Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1486 word(s) 1486 2020-12-11 07:11:42 |
2 format correct -21 word(s) 1465 2020-12-18 04:25:24 | |
3 format correct -21 word(s) 1465 2020-12-18 04:27:19 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kudo, M. PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors. Encyclopedia. Available online: https://encyclopedia.pub/entry/3656 (accessed on 28 March 2024).
Kudo M. PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors. Encyclopedia. Available at: https://encyclopedia.pub/entry/3656. Accessed March 28, 2024.
Kudo, Masatoshi. "PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors" Encyclopedia, https://encyclopedia.pub/entry/3656 (accessed March 28, 2024).
Kudo, M. (2020, December 17). PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors. In Encyclopedia. https://encyclopedia.pub/entry/3656
Kudo, Masatoshi. "PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors." Encyclopedia. Web. 17 December, 2020.
PD-1/PD-L1 antibody plus Anti-VEGF Inhibitors
Edit

A successful phase III trial for the combination of atezolizumab and bevacizumab (the IMbrave150 trial) in advanced hepatocellular carcinoma has recently been reported to show better survival benefit over sorafenib, standard of care for more than 12 years. This is a practice changing results and scientific rationale of this combination, PD-1/PD-L1 antibody plus anti-VEGF inhibitors is very important.

hepatocellular carcinoma immune checkpoint inhibitor PD-1 antibody PD-L1 antibody anti-VEGF inhibitor

1. Introduction

At the European Society for Medical Oncology (ESMO) Asia in November 2019, the positive results of the IMbrave150 study, a trial which compared the effects of the combination of atezolizumab and bevacizumab with those of sorafenib [1], drew attention to the possibility of immunotherapy with a combination of programmed cell death-1 (PD-1)/programmed death ligand 1 (PD-L1) and vascular endothelial growth factor (VEGF) inhibitors. This review outlines the scientific rationale for the therapeutic combination of PD-1/PD-L1 and VEGF antibodies, proof-of-concept results of the phase Ib trial, and results of other phase Ib trials for similar combination strategies.

2. The Rationale Underlying the Combination of PD-1/PD-L1 and VEGF Inhibitors

At tumor sites, VEGF released by hypoxic cancer cells and vascular endothelial cells promotes tumor growth, invasion, and metastasis by increasing neovascularization . Simultaneously, VEGF enhances the mobilization and proliferation of various cells, including regulatory T cells (Tregs), and the release of immunosuppressive cytokines [2][3]. It also enhances the mobilization of tumor-associated macrophages (TAMs) and their polarization to an M2 phenotype. Tregs and TAMs promote tumor growth through the release of VEGF and angiopoietin-2, among other mechanisms [4]. VEGF can also activate myeloid-derived suppressor cells (MDSCs), which in turn release more VEGF [4]. Furthermore, VEGF inhibits dendritic cell maturation and antigen presentation in the priming phase. Thus, VEGF reduces the proliferation and activation of naive CD8+ cells by suppressing dendritic cell activity even in the presence of neoantigens [4] (Figure 1). VEGF-induced Tregs, TAMs, and MDSCs reduce the proliferation and function of CD8+ cells. VEGF also prevents antigen-activated CD8+ cells from infiltrating the tumor tissue through its effects on tumor angiogenesis. In addition, VEGF creates a microenvironment that inhibits the function of T cells in the tumor during the effector phase of the immune response [4]. Furthermore, immunosuppressive cells (Tregs, TAMs, and MDSCs) promote immune escape by releasing immunosuppressive cytokines, including interleukin (IL)-10 and transforming growth factor beta (TGF-β), and by inhibiting dendritic cell maturation and activation, NK cell activation, and T cell activation and proliferation [2][3][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25] (Figure 1). The cancer immunity cycle begins with the uptake and presentation of neoantigens released from necrotic tumor cells by dendritic cells. This is followed by seven steps: (1) tumor antigen release, (2) tumor antigen uptake and presentation by dendritic cells, (3) T cell priming and activation, (4) T cell migration to the tumor, (5) T cell invasion of the tumor, (6) cancer cell recognition by T cells, and (7) attack on tumor cells by T cells, which leads to cancer cell death and release of additional tumor antigens [5] (Figure 2). VEGF promotes immune escape at almost every step of the cancer immunity cycle [6][7][8][9]. Furthermore, hepatic interstitial cells such as Kupffer cells, liver endothelial cells, and hepatic stellate cells are involved in maintaining immune tolerance in the healthy liver and may contribute to the immunosuppressive microenvironment in hepatocellular carcinoma [26].

Figure 1. Immune suppressive microenvironment induced by VEGF (modified from ref. [4] with permission).

Figure 2. The Cancer-Immunity Cycle (modified from ref. [5] with permission).

The administration of molecular targeted drugs that inhibit VEGF activity, such as multi-kinase inhibitors that inhibit VEGF receptors, leads to an increase in antigen presentation by dendritic cells [8]. These drugs also promote T cell activation in the priming phase [8] and improve the migration of T cells from the lymph nodes to the tumor site by normalizing the tumor vasculature [15]. In addition, these drugs have been found to suppress the generation of Tregs, TAMs, and MDSCs at the tumor site, and to negatively regulate the expression of immunosuppressive cytokines such as TGF-β and IL-10 [10]. VEGF inhibitors therefore reprogram the immunosuppressive tumor microenvironment into an immunostimulatory environment [8]. The administration of PD-1/PD-L1 antibodies under such conditions enhances the antitumor activity of T cells (Figures 3 and 4). As described above, the combination of VEGF and PD-1/PD-L1 inhibitors promotes antitumor immunity according to the four Rs. First, a reversal of the VEGF-mediated inhibition of dendritic cell maturation results in the effective priming and activation of T cells (Recognition) [9]. Second, anti-VEGF antibodies normalize the tumor vasculature and promote the effective infiltration of T cells into the tumor (Recruitment) [15]. Third, anti-VEGF antibodies inhibit the activity of MDSCs, Tregs, and TAMs, leading to the reprogramming of the immunosuppressive microenvironment into an immunostimulatory microenvironment (Reprogramming) [6]. Fourth, PD-1/PD-L1 antibodies enhance the ability of T cells to attack tumor cells (Restoration) (Figure 3). These four Rs lead to efficient cancer immunity and tumor growth inhibition. Proteins released by the killed tumor cells are taken up by dendritic cells, and then processed into tumor antigen peptides that are presented on major histocompatibility complex (MHC) class I molecules, leading to a progression through the cancer immunity cycle and further tumor attacks [5] (Figure 2). As described above, normalization of the VEGF-suppressed tumor microenvironment with molecular targeted agents against VEGF leads to the efficient attack on tumors by activated T cells [27] (Figures 2 and 4). In addition, non-clinical study of lenvatinib, a tyrosine kinase inhibitor (TKI), showed that the inhibition of VEGF activity reduced TAMs and Tregs in the tumor microenvironment, leading to a decrease in TGF-β and IL-10, a decreased expression of T cell exhaustion markers such as PD-1 and TIM-3, and an increased expression of immunostimulatory cytokines such as IL-12 [28][29][30][31]. These findings form the rationale for a trial of the combination of TKIs and anti-PD-1/PD-L1 antibodies.

Figure 3. Anti-VEGF antibody reprograms the tumor microenvironment from immune suppressive to immune permissive (modified from ref. [4] with permission).

Figure 4. Scientific rationale of Immune-checkpoint Inhibitors plus Anti-VEGF: 4 Roles of anti-VEGF inhibitors in Cancer Immunity cycle, Recognise, Recruitment, Reprogramme, and Restore (original Figure).

3. Results of Phase Ib Studies of Other Combinations of PD-1/PD-L1 Antibodies and VEGF Inhibitors

In addition to the trial of atezolizumab and bevacizumab described above, other studies are examining the efficacy of combined PD-1/PD-L1 and VEGF inhibition. One such study, the LEAP-002 study, is a phase III clinical trial of pembrolizumab and lenvatinib [32][35][37][38][39][40][41]. This trial is ongoing and the results are highly anticipated. In addition, multiple other clinical trials of immune checkpoint inhibitors and VEGF inhibitors have been completed (Table 1). The number of patients who received pembrolizumab and lenvatinib (n = 67) was lower than the number of patients who received atezolizumab and bevacizumab in Arm A of the phase Ib trial described above (n = 104). The ORR (40.3%), DCR (85.1%), PFS (9.7 months), and OS (20.4 months) of the combination of pembrolizumab and lenvatinib were higher than those of the combination of atezolizumab and bevacizumab [42]. Furthermore, the efficacy of the combination of nivolumab and lenvatinib (evaluated by an independent imaging committee based on RECIST 1.1), which was recently reported at the annual meeting of the American Society of Clinical Oncology, Gastrointestinal Cancers (ASCO GI), was higher than that of the other two combination therapies (ORR, 54.2%; DCR, 91.7%; PFS, 7.4 months; and OS, not reached) [43]. Of course, it is not adequate to compare the results of single-arm trials with different patient populations, small sample sizes, and short observation periods. However, the results are very promising. The ORR and PFS of the combination of camerelizumab and apatinib were 38.9% and 7.2 months, respectively [44]. However, there have been no updated reports on this combination. Moreover, the reported results of the combination of avelumab and axitinib [45] were slightly inferior to those of other combination therapies (ORR, 13.6%; PFS, 5.5 months; and OS, 12.7 months, based on RECIST 1.1). Therefore, at present, the most promising ongoing trial is the LEAP-002 study [40][41]. The decision whether or not to proceed to phase III trials of the combination of nivolumab and lenvatinib has currently drawn attention. In any case, the efficacy of all other combinations of anti-PD-1/PD-L1 antibodies and TKIs or anti-VEGF antibodies, except for the combination of avelumab and axitinib, is higher than that of nivolumab (a PD-1 antibody) alone (ORR, 15%; DCR, 55%; PFS, 3.7 months; and OS, 16.4 months) [34] or pembrolizumab alone (ORR, 18.3%; DCR, 62.2%; PFS, 3.0 months; OS, 13.9 months) [36]. Therefore, combined immunotherapy is expected to shift the paradigm as a first-line treatment option in advanced hepatocellular carcinoma [46].

Table 1. Efficacy of Immune Checkpoint Inhibitors and Combination Immunotherapy with VEGF Antibodies/Tyrosine Kinase Inhibitors in Phase 1b Trials according to RECIST 1.1.

Efficacy

Anti-PD-1 Monotherapy (Phase 3 Trial)

Anti-PD-1/PD-L1 plus TKI/Anti-VEGF (Phase 1b trial)

 

Nivolumab [34]

(n = 214)

Pembrolizumab [36]

(n = 278)

Atezolizumab + bevacizumab [33]

(n = 104)

Pembrolizumab + Lenvatinib [42]

(n = 67)

Camrelizumab + apatinib [44]

(n = 18)

Avelumab + axitinib [45]

(n = 22)

Nivolumab + Lenvatinib [43]

(n = 24)

ORR (95% CI)

15%

18.3% (14.0–23.4)

36% (26–46)

40.3% (28.5–53.0)

38.9%

13.6% (2.9–34.9)

54.2% (32.8–74.4)

DCR (95% CI)

55%

62.2%

71%

85.1% (74.3–92.6)

83.3%

68.2% (45.1–86.1)

91.7% (73.0–99.0)

PFS, months (95% CI)

3.7 (3.1–3.9)

3.0 (2.8–4.1)

7.4 (5.6–10.7)

9.7 (5.3–13.8)

7.2 (2.6–NE)

5.5 (1.9–7.4)

7.4 (3.7–NE)

OS, months (95% CI)

16.4 (13.9–18.4)

13.9 (11.6–16.0)

17.1 (13.8–NE)

 20.4 (11.0–NE)

NR

12.7 (8.0–NE)

NR

DOR, months (M)

 23.3 (3.1–34.5+)

13.8 (1.5–23.6)

NE (11.7–NE)

11.0 (5.6–11.0)

NA

5.5 (3.7–7.3)

NA

DCR, disease control rate; DOR, duration of response; NA, not available; NE; not evaluable; NR, not reached; ORR, objective response rate (RECIST 1.1); OS, overall survival; PFS, progression-fee survival. TKI, tyrosine kinase inhibitor.

References

  1. Cheng, A.L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Zhu, A.X.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; et al. IMbrave150: Efficacy and safety results from a ph III study evaluating atezolizumab (atezo) + bevacizumab (bev) vs sorafenib (Sor) as first treatment (tx) for patients (pts) with unresectable hepatocellular carcinoma (HCC). Ann. Oncol. 2019, 30 (Suppl. 9), ix186–ix187.
  2. Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat. Rev. Clin. Oncol. 2018, 15, 325–340, doi:10.1038/nrclinonc.2018.29.
  3. Chouaib, S.; Messai, Y.; Couve, S.; Escudier, B.; Hasmim, M.; Noman, M.Z. Hypoxia promotes tumor growth in linking angiogenesis to immune escape. Front. Immunol. 2012, 3, 21, doi:10.3389/fimmu.2012.00021.
  4. Voron, T.; Marcheteau, E.; Pernot, S.; Colussi, O.; Tartour, E.; Taieb, J.; Terme, M. Control of the immune response by pro-angiogenic factors. Front. Oncol. 2014, 4, 70, doi:10.3389/fonc.2014.00070.
  5. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10, doi:10.1016/j.immuni.2013.07.012.
  6. Hegde, P.S.; Wallin, J.J.; Mancao, C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin. Cancer Biol. 2018, 52, 117–124, doi:10.1016/j.semcancer.2017.12.002.
  7. Ferrara, N.; Hillan, K.J.; Gerber, H.P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400, doi:10.1038/nrd1381.
  8. Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 1996, 2, 1096–1103, doi:10.1038/nm1096-1096.
  9. Gabrilovich, D.; Ishida, T.; Oyama, T.; Ran, S.; Kravtsov, V.; Nadaf, S.; Carbone, D.P. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 1998, 92, 4150–4166.
  10. Elovic, A.E.; Ohyama, H.; Sauty, A.; McBride, J.; Tsuji, T.; Nagai, M.; Weller, P.F.; Wong, D.T. IL-4-dependent regulation of TGF-alpha and TGF-beta1 expression in human eosinophils. J. Immunol. 1998, 160, 6121–6127.
  11. Guermonprez, P.; Valladeau, J.; Zitvogel, L.; Thery, C.; Amigorena, S. Antigen presentation and T cell stimulation by dendritic cells. Annu. Rev. Immunol. 2002, 20, 621–667, doi:10.1146/annurev.immunol.20.100301.064828.
  12. Villadangos, J.A.; Schnorrer, P. Intrinsic and cooperative antigen-presenting functions of dendritic-cell subsets in vivo. Nat. Rev. Immunol. 2007, 7, 543–555, doi:10.1038/nri2103.
  13. Griffioen, A.W.; Damen, C.A.; Blijham, G.H.; Groenewegen, G. Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood 1996, 88, 667–673.
  14. Griffioen, A.W.; Damen, C.A.; Martinotti, S.; Blijham, G.H.; Groenewegen, G. Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: The role of angiogenic factors. Cancer Res. 1996, 56, 1111–1117.
  15. Goel, S.; Duda, D.G.; Xu, L.; Munn, L.L.; Boucher, Y.; Fukumura, D.; Jain, R.K. Normalization of the vasculature for treatment of cancer and other diseases. Physiol. Rev. 2011, 91, 1071–1121, doi:10.1152/physrev.00038.2010.
  16. Motz, G.T.; Santoro, S.P.; Wang, L.P.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos, G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014, 20, 607–615, doi:10.1038/nm.3541.
  17. Hodi, F.S.; Lawrence, D.; Lezcano, C.; Wu, X.; Zhou, J.; Sasada, T.; Zeng, W.; Giobbie-Hurder, A.; Atkins, M.B.; Ibrahim, N.; et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol. Res. 2014, 2, 632–642, doi:10.1158/2326-6066.Cir-14-0053.
  18. Wallin, J.J.; Bendell, J.C.; Funke, R.; Sznol, M.; Korski, K.; Jones, S.; Hernandez, G.; Mier, J.; He, X.; Hodi, F.S.; et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 2016, 7, 12624, doi:10.1038/ncomms12624.
  19. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174, doi:10.1038/nri2506.
  20. Huang, C.; Li, J.; Song, L.; Zhang, D.; Tong, Q.; Ding, M.; Bowman, L.; Aziz, R.; Stoner, G.D. Black raspberry extracts inhibit benzo(a)pyrene diol-epoxide-induced activator protein 1 activation and VEGF transcription by targeting the phosphotidylinositol 3-kinase/Akt pathway. Cancer Res. 2006, 66, 581–587, doi:10.1158/0008-5472.Can-05-1951.
  21. Ko, S.Y.; Guo, H.; Barengo, N.; Naora, H. Inhibition of ovarian cancer growth by a tumor-targeting peptide that binds eukaryotic translation initiation factor 4E. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 4336–4347, doi:10.1158/1078-0432.Ccr-08-2924.
  22. Kusmartsev, S.; Eruslanov, E.; Kubler, H.; Tseng, T.; Sakai, Y.; Su, Z.; Kaliberov, S.; Heiser, A.; Rosser, C.; Dahm, P.; et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: Link to tumor-induced immune suppression in renal cell carcinoma. J. Immunol. 2008, 181, 346–353, doi:10.4049/jimmunol.181.1.346.
  23. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; 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.
  24. Chen, X.; Zeng, Q.; Wu, M.X. Improved efficacy of dendritic cell-based immunotherapy by cutaneous laser illumination. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 2240–2249, doi:10.1158/1078-0432.Ccr-11-2654.
  25. Zou, W.; Chen, L. Inhibitory B7-family molecules in the tumour microenvironment. Nat. Rev. Immunol. 2008, 8, 467–477, doi:10.1038/nri2326.
  26. Tiegs, G.; Lohse, A.W. Immune tolerance: What is unique about the liver. J Autoimmun. 2010, 34, 1–6, doi:10.1016/j.jaut.2009.08.008.
  27. Oyama, T.; Ran, S.; Ishida, T.; Nadaf, S.; Kerr, L.; Carbone, D.P.; Gabrilovich, D.I. Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells. J. Immunol. 1998, 160, 1224–1232.
  28. Kato, Y.; Tabata, K.; Kimura, T.; Yachie-Kinoshita, A.; Ozawa, Y.; Yamada, K.; Ito, J.; Tachino, S.; Hori, Y.; Matsuki, M.; et al. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS ONE 2019, 14, e0212513, doi:10.1371/journal.pone.0212513.
  29. Kudo, M. Combination Cancer Immunotherapy in Hepatocellular Carcinoma. Liver Cancer 2018, 7, 20–27, doi:10.1159/000486487.
  30. Kudo, M. Targeted and immune therapies for hepatocellular carcinoma: Predictions for 2019 and beyond. World J. Gastroenterol. 2019, 25, 789–807, doi:10.3748/wjg.v25.i7.789.
  31. Kudo, M. Combination Cancer Immunotherapy with Molecular Targeted Agents/Anti-CTLA-4 Antibody for Hepatocellular Carcinoma. Liver Cancer 2019, 8, 1–11, doi:10.1159/000496277.
  32. Teng, M.W.; Ngiow, S.F.; Ribas, A.; Smyth, M.J. Classifying Cancers Based on T-cell Infiltration and PD-L1. Cancer Res. 2015, 75, 2139–2145, doi:10.1158/0008-5472.Can-15-0255.
  33. Hsu, C.H.; Lee, M.S.; Lee, K.H.; Numata, K.; Stein, S.; Verret, W.; Hack, S.; Spahn, J.; Liu, B.; Huang, C.; et al. Randomised efficacy and safety results for atezolizumab + bevacizumab in patients with previously untreated, unresectable hepatocellular carcinoma. Ann. Oncol. 2019, 30 (Suppl. 9), ix187.
  34. Yau, T.; Park, J.W.; Finn, R.S.; Cheng, A.L.; Mathurin, P.; Edeline, J.; Kudo, M.; Han, K.H.; Harding, J.J.; Merle, P.; et al. CheckMate 459: A randomized, multi-center phase III study of nivolumab vs sorafenib as first-line treatment in patients with advanced hepatocellular carcinoma. Ann. Oncol. 2019, 30 (Suppl. 5), v874–v875.
  35. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 2018, 19, 940–952, doi:10.1016/s1470-2045(18)30351-6.
  36. Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 193–202, doi:10.1200/jco.19.01307.
  37. Kudo, M. Pembrolizumab for the Treatment of Hepatocellular Carcinoma. Liver Cancer 2019, 8, 143–154, doi:10.1159/000500143.
  38. Kudo, M. Immune checkpoint blockade in hepatocellular carcinoma: 2017 update. Liver Cancer 2017, 6, 1–12, doi:10.1159/000449342.
  39. El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Trojan, J.; Welling, T.H.R.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502, doi:10.1016/s0140-6736(17)31046-2.
  40. Llovet, J.M.; Kudo, M.; Cheng, A.L.; Finn, R.S.; Galle, P.R.; Kaneko, S.; Meyer, T.; Qin, S.; dutcus, C.E.; Chen, E.; et al. First-Line Combination Therapy With Lenvatinib Plus Pembrolizumab for Patients with Advanced Hepatocellular Carcinoma: Phase 3 LEAP-002 Study. In Proceedings of the ILCA 13th Annual Conference, Chicago, IL, USA, 20–22 September 2019.
  41. Kudo, M. Immuno-Oncology Therapy for Hepatocellular Carcinoma: Current Status and Ongoing Trials. Liver Cancer 2019, 8, 221–238, doi:10.1159/000501501.
  42. Llovet, J.M.; Finn, R.S.; Ikeda, M.; Sung, M.W.; Baron, A.D.; Kudo, M.; Okusaka, T.; Kobayashi, M.; Kumada, H.; Kaneko, S.; et al. A phase 1b trial of lenvatinib plus pembrolizumab in unresectable hepatocellular carcinoma: Upated results. Ann. Oncol. 2019, 30 (Suppl. 5), v253–v324.
  43. Kudo, M.; Ikeda, K.; Motomura, K.; Okusaka, T.; Kato, N.; Dutcus, C.E.; Hisai, T.; Suzuki, M.; Ikezawa, H.; Iwata, T.; et al. A Phase 1b Study of Lenvatinib Plus Nivolumab in Patients With Unresectable Hepatocellular Carcinoma. In Proceedings of the ASCO-GI, San Francisco, CA, USA, 23–25 January 2020.
  44. Xu, J.M.; Zhang, Y.; Jia, R.; Wang, Y.; Liu, R.; Zhang, G.; Zhao, C.; Zhang, Y.; Zhou, J.; Wang, Q. Anti-programmed death-1 antibody SHR-1210 (S) combined with apatinib (A) for advanced hepatocellular carcinoma (HCC), gastric cancer (GC) or esophagogastric junction (EGJ) cancer refractory to standard therapy: A phase 1 trial. J. Clin. Oncol. 2018, 36, 4075.
  45. Kudo, M.; Motomura, K.; Wada, Y.; Inaba, Y.; Sakamoto, Y.; Kurosaki, M.; Umeyama, Y.; Kamei, Y.; Yoshimitsu, J.; Fujii, Y.; et al. First-line avelumab + axitinib in patients with advanced hepatocellular carcinoma: Results from a phase 1b trial (VEGF Liver 100). J. Clin. Oncol. 2019, 37 (Suppl. 15), 4072.
  46. Cheng, A.L.; Hsu, C.; Chan, S.L.; Choo, S.P.; Kudo, M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J. Hepatol. 2020, 72, 307–319, doi:10.1016/j.jhep.2019.09.025.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.1K
Revisions: 3 times (View History)
Update Date: 18 Dec 2020
1000/1000