Next Article in Journal
Pseudo-3D Receiver Deghosting of Seismic Streamer Data Based on l1 Norm Sparse Inversion
Next Article in Special Issue
Is Probiotics Supplementation an Appropriate Strategy to Modulate Inflammation in Physically Active Healthy Adults or Athletes? A Systematic Review
Previous Article in Journal
Rectification of Gas Pipeline Bridging with the Support of Experimental Stress Analysis and Means of Regression and Correlation Analysis
Previous Article in Special Issue
Galleria mellonella as a Novel In Vivo Model to Screen Natural Product-Derived Modulators of Innate Immunity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of Phytochemicals to Improve the Efficacy of Immune Checkpoint Inhibitors: Opportunities and Challenges

1
Department of Medical Oncology, Faculty of Medicine, University of Hacettepe, Ankara 06230, Turkey
2
Department of Internal Medicine, Faculty of Medicine, University of Hacettepe, Ankara 06230, Turkey
3
Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
4
Medical Oncology Unit, National Institute of Gastroenterology, “Saverio de Bellis” Research Hospital, Castellana Grotte, 70013 Bari, Italy
5
Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig 23200, Turkey
*
Author to whom correspondence should be addressed.
Appl. Sci. 2022, 12(20), 10548; https://doi.org/10.3390/app122010548
Submission received: 7 September 2022 / Revised: 12 October 2022 / Accepted: 13 October 2022 / Published: 19 October 2022
(This article belongs to the Special Issue Novel Approaches for Natural Product-Derived Immunomodulators)

Abstract

:
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy and reshaped medical oncology practice over the past decade. However, despite unprecedented and durable clinical responses, most patients eventually fail to respond to ICI therapy due to primary or acquired resistance. There is a great need for complementary alternative medicine, such as botanicals and nutritional supplements, because of their capability to modulate a myriad of molecular mechanisms to prevent immunotherapy resistance and reduce its adverse effects. Mounting evidence suggests that phytochemicals, biologically active compounds derived from plants, can favorably regulate key signaling pathways involved in tumor development and progression. In addition, phytochemicals have been found to exert anticancer effects by altering the expression of checkpoint inhibitors of the immune response. The immunomodulatory activity of phytochemicals in the tumor microenvironment has recently received immense interest. Based on these immunomodulatory activities, phytochemicals could be candidates for combination with ICIs in future clinical studies. The current review focuses on the available evidence for combining phytochemicals with a discussion on the promising opportunities to enhance the efficacy of immune checkpoint inhibitors and potential challenges resulting from these combinations.

1. Introduction

Immune checkpoint inhibitors (ICIs) have dramatically changed the oncology landscape and become a vital part of cancer care [1]. ICIs have demonstrated efficacy in several scenarios, such as monotherapy, combination with chemotherapy or targeted therapy, and first-line and subsequent treatment lines in almost all cancers [2,3,4]. The monoclonal antibodies against PD-1 and PD-L1 are the foundations of modern immunotherapy, and currently, eight anti-PD-1/PD-L1 agents are licensed in several different indications [5,6]. Still, many patients do not respond to ICIs, and resistance is inevitable in previously responding patients. Baseline and on-treatment immune dysregulation and immune exhaustion are the main proposed reasons for this treatment resistance [7]. There is an urgent need for novel approaches, such as combinations, to modify tumor microenvironments and mobilize the immune system to fight against tumors more effectively.
Phytochemicals are plant-based bioactive chemicals with diverse health-promoting effects, including cancer prevention. Removal of antioxidant stress, prevention of DNA damage, and promotion of apoptosis after DNA damage with phytochemicals are hypothesized to be the main mechanisms of the anticancer effects of phytochemicals [8,9,10]. However, besides the antioxidant effects, the impacts of phytochemicals on the immune system could be instrumental for anticancer effects, and the immunomodulatory effects of phytochemicals in tumor microenvironments have recently received a lot of interest [11,12]. The preclinical evidence has demonstrated that plant-based polysaccharides, phenols, and silibinin could improve immunosuppression in the tumor microenvironments by inhibiting myeloid-derived suppressor cell accumulation and CD4-T lymphocyte expansion, as well as change the PD-L1 expression levels in tumor microenvironments [13]. Additionally, metabolomic studies have pointed out the possible role of phytochemicals as the mediators of the immune system and gut microbiome cross-talk, as evidenced by the increased response rates in ICI-treated melanoma patients with increased anacardic acid levels. Beneficial commensal bacteria such as Faecalibacterium prausnitzii and Bacteroides thetaiotamicron were also enriched in ICI responders, supporting an association between phytochemicals and a healthier gut microbiome and driving a better immune response [14].
Based on these immunomodulatory properties, phytochemicals could be candidates for combination therapies with anti-PD-1/anti-PD-L1 agents. Both preclinical data and data from metabolome and microbiome analyses have pointed out the role of phytochemicals in mediating immunotherapy efficacy. However, the evidence is limited, with conflicting results. Therefore, we aim to review the available evidence on the phytochemical and ICI combinations and to discuss the opportunities to improve ICI efficacy and possible challenges resulting from these combinations.

2. Literature Search and Review Structure

We conducted a literature review from the PubMed, Medline, and Embase databases to perform filtering of published studies. The MeSH search terms were “immunotherapy” OR “immune checkpoint” AND “PD-1” OR “PD-L1” OR “CTLA-4” OR “phytochemical” OR “phytonutrient” OR “microbiome”. Additionally, we conducted PubMed searches for the individual phytochemical names. We included original articles in the English language that evaluate the effects of phytochemicals on immune checkpoints, ICI and phytochemical combinations, and the association between the microbiome and ICI efficacy.
The review consisted of four sections. The first section was about “Preclinical Evidence Evaluating the Effects of Phytochemicals on Immune Checkpoints”, for which we reviewed the available evidence on the effects of phytochemicals on immune checkpoints in cell line and animal models. In the second section of the review, “The Preclinical Studies Evaluating the Efficacy of Phytochemical and Immune Checkpoint Inhibitor Combinations”, we reviewed the studies evaluating phytochemical and ICI combinations. Due to a lack of human data, we included preclinical studies only. In “The Clues from the Microbiome Studies on the Benefit of Phytochemicals in Immunotherapy Efficacy”, we reviewed the association between phytochemicals and the microbiome and the therapeutic opportunities that stemmed from phytochemicals’ effects on the microbiome. In the last part of the review, we discussed the further clinical perspectives and challenges related to phytochemical and ICI combinations in the clinic and areas needing improvement to develop effective combinations.

3. Preclinical Evidence Evaluating the Effects of Phytochemicals on Immune Checkpoints

Although the effects of phytochemicals on the immune system and tumor microenvironment have been extensively studied [12,15], the impact of phytochemicals on the immune checkpoints began receiving increased interest following ICIs’ entrance into clinical practice. Phytochemicals have been suggested to have the potential to modulate the therapeutic effects of immune checkpoint inhibitors through the regulation of several signaling pathways (Figure 1). Preclinical studies with variable phytochemicals were conducted on several tumor types, including NSCLC, breast cancer, colorectal cancer, hepatocellular cancer, melanoma, head and neck cancer, and glial tumors (Table 1). While most studies have focused on phytochemicals’ effects on the PD-1 and PD-L1 pathways, one study reported the blockage of both PD-1/PD-L1 and the CTLA-4/CD80 interactions by the flavonoids eriodictyol fisetin and quercetin liquiritigenin [16]. The phytochemicals mainly caused decreases in the tumor PD-L1 expression. In contrast, Z-guggulsterone, a phytosterol, increased PD-L1 mRNA expression and transcription in a dose-dependent manner via the activation of the AKT and ERK1/2 signaling pathways [17]. While the authors suggested that increased PD-L1 expression could be an opportunity to develop combinations with synergism, the increased PD-1 expression secondary to AKT and ERK1/2 activations could reduce ICI efficacy as a result of the pivotal roles of AKT and ERK1/2 activation in resistance to immunotherapy in clinical studies [18,19].
Interestingly, Phoenix dactylifera extract (including gallic acid, coffeic acid, and ellagic acid phytochemicals) increased cardiac and kidney PD-1 expressions in a mouse model exposed to Adriamycin [20]. The Phoenix dactylifera extract led to attenuated cardiotoxicity and nephrotoxicity secondary to decreased oxidative stress and apoptotic pressure. The authors suggested that increased PD-1 levels could protect from cardiotoxicity and nephrotoxicity via reduced oxidative stress and apoptotic pressure [20]. If their observations could be confirmed in clinical trials, the phytochemicals could be used for toxicity prevention in ICI-treated patients. Additionally, increased PD-1 expression and corresponding immunosuppression could be exercised as a strategy to prevent or mitigate immune-related adverse events by immunotherapy.
The STAT pathway was the other commonly affected pathway from phytochemicals, with five studies reporting STAT pathway aberrations in addition to changes in the PD-1/PD-L1 pathway [21,22,23,24,25]. While STAT1 and STAT2 involve the constitution of the anti-tumor response via cross-talks with IFN, STAT3 has pro-oncogenic and immunosuppressive properties [26]. Phytochemicals created a consistent inhibitory effect on the STAT3 pathway, and STAT3 down-regulation was among the drivers of PD-L1 suppression in several studies [22,25]. Contrary to the inhibition of immune-suppressive STAT3 by phytochemicals, Xu et al. demonstrated an inhibitory effect on STAT1 by apigenin, a flavonoid, and observed a greater decrease in PD-L1 by apigenin than curcumin secondary to more potent inhibition of STAT1 by apigenin [23]. These observations show that different phytochemicals could act differently on STAT proteins (immune-activating vs. immune-suppressive), and various classes of phytochemicals (i.e., flavonoid and non-flavonoid polyphenols) could have different potency on the tumor immune milieu (Figure 2). Further delineation of these interactions for individual phytochemicals is paramount to designing successful phytochemical and immunotherapy combinations.
Table 1. The combined effect of dietary phytochemicals and immune checkpoint inhibitors on different types of cancer.
Table 1. The combined effect of dietary phytochemicals and immune checkpoint inhibitors on different types of cancer.
AuthorsPhytochemical GroupPhytochemical CompoundImmune Checkpoint InhibitorCancer TypeCancer ModelMechanism of EffectOutcome
Shao et al. (2017) [27] Non-flavonoid polyphenolsCurcuminAnti-PD-L1 antibody (200 µg)Bladder CancerMB49 bladder carcinoma tumor-bearing C57BL/6 mice- Increase intratumoral CD8+ T-cell infiltration
- Elevated the level of IFN-γ in the blood
- Decrease the number of intratumoral MDSCs
Prolonged survival of intraperitoneal metastasized bladder cancer
Dent et al. (2020) [28] Non-flavonoid polyphenolsCurcuminAnti-PD-1 antibody (50 mg/kg)CRCCT26 colorectal tumor-bearing C57/BL6 mice- Reduce the expression of PD-L1, PD-L2, and ODC
- Elevated the level of MHCA
Reduce tumor growth
Guo et al. (2021) [29]Non-flavonoid polyphenolsCurcuminAnti-PD-1 antibody (10 mg/kg)HCCHep3B hepatocellular tumor-bearing BALB/c female nude mice - Reduce surface PD-L1 expression
- Activate lymphocytes
- Inhibit immune evasion
- Down-regulate TGF-β1 expression
Lowered the HCC growth rate and improved tumor microenvironment
Gong et al. (2021) [30]Non-flavonoid polyphenolsCurcuminAnti-PD-1 antibody (10 mg/kg)CRCMC-38 colorectal tumor-bearing C57BL/6 miceN/A- Reduce tumor growth and tumor volume
-Reduce the risk of tumor recurrence (0% vs. 50% in the combination and IO monotherapy arms)
Hayakawa et al. (2021) [31]Non-flavonoid polyphenolsCurcuminAnti-PD-1 Ab and Anti-PD-L1 Ab (200 µg/100 µL/mouse)CRCMC-38 colorectal tumor-bearing C57BL/6 mice
CT26 colorectal tumor-bearing BALB/c mice
Inhibit STAT3 expression induced by exogenous IL-6Reduce tumor growth
Liu et al. (2021) [32]Non-flavonoid polyphenolsCurcuminAnti-PD-L1 antibody (10 µg/mL)HNSCCHuman HNSCC cell lines (SNU1076,
SNU1041, SCC15, and FaDu)
Reinvigorates defective T-cells through
multiple (PD-1 and TIM-3) and multi-level
(IC receptors and their ligands) IC axis
inhibition
Reduce the tumor volume and weight
Kang et al. (2020) [33] Non-flavonoid polyphenolsGallic acidAnti-PD-1 antibody (5 μg/mL)NSCLCA549 and H292 NSCLC cell lines- Reduce expression levels of PD-L1
- Increase IFNγ levels
Decrease NSCLC cell viability
Lasso et al. (2020) [34] Non-flavonoid polyphenolsGallotallinAnti-PD-L1 antibody (200 µg)MelanomaB16-F10 melanoma tumor-bearing C57BL/6 mice- Increase IFNγ levels
- Increase the number of activated CD4+ and CD8+ T-cells
- Decrease the number of MDSCs
- Increase PD-L1 expression
Decrease in tumor size
Zhang et al. (2019) ([35]Non-flavonoid polyphenolsResveratrolAnti-PD-L1 antibody (100 µg)Ovarian CancerHuman ovarian carcinoma cell lines
(SKOV3 and A2780) and murine
ovarian carcinoma cell line (ID8)
Induction of tumor cell apoptosisReduce tumor growth
Jiang et al. (2021) [25]FlavonoidLuteolin ApigeninAnti-PD-1 antibody (10 mg/kg)NSCLCKRAS-mutant human lung cell lines
(H358, H460, H2122, and A549)
and mice in vivo mode
Down-regulated the IFN-γ-induced PD-L1 expression by suppressing the phosphorylation of STAT3 Reduce the tumor volume and weight
Liu et al. (2020) [36] FlavonoidBilberry AnthocyaninAnti-PD-L1 antibody (200 µg)CRCMC-38 colorectal tumor-bearing C57BL/6 miceModulate the gut microbiomeTumor growth delay
Mo et al. (2021) [37]FlavonoidIcaritinAnti-PD-1 antibody (10 mg/kg)HCC, CRC, and melanomaHuman hepatocellular carcinoma,
colorectal cancer and melanoma cell
lines (HEPA1–6, MC-38, and
B16F10) and mice in vivo model
Down-regulate PD-L1 expression and reduced
nuclear translocation of NF-κB p6.
Reduce the tumor volume and weight
Tang et al. (2019) [38] FlavonoidMelafoloneAnti-PD-1 Ab (200 µg/100 µL/mouse)Lung CancerLewis lung carcinoma or CMT tumor-bearing C57BL/6 miceDown-regulate VEGF, TGF-β, and PD-L1 through COX-2 and EGFR inhibitionPromoted survival
Tumor growth inhibition
Jiang et al. (2019) [39] TerpenesLycopeneAnti-PD-1 antibody (6 mg/kg)Lung CancerLewis lung carcinoma tumor-bearing C57BL/6 mice- Increase IFNγ levels
- Inhibit the expression of PD-L1 via activating JAK
- Inhibit the phosphorylation of AKT
Reduce the tumor volume and weight
Han et al. (2019) [40]TerpenesCryptotanshinoneAnti-PD-L1 antibody 10 µg)HCCHCC-bearing mice modelDevelops long-term ani-tumor immunity and increased tumor infiltration of CD8+ T-cellTumor growth inhibition
Dong et al. (2018) [41]TerpenesDiosgeninAnti-PD-1 antibody (200 µg)MelanomaB16-F10 melanoma tumor-bearing C57BL/6 miceEnhances T-cell immune response by modulating intestinal microbiota and inducing T-cell infiltrationTumor growth inhibition
Ye et al. (2021) [42]OthersAgrocybe aegerita galectinAnti-PD-1 Ab
(200 µg intraperitoneal)
HCCH22, HepG2, and RAW264.7 cell lines
Male Balb/c mice
Increase CD4+
and CD8+ T-cells with combination
Tumor growth inhibition
Abbreviations: COX-2: Cyclooxygenase-2; EGFR: Epidermal growth factor receptor; HCC: Hepatocellular carcinoma; IFNγ: Interferon-gamma; IL-6: Interleukin 6; MDSCs: Myeloid-derived suppressor cells; NSCLC: Non-small-cell lung carcinoma; MHCA: Human major histocompatibility complex class I A; PD-L: Programmed death ligand; ODC: Ornithine decarboxylase; STAT3: Signal transducer and activator of transcription 3; TNF: Tumor necrosis factor; VEGF: Vascular endothelial growth factor.

4. Preclinical Studies Evaluating the Efficacy of Phytochemical and Immune Checkpoint Inhibitor Combinations

Several studies have evaluated the efficacy of ICI plus phytochemical combinations using variable phytochemicals in different cancer models (Table 2). The first report of a phytochemical and ICI combination strategy (bisdemethoxycurcumin with an anti-PD-L1 antibody) was published by Shao et al. in 2017 in a mouse bladder cancer model [27]. The researchers observed T-lymphocyte-based immune activation and removal of immune exhaustion via a decrease in the intratumoral myeloid-suppressor cells, leading to increased survival [27]. Later studies most frequently used curcumin, a non-flavonoid polyphenol, as the experimental phytochemical [28,29,30,31] and primarily focused on the colorectal cancer models [28,30,31,36], followed by NSCLC [33,38,39]. The intratumoral expansion of CD8+ or CD4+ lymphocytes and the increased levels of IFN-γ were consistent findings throughout the studies [34,42].
The decrease in PD-L1 levels was observed in five studies conducted with curcumin [28,29], lycopene [39], gallic acid [33], and melafolone [38] and could contribute to the synergism between ICI and phytochemicals by releasing the inhibitory breaks of immune checkpoints. In contrast, an increase in PD-L1 expression level was reported by Lasso et al. with gallatonin-rich Caesalpinia spinosa and anti-PD-L1 antibody combination [34]. The previous observations of PD-L1 expression increase with resveratrol and piceatannol in breast and colorectal cancer cell lines support the variable effects of different phytochemicals on immune checkpoint expressions [43], although the reason for contrasting effects on PD-L1 expression with the phytochemicals from the same class (non-flavonoid polyphenols) is yet to be defined.
While it could be problematic to expect a synergism between ICIs and phytochemicals, we think the differential effects of phytochemicals on PD-L1 expression levels could be beneficial to aid individualized treatment planning according to tumor microenvironments in different tumors. While decreasing immunosuppression secondary to decreased PD-L1 expression would be beneficial in a tumor-agnostic manner, increased ICI efficacy with increased tumoral PD-L1 expression levels was observed in NSCLC [33], melanoma [34], and bladder cancer [27] patients. Both the pretreatment with phytochemicals and the addition of phytochemicals at times of progression to increase PD-L1 expression should be evaluated as a treatment strategy, especially in tumors with an increased benefit with increased PD-L1 expressions.
Table 2. Summary of phytochemicals targeting PD-L1/PD-1 and CTLA-4 pathways in different types of cancer.
Table 2. Summary of phytochemicals targeting PD-L1/PD-1 and CTLA-4 pathways in different types of cancer.
Authors/YearPhytochemical GroupPhytochemical CompoundSourceCancer TypeCancer Model or Toxicity
Mechanism of Action
Liao et al. (2018) [21] Non-flavonoid polyphenolsCurcuminTurmericHNSCC4-NQO induced C57BL/6 tongue squamous cell carcinoma miceDecrease PD-L1 and p-STAT3Y705 protein expression
Deng et al. (2020) [44]Non-flavonoid polyphenolsCurcuminTurmericHCCHepG2 hepatocellular tumor-bearing BALB/c female nude mice Decreased the protein expression of PD-L1
Inhibiting the TLR4/NF-κB signaling pathway angiogenesis.
Lucas et al. (2018) [43] Non-flavonoid polyphenolsResveratrolRed wine, Grapes, Passion fruitBreast cancerCal51 triple-negative breast cancer and SW620 colon cancerIncrease the expression level of PD-L1 via HDAC3/p300-mediated nuclear factor (NF)-κB signaling
Verdura et al. (2020) [45]Non-flavonoid polyphenolsResveratrolRed wine, Grapes, Passion fruitBreast CancerJIMT-1 and MDA-MB-231 breast cancer cellsIncreased PD-L1 dysfunction
Yang et al. (2021) [46]Non-flavonoid polyphenolsResveratrolRed wine, Grapes, Passion fruitNSCLCHuman lung adenocarcinoma cell lines (A549 and H1299)Activation of SirT1 deacetylase leads to disassembly of the destruction complex, thereby enhancing the binding of β-catenin/TCF to the PD-L1 promoter
Coomb et al. (2016) [47]FlavonoidApigeninParsley, onions, grapefruit, oranges, Breast Cancer Triple-negative MDA-MB-468 BC cells, HER2(+) SK-BR-3 BC cells, and 4T1 mouse mammary carcinoma cellsInhibit IFNγ-induced PD-L1 upregulation
Xu et al. (2018) [23] FlavonoidApigeninApple, artichoke, basil, celery, cherry, grapesMelanomaB16-F10 melanoma tumor-bearing C57BL/6 miceInhibit the IFN-γ-induced activation of STAT
Decreased expression levels of PD-L1
Choi et al. (2020) [48]FlavonoidApigeninSalvia plebeiaCRChPD-L1 knock-in MC38 tumor-bearing humanized PD-1 mouse modelBlocking of PD-1/PD-L1 interaction
Rawangkan et al. (2018) [49] FlavonoidEpigallocatechin gallate (EGCG)Green teaNSCLC4-(methylnitrosamino)-1-(3-pyridyl)-1- butanone induced nonsmall-cell lung cancer A/J miceDown-regulate IFN-γ- and EGF-induced PD-L1 expression
Sellam et al. (2020) [50] FlavonoidSilibininSilybum marianumHNSCCNasopharyngeal carcinoma cell lineDown-regulation in PD-L1 expression by interfering with HIF-1α/LDH-A
Rugamba et al. (2021) [51]FlavonoidSilibininSilybum marianumNSCLCA549, H292, and H460 cell linesSuppresses the mRNA expression of PD-L1 and EMT regulators via inhibition of STAT3 phosphorylation
Wudtiwai et al. (2021) [52]FlavonoidHesperidinOrange peel and other citrus speciesOral CancerHuman OSCC cell lines (HN6 and HN15)Inhibition of phosphorylation of STAT1 and STAT3 down-regulates IFN-γ -induced PD-L1 expression
Ke et al. (2019) [24] FlavonoidBaicalinScutellaria baicalensisHCCH22 hepatocellular tumor-bearing BALB/c mice or BALB/c-nu/nu miceDecrease STAT3 activity
Down-regulate IFN-γ-induced PD-L1 expression
Song et al. (2022) [53]FlavonoidBaicalin Scutellaria baicalensisCRCHuman colon cancer cell lines (HCT-116 and CT26) and mice in vivo model Inhibition of NF-κB signaling pathway
down-regulated PD-L1 expression and MDSC, and up-regulated CD4 + and CD8 + T-cells
Liu et al. (2021) [54]FlavonoidLicochalcone AGlycyrrhiza glabraCRC Human cancer cell lines (A549, HeLa, Hep3B, and HCT116) and mice in vivo model PD-L1 expression was down-regulated by inhibition of NF-κB and Ras/Raf/MEK signaling pathways.
Hao et al. (2019) [55] Flavonoid Icaritin EpimediumMelanomaB16-F10 melanoma tumor-bearing C57BL/6 mice
MC-38 colorectal tumor-bearing C57BL/6 mice
Reduce frequency of MDSCs
Down-regulate PD-L1 expression
Mazewski et al. (2019) [56] Flavonoid Cyanidin-3-O-glucoside Blueberry, raspberry, black rice, cherry CRC Human colorectal cancer cellsDecreased PD-1 and PD-L1 protein expression
Chen et al. (2022) [57] Flavonoid MyricetinCranberry, blueberry, lemon,
garlic
Lung cancer Human lung cancer cell lines (NCI-H1650, NCI-H460, and A549)Inhibition of IFN-γ-induced PD-L1 and IDO1 expression in cancer cells by inhibiting the JAK-STAT-IRF1 axis
Kim et al. (2020) [58] Flavonoid KaempferolGeranii herba N/A PD-1 Jurkat and PD-L1/aAPC CHO-K1 cellsInhibiting PD-1/PD-L1 Interaction
Sahyon et al.(2020) [20] FlavonoidGallic acidPhoenix dactyliferaN/A Adriamycin-induced cardiotoxicity and nephrotoxicity Increased cardiac and kidney PD-1 protein percentage
Xing et al. (2018) [22] Terpenes FraxinelloneDictamnus dasycarpus Lung cancer Human A549 lung cancer cell line Inhibit PD-L1 expression by downregulating the STAT3 and HIF-1α signaling pathways
Huang et al. (2019) [59] Terpenes Platycodin Platycodon grandiflorus Lung cancer NCI–H1975 and NCI–H358 lung cancer cell lines Down-regulate the protein level of PD-L1
Zhang et al. (2019) [60] Terpenes Triptolide Tripterygium wilfordii Glioma Glioma cells Down-regulated IFN-γ-induced PD-L1 expression
Kuo et al. (2021) [61] Terpenes Triptolide Tripterygium wilfordii HNSCC OSCC cell line SAS (JCRB0260) and mice in vivo model Reduces IFN-γ-related JAK2-STAT1 pathway and decreases PD-L1 expression
Tian et al. (2021) [17] Phytosterol Z-guggulsteroneCommiphora mukul tree NSCLC Lewis lung carcinoma tumor-bearing C57BL/6 mice Inducing PD-L1 upregulation partly mediated by FXR, Akt, and Erk1/2 signaling pathways
Wang et al. (2020)
[62]
Saponins Panaxadiol Panax ginsengCRCHCT116, SW620, HT29, and HEK293 colon cancer cell line and mice in vivo modelReduces PD-L1 expression by suppressing HIF-1α and STAT3
Deng et al. (2020) [44] SaponinsGinsenosidesPanaxHCCHepG2 hepatocellular tumor-bearing BALB/c female nude mice Decreased the protein expression of PD-L1
Inhibiting the TLR4/NF-κB signaling pathway angiogenesis.
Bedi et al. (2019) [63]Alkaloid Camptothecin Camptotheca acuminitaCRC SW620, HCT116, and RKO colon cancer cells Reduces PD-L1 expression and upregulates the secretion of pro-tumorigenic cytokines
Hunakova et al. (2019) [64] IsothiocyanatesIsothiocyanateBroccoli, Brussels sprouts, cabbage, cauliflower, horseradishBreast CancerHuman triple-negative Breast Carcinoma MDA-MB-231 CellsDecrease expression levels of PD-L1
Chang et al. (2019) [65] Astragalus membranaceus extractExtractAstragalus membranaceusBreast cancer, CRCMouse breast cancer 4T1 and colorectal cancer CT26Down-regulate PD-L1 expression by suppressing the AKT signaling pathway
Li et al. (2019) [16] Rhus verniciflua Stokes extractEriodictyol, fisetin, quercetin, liquiritigeninRhus verniciflua Stokes-N/ABlocked both the PD-1/PD-L1 and the CTLA-4/CD80 interactions
Safonova et al. (2020) [66] Tussilago farfara extractRhamnogalacturonane I and neutral polysaccharides complexTussilago farfaraLung cancerLewis lung carcinoma tumor-bearing C57BL/6 miceReducing expression levels of PD-1 and PD-L1
Inhibiting PD-1/PD-L1 Interaction
Ryan et al. (2022) [67]Black raspberry extractExtractBlack raspberryHNSCCNitroquinoline-1-oxide (4NQO) induced head and neck cancer C57BL/6 miceDecreased levels of PD-L1 expression
Abbreviations: 4-NQO: 4-nitroquinoline-1-oxide; CTLA4: Cytotoxic T-Lymphocyte Associated Protein 4; EGF: Epidermal growth factor; ERK1/2: Extracellular signal-regulated kinase 1/2; FXR: farnesoid X receptor; HIF-1:hypoxia-inducible factor-1; HNSCC: Head and neck squamous cell carcinoma; IFNγ: Interferon-gamma; LDH: Lactate dehydrogenase; MDSCs: Myeloid-derived suppressor cells; NSCLC: Non-small-cell lung carcinoma; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; PD-L: Programmed death ligand; STAT3: Signal transducer and activator of transcription.

5. Clues from Microbiome Studies on the Benefit of Phytochemicals in Immunotherapy Efficacy

The commensal bacteria residing in the gastrointestinal system and their genome is called the gastrointestinal (GI) microbiome. The GI microbiome plays significant roles in self-defense and inflammation [68]. Recently, the GI microbiome has emerged as a predictor of ICI efficacy, and several studies have demonstrated better survival in ICI-treated patients enriched with beneficial commensal bacteria, including Akkermansia muciniphila, Bacteroides spp., and Faecalibacterium prausnitzii. Akkermansia muciniphila in particular was associated with a response to ICIs in four studies conducted on patients with three different tumors (RCC, NSCLC, and HCC) [69,70,71,72]. Whether the increases in these bacteria are secondary to a bystander effect or these bacteria play roles in ICI efficacy is highly debated. However, the restoration of ICI efficacy with Akkermansia muciniphila transplantation in ICI non-responders mice [70] and the recent report of improved ICI efficacy with microbiome and ICI combination in metastatic renal cell carcinoma patients [73] point to an anti-tumor role of the microbiome rather than it only being a biomarker.
Several phytochemicals such as curcumin and phenols could increase levels of beneficial commensal bacteria (Table 3) and correct the dysbiosis created by oxidative stress, as shown in alcoholic liver disease and fatty liver disease models. Similarly, phytochemicals could correct dysbiosis in patients treated with ICIs. These phytochemicals are most commonly found in fiber-rich diets (Table 3). Frankel et al. reported indirect evidence of this strategy’s possible benefit in melanoma patients [14]. Metabolomic analyses in 39 ICI-treated melanoma patients revealed that the anacardic acid levels were increased in responders. Most of these patients (five out of six) had dietary habits that explained the high anacardic acid levels. Based on these points, we think using phytochemicals to correct dysbiosis and increase levels of bacteria associated with ICI efficacy should be exploited in clinical trials. Additionally, further research is needed to delineate the optimal fiber type to consume for ICI efficacy. Recently, Nakajima et al. reported that a soluble fiber diet increased gut Bacteroides fragilis, previously associated with ICI response, while the insoluble fiber-rich diet reduced the Bacteroides fragilis levels in a mice model [74]. In addition, Li et al. previously demonstrated the enrichment of beneficial commensal Actinobacteria and Akkermansia in obese mice fed with nondigestible fructans, which are found in higher concentrations in bananas, compared to mice fed with cellulose [75,76]. Until the results of these trials become available, the recommendation of eating soluble fiber-rich diets and diets rich in phytochemicals that increase beneficial commensal bacteria could benefit ICI-treated patients.

6. Future Perspectives

Phytochemicals have several effects on promoting anti-tumor immunity and modulating the tumor microenvironment, including immune checkpoints. Based on the available preclinical evidence, phytochemicals have the potential to transform immunologically cold tumors into hot tumors and improve immunotherapy efficacy. This strategy could be especially promising for tumors that have consistently garnered less benefit from ICIs, including sarcomas and brain tumors [104,105]. The available preclinical evidence and the demonstration of higher ICI response rates in melanoma patients with increased anacardic acid levels in metabolomic studies support the progression of phytochemical and ICI combinations to clinical studies [14]. Patient-derived xenograft models would be suitable venues to assess the clinical use of phytochemicals in transforming tumor immune profiles and would be beneficial for further clinical studies.
Combining chemotherapy and ICIs and combining two ICIs (CTLA-4 and PD-1/PD-L1) has become the standard of care in the first-line treatment of NSCLC, gastric cancer, RCC, and melanoma, with improved response rates and survival [106,107,108,109,110]. The increased immune activation in the tumor microenvironment was among the main drivers of the increased efficacy of these combinations [111]. However, the efficacy of adding phytochemicals to these combinations has not been investigated yet. We think that phytochemicals could add benefits to these combinations, and further research focusing on the efficacy of adding phytochemicals to chemotherapy-ICI and ICI-ICI combinations could have implications for current clinical practice.
Another knowledge gap concerns the efficacy of using phytochemical and ICI combinations in adjuvant settings. Although it would be harder to measure the efficacy of phytochemical and ICI combinations in adjuvant settings, animal models with radiated tumors or chemoprevention models using ICI and phytochemical combinations should be designed considering the well-known efficacy of phytochemicals in chemoprevention.
Lastly, the phytochemical structure of the traditional herbs with well-known immunomodulatory effects should be thoroughly delineated to identify more candidates to combine with ICIs. For example, Artemisia products are used in China to fight malaria, allergies, and auto-immune diseases [112]. The recently conducted phytochemical analysis of Artemisia annua L. demonstrated the presence of several flavonoids, hydroxycoumarins, and phytosterols, phytochemicals with possible anticancer effects on immune checkpoints [113]. Furthermore, recent gene expression analyses in HCC about Artemisia scoparia demonstrated the increased expression of BIRC5 and secondary expression of CTLA-4 and LAG-3 immune checkpoints and a possible immune activation with this herbal medicine [114]. Further studies evaluating the phytochemical constituent of herbal medications and corresponding effects on the immune checkpoints are needed.

7. Conclusions

In conclusion, plant-based phytochemicals could be beneficial adjuncts to ICIs with improved immune activation and tumor microenvironment modulation. Further research is needed considering the difficult scenarios in the clinical practice and the areas needing improvement to answer more clinically oriented questions.

Author Contributions

D.C.G. and K.S. had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: D.C.G., A.R., A.D.R., S.A. and K.S. Data collection: D.C.G., T.K.S., A.R. and A.D.R. Statistical analysis: D.C.G., T.K.S., A.R., A.D.R., S.A. and K.S. Drafting of the manuscript: D.C.G. and K.S. Critical revision of the manuscript for important intellectual content: D.C.G., T.K.S., A.R., A.D.R., S.A. and K.S. Study supervision: D.C.G., A.R., A.D.R., S.A. and K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Huang, W.; Chen, J.-J.; Xing, R.; Zeng, Y.-C. Combination therapy: Future directions of immunotherapy in small cell lung cancer. Transl. Oncol. 2021, 14, 100889. [Google Scholar] [CrossRef] [PubMed]
  3. Atkins, M.B.; Larkin, J. Immunotherapy Combined or Sequenced with Targeted Therapy in the Treatment of Solid Tumors: Current Perspectives. JNCI J. Natl. Cancer Inst. 2016, 108, djv414. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, C.; Kulkarni, P.; Salgia, R. Combined Checkpoint Inhibition and Chemotherapy: New Era of 1st-Line Treatment for Non-Small-Cell Lung Cancer. Mol. Ther. Oncolytics 2019, 13, 1–6. [Google Scholar] [CrossRef] [Green Version]
  5. Vaddepally, R.K.; Kharel, P.; Pandey, R.; Garje, R.; Chandra, A.B. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers 2020, 12, 738. [Google Scholar] [CrossRef] [Green Version]
  6. Akinleye, A.; Rasool, Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J. Hematol. Oncol. 2019, 12, 92. [Google Scholar] [CrossRef] [Green Version]
  7. Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef]
  8. Azqueta, A.; Collins, A. Polyphenols and DNA Damage: A Mixed Blessing. Nutrients 2016, 8, 785. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, Y.-J.; Gan, R.-Y.; Li, S.; Zhou, Y.; Li, A.-N.; Xu, D.-P.; Li, H.-B. Antioxidant Phytochemicals for the Prevention and Treatment of Chronic Diseases. Molecules 2015, 20, 21138–21156. [Google Scholar] [CrossRef] [Green Version]
  10. Sahin, T.K.; Bilir, B.; Kucuk, O. Modulation of inflammation by phytochemicals to enhance efficacy and reduce toxicity of cancer chemotherapy. Crit. Rev. Food Sci. Nutr. 2021, 1–15. [Google Scholar] [CrossRef]
  11. Power Coombs, M.R.; Hoskin, D.W. Assessing the Impact of Phytochemicals on Immune Checkpoints: Implications for Cancer Immunotherapy. Methods Mol. Biol. 2020, 2111, 161–173. [Google Scholar] [CrossRef] [PubMed]
  12. Behl, T.; Kumar, K.; Brisc, C.; Rus, M.; Nistor-Cseppento, D.C.; Bustea, C.; Aron, R.A.C.; Pantis, C.; Zengin, G.; Sehgal, A.; et al. Exploring the multifocal role of phytochemicals as immunomodulators. Biomed. Pharmacother. 2021, 133, 110959. [Google Scholar] [CrossRef] [PubMed]
  13. Yin, S.Y.; Yang, N.S.; Lin, T.J. Phytochemicals Approach for Developing Cancer Immunotherapeutics. Front. Pharm. 2017, 8, 386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Frankel, A.E.; Coughlin, L.A.; Kim, J.; Froehlich, T.W.; Xie, Y.; Frenkel, E.P.; Koh, A.Y. Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients. Neoplasia 2017, 19, 848–855. [Google Scholar] [CrossRef] [PubMed]
  15. Rana, P.; Shrama, A.; Mandal, C.C. Molecular insights into phytochemicals-driven break function in tumor microenvironment. J. Food Biochem. 2021, 45, e13824. [Google Scholar] [CrossRef]
  16. Li, W.; Kim, T.I.; Kim, J.H.; Chung, H.-S. Immune Checkpoint PD-1/PD-L1 CTLA-4/CD80 are Blocked by Rhus verniciflua Stokes and its Active Compounds. Molecules 2019, 24, 4062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Tian, H.; Gui, Y.; Wei, Y.; Shang, B.; Sun, J.; Ma, S.; You, W.; Jiang, S. Z-guggulsterone induces PD-L1 upregulation partly mediated by FXR, Akt and Erk1/2 signaling pathways in non-small cell lung cancer. Int. Immunopharmacol. 2021, 93, 107395. [Google Scholar] [CrossRef]
  18. Boussiotis, V.A. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N. Engl. J. Med. 2016, 375, 1767–1778. [Google Scholar] [CrossRef] [Green Version]
  19. Luo, M.; Xia, Y.; Wang, F.; Zhang, H.; Su, D.; Su, C.; Yang, C.; Wu, S.; An, S.; Lin, S.; et al. PD0325901, an ERK inhibitor, enhances the efficacy of PD-1 inhibitor in non-small cell lung carcinoma. Acta Pharm. Sin. B 2021, 11, 3120–3133. [Google Scholar] [CrossRef]
  20. Sahyon, H.A.; Al-Harbi, S.A. Chemoprotective role of an extract of the heart of the Phoenix dactylifera tree on adriamycin-induced cardiotoxicity and nephrotoxicity by regulating apoptosis, oxidative stress and PD-1 suppression. Food Chem. Toxicol. 2020, 135, 111045. [Google Scholar] [CrossRef]
  21. Liao, F.; Liu, L.; Luo, E.; Hu, J. Curcumin enhances anti-tumor immune response in tongue squamous cell carcinoma. Arch. Oral Biol. 2018, 92, 32–37. [Google Scholar] [CrossRef]
  22. Xing, Y.; Mi, C.; Wang, Z.; Zhang, Z.H.; Li, M.Y.; Zuo, H.X.; Wang, J.Y.; Jin, X.; Ma, J. Fraxinellone has anticancer activity in vivo by inhibiting programmed cell death-ligand 1 expression by reducing hypoxia-inducible factor-1α and STAT3. Pharm. Res. 2018, 135, 166–180. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, L.; Zhang, Y.; Tian, K.; Chen, X.; Zhang, R.; Mu, X.; Wu, Y.; Wang, D.; Wang, S.; Liu, F.; et al. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. J. Exp. Clin. Cancer Res. 2018, 37, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ke, M.; Zhang, Z.; Xu, B.; Zhao, S.; Ding, Y.; Wu, X.; Wu, R.; Lv, Y.; Dong, J. Baicalein and baicalin promote antitumor immunity by suppressing PD-L1 expression in hepatocellular carcinoma cells. Int. Immunopharmacol. 2019, 75, 105824. [Google Scholar] [CrossRef]
  25. Jiang, Z.B.; Wang, W.J.; Xu, C.; Xie, Y.J.; Wang, X.R.; Zhang, Y.Z.; Huang, J.M.; Huang, M.; Xie, C.; Liu, P.; et al. Luteolin and its derivative apigenin suppress the inducible PD-L1 expression to improve anti-tumor immunity in KRAS-mutant lung cancer. Cancer Lett. 2021, 515, 36–48. [Google Scholar] [CrossRef]
  26. Avalle, L.; Pensa, S.; Regis, G.; Novelli, F.; Poli, V. STAT1 and STAT3 in tumorigenesis: A matter of balance. JAK-STAT 2012, 1, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Shao, Y.; Zhu, W.; Da, J.; Xu, M.; Wang, Y.; Zhou, J.; Wang, Z. Bisdemethoxycurcumin in combination with α-PD-L1 antibody boosts immune response against bladder cancer. Onco Targets 2017, 10, 2675–2683. [Google Scholar] [CrossRef] [Green Version]
  28. Dent, P.; Booth, L.; Roberts, J.L.; Poklepovic, A.; Hancock, J.F. (Curcumin+sildenafil) enhances the efficacy of 5FU and anti-PD1 therapies in vivo. J. Cell. Physiol. 2020, 235, 6862–6874. [Google Scholar] [CrossRef] [PubMed]
  29. Guo, L.; Li, H.; Fan, T.; Ma, Y.; Wang, L. Synergistic efficacy of curcumin and anti-programmed cell death-1 in hepatocellular carcinoma. Life Sci. 2021, 279, 119359. [Google Scholar] [CrossRef]
  30. Gong, F.; Ma, J.-C.; Jia, J.; Li, F.-Z.; Wu, J.-L.; Wang, S.; Teng, X.; Cui, Z.-K. Synergistic effect of the anti-PD-1 antibody with blood stable and reduction sensitive curcumin micelles on colon cancer. Drug Deliv. 2021, 28, 930–942. [Google Scholar] [CrossRef]
  31. Hayakawa, T.; Yaguchi, T.; Kawakami, Y. Enhanced anti-tumor effects of the PD-1 blockade combined with a highly absorptive form of curcumin targeting STAT3. Cancer Sci. 2020, 111, 4326–4335. [Google Scholar] [CrossRef]
  32. Liu, L.; Lim, M.A.; Jung, S.-N.; Oh, C.; Won, H.-R.; Jin, Y.L.; Piao, Y.; Kim, H.J.; Chang, J.W.; Koo, B.S. The effect of Curcumin on multi-level immune checkpoint blockade and T cell dysfunction in head and neck cancer. Phytomedicine 2021, 92, 153758. [Google Scholar] [CrossRef]
  33. Kang, D.Y.; Sp, N.; Jo, E.S.; Rugamba, A.; Hong, D.Y.; Lee, H.G.; Yoo, J.S.; Liu, Q.; Jang, K.J.; Yang, Y.M. The Inhibitory Mechanisms of Tumor PD-L1 Expression by Natural Bioactive Gallic Acid in Non-Small-Cell Lung Cancer (NSCLC) Cells. Cancers 2020, 12, 727. [Google Scholar] [CrossRef] [Green Version]
  34. Lasso, P.; Gomez-Cadena, A.; Urueña, C.; Donda, A.; Martinez-Usatorre, A.; Romero, P.; Barreto, A.; Fiorentino, S. An Immunomodulatory Gallotanin-Rich Fraction From Caesalpinia spinosa Enhances the Therapeutic Effect of Anti-PD-L1 in Melanoma. Front. Immunol. 2020, 11, 584959. [Google Scholar] [CrossRef]
  35. Zhang, Y.; Yang, S.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agent Cancer 2019, 14, 27. [Google Scholar] [CrossRef] [Green Version]
  36. Liu, X.; Wang, L.; Jing, N.; Jiang, G.; Liu, Z. Biostimulating Gut Microbiome with Bilberry Anthocyanin Combo to Enhance Anti-PD-L1 Efficiency against Murine Colon Cancer. Microorganisms 2020, 8, 175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mo, D.; Zhu, H.; Wang, J.; Hao, H.; Guo, Y.; Wang, J.; Han, X.; Zou, L.; Li, Z.; Yao, H.; et al. Icaritin inhibits PD-L1 expression by Targeting Protein IκB Kinase α. Eur. J. Immunol. 2021, 51, 978–988. [Google Scholar] [CrossRef] [PubMed]
  38. Tang, H.; Liu, Y.; Wang, C.; Zheng, H.; Chen, Y.; Liu, W.; Chen, X.; Zhang, J.; Chen, H.; Yang, Y.; et al. Inhibition of COX-2 and EGFR by Melafolone Improves Anti-PD-1 Therapy through Vascular Normalization and PD-L1 Downregulation in Lung Cancer. J. Pharmacol. Exp. Ther. 2019, 368, 401–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Jiang, X.; Wu, H.; Zhao, W.; Ding, X.; You, Q.; Zhu, F.; Qian, M.; Yu, P. Lycopene improves the efficiency of anti-PD-1 therapy via activating IFN signaling of lung cancer cells. Cancer Cell Int. 2019, 19, 68. [Google Scholar] [CrossRef] [Green Version]
  40. Han, Z.; Liu, S.; Lin, H.; Trivett, A.L.; Hannifin, S.; Yang, D.; Oppenheim, J.J. Inhibition of murine hepatoma tumor growth by cryptotanshinone involves TLR7-dependent activation of macrophages and induction of adaptive antitumor immune defenses. Cancer Immunol. Immunother. 2019, 68, 1073–1085. [Google Scholar] [CrossRef]
  41. Dong, M.; Meng, Z.; Kuerban, K.; Qi, F.; Liu, J.; Wei, Y.; Wang, Q.; Jiang, S.; Feng, M.; Ye, L. Diosgenin promotes antitumor immunity and PD-1 antibody efficacy against melanoma by regulating intestinal microbiota. Cell Death Dis. 2018, 9, 1039. [Google Scholar] [CrossRef] [Green Version]
  42. Ye, X.; Wang, X.; Yu, W.; Yang, Q.; Li, Y.; Jin, Y.; Su, Y.; Song, J.; Xu, B.; Sun, H. Synergistic effects of AAGL and anti-PD-1 on hepatocellular carcinoma through lymphocyte recruitment to the liver. Cancer Biol. Med. 2021, 18, 1092. [Google Scholar] [CrossRef] [PubMed]
  43. Lucas, J.; Hsieh, T.C.; Halicka, H.D.; Darzynkiewicz, Z.; Wu, J.M. Upregulation of PD-L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300-mediated NF-κB signaling. Int. J. Oncol. 2018, 53, 1469–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Deng, Z.; Xu, X.-Y.; Yunita, F.; Zhou, Q.; Wu, Y.-R.; Hu, Y.-X.; Wang, Z.-Q.; Tian, X.-F. Synergistic anti-liver cancer effects of curcumin and total ginsenosides. World J. Gastrointest. Oncol. 2020, 12, 1091–1103. [Google Scholar] [CrossRef]
  45. Verdura, S.; Cuyàs, E.; Cortada, E.; Brunet, J.; Lopez-Bonet, E.; Martin-Castillo, B.; Bosch-Barrera, J.; Encinar, J.A.; Menendez, J.A. Resveratrol targets PD-L1 glycosylation and dimerization to enhance antitumor T-cell immunity. Aging 2020, 12, 8. [Google Scholar] [CrossRef] [Green Version]
  46. Yang, M.; Li, Z.; Tao, J.; Hu, H.; Li, Z.; Zhang, Z.; Cheng, F.; Sun, Y.; Zhang, Y.; Yang, J.; et al. Resveratrol induces PD-L1 expression through snail-driven activation of Wnt pathway in lung cancer cells. J. Cancer Res. Clin. Oncol. 2021, 147, 1101–1113. [Google Scholar] [CrossRef] [PubMed]
  47. Coombs, M.R.P.; Harrison, M.E.; Hoskin, D.W. Apigenin inhibits the inducible expression of programmed death ligand 1 by human and mouse mammary carcinoma cells. Cancer Lett. 2016, 380, 424–433. [Google Scholar] [CrossRef]
  48. Choi, J.-G.; Kim, Y.S.; Kim, J.H.; Kim, T.I.; Li, W.; Oh, T.W.; Jeon, C.H.; Kim, S.J.; Chung, H.-S. Anticancer Effect of Salvia plebeia and Its Active Compound by Improving T-Cell Activity via Blockade of PD-1/PD-L1 Interaction in Humanized PD-1 Mouse Model. Front. Immunol. 2020, 11, 8556. [Google Scholar] [CrossRef]
  49. Rawangkan, A.; Wongsirisin, P.; Namiki, K.; Iida, K.; Kobayashi, Y.; Shimizu, Y.; Fujiki, H.; Suganuma, M. Green Tea Catechin Is an Alternative Immune Checkpoint Inhibitor that Inhibits PD-L1 Expression and Lung Tumor Growth. Molecules 2018, 23, 2071. [Google Scholar] [CrossRef] [Green Version]
  50. Sellam, L.S.; Zappasodi, R.; Chettibi, F.; Djennaoui, D.; Yahi-Ait Mesbah, N.; Amir-Tidadini, Z.C.; Touil-Boukoffa, C.; Ouahioune, W.; Merghoub, T.; Bourouba, M. Silibinin down-regulates PD-L1 expression in nasopharyngeal carcinoma by interfering with tumor cell glycolytic metabolism. Arch. Biochem. Biophys. 2020, 690, 108479. [Google Scholar] [CrossRef]
  51. Rugamba, A.; Kang, D.Y.; Sp, N.; Jo, E.S.; Lee, J.M.; Bae, S.W.; Jang, K.J. Silibinin Regulates Tumor Progression and Tumorsphere Formation by Suppressing PD-L1 Expression in Non-Small Cell Lung Cancer (NSCLC) Cells. Cells 2021, 10, 1632. [Google Scholar] [CrossRef]
  52. Wudtiwai, B.; Makeudom, A.; Krisanaprakornkit, S.; Pothacharoen, P.; Kongtawelert, P. Anticancer Activities of Hesperidin via Suppression of Up-Regulated Programmed Death-Ligand 1 Expression in Oral Cancer Cells. Molecules 2021, 26, 5345. [Google Scholar] [CrossRef] [PubMed]
  53. Song, L.; Zhu, S.; Liu, C.; Zhang, Q.; Liang, X. Baicalin triggers apoptosis, inhibits migration, and enhances anti-tumor immunity in colorectal cancer via TLR4/NF-κB signaling pathway. J. Food Biochem. 2022, 46, e13703. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, X.; Xing, Y.; Li, M.; Zhang, Z.; Wang, J.; Ri, M.; Jin, C.; Xu, G.; Piao, L.; Jin, H.; et al. Licochalcone A inhibits proliferation and promotes apoptosis of colon cancer cell by targeting programmed cell death-ligand 1 via the NF-κB and Ras/Raf/MEK pathways. J. Ethnopharmacol. 2021, 273, 113989. [Google Scholar] [CrossRef]
  55. Hao, H.; Zhang, Q.; Zhu, H.; Wen, Y.; Qiu, D.; Xiong, J.; Fu, X.; Wu, Y.; Meng, K.; Li, J. Icaritin promotes tumor T-cell infiltration and induces antitumor immunity in mice. Eur. J. Immunol. 2019, 49, 2235–2244. [Google Scholar] [CrossRef] [PubMed]
  56. Mazewski, C.; Kim, M.S.; Gonzalez de Mejia, E. Anthocyanins, delphinidin-3-O-glucoside and cyanidin-3-O-glucoside, inhibit immune checkpoints in human colorectal cancer cells in vitro and in silico. Sci. Rep. 2019, 9, 11560. [Google Scholar] [CrossRef] [Green Version]
  57. Chen, Y.-C.; He, X.-L.; Qi, L.; Shi, W.; Yuan, L.-W.; Huang, M.-Y.; Xu, Y.-L.; Chen, X.; Gu, L.; Zhang, L.-L.; et al. Myricetin inhibits interferon-γ-induced PD-L1 and IDO1 expression in lung cancer cells. Biochem. Pharmacol. 2022, 197, 114940. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, J.H.; Kim, Y.S.; Choi, J.-G.; Li, W.; Lee, E.J.; Park, J.-W.; Song, J.; Chung, H.-S. Kaempferol and Its Glycoside, Kaempferol 7-O-Rhamnoside, Inhibit PD-1/PD-L1 Interaction In Vitro. Int. J. Mol. Sci. 2020, 21, 3239. [Google Scholar] [CrossRef]
  59. Huang, M.-Y.; Jiang, X.-M.; Xu, Y.-L.; Yuan, L.-W.; Chen, Y.-C.; Cui, G.; Huang, R.-Y.; Liu, B.; Wang, Y.; Chen, X.; et al. Platycodin D triggers the extracellular release of programed death Ligand-1 in lung cancer cells. Food Chem. Toxicol. 2019, 131, 110537. [Google Scholar] [CrossRef]
  60. Zhang, L.; Yu, J.S. Triptolide reverses helper T cell inhibition and down-regulates IFN-γ induced PD-L1 expression in glioma cell lines. J. Neurooncol. 2019, 143, 429–436. [Google Scholar] [CrossRef]
  61. Kuo, C.S.; Yang, C.Y.; Lin, C.K.; Lin, G.J.; Sytwu, H.K.; Chen, Y.W. Triptolide suppresses oral cancer cell PD-L1 expression in the interferon-γ-modulated microenvironment in vitro, in vivo, and in clinical patients. Biomed. Pharm. 2021, 133, 111057. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, Z.; Li, M.Y.; Zhang, Z.H.; Zuo, H.X.; Wang, J.Y.; Xing, Y.; Ri, M.; Jin, H.L.; Jin, C.H.; Xu, G.H.; et al. Panaxadiol inhibits programmed cell death-ligand 1 expression and tumour proliferation via hypoxia-inducible factor (HIF)-1α and STAT3 in human colon cancer cells. Pharm. Res. 2020, 155, 104727. [Google Scholar] [CrossRef] [PubMed]
  63. Bedi, D.; Henderson, H.J.; Manne, U.; Samuel, T. Camptothecin induces PD-L1 and immunomodulatory cytokines in colon cancer cells. Medicines 2019, 6, 51. [Google Scholar] [CrossRef] [Green Version]
  64. Hunakova, L.; Horvathova, E.; Gronesova, P.; Bobal, P.; Otevrel, J.; Brtko, J. Triorganotin Isothiocyanates Affect Migration and Immune Check-point Receptors in Human Triple-negative Breast Carcinoma MDA-MB-231 Cells. Anticancer Res. 2019, 39, 4845–4851. [Google Scholar] [CrossRef] [PubMed]
  65. Chang, H.-L.; Kuo, Y.-H.; Wu, L.-H.; Chang, C.-M.; Cheng, K.-J.; Tyan, Y.-C.; Lee, C.-H. The extracts of Astragalus membranaceus overcome tumor immune tolerance by inhibition of tumor programmed cell death protein ligand-1 expression. Int. J. Med. Sci. 2020, 17, 939–945. [Google Scholar] [CrossRef] [Green Version]
  66. Safonova, E.A.; Lopatina, K.A.; Razina, T.G.; Zueva, E.P.; Gur’ev, A.M.; Belousov, M.V. Effects of Tussilago farfara L. Polysaccharides on the Expression of PD-1 (CD279) and PD-L1 (CD274) in Peripheral Blood and Tumor Tissue Lymphocytes in Mice with Lewis Lung Carcinoma. Bull. Exp. Biol. Med. 2020, 169, 378–382. [Google Scholar] [CrossRef]
  67. Ryan, N.M.; Lamenza, F.F.; Upadhaya, P.; Pracha, H.; Springer, A.; Swingler, M.; Siddiqui, A.; Oghumu, S. Black raspberry extract inhibits regulatory T-cell activity in a murine model of head and neck squamous cell carcinoma chemoprevention. Front. Immunol. 2022, 13, 2742. [Google Scholar] [CrossRef]
  68. Wu, H.-J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Derosa, L.; Iebba, V.; Albiges, L.; Fidelle, M.; Bonvalet, M.; Colomba, E.; Zitvogel, L.; Escudier, B.; Routy, B. Gut microbiome composition to predict resistance in renal cell carcinoma (RCC) patients on nivolumab. J. Clin. Oncol. 2018, 36, 4519. [Google Scholar] [CrossRef]
  71. Chung, M.W.; Kim, M.J.; Won, E.J.; Lee, Y.J.; Yun, Y.W.; Cho, S.B.; Joo, Y.E.; Hwang, J.E.; Bae, W.K.; Chung, I.J.; et al. Gut microbiome composition can predict the response to nivolumab in advanced hepatocellular carcinoma patients. World J. Gastroenterol. 2021, 27, 7340–7349. [Google Scholar] [CrossRef]
  72. Grenda, A.; Iwan, E.; Chmielewska, I.; Krawczyk, P.; Giza, A.; Bomba, A.; Frąk, M.; Rolska, A.; Szczyrek, M.; Kieszko, R.; et al. Presence of Akkermansiaceae in gut microbiome and immunotherapy effectiveness in patients with advanced non-small cell lung cancer. AMB Express 2022, 12, 86. [Google Scholar] [CrossRef] [PubMed]
  73. Dizman, N.; Meza, L.; Bergerot, P.; Alcantara, M.; Dorff, T.; Lyou, Y.; Frankel, P.; Cui, Y.; Mira, V.; Llamas, M.; et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: A randomized phase 1 trial. Nat. Med. 2022, 28, 704–712. [Google Scholar] [CrossRef]
  74. Nakajima, A.; Sasaki, T.; Itoh, K.; Kitahara, T.; Takema, Y.; Hiramatsu, K.; Ishikawa, D.; Shibuya, T.; Kobayashi, O.; Osada, T.; et al. A Soluble Fiber Diet Increases Bacteroides fragilis Group Abundance and Immunoglobulin A Production in the Gut. Appl. Environ. Microbiol. 2020, 86, e00405-20. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, T.-W.; Cephas, K.D.; Holscher, H.D.; Kerr, K.R.; Mangian, H.F.; Tappenden, K.A.; Swanson, K.S. Nondigestible Fructans Alter Gastrointestinal Barrier Function, Gene Expression, Histomorphology, and the Microbiota Profiles of Diet-Induced Obese C57BL/6J Mice. J. Nutr. 2016, 146, 949–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Shalini, R.; Abinaya, G.; Saranya, P.; Antony, U. Growth of selected probiotic bacterial strains with fructans from Nendran banana and garlic. LWT Food Sci. Technol. 2017, 83, 68–78. [Google Scholar] [CrossRef]
  77. Guven, D.C.; Aktas, B.Y.; Simsek, C.; Aksoy, S. Gut microbiota and cancer immunotherapy: Prognostic and therapeutic implications. Future Oncol. 2020, 16, 497–506. [Google Scholar] [CrossRef]
  78. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [Green Version]
  79. Zhang, C.; Wu, W.; Li, X.; Xin, X.; Liu, D. Daily Supplementation with Fresh Angelica keiskei Juice Alleviates High-Fat Diet-Induced Obesity in Mice by Modulating Gut Microbiota Composition. Mol. Nutr. Food Res. 2019, 63, e1900248. [Google Scholar] [CrossRef]
  80. González, S.; Salazar, N.; Ruiz-Saavedra, S.; Gómez-Martín, M.; de Los Reyes-Gavilán, C.G.; Gueimonde, M. Long-Term Coffee Consumption is Associated with Fecal Microbial Composition in Humans. Nutrients 2020, 12, 1287. [Google Scholar] [CrossRef]
  81. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Qiao, Y.; Sun, J.; Xia, S.; Tang, X.; Shi, Y.; Le, G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014, 5, 1241–1249. [Google Scholar] [CrossRef] [PubMed]
  83. Burns, J.; Yokota, T.; Ashihara, H.; Lean, M.E.J.; Crozier, A. Plant Foods and Herbal Sources of Resveratrol. J. Agric. Food Chem. 2002, 50, 3337–3340. [Google Scholar] [CrossRef] [PubMed]
  84. Dubin, K.; Callahan, M.K.; Ren, B.; Khanin, R.; Viale, A.; Ling, L.; No, D.; Gobourne, A.; Littmann, E.; Huttenhower, C.; et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 2016, 7, 10391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, L.; Jiang, B.; Zhong, C.; Guo, J.; Zhang, L.; Mu, T.; Zhang, Q.; Bi, X. Chemoprevention of colorectal cancer by black raspberry anthocyanins involved the modulation of gut microbiota and SFRP2 demethylation. Carcinogenesis 2018, 39, 471–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
  88. Duda-Chodak, A. The inhibitory effect of polyphenols on human gut microbiota. J. Physiol. Pharm. 2012, 63, 497–503. [Google Scholar]
  89. Gu, J.; Thomas-Ahner, J.M.; Riedl, K.M.; Bailey, M.T.; Vodovotz, Y.; Schwartz, S.J.; Clinton, S.K. Dietary Black Raspberries Impact the Colonic Microbiome and Phytochemical Metabolites in Mice. Mol. Nutr. Food Res. 2019, 63, e1800636. [Google Scholar] [CrossRef]
  90. Ullah, M.F.; Khan, M.W. Food as medicine: Potential therapeutic tendencies of plant derived polyphenolic compounds. Asian Pac. J. Cancer Prev. 2008, 9, 187–195. [Google Scholar] [PubMed]
  91. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed]
  92. Catalkaya, G.; Venema, K.; Lucini, L.; Rocchetti, G.; Delmas, D.; Daglia, M.; De Filippis, A.; Xiao, H.; Quiles, J.L.; Xiao, J.; et al. Interaction of dietary polyphenols and gut microbiota: Microbial metabolism of polyphenols, influence on the gut microbiota, and implications on host health. Food Front. 2020, 1, 109–133. [Google Scholar] [CrossRef]
  93. Anhê, F.F.; Pilon, G.; Roy, D.; Desjardins, Y.; Levy, E.; Marette, A. Triggering Akkermansia with dietary polyphenols: A new weapon to combat the metabolic syndrome? Gut Microbes 2016, 7, 146–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Sheng, L.; Jena, P.K.; Liu, H.-X.; Hu, Y.; Nagar, N.; Bronner, D.N.; Settles, M.L.; Bäumler, A.J.; Wan, Y.-J.Y. Obesity treatment by epigallocatechin-3-gallate-regulated bile acid signaling and its enriched Akkermansia muciniphila. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2018, 32, 6371–6384. [Google Scholar] [CrossRef] [Green Version]
  95. Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef] [PubMed]
  96. Peters, B.; Wilson, M.; Moran, U.; Pavlick, A.C.; Izsak, A.; Wechter, T.; Weber, J.S.; Osman, I.; Ahn, J. Gut microbiome and immunotherapy response in melanoma patients. J. Clin. Oncol. 2018, 36, 9575. [Google Scholar] [CrossRef]
  97. Fukuoka, S.; Daisuke, M.; Togashi, Y.; Sugiyama, E.; Udagawa, H.; Kirita, K.; Kamada, T.; Kawazoe, A.; Goto, K.; Doi, T.; et al. Association of gut microbiome with immune status and clinical response in solid tumor patients who received on anti-PD-1 therapies. J. Clin. Oncol. 2018, 36, 3011. [Google Scholar] [CrossRef]
  98. Romo-Vaquero, M.; Cortés-Martín, A.; Loria-Kohen, V.; Ramírez-de-Molina, A.; García-Mantrana, I.; Collado, M.C.; Espín, J.C.; Selma, M.V. Deciphering the Human Gut Microbiome of Urolithin Metabotypes: Association with Enterotypes and Potential Cardiometabolic Health Implications. Mol. Nutr. Food Res. 2019, 63, e1800958. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, S.; Al-Maghout, T.; Cao, H.; Pelzl, L.; Salker, M.S.; Veldhoen, M.; Cheng, A.; Lang, F.; Singh, Y. Gut Bacterial Metabolite Urolithin A (UA) Mitigates Ca2+ Entry in T Cells by Regulating miR-10a-5p. Front. Immunol. 2019, 10, 1737. [Google Scholar] [CrossRef] [Green Version]
  100. Maia, M.C.; Poroyko, V.; Won, H.; Almeida, L.; Bergerot, P.G.; Dizman, N.; Hsu, J.; Jones, J.; Salgia, R.; Pal, S.K. Association of microbiome and plasma cytokine dynamics to nivolumab response in metastatic renal cell carcinoma (mRCC). J. Clin. Oncol. 2018, 36, 656. [Google Scholar] [CrossRef]
  101. Lordan, C.; Thapa, D.; Ross, R.P.; Cotter, P.D. Potential for enriching next-generation health-promoting gut bacteria through prebiotics and other dietary components. Gut Microbes 2020, 11, 1–20. [Google Scholar] [CrossRef]
  102. Botticelli, A.; Vernocchi, P.; Marini, F.; Quagliariello, A.; Cerbelli, B.; Reddel, S.; Del Chierico, F.; Di Pietro, F.; Giusti, R.; Tomassini, A.; et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J. Transl. Med. 2020, 18, 49. [Google Scholar] [CrossRef] [Green Version]
  103. Sun, H.; Zhang, P.; Zhu, Y.; Lou, Q.; He, S. Antioxidant and prebiotic activity of five peonidin-based anthocyanins extracted from purple sweet potato (Ipomoea batatas (L.) Lam.). Sci. Rep. 2018, 8, 5018. [Google Scholar] [CrossRef] [Green Version]
  104. Buerki, R.A.; Chheda, Z.S.; Okada, H. Immunotherapy of Primary Brain Tumors: Facts and Hopes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 5198–5205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Birdi, H.K.; Jirovec, A.; Cortés-Kaplan, S.; Werier, J.; Nessim, C.; Diallo, J.-S.; Ardolino, M. Immunotherapy for sarcomas: New frontiers and unveiled opportunities. J. Immunother. Cancer 2021, 9, e001580. [Google Scholar] [CrossRef] [PubMed]
  106. Song, X.; Qi, W.; Guo, J.; Sun, L.; Ding, A.; Zhao, G.; Li, H.; Qiu, W.; Lv, J. Immune checkpoint inhibitor combination therapy for gastric cancer: Research progress. Oncol. Lett. 2020, 20, 46. [Google Scholar] [CrossRef] [PubMed]
  107. Liu, W.; Zhang, L.; Xiu, Z.; Guo, J.; Wang, L.; Zhou, Y.; Jiao, Y.; Sun, M.; Cai, J. Combination of Immune Checkpoint Inhibitors with Chemotherapy in Lung Cancer. OncoTargets Ther. 2020, 13, 7229–7241. [Google Scholar] [CrossRef]
  108. Huang, Z.; Su, W.; Lu, T.; Wang, Y.; Dong, Y.; Qin, Y.; Liu, D.; Sun, L.; Jiao, W. First-Line Immune-Checkpoint Inhibitors in Non-Small Cell Lung Cancer: Current Landscape and Future Progress. Front. Pharmacol. 2020, 11, 8091. [Google Scholar] [CrossRef] [PubMed]
  109. Mason, R.; Dearden, H.C.; Nguyen, B.; Soon, J.A.; Smith, J.L.; Randhawa, M.; Mant, A.; Warburton, L.; Lo, S.; Meniawy, T.; et al. Combined ipilimumab and nivolumab first-line and after BRAF-targeted therapy in advanced melanoma. Pigment Cell Melanoma Res. 2020, 33, 358–365. [Google Scholar] [CrossRef]
  110. Garje, R.; An, J.; Greco, A.; Vaddepally, R.K.; Zakharia, Y. The Future of Immunotherapy-Based Combination Therapy in Metastatic Renal Cell Carcinoma. Cancers 2020, 12, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef] [PubMed]
  112. 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] [PubMed]
  113. Abate, G.; Zhang, L.; Pucci, M.; Morbini, G.; Mac Sweeney, E.; Maccarinelli, G.; Ribaudo, G.; Gianoncelli, A.; Uberti, D.; Memo, M.; et al. Phytochemical Analysis and Anti-Inflammatory Activity of Different Ethanolic Phyto-Extracts of Artemisia annua L. Biomolecules 2021, 11, 975. [Google Scholar] [CrossRef]
  114. Mo, Z.; Cao, Z.; Yu, L.; Wang, Y.; Li, P.; Lin, Y.; Zhang, S. An Integrative Analysis Reveals the Potential Mechanism between Herbal Medicine Yinchen and Immunoregulation in Hepatocellular Carcinoma. BioMed Res. Int. 2020, 2020, 8886914. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic figure representing some interactions between phytochemicals, T-cells, and cancer cells. Phytochemicals have been suggested to have the potential to modulate the therapeutic effects of immune checkpoint inhibitors and PD-L1 expression through the regulating of several signaling pathways, such as EGFR/Akt. Some phytochemicals can regulate PD-L1 expression, while others inhibit PD-L1 glycosylation or PD-L1/PD-1 binding, such as quercetin, saponins, and fisetin. Abbreviations: EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; MHC: major histocompatibility complex; PD-1: programmed cell death protein 1; PD-L1: programmed death ligand 1 (PD-L1).
Figure 1. Schematic figure representing some interactions between phytochemicals, T-cells, and cancer cells. Phytochemicals have been suggested to have the potential to modulate the therapeutic effects of immune checkpoint inhibitors and PD-L1 expression through the regulating of several signaling pathways, such as EGFR/Akt. Some phytochemicals can regulate PD-L1 expression, while others inhibit PD-L1 glycosylation or PD-L1/PD-1 binding, such as quercetin, saponins, and fisetin. Abbreviations: EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; MHC: major histocompatibility complex; PD-1: programmed cell death protein 1; PD-L1: programmed death ligand 1 (PD-L1).
Applsci 12 10548 g001
Figure 2. Schematic figure reporting the main phytochemicals which have been tested as modulators of immune checkpoint inhibitors in cancer patients. The flavonoid polyphenols include, among others, icaritin, luteolin, bilberry anthocyanin, and melafolone. Non-flavonoid polyphenolic compounds include curcumin, gallic acid, gallotannin, and resveratrol, while terpenes include lycopene, diosgenin, and cryptotanshinone.
Figure 2. Schematic figure reporting the main phytochemicals which have been tested as modulators of immune checkpoint inhibitors in cancer patients. The flavonoid polyphenols include, among others, icaritin, luteolin, bilberry anthocyanin, and melafolone. Non-flavonoid polyphenolic compounds include curcumin, gallic acid, gallotannin, and resveratrol, while terpenes include lycopene, diosgenin, and cryptotanshinone.
Applsci 12 10548 g002
Table 3. Summary of phytochemicals as modifiers of the gut microbiome in the immunotherapy studies (adapted from Guven DC et al. [77]).
Table 3. Summary of phytochemicals as modifiers of the gut microbiome in the immunotherapy studies (adapted from Guven DC et al. [77]).
Lead Author, YearTarget Population/Patient Number (n)Main FindingsCandidate PhytochemicalPhytochemical Enriched NutrientBacteria
Vetizou et al. (2015) [78]Melanoma/25Increased levels of Bacteroides thetaiotaomicron and Bacteroides fragilis in ICI responders
Improved response to CTLA-4 blockade with FMT from patients with increased fecal Bacteroides spp. levels
Polyphenol/coumarin [79]Soluble fiber-rich diet [74]/High coffee-consumption [80] Bacteroides thetaiotaomicron
Bacteroides
fragilis
Sivan et al. (2015) [81]Melanoma/MiceIncreased levels of Bacteroides spp. in ICI responders
Similar tumor control as PD-L1 blockade with oral supplementation of Bifidobacterium spp.
Resveratrol [82] Grapes, wine, and peanuts [83]Bifidobacterium spp.
Dubin et al. (2016) [84] Melanoma/34Lower risk of colitis in Bacteroides spp. enriched patientsPolyphenol/coumarin [79]Soluble fiber-rich diet [74]/High coffee-consumption [80]Bacteroides spp.
Chaput et al. (2017) [85]Melanoma/26Longer progression-free and overall survival in Faecalibacterium spp. enriched patientsAnthocyanin [86]Black Raspberries [86]Faecalibacterium spp.
Gopalakrishnan et al. (2018) [87]Melanoma/112Higher alpha diversity and increased Ruminococcaceae levels in the feces of ICI responders
Higher buccal and fecal levels of Bacteroidales in ICI non-responders
Polyphenols [88] (Naringenin and quercetin)
Anthocyanin [89]
Onion, apple, broccoli [90]
Black Raspberries [89]
Ruminococcaceae
Bacteroidales
Matson et al. (2018) [91]Melanoma/42Increased Bifidobacterium longum in the feces of ICI responders
Tumor control, augmented T-cell responses, and improved efficacy of anti-PD-L1 blockade with oral supplementation of responders’ feces to germ-free mice
Resveratrol [92]Grapes, wine, and peanuts [83]Bifidobacterium longum
Routy et al. (2018) [69] NSCLC/140
RCC/67
Increased levels of Akkermansia muciniphila in ICI responders
Restoration of the efficacy of PD-1 blockade in antibiotic pretreated mice after FMT from ICI responders
Curcumin [93]/EGCG [94]Prebiotic nondigestible fiber-rich diet [95]Akkermansia muciniphilia
Peters et al. (2018) [96] Melanoma/26Lower risk of progression in patients with higher community diversity
Lower risk of progression in patients enriched with Faecalibacterium prausnitzii
Anthocyanin [86]Black Raspberries [86]Faecalibacterium prausnitzii
Fukuoka et al.
(2018) [97]
NSCLC/14
Gastric cancer/24
Higher alpha diversity and Ruminococcaceae levels in ICI respondersEllagitannins [98]Pomegranates, nuts [99]Ruminococcaceae
Derosa et al. (2018) [70] RCC/85Increased abundance of Akkermansia muciniphila and Bacteroides salyersiae in non-resistant renal cell carcinoma patientsCurcumin [93]/EGCG [94]Prebiotic nondigestible fiber-rich diet [95]Akkermansia muciniphila
Restoration of the efficacy of the ICI with Akkermansia muciniphila and Bacteroides salyersiae transplantation to mice with unfavorable/dysbiotic profileNot reported Bacteroides salyersiae
Maia et al. (2018) [100]RCC/20Increased abundance of Roseburia and Faecalibacterium spp. in ICI respondersAnthocyanin [86]
Polyphenols [101]
Black Raspberries [86]
Resistant Starch [101]
Faecalibacterium prausnitzii
Roseburia
Botticelli et al. (2020) [102]NSCLC/11Increased fecal Akkermansia muciniphila, Bifidobacterium longum, and Faecalibacterium prausnitzii levels in ICI respondersCurcumin [93]/EGCG [94]
Resveratrol [82], Anthocyanin [103]
Anthocyanin [86]
Prebiotic nondigestible fiber-rich diet [95]
Grapes, wine, and peanuts [83],
Chinese purple sweet potato cultivar [103]
Black Raspberries [86]
Akkermansia muciniphila
Bifidobacterium longum
Faecalibacterium prausnitzii
Liu et al.
(2020) [36]
CRC/miceThe addition of anthocyanins in the α-PD-L1 treatment showed an overrepresentation of Lachnospiraceae and RuminococcaceaeAnthocyanin [86]Black Raspberries [86]Ruminococcaceae and Lachnospiraceae
Chung et al. (2021) [71]HCC/8 Increased fecal Akkermansia levels in ICI respondersCurcumin [93]/EGCG [94]Prebiotic nondigestible fiber-rich diet [95]
Grapes, wine, and peanuts [83]
Akkermansiaceae
Grenda et al. (2022) [72]NSCLC/47Increased fecal Akkermansia levels in ICI respondersCurcumin [93]/EGCG [94]Prebiotic nondigestible fiber-rich diet [95]
Grapes, wine, and peanuts [83]
Akkermansiaceae
Abbreviations: EGCG: Epigallocatechin gallate; FMT: Fecal microbiota transfer; ICI: Immune checkpoint inhibitors; NSCLC: Non-small-cell lung carcinoma; RCC: Renal cell carcinoma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Guven, D.C.; Sahin, T.K.; Rizzo, A.; Ricci, A.D.; Aksoy, S.; Sahin, K. The Use of Phytochemicals to Improve the Efficacy of Immune Checkpoint Inhibitors: Opportunities and Challenges. Appl. Sci. 2022, 12, 10548. https://doi.org/10.3390/app122010548

AMA Style

Guven DC, Sahin TK, Rizzo A, Ricci AD, Aksoy S, Sahin K. The Use of Phytochemicals to Improve the Efficacy of Immune Checkpoint Inhibitors: Opportunities and Challenges. Applied Sciences. 2022; 12(20):10548. https://doi.org/10.3390/app122010548

Chicago/Turabian Style

Guven, Deniz Can, Taha Koray Sahin, Alessandro Rizzo, Angela Dalia Ricci, Sercan Aksoy, and Kazim Sahin. 2022. "The Use of Phytochemicals to Improve the Efficacy of Immune Checkpoint Inhibitors: Opportunities and Challenges" Applied Sciences 12, no. 20: 10548. https://doi.org/10.3390/app122010548

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