Next Article in Journal
A Computational Framework for Comprehensive Genomic Profiling in Solid Cancers: The Analytical Performance of a High-Throughput Assay for Small and Copy Number Variants
Previous Article in Journal
Development of Allogeneic Stem Cell-Based Platform for Delivery and Potentiation of Oncolytic Virotherapy
Previous Article in Special Issue
Thymic Stromal Lymphopoietin Induction Suppresses Lung Cancer Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Repurposing of Commercially Existing Molecular Target Therapies to Boost the Clinical Efficacy of Immune Checkpoint Blockade

1
Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
2
Mater Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(24), 6150; https://doi.org/10.3390/cancers14246150
Submission received: 24 October 2022 / Revised: 29 November 2022 / Accepted: 7 December 2022 / Published: 13 December 2022

Abstract

:

Simple Summary

Epithelial cancers, such as lung, breast, and colon cancers, have high mortality rates because of their ability to spread across multiple organs in the body. Besides the standard of care which includes chemotherapy and radiotherapy, approaches directed to use patient’s own immune responses against the disease called immunotherapies have emerged as a powerful treatment option. In the past 10 years, immune checkpoint blockade, a form of immunotherapy which either stimulates or removes the breaks of the immune response against cancer, is having the largest impact in the clinic. However epithelial cancers are commonly either naturally resistant or develop resistance to these types of treatments. Hence, there is an urgent need to boost the effectiveness of immune checkpoint blockers. Small molecule inhibitors are chemical molecules which are specifically designed to target important cancer proteins and unlike chemotherapy, typically have manageable toxicity. These inhibitors have shown good efficacy in reducing tumour growth but more recently, they have been shown to enhance the performance of immune cells in eliminating cancers. In this review, we have focused on tactical usage of small molecule inhibitors to boost the efficacy of immune checkpoint blockers. We believe our review will pave the way for novel research combining the two therapeutic modalities.

Abstract

Immune checkpoint blockade (ICB) is now standard of care for several metastatic epithelial cancers and prolongs life expectancy for a significant fraction of patients. A hostile tumor microenvironment (TME) induced by intrinsic oncogenic signaling induces an immunosuppressive niche that protects the tumor cells, limiting the durability and efficacy of ICB therapies. Addition of receptor tyrosine kinase inhibitors (RTKi) as potential modulators of an unfavorable local immune environment has resulted in moderate life expectancy improvement. Though the combination strategy of ICB and RTKi has shown significantly better results compared to individual treatment, the benefits and adverse events are additive whereas synergy of benefit would be preferable. There is therefore a need to investigate the potential of inhibitors other than RTKs to reduce malignant cell survival while enhancing anti-tumor immunity. In the last five years, preclinical studies have focused on using small molecule inhibitors targeting cell cycle and DNA damage regulators such as CDK4/6, CHK1 and poly ADP ribosyl polymerase (PARP) to selectively kill tumor cells and enhance cytotoxic immune responses. This review provides a comprehensive overview of the available drugs that attenuate immunosuppression and overcome hostile TME that could be used to boost FDA-approved ICB efficacy in the near future.

1. Introduction

One of the hallmarks of cancer is the ability of malignant cells to avoid immune surveillance. Tumors subvert many different normal immunosuppressive mechanisms to either block detection or suppress immune recognition. One mechanism that has proven to be targetable and enhances immune recognition is the immune checkpoint pathways. Research on regulation of normal immune responses has identified inhibitory receptors on immunocytes whose normal function is to limit healthy immune responses or block auto-immune detection and response. Antibodies that block inhibitory receptors can enhance and prolong immune responses. This research has enabled development of immune checkpoint blockade (ICB) monoclonal antibodies (mAbs) that improve clinical outcome for multiple cancers, notably metastatic melanoma, for which the 5 years survival rate can reach up to 44% with nivolumab treatment, and 26% for ipilimumab treatment [1,2]. Currently, Food and Drug Administration (FDA) approved ICB monoclonal antibodies (mAbs) in use in the clinic are: (i) ipilimumab (Yervoy®) blocking cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4); (ii) cemiplimab (Libtayo®), (iii) nivolumab (Opdivo®) and (iv) pembrolizumab (Keytruda®) inhibiting programmed death-1 (PD-1); (v) atezolizumab (Tecentriq®), (vi) avelumab (Bavencio®), and (vii) durvalumab (Imfinzi®) targeting programmed death ligand-1 (PD-L1); and very recently approved (March, 2022) (viii) relatlimab (Opdualag®), targeting lymphocyte activation gene-3 (LAG-3) and (ix) tiragolumab (Tecentriq®) against T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT) [3,4,5,6,7]. Additionally, targeting other immune checkpoint molecules such as T cell immunoglobulin and mucin-domain containing-3 (TIM-3), Signal regulatory protein α (SIRP α) and V-domain immunoglobulin (Ig) suppressor of T cell activation (VISTA) using mAbs are also currently under evaluation at either at preclinical or early phase of clinical trials [8,9,10].
The immune checkpoints (namely PD-1, CTLA-4, LAG-3, TIM-3, TIGIT) curtail overstimulation of the immune system post antigen exposure to restore normal homeostasis and avoid exacerbated immune responses. This balance is maintained by binding of these inhibitory receptors expressed by several immune subsets including T cells or NK cells, with complementary co-stimulatory ligands expressed by antigen presenting cells (APCs) and other myeloid cells, respectively. Interestingly, in cancer, tumor cells upregulate the expression of PD-L1 which binds PD-1 with high affinity resulting in the inactivation of Zeta-chain-associated protein kinase 70 (ZAP70) and CD28 and subsequent TCR signalling cascade inhibition. CTLA-4 competes with T-cell activation receptor CD28 for binding to CD80 and CD86 (co-stimulatory molecules). These receptors are highly expressed by cancer cells and upon interaction with CTLA-4 results in reduction in T cell proliferation and interleukine-2 (IL-2) production [11]. Other checkpoint molecules bind to their respective targets expressed on cancer cells to trigger immune malfunction and to facilitate immune evasion. Therefore, the rationale behind inhibition of these checkpoint interactions through engineered ICB mAbs is to override immunosuppression facilitating reactivation of the adaptive immune response [3]. However, apart from melanoma, the response rates to ICB mAbs across a variety of tumor types have generally been less than 30% and face a stiff challenge in clinic [12] (Table 1).
Immunotherapy for epithelial cancers can fail because there is an immunosuppressive tumor microenvironment (TME) as reviewed by de Miguel M, et al. [30]. Development of resistance to therapy, with local or metastatic tumor recurrence, occurs with ICB mAbs, as it does for small molecule drugs targeting cell tumor metabolism [31]. Hence, understanding how cancer cells influence the local immune environment and how small molecule cytotoxic cancer therapies can improve tumor immunogenicity is essential to design new strategies that can enhance the therapeutic effect of ICB mAbs. In this review, we focus on several anti-cancer drugs that also modulate the immune anti-tumor response and could be strategically repurposed in combination with FDA-approved ICB mAbs to improve clinical outcomes.

2. Mechanism Driving Resistance to ICB

The TME consists of a heterogeneous population of cells that collectively contribute pro-immune and immunosuppressive signalling as shown in Figure 1. Macrophages, myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), and cell-free factors including anti-inflammatory cytokines are major contributors to local immunosuppression. These factors generate a protective shield to defends the tumor from cytotoxic T cells by inhibiting their T cell trafficking and proliferation. This can be achieved by suppressing neoantigen or tumor associated antigen presentation, or by inducing T cell exhaustion. As these processes have been extensively reviewed, we will briefly highlight their importance in driving resistance to ICB [32,33]. T cell trafficking comprises a step wise process of rolling, adhesion, extravasation, and chemotaxis, governed by pro- and anti-inflammatory chemokines (chemokine (C-C motif) ligand (CCL) 5, CCL17, CCL22, chemokine (C-X-C motif) ligand (CXCL) 8, and CXCL12) and cytokines including interleukin (IL)-4, IL-6, IL-10, IL-11, and IL-13. This cocktail of secreted molecules favours mobilization of MDSCs and Tregs hindering the recruitment of cytotoxic T cells to the tumor [34]. Similarly, impaired interferon (IFN)-γ signalling accompanied by suppression of dendritic cell (DC) maturation and recruitment leads to hindered T cell proliferation and priming [35].
Additionally, impaired adaptive immune responses can result from reduced tumor antigen presentation, a consequence of downregulation of expression of major histocompatibility complex (MHC)-I on cancer cells [36]. Post-translationally, loss of β2-microglobulin is the major contributor to disruption of MHC-I folding and transport to the cell surface [37]. Interestingly, mutations within the T cell receptor binding domain of MHC, reported in colorectal cancer, block immunosurveillance by abrogating cytotoxicity [38]. Intrinsically, immunologically cold tumors, such as pancreatic and triple negative breast cancers, have low tumor mutational burden (TMB), limiting presentation of immunogenic neoantigens, and thus tumor specific cytotoxic T cell repertoires [39,40]. Over ten years of ICB’s use in the clinic, have shown that tumors lacking neoantigen presentation have poor treatment outcome, whereas tumors with high TMB and neoantigen presentation, including melanoma, have an improved ICB response [41].
Additional immunosuppressive mechanisms employed by cancer cells include expression of immunomodulatory ligands (PD-L1, CD47 and CD155) that bind to corresponding cytotoxic T cells receptors (PD-1, SIRPα and TIGIT). Persistent signalling induced by these interactions leads to T cell exhaustion. Though ICB mAbs should bypass these mechanisms, compensatory upregulation of other checkpoint pathways, such as lymphocyte-activation gene 3 (LAG-3) or CTLA-4 can follow [42]. Activation of multiple checkpoint signalling networks can contribute to treatment failure after prolonged ICB cancer treatment, highlighting the need for combination strategies to minimize drug resistance and maximise the durability and efficacy of ICB mAbs.

3. Repurposing SMIs to Improve Efficacy of ICB

Targeted therapies using small molecules (SMIs) known to inhibit molecular or biochemical pathways critical for tumor growth and maintenance also have an impact on tumor infiltrating immune effector cells. Combinations of SMIs with immune checkpoint blockade has proven to be effective in pre-clinical models, and several clinical trials are underway (Table 2) [43]. Therapies targeting receptor tyrosine kinases (RTK) can induce immunogenic modulation either by improving the cytotoxic function of the adaptive immune system, or by blocking expression of immunosuppressive molecules such as PD-L1, to enhance T cell mediated elimination of cancer cells. In addition, RTK inhibitors increase frequency and function of effector immune cells in the TME of epithelial cancers (i.e., melanoma and colon cancer) while decreasing the number and function of immune suppressor cells [44,45].
SMIs can target RTKs such as Epidermal Growth Factor Receptor (EGFR), v-raf murine sarcoma viral oncogene homolog B1 (BRAF), KIT, Human Epidermal growth factor Receptor-2 (HER-2), phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3CA)/AKT/mammalian Target Of Rapamycin (mTOR) and Anaplastic Lymphoma Kinase (ALK). EGFR inhibitor-based therapies (Sunitinib, axitinib, erlotinib, gefitinib, imatinib) can influence T cell priming, increasing memory and effector T cell phenotypes. EGFR inhibitors can also impact on T cell tumor antigen recognition, activation, and trafficking of immune cells into the tumor. Additionally, they can sensitize cancer cells to immune effector cell mediated killing and antagonize cancer-induced immune suppression [57]. EGFR inhibitors also augment DCs function and tumor antigen presentation, enabling better T cell-mediated tumor destruction [57].
SMIs blocking immune check points can directly remodel the immune response in the TME. The blockage of the innate immune checkpoint CD47/SIRPα pathway using either antibodies or SMIs has been extensively investigated [58,59,60]. However, CD47 is a ubiquitously expressed cell surface protein also found on red blood cells and antibodies against CD47 have been associated with adverse events including anaemia [60]. Unlike ICBs, SMIs targeting CD47 displayed less toxicity as they either disrupt CD47/SIRPα interaction or modulate CD47 at the transcriptional, translational, and post-translational modification levels [60]. A good example is the SMI RRx-001 which skews the phenotype of tumor infiltrating macrophages from immune-suppressive M2 to highly phagocytic M1 [61]. In contrast to anti-CD47 antibodies, RRx-001 showed no hematologic toxicities in 9 clinical trials (~300 patients involved) and has positively progressed to Phase III (NCT03699956 and NCT02489903) [62,63]. As the majority of ICBs harness the power of adaptive immunity, the combination of these agents with innate immunity modulator such as RRx-001 is a very attractive approach that will need further evaluation with pre-clinical studies.
Although, effective LAG-3 SMIs are lacking promising development, recent research revealed that SMIs blocking glycogen synthase kinase-3 (GSK-3), such as SB415286 and elraglusib, not only down-regulated PD-1 expression enhancing CD8+ T cell cytotoxicity, but also reduced LAG-3 levels on T cells in mice [64]. Interestingly, the combination of GSK-3 SMI with anti-LAG-3 mAbs had a synergistic effect and was more effective than the SMI monotherapy alone in a melanoma mouse model [65,66].
TIGIT has proven to be of clinical interest due to its dual expression on tumor and immune cells, such as NKs and CD8+ tumor infiltrating T cells, and the positive correlation between TIGIT levels and PD-1 expression on human melanoma infiltrating CD8+ T cells [67,68]. In addition to anti-TIGIT antibodies, FDA-approved TIGIT SMIs liothyronine and azelnidipine are approved by the FDA with ability to block the interaction between TIGIT and CD155 [69,70]. Although liothyronine did not inhibit tumor cell proliferation in vitro, it significantly abrogated tumor growth in an in vivo model of colon adenocarcinoma (MC-38) by increasing the levels of CD8+ T cells within the tumor, protection that was lost when either CD4+ or CD8+ T or NK cells were depleted [70]. Interestingly, azelnidipine inhibits both CD47/SIRPα and TIGIT/PVR pathways by binding SIRPα and CD155, to enhance macrophage phagocytic activity and increase the infiltration of CD8+ T cells in murine MC-38 tumors. Although both TIGIT SMIs showed encouraging effects in tumor-bearing mice, their potential off target effects in endocrine system or ion channels should be carefully monitored in future clinical trials [69,70]. The recent FDA-approval of relatlimab (anti-LAG3, Opdualag®) in combination with nivolumab and tiragolumab (anti-TIGIT, Tecentriq®) to treat metastatic melanoma and NSCLC patients, respectively. This opens the door for new combination therapies with some of the previously mentioned FDA-approved ICB and SMIs that have the potential to synergistically reverse immune suppression and enhance anti-tumor response.
The mitogen-activated protein kinase (MAPK) signalling pathway is critical for tumor cell growth, proliferation, invasion, and metastasis in multiple cancers [71,72]. Mitogen Expressing Kinase (MEK) inhibitors (PD098059, trametinib and cobimetinib) were the first drugs developed to suppress the MAPK pathway. However, despite their high potency and selectivity, clinical response to MEK inhibitors as a single agent was largely disappointing [73]. Recent studies have shown the potential of MEK inhibitors for use in immune-sensitization by up-regulation of tumor antigen expression and presentation [74,75], and through production of IL-8 and vascular endothelial growth factor (VEGF), enhancing recruitment of immune cell to the tumor site [76]. Notably, Kang et al. [77] demonstrated in human NSCLC that trametinib (MEK1/2 inhibitor) enhances MHC-class I expression via signal transducer and activator of transcription-3 (STAT3) activation and upregulates chemokines associated with T cell infiltration and homing. Interestingly, a recent study, using a murine syngeneic BRAFV600E melanoma model, demonstrated improved efficacy of PMEL (premelanosome protein)-1-specific adaptive cell therapy, when combined with the BRAF + MEK inhibitors dabrafenib and trametinib [78]. The triple combination increased tumor T cell infiltration, leading to complete tumor regression [78]. Also recently, experiments using a head and neck squamous cell carcinoma (HNSC) model demonstrated that trametinib delays tumor initiation and progression by enhancing CD8+ T cell antitumor function and promoting development of long-term memory cells when combined with anti-PD-1 [79]. Clinical trials in which RTK inhibitors have been combined with ICB mAb therapy have shown promising response (Table 2). Other SMIs with immunomodulatory capacities, discussed below, are yet to be tested in combination with ICB mAb therapy and could also prove to be effective cancer therapeutics.

4. Taking Advantage of Cell Cycle Inhibitors

Deregulation of the cell cycle is a well-known hallmark of tumorigenesis and to date, multiple SMIs have been designed to target major players known to modulate this pathway in the cancer setting. The most interesting SMIs are designed to target the aberrant activity of CDK4/6 (FDA approved palbociclib, ribociclib, and abemaciclib). Overexpression of Cyclin D1 (the binding partner of CDK4/6) alongside loss of function of p16INK4a (the endogenous CDK4/6 inhibitor), enables abnormal function of CDK4/6 leading to compromising the G1/S checkpoint of the cell cycle [80]. Though CDK4/6 inhibitors have been extensively utilised for the treatment of hormonal breast cancer, recent studies in melanoma (using mouse models) have highlighted their complementary immunotherapeutic activity [81,82,83,84,85]. Palbociclib has been shown to improve the anti-tumor efficacy of anti-PD-1/PD-L1 ICBs by enhancing MHC-I expression through type III interferon production. This drug reduces PD-L1 expression in mouse breast cancer cells and increases tumor cell production of T cell stimulants, such as CXCL10 and CXCL13 chemokines resulting in an increased lymphocyte recruitment within the TME [85,86,87]. In addition, CDK4/6i can act directly on T cells by diminished Treg proliferation and enhancing effector T cell activity through downregulation of nuclear factor of activated T cells (NFAT), that regulates transcription in Tregs [81,82,88]. In mouse model, breast tumors treated with CDK4/6i showed an enhancement of stem or memory-like cytotoxic CD8+ T cells responsible for sustained clinical responses to ICB [86].
WEE1, an important regulator of G2/M phase of the cell cycle, has been recently shown to play an important role in dictating anti-tumor immune responses in preclinical small cell lung cancer models. Using AZD1775 (WEE1 inhibitor), Taniguchi et al. [89] demonstrated that WEE1 inhibition led to activation of the stimulator of interferon genes (STING)-TANK binding kinase (TBK)-interferon regulatory factor (IRF3) pathway which increased production of type I interferons (IFN-α and IFN-β) alongside pro-inflammatory chemokines (CXCL10 and CCL5). Furthermore, WEE1 inhibition triggered upregulation of STAT1 which induced upregulation of PD-L1 and IFN-γ expression, but upon combination with anti-PD-L1 blockade induced anti-tumor immune response in a CD8+ T cell dependent-manner [89].
Polo like kinase 1 (PLK1) is an important player in the regulation of the mitotic phase of the cell cycle and its expression is deregulated during tumorigenesis. PLK1 overexpressing tumors (most epithelial cancers) have been shown to have minimal tumor infiltrates alongside low MHC-I expression [90]. Metadata analysis of publicly available genomic data from TCGA (The Cancer Genome Atlas Program) dataset on 33 different cancer types patients who were treated with PLK1 inhibitor demonstrated increased anti-tumor immunity characterized by an upregulated expression of NK (natural killer)-cell-like gene signatures and genes involved in antigen presentation such as Transporter associated with antigen processing 1 & 2 (TAP1 and TAP2) [90]. In another study using preclinical NSCLC mouse model, PLK1i (BI2536) enhanced DC maturation and T cell infiltration [91]. Aurora Kinase A (AURKA) is an upstream regulator of PLK1 and regulates centrosome maturation and spindle formation in mitosis. Interestingly, in a recent study using a murine mammary tumor model, Alisertib (AURKA inhibitor) in combination with anti-PD-L1 therapy induced tumor regression. This combination was associated with reduced numbers of tumor-promoting myeloid cells (induced apoptosis of MDSCs) alongside significant increases of active CD8+ and CD4+ T cells [92].
KRAS (Kirsten rat sarcoma), is an important oncogene and its mutation is known to drive abnormal cell cycle progression and tumorigenesis in NSCLC, pancreatic ductal adenocarcinoma, and colorectal cancer (CRC). The common missense mutations observed in KRAS oncogene are: G12, G13, and Q61 and have been extensively investigated for designing targeted therapies [93]. Ostrem et al. [94] identified docking pocket in the KRAS-G12C mutant paving way for designing multiple covalent inhibitors. AMG 510 (sotorasib) was the first drug candidate which demonstrated success in clinical trials for KRAS-mutant cancers, especially NSCLC patients with KRAS-G12C mutation (32.2% achieved objective response and 88.1% achieved disease control) [95,96,97]. Consequently, it received fast track FDA approval for treatment of NSCLC patients harbouring KRAS-G12C mutations. Interestingly, these patients have a high response rate to ICBs compared with NSCLC patients with other mutations, such as EGFR [97]. Currently, sotorasib either alone or in combination with chemotherapy and ICB is under clinical trial (NCT04625647, NCT04185883) and could prove to be highly beneficial in inducing antitumor immunity in NSCLC patients.

5. Potential Application SMIs against DNA Damage Regulatory Proteins

Conventional chemotherapies and DNA damage response inhibitors (DDRi) both increase the load of DNA damage in tumor cells triggering an innate immune response, but also promote immunosuppressive signals [98,99]. However, conventional chemotherapies, through their less targeted approach, also kill immune cells and thus are poor candidates to combine with immunotherapies. Therefore, it is proposed that DDRis have fewer healthy tissue toxicities as they target tumor-specific defects and thus represent better candidates for combination with immunotherapies. The prototypic tumor targeted DDRis are the poly ADP ribosyl polymerase (PARP) inhibitors (PARPi), specially Olaparib which is currently under clinical trial in combination with ICB (Table 2). These drugs were identified as synthetic lethal interactors initially with BReast CAncer gene 2 (BRCA2) mutations, but since have been shown to have similar synthetic lethal interaction with any mutation that results in defective homologous recombination repair (HRR) [100]. However, clinical experience suggests that germline or somatic mutations of only a subset of HRR genes including BRCA1/2 and Partner and localizer of BRCA2 (PALB2) confer sensitivity to PARPi, (olaparib) in patients [101]. One of the outcomes of olaparib treatment is increased DNA damage which triggers an innate immune response, commonly through the cGAS-STING pathway (cyclic-GMP-AMP synthase cGAS—Stimulator of Interferon genes) [102]. This can produce improved immune recognition that is further enhanced with ICB, although the effect appears to be independent of the functional status of HRR [103,104,105]. This innate immune response can be triggered by any agent that promotes DNA damage and can utilise either the canonical cGAS-STING or non-canonical pathways [106,107,108]. However, DNA damage can also trigger immunosuppressive responses such as upregulation of PD-L1 [109,110].
Ataxia telangiectasia and Rad3-related protein (ATR) and Checkpoint Kinase 1 (CHK1) are components of the cellular response to replication stress [111,112]. Although SMIs targeting ATR and CHK1 (M6620 (VX-970) and SRA737) have limited activity in patients as single agents [113,114], they have been shown to trigger innate immune signalling and can be combined with ICB to enhance anti-tumor immune responses in preclinical models and recently in clinical trials [115,116,117]. This may be a consequence of the ability of ATRi to block the DNA damage-induced expression of PD-L1 [109,110]. ATRis (AZD6738 and CHK1i (GDC-0575) also synergise with drugs that promote replication stress such as gemcitabine and cisplatin. However, when combined with standard doses of these drugs, they were associated with high levels of severe adverse haematological responses limiting their ability to be used with immunotherapies [111,118,119]. It is possible to avoid these adverse responses by using subclinical doses of replication stress promoting drugs such as gemcitabine or hydroxyurea in combination with CHK1 inhibitor [115,120]. Unfortunately, ATRis are ineffective in combination with subclinical levels of hydroxyurea or gemcitabine. Conversely, the combinations of CHK1i and subclinical dose of hydroxyurea or gemcitabine not only had little normal tissue toxicity [121,122]. By using subclinical doses of replication stress promoters’ gemcitabine and hy-droxyurea in combination of CHK1i and hydroxyurea [115,120,121]. Although, ATRi shows to have little normal tissue toxicity even in normally chemo-sensitive tissue such as immune cells, subclinical dosages are ineffective in these combinations. In preclinical models of melanoma and small cell lung cancer, combination of low dose hydroxyurea or gemcitabine with CHK1i trigger proinflam-matory cytokine and chemokine expression and enhance both innate and adaptive immune cell tumor infiltration and anti-tumor responses [115,120]. The immune responses triggered by these combinations differed depending on the cancer type, and ICB enhanced the immune response only in the small cell lung cancer models suggesting the immunosuppressive pathway differed between cancer types.

6. Use of SMIs Which Induce Epigenetic Changes

Epigenetic alterations contribute to carcinogenesis and significantly influence T and NK cell activation, differentiation, and function [123]. Therefore, strategic repurposing of epigenetically targeted drugs to boost immune cell function whilst suppressing pro-oncogenic signals could enhance clinical response to ICB mAbs [123]. Drugs targeting epigenetic alteration inhibit DNA methyltransferases (DNMTs), DNA demethylases, histone methyltransferases (HMTs), histone demethylases (HDMs) and other relevant enzymes involved in gene expression modulation [124,125,126,127,128]. In a murine B16-gp33 model, HDACi MS-275 induced NOS2 (Nitric Oxide Synthase 2)/Reactive Oxygen species (ROS) secretion and activated pro-inflammatory gene signatures which reduced the immunosuppressive function of tumor-infiltrating myeloid cells, by inducing their cell death in an IFN-γR/STAT1 signalling dependent manner [129].
Selective inhibition of Enhancer of Zeste Homolog 2 (EZH2), using CPI-1205, in a murine MC-38 cancer model disrupted the immunosuppressive function of tumor infiltrating Tregs, skewing their response towards a more pro-inflammatory phenotype. Effector CD4+ and CD8+ T cell numbers increased within the TME leading to tumor elimination [130]. Ghosh et al. [131] demonstrated that chemical inhibition of Cyclic adenosine monophosphate response element Binding Protein (CBP/EP300) bromodomain, using a series of laboratory synthesised inhibitors, led to the blockage of Treg immunosuppressive function due to reduced FOXP3 acetylation which resulted in its degradation.
Bromodomain (BRD) and extra-terminal motif (BET) proteins inhibitors (BETi) have been shown to regulate the presentation and generation of neo-antigens, expression of immune checkpoints molecules, secretion of cytokines, and the activation of immune cells in several murine and human cancer settings [132]. Mechanistically, the BET family (BRD2, BRD3, BRD4, and BRDT) transcriptionally controls a range of proinflammatory and immunoregulatory genes by recognizing acetylated histones (mainly H3 and H4) and recreating necessary transcription factors and promote phosphorylation of RNA polymerase to the chromatin site [133]. BRD4 restores anti-tumor immune responses following chemical inhibition with small-molecule bromodomain inhibitor JQ1, by down regulating PD-L1 expression in a MYC dependent manner in multiple myeloma [134]. To date, JQ1 has been shown to downregulate the BRD4-MYC axis across several epithelial cancers, in preclinical and clinical studies [135]. Downregulation of the BRD4-MYC transcription axis using JQ1 resulted in boosting of stem cell–like and central memory CD8+ T cells responses that enhanced antitumor immunity in mouse models of epithelial ovarian cancer [136]. Similarly, BRD4 inhibition led to expression of proinflammatory genes such as Baculoviral IAP Repeat Containing 2 & 3 (BIRC2 and BIRC3), which in turn led to tumor necrosis factor (TNF) production triggering apoptosis in preclinical colon cancer models, boosting anti-tumour immunity [137]. Thus, combination of these drugs with ICB mAbs could prove effective against aggressive solid tumors, although optimisation of drug combinations in animal studies will be required.

7. SMIs Paving Way for Cytotoxic Lymphocytes to Transform into Super Killers

SMIs have been developed which inhibit immune suppressive mechanisms whilst activating innate and/or adaptive immune cell pathways. These chemical therapies have advantages compared to biological therapies (antibody and cell therapies) such as lower manufacturing and administrative costs. A relevant example are two small molecule inhibitors from Curis biopharmaceutical, phase-I trial CA-170 (antagonizes VISTA and PD-L1) (NCT02812875) and CA-327 (antagonizes TIM-3 and PD-L1) [138]. In contrast to ICB mAbs, these drugs can simultaneously antagonize multiple immune checkpoint receptors, increasing their potential to prevent tumor immune escape [139].
Toll-like receptors (TLRs) trigger innate immune responses by recognising pathogen-associated antigens. TLR agonists, and particularly TLR7/8 agonists, are potential immuno-oncologic therapeutic targets [139]. Imiquimod and derivative imidazoquinolines (resiquimod, 852A, 852A and VTX-2337) have been developed for systemic delivery and are currently under clinical trial [140,141,142,143]. These TLR agonists synergise with interferons (type I or II) and induce reprogramming of M2 immune-suppressive macrophages into M1 proinflammatory type [144,145,146]. TLR5 agonist entolimod induced NK-cell-dependent activation of DCs which resulted in stimulation of CD8+ T cells, triggering durable memory against aggressive colon and mammary metastatic mouse models [147].
N-formyl-kynurenine is a potent endogenous inhibitor of T cell activation produced by catabolism of tryptophan by heme-containing dioxygenase enzyme called IDO (indoleamine 2,3-dioxygenase) and helps tumor cells to evade immunosurveillance. Kynurenine metabolic pathway upregulation results in downregulation of tryptophan uptake as a consequence of which effector T cells function is reduced. Tryptophan is critical for TCR activation and hence is important in promoting antigen recognition. However, absence of tryptophan promotes Treg function by activating aryl hydrocarbon receptor activation enabling tumor evasion. IDO is thus an important target in immune-oncology. IDO inhibitors (e.g., epacadostat) reduce tumor growth and promote the proliferation of CD8+ T cells and NK cells in human peripheral blood mononuclear cells (PBMCs) ex vivo and are currently under clinical trial (Table 2) [148].
Adenosine triphosphate (ATP) catabolism mediates immunosuppression, through inducing expression of CD39 and CD73, which regulate growth and metastasis of tumor cells. Tumor cells dephosphorylate ATP with the help of CD39 and CD73 to produce adenosine, which interacts with adenosine receptors A2aR and A2bR on cytotoxic lymphocytes and suppresses cytolysis [149]. Free ATP molecules are recognised as “danger” signals by the immune system and are known to activate the nucleotide-binding oligomerization domain (NLRP3) inflammasome in DCs and induce IL1-β, promoting an inflammatory response in cancer. SMIs against CD39 (ARL6715), CD73 (AMPCP) and adenosine receptors (CPI-444 inhibiting A2AR) have been shown to promote robust cytotoxic CD8+ T cell responses [150,151,152]. Collectively, these studies highlight the potential of small molecule-based immune therapies to “super activate” or prevent immune exhaustion which in turn enhances tumor killing.

8. Utilising SMIs to Induce Immunogenic Cell Death

Chronic exposure of damage-associated molecular patterns (DAMPs) in the TME can activate or suppress key multiple cellular pathways among cancer cells such as Caspase 3 or PIK3CA which results in immunogenic cell death (ICD) by necroptosis, ferroptosis or pyroptosis [153,154]. The release of DAMPs can be observed upon exposure to chemotherapeutic drugs, on-colytic viruses, physicochemical therapies, photodynamic therapy, and radiotherapy. An adaptive immune response can thus be triggered, initiating effector cytotoxic T cell function, and eliciting immunological memory by exposing [155]. When cells undergo ICD, there is a characteristic release of adenosine triphosphate (ATP) and high mobility group box 1 (HMGB1)) that leads to the activation of type I IFN responses and release of pro-inflammatory chemokines/cytokines (i.e., IL-1 and IL-18) [156,157,158]. As a result, immune cells can be recruited to the tumor, including cross-primed CD8+ T cells, due to availability of rich source of immunogens [159,160].
When cancer cells undergo cell death by necrosis, tumor cell DNA is released and detected by cGAS in APCs. cGAS is responsible for the production of Cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) which binds to and activated STING [161]. Consequently, it activates ICD through activation of NF-κB (nuclear factor kappa B) pathway. This pathway results in production of IFNs and pro-inflammatory cytokines, promoting recruitment and activation of T cells [107]. In a preclinical pancreatic mouse model, STING agonist DMXAA reshaped the archi-tecture of the TME enabling more infiltration of activated cytotoxic T cells while re-ducing Tregs numbers. Additionally, DMXAA induced high expression of costimulatory molecules in cross-presenting DCs which resulted in reprogramming M2 into M1 macrophages [162]. Upregulation of anti-apoptotic proteins such as B-cell lymphoma-2 (BCL-2) and its homologues Bcl-xL and Bcl-w protect against tumour cell apoptosis by inhibiting mi-tochondrial outer membrane permeabilization. Navitoclax, a BCL-2 inhibitor, has been shown to reduce immune suppression, and proliferation and survival of cancer associ-ated fibroblasts (CAFs). CAF facilitate downregulation of ICD by suppressing release of ATP and HMGB-1 when exposed to radiation or chemotherapy which can induce resistance to ICD [163].In addition, CAFs restrict CD8+ T-cell infiltration which imposes immunologically cold TME leading to insensitivity towards ICB treatment in syngeneic breast cancer mouse model [164]. Hence, eliminating CAF population using navitoclax could potentially boost the efficacy of ICB.
To date, multiple chemo-drugs such as doxorubicin, mitoxantrone, oxaliplatin, and bortezomib, have been demonstrated to effectively induce tumor cell death. Artemisinin (ART)—a clinically approved anti-malarial drug—has been shown to have cytotoxic properties against tumor cells resulting in immune mediated cell death [165,166]. In an ex vivo experiment using an endometrial carcinoma cell line, ART upregulated the expression of immunosuppressive molecules such as CD155 (expressed on tumor cells) whilst downregulating TIGIT on NK cells which overall enhanced cytotoxicity when tumor and NK cells were cocultured [167].
Azacytidine and romidepsin (FDA-approved drugs) in combination with IFNα2 (ARI) have been shown to induce ICD in colorectal cancers cells in vitro, which in turn resulted in DCs stimulation due to upregulation of IFN. Increased DCs trafficking facilitated T cell cross-priming in tumor draining lymph nodes in a syngeneic colon cancer mouse model [168,169]. In a recent study, Zhang et al. [170] compared 4 SMIs (bortezomib and obatoclax mesylate vs. BI 2536 (BI) and (S)-(+)-camptothecin (CPT)). They demonstrated that BI and CPT triggered immune mediated cell death (pyroptosis) in syngeneic colon cancer mouse model leading to a greater CD8+ T cell accumulation at the tumor site compared to bortezomib or obatoclax mesylate which, conversely, did not induce ICD. Apurinic/apyrimidinic endonuclease 1 (APE1) inhibitor NO.0449-0145 has been shown to induce ICD in NSCLC preclinical models justifying investigation of the efficacy of this inhibitor in boosting anti-tumour immunity [171]. Cancer therapies that induce ICD should demonstrate enhanced effectiveness when combined with ICB mAbs, and it is likely that further promising combinatory therapies will be developed soon.

9. Future Perspective

The clinical efficacy and durability of ICB-based cancer immunotherapy has revolutionised the way solid tumors are treated and managed over the last decade. However, increasing the efficacy of ICB in a broader patient cohort continues to be challenging. With advancing technology, it is now understood that inherent oncogenic properties of cancer cells dictate the TME to alter the immune architecture. Targeting oncogenic signalling—especially the RTK signalling cascade—with SMIs in combination with ICB mAbs has gained traction and a wide range of RTKi are currently in phase III/IV clinical trials (currently recruiting or published). Several of these have shown significantly better clinical efficacy compared to ICB alone (Table 1 and Table 2). However, over the last decade, other SMIs known to inhibit oncogenic properties such as deregulated cell cycle, abnormal DNA damage regulation, epigenetic aberrations, metabolic abnormality, upregulated immune evasion molecules and suppressors of cell death mechanism have been found to modulate anti-tumor immunity in preclinical syngeneic epithelial cancer models (Figure 1). The major areas of research focus should be to: (i) elaborate on tissue-specific oncogene-related immune effects; (ii) delineate and functionally validate biomarkers which can predict response and resistance to oncogene targeting; (iii) generate and characterise highly dependable animal models to mimic human immune response to tumors (i.e., humanised mouse models) and (iv) develop multiplexed assays to incorporate immune and tumor intrinsic molecular changes in response to combination therapy. In addition, a comprehensive understanding of the TME architecture with spatial orientation of immune cells and their interaction with the cancer cells should be carefully examined for selecting the most ideal drugs for combination therapy of cancer.
Intermittent dosing of these SMIs which would accommodate treatment-free interval for the administration of ICB mAbs to potentiate high antigen expression and cytotoxic T cell infiltration is currently needed to avoid drug resistance and maximize treatment efficacy. As such optimization of these potential combinations should be investigated both at preclinical and clinical level including designing appropriate treatment schedules to attain enhanced anticancer immunosurveillance with minimum risk of toxicities. Using SMIs to induce an immunomodulatory effect in conjunction with making the hostile TME favourable to an anti-tumor response, provides a strong rationale for their combination with ICB mAbs. Combination therapy has the potential to synergistically inhibit malignant cells alongside augmenting the immune recognition and elimination of the tumor. Furthermore, with the ability to induce long-term antitumor memory, combination therapy may lead to greater rates of cure. Target therapies in combination with ICB mAbs might prove to be “game-changing” for patients with aggressive disease in the near future.

Author Contributions

D.S., P.M. and B.G.—bibliographic research, draft of the manuscript. D.S., B.G. and J.L.G.C.—conceptualization. X.L., P.M., Q.W., B.G., I.H.F. and J.L.G.C.—reviewing and editing of final drafts. J.L.G.C.—funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Garnet Passe and Rodney Williams Memorial Foundation Co-joint grant (2019_CG_Gonzalez Cruz_Perry), Princess Alexandra Research Foundation 2021 Research Award and NHMRC Investigator grant (Frazer_2020).

Conflicts of Interest

The funders had no role in the writing of the manuscript.

References

  1. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef]
  2. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Huo, J.-L.; Wang, Y.-T.; Fu, W.-J.; Lu, N.; Liu, Z.-S. The promising immune checkpoint LAG-3 in cancer immunotherapy: From basic research to clinical application. Front. Immunol. 2022, 13, 956090. [Google Scholar] [CrossRef] [PubMed]
  5. Chauvin, J.-M.; Zarour, H.M. TIGIT in cancer immunotherapy. J. Immunother. Cancer 2020, 8, e000957. [Google Scholar] [CrossRef] [PubMed]
  6. Chocarro, L.; Bocanegra, A.; Blanco, E.; Fernández-Rubio, L.; Arasanz, H.; Echaide, M.; Garnica, M.; Ramos, P.; Piñeiro-Hermida, S.; Vera, R.; et al. Cutting-Edge: Preclinical and Clinical Development of the First Approved Lag-3 Inhibitor. Cells 2022, 11, 2351. [Google Scholar] [CrossRef]
  7. Cho, B.C.; Abreu, D.R.; Hussein, M.; Cobo, M.; Patel, A.J.; Secen, N.; Lee, K.H.; Massuti, B.; Hiret, S.; Yang, J.C.H.; et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): Primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022, 23, 781–792. [Google Scholar] [CrossRef]
  8. Kuang, Z.; Li, L.; Zhang, P.; Chen, B.; Wu, M.; Ni, H.; Yi, S.; Zou, J.; Liu, J. A novel antibody targeting TIM-3 resulting in receptor internalization for cancer immunotherapy. Antib. Ther. 2020, 3, 227–236. [Google Scholar] [CrossRef]
  9. Kuo, T.C.; Chen, A.; Harrabi, O.; Sockolosky, J.T.; Zhang, A.; Sangalang, E.; Doyle, L.V.; Kauder, S.E.; Fontaine, D.; Bollini, S.; et al. Targeting the myeloid checkpoint receptor SIRPα potentiates innate and adaptive immune responses to promote anti-tumor activity. J. Hematol. Oncol. 2020, 13, 160. [Google Scholar] [CrossRef]
  10. Mehta, N.; Maddineni, S.; Kelly, R.L.; Lee, R.B.; Hunter, S.A.; Silberstein, J.L.; Sperberg, R.A.P.; Miller, C.L.; Rabe, A.; Labanieh, L.; et al. An engineered antibody binds a distinct epitope and is a potent inhibitor of murine and human VISTA. Sci. Rep. 2020, 10, 15171. [Google Scholar] [CrossRef]
  11. Li, J.; Yang, Y.; Inoue, H.; Mori, M.; Akiyoshi, T. The expression of costimulatory molecules CD80 and CD86 in human carcinoma cell lines: Its regulation by interferon γ and interleukin-10. Cancer Immunol. Immunother. 1996, 43, 213–219. [Google Scholar] [CrossRef] [PubMed]
  12. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; Van den Eertwegh, A.J.M.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H.; et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Beer, T.M.; Kwon, E.D.; Drake, C.G.; Fizazi, K.; Logothetis, C.; Gravis, G.; Ganju, V.; Polikoff, J.; Saad, F.; Humanski, P.; et al. Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2017, 35, 40–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Bristol-Myers Squibb Announces Phase 3 CheckMate-498 Study Did Not Meet Primary Endpoint of Overall Survival with Opdivo (nivolumab) Plus Radiation in Patients with Newly Diagnosed MGMT-Unmethylated Glioblastoma Multiforme. Available online: https://news.bms.com/news/corporate-financial/2019/Bristol-Myers-Squibb-Announces-Phase-3-CheckMate--498-Study-Did-Not-Meet-Primary-Endpoint-of-Overall-Survival-with-Opdivo-nivolumab-Plus-Radiation-in-Patients-with-Newly-Diagnosed-MGMT-Unmethylated-Glioblastoma-Multiforme/default.aspx (accessed on 5 September 2019).
  16. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  17. Popat, S.; Curioni-Fontecedro, A.; Dafni, U.; Shah, R.; O’Brien, M.; Pope, A.; Fisher, P.; Spicer, J.; Roy, A.; Gilligan, D.; et al. A multicentre randomised phase III trial comparing pembrolizumab versus single-agent chemotherapy for advanced pre-treated malignant pleural mesothelioma: The European Thoracic Oncology Platform (ETOP 9-15) PROMISE-meso trial. Ann. Oncol. 2020, 31, 1734–1745. [Google Scholar] [CrossRef]
  18. Winer, E.P.; Lipatov, O.; Im, S.-A.; Goncalves, A.; Muñoz-Couselo, E.; Lee, K.S.; Schmid, P.; Tamura, K.; Testa, L.; Witzel, I.; et al. Pembrolizumab versus investigator-choice chemotherapy for metastatic triple-negative breast cancer (KEYNOTE-119): A randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 499–511. [Google Scholar] [CrossRef]
  19. Shitara, K.; Özgüroğlu, M.; Bang, Y.-J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.-H.; Fornaro, L.; Olesiński, T.; Caglevic, C.; Chung, H.C.; et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. Lancet 2018, 392, 123–133. [Google Scholar] [CrossRef]
  20. Shitara, K.; Van Cutsem, E.; Bang, Y.-J.; Fuchs, C.; Wyrwicz, L.; Lee, K.-W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; et al. Efficacy and Safety of Pembrolizumab or Pembrolizumab Plus Chemotherapy vs Chemotherapy Alone for Patients With First-line, Advanced Gastric Cancer: The KEYNOTE-062 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1571–1580. [Google Scholar] [CrossRef]
  21. Powles, T.; Csőszi, T.; Özgüroğlu, M.; Matsubara, N.; Géczi, L.; Cheng, S.Y.-S.; Fradet, Y.; Oudard, S.; Vulsteke, C.; Barrera, R.M.; et al. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): A randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 931–945. [Google Scholar] [CrossRef]
  22. 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. [Google Scholar] [CrossRef] [PubMed]
  23. Update on KESTREL Phase III Trial of Imfinzi with or without Tremelimumab in the 1st-Line Treatment of Recurrent or Metastatic Head and Neck Cancer. Available online: https://www.astrazeneca.com/media-centre/press-releases/2021/update-on-kestrel-phase-iii-trial-for-imfinzi.html (accessed on 5 February 2021).
  24. Lee, N.Y.; Ferris, R.L.; Psyrri, A.; I Haddad, R.; Tahara, M.; Bourhis, J.; Harrington, K.; Chang, P.M.-H.; Lin, J.-C.; Razaq, M.A.; et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: A randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 2021, 22, 450–462. [Google Scholar] [CrossRef] [PubMed]
  25. On the Phase III NEPTUNE Trial of Imfinzi Plus Tremelimumab in Stage IV Non-Small Cell Lung Cancer. Available online: https://www.astrazeneca.com/media-centre/press-releases/2019/update-on-the-phase-iii-neptune-trial-of-imfinzi-plus-tremelimumab-in-stage-iv-non-small-cell-lung-cancer-21082019.html#! (accessed on 21 August 2019).
  26. Owonikoko, T.K.; Park, K.; Govindan, R.; Ready, N.; Reck, M.; Peters, S.; Dakhil, S.R.; Navarro, A.; Rodríguez-Cid, J.; Schenker, M.; et al. Nivolumab and Ipilimumab as Maintenance Therapy in Extensive-Disease Small-Cell Lung Cancer: CheckMate 451. J. Clin. Oncol. 2021, 39, 1349–1359. [Google Scholar] [CrossRef]
  27. Phase 3 Trial in Squamous Non Small Cell Lung Cancer Subjects Comparing Ipilimumab Plus Paclitaxel and Carboplatin Versus Placebo Plus Paclitaxel and Carboplatin. Available online: https://clinicaltrials.gov/ct2/show/results/NCT02279732 (accessed on 28 August 2019).
  28. Govindan, R.; Szczesna, A.; Ahn, M.-J.; Schneider, C.-P.; Mella, P.F.G.; Barlesi, F.; Han, B.; Ganea, D.E.; Von Pawel, J.; Vladimirov, V.; et al. Phase III Trial of Ipilimumab Combined With Paclitaxel and Carboplatin in Advanced Squamous Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2017, 35, 3449–3457. [Google Scholar] [CrossRef] [PubMed]
  29. Reck, M.; Luft, A.; Szczesna, A.; Havel, L.; Kim, S.-W.; Akerley, W.; Pietanza, M.C.; Wu, Y.-L.; Zielinski, C.; Thomas, M.; et al. Phase III Randomized Trial of Ipilimumab Plus Etoposide and Platinum versus Placebo Plus Etoposide and Platinum in Extensive-Stage Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 3740–3748. [Google Scholar] [CrossRef]
  30. de Miguel, M.; Calvo, E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell 2020, 38, 326–333. [Google Scholar] [CrossRef]
  31. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [Green Version]
  32. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  33. Pitt, J.; Marabelle, A.; Eggermont, A.; Soria, J.-C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef]
  34. Hunter, M.C.; Teijeira, A.; Halin, C. T Cell Trafficking through Lymphatic Vessels. Front. Immunol. 2016, 7, 613. [Google Scholar] [CrossRef]
  35. Chow, K.T.; Gale, M. SnapShot: Interferon Signaling. Cell 2015, 163, 1808–1808.e1. [Google Scholar] [CrossRef] [PubMed]
  36. Rooney, M.S.; Shukla, S.A.; Wu, C.J.; Getz, G.; Hacohen, N. Molecular and Genetic Properties of Tumors Associated with Local Immune Cytolytic Activity. Cell 2015, 160, 48–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef] [PubMed]
  38. Giannakis, M.; Mu, X.J.; Shukla, S.A.; Qian, Z.R.; Cohen, O.; Nishihara, R.; Bahl, S.; Cao, Y.; Amin-Mansour, A.; Yamauchi, M.; et al. Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma. Cell Rep. 2016, 17, 1206. [Google Scholar] [CrossRef] [PubMed]
  39. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [Green Version]
  40. Riaz, N.; Morris, L.; Havel, J.J.; Makarov, V.; Desrichard, A.; Chan, T.A. The role of neoantigens in response to immune checkpoint blockade. Int. Immunol. 2016, 28, 411–419. [Google Scholar] [CrossRef] [Green Version]
  41. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The Three Es of Cancer Immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef]
  42. Huang, R.-Y.; Francois, A.; McGray, A.R.; 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 2016, 6, e1249561. [Google Scholar] [CrossRef] [Green Version]
  43. Wang, M.; Liu, Y.; Cheng, Y.; Wei, Y.; Wei, X. Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim. Et Biophys. Acta (BBA)—Rev. Cancer 2018, 1871, 199–224. [Google Scholar] [CrossRef]
  44. Kwilas, A.R.; Donahue, R.N.; Tsang, K.Y.; Hodge, J.W. Immune consequences of tyrosine kinase inhibitors that synergize with cancer immunotherapy. Cancer Cell Microenviron. 2015, 2, e677. [Google Scholar] [CrossRef]
  45. Sinha, D.; Smith, C.; Khanna, R. Joining Forces: Improving Clinical Response to Cellular Immunotherapies with Small-Molecule Inhibitors. Trends Mol. Med. 2020, 27, 75–90. [Google Scholar] [CrossRef] [PubMed]
  46. Pembrolizumab in Combination with Epacadostat or Placebo in Cisplatin-ineligible Urothelial Carcinoma (KEYNOTE-672/ECHO-307). Available online: https://clinicaltrials.gov/ct2/show/results/NCT03361865 (accessed on 16 December 2021).
  47. Long, G.V.; Dummer, R.; Hamid, O.; Gajewski, T.F.; Caglevic, C.; Dalle, S.; Arance, A.; Carlino, M.S.; Grob, J.-J.; Kim, T.M.; et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): A phase 3, randomised, double-blind study. Lancet Oncol. 2019, 20, 1083–1097. [Google Scholar] [CrossRef] [PubMed]
  48. Efficacy and Safety Study of Pembrolizumab (MK-3475) with or without Lenvatinib (MK-7902/E7080) in Adults with Programmed Cell Death-Ligand 1 (PD-L1)-Positive Treatment-naïve Nonsmall Cell Lung Cancer (NSCLC) (MK-7902-007/E7080-G000-314/LEAP-007). Available online: https://clinicaltrials.gov/ct2/show/results/NCT03829332 (accessed on 28 October 2022).
  49. Makker, V.; Colombo, N.; Herráez, A.C.; Santin, A.D.; Colomba, E.; Miller, D.S.; Fujiwara, K.; Pignata, S.; Baron-Hay, S.; Ray-Coquard, I.; et al. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. N. Engl. J. Med. 2022, 386, 437–448. [Google Scholar] [CrossRef]
  50. Rini, B.I.; Plimack, E.R.; Stus, V.; Gafanov, R.; Hawkins, R.; Nosov, D.; Pouliot, F.; Alekseev, B.; Soulières, D.; Melichar, B.; et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1116–1127. [Google Scholar] [CrossRef]
  51. Powles, T.; Plimack, E.R.; Soulières, D.; Waddell, T.; Stus, V.; Gafanov, R.; Nosov, D.; Pouliot, F.; Melichar, B.; Vynnychenko, I.; et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): Extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020, 21, 1563–1573. [Google Scholar] [CrossRef] [PubMed]
  52. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115. [Google Scholar] [CrossRef] [PubMed]
  53. Eng, C.; Kim, T.W.; Bendell, J.; Argilés, G.; Tebbutt, N.C.; Di Bartolomeo, M.; Falcone, A.; Fakih, M.; Kozloff, M.; Segal, N.H.; et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019, 20, 849–861. [Google Scholar] [CrossRef]
  54. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Oyervides Juárez, V.M.; Hsieh, J.J.; Basso, U.; Shah, A.Y.; et al. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2021, 384, 829–841. [Google Scholar] [CrossRef]
  55. LBA76—Overall Survival (OS) Results from the Phase III TROPiCS-02 Study of Sacituzumab Govitecan (SG) vs. Treatment of Physician’s Choice (TPC) in Patients (pts) with HR+/HER2-Metastatic Breast Cancer (mBC). Available online: https://oncologypro.esmo.org/meeting-resources/esmo-congress/overall-survival-os-results-from-the-phase-iii-tropics-02-study-of-sacituzumab-govitecan-sg-vs-treatment-of-physician-s-choice-tpc-in-patient (accessed on 9 September 2022).
  56. Merck and Eisai Provide Update on Phase 3 LEAP-002 Trial Evaluating KEYTRUDA® (pembrolizumab) Plus LENVIMA® (lenvatinib) versus LENVIMA Monotherapy in Patients with Unresectable Hepatocellular Carcinoma. Available online: https://www.merck.com/news/merck-and-eisai-provide-update-on-phase-3-leap-002-trial-evaluating-keytruda-pembrolizumab-plus-lenvima-lenvatinib-versus-lenvima-monotherapy-in-patients-with-unresectable-hepatocellul/ (accessed on 3 August 2022).
  57. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef] [Green Version]
  58. Qu, T.; Li, B.; Wang, Y. Targeting CD47/SIRPα as a therapeutic strategy, where we are and where we are headed. Biomark. Res. 2022, 10, 20. [Google Scholar] [CrossRef]
  59. Zhao, H.; Song, S.; Ma, J.; Yan, Z.; Xie, H.; Feng, Y.; Che, S. CD47 as a promising therapeutic target in oncology. Front. Immunol. 2022, 13, 757480. [Google Scholar] [CrossRef] [PubMed]
  60. Yu, W.-B.; Ye, Z.-H.; Chen, X.; Shi, J.-J.; Lu, J.-J. The development of small-molecule inhibitors targeting CD47. Drug Discov. Today 2020, 26, 561–568. [Google Scholar] [CrossRef] [PubMed]
  61. Cabrales, P. RRx-001 Acts as a Dual Small Molecule Checkpoint Inhibitor by Downregulating CD47 on Cancer Cells and SIRP-α on Monocytes/Macrophages. Transl. Oncol. 2019, 12, 626–632. [Google Scholar] [CrossRef] [PubMed]
  62. Oronsky, B.; Reid, T.R.; Larson, C.; Caroen, S.; Quinn, M.; Burbano, E.; Varner, G.; Thilagar, B.; Brown, B.; Coyle, A.; et al. REPLATINUM Phase III randomized study: RRx-001 + platinum doublet versus platinum doublet in third-line small cell lung cancer. Futur. Oncol. 2019, 15, 3427–3433. [Google Scholar] [CrossRef]
  63. Tomita, Y.; Oronsky, B.; Abrouk, N.; Cabrales, P.; Reid, T.R.; Lee, M.-J.; Yuno, A.; Baker, J.; Lee, S.; Trepel, J.B. In small cell lung cancer patients treated with RRx-001, a downregulator of CD47, decreased expression of PD-L1 on circulating tumor cells significantly correlates with clinical benefit. Transl. Lung Cancer Res. 2021, 10, 274–278. [Google Scholar] [CrossRef]
  64. Smith, W.M.; Purvis, I.J.; Bomstad, C.N.; Labak, C.M.; Velpula, K.K.; Tsung, A.J.; Regan, J.N.; Venkataraman, S.; Vibhakar, R.; Asuthkar, S. Therapeutic targeting of immune checkpoints with small molecule inhibitors. Am. J. Transl. Res. 2019, 11, 529–541. [Google Scholar]
  65. Shaw, G.; Cavalcante, L.; Giles, F.J.; Taylor, A. Elraglusib (9-ING-41), a selective small-molecule inhibitor of glycogen synthase kinase-3 beta, reduces expression of immune checkpoint molecules PD-1, TIGIT and LAG-3 and enhances CD8+ T cell cytolytic killing of melanoma cells. J. Hematol. Oncol. 2022, 15, 1–13. [Google Scholar] [CrossRef]
  66. Rudd, C.E.; Chanthong, K.; Taylor, A. Small Molecule Inhibition of GSK-3 Specifically Inhibits the Transcription of Inhibitory Co-receptor LAG-3 for Enhanced Anti-tumor Immunity. Cell Rep. 2020, 30, 2075–2082.e4. [Google Scholar] [CrossRef] [Green Version]
  67. Johnston, R.J.; Comps-Agrar, L.; Hackney, J.; Yu, X.; Huseni, M.; Yang, Y.; Park, S.; Javinal, V.; Chiu, H.; Irving, B.; et al. The Immunoreceptor TIGIT Regulates Antitumor and Antiviral CD8+ T Cell Effector Function. Cancer Cell 2014, 26, 923–937. [Google Scholar] [CrossRef] [Green Version]
  68. Chauvin, J.-M.; Pagliano, O.; Fourcade, J.; Sun, Z.; Wang, H.; Sander, C.; Kirkwood, J.M.; Chen, T.-H.T.; Maurer, M.; Korman, A.J.; et al. TIGIT and PD-1 impair tumor antigen–specific CD8+ T cells in melanoma patients. J. Clin. Investig. 2015, 125, 2046–2058. [Google Scholar] [CrossRef]
  69. Zhou, X.; Jiao, L.; Qian, Y.; Dong, Q.; Sun, Y.; Zheng, W.; Zhao, W.; Zhai, W.; Qiu, L.; Wu, Y.; et al. Repositioning Azelnidipine as a Dual Inhibitor Targeting CD47/SIRPα and TIGIT/PVR Pathways for Cancer Immuno-Therapy. Biomolecules 2021, 11, 706. [Google Scholar] [CrossRef]
  70. Zhou, X.; Du, J.; Wang, H.; Chen, C.; Jiao, L.; Cheng, X.; Zhou, X.; Chen, S.; Gou, S.; Zhao, W.; et al. Repositioning liothyronine for cancer immunotherapy by blocking the interaction of immune checkpoint TIGIT/PVR. Cell Commun. Signal. 2020, 18, 142. [Google Scholar] [CrossRef]
  71. Lee, S.; Rauch, J.; Kolch, W. Targeting MAPK Signaling in Cancer: Mechanisms of Drug Resistance and Sensitivity. Int. J. Mol. Sci. 2020, 21, 1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bedognetti, D.; Roelands, J.; Decock, J.; Wang, E.; Hendrickx, W. The MAPK hypothesis: Immune-regulatory effects of MAPK-pathway genetic dysregulations and implications for breast cancer immunotherapy. Emerg. Top. Life Sci. 2017, 1, 429–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Cheng, Y.; Tian, H. Current Development Status of MEK Inhibitors. Molecules 2017, 22, 1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kono, M.; Dunn, I.S.; Durda, P.J.; Butera, D.; Rose, L.B.; Haggerty, T.J.; Benson, E.M.; Kurnick, J.T. Role of the Mitogen-Activated Protein Kinase Signaling Pathway in the Regulation of Human Melanocytic Antigen Expression. Mol. Cancer Res. 2006, 4, 779–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Boni, A.; Cogdill, A.P.; Dang, P.; Udayakumar, D.; Njauw, C.-N.J.; Sloss, C.M.; Ferrone, C.R.; Flaherty, K.T.; Lawrence, D.P.; Fisher, D.E.; et al. Selective BRAFV600E Inhibition Enhances T-Cell Recognition of Melanoma without Affecting Lymphocyte Function. Cancer Res 2010, 70, 5213–5219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liu, C.; Peng, W.; Xu, C.; Lou, Y.; Zhang, M.; Wargo, J.A.; Chen, J.Q.; Li, H.S.; Watowich, S.S.; Yang, Y.; et al. BRAF Inhibition Increases Tumor Infiltration by T cells and Enhances the Antitumor Activity of Adoptive Immunotherapy in Mice. Clin. Cancer Res. 2013, 19, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Kang, S.-H.; Keam, B.; Ahn, Y.-O.; Park, H.-R.; Kim, M.; Kim, T.M.; Kim, D.-W.; Heo, D.S. Inhibition of MEK with trametinib enhances the efficacy of anti-PD-L1 inhibitor by regulating anti-tumor immunity in head and neck squamous cell carcinoma. Oncoimmunology 2018, 8, e1515057. [Google Scholar] [CrossRef] [Green Version]
  78. Hu-Lieskovan, S.; Mok, S.; Moreno, B.H.; Tsoi, J.; Robert, L.; Goedert, L.; Pinheiro, E.M.; Koya, R.C.; Graeber, T.G.; Comin-Anduix, B.; et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAFV600E melanoma. Sci. Transl. Med. 2015, 7, 279ra41. [Google Scholar] [CrossRef] [Green Version]
  79. Prasad, M.; Zorea, J.; Jagadeeshan, S.; Shnerb, A.B.; Mathukkada, S.; Bouaoud, J.; Michon, L.; Novoplansky, O.; Badarni, M.; Cohen, L.; et al. MEK1/2 inhibition transiently alters the tumor immune microenvironment to enhance immunotherapy efficacy against head and neck cancer. J. Immunother. Cancer 2022, 10, e003917. [Google Scholar] [CrossRef] [PubMed]
  80. George, M.A.; Qureshi, S.; Omene, C.; Toppmeyer, D.L.; Ganesan, S. Clinical and Pharmacologic Differences of CDK4/6 Inhibitors in Breast Cancer. Front. Oncol. 2021, 11, 693104. [Google Scholar] [CrossRef] [PubMed]
  81. Goel, S.; DeCristo, M.J.; Watt, A.C.; BrinJones, H.; Sceneay, J.; Li, B.B.; Khan, N.; Ubellacker, J.M.; Xie, S.; Metzger-Filho, O.; et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017, 548, 471–475. [Google Scholar] [CrossRef] [Green Version]
  82. Deng, J.; Wang, E.S.; Jenkins, R.W.; Li, S.; Dries, R.; Yates, K.; Chhabra, S.; Huang, W.; Liu, H.; Aref, A.R.; et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018, 8, 216–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhang, J.; Bu, X.; Wang, H.; Zhu, Y.; Geng, Y.; Nihira, N.T.; Tan, Y.; Ci, Y.; Wu, F.; Dai, X.; et al. Cyclin D–CDK4 kinase destabilizes PD-L1 via cullin 3–SPOP to control cancer immune surveillance. Nature 2018, 553, 91–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Schaer, D.A.; Beckmann, R.P.; Dempsey, J.A.; Huber, L.; Forest, A.; Amaladas, N.; Li, Y.; Wang, Y.C.; Rasmussen, E.R.; Chin, D.; et al. The CDK4/6 Inhibitor Abemaciclib Induces a T Cell Inflamed Tumor Microenvironment and Enhances the Efficacy of PD-L1 Checkpoint Blockade. Cell Rep. 2018, 22, 2978–2994. [Google Scholar] [CrossRef] [Green Version]
  85. Uzhachenko, R.V.; Bharti, V.; Ouyang, Z.; Blevins, A.; Mont, S.; Saleh, N.; Lawrence, H.A.; Shen, C.; Chen, S.-C.; Ayers, G.D.; et al. Metabolic modulation by CDK4/6 inhibitor promotes chemokine-mediated recruitment of T cells into mammary tumors. Cell Rep. 2021, 35, 108944. [Google Scholar] [CrossRef]
  86. Heckler, M.; Ali, L.R.; Clancy-Thompson, E.; Qiang, L.; Ventre, K.S.; Lenehan, P.; Roehle, K.; Luoma, A.; Boelaars, K.; Peters, V.; et al. Inhibition of CDK4/6 Promotes CD8 T-cell Memory Formation. Cancer Discov. 2021, 11, 2564–2581. [Google Scholar] [CrossRef]
  87. Lelliott, E.J.; Sheppard, K.E.; McArthur, G.A. Harnessing the immunotherapeutic potential of CDK4/6 inhibitors in melanoma: Is timing everything? npj Precis. Oncol. 2022, 6, 26. [Google Scholar] [CrossRef]
  88. Lelliott, E.J.; Kong, I.Y.; Zethoven, M.; Ramsbottom, K.M.; Martelotto, L.G.; Meyran, D.; Jiang Zhu, J.; Costacurta, M.; Kirby, L.; Sandow, J.J.; et al. CDK4/6 inhibition promotes anti-tumor immunity through the induction of T cell memory. Cancer Discov. 2021, 11, 2582–2601. [Google Scholar] [CrossRef]
  89. Taniguchi, H.; Caeser, R.; Chavan, S.S.; Zhan, Y.A.; Chow, A.; Manoj, P.; Uddin, F.; Kitai, H.; Qu, R.; Hayatt, O.; et al. WEE1 inhibition enhances the antitumor immune response to PD-L1 blockade by the concomitant activation of STING and STAT1 pathways in SCLC. Cell Rep. 2022, 39, 110814. [Google Scholar] [CrossRef] [PubMed]
  90. Li, M.; Liu, Z.; Wang, X. Exploration of the Combination of PLK1 Inhibition with Immunotherapy in Cancer Treatment. J. Oncol. 2018, 2018, 3979527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Zhou, J.; Yang, Q.; Lu, L.; Tuo, Z.; Shou, Z.; Cheng, J. PLK1 Inhibition Induces Immunogenic Cell Death and Enhances Immunity against NSCLC. Int. J. Med Sci. 2021, 18, 3516–3525. [Google Scholar] [CrossRef]
  92. Yin, T.; Zhao, Z.-B.; Guo, J.; Wang, T.; Yang, J.-B.; Wang, C.; Long, J.; Ma, S.; Huang, Q.; Zhang, K.; et al. Aurora A Inhibition Eliminates Myeloid Cell–Mediated Immunosuppression and Enhances the Efficacy of Anti–PD-L1 Therapy in Breast Cancer. Cancer Res. 2019, 79, 3431–3444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tani, T.; Kitajima, S.; Conway, E.B.; Knelson, E.H.; Barbie, D.A. KRAS G12C inhibition and innate immune targeting. Expert Opin. Ther. Targets 2021, 25, 167–174. [Google Scholar] [CrossRef] [PubMed]
  94. Ostrem, J.M.; Peters, U.; Sos, M.L.; Wells, J.A.; Shokat, K.M. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013, 503, 548–551. [Google Scholar] [CrossRef] [Green Version]
  95. Canon, J.; Rex, K.; Saiki, A.Y.; Mohr, C.; Cooke, K.; Bagal, D.; Gaida, K.; Holt, T.; Knutson, C.G.; Koppada, N.; et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019, 575, 217–223. [Google Scholar] [CrossRef]
  96. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef]
  97. Gainor, J.F.; Shaw, A.T.; Sequist, L.V.; Fu, X.; Azzoli, C.G.; Piotrowska, Z.; Huynh, T.G.; Zhao, L.; Fulton, L.; Schultz, K.R.; et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non–Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res. 2016, 22, 4585–4593. [Google Scholar] [CrossRef] [Green Version]
  98. Shi, C.; Qin, K.; Lin, A.; Jiang, A.; Cheng, Q.; Liu, Z.; Zhang, J.; Luo, P. The role of DNA damage repair (DDR) system in response to immune checkpoint inhibitor (ICI) therapy. J. Exp. Clin. Cancer Res. 2022, 41, 268. [Google Scholar] [CrossRef]
  99. Bracci, L.; Schiavoni, G.; Sistigu, A.; Belardelli, F. Immune-based mechanisms of cytotoxic chemotherapy: Implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2013, 21, 15–25. [Google Scholar] [CrossRef] [PubMed]
  100. Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
  101. Tung, N.M.; Robson, M.E.; Ventz, S.; Santa-Maria, C.A.; Nanda, R.; Marcom, P.K.; Shah, P.D.; Ballinger, T.J.; Yang, E.S.; Vinayak, S.; et al. TBCRC 048: Phase II Study of Olaparib for Metastatic Breast Cancer and Mutations in Homologous Recombination-Related Genes. J. Clin. Oncol. 2020, 38, 4274–4282. [Google Scholar] [CrossRef] [PubMed]
  102. Ding, L.; Kim, H.-J.; Wang, Q.; Kearns, M.; Jiang, T.; Ohlson, C.E.; Li, B.B.; Xie, S.; Liu, J.F.; Stover, E.H.; et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep. 2018, 25, 2972–2980.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Higuchi, T.; Flies, D.B.; Marjon, N.A.; Mantia-Smaldone, G.; Ronner, L.; Gimotty, P.A.; Adams, S.F. CTLA-4 Blockade Synergizes Therapeutically with PARP Inhibition in BRCA1-Deficient Ovarian Cancer. Cancer Immunol. Res. 2015, 3, 1257–1268. [Google Scholar] [CrossRef] [Green Version]
  104. Pilié, P.G.; Gay, C.M.; Byers, L.A.; O’Connor, M.J.; Yap, T.A. PARP Inhibitors: Extending Benefit beyond BRCA-Mutant Cancers. Clin. Cancer Res. 2019, 25, 3759–3771. [Google Scholar] [CrossRef] [Green Version]
  105. Shen, J.; Zhao, W.; Ju, Z.; Wang, L.; Peng, Y.; Labrie, M.; Yap, T.A.; Mills, G.B.; Peng, G. PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness. Cancer Res. 2019, 79, 311–319. [Google Scholar] [CrossRef] [Green Version]
  106. Dunphy, G.; Flannery, S.M.; Almine, J.F.; Connolly, D.J.; Paulus, C.; Jønsson, K.L.; Jakobsen, M.R.; Nevels, M.M.; Bowie, A.G.; Unterholzner, L. Non-canonical Activation of the DNA Sensing Adaptor STING by ATM and IFI16 Mediates NF-κB Signaling after Nuclear DNA Damage. Mol. Cell 2018, 71, 745–760. [Google Scholar] [CrossRef] [Green Version]
  107. Li, T.; Chen, Z.J. The cGAS–cGAMP–STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299. [Google Scholar] [CrossRef]
  108. Vanpouille-Box, C.; Demaria, S.; Formenti, S.C.; Galluzzi, L. Cytosolic DNA Sensing in Organismal Tumor Control. Cancer Cell 2018, 34, 361–378. [Google Scholar] [CrossRef] [Green Version]
  109. Hsieh, R.C.-E.; Krishnan, S.; Wu, R.-C.; Boda, A.R.; Liu, A.; Winkler, M.; Hsu, W.-H.; Lin, S.H.; Hung, M.-C.; Chan, L.-C.; et al. ATR-mediated CD47 and PD-L1 up-regulation restricts radiotherapy-induced immune priming and abscopal responses in colorectal cancer. Sci. Immunol. 2022, 7, eabl9330. [Google Scholar] [CrossRef]
  110. Sato, H.; Niimi, A.; Yasuhara, T.; Permata, T.B.M.; Hagiwara, Y.; Isono, M.; Nuryadi, E.; Sekine, R.; Oike, T.; Kakoti, S.; et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 2017, 8, 1751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. da Costa, A.A.B.A.; Chowdhury, D.; Shapiro, G.I.; D’Andrea, A.D.; Konstantinopoulos, P.A. Targeting replication stress in cancer therapy. Nat. Rev. Drug Discov. 2022, 1–21. [Google Scholar] [CrossRef] [PubMed]
  112. Gaillard, H.; Garcia-Muse, T.; Aguilera, A. Replication stress and cancer. Nat. Rev. Cancer 2015, 15, 276–289. [Google Scholar] [CrossRef] [PubMed]
  113. Plummer, E.R.; Kristeleit, R.S.; Cojocaru, E.; Haris, N.; Carter, L.; Jones, R.H.; Blagden, S.P.; Evans, T.J.; Arkenau, H.-T.; Sarker, D.; et al. A first-in-human phase I/II trial of SRA737 (a Chk1 Inhibitor) in subjects with advanced cancer. J. Clin. Oncol. 2019, 37, 3094. [Google Scholar] [CrossRef]
  114. Yap, T.A.; O’Carrigan, B.; Penney, M.S.; Lim, J.S.; Brown, J.S.; Luken, M.J.D.M.; Tunariu, N.; Perez-Lopez, R.; Rodrigues, D.N.; Riisnaes, R.; et al. Phase I Trial of First-in-Class ATR Inhibitor M6620 (VX-970) as Monotherapy or in Combination With Carboplatin in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2020, 38, 3195–3204. [Google Scholar] [CrossRef] [PubMed]
  115. Sen, T.; Della Corte, C.M.; Milutinovic, S.; Cardnell, R.J.; Diao, L.; Ramkumar, K.; Gay, C.M.; Stewart, C.A.; Fan, Y.; Shen, L.; et al. Combination Treatment of the Oral CHK1 Inhibitor, SRA737, and Low-Dose Gemcitabine Enhances the Effect of Programmed Death Ligand 1 Blockade by Modulating the Immune Microenvironment in SCLC. J. Thorac. Oncol. 2019, 14, 2152–2163. [Google Scholar] [CrossRef]
  116. Kim, R.; Kwon, M.; An, M.; Kim, S.; Smith, S.; Loembé, A.; Mortimer, P.; Armenia, J.; Lukashchuk, N.; Shah, N.; et al. Phase II study of ceralasertib (AZD6738) in combination with durvalumab in patients with advanced/metastatic melanoma who have failed prior anti-PD-1 therapy. Ann. Oncol. 2021, 33, 193–203. [Google Scholar] [CrossRef]
  117. Tang, Z.; Pilié, P.G.; Geng, C.; Manyam, G.C.; Yang, G.; Park, S.; Wang, D.; Peng, S.; Wu, C.; Peng, G.; et al. ATR Inhibition Induces CDK1–SPOP Signaling and Enhances Anti–PD-L1 Cytotoxicity in Prostate Cancer. Clin. Cancer Res. 2021, 27, 4898–4909. [Google Scholar] [CrossRef]
  118. Italiano, A.; Infante, J.; Shapiro, G.; Moore, K.; LoRusso, P.; Hamilton, E.; Cousin, S.; Toulmonde, M.; Postel-Vinay, S.; Tolaney, S.; et al. Phase I study of the checkpoint kinase 1 inhibitor GDC-0575 in combination with gemcitabine in patients with refractory solid tumors. Ann. Oncol. 2018, 29, 1304–1311. [Google Scholar] [CrossRef]
  119. Wallez, Y.; Dunlop, C.R.; Johnson, T.I.; Koh, S.-B.; Fornari, C.; Yates, J.W.; Fernández, S.B.D.Q.; Lau, A.; Richards, F.M.; Jodrell, D.I. The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression. Mol. Cancer Ther. 2018, 17, 1670–1682. [Google Scholar] [CrossRef] [PubMed]
  120. Proctor, M.; Cruz, J.G.; Daignault-Mill, S.; Veitch, M.; Zeng, B.; Ehmann, A.; Sabdia, M.; Snell, C.; Keane, C.; Dolcetti, R.; et al. Targeting Replication Stress Using CHK1 Inhibitor Promotes Innate and NKT Cell Immune Responses and Tumour Regression. Cancers 2021, 13, 3733. [Google Scholar] [CrossRef] [PubMed]
  121. Oo, Z.Y.; Proctor, M.; Stevenson, A.J.; Nazareth, D.; Fernando, M.; Daignault, S.M.; Lanagan, C.; Walpole, S.; Bonazzi, V.; Škalamera, D.; et al. Combined use of subclinical hydroxyurea and CHK1 inhibitor effectively controls melanoma and lung cancer progression, with reduced normal tissue toxicity compared to gemcitabine. Mol. Oncol. 2019, 13, 1503–1518. [Google Scholar] [CrossRef] [Green Version]
  122. Nazareth, D.; Jones, M.J.K.; Gabrielli, B. Everything in Moderation: Lessons Learned by Exploiting Moderate Replication Stress in Cancer. Cancers 2019, 11, 1320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Dai, E.; Zhu, Z.; Wahed, S.; Qu, Z.; Storkus, W.J.; Guo, Z.S. Epigenetic modulation of antitumor immunity for improved cancer immunotherapy. Mol. Cancer 2021, 20, 171. [Google Scholar] [CrossRef]
  124. Nikolich-Žugich, J. The twilight of immunity: Emerging concepts in aging of the immune system. Nat. Immunol. 2018, 19, 10–19. [Google Scholar] [CrossRef]
  125. Wilson, C.B.; Rowell, E.; Sekimata, M. Epigenetic control of T-helper-cell differentiation. Nat. Rev. Immunol. 2009, 9, 91–105. [Google Scholar] [CrossRef]
  126. Kakaradov, B.; Arsenio, J.; Widjaja, C.E.; He, Z.; Aigner, S.; Metz, P.J.; Yu, B.; Wehrens, E.J.; Lopez, J.; Kim, S.H.; et al. Early transcriptional and epigenetic regulation of CD8+ T cell differentiation revealed by single-cell RNA sequencing. Nat. Immunol. 2017, 18, 422–432. [Google Scholar] [CrossRef] [Green Version]
  127. Veazey, K.J.; Muller, D.; Golding, M.C. Prenatal alcohol exposure and cellular differentiation: A role for Polycomb and Trithorax group proteins in FAS phenotypes? Alcohol Res. 2013, 35, 77–85. [Google Scholar]
  128. Henning, A.; Roychoudhuri, R.; Restifo, N.P. Epigenetic control of CD8+ T cell differentiation. Nat. Rev. Immunol. 2018, 18, 340–356. [Google Scholar] [CrossRef]
  129. Nguyen, A.; Ho, L.; Workenhe, S.T.; Chen, L.; Samson, J.; Walsh, S.R.; Pol, J.; Bramson, J.; Wan, Y. HDACi Delivery Reprograms Tumor-Infiltrating Myeloid Cells to Eliminate Antigen-Loss Variants. Cell Rep. 2018, 24, 642–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Wang, D.; Quiros, J.; Mahuron, K.; Pai, C.-C.; Ranzani, V.; Young, A.; Silveria, S.; Harwin, T.; Abnousian, A.; Pagani, M.; et al. Targeting EZH2 Reprograms Intratumoral Regulatory T Cells to Enhance Cancer Immunity. Cell Rep. 2018, 23, 3262–3274. [Google Scholar] [CrossRef] [PubMed]
  131. Ghosh, S.; Taylor, A.; Chin, M.; Huang, H.-R.; Conery, A.R.; Mertz, J.A.; Salmeron, A.; Dakle, P.J.; Mele, D.; Cote, A.; et al. Regulatory T Cell Modulation by CBP/EP300 Bromodomain Inhibition. J. Biol. Chem. 2016, 291, 13014–13027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Boyson, S.; Gao, C.; Quinn, K.; Boyd, J.; Paculova, H.; Frietze, S.; Glass, K. Functional Roles of Bromodomain Proteins in Cancer. Cancers 2021, 13, 3606. [Google Scholar] [CrossRef]
  133. Wang, N.; Wu, R.; Tang, D.; Kang, R. The BET family in immunity and disease. Signal Transduct. Target. Ther. 2021, 6, 23. [Google Scholar] [CrossRef] [PubMed]
  134. Delmore, J.E.; Issa, G.C.; Lemieux, M.E.; Rahl, P.B.; Shi, J.; Jacobs, H.M.; Kastritis, E.; Gilpatrick, T.; Paranal, R.M.; Qi, J.; et al. BET Bromodomain Inhibition as a Therapeutic Strategy to Target c-Myc. Cell 2011, 146, 904–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Jiang, G.; Deng, W.; Liu, Y.; Wang, C. General mechanism of JQ1 in inhibiting various types of cancer. Mol. Med. Rep. 2020, 21, 1021–1034. [Google Scholar] [CrossRef] [Green Version]
  136. Zhu, H.; Bengsch, F.; Svoronos, N.; Rutkowski, M.R.; Bitler, B.G.; Allegrezza, M.J.; Yokoyama, Y.; Kossenkov, A.V.; Bradner, J.E.; Conejo-Garcia, J.R.; et al. BET Bromodomain Inhibition Promotes Anti-tumor Immunity by Suppressing PD-L1 Expression. Cell Rep. 2016, 16, 2829–2837. [Google Scholar] [CrossRef] [Green Version]
  137. Tan, Y.-F.; Wang, M.; Chen, Z.-Y.; Wang, L.; Liu, X.-H. Inhibition of BRD4 prevents proliferation and epithelial–mesenchymal transition in renal cell carcinoma via NLRP3 inflammasome-induced pyroptosis. Cell Death Dis. 2020, 11, 1–17. [Google Scholar] [CrossRef] [Green Version]
  138. Sasikumar, P.G.; Sudarshan, N.S.; Adurthi, S.; Ramachandra, R.K.; Samiulla, D.S.; Lakshminarasimhan, A.; Ramanathan, A.; Chandrasekhar, T.; Dhudashiya, A.A.; Talapati, S.R.; et al. PD-1 derived CA-170 is an oral immune checkpoint inhibitor that exhibits preclinical anti-tumor efficacy. Commun. Biol. 2021, 4, 699. [Google Scholar] [CrossRef]
  139. Chae, Y.K.; Arya, A.; Iams, W.; Cruz, M.R.; Chandra, S.; Choi, J.; Giles, F. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J. Immunother. Cancer 2018, 6, 39. [Google Scholar] [CrossRef]
  140. Dietsch, G.N.; Randall, T.D.; Gottardo, R.; Northfelt, D.W.; Ramanathan, R.K.; Cohen, P.A.; Manjarrez, K.L.; Newkirk, M.; Bryan, J.K.; Hershberg, R.M. Late-Stage Cancer Patients Remain Highly Responsive to Immune Activation by the Selective TLR8 Agonist Motolimod (VTX-2337). Clin. Cancer Res. 2015, 21, 5445–5452. [Google Scholar] [CrossRef] [PubMed]
  141. Northfelt, D.W.; Ramanathan, R.K.; Cohen, P.A.; Von Hoff, D.D.; Weiss, G.J.; Dietsch, G.N.; Manjarrez, K.L.; Randall, T.D.; Hershberg, R.M. A Phase I Dose-Finding Study of the Novel Toll-like Receptor 8 Agonist VTX-2337 in Adult Subjects with Advanced Solid Tumors or Lymphoma. Clin. Cancer Res. 2014, 20, 3683–3691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Dudek, A.Z.; Yunis, C.; Harrison, L.I.; Kumar, S.; Hawkinson, R.; Cooley, S.; Vasilakos, J.P.; Gorski, K.S.; Miller, J.S. First in Human Phase I Trial of 852A, a Novel Systemic Toll-like Receptor 7 Agonist, to Activate Innate Immune Responses in Patients with Advanced Cancer. Clin. Cancer Res. 2007, 13, 7119–7125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Donin, N.M.; Chamie, K.; Lenis, A.T.; Pantuck, A.J.; Reddy, M.; Kivlin, D.; Holldack, J.; Pozzi, R.; Hakim, G.; Karsh, L.I.; et al. A phase 2 study of TMX-101, intravesical imiquimod, for the treatment of carcinoma in situ bladder cancer. Urol. Oncol. Semin. Orig. Investig. 2016, 35, 39.e1–39.e7. [Google Scholar] [CrossRef]
  144. Van Dalen, F.J.; van Stevendaal, M.H.M.E.; Fennemann, F.L.; Verdoes, M.; Ilina, O. Molecular repolarisation of tu-mour-associated macrophages. Molecules 2019, 24, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Müller, E.; Christopoulos, P.F.; Halder, S.; Lunde, A.; Beraki, K.; Speth, M.; Øynebråten, I.; Corthay, A. Toll-Like Receptor Ligands and Interferon-γ Synergize for Induction of Antitumor M1 Macrophages. Front. Immunol. 2017, 8, 1383. [Google Scholar] [CrossRef]
  146. Müller, E.; Speth, M.; Christopoulos, P.F.; Lunde, A.; Avdagic, A.; Øynebråten, I.; Corthay, A. Both Type I and Type II Interferons Can Activate Antitumor M1 Macrophages When Combined With TLR Stimulation. Front. Immunol. 2018, 9, 2520. [Google Scholar] [CrossRef] [Green Version]
  147. Brackett, C.M.; Kojouharov, B.; Veith, J.; Greene, K.F.; Burdelya, L.G.; Gollnick, S.O.; Abrams, S.I.; Gudkov, A.V. Toll-like receptor-5 agonist, entolimod, suppresses metastasis and induces immunity by stimulating an NK-dendritic-CD8+ T-cell axis. Proc. Natl. Acad. Sci. USA 2016, 113, E874–E883. [Google Scholar] [CrossRef] [Green Version]
  148. Jochems, C.; Fantini, M.; Fernando, R.I.; Kwilas, A.R.; Donahue, R.N.; Lepone, L.M.; Grenga, I.; Kim, Y.-S.; Brechbiel, M.W.; Gulley, J.L.; et al. The IDO1 selective inhibitor epacadostat enhances dendritic cell immunogenicity and lytic ability of tumor antigen-specific T cells. Oncotarget 2016, 7, 37762–37772. [Google Scholar] [CrossRef] [Green Version]
  149. Deaglio, S.; Dwyer, K.M.; Gao, W.; Friedman, D.; Usheva, A.; Erat, A.; Chen, J.-F.; Enjyoji, K.; Linden, J.; Oukka, M.; et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 2007, 204, 1257–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Adamiak, M.; Bujko, K.; Brzezniakiewicz-Janus, K.; Kucia, M.; Ratajczak, J.; Ratajczak, M.Z. The Inhibition of CD39 and CD73 Cell Surface Ectonucleotidases by Small Molecular Inhibitors Enhances the Mobilization of Bone Marrow Residing Stem Cells by Decreasing the Extracellular Level of Adenosine. Stem Cell Rev. Rep. 2019, 15, 892–899. [Google Scholar] [CrossRef] [PubMed]
  151. Leone, R.D.; Sun, I.-M.; Oh, M.-H.; Wen, J.; Englert, J.; Powell, J.D. Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol. Immunother. 2018, 67, 1271–1284. [Google Scholar] [CrossRef]
  152. Kjaergaard, J.; Hatfield, S.; Jones, G.; Ohta, A.; Sitkovsky, M. A2A Adenosine Receptor Gene Deletion or Synthetic A2A Antagonist Liberate Tumor-Reactive CD8+ T Cells from Tumor-Induced Immunosuppression. J. Immunol. 2018, 201, 782–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Tang, R.; Xu, J.; Zhang, B.; Liu, J.; Liang, C.; Hua, J.; Meng, Q.; Yu, X.; Shi, S. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J. Hematol. Oncol. 2020, 13, 110. [Google Scholar] [CrossRef] [PubMed]
  154. Garg, A.D.; Galluzzi, L.; Apetoh, L.; Baert, T.; Birge, R.B.; Bravo-San Pedro, J.M.; Breckpot, K.; Brough, D.; Chaurio, R.; Cirone, M.; et al. Molecular and Translational Classifications of DAMPs in Immunogenic Cell Death. Front. Immunol. 2015, 6, 588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Zhou, J.; Wang, G.; Chen, Y.; Wang, H.; Hua, Y.; Cai, Z. Immunogenic cell death in cancer therapy: Present and emerging inducers. J. Cell. Mol. Med. 2019, 23, 4854–4865. [Google Scholar] [CrossRef] [PubMed]
  156. Hu, B.; Elinav, E.; Huber, S.; Booth, C.J.; Strowig, T.; Jin, C.; Eisenbarth, S.C.; Flavell, R.A. Inflammation-induced tumorigenesis in the colon is regulated by caspase-1 and NLRC4. Proc. Natl. Acad. Sci. USA 2010, 107, 21635–21640. [Google Scholar] [CrossRef] [Green Version]
  157. Janowski, A.M.; Ekolb, R.; Ezhang, W.; Sutterwala, F.S. Beneficial and Detrimental Roles of NLRs in Carcinogenesis. Front. Immunol. 2013, 4, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Dunn, J.H.; Ellis, L.Z.; Fujita, M. Inflammasomes as molecular mediators of inflammation and cancer: Potential role in melanoma. Cancer Lett. 2012, 314, 24–33. [Google Scholar] [CrossRef] [PubMed]
  159. Yatim, N.; Jusforgues-Saklani, H.; Orozco, S.; Schulz, O.; da Silva, R.B.; e Sousa, C.R.; Green, D.R.; Oberst, A.; Albert, M.L. RIPK1 and NF-κB signaling in dying cells determines crosspriming of CD8+ T cells. Science 2016, 350, 328–334. [Google Scholar] [CrossRef] [Green Version]
  160. Aaes, T.L.; Kaczmarek, A.; Delvaeye, T.; De Craene, B.; De Koker, S.; Heyndrickx, L.; Delrue, I.; Taminau, J.; Wiernicki, B.; De Groote, P.; et al. Vaccination with Necroptotic Cancer Cells Induces Efficient Anti-tumor Immunity. Cell Rep. 2016, 15, 274–287. [Google Scholar] [CrossRef] [PubMed]
  161. Collins, A.C.; Cai, H.; Li, T.; Franco, L.H.; Li, X.-D.; Nair, V.R.; Scharn, C.R.; Stamm, C.E.; Levine, B.; Chen, Z.J.; et al. Cyclic GMP-AMP Synthase Is an Innate Immune DNA Sensor for Mycobacterium tuberculosis. Cell Host Microbe 2015, 17, 820–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Jing, W.; McAllister, D.; Vonderhaar, E.P.; Palen, K.; Riese, M.J.; Gershan, J.; Johnson, B.D.; Dwinell, M.B. STING agonist inflames the pancreatic cancer immune microenvironment and reduces tumor burden in mouse models. J. Immunother. Cancer 2019, 7, 115. [Google Scholar] [CrossRef] [PubMed]
  163. Gorchs, L.; Hellevik, T.; Bruun, J.-A.; Camilio, K.-A.; Al-Saad, S.; Stuge, T.-B.; Martinez-Zubiaurre, I. Cancer-Associated Fibroblasts from Lung Tumors Maintain Their Immunosuppressive Abilities after High-Dose Irradiation. Front. Oncol. 2015, 5, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Jenkins, L.; Jungwirth, U.; Avgustinova, A.; Iravani, M.; Mills, A.P.; Haider, S.; Harper, J.; Isacke, C.M. Cancer-Associated Fibroblasts Suppress CD8+ T-cell Infiltration and Confer Resistance to Immune-Checkpoint Blockade. Cancer Res. 2022, 82, 2904–2917. [Google Scholar] [CrossRef] [PubMed]
  165. Efferth, T. From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin. Cancer Biol. 2017, 46, 65–83. [Google Scholar] [CrossRef]
  166. Efferth, T. Cancer combination therapy of the sesquiterpenoid artesunate and the selective EGFR-tyrosine kinase inhibitor erlotinib. Phytomedicine 2017, 37, 58–61. [Google Scholar] [CrossRef]
  167. Zhang, J.; Zhou, L.; Xiang, J.-D.; Jin, C.-S.; Li, M.-Q.; He, Y.-Y. Artesunate-induced ATG5-related autophagy enhances the cytotoxicity of NK92 cells on endometrial cancer cells via interactions between CD155 and CD226/TIGIT. Int. Immunopharmacol. 2021, 97, 107705. [Google Scholar] [CrossRef]
  168. Buoncervello, M.; Romagnoli, G.; Buccarelli, M.; Fragale, A.; Toschi, E.; Parlato, S.; Lucchetti, D.; Macchia, D.; Spada, M.; Canini, I.; et al. IFN-α potentiates the direct and immune-mediated antitumor effects of epigenetic drugs on both metastatic and stem cells of colorectal cancer. Oncotarget 2016, 7, 26361–26373. [Google Scholar] [CrossRef]
  169. Fragale, A.; Romagnoli, G.; Licursi, V.; Buoncervello, M.; Del Vecchio, G.; Giuliani, C.; Parlato, S.; Leone, C.; De Angelis, M.; Canini, I.; et al. Antitumor Effects of Epidrug/IFNα Combination Driven by Modulated Gene Signatures in Both Colorectal Cancer and Dendritic Cells. Cancer Immunol. Res. 2017, 5, 604–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Zhang, Y.; Xu, Z.; Feng, W.; Gao, H.; Xu, Z.; Miao, Y.; Li, W.; Chen, F.; Lv, Z.; Huo, J.; et al. Small molecule inhibitors from organoid-based drug screen induce concurrent apoptosis and gasdermin E-dependent pyroptosis in colorectal cancer. Clin. Transl. Med. 2022, 12, e812. [Google Scholar] [CrossRef] [PubMed]
  171. Long, K.; Gu, L.; Li, L.; Zhang, Z.; Li, E.; Zhang, Y.; He, L.; Pan, F.; Guo, Z.; Hu, Z. Small-molecule inhibition of APE1 induces apoptosis, pyroptosis, and necroptosis in non-small cell lung cancer. Cell Death Dis. 2021, 12, 503. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tackling immunosuppression imposed by hostile TME using Small Molecule Inhibitors (SMIs) to improve ICB. The TME recruits immunosuppressive cells, for example, Treg, TAM, MDSC, and CAF. Those inhibitory cells release immunosuppressive factors (e.g., TGF-β, IL-4, IL-5, IL-6, IL-10, IL-13, PG, IDO, and EGF), and further cause the dysregulation of immune checkpoints, inhibition of tumor antigen presentation, and suppression of T cell activation. Among the inhibitors for immune checkpoints, atezolizumab, avelumab, and durvalumab target PD-L1, whereas semiplimab, nivolumab, and pembrolizumab target PD-1. There are also inhibitors for VISTA (CA-170), TIM-3 (CA-327), CTLA-4 (Ipilimumab), A2AR (CPI-444), CD39 (ARL6715), and CD73 (AMPCP). Such immunosuppressive environment suppresses the killing of tumor cells by CD8+ T cells and NK cells, enabling immune evasion. The inhibitors currently in use for inhibiting the shown intrinsic oncogenic signals (highlighted in red) have been shown to modulate immune response by overcoming immunosuppression. Therefore, these SMIs can be a powerful to enhance the clinical efficacy of ICB. Abbreviations: TME, tumor microenvironment; SMIs, small molecule inhibitors; Treg, regulatory T cells; TAM, tumor associated macrophages; MDSC, myeloid-derived suppressor cells; CAF, carcinoma associated fibroblasts; APC, antigen-presenting cell; HMT, methyltransferases; HDAC, histone deacetylases; HAT, histone acetyltransferases. Figure generated with BioRender®.
Figure 1. Tackling immunosuppression imposed by hostile TME using Small Molecule Inhibitors (SMIs) to improve ICB. The TME recruits immunosuppressive cells, for example, Treg, TAM, MDSC, and CAF. Those inhibitory cells release immunosuppressive factors (e.g., TGF-β, IL-4, IL-5, IL-6, IL-10, IL-13, PG, IDO, and EGF), and further cause the dysregulation of immune checkpoints, inhibition of tumor antigen presentation, and suppression of T cell activation. Among the inhibitors for immune checkpoints, atezolizumab, avelumab, and durvalumab target PD-L1, whereas semiplimab, nivolumab, and pembrolizumab target PD-1. There are also inhibitors for VISTA (CA-170), TIM-3 (CA-327), CTLA-4 (Ipilimumab), A2AR (CPI-444), CD39 (ARL6715), and CD73 (AMPCP). Such immunosuppressive environment suppresses the killing of tumor cells by CD8+ T cells and NK cells, enabling immune evasion. The inhibitors currently in use for inhibiting the shown intrinsic oncogenic signals (highlighted in red) have been shown to modulate immune response by overcoming immunosuppression. Therefore, these SMIs can be a powerful to enhance the clinical efficacy of ICB. Abbreviations: TME, tumor microenvironment; SMIs, small molecule inhibitors; Treg, regulatory T cells; TAM, tumor associated macrophages; MDSC, myeloid-derived suppressor cells; CAF, carcinoma associated fibroblasts; APC, antigen-presenting cell; HMT, methyltransferases; HDAC, histone deacetylases; HAT, histone acetyltransferases. Figure generated with BioRender®.
Cancers 14 06150 g001
Table 1. Selected immunotherapy clinical trials, over the last ten years, which demonstrated no benefit. Abbreviations: HR—hazard ratio (Hazard ratio is the ratio of hazard rates between two different treatment groups. A hazard ratio of 1 indicates there is no difference in hazard rates between the two groups. Hazard ratios below 1 indicate that the treatment might be favorable). NSCLC—non-small cell lung cancer, OS—overall survival, PD-1—programmed death 1, PFS—progression free survival, SCLC—small cell lung cancer, SOC—standard of care.
Table 1. Selected immunotherapy clinical trials, over the last ten years, which demonstrated no benefit. Abbreviations: HR—hazard ratio (Hazard ratio is the ratio of hazard rates between two different treatment groups. A hazard ratio of 1 indicates there is no difference in hazard rates between the two groups. Hazard ratios below 1 indicate that the treatment might be favorable). NSCLC—non-small cell lung cancer, OS—overall survival, PD-1—programmed death 1, PFS—progression free survival, SCLC—small cell lung cancer, SOC—standard of care.
NCTICBTrial ArmsPopulationSizeResultsRef.
NCT00861614Ipilimumab (CTLA-4)Ipilimumab vs. placeboCastration resistant prostate cancer with previous treatment with docetaxel988Median OS was 11.2 months (95% CI 9.5–12.7) with ipilimumab and 10.0 months (8.3–11) with placebo (hazard ratio [HR] 0.85, 0.72–1.00; p = 0.053)[13]
NCT01057810Ipilimumab
(CTLA-4)
Ipilimumab vs. placeboCastration-resistant prostate cancer—asymptomatic or minimally symptomatic with metastatic chemotherapy-naive400Median OS was 28.7 months (95% CI, 24.5 to 32.5 months) in the ipilimumab arm versus 29.7 months (95% CI, 26.1 to 34.2 months) in the placebo arm (hazard ratio, 1.11; 95.87% CI, 0.88 to 1.39; p = 0.3667)[14]
NCT02617589Nivolumab
(PD-1)
Nivolumab vs. temozolomideNewly diagnosed MGMT-unmethylated Glioblastoma560Press release—did not meet primary end points of OS or PFS.[15]
NCT02017717Nivolumab
(PD-1)
Nivolumab vs. bevacizumabGrade IV Glioblastoma529median OS (mOS) was comparable between groups: nivolumab, 9.8 months (95% CI, 8.2–11.8); bevacizumab, 10.0 months (95% CI, 9.0–11.8); HR, 1.04 (95% CI, 0.83–1.30); p = 0.76.[16]
NCT02991482Pembrolizumab (PD-1)Pembrolizumab vs. SOCAdvanced malignant mesothelioma previously treated with platinum-based chemotherapy144No difference in OS was detected between groups (HR = 1.12, 95% CI: 0.74–1.69; p = 0.59)[17]
NCT02555657Pembrolizumab (PD-1)Pembrolizumab vs. SOCMetastatic triple negative breast cancer, previous treatment with two systemic therapies622In the overall population, median overall survival was 9.9 months (95% CI 8.3–11.4) for the pembrolizumab group and 10.8 months (9.1–12.6) for the chemotherapy group (HR 0.97 [95% CI 0.82–1.15]).[18]
NCT02370498Pembrolizumab (PD-1)Pembrolizumab vs. SOCAdvanced gastric/gastroesophageal junction adenocarcinoma progressive after platinum-based chemotherapy592Median overall survival was 9.1 months (95% CI 6.2–10.7) with pembrolizumab and 8.3 months (7.6–9.0) with paclitaxel (hazard ratio [HR] 0.82, 95% CI 0.66–1.03; one-sided p = 0.0421).[19]
NCT02494583Pembrolizumab (PD-1)Pembrolizumab vs. pembrolizumab plus SOC vs. SOCAdvanced Gastric or Gastroesophageal Junction Adenocarcinoma—first-Line Monotherapy and Combination Therapy763Pembrolizumab plus chemotherapy was not superior to chemotherapy for OS in patients with CPS of 1 or greater (12.5 vs. 11.1 months; HR, 0.85; 95% CI, 0.70–1.03; p = 0.05) or CPS of 10 or greater (12.3 vs. 10.8 months; HR, 0.85; 95% CI, 0.62–1.17; p = 0.16)[20]
NCT02853305Pembrolizumab (PD-1)Pembrolizumab vs. pembrolizumab plus SOC vs. SOCAdvanced or metastatic urothelial carcinoma with no previous systemic therapy1010Pembrolizumab plus chemotherapy versus chemotherapy did not significantly improve overall survival, with a median overall survival of 17.0 months (14.5–19.5) in the pembrolizumab plus chemotherapy group versus 14.3 months (12.3–16.7) in the chemotherapy group (0.86, 0.72–1.02; p = 0.0407).[21]
NCT02702401Pembrolizumab (PD-1)Pembrolizumab vs. placeboAdvanced hepatocellular carcinoma previously systemically treated413OS and PFS did not reach statistical significance per specified criteria. Median OS was 13.9 months (95% CI, 11.6 to 16.0 months) for pembrolizumab versus 10.6 months (95% CI, 8.3 to 13.5 months) for placebo (hazard ratio [HR], 0.781[22]
NCT02551159Durvalumab (PD-1)Durvalumab vs. SOCRecurrent/metastatic head neck squamous cell carcinoma—first line with high PD-1 expression823Press release—did not meet the primary endpoint of improving overall survival (OS) versus the EXTREME treatment regimen (chemotherapy plus cetuximab)[23]
NCT02952586Avelumab (PD-L1)Avelumab + SOC vs. SOCLocally advanced head neck squamous cell carcinoma697Median progression-free survival was not reached (95% CI 16.9 months–not estimable) in the avelumab group and not reached (23.0 months–not estimable) in the placebo group (stratified hazard ratio 1.21 [95% CI 0.93–1.57] favouring the placebo group; one-sided p = 0.92).[24]
NCT02542293Durvalumab (PD-1) + Tremelimumab (CTLA-4)Combination immunotherapy vs. SOCMetastatic NSCLC—first line953Press release—did not meet primary endpoints[25]
NCT02538666Nivolumab (PD-1) and Ipilimumab (CTLA-4)Combination immunotherapy vs. placeboExtensive disease NSCLC—as maintenance therapy post platinum-based chemotherapy1212OS was not significantly prolonged with nivolumab plus ipilimumab versus placebo (hazard ratio [HR], 0.92; 95% CI, 0.75 to 1.12; p = 0.37; median, 9.2 v 9.6 months)[26]
NCT02279732Ipilimumab
(CTLA-4)
Ipilimumab + SOC vs. placebo + SOCMetastatic or recurrent squamous NSCLC342ClinicalTrail.Gov result posted. Recruitment stopped at 204 patients and primary end point not analysed.[27]
NCT01285609Ipilimumab
(CTLA-4)
Ipilimumab + SOC vs. placebo + SOCMetastatic or recurrent squamous NSCLC1289Median OS was 13.4 months for chemotherapy plus ipilimumab and 12.4 months for chemotherapy plus placebo (hazard ratio, 0.91; 95% CI, 0.77 to 1.07; p = 0.25).[28]
NCT01450761Ipilimumab
(CTLA-4)
Ipilimumab + SOC vs. placebo + SOCNewly diagnosed extensive-stage SCLC1351Median OS was 11.0 months for chemotherapy plus ipilimumab versus 10.9 months for chemotherapy plus placebo (hazard ratio, 0.94; 95% CI, 0.81 to 1.09; p = 0.3775).[29]
Table 2. Summary of trials combining immunotherapy and small molecule inhibitors (published or currently under trial in last 5 years). The mechanisms of action of the drug are indicated in brackets. Abbreviations: AKT—protein kinase B, ALK—anaplastic lymphoma kinase, BRAF—B-Raf, CTLA-4—cytotoxic T-lymphocyte-associated protein 4, HCC—hepatocellular carcinoma, HDAC—Histone deacetylases, HR—hazard ratio, IDO1—indoleamine 2,3-dioxygenase 1, MEK—mitogen-activated protein kinase, NSCLC—non-small cell lung cancer, ORR—objective response rate, PARP—poly ADP ribose polymerase, PD-1—programmed death-1; PFS—progression free survival, SCC—squamous cell carcinoma, SCLC—small cell lung cancer, SOC—standard of care, TACE—transarterial chemoembolism, TKR—tyrosine kinase receptor.
Table 2. Summary of trials combining immunotherapy and small molecule inhibitors (published or currently under trial in last 5 years). The mechanisms of action of the drug are indicated in brackets. Abbreviations: AKT—protein kinase B, ALK—anaplastic lymphoma kinase, BRAF—B-Raf, CTLA-4—cytotoxic T-lymphocyte-associated protein 4, HCC—hepatocellular carcinoma, HDAC—Histone deacetylases, HR—hazard ratio, IDO1—indoleamine 2,3-dioxygenase 1, MEK—mitogen-activated protein kinase, NSCLC—non-small cell lung cancer, ORR—objective response rate, PARP—poly ADP ribose polymerase, PD-1—programmed death-1; PFS—progression free survival, SCC—squamous cell carcinoma, SCLC—small cell lung cancer, SOC—standard of care, TACE—transarterial chemoembolism, TKR—tyrosine kinase receptor.
NCTTrial NameICBSMITrial ArmsPopulationSizeStatusOutcomesRefs.
Published/Completed Trials
NCT03361865ECHO-007Pembrolizumab (PD-1)Epacadostat (IDO1)1. Pembrolizumab + Epacadostat
2. Pembrolizumab
Cisplatin-ineligible advanced or metastatic urothelial Carcinoma93Completed, not publishedSource—ClinicalTrials.Gov ORR 31.8
(22.46 to 55.24) vs. 24.5
(15.33 to 43.67)
[46]
NCT02752074ECHO-301Pembrolizumab (PD-1)Epacadostat (IDO1)1. Pembrolizumab + Epacadostat
2. Pembrolizumab
Unresectable or metastatic melanoma706CompletedNo significant difference in PFS or OS[47]
NCT03829332LEAP-007Pembrolizumab (PD-1)Lenvatinib
(TKR)
1. Pembrolizumab + lenvatinib + SOC
2. Pembrolizumab + SOC
Treatment-naïve, Metastatic NSCLC623Completed, not publishedClinicalTrials.Gov PFS 6.6 months (Combination) vs. 4.2 months (Pembrolizumab monotherapy) HR 0.78 (p = 0.006). No benefit to overall survival. [48]
NCT03517449KEYNOTE-775Pembrolizumab (PD-1)Lenvatinib (TKR)1. Pembrolizumab + lenvatinib
2. SOC
Advanced, recurrent or metastatic endometrial cancer.827CompletedPFS combo 7.2 vs. SOC 3.8 months; hazard ratio, 0.56; 95% CI, 0.47 to 0.66; p < 0.001. OS 8.3 vs. 11.4 months; hazard ratio, 0.62; 95% CI, 0.51 to 0.75; p < 0.001[49]
NCT02853331KEYNOTE-426Pembrolizumab
(PD-1)
Axitinib
(TKR)
1. Pembrolizumab + Axitinib
2. Sunitinib
First-line in Locally Advanced or Metastatic Renal Cell Carcinoma861CompletedPFS -15.1 months pembrolizumab + axitinib group vs. 11.1—month sunitinib group (HR for disease progression or death, 0.69; 95% CI, 0.57 to 0.84; p < 0.001[50,51]
NCT02684006JAVELIN Renal 101Avelumab
(PD-L1)
Sunitinib
(TKR)
1. Avelumab + axitinib
2. Sunitinib
First-line in Locally Advanced Renal Cell Carcinoma888Completed Median PFS l 13.8 months combination vs. 8.4 months monotherapy (hazard ratio, 0.69; 95% CI, 0.56 to 0.84; p < 0.001[52]
NCT02788279IMblaze370Atezolizumab
(PD-L1)
Cobimetinib
(MEK)
1. Atezolizumab
2. Cobimetinib + Atezolizumab
3. Regorafenib
Previously Treated Unresectable Locally Advanced or Metastatic Colorectal Adenocarcinoma363Completed Not significant difference. Median overall survival was 8.87 months with atezolizumab plus cobimetinib, 7, 10 months with atezolizumab, and 8.51 months with regorafenib; HR 1.00 for the combination versus regorafenib and HR 1.19 (p = 0.34) for atezolizumab versus regorafenib[53]
NCT03141177CheckMate 9ERNivolumab (PD-1)Cabozantinib
(TKR)
1. Nivolumab and Cabozantinib
2. Sunitinib
3. Nivolumab, Ipilimumab, Cabozantinib (discontinued)
First line Advanced or Metastatic Renal Cell Carcinoma 701CompletedPFS 16.6 months (95% CI, 12.5 to 24.9) with nivolumab + cabozantinib vs. 8.3 months (95% CI, 7.0 to 9.7) sunitinib (HR 0.51; 95% CI, 0.41 to 0.64; p < 0.001). OS at 12 months 85.7% (95% CI, 81.3 to 89.1) with nivolumab + cabozantinib vs. 75.6% (95% CI, 70.5 to 80.0) with sunitinib (HR 0.60; 98.89% CI, 0.40 to 0.89; p = 0.001). [54]
NCT03937219COSMIC-313Nivolumab (PD-1_ and Ipilimumab (CTLA-4)Cabozantinib
(TKR)
1. Cabozantinib + nivolumab + ipilimumab followed by cabozantinib + nivolumab
2. nivolumab + ipilimumab followed by nivolumab
First line Advanced or Metastatic Renal Cell Carcinoma of Intermediate or Poor Risk840Completed. Collecting OS data press release/meeting abstract. Primary PFS endpoint (HR 0.73, 95% CI, 0.57–0.94; p = 0.013) in favour of combination[55]
NCT03713593LEAP-002Pembrolizumab
(PD-1)
Lenvatinib
(TKR)
1. lenvatinib plus pembrolizumab
2. Lenvatinib + placebo
First-line Therapy for Advanced HCC794Completed Press release—did not meet primary outcome measures[56]
In Progress Trials (by Tumor Type)
NCT04335006 Carelizumab
(PD-1)
Apatinib
(TKR)
1. Carelizumab + Nab-paclitaxel + Apatinib
2. Carelizumab + Nab-paclitaxel
3. Nab-paclitaxel
Advanced or metastatic Triple Negative Breast Cancer 780Recruiting PFS
NCT04177108 Atezolizumab (PD-L1)Ipatasertib
(AKT)
1. Paclitaxel, Atezolizumab and Ipatasertib
2. Paclitaxel, ipatasertib b
3. Paclitaxel
Locally Advanced Unresectable or Metastatic Triple-Negative Breast Cancer.242Active, not recruitingPFS, OS
NCT03740165KEYLYNK-001Pembrolizumab
(PD-1)
Olaparib
(PARP)
1. Pembrolizumab + Olaparib + SOC
2. Pembrolizumab + SOC
3. SOC
BRCA Non-mutated Advanced Epithelial Ovarian Cancer1284Active, not recruitingPFS
NCT05145218 TQB2450
(PD-L1)
Anlotinib
(TKR)
1. TQB2450 + Anlotinib
2. Paclitaxel
Recurrent platinum-resistant ovarian cancer405RecruitingPFS, OS
NCT03651206ROCSANDostarlimab
(PD-1)
Niraparib
(PARP)
1. Niraparib
2. Niraparib + TSR-042 (Dostarlimab)
3. SOC
Metastatic or Recurrent Endometrial or Ovarian Carcinosarcoma196Recruiting RR, OS
NCT03598270 Atezolizumab
(PD-L1)
Niraparib
(PARP)
1. SOC
2. SOC + Atezolizumab with maintaince atezolizumab + niraparib
Recurrent ovarian cancer414Active, not recruitingPFS
NCT03793166PDGREEINivolumab (PD-1)Cabozantinib
(TKR)
1. Nivolumab
2. Nivolumab + Cabozantinib
Metastatic clear cell renal cancer1046RecruitingOS
NCT04523272 TQB2450
(PD-L1)
Anlotinib
(TKR)
1. TQB2450 + Anlotinib
2. Sunitinib
Locally advanced clear cell renal cancer418RecruitingPFS
NCT05219318SPICIPD-1/PD-L1 ICIVEGFR-Tyrosine Kinase Inhibitor1. Treatment pause post-12 months of therapy.
2. PD-1/PD-L1 inhibitor + TKI
Good or Intermediate Risk Metastatic Renal Cell Carcinoma372Not yet recruitingPFS
NCT04338269CONTACT-03Atezolizumab
(PD-L1)
Cabozantinib
(TKR)
1. Atezolizumab + cabazntinib
2. cabazantinib
Inoperable, Locally Advanced, or Metastatic Renal Cell Carcinoma523Active, not recruitingPFS, OS
NCT04987203 Nivolumab
(PD-1)
Tivozanib
(TKR)
1. Nivolumab + Tivozanib
2. Tivozanib
Locally advanced or metastatic Renal cell carcinoma-with progression following at least 6 weeks of treatment with ICI 326RecruitingPFS
NCT03898180 LEAP-011Pembrolizumab
(PD-1)
Lenvatinib
(TKR)
1. Pembrolizumab + Lenvatinib
2. Pembrolizumab monotherapy
3. Placebo + pembrolizumab
First-line Cisplatin-ineligible Participants with PDL1 expression. Ineligible for Platinum-containing Chemotherapy Urothelial Carcinoma487Active, not recruitingPFS, OS
NCT03834519KEYLYNK-010Pembrolizumab
(PD-1)
Olaparib
(PARP)
1. Pembrolizumab + Olaparib
2. Abiraterone + Prednisone or Enzalutamide
Metastatic Castration-resistant Prostate Cancer793Active, not recruitingPFS, OS
NCT03976375LEAP-008Pembrolizumab
(PD-1)
Lenvatinib
(TKR)
1. Pembrolizumab + Lenvatinib
2. Docetaxel
3. Lenvatinib monotherapy
Metastatic NSCLC405Active, not recruitingOS, PFS
NCT03178552 Atezolizumab
(PD-L1)
Cobimetinib (MEK), Alectinib (ALK), Entrectinib (ROS1), Vemurafenib (BRAF), GDC-6036 (KRAS)Multiple trial arms including different combinationsAdvanced or metastatic NSCLC1000RecruitingORR
NCT04471428 Atezolizumab
(PD-L1)
Cabozantinib (TKR)1. Atezolizuman + cabozantinib
2. Docetaxel
Metastatic NSCLC366Active, not recruitingOS
NCT04921358SAFFRON-301:Tislelizumab
(PD-1)
Sitravatinib
(TKR)
1. Tislelizumab + Sitravatinib
2. Docetaxel
Metastatic NSCLC420RecruitingOS, PFS
NCT03348904 Nivolumab
(PD-1)
Epacadostat
(IDO1)
1. Nivolumab + epacadostat + platnium
2. Platinum chemotherapy
3. Platinum + Nivolumab
Metastatic or recurrent NSCLC2Terminated early
NCT04380636KEYLYNK-012Pembrolizumab
(PD-1)
Olaparib
(PARP)
1. pembrolizumab + chemoradiation → pembrolizumab + olaparib placebo
2. pembrolizumab + chemoradiation → pembrolizumab + olaparib
3. chemoradiation → durvalumab
Unresectable, locally advanced NSCLC870RecruitingPFS, OS
NCT03906071SAPPHIRE Nivolumab
(PD-1)
Sitravatinib
(TKR)
1. Nivolumab and Sitravatinib
2. Docetaxel
Advanced or metastatic NSCLC532Active, not recruitingOS
NCT03976362KEYLYNK-008Pembrolizumab
(PD-1)
Olaparib
(PARP)
1. Pembrolizumab + Carboplatin + Taxane + Maintenance Olaparib
2. Pembrolizumab + Carboplatin + Taxane + Maintenance placebo
First-line Metastatic NSCLC857Active, not recruitingPFS, OS
NCT03976323KEYLYNK-006Pembrolizumab (PD-1)Olaparib
(PARP)
1. Pembrolizumab + Pemetrexed + Platinum Therapy + Maintenance Olaparib
2. Pembrolizumab + Pemetrexed + Platinum Therapy + Maintenance Pemetrexed
First-line Metastatic NSCLC1005Active, not recruitingPFS, OS
NCT03829319LEAP-006Pembrolizumab
(PD-1)
Lenvatinib
(TKR)
1. Pembrolizumab + lenvatinib + SOC
2. Pembrolizumab + SOC
Metastatic Nonsquamous NSCLC726Active, not recruitingSafety, PFS, OS
NCT05042375 Camrelizumab
(PD-1)
Famitinib
(TKR)
1. camrelizumab + famitinib
2. pembrolizumab
3. camrelizumab
PD-L1-Positive Recurrent or Metastatic NSCLC450Not yet recruitingPFS
NCT05346952 TQB2450
(PD-L1)
Anlotinib (TKR)1. TQB2450 + carboplatin + pemetrexed
2. TQB2450 + Anlotinib + Pemetrexed
First-line Treatment on Patient with Advanced Non-squamous NSCLC390RecruitingPFS, OS
NCT05106335 Camrelizumab (PD-1)Famitinib
(TKR)
1. Camerlizumab + famitinib
2. famitinib
3. docetaxel
Advanced NSCLC524RecruitingOS
NCT04234607ETER701TQB2450
(PD-L1)
Anlotinib (TKR)1. TQB2450 + Anlotinib + etoposide + carboplatin
2. Anlotinib + etoposide + carboplatin
3. etoposide + carboplatin
Extensive SCLC738Not yet recruitingPFS, OS
NCT04624204KEYLYNK-013Pembrolizumab (PD-1)Olaparib
(PARP)
1. Pembrolizumab + SOC
2. Pembrolizumab + Olaparib + SOC
3. SOC
Newly Diagnosed Treatment-Naïve Limited-Stage SCLC672RecruitingPFS, OS
NCT04674683 Nivolumab
(PD-1)
HBI-8000
(HDAC)
1. HBI-8000 + nivolumab
2. Placebo + nivolumab
Unresectable or metastatic melanoma480RecruitingORR, PFS
NCT03820986LEAP-003Pembrolizumab (PD-1)Lenvatinib
(TKR)
1. Lenvatinib + pembrolizumab
2. Pembrolizumab + placebo
First-line in adults With Advance Melanoma660Active, not recruitingPFS, OS
NCT03813784 SHR-1210 (PD-1)Apatinib
(TKR)
1. SHR-1210 + Apatinib + SOC
2. SOC
3. SOC + SHR-1210
Advanced or metastatic gastric cancer887Active, not recruitingOS
NCT04949256LEAP-014Pembrolizumab (PD-1) Lenvatinib
(TKR)
1. Pembrolizumab + Lenvatinib + Chemotherapy
2. Pembrolizumab + Chemotherapy
First-line Metastatic Esophageal Carcinoma862RecruitingSafety, PFS, OS
NCT04662710LEAP-015 Pembrolizumab (PD-1)Lenvatinib
(TKR)
1. Lenvatinib + Pembrolizumab + SOC
2. SOC
First-line in Advanced/Metastatic Gastroesophageal Adenocarcinoma790RecruitingPFS, OS
NCT04879368INTEGRATEIIbNivolumab
(PD-1)
Regorafenib
(TKR)
1. Nivolumab + regorafenib
2. SOC
Refractory Advanced Gastro-Oesophageal Cancer450RecruitingOS
NCT05049681 SHR-1210
(PD-1)
Apatinib
(TKR)
1. SHR-1210 + Apatinib
2. SHR-1210
Locally advanced/unresectable, recurrence or metastatic esophegeal SCC234Not yet recruitingOS
NCT04776148LEAP-17Pembrolizumab (PD-1)Lenvatinib
(TKR)
1. lenvatinib + pembrolizumab
2. SOC
Metastatic Colorectal Cancer424Active, not recruitingOS
NCT04669496 Toripalimab (PD-1)Lenvatinib
(TKR)
1. Neoadjuvant GEMOX + Lenvatinib + Toripalimab
2. No neoadjuvant therapy
Resectable Intrahepatic Cholangiocarcinoma with High-risk Recurrence Factors178RecruitingPFS
NCT04246177LEAP-012Pembrolizumab (PD-1) Lenvatinib
(TKR)
1. Lenvatinib plus Pembrolizumab plus TACE
2. Oral Placebo plus IV Placebo plus TACE
Incurable Locally Advanced HCC950RecruitingPFS, OS
NCT04523493 Toripalimab
(PD-1)
Lenvatinib
(TKR)
1. Toripalimab + Lenvatinib
2. Lenvatinib
First-line Therapy for Advanced HCC519RecruitingPFS, OS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sinha, D.; Moseley, P.; Lu, X.; Wright, Q.; Gabrielli, B.; Frazer, I.H.; Cruz, J.L.G. Repurposing of Commercially Existing Molecular Target Therapies to Boost the Clinical Efficacy of Immune Checkpoint Blockade. Cancers 2022, 14, 6150. https://doi.org/10.3390/cancers14246150

AMA Style

Sinha D, Moseley P, Lu X, Wright Q, Gabrielli B, Frazer IH, Cruz JLG. Repurposing of Commercially Existing Molecular Target Therapies to Boost the Clinical Efficacy of Immune Checkpoint Blockade. Cancers. 2022; 14(24):6150. https://doi.org/10.3390/cancers14246150

Chicago/Turabian Style

Sinha, Debottam, Philip Moseley, Xuehan Lu, Quentin Wright, Brian Gabrielli, Ian H. Frazer, and Jazmina L. G. Cruz. 2022. "Repurposing of Commercially Existing Molecular Target Therapies to Boost the Clinical Efficacy of Immune Checkpoint Blockade" Cancers 14, no. 24: 6150. https://doi.org/10.3390/cancers14246150

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop