Skip to main content
Gene Expression logoLink to Gene Expression
. 2020 Jun 12;20(1):53–65. doi: 10.3727/105221620X15880179864121

Immune Checkpoint Inhibitors in Hepatocellular Cancer: Current Understanding on Mechanisms of Resistance and Biomarkers of Response to Treatment

Amblessed E Onuma *, Hongji Zhang *,, Hai Huang *, Terence M Williams , Anne Noonan §, Allan Tsung *
PMCID: PMC7284108  PMID: 32340652

Abstract

Hepatocellular carcinoma (HCC) is the most common primary liver malignancy worldwide and a leading cause of death worldwide. Its incidence continues to increase in the US due to hepatitis C infection and nonalcoholic steatohepatitis. Liver transplantation and resection remain the best therapeutic options for cure, but these are limited by the shortage of available organs for transplantation, diagnosis at advanced stage, and underlying chronic liver disease found in most patients with HCC. Immune checkpoint inhibitors (ICIs) have been shown to be an evolving novel treatment option in certain advanced solid tumors and have been recently approved for inoperable, advanced, and metastatic HCC. Unfortunately, a large cohort of patients with HCC fail to respond to immunotherapy. In this review, we discuss the ICIs currently approved for HCC treatment and their various mechanisms of action. We will highlight current understanding of mechanism of resistance and limitations to ICIs. Finally, we will describe emerging biomarkers of response to ICIs and address future direction on overcoming resistance to immune checkpoint therapy.

Key words: Checkpoint inhibitor, Immunotherapy, Hepatocellular carcinoma (HCC), Biomarker, Resistance

INTRODUCTION

Hepatocellular carcinoma (HCC) is a primary tumor of the liver that most often occurs in the setting of chronic liver disease. Globally, HCC accounts for the third and seventh most common malignancy in men and women, respectively, and is the fourth leading cause of malignancy-related death1,2. In the US, the death rate from HCC increased by 43% between 2000 and 20163. The incidence of HCC remains on the rise and is estimated to reach 27,000 by the end of 20204. Although hepatitis B virus (HBV) is the most common cause of HCC worldwide, the vast majority of HCC in the US is due to hepatitis C virus (HCV), while the incidence of HCC secondary to nonalcoholic fatty liver disease (NAFLD) is on the rise5–9. This steep increase in incidence and mortality in HCC has led to investigation of better treatment strategies to combat this deadly disease.

Treatment of HCC is challenging due to the complex pathophysiology of the disease. Curative intent treatment options include orthotopic liver transplantation and surgical resection for early stage disease10. Liver transplantation is an important therapeutic option; however, there are limitations due to the shortage of organs for transplantation. Surgical resection is another potentially curative treatment modality for early, localized disease. The majority of patients with HCC are not eligible for resection due to advanced disease at presentation and underlying liver dysfunction. Two landmark trials, Sorafenib HCC Assessment Randomized Protocol (SHARP) and Asia-Pacific (AP), investigated the efficacy of the multikinase inhibitor sorafenib, which led to its approval in 2007 as monotherapy in patients with Childs A cirrhosis and unresectable or metastatic HCC11–13. It is known that the overall survival (OS) benefit from sorafenib is much higher in patients with HCC related to HCV than in those with other underlying etiologies for HCC14. Despite its poor side effect profile and improvement in OS of less than 3 months compared to placebo, sorafenib remains one of the frontline systemic therapies. Lenvatinib was approved as an alternative first-line therapy as it was confirmed to be noninferior to sorafenib in the REFLECT study15. Multitarget tyrosine inhibitors (regorafenib and cabozantinib) and vascular endothelial growth factor (VEGF) receptor inhibitors (ramucirumab) have all been approved by the Food and Drug Administration (FDA) as single-agent second-line systemic therapy for patients who have failed sorafenib16–19. Despite these options, better and effective alternatives are needed to improve patient survival.

Recently, immune checkpoint inhibitors (ICIs) have emerged as alternatives for patients with adequate performance status who progress on first-line therapy. On September 22, 2017, the FDA approved nivolumab as an adjunct treatment for patients who have failed treatment with sorafenib. This was followed by approval of pembrolizumab in November 2018. These two immunotherapies, which fall under the broad category of programmed cell death protein-1 (PD-1) inhibitors, have shown some promise in the treatment of advanced HCC. Drug combination treatments have found some success in HCC as combination ipilimumab [cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor] and nivolumab was recently approved by the FDA in late 2019. Results of the IMbrave150 trial presented at the European Society of Medical Oncology conference in November 2019 show that atezolizumab in combination with bevacizumab was superior in prolonged OS and progression-free survival (PFS) to current standard of care, sorafenib20. While awaiting the final publication of the IMbrave150 study and regulatory approval, this result is exciting as this combination may soon be the frontline therapy for advanced HCC. Despite the success of ICIs, there remains a large cohort of HCC patients that do not respond to ICIs, and the challenge remains to find cellular and molecular cues that could predict which patients would benefit from these therapies21. In this review, we will discuss ICIs currently approved for HCC treatment and new options that are currently in development. We will highlight current understanding of mechanism of resistance and limitations to ICIs. Finally, we will describe emerging biomarkers of response to ICIs and address future directions on overcoming resistance to immune checkpoint therapy.

ICIs: DISCOVERY AND EVOLUTION

Immune checkpoints are membrane-bound molecules expressed in different cells types such as natural killer (NK) cells, dendritic cells (DC), tumor-associated macrophages, monocytes, and myeloid-derived suppressor cells (MDSC) including B and T cells22. These immune checkpoint proteins apply a physiologic break that prevent activation of these cells, limiting widespread off-target tissue damage. The intensity of immune response and activation of cytotoxic immune response depends on the balance between costimulatory signals and immune checkpoints23,24. It has been found that immune checkpoint proteins can be dysregulated by tumors as an important mechanism of immune resistance25. The discovery of cancer therapy by inhibition of this negative immune regulation led to the 2018 Nobel Prize in Physiology or Medicine to be jointly awarded to Drs. James P. Allison and Tasuku Honjo26. In turn, T cells have been the major focus of immune checkpoint therapy because of three major reasons: their capacity for selective recognition of peptides derived from proteins in all cellular compartments; their ability to directly kill antigen-expressing cells through cytotoxic CD8+ T cells; and, finally, their capacity to mount diverse immune responses through CD4+ helper T cells, which link adaptative and innate immunity25. The immune checkpoints most commonly studied in human cancers are CTLA-4, PD-1/programmed death-ligand-1 (PD-L1), lymphocyte activation gene 3 (LAG-3), T-cell membrane protein 3 (TIM-3), and B- and T-lymphocyte attenuator (BTLA). These molecules and their functions have been well described in the literature25,27–30. In this review, we will focus on the two major classes (PD-1/PD-L1 and CTLA-4) that have been studied in HCC.

ICIs IN HCC

The success of ICIs in a number of malignancies have opened the prospects of ICIs as a potential immunotherapeutic strategy for treating HCC29. The liver possesses a unique immune biology that allows for the use of checkpoint therapy. First, HCC arises in the background of chronically inflamed livers. Patients with chronic inflamed liver disease have been shown to overexpress PD-1 in the intrahepatic lymphocytes, while the ligands, PD-L1 and PD-L2, were found to be overexpressed in Kupffer cells, liver sinusoidal endothelial cells, and leukocytes31,32. Additionally, the liver has an abundance of Kupffer cells, DC, and naive T cells that are prone to dysregulation in cytokine secretion, antigen and immune checkpoint expression in the local immune microenvironment29,33,34. This upregulation of checkpoint proteins in the liver makes ICIs a plausible option for the treatment of HCC as checkpoint inhibitors have been developed to block these inhibitory molecules expressed on the surface of these cells, thereby generating antitumor activity.

There are two classes of ICIs that are currently being used clinically or are part of active investigation in the treatment of advanced HCC. These immunotherapies belong to the CTLA-4 and PD-1/PD-L1 inhibition pathway. Monoclonal antibodies against CTLA-4 and PD-1/PD-L1 have been used successfully in the treatment of advanced melanoma, renal cell carcinoma, non-small cell lung cancer, and others including microsatellite instability-high (MSI-H) colorectal cancers35–40. Checkpoint inhibitors are indicated for use as second-line treatment of HCC in patients who have failed first-line sorafenib. There are ongoing trials looking at ICIs either as monotherapy or combination therapy for first-line treatment of advanced HCC.

Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA-4)

CTLA-4 is an intracellular protein in resting T cells. When the T-cell receptor is activated by CD28, CTLA-4 is translocated to the cell surface. Once CTLA-4 is expressed on the surface of T cells, it binds to CD80 and CD86, preventing the binding of CD28 to these critical costimulatory molecules, mediating inhibitory signals to the T cell resulting in arrest of both proliferation and activation36,41,42. CTLA-4 signaling also promotes tumor development by inhibiting the binding of antigen presentation by antigen-presenting cells42. Furthermore, CTLA-4 signaling may also stimulate the expression of immune regulatory cytokine such as transforming growth factor-β (TGF-β)43. CTLA-4 is thought to affect Treg activation and differentiation as this receptor is constitutively expressed in Tregs. This is further supported by the fact that blockade of CTLA-4 or Treg-specific knockout inhibits their ability to regulate both antitumor activity and autoimmunity44,45.

CTLA-4 inhibitors (ipilimumab and tremelimumab) were introduced into clinical trials in the 2000s after preclinical models showed that inhibition of this molecular brake with an antibody could allow for T-cell activation and proliferation41. Clinical activity of anti-CTLA-4 therapy is more pronounced in advance metastatic melanoma with greater than 15% objective response rate lasting more than 10 years even after therapy was discontinued36,46,47. Tremelimumab was investigated as monotherapy for patients with HCC secondary to HCV-induced cirrhosis in a phase II clinical trial (NCT01008358)48. Partial response rate was observed in 17.6%, while time to progression was 6.48 months with good safety profile noted in the trial48. A randomized, multicenter phase III study (NCT04039607) looking at nivolumab in combination with ipilimumab compared to sorafenib or lenvatinib as first-line treatment is currently ongoing and is estimated to be completed by September 2023. Although this class of immunotherapy has not been approved for use as a single-agent therapy in HCC yet, combination of ipilimumab and nivolumab was granted priority review in November 2019 by the FDA with final approval for use granted on March 11, 202049. This approval was based on positive results from CheckMate 040 (NCT01658878), which showed objective response rate of 31% in the combination group compared to 14% in the nivolumab monotherapy group49,50. There are ongoing trials looking at combination therapy with PD-1/PD-L1 blockade.

PD-1/PD-L1

PD-1 is an immunosuppressive receptor that is expressed on activated T cells, B cells, NK cells, Tregs, MDSC, and DC51. It was initially thought to be a receptor that induced cell death of activated T cell, hence the name programmed cell death protein52. However, it was later discovered that it is an immune checkpoint with its inhibitory function mediated by tyrosine phosphatase SHP-253. PD-1 has two ligands: the first is PD-L1 (also known as CD274 or B7-H1), which is generally expressed by multiple somatic cells when exposed to proinflammatory cytokines53. This ligand is mainly responsible for the suppression of T-cell migration, proliferation, and secretion of cytotoxic mediators54,55. The second ligand is PD-L2 (CD273 or B7-DC), which is infrequently expressed in antigen-presenting cells53. T-cell function depends on the level of PD-1 activity56. Cancer cells have evolved to hijack PD-1/PD-L1 signaling by constitutively expressing PD-L1 or PD-L2 to activate PD-1 in tumor-infiltrating lymphocytes (TILs) and evade immune surveillance57,58. High PD-L1 expression on tumor cells has been associated with recurrence in HCC, in addition to tumor aggressiveness, and poor prognosis in patients who have never received immunotherapy59–63.

Nivolumab and pembrolizumab are the only two PD-1 inhibitors currently approved for use in HCC treatment, although there are other PD-1 inhibitors (tislelizumab and camrelizumab) currently in clinical trials. Nivolumab was the first PD-1 inhibitor to be approved based on results of the CheckMate 040 trial (NCT10658878) that concluded on August 201664. This trial was a phase I/II, dose escalation, and expansion trial of nivolumab in adults with histologically confirmed HCC with or without HCV or HBV. The authors of this study saw an objective response rate of 20% in the dose expansion phase and 15% in the dose escalation phase64. This trial was followed up by the CheckMate 459 trial (NCT02576509), which was a phase III multicenter study comparing the efficacy of nivolumab versus sorafenib as first-line treatment in patients with advanced HCC. The authors of this study reported that although the primary endpoint of OS was not statistically significant (median OS of 16.4 months in nivolumab versus 14.7 months in the sorafenib group), nivolumab demonstrated clinical improvements in OS, objective response rate, and complete response rate as first-line treatment of advance HCC65. Pembrolizumab was approved by the FDA in November 2018 after the Keynote-224 trial, a nonrandomized phase II trial of pembrolizumab in patients with pathologically confirmed HCC who were previously treated with sorafenib and were either intolerant or showed signs of progression66. The trial reported overall objective response rate of 17%, stable disease in 44% of the cohort, while 33% had progressive disease66. This trial was followed by Keynote-240, a phase III trial of pembrolizumab as second-line therapy versus placebo for patients previously treated with sorafenib67. After a median follow-up of 13.8 months, OS did not meet statistical significance (13.9 months in the pembrolizumab group compared to 10.6 months in the placebo group), while the objective response rate was 17%, similar to the results from the phase II Keynote-224 trial. Various PD-L1 inhibitors (avelumab, atezolizumab, and durvalumab) are currently in clinical trials as of January 2020 as either monotherapy or combination therapy with other ICIs.

Immune-related adverse events are the side effects of the unbalanced immune system stemming from immunotherapy use, which may affect the intestine, endocrine glands, liver, and various tissues. Grade 3/4 treatment-related adverse events were reported in 25% of patients in the CheckMate 040 trial (pemphigoid, adrenal insufficiency, and liver disorder) and 24% of patients in the Keynote-224 trial (hypertransaminasemia and fatigue). One treatment-related death was reported in the Keynote-224 trial, which was associated with ulcerative esophagitis66. Both medications are well tolerated with few side effects and no dose-limiting toxicities similar to patients with melanoma and NSCLC treated with ICIs68.

MECHANISMS OF RESISTANCE AND OVERCOMING RESISTANCE TO ICIs

While the mechanisms of resistance to ICIs in other cancers (melanoma, non-small cell lung cancer) are well described, there are limited data on mechanism of resistance for ICIs in HCC, probably due to its recent approval for use in HCC treatment69–71. These mechanisms of resistance can be categorized into tumor intrinsic and extrinsic factors. Tumor intrinsic mechanisms arise from changes in the tumor such as expression of PD-L1, downregulation of major histocompatibility complex (MHC) class I molecules and changes in oncogenic signaling pathways such as activation of β-catenin signaling25,72,73. While more studies are needed, β-catenin activation due to mutations in CTNNB1 gene may be influencing the immune microenvironment in HCC, at least in part through modulation of nuclear factor κB (NF-κB) signaling pathway. A direct complex of β-catenin and NF-κB subunit p65 has been shown in the liver and in HCC74. Increased β-catenin levels due to mutations [also observed as an increase in its target glutamine synthetase (GS)] was shown to enhance its association with NF-κB, which in turn decreased NF-κB activity in HCC cells. Further, GS-positive HCCs showed less p65 immunostaining and vice versa, suggesting that CTNNB1-mutated HCC may have decreased immune cell infiltration, at least in part due to reduced NF-κB activity. Extrinsic factors arise from changes in the tumor microenvironment (TME) such as contributions from Tregs, MDSC, upregulation of coinhibitory molecules on lymphocytes, and contribution from the gut microbiome75. Table 1 summarizes known mechanism of resistance to ICIs. We assume that the mechanisms of resistance will be similar to those found in other tumors, but as more patients with HCC are treated with ICIs, we may uncover newer mechanisms of resistance.

Table 1.

Summary of Known Resistance Mechanisms to Checkpoint Inhibitors

Categories of Resistance Mechanism of Resistance
Tumor intrinsic factors Downregulation of antigen processing and presentation: HLA deletion144,145, β2 microglobulin146
Downregulation of cytokines and signaling pathways: loss of JAK1/2 function69,147, deletion of interferon IFNGR1/2, IRF1 148
β-Catenin activation (due to mutations in CTNNB1 gene)73
Tumor extrinsic factors TILs exclusion by PTEN deletion and VEGF upregulation149
Expression of alternative coinhibitory checkpoint receptors like TIM-3, LAG-3, TIGIT, VISTA, and BTLA69,126
Decreased TILs to Treg ratio150–152
Downregulation of dendritic cell recruitment through β-catenin signaling110
Increased immunosuppressive cells such as MDSCs, Tregs151,153,154
Epithelial-to-mesenchymal transition155
Microbiome75, 143

BIOMARKERS FOR RESPONSE TO IMMUNE CHECKPOINT THERAPY STUDIED IN HCC

Based on published results of the clinical trials of ICIs in patients with HCC, we know that there remains a large proportion of patients who do not benefit from this class of treatment, and the challenge remains to find cellular and molecular cues that could help predict which patients would benefit from these therapies. Prognostic biomarkers of response to ICIs in various cancers have been extensively reviewed76–79. However, there are few studies on predictive biomarkers of response to ICI treatment in HCC owing to that fact that ICI therapy is still in its infancy in HCC. We will summarize emerging major biomarkers of response to treatment and highlight their application in HCC.

PD-L1 Expression

This is one of the earliest and the most commonly used predictive biomarker in immunotherapy. High PD-L1 expression has been associated with improved objective response rate and survival in patients with melanoma, non-small cell lung cancer, and head and neck squamous cell lung cancer80–82. In fact, PD-L1 testing by immunohistochemistry has been approved by the FDA as a companion diagnostic when considering the use of anti-PD1 therapy in non-small cell lung cancer83,84. PD-L1 has been previously investigated in HCC prior to initiation of immune checkpoint therapy. In HCC tissues, PD-L1 is found to be expressed by both the tumor cells and macrophages59,85. Previous studies have shown that PD-L1 expression is generally low in the tumor (roughly 10% of tumor cells), and there is heterogeneity in PD-L1 immunohistochemical detection in HCC84,86. A meta-analysis study by Gu et al. surmised that higher PD-L1 levels predict poor differentiation, higher alpha-fetoprotein, vascular invasion, and poorer survival in HCC87,88. Finkelmeier et al. studied circulating levels of PD-L1 and concluded that a high soluble PD-L1 level may be a prognostic indicator for poor prognosis89.

All this background evidence of PD-L1 as a prognostic biomarker was promising. However, when PD-L1 expression was evaluated in the CheckMate 040 and Keynote-224 trials, it failed to have an impact on the objective response rates to anti-PD-1 therapy64,66,90. This was further confirmed by a study by Feun et al., where response to anti-PD-1 had no correlation with PD-L1 tumor staining in advanced HCC91. However, it is worthwhile to understand why the use of PD-L1 as a biomarker failed to predict response to treatment in these clinical trials. One reason for this failure was because different assays were used at the different institutions for the detection of PD-L1 as well as varying cutoffs in assessing positive staining, thus making it hard to interpret the results83,84,92. In the Keynote-224 trial, two different methods were used to investigate PD-L1 expression as a potential biomarker. One method was the combined positive score (CPS), which was calculated by dividing the number of PD-L1-positive cells (tumor cells, lymphocytes, and macrophages) by the total number of viable tumor cells and multiplying by 100. The other method, tumor proportion score (TPS), was calculated by dividing the number of PD-L1-expressing tumor cells by the total number of viable tumor cells and multiplying by 10066. The authors of this trial found that CPS was associated with response to anti-PD-1, while TPS was not significant, suggesting that inclusion of immune cell scoring could improve predictive value of a PD-L1 immunohistochemistry assay66. Additionally, we know that patients with PD-L1-negative tumors also respond to PD-1/PD-L1 blockade93. Furthermore, PD-L1 expression is inducible and can change during the course of HCC and during treatment94. It has been shown that interferon-γ (IFN-γ) induces upregulation of PD-L1 expression in melanoma and ovarian cancer and is a biomarker of response to checkpoint therapy in numerous cancers95–98. However, this needs to be investigated in HCC.

Tumor Mutational Burden (TMB)

The landscape of biomarker discovery for checkpoint therapy is constantly evolving, and there is more recognition that interactions between the microenvironment, genetic, and systemic factors play a role in determining response to these therapies. It is reasonable to assume that, in the near future, immunotherapy selections will be based on some combination of TMB, cell surface marker, or blood markers99,100. TMB is defined as total number of unique mutations in the tumor exome101. Known causes of TMB include microsatellite instability (MSI), or DNA mismatch repair gene deficiency and somatic mutations that arise in DNA polymerase102,103. MSI is an FDA-approved indication for use of PD-1 checkpoint inhibitor in solid tumors like colorectal cancer, given that there is a stronger response in MSI-high tumors compared to MSI-low tumors104. TMB such as changes in DNA damage response genes and DNA polymerase epsilon (POLE) and delta (POLD) have been investigated as a biomarker in multiple tumor types103,105,106. Ang et al. performed a comprehensive genomic profiling of 755 patients with advanced HCC to evaluate the frequency of genomic biomarkers. The most commonly altered genes were TERT (44%), TP53 (35%), CTNNB1 (31%), ARID1A (12%), and MYC (12%), with median TMB for the entire cohort being four mutations per megabase103. Only 27 (4%) patients had POLE or POLD alterations, and there was no significant correlation between TMB and responders, progressors, or stable disease103. High tumor burden is hypothesized to generate elevated neoantigen expression by cancer cells that are not subject to immune tolerance, marking them as targets for clearance by the immune system103,107. The reported prevalence of MSI in HCC is approximately 0.80%–3% and, together with the relatively lower median TMB and lack of DNA repair mutations, provides an explanation for why TMB has failed to predict response in the majority of HCC patients108.

Signaling Pathways

Next-generation sequencing (NGS) of HCC tumors in patients treated with ICIs (either anti-PD/PD-L1 monotherapy, anti-CTLA-4 monotherapy, or combination anti-PD-1 with LAG-3, KIR, or CTLA-4) has identified alterations in Wnt/β-catenin signaling pathway to be associated with lower disease control rates, shorter median PFS, and OS109. WNT/β-catenin alterations may play a role in ICI response in HCC, as this signaling pathway is known to render tumors immunologically cold by way of T-lymphocyte exclusion110. Although the authors did not find any other pathways that correlated with responsiveness or resistance to ICI treatment, their work represents an attempt to use NGS to identify potential genetic markers that could improve current HCC treatment. Tumor cell biodiversity has been implicated in patient outcome after treatment with checkpoint inhibitors in HCC and intrahepatic cholangiocarcinoma111. In this study, single-cell RNA sequencing of liver cancer specimens from 19 patients treated with ICIs revealed a diverse landscape of TME. Tumors with less intratumoral diversity had a higher OS compared to highly diverse tumors111. Additionally, they found that T-cell dysfunction in these tumors was linked to increased VEGF expression, and combination therapy of ICIs with anti-VEGF therapy could potentially improve therapeutic outcomes. This provided the preclinical rationale for the phase III IMbrave150 trial, which compared combination atezoliumab and bevacizumab to sorafenib in patients with unresectable HCC who have not received prior systemic therapy20. In this study, 336 patients were treated with atezoliumab at a dose of 1,200 mg plus 15 mg/kg of bevacizumab intravenously, while the sorafenib group received 400 mg twice daily. The coprimary endpoints were OS and PFS by RECIST v1.1. With a median follow-up of 8.6 months, the trial demonstrated statistically significant and clinically meaningful improvement in both OS (median OS for the atezoliumab + bevacizumab group not reached versus 13.2 months in the sorafenib group) and PFS (median PFS of 6.8 months in the atezoliumab + bevacizumab group versus 4.3 months in the sorafenib group20. Grade 3–4 adverse events occurred in 57% of patients in the atezoliumab + bevacizumab group versus 55% in the sorafenib group. The safety profile of the combination was similar with known safety profile of each individual drug. This study is practice changing as this combination treatment may soon replace sorafenib as first-line treatment for advanced HCC pending regulatory approval.

Tumor-Infiltrating Lymphocytes

Intratumoral immune cell density have previously been correlated with survival in HCC. High densities of CD3+ and CD8+ T cells both intratumor and along the tumor margin of HCC samples were found to be significantly associated with lower recurrence rates and prolonged recurrence-free survival112–114. Additionally, tumor-infiltrating NK cells have also been associated with prolonged survival in HCC115. Similarly, higher densities of TILs have been associated with good prognosis in other types of cancers such as colorectal cancer and melanoma116,117. These findings suggest that both baseline and posttreatment TIL density could be important markers of response to ICI treatment. Indeed, this has led to the investigation of TILs as a biomarker of response to ICIs in numerous cancers. In patients with melanoma treated with pembrolizumab, Tumeh et al. found that presence of CD8+ TILs, which also was associated with higher PD-1/PD-L1 expression, correlated with radiographic reduction in tumor size118.

TILs have also been investigated as a biomarker of response to ICIs in HCC. Kaseb et al. found that clinical response in patients who received combination checkpoint inhibitor (nivolumab and ipilimumab) followed by surgical resection of HCC tumors correlated with an increase in CD8+ T-cell infiltration and specifically with two effector T-cell clusters (CD3+ CD8+ CD45RO+Eomes+ and CD3+ CD8+ CD45RO+Eomes+ CD57+ CD38low clusters)119. However, it is important to note that T-cell infiltration was already present in the tumor prior to treatment, likely due to chronic HCV infection. This study is currently ongoing, and final results may provide insight on the use of TILs as a biomarker. Further work is needed to clarify if background viral infection plays a role in the utility of TILs as a prognostic biomarker in HCC.

Circulating Soluble Factors

Circulating soluble factors such as cytokines have been investigated as biomarkers of response to ICIs. Recently, Feun et al. analyzed several representative circulating biomarkers before treatment and after 60 to 90 days of treatment with pembrolizumab in unresectable HCC patients and at the time of tumor response or disease progression91. Using enzyme-linked immunosorbent assay (ELISA), they detected interleukin-1β (IL-1β), IL-6, IL-8, IL-12, IL-18, IFN-γ, TGF-β, IL-10, CXCL9, CCL4, CCL5, PD-L1, and PD-L2 from patient plasma. They found that low baseline levels of TGF-β were significantly associated with improved OS and PFS after treatment with pembrolizumab. This study is in line with a previous study by Mariathasan et al., which shows that TGF-β attenuates tumor response to PD-L1 inhibition by excluding CD8+ effector T cells from the parenchyma120. TGF-β is known to promote immunosuppression through various mechanisms such as impaired differentiation or activation of innate and adaptive immune cells, inhibition of cytotoxic T-cell function, and impaired regulation of cytokine production121. Taken together, this suggests that TGF-β could be used as a predictive biomarker for response to PD-1/PD-L1 blockade.

Epithelial-to-Mesenchymal Transition

The TME has been shown to play a role in the mechanism of resistance to ICIs. Epithelial-to-mesenchymal transition (EMT) has been implicated as a resistance mechanism in HCC. EMT is known to promote immune evasion of cancer cells through Snail signaling122,123. Ueno et al. have shown that there is an association between EMT and PD-L1 expression in extrahepatic cholangiocarcinoma124. Shrestha et al. analyzed 422 HCC patient samples from the The Cancer Genome Atlas (TCGA) liver cancer database and found that high expression of PD-L1 and EMT markers (vimentin and E-cadherin) was significantly correlated with poor survival29. In fact, drugs that inhibit both PD-L1 expression and EMT have been developed for use in non-small cell lung cancer125. This correlation between PD-L1 expression and EMT presents an opportunity to investigate EMT as a potential biomarker for ICI response.

OVERCOMING RESISTANCE

Despite the success of ICIs, approximately 85% of HCC patients do not respond to ICIs. Newer approaches to overcome resistance to ICIs are desperately needed. One proposed mechanism of resistance to ICIs is the overexpression of alternate immune checkpoints such as T-cell immunoglobulin, mucin domain-3 protein TIM-3, and LAG-3126–128. A study by Thommen et al. showed a positive association between progressive T-cell exhaustion and increased coexpression of these alternate checkpoints in non-small cell lung cancer including BTLA129. This work provides a rationale for combination checkpoint therapy to increase efficacy of ICI therapy. In a preclinical study, pembrolizumab in combination with lenvatinib was shown to suppress tumor-associated macrophages, regulatory T cells resulting in decrease in TGF-β, IL-10, and downregulation of PD-1 and Tim3130. The combination of lenvatinib and pembrolizumab is on fast track designation by the FDA for the treatment of unresectable HCC. Combination therapy involving ICIs and antiangiogenic medications may work synergistically because VEGF-A inhibition increases tumor infiltration and survival of cytotoxic T lymphocytes, thereby producing a favorable microenvironment for ICIs to function131. Currently, combination atezoliumab and bevacizumab is also on fast track designation by the FDA for first-line treatment of HCC. Table 2 lists ongoing classes and examples of combination therapies that are being investigated in HCC cancer. In fact, it is possible that immunotherapy in HCC may move to combination triple therapy as one study has shown the efficacy of combination of PD-L1 blockade and CD137 plus OX40 (immunostimulatory agonists) against spontaneous liver cancer in transgenic mice132. One of the major limitations of combination therapy is significant immune-related adverse events from the treatment. Although these toxicities are rare, clinicians should always monitor for these events.

Table 2.

Classes of Therapies in Combination Therapy With Checkpoint Inhibitors in Hepatocellular Carcinoma

Class of Therapies Examples of Combinations Currently Tested Rationale
Combination with another checkpoint inhibitor Anti-PD-1/PD-L1 and CTLA-4; -PD-L1 and TIM-3; -PD-L1 and LAG-3 Inhibition of alternate inhibitory pathway in immune cells; increase number of activated CD8+ T cells156
Combination with multikinase inhibitor or antiangiogenic drug Anti-PD-1/PD-L1 with sorafenib; Anti-PD-1/PD-L1 with levantinib; Anti-PD-L1 and apatinib; Anti-PD-L1 with bevacizumab (IMbrave150) Reduces immunosuppressive Tregs and MDSCs; antiangiogenic properties may increase tumor hypoxia and enhance expression of immune checkpoint molecules157
Combination with local therapy Anti-PD-1 with TACE; Anti-CTLA-4 with RFA/TACE Enhanced immune cell activation and recruitment158; upregulation of soluble PD-L1159
Combination with oncolytic virus Anti-PD-1 with Pexa-Vec (JX-594) Promotion of NK and T-cell tumor infiltration160
Combination with polypeptide Anti-PD-1 with DSP-7888 (NCT03311334) Polypeptide HCC vaccine to expand preexisting neoantigen-specific T-cell population
Combination with antibiotics Anti-PD-1 with vancomycin and tadalafil (NCT03785210) Oral antibiotic alters gut commensal bacteria inducing antitumor effect

Another strategy to improve efficacy of ICIs is by priming adaptive response through treatments that release tumor antigens such as the addition of radiofrequency ablation (RFA), transarterial chemoembolization (TACE), transarterial radioembolization (TARE), stereotactic body radiation therapy (SBRT), or conventional chemotherapy51. Adaptive immune response can be primed by vaccines that use tumor-specific peptides to increase antigen presentation133. In a preclinical study, Chen et al. evaluated the effect of microwave ablation of subcutaneous hepatoma followed by combination intratumoral microspheres encapsulating GM-CSF and anti-CTLA-4 administration34. They found that this combination therapy resulted in local eradication of tumors and, surprisingly, led to rejections of tumors following rechallenge, including distant metastasis34. Radiation therapy has previously been shown to have a synergist effect in combination with ICIs134. There are several ongoing clinical trials (NCT03143270 and NCT01853618) looking at combination ICIs with local therapy (TACE or RFA)35. Tremelimumab in combination with tumor RFA leads to accumulation of intratumoral CD8+ T cells and is a potential treatment combination for patients with advanced HCC35. The results of these studies may shed light on another therapeutic option to improve the efficacy of ICIs in HCC.

T cell stimulation by delivering an oncolytic virus into the tumor can promote tumor infiltration and maturation of T and NK cells135. The oncolytic virus, which can sometimes be genetically modified, selectively targets and kills tumor cells in addition to stimulating the host’s immune system136,137. In melanoma, tamilogene laherparepvec (T-VEC) was the first approved oncolytic virus to be used in combination with ipilimumab or pembrolizumab and was found to have better efficacy than with monotherapy ICI138. JX-594, an oncolytic poxvirus, has been tried in patients with refractory primary and metastatic liver cancer and was found to be well tolerated139. There is an ongoing phase I/IIa clinical trial (NCT03071094) looking at the safety and efficacy of combination nivolumab with oncolytic viral therapy (Pexa-Vec) in advanced HCC.

The importance of gut microbiota in modulating key processes of inflammation and immunity has been a focus of recent studies. Alterations in the gut microbiota have been implicated in the progression of chronic liver disease and in the development of HCC140,141. Some studies have highlighted the relationship between gut microbiota and response to treatment with ICIs142,143. Sivan et al. found that oral administration of Bifidobacterium in combination with anti-PD-L1 therapy improved tumor control by increasing the accumulation of CD8+ T cells in melanoma142. It is unclear whether gut microbiota has a role in ICI response in HCC. There is an ongoing phase II trial (NCT03785210) looking at combination oral vancomycin, tadalafil, and nivolumab in patients with advanced HCC. The result of this study will hopefully shed light on the efficacy of altering the gut microbiome in patients with HCC who are on checkpoint inhibition.

FUTURE PERSPECTIVES

Since its introduction, ICI therapy has significantly changed the treatment of numerous malignancies especially in situations where there have been fewer alternatives. Despite its success in various malignancy, only very few patients with advanced HCC benefit from checkpoint inhibition. Understanding the mechanism of resistance and proper patient selection will hopefully provide better treatment results. The future of ICIs is reassuring as there are many ongoing clinical trials that could discover better biomarkers of response, combinatory treatments, and uncover newer methods of overcoming resistance to immunotherapy.

ACKNOWLEDGMENTS

This work was supported by the National Institutes of Health T32AI 106704-01A1 (to A.E.O.) and R01-CA214865-01 (to A.T.).

Footnotes

The authors declare no conflicts of interest.

REFERENCES

  • 1. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, Allen C, et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: Results from the Global Burden of Disease Study 2015. JAMA Oncol. 2017;3(12):1683–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, MacIntyre MF, et al. The global burden of cancer 2013. JAMA Oncol. 2015;1(4):505–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380(15):1450–62. [DOI] [PubMed] [Google Scholar]
  • 4. Liver EAFTSOT, Cancer EOFRATO. EASL-EORTC clinical practice guidelines: Management of hepatocellular carcinoma. J Hepatol. 2012;56(4):908–43. [DOI] [PubMed] [Google Scholar]
  • 5. Mittal S, El-Serag HB. Epidemiology of hepatocellular carcinoma: Consider the population. J Clin Gastroenterol. 2013;47(Suppl):S2–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Davila JA, Morgan RO, Shaib Y, McGlynn KA, El-Serag HB. Hepatitis C infection and the increasing incidence of hepatocellular carcinoma: A population-based study. Gastroenterology 2004;127(5):1372–80. [DOI] [PubMed] [Google Scholar]
  • 7. Maucort-Boulch D, de Martel C, Franceschi S, Plummer M. Fraction and incidence of liver cancer attributable to hepatitis B and C viruses worldwide. Int J Cancer 2018;142(12):2471–7. [DOI] [PubMed] [Google Scholar]
  • 8. Ghouri YA, Mian I, Rowe JH. Review of hepatocellular carcinoma: Epidemiology, etiology, and carcinogenesis. J Carcinog. 2017;16:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Kim HS, El-Serag HB. The epidemiology of hepatocellular carcinoma in the USA. Curr Gastroenterol Rep. 2019;21(4):17. [DOI] [PubMed] [Google Scholar]
  • 10. Akateh C, Black SM, Conteh L, Miller ED, Noonan A, Elliott E, et al. Neoadjuvant and adjuvant treatment strategies for hepatocellular carcinoma. World J Gastroenterol. 2019;25(28):3704–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–90. [DOI] [PubMed] [Google Scholar]
  • 12. Marrero JA, Kudo M, Venook AP, Ye SL, Bronowicki JP, Chen XP, et al. Observational registry of sorafenib use in clinical practice across Child-Pugh subgroups: The GIDEON study. J Hepatol. 2016;65(6):1140–7. [DOI] [PubMed] [Google Scholar]
  • 13. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10(1):25–34. [DOI] [PubMed] [Google Scholar]
  • 14. Jackson R, Psarelli EE, Berhane S, Khan H, Johnson P. Impact of viral status on survival in patients receiving sorafenib for advanced hepatocellular cancer: A meta-analysis of randomized phase iii trials. J Clin Oncol. 2017;35(6):622–8. [DOI] [PubMed] [Google Scholar]
  • 15. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018;391(10126):1163–73. [DOI] [PubMed] [Google Scholar]
  • 16. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;389(10064):56–66. [DOI] [PubMed] [Google Scholar]
  • 17. Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med. 2018;379(1):54–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Zhu AX, Park JO, Ryoo BY, Yen CJ, Poon R, Pastorelli D, et al. Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (REACH): A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2015;16(7):859–70. [DOI] [PubMed] [Google Scholar]
  • 19. Bruix J, Tak WY, Gasbarrini A, Santoro A, Colombo M, Lim HY, et al. Regorafenib as second-line therapy for intermediate or advanced hepatocellular carcinoma: Multicentre, open-label, phase II safety study. Eur J Cancer 2013;49(16):3412–9. [DOI] [PubMed] [Google Scholar]
  • 20. Cheng A-Lea. 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). ESMO 2019 Annual Congress, Late breaking abstract LBA38. Ann Oncol. 2019;30(Suppl 9). [Google Scholar]
  • 21. Rolfes V, Idel C, Pries R, Plötze-Martin K, Habermann J, Gemoll T, et al. PD-L1 is expressed on human platelets and is affected by immune checkpoint therapy. Oncotarget 2018;9(44):27460–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Greten TF, Sangro B. Targets for immunotherapy of liver cancer. J Hepatol. 2018;68(1):157–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Hato T, Goyal L, Greten TF, Duda DG, Zhu AX. Immune checkpoint blockade in hepatocellular carcinoma: Current progress and future directions. Hepatology 2014;60(5):1776–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13(4):227–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12(4):252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. The Nobel Prize in Physiology or Medicine 2018 [press release]. Nobel Media AB9 February 2020.
  • 27. Le Mercier I, Lines JL, Noelle RJ. Beyond CTLA-4 and PD-1, the generation z of negative checkpoint regulators. Front Immunol. 2015;6:418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Fourcade J, Sun Z, Pagliano O, Guillaume P, Luescher IF, Sander C, et al. CD8(+) T cells specific for tumor antigens can be rendered dysfunctional by the tumor microenvironment through upregulation of the inhibitory receptors BTLA and PD-1. Cancer Res. 2012;72(4):887–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Shrestha R, Prithviraj P, Anaka M, Bridle KR, Crawford DHG, Dhungel B, et al. Monitoring immune checkpoint regulators as predictive biomarkers in hepatocellular carcinoma. Front Oncol. 2018;8:269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ngiow SF, von Scheidt B, Akiba H, Yagita H, Teng MW, Smyth MJ. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Cancer Res. 2011;71(10):3540–51. [DOI] [PubMed] [Google Scholar]
  • 31. Kassel R, Cruise MW, Iezzoni JC, Taylor NA, Pruett TL, Hahn YS. Chronically inflamed livers up-regulate expression of inhibitory B7 family members. Hepatology 2009;50(5):1625–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Wang BJ, Bao JJ, Wang JZ, Wang Y, Jiang M, Xing MY, et al. Immunostaining of PD-1/PD-Ls in liver tissues of patients with hepatitis and hepatocellular carcinoma. World J Gastroenterol. 2011;17(28):3322–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Nemeth E, Baird AW, O’Farrelly C. Microanatomy of the liver immune system. Semin Immunopathol. 2009;31(3):333–43. [DOI] [PubMed] [Google Scholar]
  • 34. Chen Z, Shen S, Peng B, Tao J. Intratumoural GM-CSF microspheres and CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a subcutaneous murine hepatoma model. Int J Hyperthermia 2009;25(5):374–82. [DOI] [PubMed] [Google Scholar]
  • 35. Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66(3):545–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359(6382):1350–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Cohen R, Rousseau B, Vidal J, Colle R, Diaz LA, André T. Immune checkpoint inhibition in colorectal cancer: Microsatellite instability and beyond. Target Oncol. 2020;15(1):11–24. [DOI] [PubMed] [Google Scholar]
  • 41. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol. 2001;19:565–94. [DOI] [PubMed] [Google Scholar]
  • 42. Schneider H, Downey J, Smith A, Zinselmeyer BH, Rush C, Brewer JM, et al. Reversal of the TCR stop signal by CTLA-4. Science 2006;313(5795):1972–5. [DOI] [PubMed] [Google Scholar]
  • 43. Chen W, Jin W, Wahl SM. Engagement of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) induces transforming growth factor beta (TGF-beta) production by murine CD4(+) T cells. J Exp Med. 1998;188(10):1849–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008;322(5899):271–5. [DOI] [PubMed] [Google Scholar]
  • 45. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206(8):1717–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase ii and phase iii trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015;33(17):1889–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Eroglu Z, Kim DW, Wang X, Camacho LH, Chmielowski B, Seja E, et al. Long term survival with cytotoxic T lymphocyte-associated antigen 4 blockade using tremelimumab. Eur J Cancer 2015;51(17):2689–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Sangro B, Gomez-Martin C, de la Mata M, Iñarrairaegui M, Garralda E, Barrera 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(1):81–8. [DOI] [PubMed] [Google Scholar]
  • 49. FDA Pipeline: Recent designations in hepatocellular carcinoma, cholangiocarcioma, lymphoma, and more [press release]. 2019. [Google Scholar]
  • 50. Yau T, Kang Y-K, Kim T-Y. Nivolumab (NIVO) + ipilimumab (IPI) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): Results from CheckMate 040. J Clin Oncol. 2019;37(Suppl; abstr 4012). [Google Scholar]
  • 51. Iñarrairaegui M, Melero I, Sangro B. Immunotherapy of hepatocellular carcinoma: Facts and hopes. Clin Cancer Res. 2018;24(7):1518–24. [DOI] [PubMed] [Google Scholar]
  • 52. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol. 2016;34:539–73. [DOI] [PubMed] [Google Scholar]
  • 54. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 2007;27(1):111–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Xu F, Jin T, Zhu Y, Dai C. Immune checkpoint therapy in liver cancer. J Exp Clin Cancer Res. 2018;37(1):110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wei F, Zhong S, Ma Z, Kong H, Medvec A, Ahmed R, et al. Strength of PD-1 signaling differentially affects T-cell effector functions. Proc Natl Acad Sci USA 2013;110(27):E2480–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA 2002;99(19):12293–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8(6):467–77. [DOI] [PubMed] [Google Scholar]
  • 59. Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, Nakajima Y, et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res. 2009;15(3):971–9. [DOI] [PubMed] [Google Scholar]
  • 60. Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, Costentin C, et al. Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship With clinical and pathological features. Hepatology 2016;64(6):2038–46. [DOI] [PubMed] [Google Scholar]
  • 61. Jung HI, Jeong D, Ji S, Ahn TS, Bae SH, Chin S, et al. Overexpression of PD-L1 and PD-L2 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Res Treat. 2017;49(1):246–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Chang H, Jung W, Kim A, Kim HK, Kim WB, Kim JH, et al. Expression and prognostic significance of programmed death protein 1 and programmed death ligand-1, and cytotoxic T lymphocyte-associated molecule-4 in hepatocellular carcinoma. APMIS 2017;125(8):690–8. [DOI] [PubMed] [Google Scholar]
  • 63. Sideras K, Biermann K, Verheij J, Takkenberg BR, Mancham S, Hansen BE, et al. PD-L1, galectin-9 and CD8. Oncoimmunology 2017;6(2):e1273309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, 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(10088):2492–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Yau T, Park J, Finn R, Cheng A, Mathurin P, Edeline J, et al. CheckMate 459: A randomized, multi-center phase 3 study of nivolumab (NIVO) vs sorafenib (SOR) as first-line (1L) treatment in patients (pts) with advanced hepatocellular carcinoma (aHCC). Ann Oncol. 2019;30(Suppl 5):v874–5. [Google Scholar]
  • 66. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, 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(7):940–52. [DOI] [PubMed] [Google Scholar]
  • 67. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, 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(3):193–202. [DOI] [PubMed] [Google Scholar]
  • 68. Brown ZJ, Heinrich B, Steinberg SM, Yu SJ, Greten TF. Safety in treatment of hepatocellular carcinoma with immune checkpoint inhibitors as compared to melanoma and non-small cell lung cancer. J Immunother Cancer 2017;5(1):93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of resistance to immune checkpoint blockade: Why does checkpoint inhibitor immunotherapy not work for all patients? Am Soc Clin Oncol Educ Book 2019;39:147–64. [DOI] [PubMed] [Google Scholar]
  • 70. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168(4):707–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Pu X, Wu L, Su D, Mao W, Fang B. Immunotherapy for non-small cell lung cancers: Biomarkers for predicting responses and strategies to overcome resistance. BMC Cancer 2018;18(1):1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Topalian SL, Wolchok JD, Chan TA, Mellman I, Palucka K, Banchereau J, et al. Immunotherapy: The path to win the war on cancer? Cell 2015;161(2):185–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ruiz de Galarreta M, Bresnahan E, Molina-Sánchez P, Lindblad KE, Maier B, Sia D, et al. β-Catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 2019;9(8):1124–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Nejak-Bowen K, Kikuchi A, Monga SP. Beta-catenin-NF-κB interactions in murine hepatocytes: A complex to die for. Hepatology 2013;57(2):763–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer 2019;7(1):193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019;19(3):133–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Buder-Bakhaya K, Hassel JC. Biomarkers for clinical benefit of immune checkpoint inhibitor treatment—A review from the melanoma perspective and beyond. Front Immunol. 2018;9:1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Jeyakumar G, Kim S, Bumma N, Landry C, Silski C, Suisham S, et al. Neutrophil lymphocyte ratio and duration of prior anti-angiogenic therapy as biomarkers in metastatic RCC receiving immune checkpoint inhibitor therapy. J Immunother Cancer 2017;5(1):82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Spencer KR, Wang J, Silk AW, Ganesan S, Kaufman HL, Mehnert JM. Biomarkers for immunotherapy: Current developments and challenges. Am Soc Clin Oncol Educ Book 2016;35:e493–503. [DOI] [PubMed] [Google Scholar]
  • 80. 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(7528):563–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, et al. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol. 2016;34(34):4102–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Gandini S, Massi D, Mandalà M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016;100:88–98. [DOI] [PubMed] [Google Scholar]
  • 83. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–e51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016;16(5):275–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206(6):1327–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Pinato DJ, Mauri FA, Spina P, Cain O, Siddique A, Goldin R, et al. Clinical implications of heterogeneity in PD-L1 immunohistochemical detection in hepatocellular carcinoma: The Blueprint-HCC study. Br J Cancer 2019;120(11):1033–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Zhong JH, Luo CP, Zhang CY, Li LQ. Strengthening the case that elevated levels of programmed death ligand 1 predict poor prognosis in hepatocellular carcinoma patients. J Hepatocell Carcinoma 2017;4:11–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Liu X, Qin S. Immune checkpoint inhibitors in hepatocellular carcinoma: Opportunities and challenges. Oncologist 2019;24(Suppl 1):S3–S10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Finkelmeier F, Canli Ö, Tal A, Pleli T, Trojan J, Schmidt M, et al. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer 2016;59:152–9. [DOI] [PubMed] [Google Scholar]
  • 90. Tai D, Choo SP, Chew V. Rationale of immunotherapy in hepatocellular carcinoma and its potential biomarkers. Cancers (Basel) 2019;11(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Feun LG, Li YY, Wu C, Wangpaichitr M, Jones PD, Richman SP, et al. Phase 2 study of pembrolizumab and circulating biomarkers to predict anticancer response in advanced, unresectable hepatocellular carcinoma. Cancer 2019;125(20):3603–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, et al. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 2018;17(1):129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Sharma P. Immune checkpoint therapy and the search for predictive biomarkers. Cancer J. 2016;22(2):68–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Vilain RE, Menzies AM, Wilmott JS, Kakavand H, Madore J, Guminski A, et al. Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin Cancer Res. 2017;23(17):5024–33. [DOI] [PubMed] [Google Scholar]
  • 95. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 2017;19(6):1189–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, et al. IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br J Cancer 2015;112(9):1501–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Karachaliou N, Gonzalez-Cao M, Crespo G, Drozdowskyj A, Aldeguer E, Gimenez-Capitan A, et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther Adv Med Oncol. 2018;10:1758834017749748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127(8):2930–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Friedman CF, Postow MA. Emerging tissue and blood-based biomarkers that may predict response to immune checkpoint inhibition. Curr Oncol Rep. 2016;18(4):21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359(6371):91–7. [DOI] [PubMed] [Google Scholar]
  • 101. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature 2013;500(7463):415–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Berland L, Heeke S, Humbert O, Macocco A, Long-Mira E, Lassalle S, et al. Current views on tumor mutational burden in patients with non-small cell lung cancer treated by immune checkpoint inhibitors. J Thorac Dis. 2019;11(Suppl 1):S71–S80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Ang C, Klempner SJ, Ali SM, Madison R, Ross JS, Severson EA, et al. Prevalence of established and emerging biomarkers of immune checkpoint inhibitor response in advanced hepatocellular carcinoma. Oncotarget 2019;10(40):4018–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Gong J, Wang C, Lee PP, Chu P, Fakih M. Response to PD-1 blockade in microsatellite stable metastatic colorectal cancer harboring a POLE mutation. J Natl Compr Canc Netw. 2017;15(2):142–7. [DOI] [PubMed] [Google Scholar]
  • 106. Rayner E, van Gool IC, Palles C, Kearsey SE, Bosse T, Tomlinson I, et al. A panoply of errors: Polymerase proofreading domain mutations in cancer. Nat Rev Cancer 2016;16(2):71–81. [DOI] [PubMed] [Google Scholar]
  • 107. Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: Building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125(9):3413–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, et al. Prospective genotyping of hepatocellular carcinoma: Clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin Cancer Res. 2019;25(7):2116–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015;523(7559):231–5. [DOI] [PubMed] [Google Scholar]
  • 111. Ma L, Hernandez MO, Zhao Y, Mehta M, Tran B, Kelly M, et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell 2019;36(4):418–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Gabrielson A, Wu Y, Wang H, Jiang J, Kallakury B, Gatalica Z, et al. Intratumoral CD3 and CD8 T-cell densities associated with relapse-free survival in HCC. Cancer Immunol Res. 2016;4(5):419–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: A systematic review with meta-analysis. Br J Cancer 2011;105(1):93–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Zeng DQ, Yu YF, Ou QY, Li XY, Zhong RZ, Xie CM, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes for clinical therapeutic research in patients with non-small cell lung cancer. Oncotarget 2016;7(12):13765–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Chew V, Tow C, Teo M, Wong HL, Chan J, Gehring A, et al. Inflammatory tumour microenvironment is associated with superior survival in hepatocellular carcinoma patients. J Hepatol. 2010;52(3):370–9. [DOI] [PubMed] [Google Scholar]
  • 116. Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58(16):3491–4. [PubMed] [Google Scholar]
  • 117. Taylor RC, Patel A, Panageas KS, Busam KJ, Brady MS. Tumor-infiltrating lymphocytes predict sentinel lymph node positivity in patients with cutaneous melanoma. J Clin Oncol. 2007;25(7):869–75. [DOI] [PubMed] [Google Scholar]
  • 118. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515(7528):568–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Kaseb AO, Vence L, Blando J, Yadav SS, Ikoma N, Pestana RC, et al. Immunologic correlates of pathologic complete response to preoperative immunotherapy in hepatocellular carcinoma. Cancer Immunol Res. 2019;7(9):1390–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554(7693):544–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery JP, et al. New insights into the role of EMT in tumor immune escape. Mol Oncol. 2017;11(7):824–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 2009;15(3):195–206. [DOI] [PubMed] [Google Scholar]
  • 124. Ueno T, Tsuchikawa T, Hatanaka KC, Hatanaka Y, Mitsuhashi T, Nakanishi Y, et al. Prognostic impact of programmed cell death ligand 1 (PD-L1) expression and its association with epithelial-mesenchymal transition in extrahepatic cholangiocarcinoma. Oncotarget 2018;9(28):20034–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Liakopoulou C, Kazazis C, Vallianou NG. Silimarin and cancer. Anticancer Agents Med Chem. 2018;18(14):1970–4. [DOI] [PubMed] [Google Scholar]
  • 126. Shayan G, Srivastava R, Li J, Schmitt N, Kane LP, Ferris RL. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology 2017;6(1):e1261779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Huang RY, Francois A, McGray AR, Miliotto A, Odunsi K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology 2017;6(1):e1249561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Thommen DS, Schreiner J, Müller P, Herzig P, Roller A, Belousov A, et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res. 2015;3(12):1344–55. [DOI] [PubMed] [Google Scholar]
  • 130. Kato Yea. Lenvatinib mesilate (LEN) enhanced antitumor activity of a PD-1 blockade agent by potentiating Th1 immune response. Ann Oncol. 2016;27(Suppl 6). [Google Scholar]
  • 131. Hilmi M, Neuzillet C, Calderaro J, Lafdil F, Pawlotsky JM, Rousseau B. Angiogenesis and immune checkpoint inhibitors as therapies for hepatocellular carcinoma: Current knowledge and future research directions. J Immunother Cancer 2019;7(1):333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Morales-Kastresana A, Sanmamed MF, Rodriguez I, Palazon A, Martinez-Forero I, Labiano S, et al. Combined immunostimulatory monoclonal antibodies extend survival in an aggressive transgenic hepatocellular carcinoma mouse model. Clin Cancer Res. 2013;19(22):6151–62. [DOI] [PubMed] [Google Scholar]
  • 133. Buonaguro L, Consortium H. Developments in cancer vaccines for hepatocellular carcinoma. Cancer Immunol Immunother. 2016;65(1):93–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Jhawar SR, Thandoni A, Bommareddy PK, Hassan S, Kohlhapp FJ, Goyal S, et al. Oncolytic viruses—Natural and genetically engineered cancer immunotherapies. Front Oncol. 2017;7:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Prestwich RJ, Harrington KJ, Pandha HS, Vile RG, Melcher AA, Errington F. Oncolytic viruses: A novel form of immunotherapy. Expert Rev Anticancer Ther. 2008;8(10):1581–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Aurelian L. Oncolytic viruses as immunotherapy: Progress and remaining challenges. Onco Targets Ther. 2016;9:2627–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Puzanov I, Milhem MM, Minor D, Hamid O, Li A, Chen L, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34(22):2619–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: A phase I trial. Lancet Oncol. 2008;9(6):533–42. [DOI] [PubMed] [Google Scholar]
  • 140. Yu LX, Schwabe RF. The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 2017;14(9):527–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, et al. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol. 2018;15(7):397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015;350(6264):1084–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015;350(6264):1079–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7(12):1420–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer 2018;118(1):9–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ozcan M, Janikovits J, von Knebel Doeberitz M, Kloor M. Complex pattern of immune evasion in MSI colorectal cancer. Oncoimmunology 2018;7(7):e1445453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7(2):188–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Gao J, Shi LZ, Zhao H, Chen J, Xiong L, He Q, et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016;167(2):397–404.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. George S, Miao D, Demetri GD, Adeegbe D, Rodig SJ, Shukla S, et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 2017;46(2):197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116(7):1935–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210(9):1695–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Viehl CT, Moore TT, Liyanage UK, Frey DM, Ehlers JP, Eberlein TJ, et al. Depletion of CD4+CD25+ regulatory T cells promotes a tumor-specific immune response in pancreas cancer-bearing mice. Ann Surg Oncol. 2006;13(9):1252–8. [DOI] [PubMed] [Google Scholar]
  • 153. Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother. 2014;63(3):247–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J Leukoc Biol. 2017;102(3):727–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2017;168(3):542. [DOI] [PubMed] [Google Scholar]
  • 156. Zhou G, Sprengers D, Boor PPC, Doukas M, Schutz H, Mancham S, et al. Antibodies against immune checkpoint molecules restore functions of tumor-infiltrating T cells in hepatocellular carcinomas. Gastroenterology 2017;153(4):1107–19.e10. [DOI] [PubMed] [Google Scholar]
  • 157. Hato T, Zhu AX, Duda DG. Rationally combining anti-VEGF therapy with checkpoint inhibitors in hepatocellular carcinoma. Immunotherapy 2016;8(3):299–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Chew V, Lee YH, Pan L, Nasir NJM, Lim CJ, Chua C, et al. Immune activation underlies a sustained clinical response to Yttrium-90 radioembolisation in hepatocellular carcinoma. Gut 2019;68(2):335–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Kim HJ, Park S, Kim KJ, Seong J. Clinical significance of soluble programmed cell death ligand-1 (sPD-L1) in hepatocellular carcinoma patients treated with radiotherapy. Radiother Oncol. 2018;129(1):130–5. [DOI] [PubMed] [Google Scholar]
  • 160. LaRocca CJ, Warner SG. Oncolytic viruses and checkpoint inhibitors: Combination therapy in clinical trials. Clin Transl Med. 2018;7(1):35. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Gene Expression are provided here courtesy of Cognizant Communication Corporation

RESOURCES