, Belsey J, Gibson D, Morgan J, Motzer R. The role of tivozanib in advanced renal cell carcinoma therapy. Expert Rev Anticancer Ther. 2018;11:1113–1114.
    1. Stankov K, Popovic S, Mikov M. C-KIT signaling in cancer treatment. Curr Pharm Des. 2014;20(17):2849–2880. doi:10.2174/13816128113199990593.
    1. Ali S, Ali S. Role of c-kit/SCF in cause and treatment of gastrointestinal stromal tumors (GIST). Gene. 2007;401(1–2):38–45. doi:10.1016/j.gene.2007.06.017.
    1. Foster BM, Zaidi D, Young TR, Mobley ME, Kerr BA. CD117/c-kit in Cancer Stem Cell-Mediated Progression and Therapeutic Resistance. Biomedicines. 2018;6.
    1. Abbaspour BM, Kamalidehghan B, Saleem M, Huri HZ, Ahmadipour F. Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. Drug Des Devel Ther. 2016;10:2443–2459. doi:10.2147/DDDT.S89114.
    1. Pan PY, Wang GX, Yin B, Ozao J, Ku T, Divino CM, Chen SH. Reversion of immune tolerance in advanced malignancy: modulation of myeloid-derived suppressor cell development by blockade of stem-cell factor function. Blood. 2008;111(1):219–228. doi:10.1182/blood-2007-04-086835.
    1. 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. 2011;77(1):12–19. doi:10.1016/j.critrevonc.2010.02.004.
    1. Ozao-Choy J, Ma G, Kao J, Wang GX, Meseck M, Sung M, Schwartz M, Divino CM, Pan PY, Chen SH. 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. 2009;69(6):2514–2522. doi:10.1158/0008-5472.CAN-08-4709.
    1. Pawlowski N, Hoerzer H, Singh HJ, Hilf N. Impact of various first- and second-generation tyrosine-kinase inhibitors on frequency and functionality of immune cells. Proc 104th Annu Meeting Am Assoc Cancer Res. 2013;73:8.
    1. Immune Checkpoint KM. Inhibition in Hepatocellular Carcinoma: Basics and Ongoing Clinical Trials. Oncology. 2017;92(Suppl 1):50–62. doi:10.1159/000451016.
    1. Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12:681–700.
    1. Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y. Higher Frequencies of GARP(+)CTLA-4(+)Foxp3(+) T Regulatory Cells and Myeloid-Derived Suppressor Cells in Hepatocellular Carcinoma Patients Are Associated with Impaired T-Cell Functionality. Cancer Res. 2013;73(8):2435–2444. doi:10.1158/0008-5472.CAN-12-3381.
    1. Lugade AA, Kalathil S, Miller A, Iyer R, Thanavala Y. High immunosuppressive burden in advanced hepatocellular carcinoma patients: Can effector functions be restored? Oncoimmunology. 2013;2(7):e24679. doi:10.4161/onci.24679.
    1. Kalathil SG, Lugade AA, Miller A, Iyer R, Thanavala YPD. 1(+) and Foxp3(+) T cell reduction correlates with survival of HCC patients after sorafenib therapy. JCI Insight. 2016;1:11:e86182.
    1. Fountzilas C, Gupta M, Lee S, Krishnamurthi S, Estfan B, Wang K, Attwood K, Wilton J, Bies R, Bshara W, et al.. A multicentre phase 1b/2 study of tivozanib in patients with advanced inoperable hepatocellular carcinoma. Br J Cancer. 2020;122:963–970. doi:10.1038/s41416-020-0737-6.
    1. Bukowski RM. Third generation tyrosine kinase inhibitors and their development in advanced renal cell carcinoma. Front Oncol. 2012;2:13. doi:10.3389/fonc.2012.00013.
    1. Nosov DA, Esteves B, Lipatov ON, Lyulko AA, Anischenko AA, Chacko RT, Doval DC, Strahs A, Slichenmyer WJ, Bhargava P. Antitumor activity and safety of tivozanib (AV-951) in a phase II randomized discontinuation trial in patients with renal cell carcinoma. J Clin Oncol. 2012;30:1678–1685. doi:10.1200/JCO.2011.35.3524.
    1. Rini BI, Pal SK, Escudier BJ, Atkins MB, Hutson TE, Porta C, Verzoni E, Needle MN, McDermott DF. Tivozanib versus sorafenib in patients with advanced renal cell carcinoma (TIVO-3): a phase 3, multicentre, randomised, controlled, open-label study. Lancet Oncol. 2020;21(1):95–104. doi:10.1016/S1470-2045(19)30735-1.
    1. Zhu AX, Kang Y-K, Yen C-J, Finn RS, Galle PR, Llovet JM, Assenat E, Brandi G, Pracht M, Lim HY, et al.. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20:282–296. doi:10.1016/S1470-2045(18)30937-9.
    1. Kalathil SG, Hutson A, Barbi J, Iyer R, Thanavala Y. Augmentation of IFN-γ+ CD8+ T cell responses correlates with survival of HCC patients on sorafenib therapy. JCI Insight. 2019;4(15):15:e130116. doi:10.1172/jci.insight.130116.
    1. Lee WC, Wang YC, Cheng CH, Wu TH, Lee CF, Wu TJ, Chou HS, Chan KM. Myeloid-derived suppressor cells in the patients with liver resection for hepatitis B virus-related hepatocellular carcinoma. Sci Rep. 2019;9:2269. doi:10.1038/s41598-019-38785-3.
    1. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, et al.. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382:1894–1905. doi:10.1056/NEJMoa1915745.
    1. Lee MS, Ryoo BY, Hsu CH, Numata K, Stein S, Verret W, Hack SP, Spahn J, Liu B, Abdullah H, et al.. Atezolizumab with or without bevacizumab in unresectable hepatocellular carcinoma (GO30140): an open-label, multicentre, phase 1b study. Lancet Oncol. 2020;21:808–820. doi:10.1016/S1470-2045(20)30156-X.
    1. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018;391(10127):1301–1314. doi:10.1016/S0140-6736(18)30010-2.
    1. Kalathil SG, Lugade AA, Pradhan V, Miller A, Parameswaran GI, Sethi S, Thanavala Y. T-Regulatory Cells and Programmed Death 1 + T Cells Contribute to Effector T-Cell Dysfunction in Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2014;190(1):40–50. doi:10.1164/rccm.201312-2293OC.

Source: PubMed

3
購読する