Immunotherapy for Hepatocellular Carcinoma: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Oncology
Contributor:

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death since most patients are diagnosed at advanced stage and the current systemic treatment options using molecular-targeted drugs remain unsatisfactory. However, the recent success of cancer immunotherapies has revolutionized the landscape of cancer therapy. Since HCC is characterized by metachronous multicentric occurrence, immunotherapies that induce systemic and durable responses could be an appealing treatment option. Despite the suppressive milieu of the liver and tumor immunosurveillance escape mechanisms, clinical studies of checkpoint inhibitors in patients with advanced HCC have yielded promising results.

  • hepatocellular carcinoma
  • immunotherapy
  • immune checkpoint inhibitor
  • PD-1
  • CTLA-4
  • combination therapy

1. Introduction

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and poses a serious health problem worldwide [1]. Although various surveillance systems and treatment strategies have been developed and are recommended by guidelines, including surgical resection, radiofrequency ablation (RFA), transarterial chemoembolization (TACE), systemic therapy, and liver transplantation, the prognosis of HCC remains poor due to high levels of high intra- and extra-hepatic recurrence and metastasis [2][3]. Systemic therapies using molecular-targeted agents (MTAs) have been considered efficient and are recommended for patients with advanced-stage HCC [4]; however, the regimens currently available are often unsatisfactory. Therefore, a novel approach that uses a different mechanism to these conventional therapies is required to improve the prognosis of HCC.

The recent development of cancer immunotherapies using immune checkpoint inhibitors (ICIs) targeting cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and anti-programmed cell death protein-1 (PD-1) has dramatically changed the landscape of cancer therapy and was awarded the Nobel Prize in 2018. Several monoclonal antibodies (mAbs) targeting CTLA-4, PD-1, or its ligand programmed cell death-ligand 1 (PD-L1) have now been approved by the FDA for various types of cancers [5]. The liver is a tolerogenic organ [6] that is relevant to successful allograft acceptance after transplantation. Thus, the development of antitumor immunity against HCC might be speculated to be synergistically impeded by this tolerogenic nature of the liver and the immunosuppressive tumor microenvironment of HCC. However, the potential of cancer immunotherapy to induce systemic and durable antitumor responses may make it an ideal therapeutic option for HCC characterized by metachronous multicentric occurrence. Indeed, several ICI therapies targeting PD-1/PD-L1 and CTLA-4 have already demonstrated promising activity against HCC and manageable safety in clinical trials, thus have been approved by the FDA. Combination ICI-based strategies have also shown promising results, while other classes of immunotherapies have begun to emerge and are being tested in preclinical and clinical studies.

2. Tolerogenic Liver Immune Environment and HCC Immune Evasion Mechanisms

The liver is a tolerogenic organ in which a unique immune environment prevents the overactivation of the immune system to antigens derived from food and bacterial products in the portal flow [6]. Immune tolerance in the liver is induced by non-parenchymal cells. Kupffer cells (KCs) are liver-resident macrophages that play a role in pathogen clearance mediated by innate immune activation [7]. However, under physiological conditions, KCs induce tolerance by impairing T cell activation or preferentially expanding regulatory T cells (Tregs) by secreting immunosuppressive factors such as IL-10, TGF- β, and prostaglandin E2 [8][9]. Liver sinusoidal endothelial cells (LSECs), which act as antigen-presenting cells (APCs) and form a cellular barrier between the liver parenchyma and sinusoid [10], are characterized by low co-stimulatory molecule levels, high immune checkpoint molecule levels, and immunosuppressive cytokine production, all of which impede their potential for T cell activation and induce immune tolerance [11][12]. Hepatic dendritic cells (DCs) mediate the induction of T cell tolerance rather than their activation [13], presumably, as they are under the influence of IL-10 and TGF-β secreted by KCs and LSECs [14]. In addition to these non-parenchymal cells, hepatocytes also function as APCs by directly interacting with and presenting antigens to naïve T cells; however, hepatocytes predispose T cells towards tolerance because they lack co-stimulatory molecule expression [15]. Together, these immunosuppressive features of the liver might impede the development of antitumor immunity.

HCC evades host immunosurveillance via multiple mechanisms; for instance, HCC cells silence the expression of tumor antigens or antigen presentation-related molecules so that cytotoxic T cells (CTLs) cannot recognize tumor cells [16][17]. HCC cells also escape immunosurveillance by expressing immune checkpoint molecules such as PD-L1 and producing various immunoinhibitory molecules, including TGF-β, IL-10, indoleamine 2, 3-dioxygenase, arginase, and adenosine [18][19]. Immunosuppressive stromal cells are also a critical component of immune dysregulation. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that inhibit T cell activation via iNOS, ROS, and increased arginase activity, and induce Treg expansion by producing IL-10 and TGF-β [20]. Moreover, the frequency of MDSCs in HCC patients has been reported to correlate with tumor progression [21]. Macrophages are generally categorized as having an M1 or M2 state; M1 macrophages display an antitumor phenotype by producing high and low levels of IL-12 and IL-10, respectively, whereas M2 macrophages exhibit a tumor supportive phenotype with opposite cytokine profiles. During HCC progression, hepatic macrophages are skewed from an M1 phenotype to an M2 phenotype characteristic of tumor-associated macrophages (TAMs), which act as immune suppressor cells and support tumor growth by promoting angiogenesis and tumor invasion [22]. Tregs can also impede immune surveillance against HCC due to their immunosuppressive functions; indeed, they have been shown to densely infiltrate the tumor site in patients with HCC, with the number of intratumoral Tregs acting as an independent prognostic factor of overall survival (OS) and disease-free survival (DFS) in those patients [23].

Even under these immunosuppressive conditions, several studies have shown that antitumor immunity exists in patients with HCC. For instance, T cells specific for four different tumor-associated antigens (TAAs) were detected in both the tumor tissue and peripheral blood of patients with HCC, with the breadth of T cell response correlating with survival [24]. Another study found that the intratumoral density of activated CTLs in patients with HCC after resection was associated with OS and that the intratumoral balance between CTLs and Tregs was associated with OS and DFS [25]. These observations suggest that the immunogenic potential of HCC could be controlled by optimized immunotherapy.

3. PD-1/PD-L1 and CTLA-4-Blockade Therapies

3.1. Basic Immunobiology of PD-1 and CTLA-4

Immune checkpoint molecules—among which, PD-1 and CTLA-4 are the best studied—play essential roles in preventing T cell overactivation by interacting with APCs and other cell types. PD-1 is a member of the CD28 family that is expressed on activated T cells, B cells, and myeloid cells and negatively regulates the immune system. The engagement of PD-1 by its ligand PD-L1 leads to the transmission of suppressive signals into T cells and the induction of peripheral tolerance [26]. In the liver, PD-L1 is constitutively expressed on liver non-parenchymal cells such as LSECs and KCs [27]; however, PD-L1 is aberrantly expressed in various tumors, including HCC tumor cells, allowing them to escape from host immune surveillance. Indeed, it has been demonstrated that tumor PD-L1 expression is associated with HCC prognosis after curative surgical treatment, suggesting that the PD-1/PD-L1 pathway is an immune escape mechanism in HCC [19]. Another member of the CD28 family, CTLA-4, is induced on naïve T cells by antigen activation but is constitutively expressed on Tregs [28]. CTLA-4 binds to CD80 and CD86 more tightly than CD28, which provides a positive signal required for T cell activation; therefore, CTLA-4 induces peripheral tolerance by counteracting CD28-mediated costimulatory signals [28]. Importantly, the expression of CTLA-4 on Tregs depletes APCs of CD80 and CD86, leaving them with a reduced ability to prime naïve T cells . The intensive study of PD-1- and CTLA-4-mediated immunosuppression culminated in the dramatic success of cancer immunotherapies [29] and many clinical trials of ICI mono- and combination therapies targeting PD-1/PD-L1 and CTLA-4 in HCC have now been conducted.

3.2. ICI Monotherapies Directed Against PD-1 and CTLA-4

Many clinical trials have been conducted for ICI monotherapies in HCC (Table 1) and the first to be approved by the FDA was the anti-PD-1 mAb nivolumab. A phase I/II trial of nivolumab in patients with advanced HCC (CheckMate-040) showed promising results. In the dose-expansion phase in which a total of 214 patients in 4 cohorts were enrolled, the objective response rate (ORR) was 20%, the disease control rate (DCR) was 64%, and progression free survival (PFS) was 4.1 months [30]. Since adverse events (AEs) were fairly mild [30], nivolumab was approved by the FDA in September 2017 as a second-line treatment for unresectable HCC after sorafenib failure, based on subgroup analysis in CheckMate-040. However, a phase III trial (CheckMate-459) evaluating nivolumab versus sorafenib as first-line treatments in patients with unresectable HCC revealed that the trial did not achieve statistical significance for its primary OS endpoint as per the prespecified analysis [31]. The CheckMate-9DX trial is currently evaluating adjuvant nivolumab versus a placebo in HCC patients at high risk of recurrence after curative hepatic resection or ablation.

Pembrolizumab is another anti-PD-1 mAb that was granted accelerated approval by the FDA in May 2017 for patients with unresectable or metastatic microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) solid tumors that continued to progress after conventional treatment, based on the data from five clinical trials [32]. A phase II trial (KEYNOTE-224) revealed the potential of pembrolizumab against HCC after sorafenib failure, with an ORR of 17% with one complete response (CR), a DCR of 61%, and AEs (>grade 3) reported in 26% of patients [33]. Based on this data, pembrolizumab was granted accelerated approval by the FDA in November 2018 as a second-line treatment after sorafenib. A phase III trial (KEYNOTE-240) comparing pembrolizumab to a placebo as a second-line treatment demonstrated that pembrolizumab was associated with a longer median OS and PFS; however, these findings were not deemed statistically significant according to the prespecified statistical plan [34]. Two further phase III trials are currently ongoing: KEYNOTE-394 is evaluating pembrolizumab versus a placebo and best supportive care in Asian patients with systemically treated advanced HCC, while KEYNOTE-937 is evaluating pembrolizumab versus a placebo as an adjuvant therapy in HCC patients after curative treatment.

In addition, the anti-PD-L1 mAb Durvalumab was tested in a phase I/II trial (NCT01693562) of patients with advanced HCC who had been previously treated with sorafenib, achieving an OS rate of 10.3% in 39 patients [35]. The investigational IgG4 anti-PD-1 Ab, tislelizumab (BGB-A317), was designed to bind minimally to FcγR on macrophages in order to abrogate antibody-dependent phagocytosis, which is a potential mechanism of anti-PD-1 therapy resistance. Tislelizumab has demonstrated a good preliminary safety profile and antitumor activity in a phase I trial and a phase III trial (RATIONALE 301) of tislelizumab versus sorafenib as a first-line treatment in patients with unresectable HCC is currently underway [36].

The anti-CTLA-4 mAb, tremelimumab, has been tested in a small phase II pilot trial (NCT01008358) of HCV-infected patients with advanced HCC, demonstrating partial response (PR) and stable disease (SD) rates of 17.6 and 58.8%, respectively. Moreover, the treatment was well tolerated, and no patients needed steroids due to severe immune-related AEs (irAEs) [37].

Table 1. Summary of clinical trials of ICI monotherapy for HCC.

Trial identifier

Target

Drugs

Phase

N

Patient Group

ORR

DCR

PFS
(Median,mo)

OS
(Median,mo)

NCT01658878
(CheckMate040) [30]

PD-1

Nivolumab

I/II

214*

Naive/Pre-treated

20.0%

64.0%

4

NR

NCT02576509
(CheckMate459) [31]

PD-1

Nivolumab vs. Sorafenib

III

743

Naïve

15% vs. 7%

N/A

3.7 vs. 3.8

16.4 vs. 14.7

NCT03383458
(CheckMate 9DX)

PD-1

Nivolumab vs. Placebo

III

530

Adjuvant

N/A

N/A

N/A

N/A

NCT02702414
(KEYNOTE-224) [33]

PD-1

Pembrolizumab

II

104

Pre-treated

17.0%

61.0%

4.9

12.9

NCT02702401
(KEYNOTE-240) [34]

PD-1

Pembrolizumab vs. Placebo

III

413

Pre-treated

18.3% vs. 4.4%

62.2% vs. 53.3%

3.0 vs. 2.8

13.9 vs. 10.6

NCT03062358
(KEYNOTE-394)

PD-1

Pembrolizumab vs. Placebo

III

N/A

Pre-treated

N/A

N/A

N/A

N/A

NCT03867084
(KEYNOTE-937)

PD-1

Pembrolizumab vs. Placebo

III

N/A

Adjuvant

N/A

N/A

N/A

N/A

NCT03412773
(RATIONALE-301) [36]

PD-1

Tislelizumab vs. Sorafenib

III

N/A

Naïve

N/A

N/A

N/A

N/A

NCT01693562
[35]

PD-L1

Durvalumab

I/II

39

Pre-treated

10.3%

33.3%

NA

13.2

NCT01008358
[37]

CTLA-4

Tremelimumab

II

20

Pre-treated

17.6%

76.4%

6.48

8.2

N, number of patients; N/A; not available; NR, not reached; * dose-expansion phase.

This entry is adapted from the peer-reviewed paper 10.3390/cancers12040775

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424, doi:10.3322/caac.21492.
  2. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236, doi:10.1016/j.jhep.2018.03.019.
  3. Zheng, Z.; Liang, W.; Wang, D.; Schroder, P.M.; Ju, W.; Wu, L.; Shang, Y.; Guo, Z.; He, X. Adjuvant chemotherapy for patients with primary hepatocellular carcinoma: A meta-analysis. Int. J. Cancer 2015, 136, E751–E759, doi:10.1002/ijc.29203.
  4. Eso, Y.; Marusawa, H. Novel approaches for molecular targeted therapy against hepatocellular carcinoma. Hepatol. Res. 2018, 48, 597–607, doi:10.1111/hepr.13181.
  5. Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39, doi:10.1016/j.intimp.2018.06.001.
  6. Crispe, I.N. Liver antigen-presenting cells. J. Hepatol. 2011, 54, 357–365, doi:10.1016/j.jhep.2010.10.005.
  7. Knolle, P.A.; Gerken, G. Local control of the immune response in the liver. Immunol. Rev. 2000, 174, 21–34, doi:10.1034/j.1600-0528.2002.017408.x.
  8. Krenkel, O.; Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 2017, 17, 306–321, doi:10.1038/nri.2017.11.
  9. Breous, E.; Somanathan, S.; Vandenberghe, L.H.; Wilson, J.M. Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 2009, 50, 612–621, doi:10.1002/hep.23043.
  10. Limmer, A.; Knolle, P.A. Liver sinusoidal endothelial cells: A new type of organ-resident antigen-presenting cell. Arch. Immunol. Ther. Exp. 2001, 49, S7–S11.
  11. Diehl, L.; Schurich, A.; Grochtmann, R.; Hegenbarth, S.; Chen, L.; Knolle, P.A. Tolerogenic maturation of liver sinusoidal endothelial cells promotes B7-homolog 1-dependent CD8+ T cell tolerance. Hepatology 2008, 47, 296–305, doi:10.1002/hep.21965.
  12. von Oppen, N.; Schurich, A.; Hegenbarth, S.; Stabenow, D.; Tolba, R.; Weiskirchen, R.; Geerts, A.; Kolanus, W.; Knolle, P.; Diehl, L. Systemic antigen cross-presented by liver sinusoidal endothelial cells induces liver-specific CD8 T-cell retention and tolerization. Hepatology 2009, 49, 1664–1672, doi:10.1002/hep.22795.
  13. Bamboat, Z.M.; Stableford, J.A.; Plitas, G.; Burt, B.M.; Nguyen, H.M.; Welles, A.P.; Gonen, M.; Young, J.W.; DeMatteo, R.P. Human liver dendritic cells promote T cell hyporesponsiveness. J. Immunol. 2009, 182, 1901–1911, doi:10.4049/jimmunol.0803404.
  14. Lau, A.H.; Thomson, A.W. Dendritic cells and immune regulation in the liver. Gut 2003, 52, 307–314, doi:10.1136/gut.52.2.307.
  15. Holz, L.E.; Benseler, V.; Bowen, D.G.; Bouillet, P.; Strasser, A.; O’Reilly, L.; d’Avigdor, W.M.; Bishop, A.G.; McCaughan, G.W.; Bertolino, P. Intrahepatic murine CD8 T-cell activation associates with a distinct phenotype leading to Bim-dependent death. Gastroenterology 2008, 135, 989–997, doi:10.1053/j.gastro.2008.05.078.
  16. Matsui, M.; Machida, S.; Itani-Yohda, T.; Akatsuka, T. Downregulation of the proteasome subunits, transporter, and antigen presentation in hepatocellular carcinoma, and their restoration by interferon-gamma. J. Gastroenterol. Hepatol. 2002, 17, 897–907, doi:10.1046/j.1440-1746.2002.02837.x.
  17. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998, doi:10.1038/ni1102-991.
  18. Prieto, J.; Melero, I.; Sangro, B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 681–700, doi:10.1038/nrgastro.2015.173.
  19. Gao, Q.; Wang, X.Y.; Qiu, S.J.; Yamato, I.; Sho, M.; Nakajima, Y.; Zhou, J.; Li, B.Z.; Shi, Y.H.; Xiao, Y.S.; et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin. Cancer Res. 2009, 15, 971–979, doi:10.1158/1078-0432.ccr-08-1608.
  20. Meirow, Y.; Kanterman, J.; Baniyash, M. Paving the Road to Tumor Development and Spreading: Myeloid-Derived Suppressor Cells are Ruling the Fate. Front. Immunol. 2015, 6, 523, doi:10.3389/fimmu.2015.00523.
  21. Arihara, F.; Mizukoshi, E.; Kitahara, M.; Takata, Y.; Arai, K.; Yamashita, T.; Nakamoto, Y.; Kaneko, S. Increase in CD14+HLA-DR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol. Immunother. 2013, 62, 1421–1430, doi:10.1007/s00262-013-1447-1.
  22. Degroote, H.; Van Dierendonck, A.; Geerts, A.; Van Vlierberghe, H.; Devisscher, L. Preclinical and Clinical Therapeutic Strategies Affecting Tumor-Associated Macrophages in Hepatocellular Carcinoma. J. Immunol. Res. 2018, 2018, 7819520, doi:10.1155/2018/7819520.
  23. Chen, K.J.; Lin, S.Z.; Zhou, L.; Xie, H.Y.; Zhou, W.H.; Taki-Eldin, A.; Zheng, S.S. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS ONE 2011, 6, e24671, doi:10.1371/journal.pone.0024671.
  24. Flecken, T.; Schmidt, N.; Hild, S.; Gostick, E.; Drognitz, O.; Zeiser, R.; Schemmer, P.; Bruns, H.; Eiermann, T.; Price, D.A.; et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology 2014, 59, 1415–1426, doi:10.1002/hep.26731.
  25. Gao, Q.; Qiu, S.J.; Fan, J.; Zhou, J.; Wang, X.Y.; Xiao, Y.S.; Xu, Y.; Li, Y.W.; Tang, Z.Y. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J. Clin. Oncol. 2007, 25, 2586–2593, doi:10.1200/jco.2006.09.4565.
  26. Okazaki, T.; Honjo, T. PD-1 and PD-1 ligands: From discovery to clinical application. Int. Immunol. 2007, 19, 813–824, doi:10.1093/intimm/dxm057.
  27. Iwai, Y.; Terawaki, S.; Ikegawa, M.; Okazaki, T.; Honjo, T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J. Exp. Med. 2003, 198, 39–50, doi:10.1084/jem.20022235.
  28. Fritz, J.M.; Lenardo, M.J. Development of immune checkpoint therapy for cancer. J. Exp. Med. 2019, 216, 1244–1254, doi:10.1084/jem.20182395.
  29. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61, doi:10.1126/science.aaa8172.
  30. 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.; 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.
  31. 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. LBA38_PR-CheckMate 459: A randomized, multi-center phase III study of nivolumab (NIVO) vs. sorafenib (SOR) as first-line (1L) treatment in patients (pts) with advanced hepatocellular carcinoma (aHCC). Ann. Oncol. 2019, 30, v874–v875, doi:10.1093/annonc/mdz394.029.
  32. Prasad, V.; Kaestner, V.; Mailankody, S. Cancer Drugs Approved Based on Biomarkers and Not Tumor Type-FDA Approval of Pembrolizumab for Mismatch Repair-Deficient Solid Cancers. JAMA Oncol. 2018, 4, 157–158, doi:10.1001/jamaoncol.2017.4182.
  33. 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.
  34. 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. 2020, 38, 193–202, doi:10.1200/jco.19.01307.
  35. Wainberg, Z.A.; Segal, N.H.; Jaeger, D.; Lee, K.-H.; Marshall, J.; Antonia, S.J.; Butler, M.; Sanborn, R.E.; Nemunaitis, J.J.; Carlson, C.A.; et al. Safety and clinical activity of durvalumab monotherapy in patients with hepatocellular carcinoma (HCC). J. Clin. Oncol. 2017, 35, 4071, doi:10.1200/JCO.2017.35.15_suppl.4071.
  36. Qin, S.; Finn, R.S.; Kudo, M.; Meyer, T.; Vogel, A.; Ducreux, M.; Macarulla, T.M.; Tomasello, G.; Boisserie, F.; Hou, J.; et al. RATIONALE 301 study: Tislelizumab versus sorafenib as first-line treatment for unresectable hepatocellular carcinoma. Future Oncol. 2019, 15, 1811–1822, doi:10.2217/fon-2019-0097.
  37. Sangro, B.; Gomez-Martin, C.; de la Mata, M.; Iñarrairaegui, M.; Garralda, E.; Barrera, P.; Riezu-Boj, J.I.; Larrea, E.; Alfaro, C.; Sarobe, P.; et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J. Hepatol. 2013, 59, 81–88, doi:10.1016/j.jhep.2013.02.022.
More
This entry is offline, you can click here to edit this entry!
Video Production Service