4,816
views
1
recommends
+1 Recommend
1 collections
    1
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Role of natural products in tumor therapy from basic research and clinical perspectives

      Published
      review-article
      Bookmark

            Abstract

            Cancer is the leading cause of morbidity and mortality worldwide and is an important barrier to lengthening life expectancy in every country. Natural products are receiving increased attention from researchers globally and increasing numbers of natural products are approved for clinical studies involving cancer in recent years. To gain more insight into natural products that have undergone clinical trials for cancer treatment, a comprehensive search was conducted. The https://clinicaltrials.gov website was searched for relevant clinical trials and natural product information up to December 2022. The search terms included different types of cancers, such as colorectal, lung, breast, gynecologic, kidney, bladder, melanoma, pancreatic, hepatocellular, gastric and haematologic. Then, PubMed and Web of Science were searched for relevant articles up to February 2024. Hence, we listed existing clinical trials about natural products used in the treatment of cancers and discussed the preclinical and clinical studies of some promising natural products and their targets, indications, and underlying mechanisms of action. Our intent was to provide basic information to readers who are interested or majoring in natural products and obtain a deeper understanding of the progress and actions of natural product mechanisms of action.

            Main article text

            1. INTRODUCTION

            Cancer is a major public health problem worldwide. According to an estimate from the World Health Organization (WHO) in 2019, cancer ranks as the first or second leading cause of death in many countries [1]. In the past few decades, significant advances have been made in the treatment of cancer, such as surgical resection, chemotherapy, radiotherapy, immunotherapy, biotherapy, and molecular-targeted therapy [2]. However, current treatments are still not achieving the expected optimal outcomes. In recent years numerous natural products have been approved by the United States (US) Food and Drug Administration (FDA) for cancer treatment. Natural products refer to the compounds extracted and/or optimized from nature and have a wide range of sources, including plants, animals, marine organisms, and microorganisms. Natural products play an important role and have an irreplaceable status in drug development and design. Notably, there are > 10,000 species of Chinese materia medica with a one thousand year-long history of clinical use with a strong theoretical basis in Chinese Medicine. In the clinical setting Chinese medicine is sometimes used to treat cancer with an unexpected good curative effect but the results are difficult to explain and repeat. As such, the identification of the active ingredients in these Chinese materials not only contribute to the research and promotion of Chinese medicine but is also an important component of drug research in China. In contrast, natural products are an important source of dietary supplements and this industry is growing very rapidly, with annual sales now in the multi-billions of dollars [3]. In the US nearly one-half of patients begin taking new dietary supplements when they are diagnosed with cancer [4]. Although these dietary supplements are marketed and used by patients with cancer, dietary supplements have not been approved by the US FDA for preventing cancer, stopping cancer growth, or preventing cancer recurrence. Dietary supplements may help to improve health but may also be ineffective or risky. In the future the ongoing research involving natural products will facilitate the accumulation of substantial evidence to support the use of dietary supplements. Indeed, in recent years many natural products have been extensively investigated to cure different types of cancer (Figure 1). However, few studies have systematically collated and analyzed clinical trial results with respect to the use of natural products for the treatment of cancer. In this paper we comprehensively collected and summarized existing clinical trial data about natural products used in the treatment of cancer and discussed the preclinical and clinical studies of some promising natural products, the targets, indications for use, and detailed mechanisms of action (Figure 2). This review is intended to provide basic information to readers who are interested or majoring in natural products and obtain a deeper understanding of the progress and mechanisms of actions underlying natural products.

            Next follows the figure caption
            Figure 1 |

            Schematic representation of the typical natural products in different types of cancers. This figure is created with BioRender.com.

            Next follows the figure caption
            Figure 2 |

            The chemical structure of typical natural products.

            2. COLORECTAL CANCER

            Colorectal cancer (CRC) is the second most significant cause of cancer-related mortality worldwide [5, 6]. Currently, surgical resection, chemotherapy, and adjuvant therapy are common treatment strategies for CRC. Nevertheless, side effects, such as cytotoxicity and resistance, limit utilization of these treatment modalities. There is a clear need to develop new drugs and/or new therapeutic combinations for CRC patients. Table 1 lists the preclinical studies involving natural products and their derivatives in the context of CRC. Table 2 further clarifies the clinical trials involving natural products in different types of cancers.

            Table 1 |

            Preclinical studies of natural products and their derivatives in colorectal cancer.

            CompoundsSourceExperiments
            Effects and mechanismsRef.
            In vivo In vitro
            Curcumin Curcuma longa L.Xenograft NOD/SCID mice model for SW620-Luc2HCT116, RKO, SW48, CCD18Co, SW620-Luc2ROS↑, KEAP1↑, NRF2↑, ARE↑, miR-34a/b/c↑, apoptosis↑, senescence↑; EMT↓, metastasis↓[10]
            Xenograft BALB/c nude mice for CT26CT26BAX↑, cleaved caspase 3↑, granzyme B↑, IL-6↑, IL-1β↑; NF-κB↓, PD-L1↓; immunomodulation[12]
            Xenograft BALB/c nude mice for CL-188CL-188, DLD-1Tumor necrosis↑; Notch1↓, TGF-β↓, proliferation↓, metastasis↓[13]
            Xenograft nude mice for SW620SW620Apoptosis↑; proliferation↓, p-ERK↓, STAT1↓, L1↓; enhancing the anti-cancer effect of 5-FU[14]
            Analogue DMC-BHXenograft nude mice for HCT116 and HT-29HCT116, HT29Apoptosis↑; PI3K↓, AKT↓, mTOR↓; proliferation↓, invasion↓[11]
            Analogue WZ26Xenograft BALB/c nude mice for HCT116HCT116, RKOROS↑, JNK↑, DNA damage↑, cisplatin-induced cell death↑; TrxR1↓[15]
            ResveratrolGrapes, peanuts, blueberries, and Reynoutria japonica (-)HCT116 WT, HCT116 p53-/-, MRC-5p53↑, p21↑, BAX↑, cytochrome C↑, cleaved caspase 3↑, apoptosis↑; Sirt-1↓[17]
            (-)HCT116, 5-FU-resistant HCT116R, MRC-5Caspase 3↑, chemosensitisation to 5-FU↑; β1-integrin receptors↓, NF-κB↓, VEGF↓, HIF-1α↓, CD44↓, CD133↓, ALDH1↓[22]
            (-)Caco2, CEM/ADR5000caspase 3/8/6/9↑; P-gp↓, MRP1↓, BCRP↓, CYP3A4↓, GST↓, hPXR↓[23]
            Analogue DMU-218(-)DLD-1, LOVOCaspase 3, Smac/Diablo, Fadd, Hsp60↑; Hsp27, Bcl-2, Bcl-xL↓; inducing cell cycle arrest and apoptosis[19]
            Analogue CS(-)HCT116, CCD-112p53↑, p21↑, Fas death receptor↑, FADD↑, caspase 3/8/9↑, cleaved PARP↑, cell cycle arrest↑, apoptosis↑[18]
            Oxyresveratrol(-)HT29, HCT116miR-3612↑, E-cadherin↑; snail↓, miR-3687↓, miR-301a-3p↓, migration↓[21]
            3-β-D-glucoside of trans-resveratrol(-)HT29, SW480, Caco2, HUVECIL-10↓, IL-8↓, E-selectin↓, VCAM-1↓, producing anti-proliferative and pro-apoptotic effects[20]
            QuercetinOnions, asparagus, red leaf lettuce, and Ginkgo biloba L.Xenograft BALB/c nude mice for RKORKO, SW480, HCT116E-cadherin↑, p38↑, p-JNK↑, p-ERK↑; N-cadherin↓, vimentin↓[29]
            (-)HCT116, HT29Sprouty2↑, PTEN↑, SFRP1↑, ZBTB10↑; Sp1↓, miR-27a↓, miR-23a↓, miR-24-2↓[30]
            DMH-exposed Wistar rats(-)BAX↑, PARP↑, APC↑; Bcl-2↓, β-catenin↓[31]
            APCMin/+ mice(-)Wnt/β-catenin pathway inhibition[32]
            (-)HT29ESR2↑, GPR30 genes↑; inducing cell cycle arrest in the G0/G1 phase, inhibiting BPA-exposed HT29 cells viability[33]
            Xenograft BALB/c nude mice for HCT116HCT116Oxaliplatin-induced ROS production↑, oxaliplatin-mediated proliferation suppression↑; GSH↓[34]
            (-)COLO 320, MCF7, 3T3-L1Caspase 3↑, cell death↑, mitochondrial depolarization↑; controlling the growth of cells, arresting cell cycle[35]
            RutinHedyotis diffusa Wild(-)HT29BAX↑, p53↑, caspases 3/8/9↑, MAPK↑; NF-κB↓, IKK-ɑ↓, IKK-β↓[36]
            GenisteinSoy(-)HT29, SW620H2O2↑, oxidative stress↑, inflammation↑; cell viability↓[39]
            Cannabidiol Cannabis sativa L.Xenograft C57BL/6 mice for MC38MC38M1-like macrophages↑; M2-like macrophages↓, PI3K/AKT signaling↓[43]
            (-)HT29, SW480, HCT116, HCT15Cleaved caspase 3↑, BAX↑, p-p53↑, ATF4↑, CHOP↑, endoplasmic reticulum stress↑, Atg7↑, p-Beclin-1↑, LC3-II↑, p-JNK↑, p-p38↑, p-ERK↑; Bcl-xL↓, IAP-1↓, survivin↓, Alix↓[44]
            Xenograft SCID mice for HCT116 p53wt and HCT116 p53−/− HCT116 p53wt, LS174T p53wt), HCT116 p53−/−, SW480 p53mutROS↑, cleaved PARP1↑, p21↑; CDK2↓; inducing autophagy, Hsp70 and KEAP1-NRF2 signaling pathway[45]
            (-)LoVo, SW480MTs↑, MT2A↑[46]
            (-)HCT116, SW480, SW620, Caco2, CCD18COcleavage PARP↑, BiP↑, IRE1α↑, eIF2α↑, ATF3↑, ATF4↑, cell arrest↑, apoptosis↑, endoplasmic reticulum stress↑; cyclin D1↓, cyclin D3↓, CDK2↓, CDK4↓, CDK6↓; cell viability↓[47]
            EGCGTeaXenograft BALB/c nude mice for SW480; azoxymethane-induced and dextran sulfate sodium-promoted mouse modelHCT116, SW480, HCT15, HT29, LoVo, Caco2, HEK293ESE-1↑[53]
            (-)HCT116, RKO, SW620, HEK293TYAP↑, CTGF↑, CYR61↑, ABCB1↑, ABCC1↑, vimentin↑, Slug↑; LATS1/2↓, p-LATS1/2↓; the activation of YAP promoted proliferation, EMT and drug exportation transporters[55]
            (-)HT29, SW480, HCT15, HCT116, Caco2NOX1↓, MMP-2↓, MMP-9↓; inhibiting the activation of the EGFR and downstream of NF-κB, AKT, and ERK1/2[54]
            (-)CT-26Plasminogen↑; tetranectin↓; proliferation↓[51]
            Andrographolide Andrographis paniculata Xenograft nude mice for HCT116/5-FURHCT116, HCT116/5-FURBAX↑, PARP↑, caspase 3↑[62]
            Xenograft BALB/c mice for CT26CT26IFN-γ↑, FasL↑, perforin↑, Granzyme B↑; COX2↓, PGE2↓;[63]
            (-)HT29, HCT15, HCT116IRE-1↑, PERK↑, ATF6↑; VEGFR1↓, FoxM1↓, PTTG1↓;[60]
            Xenograft 002019-NU/J nude mice for HT29HT29, HCT15, HCT116ROS↑; β-catenin↓, cyclin D1↓, c-Myc↓, TCF4↓, LGR5↓, LEF1↓, Axin-1↓, mitochondrial membrane potential↓, ATP level↓[61]
            Analogue AGS-30Xenograft nude mice for HT29HT29, HCT116,ROS↑, p-JNK1/2↑, cleaved caspase 3/9↑, cleaved PARP↑[57]
            Transgenic zebrafish model, rat aortic ring model, mouse Matrigel plug model, and xenograft nude mice for HT29HT29, HUVECsVEGF↓, p-ERK1/2↓, p-AKT↓, p-VEGFR2↓, p-mTOR↓, p-MEK1/2↓, p-p38↓, inhibiting blood vessel formation, AKT/mTOR and ERK-dependent pathways, as well as VEGF signaling[59]
            Table 2 |

            Clinical trials of natural products in different types of cancers.

            Types of cancersNatural productsGroupClinical status/Study typeStudy populationSample sizeExperiments resultsRef.
            Colorectal cancerLipid carrier containing curcuminCurcumin plus bevacizumab/FOLFIRI (folinic acid, 5-FU, irinotecan)A prospective, observational, single-group analysisColorectal cancer patients44OS 30.7 months, median PFS 12.8 months, none of the patients achieved a CR, 9 patients achieved a PR, presenting comparable long-term survival outcomes with acceptable toxicity outcomes[7]
            CurcuminCurcumin plus FOLFOX (folinic acid, 5-FU, oxaliplatin) or FOLFOX groupA phase IIa TrialColorectal cancer patients28CXCL1, PFS (-); OS↑[8]
            Curcumin plus anthocyanin or placebo groupA randomized, double-blind, placebo-controlled, phase II presurgical trialColorectal adenomas patients35IGF-1, 25OHD, IL-10, IL-6, TNFα, IGFBP-3, Leptin, Adiponectin, L/A, hs-CRP, HOMA-index (-); did not directly modulate inflammatory and metabolic biomarkers[9]
            GenisteinGenistein plus FOLFOX or genistein plus FOLFOX–bevacizumabA phase I/II pilot studyMetastatic colorectal cancer patients13BOR, 61.5%; median PFS, 11.5 months[40]
            EGCGGTE group or placebo groupA single-center prospective randomized open-labelled studyPatients with complete removal of colorectal adenomas by endoscopic polypectomy176The number of relapsed adenomas↓; body mass index, dietary intake, serum lipid profiles, fasting serum glucose, and serum C-reactive protein level (-)[49]
            GTE (standardized to contain 150 mg EGCG) group or placebo groupA prospective, randomized, double-blind, placebo-controlled, multicentre trial50 and 80 years and had ≥1 histologically confirmed colorectal adenoma(s) removed within 6 months1001The GTE and placebo groups did not have a statistically significant difference in the adenoma rate and adverse events[50]
            Lung cancerEGCGProphylactic EGCG or therapeutic EGCG group after the occurrence of esophagitis or conventional therapy groupA prospective, three-arm, randomized trialMedically inoperable stage IIIA or IIIB or limited stage small cell lung cancer patients83Adjusted esophagitis index (AEI)↓, pain index (API)↓, dysphagia index (ADI)↓[84]
            EGCG or conventional therapy groupThe 5-year survival analysis of phase II studyLung cancer patients83Objective response rate (ORR) was higher in the EGCG group; PFS (-), OS (-)[83]
            Genistein1.8-2.0 Gy radiation therapy, concurrent weekly paclitaxel/carboplatin, plus BIO 300 (cohort 1, 500 mg/d; cohort 2, 1000 mg/d; cohort 3, 1500 mg/d)A open-label, single-arm, ascending dose phase Ib/IIa studyStage II-IV NSCLC patients21Tumor response rate was 65%, CR rate was 20%, a dose-dependent decrease in the serum TGF-β1 level, no serious AEs or dose-limiting toxicities; low toxicity rates[101]
            GossypolGossypol plus docetaxel and cisplatin or docetaxel and cisplatin alone groupA randomized, double-blind, placebo-controlled studyAdvanced NSCLC patients with high APE1 expressio.62PFS (-), OS (-), serious adverse events (-)[106]
            Mistletoe Viscum album L. treatmentA real-world data studyLung cancer patients112Improved poor quality of life, especially in combination with radiation; pain, nausea, and vomiting were reduced[112]
            Viscum album L. plus chemotherapy or chemotherapy alone groupA non-controlled, non-randomized observational multicenter cohort studyStage IV NSCLC patients158Median survival (17.0 months vs. 8 months), OS prolonged, 1- and 3-year OS rates were greater[113]
            Viscum album extract (Helixor-M) was instilled via pleural catheterA prospective observational studyLung cancer with malignant pleural effusions patients52Recurrence rate, 81% (42/52); 1-month recurrence rate, 48% (20/42); adverse events included pain medication, 13 (25%); fever > 38 °C, 8 (15%); oxygen demand, 6 (12%); pneumothorax, 2 (4%)[111]
            Breast cancerSulforaphaneSulforaphane supplement by dietary cruciferous vegetable intakeA randomized controlled trialWomen had abnormal mammogram findings and were scheduled for breast biopsy54Total cruciferous vegetable intake was inversely associated with Ki-67 protein expression; other biomarkers (HDAC3, HDAC6, H3K9, H3K18, and p21) had no correlation[118]
            ITC-rich broccoli sprout extract (BSE) (200 μmol ITC per day) or placebo groupA randomized pilot intervention studyPostmenopausal breast cancer patients30High compliance (100%) and low toxicity (no grade 4 adverse events), increased trend in cleaved caspase 3, decreased trend in Ki-67, NQO1, and ER-ɑ were observed but the differences were not statistically significant[119]
            CurcuminPaclitaxel plus curcumin or paclitaxel plus placeboA randomized, double-blind, placebo-controlled, parallel-group comparative clinical studyBreast cancer patients150ORR↑, reduced fatigue, the self-assessed overall physical performance was significantly higher[126]
            EGCGEGCG or placebo groupA double-blind, placebo-controlled, phase II randomized clinical trialBreast cancer patients receiving postoperative radiotherapy180The occurrence of grade 2 or worse RID was significantly lower in the EGCG group, RID index↓, symptom indexes↓, 4 patients (3.6%) had adverse events[138]
            Daily intake of GTE 1315 mg (EGCG 800 mg) or placebo groupA randomized, double-blinded, placebo-controlled phase II clinical trialHealthy postmenopausal women1075Both percent MD and absolute MD were not changed in all women; percent MD was decreased in younger women (50–55 years)[136]
            Daily intake of GTE 1315 mg (EGCG 800 mg) or placebo groupA placebo-controlled double-blind randomized clinical trialHealthy postmenopausal women1075Did not reduce sex steroid hormones; increased estradiol concentration[137]
            GenisteinIsoflavone (containing 137.5 mg genistein) or placebo groupA phase I double-blind clinical trialHealthy postmenopausal women30There were no significant changes in estrogenic effects, DNA strand breakage, apurinic/apyrimidinic sites, or apoptosis[146]
            Isoflavones (containing 11.57% genistein) or placebo groupA double-blind, randomized, placebo-controlled intervention studyBreast cancer patients (n=66) and high-risk women (n=29)95No statistically significant impact on mammographic density and fibroglandular tissue volume[147]
            TocotrienolStandard neoadjuvant treatment alone or in combination with delta-tocotrienolA open-label, randomized phase II trialNewly diagnosed, histologically verified breast cancer patients80There was no difference in OS, IDFS, and adverse events, and no association between ctDNA status and pathological treatment response[160]
            Tocotrienol-rich fraction (TRF) plus tamoxifen or placebo plus tamoxifenA double-blinded, placebo-controlled pilot trialBreast cancer patients with ER-positive tumors240There was no difference in BC-specific survival, disease-free survival, cancer recurrence; mortality was decreased but no statistical significant difference was detected[161]
            ArtesunateArtesunate dose group:100 mg/d, 150 mg/d, 200 mg/dOpen uncontrolled phase I StudyPatients with metastatic breast cancer13No major safety concerns[167]
            Artesunate administered at doses of 150-300 mg daily for 1.5 yearsA case reportPatients with metastatic breast cancer1The patient experienced a stabilization of her disease for 1.5 years, causing no or minimal side-effects[168]
            MistletoeTargeted therapy or targeted therapy plus Viscum album L.A real-world data observational cohort studyBreast and gynecological cancer patients242No adverse events, improving targeted therapy adherence, does not negatively alter the safety profile of targeted therapies[179]
            Two different European mistletoe extract groups or a control groupA prospective, randomized, open-label clinical trialBreast cancer patients95Fewer fever symptoms; improved pain and appetite loss scores; no significant differences in relapse or metastasis[180]
            Cervical cancer/Endometrial cancerCurcuminIDC, pembrolizumabAn investigator-initiated, non-randomized, open-label, multicohort, non-comparative, multisite, phase II studyCervical cancer patients (n = 18), endometrial cancer patients (n = 25)43irORR was 11.1% in cervical cancer and 12% in endometrial cancer, grade≥3 adverse events were 10 (55.6%) in cervical cancer patients and 9 (36.0%) in endometrial cancer patients[183]
            Cervical cancerBryostatin-1Bryostatin-1 and cisplatinA multi-institutional, prospective, single-arm trialMetastatic or recurrent cervical cancer patients1480% (8/10) patients had progressive disease and 20% (2/10) had stable disease; there were no treatment responses[191]
            Polyphenon EPolyphenon E or placebo groupA phase II randomized, double-blind, placebo-controlled trialWomen with persistent high-risk HPV infection and low-grade cervical intraepithelial neoplasia98Polyphenon E vs. placebo group: Progression, 6 (14.6%) vs. 3(7.7%; CR, 7 (17.1%) vs. 6 (14.6%)[192]
            Ovarian cancerBryostatin-1Bryostatin-1 in combination with cisplatinA multi-centered phase II studyRecurrent or persistent epithelial ovarian cancer patients8Median PFS (3 months) and median OS (8.2 months); all patients experienced grade 3 or 4 adverse events[196]
            Endometrial cancerCurcuminCurcumin phytosome orally for 2 weeksA open-label, non-randomized phase II studyEndometrial cancer patients7Minor immunomodulatory effects, a non-significant trend to improved quality of life[197]
            GenisteinDietary exposures were divided into quintiles for legumes, tofu, total isoflavones, glycitein, daidzein, and genistein for all women in the cohortA longitudinal multiethnic Cohort StudyNon-hysterectomized postmenopausal women46027A reduced risk of cancer was associated with the intake of total isoflavone, daidzein, or genistein (highest vs. lowest quintile)[200]
            Kidney cancerIsoquercetinAdministration of isoquercetin and sunitinibA phase I trialkidney cancer patients12A statistically significant improvement in fatigue score[207]
            Pancreatic cancerCurcuminGemcitabine and Meriva®, a patented preparation of curcumin complexed with phospholipidsA single centre, single arm prospective phase II trialPancreatic cancer patients52Response rate, 27.3%; cases with stable disease, 34.1%; total disease control rate, 61.4%, median PFS, 8.4 months, median OS, 10.2 months; grade 3/4 toxicity (neutropenia, 38.6%; anemia, 6.8%); no significant changes in quality of life[210]
            GenisteinAXP107-11 in combination with standard gemcitabine treatmentA phase Ib, single site, open-label, clinical studyPancreatic cancer patients16No hematologic or non-hematologic toxicity; median overall survival time, 4.9 months (range 1.5-19.5 months); 44% (7/16) survived longer > 6 months; 19% were alive at the 1-year follow-up evaluation[216]
            MelanomaSFNThree dosage groups (50, 100, or 200 μmol of oral sprout extract (SFN) daily for 28 daysA phase II studyMelanoma patients had at least 2 clinically atypical nevi ≥ 4 mm17No dose-limiting toxicities; the levels of SFN dose-dependent increase in plasma and skin, IP-10↓, MCP-1↓, MIG↓, MIP-1β↓[233]
            Multiple myelomaCurcuminMPC group (melphalan, prednisone, and curcumin) or MP group (melphalan, prednisone, and placebo)A randomized, single-blind & parallel design of the studyMultiple myeloma patients33Improving overall remission, NF-κB↓, VEGF↓, TNF-α ↓, and IL-6[253]
            Curcumin and piperineNewly diagnosed patients were divided into control or treatment group (curcumin/piperine); pCR patients were the chemotherapy groupA clinical trialNewly diagnosed MM (n=20) and pCR patients (n=12)32OCT-4A↓, NANOG↓,SOX2↓, cell cycle S and G0/G1 phases↓, cell cycle G2/M phases↑, inhibition of multiple myeloma CSC proliferation[258]
            Hepatocellular carcinomaHuaier granuleHuaier granule or non-huaier treatmentA multicentre, randomised, controlled, phase IV trialAfter curative resection of hepatocellular carcinoma patients1044Prolonging RFS; reducing extrahepatic recurrence[260]
            Huaier granule or non-huaier treatmentA propensity score analysisAfter curative resection of hepatocellular carcinoma patients1111Prolonging overall survival >5 years[261]
            Huaier granule or non-huaier treatmentA single-center cohort studyHepatocellular carcinoma patients826Improving OS rate[262]
            SRL-based therapy or tacrolimus-based therapyA single center experienceHCC patients who underwent liver transplantation (LT)36α-fetoprotein↓, FoxP3+ Treg↓, CD8+/CD3+ T cells↑, longer recurrence and survival times[263]

            Complete response (CR), partial response (PR), best overall response rate (BOR), progression-free survival (PFS), overall survival (OS), objective response rate (ORR), invasive disease-free survival (IDFS), recurrence-free survival (RFS), complete response (pCR) 1 year after chemotherapy.

            2.1 Curcumin

            Curcumin is a natural polyphenolic compound isolated from Curcuma longa L. Ginger plant has been widely used clinically in Chinese medicine for many years. In recent years, numerous studies have demonstrated the potential of curcumin to modulate the development and progression of several cancers, including CRC. A prospective, observational, single-group analysis showed that curcumin in combination with bevacizumab/folinic acid, fluorouracil (5-FU), and irinotecan (FOLFIRI)in patients with CRC had comparable long-term survival outcomes with acceptable toxicity outcomes (NCT02439385) [7]. Another double-group phase IIa trial showed that curcumin combined with folinic acid, 5-FU, and oxaliplatin (FOLFOX)) chemotherapy exhibited a longer overall survival (OS) than the FOLFOX alone group (NCT01490996) [8]. However, in patients with colorectal adenomas undergoing surgery, the administration of a combination of curcumin and anthocyanin did not directly modulate inflammatory and metabolic biomarkers [9]. In CRC cell and animal studies, curcumin was shown to activate the ROS/KEAP1/NRF2/miR-34a/b/c cascade, thereby suppressing epithelial-mesenchymal transition (EMT) and metastasis, and inducing apoptosis and senescence [10]. The curcumin analog, DMC-BH, has also shown promising anti-tumor effects via inactivation of the PI3K/AKT/mTOR signaling pathway [11]. The synergistic anti-tumor effects of curcumin have also attracted much attention. For example, curcumin enhanced the therapeutic efficacy of radiotherapy and augmented the radiotherapy-induced abscopal effect in mice with CRC by acting as an immunomodulator [12]. Similarly, the combination of curcumin and luteolin synergistically inhibited colon cancer by reducing the Notch1 and TGF-β signaling pathways [13]. Remarkably, a low concentration of curcumin potentiated the anti-cancer effect of 5-FU against CRC, which inhibited p-ERK, STAT1, and L1 expression [14]. Curcuminoid WZ26 augments the anti-cancer effect of cisplatin by regulating the TrxR1/ROS/JNK pathway [15]. These results further indicated that curcumin is a promising compound for the treatment of CRC. However, the use of curcumin in the pharmaceutical field has been hindered due to its bioavailability and stability. Therefore, it is essential to explore better curcumin derivatives or new dosage forms.

            2.2 Resveratrol

            Resveratrol is a non-flavonoid polyphenol that is widely distributed in the leaves and skins of grapes. Resveratrol has been marketed as a dietary supplement and commonly used to treat high cholesterol, cancer, and heart disease but there is no substantial scientific evidence to support these application. Of importance, the mechanism underlying the resveratrol effect in cancer prevention and management has garnered increasing attention [16]. Brockmueller et al. [17] verified that higher resveratrol concentrations in vitro boosts CRC cell apoptosis by mediating a negative regulatory loop between p53 and Sirt-1. To overcome the low bioavailability of resveratrol, researchers have conducted related studies on resveratrol analogues. For example, resveratrol derivatives (CS and DMU-281) exert a marked anti-cancer activity on CRC cells, effectively inducing apoptosis and cell cycle arrest [18, 19]. Similarly, the 3-β-D-glucoside of trans-resveratrol exerts anti-proliferative and pro-apoptotic effects on CRC cells by regulating modulation of the tumor microenvironment [20]. Additionally, Lin et al. [21] reported that oxyresveratrol inhibits CRC cell migration by regulating EMT and microRNA (miRNAs). Resveratrol also has an important role in overcoming drug resistance in CRC. For example, resveratrol increases the chemosensitivity of CRC cells to 5-FU by targeting the β1-integrin/HIF-1α axis [22]. Moreover, resveratrol inhibits drug-related metabolic enzyme (CYP3A4 and glutathione-S-transferase [GST]) activity [23]. Previously, we reported that PD-1/PD-L1 interaction blockade is an effective cancer immunotherapy [2427] but the response rate of patients to anti-PD-1/PD-L1 immunotherapy may be affected by PD-L1 expression. Thus, identifying drugs that modulate PD-L1 expression is warranted. Notably, Lucas et al. [28] showed that resveratrol plus piceatannol upregulated the expression of PD-L1 in CRC cells. These results indicated that resveratrol may be a potential compound in cancer immunotherapy that may expand indications and enhance the efficiency of cancer vaccines.

            2.3 Quercetin

            Quercetin is a natural flavonoid molecule frequently found in fruits and vegetables (e.g., onions). Quercetin is also marketed as a dietary supplement used for heart and blood vessels, cancer, arthritis, bladder infections and diabetes but there is a lack of scientific evidence to support most of these uses. A randomized clinical trial is underway to assess the efficacy of sulindac, curcumin, rutin, and quercetin in preventing colon cancer but the results have not been announced (NCT00003365). Mechanistic studies showed that quercetin inhibits the migration and invasion of colon cancer cells by regulating the JNK signaling pathway [29]. Fosso et al. [30] further found that quercetin exhibits anti-proliferative and pro-apoptotic effects by counteracting the Sp1-miR-27a axis in CRC cells. It is known that 1,2-dimethyl hydrazine (DMH) is a cogent environmental toxicant. In a DMH-exposed colon cancer rat model, quercetin was shown to ameliorate ROS formation, inflammation, and hyperproliferation by targeting the adenomatous polyposis coli (APC) and β-catenin pathways [31]. This finding is consistent with the results from Benito et al. [32], who demonstrated that micro-encapsulated probiotics in combination with quercetin inhibit the development of CRC in APCMin/+ mice by inhibition of the Wnt/β-catenin signaling pathway. Similarly, recent studies have reported that bisphenol A (BPA) may have toxic effects on the body and colon. Quercetin and its fermented extract exhibited excellent inhibitory effect on the viability of BPA-exposed HT29 colon cancer cells [33]. Additionally, quercetin enhanced the chemotherapeutic impact of oxaliplatin in HCT116 cells by increasing intracellular ROS through its suppressive action on glutathione levels [34]. Moreover, amalgamation of quercetin with anastrozole and capecitabine induce activation of caspase 3 expression in vitro [35]. These results indicate that quercetin is a promising chemotherapeutic intervention in the treatment of CRC.

            2.4 Rutin

            Rutin, which is extracted from Hedyotis diffusa Wild, has also been a focus of CRC treatment. Nafees et al. [36] showed that rutin in combination with silibinin produces synergistic anti-cancer effects on HT29 CRC cells by regulating expression of apoptosis, inflammation, and MAPK pathway proteins. Recently, a new colonic delivery method for rutin was studied by formulating rutin into frankincense-based compression-coated tablets and showed that the anti-cancer effect was improved [37]. Interestingly, network pharmacology and analysis of gene biological information showed that the potential intracellular signaling pathways of rutin in human SW480 cells may be associated with miRNAs-long noncoding RNAs (lncRNAs)-messenger RNAs-transcription factors [38]. Further research is required to validate the anti-cancer effect of rutin on CRC in vivo.

            2.5 Genistein

            Genistein, a polyphenolic isoflavone compound, is abundantly present in soy or soy-based products. People residing in Asian countries usually have a high intake of genistein in the daily diet because many traditional foods are made from soybeans [39]. Recently, the anti-cancer activity of genistein has undergone extensive research. A phase I/II pilot study showed that genistein in combination with FOLFOX or FOLFOX–bevacizumab is safe and well-tolerated [40]. Moreover, genistein in combination with FOLFOX or FOLFOX–bevacizumab enhanced response rates and progression-free survival (PFS) in the treatment of metastatic CRC (NCT01985763) [40]. Another nested case-control study determined that high plasma levels of isoflavones (genistein and daidzein) are associated with a decreased risk of CRC [41]. Alorda-Clara et al. [39] demonstrated that high concentrations of genistein decrease cell viability depending on the regulation of oxidative stress and inflammation in colon cancer cells. Network pharmacology and bioinformatics analyses further uncovered the molecular mechanisms underlying genistein against CRC [42]. The potential drug targets associated with autophagy, including epidermal growth factor receptor (EGFR) and estrogen receptor (ER) [42]. Hence, genistein is a promising natural compound that may become a drug and supplement in cancer treatment. While some researchers have studied the anti-cancer activity of genistein, the precise mechanism by which genistein might prevent cancer has not been established.

            2.6 Cannabidiol

            Cannabidiol (CBD), which is isolated from the Cannabis sativa L., has been approved as a drug in the US and is utilized for controlling seizures among patients with Lennox-Gastaut syndrome. Cin recent years several clinical trials have attempted to determine whether CBD provides benefits to patients with CRC and breast cancer (BC) in recent years (NCT03607643; NCT04398446; NCT04482244; NCT05016349; NCT04754399). Single-cell analyses revealed that CBD inhibits alternative activation of macrophages and shifts metabolic processes by inhibiting the PI3K-AKT pathway, which rewires the tumor microenvironment and restores the intrinsic anti-tumor properties of macrophages. Accordingly, CBD enhances the response of anti-PD-1 immunotherapy to CRC in vivo [43]. Cannabidiol was shown to induce apoptosis and autophagy in vitro by activating the JNK/p38/ERK MAPK pathway [44]. Further research revealed that CBD-induced ROS production is dependent on the p53 pathway. An Hsp70 inhibitor was shown to contribute to shifting CBD-induced autophagy toward apoptosis, which improved programmed tumor cell death in p53wt CRC cells [45]. Additionally, metallothionein family proteins, as regulators of zinc, have an important role in facilitating the anti-cancer effects of CBD [46]. CBD-induced cycle cell arrest and apoptosis are dependent on the activation of cannabinoid receptor type 2 (CB2) but not cannabinoid receptor type 1 (CB1) [47]. A corollary study should determine the impact of CBD in combination with another chemotherapy agent in animal and clinical trials.

            2.7 Epigallocatechin-3-gallate (EGGG)

            Tea, as one of the most widely consumed beverages worldwide, is generally thought to have health benefits. Most studies involving tea and cancer prevention have focused on green tea. EGCG is the most active and abundant catechins in green tea. The other catechins include epigallocatechin (EGC), epicatechin-3-gallate (ECG), and epicatechin. The catechins have the capacity to scavenge free radicals, which may protect cells from ROS-induced DNA damage [48]. At present the chemoprophylaxis effects of EGCG or green tea extract (GTE) in CRC patients are receiving attention in clinical trials (NCT02891538; NCT01239095; NCT01360320; NCT02321969). One clinical trial reported that a GTE supplement decreased the number of relapsed adenomas and exhibited a favorable outcome for the prevention of colorectal adenomas (NCT02321969) [49]. Conversely, another randomized, placebo-controlled clinical trial reported that GTE is well-tolerated but no statistically significant difference was detected in the adenoma rate (NCT01360320) [5052]. Additional mechanistic studies showed that EGCG was an activator of epithelial-specific E26 transformation-specific sequence (ETS) transcription factor-1 (ESE-1), and overexpression of ESE-1 suppressed tumor development and cell migration/invasion [53]. Another study reported that the ECG and EGCG dimers inhibit CRC cell invasion and metastasis by downregulating MMP-2 and MMP-9 expression via a NOX1/EGFR-dependent mechanism [54]. Moreover, Iram et. al [51] demonstrated that EGCG suppressed CRC cell proliferation by targeting tetranectin. Interestingly, the activation of Yes associated protein (YAP) protein induced by EGCG may impede the anti-tumor effect of EGCG [55]. Although EGCG has been shown to inhibit CRC tumorigenesis in laboratory and animal studies, the results of human epidemiologic and clinical studies have been inconclusive.

            2.8 Andrographolide

            Andrographolide is a natural product from the traditional Chinese medicine, Andrographis paniculata. In the past several decades, researchers have focused on the potential role of andrographolide and its derivatives in inflammatory diseases and cancer treatment [56]. However, a phase II clinical trial investigating the efficacy and safety of andrographolide plus capecitabine in CRC patients was terminated due to a low accrual rate (NCT01993472). Nevertheless, several researchers have synthesized a series of andrographolide derivatives that exhibit favorable anti-cancer effects [57, 58]. Specifically, the andrographolide derivative, AGS-30, was shown to induce apoptosis in CRC cells by activating the ROS-dependent JNK signaling pathway [57]. Another study showed that AGS-30 suppresss tumor growth and angiogenesis by inhibiting AKT/mTOR and ERK-dependent pathways, as well as interrupting VEGF signaling [59]. Alternatively, a series of studies demonstrated that andrographolide and melatonin had a synergistic effect on promoting the death of CRC cells by inducing endoplasmic reticulum stress and suppressing angiogenic activity [60]. Similarly, the combined action synergized to inhibit the colospheroid phenotype by targeting the Wnt/β-catenin signaling pathway [61]. We previously demonstrated andrographolide reversed 5-FU resistance in human CRC by elevating BAX expression [62]. Moreover, andrographolide potentiated PD-1 blockade immunotherapy by inhibiting COX2-mediated PGE2 release [63]. Overall, these findings indicate that andrographolide and its derivatives inhibit the development of CRC in vivo and in vitro. A clinical trial of andrographolide derivatives merits further investigation. We look forward to more experiments to determine the role of andrographolide in regulating the immunologic microenvironment and immunosuppression in CRC [64]. Natural products have an important role in the treatment of CRC, of which quercetin, resveratrol, and CBD have been marketed as drugs and supplements. Mechanistic studies have shown that quercetin, resveratrol, and CBD hinder the progression of CRC via multiple signaling pathways. Well-designed, rigorous clinical studies are needed to optimize dosage, formulations, and the appropriate target population.

            3. LUNG CANCER

            Lung cancer is the most common cancer and the leading cause of cancer-related deaths worldwide [65]. Lung cancer is categorized as small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC) based on the main histotype, prognosis, and therapy. Although significant progress has been made in the diagnosis and treatment of lung cancer in recent years, the prognosis for patients remains unsatisfactory. Efforts are currently underway to examine natural products for treating the possibility of lung cancer, many of which have proceeded to clinical trials (Table 3).

            Table 3 |

            Preclinical studies of natural products and their derivatives in lung cancer.

            CompoundsSourceExperiments
            Effects and mechanismsRef.
            In vivo In vitro
            SulforaphaneBroccoli, cauliflower(-)A549, SK-1p-ERK1/2↑, claudin-7↑; claudin-5↓; SFN-Cys inhibited invasion via microtubule-mediated claudin dysfunction, SFN-NAC inhibited invasion via microtubule-mediated inhibition of autolysosome formation[67]
            (-)A549, SK-1Proteasomes↑, LC3 II/I↑; FASN↓, ACACA↓, ACLY↓, SREBP1↓, Bnip3↓, NIX↓, α/β-tubulin↓[68]
            Xenograft BALB/c nude mice for CSE-transformed HBEHBEIL-6↓, ΔNp63α↓, Notch↓, Hes1↓, CD133↓, Nanog↓[70]
            (-)A549p53↑, cleaved caspase 3↑, cleaved PARP↑, apoptosis↑, cell cycle arrest↑, ROS↑; survivin↓, cell growth↓[71]
            (-)A549, taxol-resistant A549p-ERK1/2↑; βIII-tubulin↓, XIAP↓, tau↓, stathmin1↓, α-tubulin↓[72]
            Xenograft BALB/c nude mice for CL1-5; 24 lung tissues samples of lung cancer patientsCL1-0, CL1-5γH2AX↑, ROS↑[73]
            Curcumin Curcuma longa L.32 specimens of lung cancer tissue and the corresponding healthy tissuesH1299, A549TAp63α↑, E-cadherin↑, ZO-1↑; miR-19↓, vimentin↓, N-cadherin↓[78]
            (-)A549, HepG2Caspase 3↑, apoptosis↑; p-PI3K↓, AKT↓, proliferation↓, migration↓[80]
            (-)A549, docetaxel/vincristine-resistant A549p-p38↑, p-ERK↑, cleaved PARP↑, p-eIF2α↑, ROS↑, apoptosis↑[79]
            Analogue EF24Xenograft BALB/c nude mice for A549A549, SPC-A1, H460 and H520ROS↑, apoptosis↑, mitochondrial fssion↑, autophagy↑; proliferation↓[75]
            Analogue EB30(-)A549, NCI-H292, HBEAIF↑, BAX↑, cleaved PARP↑, cleaved caspase-3/9↑, p-ERK↑, p-90RSK↑, apoptosis↑, cell cycle arrest↑, ROS↑; Bcl-2↓, Bcl-xL↓, p-AKT↓, p-P70S6K↓, proliferation↓, migration↓[74]
            Analogue MS13(-)NCI-H520, NCI-H23, MRC-9Caspase 3↑, anti-proliferative activity↑, apoptosis↑; Bcl-2↓[76]
            Curcumin mono-carbonyl analog 2cXenograft BALB/c nude mice for NCI–H460NCI–H460, HepG2, HT-1080, A549, MRC-5BAX↑, caspase-3/9↑; GPX4↓, Bcl-2↓, TrxR↓; inducing ROS-dependent apoptosis and ferroptosis[77]
            Diarylheptanoid 35dXenograft SCID mice for HCC827H460, H1299, H1650, HCC827, H1975Hsp70↑, bag3↑, p62↑; EGFR↓[81]
            Derivative 35dXenograft SCID mice for HCC827HCC827CPTI↑, CPTII↑, Hsp60↑, long-chain acylcarnitines↑, mitochondrial dysfunction↑; short-chain acylcarnitines↓, Tom20↓, fatty acyl metabolism↓[82]
            EGCGTeaMulti-dose urethane-induced C57BL/6J mice lung tumorigenesis with high-fat dietA549, RAW264.7STAT1↓, SLC7A11↓; apoptosis↑, ferroptosis↑, leptin-induced proliferation↓, invasion↓, migration↓[91]
            Xenograft BALB/c nude mice for NCI-H1975 or AREGFR-mutant NCI-H1975, H1975 AR cell, HEK293AMPK pathway↑, mitochondrial membrane depolarization↑, apoptosis↑; AKT/mTOR and MAPK/ERK pathways↓, altering cancer energy metabolism[92]
            Chlorogenic acidGreen coffee, apples, artichoke, and Lonicera japonica ThunbXenograft BALB/c nude mice for A549A549cIAP1↓, cIAP2↓, proliferation↓, inhibiting the binding of ANXA2 to p50[93]
            Xenograft NOD/SCID mice for Huh7 and H446NCI-H446, Huh7, Bel-7402, HEK293T, HCT116, U87MG, M059J, CCC-HEL-1, NCI-H358, WI-38, MRC-5, A549-5FU, SK-LU-1, MIHA, iPS, HH, rat C6 and mouse G422 glioma cellsSUMO1↑, p21↑, Tuj1↑, GFAP↑, p53↑, KHSRP↑, cell differentiation↑; miR-17 family↓, p-c-Myc↓, EPCAM↓, CD44↓, proliferation rate↓, migration↓, invasion↓, mitochondrial ATP production↓[94]
            Xenograft BALB/c nude mice for LAC-1(-)IL-6↓, alleviating cancer-related neurological complications[95]
            Genisteinsoy(-)BEAS-2BReducing ROS and DNA damage, upregulating p-NRF2 nuclear translocation and catalase activity[104]
            Xenograft BALB/c nude mice for A549H292, A549, 16HBEApoptosis↑; circ_0031250↓, miR-873-5p↓, FOXM1↓, cell viability↓, proliferation↓ migration↓, invasion↓[102]
            (-)A549, 95DFOXO3a↑, PUMA↑, ROS↑, cytochrome c↑, BAX↑; mitochondrial membrane potential↓, mitochondrial activity↓, Bcl-2↓[103]
            GossypolCotton seeds (Gossypium herbaceum L.)(-)A549, NCI H460LC3-II↑, ROS↑, autophagy↑, apoptosis↑; p62↓[108]
            (-)H1975 (EGFR L858R/T790 M), H441, A549Apoptosis↑; YAP↓, TAZ↓, p-EGFR↓, p-ERK1/2↓; cell growth↓[109]
            Gossypol acetateXenograft BALB/c nude mice for A549HEK-293FT, H1944, H1299, A549, H460, H23E2F1↓, LRPPRC↓, CDK6↓; suppressing cell cycle G1/S transition, oxidative phosphorylation, cancer stem cells[107]
            Mistletoe Viscum album L.Xenograft NU/J nude mice for H82H82, H69, H196, SHP77Apoptosis↑; cleaved PARP↑, C-myc↓, N-myc↓, cell growth↓[114]
            3.1 Sulforaphane

            Sulforaphane (SFN) is a primary isothiocyanate (ITC) derived from cruciferous vegetables, especially broccoli and cauliflower [66]. SFN is metabolized into SFN-N-acetyl-cysteine (SFN-NAC) and SFN-cysteine (SFN-Cys) in the blood. A mechanistic study showed that SFN-Cys inhibits NSCLC cell invasion by microtubule-mediated claudin dysfunction, while SFN-NAC inhibits invasion by microtubule-mediated inhibition of autolysosome formation [67]. Similarly, SFN triggers NSCLC cell apoptosis by downregulating fatty acid synthase and inhibiting microtubule-mediated mitophagy [68, 69]. SFN intervention suppresses tobacco smoke-induced cancer stem cell (CSC)-like properties of human bronchial epithelial (HBE) cells via the IL-6/ΔNp63α/Notch axis in vivo, [70]. The synergistic anti-tumor effects of SFN with other drugs have also attracted much attention. SFN in combination with the allyl, ITC, produces superior protective effects against carcinogenesis [71]. SFN metabolites decrease the resistance of NSCLC cells to paclitaxel in usual doses by activating caspase 3-induced microtubule disruption [72]. A higher level of EGFR showed resistance to SFN induced ROS-mediated apoptosis [73]. These findings indicate that the antitumor effects of SFN and its metabolites involved in multiple targets and pathways. A future study may determine SFN function in the lung cancer immunologic microenvironment.

            3.2 Curcumin

            Accumulating evidence has shown that curcumin has anti-cancer effects by inhibiting lung cancer cell growth and inducing apoptosis [7478]. Xie et al. [78] demonstrated that curcumin suppresses tobacco smoke-induced EMT progress by increasing TAp63α expression and reducing miR-19 expression. Recently, a series of curcumin derivatives (EF24, EB30, and MS13) were synthesized by researchers and showed excellent anti-cancer effects on lung cancer. The underlying mechanism includes suppression of the PI3K/AKT pathway, activation of ERK1/2, and accumulation of ROS [7476]. Another curcumin analog (2c) has been designed that exhibits selective ROS generation compared to normal cells through the inhibition of intracellular TrxR, thereby mitigating side effects and improving the safety profile [77]. In addition, existing evidence indicates that curcumin promotes apoptosis of chemoresistant lung cancer cells via ROS-regulated p38 MAPK phosporylation [79]. A triple combination (curcumin, thymoquinone, and 3, 3′-diindolylmethane) better attenuates lung and liver cancer progression compared to a double combination [80]. Moreover, diarylheptanoid 35d was shown to overcome EGFR tailored tyrosine kinase inhibitor (TKI) resistance in EGFR-mutant lung adenocarcinoma by inducing Hsp70-mediated lysosomal degradation of EGFR [81]. Similarly, Hsieh et al. [82] further demonstrated that the curcumin derivative, 35d, enhanced the sensitivity of osimertinib (a TKI inhibitor) to lung cancer cells by disrupting fatty acyl metabolism and inducing mitochondrial stress. These results from in vivo and in vitro experiments are encouraging and we anticipate publication of additional clinical outcomes involving curcumin and its analogues.

            3.3 EGCG

            Accumulating evidence reveals that EGCG has a strong protective effect against lung cancer. Several clinical studies showed that EGCG is a radioprotective agent, prevents radiation-induced esophagitis, and enhances objective response rate (ORR) but without significantly prolonging PFS and OS in lung cancer patients [83, 84]. A number of in vitro and in vivo studies have confirmed that EGCG inhibits CSC-like properties and self-renewal ability of lung cancer [8588]. Moreover, EGCG has been shown to inhibit lung cancer cell proliferation and migration by suppressing EGFR signaling [8990]. Alternatively, Li et al. [91] reported that EGCG alleviates obesity-exacerbated lung cancer progression. The underlying mechanism is associated with the STAT1/SLC7A11-mediated ferroptosis pathway and gut microbiota. EGCG also has a critical role in overcoming drug resistance in tumors. According to Zhou et al. [92], EGCG circumvents drug-induced resistance in NSCLC by modulating glucose metabolism and the AMPK/AKT/MAPK axis. Overall, EGCG may be a potential anti-cancer agent. The potential research direction in the future may involve examining the role of EGCG in regulating the lung cancer immunologic microenvironment.

            In addition, polyphenon E serves as a standardized, highly reproducible green tea polyphenol mixture containing 60% EGCG. Polyphenon E is the recommended form of green tea for clinical chemoprevention trials and has entered multiple phase I/II lung cancer clinical studies (NCT00363805; NCT00707252). Currently, a limited number of studies have been conducted to determine the anti-cancer mechanism underlying polyphenon E in lung cancer. The components of green tea are complex and the study of single component EGCG or multiple components of polyphenol E are necessary, which are more conducive to understand the mechanism underlying the green tea anti-cancer effect.

            3.4 Chlorogenic acid

            Chlorogenic acid (CGA) is a phenolic acid that is widely distributed in green coffee, apples, artichoke, and Lonicera japonica Thunb. Green coffee is used for obesity, diabetes, high blood pressure, and high cholesterol but there remains a lack of good scientific evidence. Recently, a phase I/II study was conducted to investigate the safety and efficacy of injected anCGA in the treatment of advanced lung cancer (NCT03751592). A mechanistic study verified that CGA inhibits binding of annexin A2 (ANXA2) to p50 and attenuates related anti-apoptotic genes, thereby suppressing the proliferation of A549 in vivo and vitro [93]. It is known that inducing cancer differentiation is a promising approach to treat cancer. Further studies demonstrated that CGA treatment prevents the development of lung cancer by inducing lung cancer differentiation [94]. Alternatively, orally administered dietary Ilex paraguariensis led to a dose-dependent brain accumulation of CGA and quercetin in mice with lung adenocarcinoma, in different neuroprotective effects occurred in the telencephalon and diencephalon [95]. These findings support plant-based strategies to improve prognosis in lung cancer.

            3.5 Soy isoflavone and genistein

            Soy isoflavone, a type of flavonoid, is classified as a plant estrogen extracted from soybeans, which consists of genistein, daidzein, and glycitein. Chei et al. [96, 97] conducted a pooled analysis of four cohort studies from Japan and China that revealed a significant association between higher intake of isoflavone and soy food and a reduced risk of lung cancer among individuals who have never smoked. These findings were consistent with several meta-analyses that concluded higher intake of soy and soy isoflavone are inversely associated with the risk of lung cancer incidence and mortality [98100]. Moreover, genistein, as one of the predominant soy isoflavones, has also entered clinical trials (NCT01628471; NCT00769990; NCT00533949). A multicenter phase Ib/IIa clinical trial demonstrated that the radioprotectant, BIO 300 oral suspension (synthetic genistein nanosuspension), is an effective radioprotector for patients with NSCLC receiving concurrent chemoradiotherapy. BIO 300 oral suspension exhibits low toxicity rates, along with the pharmacodynamic results and high tumor response rates [101]. Additionally, Yu et al. [102] found that genistein induced apoptosis and repressed NSCLC progression by inhibiting circ_0031250/miR-873-5p/FOXM1axis. Chan et al. [103] found that genistein induced mitochondrial apoptosis and FOXO3a/PUMA expression in NSCLC cells. Interestingly, genistein and procyanidin B2 alleviate carcinogen-induced ROS and DNA damage by activating NRF2/ARE signaling in normal bronchial epithelial cells, which provided a research basis for cancer prevention through dietary modifications [104]. Hence, soy is considered to be a healthy diet component, but soy isoflavone and genistein as dietary supplements need additional supportive clinical data.

            3.6 Gossypol

            Gossypol is a natural polyphenolic compound extracted from cotton seeds, roots, and stems [105]. Use of gossypol acetate, as a gynecologic medicine, was discontinued owing to serious side effects but gossypol acetate is currently receiving attention again. Recently, a randomized, double-blind, placebo-controlled study showed that gossypol in combination with docetaxel and cisplatin was well-tolerated and had a better median PFS and OS than the control group. Because there were no significant differences between the gossypol, docetaxel, and cisplatin group and the control group, future studies with larger sample sizes should be conducted [106]. In the past few years gossypol has made significant progress in improving chemoradiotherapeutic sensitivity. For example, gossypol acetate was shown to enhance the sensitivity of CDK4/6 inhibitors (cancer therapeutic drugs) by inhibiting the LRPPRC-CDK6 loop in lung cancer cells. Gossypol acetate is considered to be a degrader of LRPPRC [107]. Gossypol can also enhance the cytotoxic effect of sorafenib in lung cancer by promoting autophagy and apoptosis [108]. In addition, gossypol overcomes TKI resistance in EGFR L858R/T790 mutation NSCLC cells by inhibiting YAP/TAZ and EGFR [109]. These findings suggest that gossypol is a potential natural product for the clinical management of lung cancer.

            3.7 Mistletoe

            European mistletoe (Santalaceae: Viscum album L.), commonly known as mistletoe, is frequently prescribed as an unconventional cancer therapy in central Europe [110]. Mistletoe is a universal name for various species of semi-parasitic plants which grow on host trees, such as apple, elm, oak, and pine [111]. A real-world data questionnaire-based study assessed the poor quality of life for lung cancer at first diagnosis and 12 months later. The results showed that add-on Viscum album L. therapy improved poor quality of life in lung cancer patients, especially when paired with radiation therapy. Specifically, the occurrence of pain, nausea, and vomiting decreased remarkably [112]. This finding is in agreement with Lee et al. [111] who showed that Viscum album extract is an effective and tolerable procedure for controlling malignant pleural effusions (MPEs) in lung cancer patients. Of note, another real-world observational multicenter analysis demonstrated that Viscum album L. in combination with chemotherapy significantly enhanced OS for stage IV NSCLC patients [113]. It was further shown that mistletoe lectin inhibits the growth of Myc-amplified SCLC [114]. Although mistletoe has entered clinical trials for the treatment of lung cancer patients, the anti-lung cancer mechanism is still unclear and more preclinical trials are needed to better define the tumor microenvironment. In conclusion, natural products with the unique advantages of multi-ingredient, multi-target, multi-pathway therapy have been widely investigated in lung cancer. In the absence of clinically ideal drugs for the prevention and treatment of lung cancer, it is a potential direction to explore natural products to prevent or delay the process of lung cancer, and dietary supplements are a good adjuvant treatment.

            4. BREAST CANCER

            BC is the most common cancer among women with a high incidence and prevalence. The four subtypes of BC are widely recognized: luminal A; luminal B; human epidermal growth factor (HER2)-positive; and triple negative [115]. Although treatment options have rapidly developed in recent years, relapse and metastasis remain the main causes of BC death. The advances in natural products, particularly in the field of cancer therapy, provide a promising research direction for the treatment of BC (Table 4).

            Table 4 |

            Preclinical studies of natural products and their derivatives in breast cancer.

            CompoundsSourceExperiments
            Effects and mechanismsRef.
            In vivo In vitro
            SulforaphaneBroccoli, cauliflowerXenograft BALB/c nude mice for MCF7MCF7Sulfate-related metabolites↑, glutathione-related metabolites↑, genus Lactobacillus↑; tryptophan metabolites↓, methyl-purine metabolites↓, bacterium Desulfovibrio↓, inhibiting the activation of the AHR pathway and influencing microbial diversity[120]
            C3(1)-SV40 Tag (FVB-Tg(C3-TAg) cJeg/JegJ) (C3) mice, FVB/N-Tg (MMTVneu) 202NK1/J-(HER2/neu) miceER(-) BC cell lines (MDA-MB-157 and MDA-MB-231), ER(+) BC cell lines (MCF7), MCF10ADNMT3B↑, p16↑, p53↑, MYC↑; DNMT3A↓, HDAC1↓, HDAC6↓, KAT2A↓, EZH2↓, HDAC1↓, HDAC3↓, HDAC8↓, tumor growth↓[123]
            (-)MDA-MB-231, MDA-MB-157paxillin↓, IQGAP1↓, FAK↓, PAK2↓, ROCK↓, p-ERK↓, p-MEK↓, inhibiting actin stress fiber formation[121]
            Xenograft BALB/c nude mice for 4T14T1NRF2↑, HO-1↑, GCLC↑; COX-2↓, PGE2↓; increasing cytotoxic CD8+ T cells[122]
            Curcumin Curcuma longa L.(-)MCF7Vimentin↓, α-SMA↓, fibronectin↓, CXCL12↓, CXCR4↓, NF-κB↓, SHH↓[130]
            (-)MCF7 (ER-positive), MDA-MB-231MAPKAPK3, AKT3, CDK5, IGF1R, and MAPK11 may be potential therapeutic targets of curcumin[135]
            Analogue PAC(-)MCF7, MDA-MB-231BAX↑, apoptosis↑; Bcl-2↓, migration↓, modulating the DNA repair gene expression[131]
            EGCGTea(-)ADMSC, MDA-MB-231p-smad2↓, p-NF-κB↓, snail↓, slug↓, CCL2↓, CCL5↓, CXCL8↓, IL-1β↓, IL-6↓, VEGFα↓, HIF-1α↓, COX2↓, IDO↓[141]
            Normal and breast cancer tissues, xenograft BALB/c nude mice for MDAMB-231 cellsT47D, MCF7, SKBR3, MDA-MB453, UACC-812, MDA-MB-231, BT-549, MDA-MB-468Cytoplasmic YAP1 inhibition of cell proliferation and promotion of autophagic death; YAP1 promotion of autophagosome formation, autophagic death, assembly of the ESCRT-III complex subunits (CHMP2B and VPS4B) in the cytoplasm[144]
            C3(1)-SV40 Tag (FVB-Tg(C3-1-TAg) cJeg/JegJ) (C3), FVB/N-Tg (MMTVneu)202NK1/J (HER2/neu)ER(-) BC cell lines (MDA-MB-157 and MDA-MB-231), ER(+) BC cell line (MCF7)p16↑, p53↑, MYC↑; BMI1↓, HDAC1↓, HDAC3↓, DNMT3A↓, HDAC3↓, changing the expression of tumor- and epigenetic-related proteins in C3 mice, preventing ER-negative mammary cancer in transgenic mice[123]
            Xenograft nude mice for MCF7/ADRMCF7/ADR, MCF7, H9c2ATP↑, SOD↑, TNF-α↑, iFN-γ↑; IL-10↓, P-gp↓, HMGB1↓, MDA↓,[145]
            GenisteinsoyPreclinical patient-derived xenograft orthotopic mouse models(-)Cd74↓, Lp1↓, Sat1↓, Tap63↓, Dnmt3b↓, Tet2↓, Hdac2↓, Dnmts↓, changing genome-wide transcriptomic, DNA methylation, multiple epigenetic-related genes[148]
            (-)MCF7, T47DHER2↑, p-ERK1/2↑, p-EZH2(Ser21)↑, IL-6↑, IL-8↑; H3K27me3↓[150]
            (-)MCF7, BJHigh concentrations of genistein (50 μM and higher) destroyed MCF7 BC cells, but it harmful to dermal fibroblasts at longer exposure times (48 h)[151]
            (-)HFF-1Cleaved PARP↑, ER-β↑, androgen receptor↑; cyclin E↓, pro-caspase 7↓, Aromatase↓, ER-α↓[149]
            Cannabidiol Cannabis sativa L.Xenograft nude mice for MDAMB-231 cellsTNBC, MCF10A, MDA-MB-468, MDAMB-231Caspase 9↑, GADD45-α↑, p-p38↑, p-p53↑; integrin-α5↓, integrin-β5↓, integrin-β1↓, fbronectin↓, vimentin↓, Beclin1↓, LOX↓[153]
            (-)MCF7, MDA-MB-231, T47D, SK-BR-3, MCF10A, HUVECsVHL↑, HIF-1α↓, Src↓, proliferation↓, invasion↓, migration↓; suppressing angiogenesis and stem cell-like properties↓[152]
            (-)MDA-MB-231CBD at low threshold concentration (5 μM) protected BC cells; above this concentration, CBD formed aggregates, anti-proliferation, arrested the cell cycle, and triggered autophagy; at higher doses, CBD caused bubbling cell death[158]
            Xenograft nude mice for MCF7-derived malignant phenotype (6D cells)(-)p53↑; Ki67↓, Bcl2↓, CB1↓, p-AKT↓, BIRC3↓, ∆NP63α↓[157]
            (-)MCF7Lactate dehydrogenase↑, apoptosis↑; ATP↓[154]
            (-)MCF7aro, HFF-1Caspase 7/8/9↑, AR↑, apoptosis↑; ER-α↓, aromatase↓, EGR3↓, ERK1/2↓[155]
            TocotrienolPalm oil, rice bran, oat, wheat germ, barley and rye(-)MDA-MB-231, MCF7Cytochrome C↑, cleaved PARP-1↑, BAX/Bcl-2↑, cleaved caspase 3↑; p-PI3K↓, p-GSK-3↓[159]
            (-)Doxorubicin resistant MCF7mdr1↓, P-gp↓, inhibition of NF-κB activation and reduction of NF-κB transcriptional activity[163]
            (-)MDA-MB-231Reducing lipid droplet biogenesis; potentiating lipophagy[164]
            Xenograft BALB/c nude mice for 4TI4T1MIG-6↑, Cadherin 13↑, CD4+/CD127+ ↑, CD4+/CD25+↓, CD4+/CD25+/CD73+↓[165]
            Artesunate Artemisia annua L.Xenograft BALB/c nude mice for 4TIL-929, 4 T1MMP-9↓, MMP-2↓, MMP-14↓, FAP↓, fibronectin↓, vimentin↓, α-SMA↓, S100A4↓, p-Smad3↓,TGF-β1↓[169]
            A mammary hyperplasia modelMCF10ABAX↑; p-AKT↓, p-NF-κB↓, COX-2↓, PCNA↓, proliferation↓, inflammation↓, fibrosis↓[173]
            (-)MCF7, HeLa, HCT116, HepG2, A549, MCF7/ADR, A549/TAX, A549/DDPp62↑, LC3↑; m-cathepsin L↓, m-cathepsin D↓, inducing autophagosome accumulation but decreasing the formation of autolysosomes, inhibiting lysosomal function[172]
            Xenograft BALB/c nude mice for 4TI4T1Cleaved caspase3↑, cleaved PARP↑, p-H2AX↑, p-p38↑; MMPs↓, uPA↓ p-STAT3↓, FOXM1↓, RAD51↓, survivin↓[181]
            (-)MCF7, 4T1, RAW 264.7Artesunate influenced cell viability, ROS production, cell cycle arrest, and inflammatory responses in a fever-range hyperthermia-dependent manner[182]
            4.1 Sulforaphane

            In recent years the anti-BC effects of SFN have been widely investigated in preclinical and clinical trials [116, 117]. A clinical trial showed that dietary cruciferous vegetable intake (SFN supplement) is inversely associated with Ki-67 protein (a biomarker of cell proliferation) expression among women scheduled for breast biopsies. This finding indicated that SFN supplements from cruciferous vegetables have potent anti-BC activity (NCT00843167) [118]. Similarly, another randomized pilot intervention study found that ITC-rich broccoli sprout extract supplement has high compliance (100%) and low toxicity (no grade 4 adverse event) rates in BC patients. Large differences in biomarker expression of NQO1, ER-α, ER-β, and cleaved caspase 3 were observed but the differences were not statistically significant due to small sample sizes (NCT01753908) [119]. A mechanistic study demonstrated that SFN influences metabolism and methylation, alters the diversity of gut microbiota, and indirectly affects the activation of aryl hydrocarbon receptor (AHR) by tryptophan metabolism, thereby inhibiting the progression of BC [120]. Moreover, SFN suppresses TGF-β1-induced migration and invasion by inhibiting the RAF/MEK/ERK pathway in BC cells [121]. Alternatively, the adjuvant administration of SFN enhances the therapeutic effects of doxorubicin against BC. This finding might be explained by blocking myeloid-derived suppressor cell (MDSC) accumulation and suppressive activities, increasing in cytotoxic CD8+ T cells [122]. Strikingly, Li et al. [123] verified that paternal exposure to combined SFN and EGCG synergistically increases tumor latency and prevents the growth of ER(-) BC in control-fed female offspring. These results indicated that SFN shows great promise as a cancer-fighting treatment in BC. Although SFN is not currently being marketed as a dietary supplement, SFN remains a promising natural product, the bioavailability and stability of which need further optimization.

            4.2 Curcumin

            Several recent studies have reported that curcumin is used for the treatment of BC [124, 125]. For example, a randomized, placebo-controlled clinical study reported that outcomes in BC patients treated with curcumin in combination with paclitaxel was superior to paclitaxel-placebo, exhibiting a higher ORR, improved physical performance, and reduced fatigue following a 12-week treatment period [126]. Additionally, curcumin was shown to reduce radiation-induced dermatitis (RID) in BC patients compared to placebo-treated groups, although this result has not been published (NCT01042938). Extensive in vitro and animal experiments have verified that curcumin and its analogs exhibit inhibitory effects on the proliferation, migration, and invasion of BC cells. These actions involve various molecular targets, including the lncRNA HCG11/Sp1 axis, hedgehog/Gli1 signaling pathway, and TGF-β signaling inhibition [127129]. Furthermore, Jang et al. [130] reported that curcumin remodels the microenvironment of BC by interrupting a positive feedback loop between adipose-derived mesenchymal stem and cancer cells. This finding reflected inhibition of the CXCL12/CXCR4 axis [130]. The curcumin analog, PAC, was shown to inhibit BC by modulating DNA repair pathway gene expression [131]. Of note, a series of studies have demonstrated that curcumin has a pivotal role in augmenting the therapeutic efficiency of chemotherapy drugs in BC [132134]. Additional high-throughput assays were performed to investigate the potential of MAPKAK3, p-AKT3, CDK5, IGF1R, and MAPK11 as prognostic markers and therapeutic targets for curcumin in treating BC patients [135]. Validation of these findings is necessary both in vitro and in vivo.

            4.3 EGCG

            A growing body of research has underscored the prospective role of EGCG or GTE, particularly genistein, in reducing the risk of BC. For example, a clinical trial has shown that supplementation with GTE (containing 64% EGCG) among healthy postmenopausal women did not exhibit significant effects on reduction of mammographic density (MD). However, supplementation with GTE did result in an elevation of circulating estradiol concentrations. Notably, a reduction in percent MD was observed among younger women [50-55 years of age] (NCT00917735) [136, 137]. Another double-blind, placebo-controlled, phase II randomized clinical trial revealed that topical EGCG solution is safe for external use on the skin and reduces the occurrence of RID compared to placebo groups (NCT02580279) [138]. Hence, GTE or EGCG may have a complex influence on the progression of BC, which requires more preclinical and clinical evidence. Some researchers have confirmed that EGCG exhibits suppressive effects on tumor growth in BC through diverse mechanisms. This effect encompasses attenuating MDSCs-mediated immunosuppression, halting the acquisition of cancer-associated adipocyte (CAA)-like phenotype, or inhibiting the expression of proline dehydrogenase [139141]. Both green tea extract and EGCG inhibit BC cell migration [142, 143]. Guo et al. [144] reported that EGCG effectively maintains YAP1 within the cytoplasm, enabling assembly of the ESCRT-III complex and ultimately stimulating autophagic apoptosis in BC cells. Alternatively, polyethylene glycol-doxorubicin/EGCG/folic acid inhibits the expression of P-glycoprotein (P-gp) and reverses the multidrug resistance (MDR) of BC cells, thereby enhancing therapeutic efficacy of doxorubicin [145]. Remarkably, paternal consumption of combined botanicals (SFN or EGCG-rich) contribute to the prevention of ER-negative mammary cancer in transgenic mice [123]. Therefore, natural products as beneficial dietary components may facilitate cancer prevention.

            4.4 Genistein

            Recently, a phase I clinical trial reported that soy isoflavone, at a dose of 900 mg/day, is safe and well-tolerated among healthy postmenopausal women(NCT00099008) [146]. However, another intervention study concluded that a 12-month soy supplement did not exert a statistically significant impact on breast MRI fibroglandular tissue density or MD (NCT00290758; NCT01219075) [147]. Notably, the lifetime exposure to dietary genistein, starting at conception, decreases the risk of BC risk in mice by modulating epigenetic mechanisms [148]. Genistein was shown to enhance the anti-cancer properties of exemestane in vitro [149]. Paradoxically, long-term low-level genistein results in endocrine resistance in BC cells by suppressing H3K27 trimethylation, thus posing potential health risks [150]. Although genistein had high potential for use in the treatment of skin problems during and after BC treatment, genistein is not completely safe. High concentrations of genistein (≥50 μM) destroys MCF7 BC cells but is harmful to dermal skin fibroblasts at longer exposure times (48 h) [151]. These results indicate that genistein, as a partial agonist of ER-α, might be beneficial to tumor prevention under certain conditions but it may possess potential risks to BC patients in other cases, which may vary depending on the concentration, concurrent therapies, and BC subtype.

            4.5 Cannabidiol

            There have been several isolated reports on the anti-cancer function of CBD through distinct mechanisms. For example, Jo et al. [152] demonstrated that CBD inhibits angiogenesis and CSC properties of BC cells by regulating Src/VHL/HIF-1α signaling. Surapaneni et al. [153] discovered that CBD induces apoptosis, cell cycle G1 arrest, migration suppression, and doxorubicin sensitivity enhancement. The main mechanism of action for CBD involves activation of GADD45α/p38/p53, downregulation of integrin-α5, -β5, and -β1, and inhibition of autophagy [153]. A series of studies have highlighted the promising synergistic effects of combining CBD with exemestane, rimonabant, or photodynamic therapy in vitro in improving the treatment of BC. These findings endorse the therapeutic potential of CBD for cancer treatment [154156]. Furthermore, CBD has been shown to effectively block in vivo development of BC [157]. Interestingly, D’Aloia et al. [158] found that the effect of CBD on cultured BC cells depended on a specific threshold concentration. At doses above this threshold, CBD exhibits a potent cytotoxic effect, ultimately inducing the bubbling cell death [158]. CBD not only exhibits anti-cancer effects on BC but may also improve chemotherapy-mediated complications and decrease drug resistance. We expect more clinical evidence to support the application of CBD in BC patients.

            4.6 Tocotrienol

            Vitamin E, a liposoluble micronutrient, is categorized into subgroups of tocopherols and tocotrienols, each containing four isoforms: α; β; γ; and δ. In general, tocotrienols are abundant in palm oil, rice bran, oats, wheat germ, barley, and rye, whereas tocopherols are mainly present in vegetable oils, such as corn, olive, and sunflower oils [159]. At present, the safety and effectiveness of tocotrienols in the treatment of BC are being evaluated in clinical trials (NCT04496492; NCT03855423). Regrettably, a recent clinical trial concluded that δ-tocotrienol does not improve the effectiveness of neoadjuvant BC treatment or mitigate the incidence of adverse effects (NCT02909751) [160]. This finding is consistent with the results from Nesaretnam et al. [161], who demonstrated that adjuvant tocotrienol therapy does not have an impact on BC-specific survival in women with early BC (NCT01157026). It is important to note that accruing evidence suggest the anti-cancer properties of tocotrienols in vitro. For example, based on label-free quantitative proteomics analysis, γ-tocotrienol is thought to be a potential proteasome inhibitor, the inhibitory action of which contributes to the induction of apoptosis [162]. In addition, γ-tocotrienol reverses MDR in doxorubicin-resistant BC cells by regulating the NF-κB-P-gp axis [163]. Moreover, omega 3-docosahexaenoic acid (DHA) together with δ-tocotrienol decreases lipid droplet biogenesis and potentiates lipophagy lipid droplets, possibly exerting a beneficial effect on inhibiting BC malignancy [164]. Conversely, spirulina (Arthrospira) platensis plus γ-tocotrienol does not show any synergistic anti-cancer effects in vivo. Compared to combination groups, the γ-tocotrienol alone group induced more necrotic cells based on histopathologic analysis [165]. Interestingly, Idriss et al. [159] reported that β-tocotrienol exhibits more cytotoxic effects than γ-tocotrienol on BC cells and β-tocotrienol induces apoptosis via a P53-independent PI3K dependent pathway. These preclinical investigations suggest that tocotrienol may be a promising anti-cancer agent for BC. Furthermore, the four isoforms of tocotrienols may have distinct roles in the treatment of cancer. Future research, including additional animal studies and larger clinical trials, is necessary to fully evaluate the therapeutic potential of tocotrienols in BC.

            4.7 Artesunate

            Artesunate is a semi-synthetic derivative of the Chinese herb, Artemisia annua L., that has an excellent safety profile in the treatment of malaria [166]. A recent phase I study demonstrated that chronic oral intake of artesunate (up to 200 mg/day [2.2-3.9 mg/kg/day]) over a period of 37 months did not elicit any significant safety concerns among patients with metastatic BC (NCT00764036) [167]. Another clinical case reported that in a patient with metastatic BC oral artesunate maintained disease stabilization for a duration of 1.5 years. Biomarker profiling revealed that carcinoembryonic antigen may be a potential target of artesunate [168]. A mechanistic study demonstrated that artesunate and dihydroartemisinin suppresses TGF-β signaling, subsequently inactivating cancer-associated fibroblasts, leading to inhibition of cancer growth and metastasis [169]. Other research showed that artesunate exhibits synergistic interactions with doxorubicin or TP-0903, a mesenchymal-epithelial transition and receptor tyrosine kinase inhibitor, and enhances the cytotoxic effect on BC cells [170, 171]. Recent evidence suggests a connection between enhanced lysosomal function and paclitaxel resistance in cancer cells. This resistance can be overcome with artesunate or other inhibitors of lysosomal function [172]. Simultaneously, artesunate was shown to effectively mitigate the proliferation of mammary hyperplasia by suppressing the NF-κB and AKT signaling pathways in a rat model of mammary hyperplasia [173]. Hence, artesunate is a potential natural product or dietary supplement. The effectiveness of artesunate in preventing BC needs to be supported by additional clinical data.

            4.8 Mistletoe

            Mistletoe, as a complementary and alternative medicine, has become increasingly popular among cancer patients [174176]. Accumulating evidence indicates that mistletoe may have a positive impact on the quality of life among patients with BC, such as improving fatigue, insomnia, and physical function [177, 178]. Moreover, a real-world data study demonstrated that add-on Viscum album L. did not negatively impact the safety profile of targeted therapies in breast and gynecologic cancer patients [179]. Similarly, other clinical trials revealed that mistletoe is safe and decreases chemotherapy side effects but does not influence the frequency of relapse and metastasis within 5 years [180]. Cellular and animal experiments have demonstrated that mistletoe induces apoptosis and inhibits metastasis by targeting the STAT3-FOXM1 pathway in BC [181]. Mistletoe exerts distinct effects on BC cells and macrophages, influencing cell viability, ROS generation, cell cycle arrest, and inflammation, all of which are modulated by changing in fever-range hyperthermia [182]. Although clinical trials have been conducted to investigate the potential of mistletoe in mitigating the side effects and enhancing anti-cancer effects, only a few numbers of animal studies have delved into the underlying molecular targets and mechanisms of its actions.

            5. GYNECOLOGIC TUMOR

            Gynecologic tumors are common tumors in women, affecting the health of women worldwide. Cervical, ovarian, and endometrial cancer (EC) are common gynecologic tumors. In recent years, natural products used in the treatment of gynecologic tumors have also received attention (Table 5).

            Table 5 |

            Preclinical studies of natural products and their derivatives in other types of cancers.

            Type of cancerCompoundsSourceExperiments
            Effects and mechanismsRef.
            In vivo In vitro
            Cervical cancerCurcumin Curcuma longa L.(-)HeLa, CaSkip53↑, p21↑, BAX↑, cleaved caspase 3↑, E-cadherin↑, Bcl-2↓, N-cadherin↓,vimentin↓, viral oncoproteins E6 and E7↓[186]
            The laying hen develops ovarian cancer spontaneously, diet supplemented with whole flax seed, defatted flax meal or flax oil(-)CYP1A1↑, 2MeOE2↑, p-p38↑, p-ERK1/2↑, CYP3A4↓, a whole flaxseed supplemented diet decreased the onset and severity of ovarian cancer[193]
            The laying hen develops ovarian cancer spontaneously, diet supplemented with whole flax seed, defatted flax meal or flax oilBG1, HeyC2, TOV112D2MeOE2↑, DHA↑, 2MeOE2 and DHA both have anti-angiogenic effects. 2MeOE2 has pro-apoptotic effects, anti-cancer actions of 2MeOE2 are dependent on p38-MAPK pathway[194]
            Analogue CLEFMAXenograft SCID mice for HCC827HCC827CPTI↑, CPTII↑, Hsp60↑, long-chain acylcarnitines↑, mitochondrial dysfunction↑; short-chain acylcarnitines↓, Tom20↓; disrupting the fatty acyl metabolism[187]
            Analogue EF-24(-)HeLa, SiHaMMP-9↓, p-p38↓, migration↓[188]
            Endometrial cancerCurcumin Curcuma longa L.Xenograft NOD-SCID mice for IshikawaIshikawa, HEC-1B, VEROSlit-2↑, ROS↑, apoptosis↑; SDF-1↓, CXCR4↓, MMP-2↓, MMP-9↓, CTGF↓, migration↓[198]
            Analogue CP41Xenograft BALB/c nude mice for HEC-1BAN3CA, HEC-1BH3F3A ↑,p-p38↑, p-ERK↑, p-JNK↑, BAX↑, cleaved caspase 3↑, ROS↑, mitochondrial impairment↑, endoplasmic reticulum stress↑; Bcl-2↓, proteasome↓[199]
            GenisteinSoyXenograft BALB/c nude mice for Ishikawa, EC tissues of 51 patients aged<40 yearsIshikawaPR↑, FOXO1↑, c-Jun↑, p-JNK↑, cleaved caspase 3↑, p-cdc2↑; p-histone H3↓,C/EBPβ↓, Ki-67↓, inducing cell cycle arrest in G2 and apoptosis[201]
            PterostilbeneBlueberries, grapesEC tissues of 51 patientsHTB-111, Ishikawacaspase 3/8/9↑, BCL2L14↑, miR-663b↓[203]
            Kidney cancerGenisteinSoy(-)786-O, ACHNCleaved caspase 3/8/9↑, BAX↑; CD133↓, CD44↓, ALDH1A1↓, Oct4↓, Nanog↓, PCNA↓, cyclin D1↓, Bcl-2↓, renal CSCs↓[206]
            Pancreatic cancerCurcumin Curcuma longa L.(-)PANC1, AsPC-1TFPI-2↑, E-cadherin↑; p-ERK↓, p-JNK↓, N-cadherin↓, vimentin↓[213]
            (-)p53Y220C mutant cancer line BxPC-3Caspase 3↑, stabilizing p53Y220C mutant and restore its function[215]
            (-)p53Y220C mutant cancer line BxPC-3Cleaved caspase 3/7↑, cleaved PARP↑, BAX↑; p-Bcl2↓, rescuing mutant p53Y220C[214]
            Analogue FN2(-)PANC1BAX↑, apoptosis↑; Bcl-2↓, p-p65↓, p-AKT↓, cell growth↓[208]
            Analogue C66(-)HPNE, BxPC3, SW1990 PANC1IL-1β↓, IL-6↓, IL-8↓, IL-15↓, TNF-α↓, COX2↓, p-JNK↓[212]
            GenisteinSoyXenograft NOD/SCID nude mice for genistein-resistant PaCa and PANC1 cellsPANC1, PaCaDEPTOR↑; ELK1↓; inhibiting PI3K/AKT/mTOR pathway, enhancing the sensitivity of PC cells to genistein[218]
            (-)MiaPaCa2 and PANC1, H6C7Cytosolic cytochrome c↑, cleaved caspase 3/9↑, ROS↑; STAT3↓, survivin↓, mitochondrial membrane potential↓, cyclin D1↓,ALDH1A1↓, cell viability↓; triggering cell cycle arrest in G0/G1 phase[219]
            Analogue AXP107-1Patient-derived xenograft BALB/c) nude mice modelMiaPaCa2, PANC1Cleaved PARP↑, cleaved caspase 3/9↑, GPER1↑; MUC1↓; reducing chemoresistance of gemcitabine[217]
            Cannabidiol Cannabis sativa L.Xenograft PAK1 wild type and knockout C57BL6 mice for murine PC cells TB33117PANC1, CFPAC1, Capan2, SW1990, HPAF11, MiaPaCa2, PSCsPD-L1↓, PAK1↓, pPAK1↓; inhibiting the proliferation of PC, PSC, and PSC-stimulated PC cells[220]
            Xenograft C57BL/6NTac mice for PANC02PANC02Enhancing radiation therapy treatment outcomes[221]
            (-)PANC1, MiaPaCa2CBD increased the chemosensitivity of gemcitabine and paclitaxel; oxygen-ozone enhanced the cytotoxicity of CBD; the combination of CBD with oxygen-ozone resulted in an upregulation of the CDKN2A, MAP2K1, and ERBB2 genes, a downregulation of the BRAF, RHOA, AKT1, AKT2, PIK3CB, PIK3CD, PIK3R1, and PIK3R2 genes[222]
            (-)PANC1, U266IL-2 determined the expression of CB2 receptor, CBD increased the cytotoxicity of CIK cells[223]
            Bladder cancerSFNBroccoli, cauliflower32 bladder tumors and 20 adjacent non-tumor tissue samples; nitrosamine-induced bladder tumor model in C57BL/6 miceRT4, T24, UMUC-3HK2↓, PDH↓, AKT1↓, p-AKT↓, N-PKM2↓ SFN inhibited ATP production by inhibiting glycolysis and mitochondrial oxidative phosphorylation (OXPHOS)[225]
            GenisteinSoy(-)SV-HUC-1E-cadherin↑; vimentin↓, Snail↓, Slug↓, CD44↓, Cyclin D1↓, PCNA↓, p-ERK↓, p-AKT↓, p-STAT3↓[230]
            MelanomaSFNBroccoli, cauliflowerThe zebrafish embryos were exposed tp SFN and phenylthioureaB16F10, A375Tyrosinase↑, melanin biosynthesis↑; MITF↑, PKCβ1↑, PCNA↓[234]
            (-)A375Anti-carcinogenic activities of sulforaphane are influenced by nerve growth factor[235]
            LeukemiasCurcumin Curcuma longa L.(-)JurkatCannabidiol, curcumin, and quercetin induced mitochondrial membrane potential loss and Ca2+ overload, curcumin, and quercetin caused direct mitochondrial uncoupling[237]
            (-)MOLT-4, BJThe combination of curcumin, genistein, resveratrol, and quercetin has cooperative anti-neoplastic activity without a significant effect on non-tumor cells[239]
            Xenograft BALB/c nude mice for HL-60HL60Cleaved caspase3↑, BAX↑, p27↑; HOTAIR↓, WT1↓,miR-20a-5p↓, Bcl-2↓[240]
            Xenograft NOD/SCID mice for MOLM13MOLM13, OCI-AML2, HL60ROS↑, mitochondrial dysfunction↑, p21↑, p-JNK↑, p-p38↑, cleaved caspase 3↑, cleaved PARP↑; CHK1↓, RAD51↓, Cyclin-D↓, XIAP↓, c-Myc↓; enhancing cytarabine sensitivity[241]
            LymphomasCurcumin Curcuma longa L.(-)CH12F3γH2AX↑, PARP1↑, PCNA↑, caspase 3/9↑, Rad51↓, inducing DNA breaks, sensitizing lymphoma cells to various DNA damage drugs[249]
            Xenograft BALB/c nude mice for RajiU937, RajiCleaved caspase 3↑, cleaved caspase 9↑, BAX↑, E-cadherin↑; p-Smad3↓, N-cadherin↓, Bcl-2↓[250]
            131 DLCBL tissue samples, xenograft BALB/C nude mice for RajiOCI-LY8, RajiCD59↓, pCREB↓, inhibiting the activation of NF-kB[251]
            Multiple myelomaGZ17-6.02 (isovanillin, harmine and curcumin) Curcuma longa L.(-)ALMC1, ANBL6, U266p-ATG13↑, p-S318↑,Beclin1↑, ATG5↑, BAK↑, BIM↑; Bcl-XL↓, MCL1↓, HDACs1/2/3↓, causing autophagosome formation and autophagic flux, regulating histone H3 acetylation and methylation[254]
            Cannabidiol Cannabis sativa L.(-)KMS-12 PE, U266CB2 receptor is highly expressed on CIK cells and MM cells[259]
            5.1 Cervical cancer
            5.1.1 Curcumin

            Recently, several clinical trials have commenced to assess the safety and effectiveness of curcumin in the treatment of cervical cancer, as well as its potential as a complementary medicine in conjunction with standard therapy (NCT02554344; NCT04294836). For example, a phase II study showed that the combination of pembrolizumab, stereotactic body radiotherapy, and low-dose cyclophosphamide, aspirin, lansoprazole, vitamin D, and curcumin (IDC) did not meet expected clinical activity in cervical and EC patients but some difficult-to-treat patients may have derived benefit from this therapeutic regimen, with durable responses (NCT03192059) [183]. Mechanistic studies found that curcumin exhibits a cytotoxic effect on cervical cancer cells in vitro through activation of p53 and p21 signaling [184, 185]. Similarly, in papillomavirus (HPV)-positive cervical cancer cells, curcumin suppresses proliferation and migration by directly targeting the viral oncoprotein E6 protein, subsequently activating p53 and p21 [186]. Interestingly, Lee et al. [187] discovered that the curcumin derivative, CLEFMA, triggers intrinsic and extrinsic apoptotic pathways by activating ERK1/2 and p38 signaling [187]. Conversely, EF24 suppresses cellular migration and MMP-9 expression by inhibiting p38 signaling [188]. While curcumin shows excellent anti-cancer effects based on preclinical evidence, more scientific evidence is required to determine whether curcumin can be used as a dietary supplement for the prevention or treatment of cervical cancer.

            5.1.2 Other natural products

            Bryostatin1 is a putative anti-cancer compound derived from marine animals [189]. However, current clinical trials have not met expectations [190]. Nezhat et al. [191] reported that bryostatin-1 and cisplatin combination therapy is not effective in metastatic or recurrent cervical cancer patients (NCT00005965). Likewise, the clinical evaluation of polyphenol E was also disappointing. According to Garcia [192], the administration of polyphenol E did not promote the clearance of persistent high-risk HPV infection and low-grade cervical intraepithelial neoplasia among women, compared to the placebo groups (NCT00303823). Therefore, the underlying mechanisms of these natural products is seldom mentioned in recent studies.

            5.2 Ovarian Cancer
            5.2.1 Flaxseed

            Flaxseed, isolated from Linum usitatissimum L., is composed of total fat (polyunsaturated, monounsaturated, and saturated fatty acids), carbohydrate (total dietary fiber and sugars), protein, water, minerals, vitamins, and lignans. In the laying hen a whole flaxseed supplemented diet decreased the onset and severity of spontaneous ovarian cancer. This flaxseed-enriched diet stimulated the systemic production of 2-methoxyestradiol (2MeOE 2) and DHA, thereby promoting apoptosis and decreasing angiogenesis in ovarian tumors but not in normal ovarian tissues [193, 194]. Another LC-MS/MS metabolomics approach also demonstrated that flaxseed increased animal lifespan and reduced ovarian cancer severity [195]. These findings indicate that flaxseed may be a healthy dietary habit to weaken ovarian cancer severity.

            5.2.2 Other natural products

            Although bryostatin-1 and squalamine have been investigated in clinical trials of ovarian cancer, these clinical trial results have not been published. Moreover, current clinical trials are experiencing disappointment. For example, Morgan et al. [196] observed that bryostatin-1 in combination with cisplatin treatment of patients with recurrent or persistent ovarian cancer had a modest response rate, but toxicities were insufferable and precluded tolerability. Consequently, these two natural products have rarely been mentioned in recent clinical and animal research pertaining to ovarian cancer.

            5.3 Endometrial Cancer
            5.3.1 Curcumin

            Recently, clinical investigations have been conducted on the potential therapeutic effect of curcumin for EC. Specifically, seven EC patients who consumed curcumin orally for 2 weeks exhibited minor immunomodulatory effects without a significant trend in quality of life improvement (NCT02017353) [197]. Curcumin was shown to inhibit cellular migration in vivo by inducing Slit-2 mediated downregulation of SDF-1 and CXCR4 [198]. Further research demonstrated that the curcumin analogue, CP41, induced apoptosis by activating the H3F3A/proteasome-MAPK signaling pathway and augmenting oxidative stress. Additionally, CP41 enhanced the sensitivity of EC to chemotherapeutic agents through mediation of H3F3A [199]. These findings suggest that curcumin holds considerable promise as a natural therapeutic agent for the treatment of EC.

            5.3.2 Genistein

            Previous research has revealed that soy isoflavone containing genistein, daidzein, and glycitein is safe and well-tolerated in healthy postmenopausal women. However, limited endometrial biopsy samples have prevented the trial from evaluating the effectiveness of isoflavone in preventing EC in this group (NCT00099008) [146]. Another multiethnic cohort study demonstrated that the highest intake of total isoflavones, daidzein, or genistein was associated with a decreased risk of EC in non-hysterectomized postmenopausal women [200]. Currently, there is a scarcity of prospective studies on overall or individual isoflavones, particularly genistein. Furthermore, immunohistochemistry analysis found that elevated progesterone receptor (PR) expression significantly increased rates of progression-free and OS among 31 young EC patients. Genistein was shown to increase long-time PR expression in vivo and inhibit cell proliferation in an ER-independent manner [201]. Although genistein has been assessed in different clinical trials, the precise mechanism by which genistein might help prevent EC has not been established. Additional laboratory and animal studies are clearly needed.

            5.3.3 Pterostilbene

            Pterostilbene, a resveratrol analog, is extracted mainly from blueberries and grapes. Existing evidence indicates that pterostilbene-mediated endometriotic cell apoptosis modulation has been confirmed to be more potent than resveratrol [202]. Of note, a clinical trial is presently underway to assess the effectiveness of megestrol acetate with or without pterostilbene in the treatment of patients with EC undergoing hysterectomy (NCT03671811). Furthermore, Wang et al. [203] concluded that EC patients with high miR-663b expression have a significantly poor prognosis. An in vitro study revealed that pterostilbene effectively suppresses cell viability and induces apoptosis via the miR-663b/BCL2L14 signaling pathway [203]. As a result, pterostilbene holds promising potential in the treatment of EC but further clinical and animal studies are imperative to validate the therapeutic effectiveness.

            In conclusion, there are relatively few clinical studies on natural products for use in gynecologic tumors. Notably, Chinese medicine treats some chronic gynecologic diseases with good curative effect, employing theories that include preventive measures, the reinforcement of vital qi to eliminate pathogenic factors, and the enhancement of bodily resistance. The theoretical guidance of Chinese medicine will greatly inspire the research of natural products. Targeting natural products may be a promising direction to enhance health management and prevention of gynecologic tumors.

            6. KIDNEY CANCER

            Kidney cancer is not a single disease. Kidney cancer represents several distinct types of cancer. Renal cell carcinoma (RCC) is the most common subtype, accounting for 85% of all cases [204]. In addition to surgery, there has been an explosion in target therapies that control tumor growth and the creation of blood vessels in the past decade [205]. In addition, the use of natural products for the treatment of kidney cancer has attracted attention.

            6.1 Genistein

            A recent clinical trial examined the impact of combining genistein with interleukin-2 (IL-2) in patients diagnosed with metastatic melanoma or kidney cancer (NCT00276835). Existing evidence demonstrated that genistein exhibits an interventional inhibitory effect on renal CSCs in vitro by suppressing the sonic hedgehog (Shh) pathway [206]. Although genistein has obtained attention in clinical trials of kidney cancer, mechanistic studies are lacking.

            6.2 Other natural products

            Sunitinib is recognized as the standard first-line therapy for advanced RCC. Nevertheless, sunitinib is associated with numerous side effects, one of which is fatigue, which occurs frequently. A recent phase I clinical trial has demonstrated that isoquercetin is safe and effective in reducing fatigue among kidney cancer patients receiving sunitinib treatment. This finding holds significant promise for improving the quality of life for these patients (NCT02446795) [207]. Other natural products contain shark cartilage (AE-941) and bryostatin 1. Shark cartilage has gradually lost interest among researchers. The anti-cancer effect of bryostatin 1 on kidney cancer has not been verified in vivo or in vitro. To date, natural product clinical trials in kidney cancer therapy are limited. It must be recalled that Chinese medicine has a thorough theoretical system with a focus on kidney disease in the ancient books of Yellow Emperors Classic of Medicine. Based on these rich theories and abundance of Chinese medicinal herbs, the search for compound Chinese medicine and monomers suitable for the treatment of kidney diseases and cancer has unique advantages.

            7. PANCREATIC CANCER

            Pancreatic cancer (PC) is a fatal malignant tumor with a 5-year survival rate of 5% [208]. PC has an extremely poor prognosis due to extensive local invasion, early systemic dissemination, and resistance to chemotherapeutic drugs. Surgery is the most effective treatment of PC, but 80%-85% of patients are diagnosed with advanced and unresectable disease [209]. Hence, considering natural products as a research direction is a promising breakthrough.

            7.1 Curcumin

            Presently, clinical trials exploring the therapeutic potential of curcumin in PC treatment have been conducted (NCT00192842, NCT00486460, NCT00094445). Of note, a prospective phase II trial has demonstrated that the phytosome complex of curcumin, as a complementary therapy in first-line therapy of advanced PC, had improved safety and efficacy compared to gemcitabine [210]. The curcumin analogue, FN2, also augmented the inhibitory effect of gemcitabine on PC cells [208, 211]. Another curcumin derivative, C66, has good anti-inflammatory activity. C66 was shown to inhibit the progression of PC cells via inhibition of JNK-mediated inflammation [212]. Curcumin inhibits ERK- and JNK-mediated EMT in vitro by upregulating TFPI-2, subsequently suppressing the migration and invasion of PC cells [213]. In addition, the p53Y220C mutation, a commonly identified variant in the p53 gene, results in the inactivation of the tumor suppressor protein, p53. This mutant p53Y220C is frequently observed in numerous tumors, including PC. Notably, curcumin has the potential to rescue mutant p53Y220C, thereby activating apoptosis [214, 215]. Therefore, curcumin possesses potential significance in the prevention and treatment of PC but additional clinical evidence is required to support its application.

            7.2 Genistein

            AXP107-1, a novel crystal form of genistein, has been demonstrated in a phase Ib clinical trial to be safe in patients with PC when used in combination with gemcitabine with no signs of hematologic or non-hematologic toxicity and with elevated serum levels [216]. The efficacy requires a larger sample size for verification. Of note, animal studies have verified that AXP107-1 in combination with gemcitabine has synergistic anti-cancer effects by activating GPER1 signaling [217]. In addition, Li et al. [218] showed that downregulation of ELK1 leads to upregulation of DEPTOR, which in turn enhances the sensitivity of PC cells to genistein through inhibition of mTOR signaling [218]. The anti-cancer effects of genistein in vitro are associated with induction of ROS-mediated mitochondrial apoptosis, cell cycle arrest, and regulation of STAT3 signaling [219]. In conclusion, these results predict that both curcumin and genistein are hot candidate adjuvant drugs for the treatment of PC. New delivery systems for genistein or curcumin contribute to improving bioavailability, stability, and application.

            7.3 Cannabidiol

            In recent years the treatment of CBD for PC has also attracted attention. For example, CBD was shown to inhibit the proliferation of PC, pancreatic stellate cell (PSC), and PSC-stimulated PC cells by targeting the P-21 activated kinase 1 (PAK1)-dependent pathway. Furthermore, CBD also inhibits the expression of PD-L1 via downregulating PAK1 activity, thereby potentiating the immune checkpoint blockade of PC [220]. Additionally, Alfonzetti et al. [221] demonstrated that CBD augments the sensitivity of radiotherapy in vivo and vitro. Likewise, the combination of CBD and oxygen-ozone exhibited a synergistic augmentation in cytotoxic effects on PDAC cell lines [222]. It is known that cytokine-induced killer cells (CIKs) are pivotal cytotoxic immunologic effector cells and have shown encouraging synergistic effects when combined with cancer-associated inhibitors and blockade. Specially, a low dose of CBD was shown to significantly enhance the cytotoxic function of CIKs without exerting any associated mediators in pancreatic and myeloma cells [223]. CBD treatment may help increase the therapeutic response for non-responder patients by activation of CIK therapy. CBD is a promising adjuvant therapy medicine that can enhance the effectiveness of chemoradiotherapy in PC through various pathways and mechanisms.

            8. BLADDER CANCER

            Bladder cancer is the 10th most common cancer type worldwide with a 5-year survival rate of approximately 70% [224]. Huang et al. [225] showed that patients with stage T1 bladder cancer have unique glucose metabolic abnormalities. SFN significantly downregulates glucose metabolism by blocking the AKT1/HK2 axis [225]. Alternatively, Wang et al. [226] confirmed that upregulation of FAT atypical cadherin 1 (FAT1) is associated with a lower survival rate of patients with bladder cancer. In vitro studies further demonstrated that SFN inhibit FAT1 expression, thereby suppressing the viability and metastasis, and promoting apoptosis of bladder cancer cells [226]. In addition, a series of studies have indicated that SFN has a crucial role in overcoming drug resistance, particularly in the case of resistance to cisplatin, gemcitabine, or everolimus [227229]. These laboratory and animal studies indicated that SFN has an important role in inhibiting the development of bladder cancer. However, the clinical trials of SFN or broccoli sprout extract in bladder cancer were terminated due to low accrual. Thus, a new delivery system for SFN may be necessary to improve bioavailability and decrease side effects, such as liposomes, cyclodextrin inclusion, and nanotechnologic approaches. Other natural compounds that have attracted attention in clinical trials for bladder cancer therapy include genistein and polyphenol E. Genistein has been shown to inhibit arsenic-induced EMT phenotype and CD44 expression in vitro by inhibiting HER2 phosphorylation [230]. Genistein and polyphenol E have received relatively little attention compared to SFN in vitro and in vivo.

            9. MELANOMA

            Melanoma is the most severe form of skin cancer [231]. The 5-year survival rate for cutaneous melanoma is 93% but patients with distant metastases have a 5-year survival rate of only 27% [232]. A clinical trial has demonstrated that the oral broccoli sprout extract, SFN, at a daily dose up to 200 μmol, is safe and well-tolerated in patients with melanoma and achieved dose-dependent levels in plasma and skin [233]. In a melanoma cell and zebrafish model, SFN has been shown to induce cell differentiation, melanogenesis, and inhibit proliferation by regulating the expression of MITF, PKCβ1, and tyrosinase [234]. However, when SFN combined with other biological elements, such as nerve growth factor, the anti-carcinogenic activities were partly reversed [235]. The next study should explore the role of SFN in combination with other chemotherapy drugs. Apart from SFN, other natural products include genistein and bryostatin-1 but have fewer clinical and animal results. In conclusion nrelatively few natural products have progressed to the clinical trial phase for the treatment of bladder cancer and melanoma.

            10. HEMATOLOGIC MALIGNANCIES

            10.1 Leukemia

            Leukemia is a group of malignant disorders of the blood, bone marrow, and lymphoid system. Leukemia is further divided into four major types: acute myeloid leukemia (AML); acute lymphocytic leukemia (ALL); chronic myeloid leukemia (CML); and chronic lymphocytic leukemia (CLL) [236]. In recent years the potential of natural products in the treatment of hematologic tumors has garnered widespread interest. Specifically, curcumin has emerged as a promising candidate for clinical trials. In ALL cells, three phenolic compounds (curcumin, CBD, and quercetin) display anti-leukemic activity by promoting mitochondrial membrane potential loss and mitochondrial Ca2+ overload. Notably, curcumin and quercetin further facilitate mitochondrial uncoupling [237]. This mechanism may enable curcumin to reduce glucocorticoid resistance in ALL cells [238]. Similarly, further research showed that the “cocktail” of four phenolic compounds (curcumin, genistein, resveratrol, and quercetin) has a cooperative anti-neoplastic activity on ALL cells without a significant effect on non-tumor cells [239]. In the context of AML, curcumin was shown to attenuate adriamycin resistance by suppressing the HOTAIR/miR-20a-5p/WT1 pathway in AML cells [240]. Moreover, mitocurcumin utilized oxidative stress to upregulate JNK/p38 to lead to apoptosis and overcome cytarabine resistance via ROS/p21/CHK1 in AML [241]. Similarly, curcumin and its analogs also exhibit chemopotentiating properties in other leukemia subtypes, such as CLL and CML [242, 243]. In addition, a variety of curcumin analogues, including EF-24, C1206, C212, and DMC, have been shown to possess anti-leukemic properties in leukemia [244247]. These results indicate that curcumin is a promising anti-cancer medicine against various subtypes of leukemia. Conversely, other natural products exhibit limited clinical and animal-based results in the treatment of leukemias.

            10.2 Lymphomas

            Lymphomas arise from an aberrant clonal proliferation of lymphocytes and can present within any organ in the body. According to the WHO classification, mature lymphoid neoplasms are mainly divided into non-Hodgkin’s lymphoma (NHL), which accounts for approximately 90% of all lymphomas, and Hodgkin’s lymphoma (HL), which accounts for approximately 10% [248]. Curcumin triggers caspase 3-dependent apoptosis in vitro and induces DNA damage through impairing Rad51-dependent homologous recombination. More importantly, curcumin enhances the sensitivity of lymphoma cells to various DNA damage drugs, including hydroxyurea, camptothecin, and cisplatin [249]. Curcumin in combination with homoharringtonine synergistically inhibit lymphoma cell growth in vivo by inhibiting the TGF-β/Smad3 signaling pathway [250]. It is known that CD20 is a prerequisite for rituximab anti-tumor activity. Inhibiting the expression of CD59 is a good strategy for overcoming resistance [251]. Both curcumin and perillyl alcohol suppress activation of NF-κB and CREB, and subsequently restrain expression of CD59 but not CD20, thereby sensitizing rituximab-resistant B lymphoma cells [251]. In conclusion, curcumin may be a potential adjuvant drug to enhance the efficacy of chemotherapy drugs in leukemias and lymphomas but clinical application needs additional data support.

            10.3 Multiple Myeloma

            Multiple myeloma (MM) is described as an incurable malignant disease that accounts for approximately 10%-15% of all hematologic malignancies [252]. Currently, immunotherapy, particularly chimeric antigen receptor T cell therapy, has an increasingly significant role in the treatment of hematologic tumors, including MM. Natural products retain unique benefits in enhancing the potency of other drugs while mitigating adverse effects. A pilot randomized clinical trial demonstrated that the combination of curcumin with melphalan and prednisone is more effective on improving overall remission and reducing the levels of NF-κB, VEGF, TNF-α, and IL-6 among MM patients compared to the group receiving only melphalan and prednisone [253]. GZ17-6.02, a synthetically manufactured compound containing isovanillin, harmine, and curcumin, has been shown to enhance the toxicity of proteasome inhibitors to kill MM cells in vitro [254]. Additionally, a number of studies have demonstrated that omega-3 fatty acid derivatives of DHA or eicosapentaenoic acid (EPA) not only alleviate bortezomib resistance by facilitating glutathione degradation but also increase bortezomib cytotoxicity. More importantly the co-incubation time is also crucial in this process [255257]. Furthermore, Mekkawy et al. [258] reported that bone marrow-derived MM CSCs exhibit potential as a prognostic indicator for predicting recurrent MM incidence among MM patients. Treatment with curcumin and piperine induce cell cycle arrest and apoptosis in MM CSCs [258]. Schmidt-Wolf et al. [259] verified that CB2 receptor is highly expressed on CIK cells as well as on MM cells. A low concentration of CBD could enhance the cytotoxic function of CIKs [259]. Hence, a thorough investigation into the precise mechanism by which CBD modulates CIKs in MM is imperative. In the future, we also look forward to more clinical data and application of natural products in the treatment of hematologic tumors.

            11. OTHER TUMORS

            Hepatocellular carcinoma (HCC) is among the most common and deadliest cancers and treatment options are limited. Huaier, a traditional herb, has enjoyed widespread application in Chinese medicine for approximately 1600 years. Multiple clinical trials have demonstrated that the administration of huaier granules extends recurrence-free survival (RFS) and OS after curative resection of HCC. Furthermore, this treatment also reduces extrahepatic recurrences [260262]. Similarly, SRL therapy (sirolimus, thymalfasin, and huaier granules) has been shown to be safe and effective in preventing HCC recurrence following liver transplantatio, without significant adverse events [263]. These results suggest that huaier granules are a promising candidate natural product for the treatment of HCC. In addition to huaier granules, other natural products have been advanced to clinical trials, including icaritin, ginsenoside Rg3, artesunate, CBD, coriolus versicolor, and Xiang Sha Liu Jun Zi decoction. Gastric carcinoma accounts for approximately 6% of cancers worldwide and is the 3rd leading cause of cancer-related deaths [264]. Recently, huaier granules, ginsenoside Rg3, mistletoe, and bryostatin 1 have moved into clinical trials for gastric carcinoma but these results have not been published or completed.

            12. CONCLUSION AND PERSPECTIVE

            Natural products are structurally diverse and have a wide range of sources and unique pharmacologic and biological activities [265, 266]. Currently, natural product-based pharmaceuticals and diet supplement development strategies play an important role in modern new drug development, accounting for a significant share of the market. For example, resveratrol and quercetin have been marketed as dietary supplements. The development of natural products is driving more dietary supplements to market. Remarkably, arsenic, a traditional Chinese medicine, has demonstrated remarkable effectiveness in the treatment of relapsed acute promyelocytic leukemia, highlighting its significant medical value [267]. Other natural products, such as paclitaxel, vindesine, and homoharringtonine, have been effectively utilized in the clinical management of cancer. It follows that research on the active ingredients of Chinese materia medica and natural products contributes to inspiring more new cancer therapeutic drugs.

            Apart from the natural products listed in Table 6, numerous emerging and well-known natural compounds, such as alfa-mangostin, garcinol, diosgenin, eugenol, xanthochymol, rosmarinic acid, and capsaicin, have demonstrated significant potential for entering clinical research in the treatment of cancers. For example, capsaicin has been used in clinical trials to assess its effectiveness in treating neuropathic pain among BC patients (NCT03794388, NCT05726929). These numerous emerging and well-known natural products provide a wide range of options for clinical trials of cancer treatments. In conclusion, natural products have great potential and market prospects in the field of new drug research. It is not only beneficial to the research and promotion of Chinese medicine but may also be developed as dietary supplements or adjuvant drugs for chemoradiotherapy. In this review, we have presented a comprehensive overview of the clinical trial progress in the field of natural product-relevant cancer therapy. The aim was to provide readers with a comprehensive resource of valuable information on natural drug candidates, deepen our understanding of the role of natural products in cancer treatment, and offer hope for the discovery of novel drug candidates for cancer therapy.

            Table 6 |

            Natural products that have been tested in cancer treatment of clinical trials.

            DiseaseNatural productReferences (https://clinicaltrials.gov)
            Colorectal cancerCurcumin, curcumin C3 complexNCT02724202*, NCT00973869*, NCT01859858*, NCT01333917,NCT00027495*, NCT00003365*, NCT01294072*, NCT00295035, NCT05472753*, NCT02439385*, NCT01490996
            ResveratrolNCT00256334, NCT00433576
            QuercetinNCT00003365*
            RutinNCT00003365*
            GenisteinNCT01985763
            CannabidiolNCT03607643, NCT04398446
            Epigallocatechin gallate, green tea extractNCT02891538, NCT01239095, NCT01360320*, NCT02321969*, NCT01606124
            AndrographolidesNCT01993472
            Mistletoe extractNCT00049608*
            Oligofructose-enriched inulinNCT00335504
            Topical mentholNCT01855607
            Shark cartilage (BenFin)NCT00026117
            HydroxytyrosolNCT05472753*
            ArtesunateNCT03093129, NCT02633098
            Lung cancerSulforaphane, broccoli sprout extractNCT03232138*, NCT00255775*
            Curcumin, curcumin C3 complex, Theracurmin 2XNCT01048983, NCT03598309, NCT02321293*, NCT04871412*
            Epigallocatechin gallate, green tea extract, polyphenon ENCT02577393, NCT04871412*, NCT01317953*, NCT00611650, NCT00573885, NCT00363805*, NCT00707252
            Coriolus VersicolorNCT04871412*
            Chlorogenic acidNCT03751592
            Soy isoflavonesNCT01958372*
            R-(-)-gossypol acetic acid, GossypolNCT00544596, NCT00397293, NCT00544960, NCT00934076, NCT00988169, NCT00773955, NCT01977209
            Mistletoe extractNCT00052325*, NCT00079794, NCT00516022, NCT00049608*
            GenisteinNCT00769990*, NCT01628471
            Ginsenoside H dripping pillsNCT02714608
            Berry powderNCT00681512*, NCT01426620*
            Shark cartilage extract AE-941NCT00005838
            FlaxseedNCT00955942*, NCT02475330*
            Lindera obtusiloba extractNCT04348149*
            AntroquinonolNCT02047344, NCT01134016
            Breast cancerSulforaphane, broccoli sprout extractNCT00894712, NCT03934905, NCT00982319, NCT00843167, NCT01753908, NCT03775525
            Curcumin, curcumin C3 complexNCT03980509, NCT03847623*, NCT03072992, NCT03865992*, NCT01740323, NCT01975363*, NCT00852332*, NCT02556632, NCT01042938
            Epigallocatechin gallate, green tea extract, tea capsule, polyphenolNCT02580279, NCT00516243, NCT00917735, NCT00949923*, NCT03482401*
            Genistein, soy isoflavonesNCT00244933*, NCT00099008*, NCT00290758, NCT00036686*, NCT00200824*, NCT04880369*, NCT00513916*, NCT00343434*, NCT01219075*, NCT00204490*, NCT00769990*
            CannabidiolNCT04482244, NCT05016349, NCT04754399, NCT04398446
            Tocotrienol, tocotrienol-rich fraction (TRF)NCT04496492, NCT02909751*, NCT03855423*, NCT01157026*
            ArtesunateNCT00764036
            Viscum album pini (mistletoe extract)NCT00176046, NCT00049608*
            Omega-3 fatty acid, docosahexaenoic acid (DHA)NCT00114296*, NCT02295059*, NCT01282580*, NCT01784042, NCT02101970*, NCT02278965, NCT02150525*, NCT05331807*, NCT00930527*, NCT02795572*, NCT02831582*, NCT01478477*, NCT02352779*, NCT01869764, NCT04268134*, NCT01881048*, NCT00627276*, NCT01849250, NCT01548534*, NCT01127867*, NCT01049295*, NCT03831178*, NCT03383835*
            Extra-virgin olive oil, hydroxytyrosolNCT04174391*, NCT02068092
            Seaweed and soy proteinNCT01204957*
            Perillyl alcoholNCT00003219, NCT00022425
            Shark cartilage (BenFin)NCT00026117
            Black cohoshNCT00060320*, NCT01628536*
            FlaxseedNCT00612560*, NCT00956813*, NCT00010829*, NCT00794989*
            CrocinNCT05504148
            MentholNCT05429814, NCT01855607
            White button mushroom extractNCT00709020, NCT04913064
            FADA (active fraction of ficus septica leaf)NCT05033925*
            Bryostatin 1NCT00003205
            Huaier granuleNCT02627248, NCT02615457, NCT04790305
            IH636 grape seed proanthocyanidin extractNCT00041223*, NCT00100893*
            QS21NCT00004156, NCT00470574, NCT00030823, NCT00003357
            CapsaicinNCT03794388
            Cervical cancerCurcuminNCT03192059*, NCT02554344, NCT04294836
            Bryostatin 1NCT00005965
            Green tea extractNCT00303823*
            Ovarian cancerFlaxseedNCT02324439
            Omega-3 fatty acidNCT01821833*
            Bryostatin 1NCT00004008, NCT00006942
            Squalamine lactateNCT00021385
            CannabidiolNCT04398446
            Endometrial cancerGenisteinNCT00099008*
            FlaxseedNCT00010829*
            CurcuminNCT02017353*, NCT03192059*
            PterostilbeneNCT03671811
            Pancreatic cancerCurcuminNCT00192842, NCT00486460, NCT00094445
            GenisteinNCT00376948*, NCT00882765*
            CannabidiolNCT03607643, NCT04398446
            AntroquinonolNCT03310632
            Mistletoe extract (Iscador Qu)NCT01448668, NCT02948309, NCT00049608*
            Perillyl alcoholNCT00003769
            Bryostatin 1NCT00031694
            Broccoli sprout grainNCT01879878*
            Bladder cancerSulforaphane, broccoli sprout extractNCT03517995, NCT01108003
            GenisteinNCT01489813, NCT00118040
            Green tea extract, polyphenon ENCT00666562*, NCT00088946*
            MelanomaSulforaphaneNCT01568996
            GenisteinNCT00769990*, NCT00276835*
            Bryostatin 1NCT00006022, NCT00112476
            LeukemiasCurcuminNCT05045443, NCT02100423
            Omega-3 fatty acid, docosahexaenoic acid (DHA) Eicosapentaenoic acid (EPA)NCT00899353*, NCT02373579, NCT00003077*, NCT01051154*, NCT04006847
            Bryostatin 1NCT00017342, NCT00003079, NCT00003174, NCT00087425, NCT00136461, NCT00002908, NCT00003171, NCT00003166, NCT00012376, NCT00005580
            Polyphenon ENCT00262743
            Sheng-Yu-TangNCT02580071
            R-(-)-gossypol acetic acid (AT-101)NCT01003769, NCT00275431
            CordycepinNCT00709215, NCT00003005
            LymphomasCurcuminNCT00969085, NCT02100423
            SGX301(synthetic hypericin)NCT02448381, NCT05380635
            GenisteinNCT02624388
            Broken ganoderma lucidum spore powderNCT04914143
            Omega-3 fatty acidNCT00003077*
            Bryostatin 1NCT00002725, NCT00022555, NCT00003993, NCT00058305, NCT00003936, NCT00003079, NCT00087425, NCT00002908, NCT00003166, NCT00005580
            Perillyl alcoholNCT00002862
            R-(-)-gossypol acetic acid (AT-101)NCT00891072, NCT05338931, NCT00440388, NCT00275431
            Multiple myelomCurcuminNCT01269203, NCT04731844, NCT00113841
            Omega-3 fatty acidNCT00899353*, NCT00003077*
            Green tea extractNCT00942422*
            PiperineNCT04731844
            BioperineNCT00113841
            Bryostatin 1NCT00002907, NCT00003166
            Shark cartilage extract AE-941NCT00022282
            PsoralenNCT00005092
            Agaricus blazei murillNCT00970021*
            CannabidiolNCT03607643
            Hepatocellular carcinomaHuaier granuleNCT01760616, NCT03356236, NCT01770431
            IcaritinNCT03236649, NCT01972672, NCT05594927, NCT03236636
            Ginsenoside Rg3NCT04523467, NCT01717066
            CannabidiolNCT03607643
            ArtesunateNCT02304289
            Coriolus cersicolorNCT01097083
            Xiang Sha Liu Jun Zi Decoction dry powderNCT04562428
            Gastric carcinomaHuaier granuleNCT05498766
            Ginsenoside Rg3NCT01757366
            Mistletoe extractNCT01401075
            Bryostatin 1NCT00006389, NCT00005599, NCT00006081
            Theracurmin 2XNCT04871412*
            Green tea extractNCT04871412*
            Coriolus VersicolorNCT04871412*

            *Natural products used in this clinical trial were dietary supplements.

            The literature search conducted on the website https://clinicaltrials.gov this time covered data through December 2022.

            CONFLICT OF INTEREST

            All authors declare no conflicts of interest.

            REFERENCES

            1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al.. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021. Vol. 71:209–249

            2. Liu Y, Yang S, Wang K, Lu J, Bao X, Wang R, et al.. Cellular Senescence and Cancer: Focusing on Traditional Chinese Medicine and Natural Products. Cell Proliferation. 2020. Vol. 53:e12894

            3. Pawar RS, Grundel E. Overview of Regulation of Dietary Supplements in the USA and Issues of Adulteration with Phenethylamines (PEAs). Drug Testing and Analysis. 2016. Vol. 9:500–517

            4. Patterson RE, Neuhouser ML, Hedderson MM, Schwartz SM, Standish LJ, Bowen DJ. Changes in Diet, Physical Activity, and Supplement use Among Adults Diagnosed with Cancer. Journal of the American Dietetic Association. 2003. Vol. 103:323–328

            5. Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, et al.. Colon Cancer and Colorectal Cancer: Prevention and Treatment by Potential Natural Products. Chemico-Biological Interactions. 2022. Vol. 368:110170

            6. Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, et al.. Small Molecule-Driven Mitophagy-Mediated NLRP3 Inflammasome Inhibition is Responsible for the Prevention of Colitis-Associated Cancer. Autophagy. 2014. Vol. 10:972–985

            7. Jeon Y, Sym SJ, Yoo BK, Baek JH. Long-Term Survival, Tolerability, and Safety of First-Line Bevacizumab and FOLFIRI in Combination With Ginsenoside-Modified Nanostructured Lipid Carrier Containing Curcumin in Patients With Unresectable Metastatic Colorectal Cancer. Integrative Cancer Therapies. 2022. Vol. 21:15347354221105498

            8. Howells LM, Iwuji COO, Irving GRB, Barber S, Walter H, Sidat Z, et al.. Curcumin Combined with FOLFOX Chemotherapy Is Safe and Tolerable in Patients with Metastatic Colorectal Cancer in a Randomized Phase IIa Trial. The Journal of Nutrition. 2019. Vol. 149:1133–1139

            9. Macis D, Briata IM, D’Ecclesiis O, Johansson H, Aristarco V, Buttiron Webber T, et al.. Inflammatory and Metabolic Biomarker Assessment in a Randomized Presurgical Trial of Curcumin and Anthocyanin Supplements in Patients with Colorectal Adenomas. Nutrients. 2023. Vol. 15:3894

            10. Liu C, Rokavec M, Huang Z, Hermeking H. Curcumin Activates a ROS/KEAP1/NRF2/miR-34a/b/c Cascade to Suppress Colorectal Cancer Metastasis. Cell Death and Differentiation. 2023. Vol. 30:1771–1785

            11. Liu G, Chen J, Bao Z. Promising Antitumor Effects of the Curcumin Analog DMC-BH on Colorectal Cancer Cells. Aging. 2023. Vol. 15:2221–2236

            12. Shih KC, Chan HW, Wu CY, Chuang HY. Curcumin Enhances the Abscopal Effect in Mice with Colorectal Cancer by Acting as an Immunomodulator. Pharmaceutics. 2023. Vol. 15:1519

            13. Aromokeye R, Si H. Combined Curcumin and Luteolin Synergistically Inhibit Colon Cancer Associated with Notch1 and TGF-beta Signaling Pathways in Cultured Cells and Xenograft Mice. Cancers. 2022. Vol. 14:3001

            14. Zheng X, Yang X, Lin J, Song F, Shao Y. Low Curcumin Concentration Enhances the Anticancer Effect of 5-Fluorouracil Against Olorectal Cancer. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2021. Vol. 85:153547

            15. Zhang T, Zheng P, Shen X, Shao R, Wang B, Shen H, et al.. Curcuminoid WZ26, a TrxR1 Inhibitor, Effectively Inhibits Colon Cancer Cell Growth and Enhances Cisplatin-Induced Cell Death through the Induction of ROS. Free Radical Biology & Medicine. 2019. Vol. 141:93–102

            16. Wu SX, Xiong RG, Huang SY, Zhou DD, Saimaiti A, Zhao CN, et al.. Effects and Mechanisms of Resveratrol for Prevention and Management of Cancers: An Updated Review. Critical Reviews in Food Science and Nutrition. 2023. Vol. 63:12422–12440

            17. Brockmueller A, Buhrmann C, Shayan P, Shakibaei M. Resveratrol Induces Apoptosis by Modulating the Reciprocal Crosstalk between p53 and Sirt-1 in the CRC Tumor Microenvironment. Frontiers in Immunology. 2023. Vol. 14:1225530

            18. Cheah FK, Leong KH, Thomas NF, Chin HK, Ariffin A, Awang K. Resveratrol Analogue, (E)-N-(2-(4-Methoxystyryl) Phenyl) Furan-2-Carboxamide Induces G(2)/M Cell Cycle Arrest through the Activation of p53-p21(CIP1/WAF1) in Human Colorectal HCT116 Cells. Apoptosis: An International Journal on Programmed Cell Death. 2018. Vol. 23:329–342

            19. Jozkowiak M, Skupin-Mrugalska P, Nowicki A, Borys-Wojcik S, Wierzchowski M, Kaczmarek M, et al.. The Effect of 4’-Hydroxy-3,4,5-Trimetoxystilbene, the Metabolite of Resveratrol Analogue DMU-212, on Growth, Cell Cycle and Apoptosis in DLD-1 and LOVO Colon Cancer Cell Lines. Nutrients. 2020. Vol. 12:1327

            20. De Gregorio A, Krasnowska EK, Zonfrillo M, Ravagnan G, Bordignon V, Bonmassar E, et al.. Influence of Polydatin on the Tumor Microenvironment In Vitro: Studies with a Colon Cancer Cell Model. International Journal of Molecular Sciences. 2022. Vol. 23:8442

            21. Lin TA, Lin WS, Chou YC, Nagabhushanam K, Ho CT, Pan MH. Oxyresveratrol Inhibits Human Colon Cancer Cell Migration through Regulating Epithelial-Mesenchymal Transition and microRNA. Food & Function. 2021. Vol. 12:9658–9668

            22. Brockmueller A, Girisa S, Kunnumakkara AB, Shakibaei M. Resveratrol Modulates Chemosensitisation to 5-FU via beta1-Integrin/HIF-1alpha Axis in CRC Tumor Microenvironment. International Journal of Molecular Sciences. 2023. Vol. 24:4988

            23. El-Readi MZ, Eid S, Abdelghany AA, Al-Amoudi HS, Efferth T, Wink M. Resveratrol Mediated Cancer Cell Apoptosis, and Modulation of Multidrug Resistance Proteins and Metabolic Enzymes. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2019. Vol. 55:269–281

            24. Liu Q, Qu J, Zhao M, Xu Q, Sun Y. Targeting SHP2 as a Promising Strategy for Cancer Immunotherapy. Pharmacological Research. 2020. Vol. 152:104595

            25. Zhao M, Guo W, Wu Y, Yang C, Zhong L, Deng G, et al.. SHP2 Inhibition Triggers Anti-Tumor Immunity and Synergizes with PD-1 Blockade. Acta Pharmaceutica Sinica B. 2019. Vol. 9:304–315

            26. Pan J, Zhou L, Zhang C, Xu Q, Sun Y. Targeting Protein Phosphatases for the Treatment of Inflammation-Related Diseases: From Signaling to Therapy. Signal Transduction and Targeted Therapy. 2022. Vol. 7:177

            27. Deng G, Zhou L, Wang B, Sun X, Zhang Q, Chen H, et al.. Targeting Cathepsin B by Cycloastragenol Enhances Antitumor Immunity of CD8 T Cells via Inhibiting MHC-I Degradation. Journal for Immunotherapy of Cancer. 2022. Vol. 10:e004874

            28. Lucas J, Hsieh TC, Halicka HD, Darzynkiewicz Z, Wu JM. Upregulation of PD-L1 Expression by Resveratrol and Piceatannol in Breast and Colorectal Cancer Cells Occurs via HDAC3/p300-Mediated NF-kappaB Signaling. International Journal of Oncology. 2018. Vol. 53:1469–1480

            29. Trinh NT, Nguyen TMN, Yook JI, Ahn SG, Kim SA. Quercetin and Quercitrin from Agrimonia pilosa Ledeb Inhibit the Migration and Invasion of Colon Cancer Cells through the JNK Signaling Pathway. Pharmaceuticals. 2022. Vol. 15:364

            30. Fosso E, Leo M, Muccillo L, Mandrone VM, Di Meo MC, Molinario A, et al.. Quercetin’s Dual Mode of Action to Counteract the Sp1-miR-27a Axis in Colorectal Cancer Cells. Antioxidants. 2023. Vol. 12:1547

            31. Shree A, Islam J, Sultana S. Quercetin Ameliorates Reactive Oxygen Species Generation, Inflammation, Mucus depletion, Goblet Disintegration, and Tumor Multiplicity in Colon Cancer: Probable Role of Adenomatous Polyposis Coli, Beta-Catenin. Phytotherapy Research: PTR. 2021. Vol. 35:2171–2184

            32. Benito I, Encio IJ, Milagro FI, Alfaro M, Martinez-Penuela A, Barajas M, et al.. Microencapsulated Bifidobacterium Bifidum and Lactobacillus Gasseri in Combination with Quercetin Inhibit Colorectal Cancer Development in Apc(Min/+) Mice. International Journal of Molecular Sciences. 2021. Vol. 22:4906

            33. Garcia-Gutierrez N, Luna-Barcenas G, Herrera-Hernandez G, Campos-Vega R, Lozano-Herrera SJ, Sánchez-Tusié AA, et al.. Quercetin and Its Fermented Extract as a Potential Inhibitor of Bisphenol A-Exposed HT-29 Colon Cancer Cells’ Viability. International Journal of Molecular Sciences. 2023. Vol. 24:5604

            34. Lee J, Jang CH, Kim Y, Oh J, Kim JS. Quercetin-Induced Glutathione Depletion Sensitizes Colorectal Cancer Cells to Oxaliplatin. Foods. 2023. Vol. 12:1733

            35. Rani Inala MS, Pamidimukkala K. Amalgamation of Quercetin with Anastrozole and Capecitabine: A Novel Combination to Treat Breast and Colon Cancers - An In Vitro Study. Journal of Cancer Research and Therapeutics. 2023. Vol. 19 Supplement:S93–S105

            36. Nafees S, Mehdi SH, Zafaryab M, Zeya B, Sarwar T, Rizvi MA. Synergistic Interaction of Rutin and Silibinin on Human Colon Cancer Cell Line. Archives of Medical Research. 2018. Vol. 49:226–234

            37. Ismail A, El-Biyally E, Sakran W. An Innovative Approach for Formulation of Rutin Tablets Targeted for Colon Cancer Treatment. AAPS PharmSciTech. 2023. Vol. 24:68

            38. Nasri Nasrabadi P, Zareian S, Nayeri Z, Salmanipour R, Parsafar S, Gharib E, et al.. A Detailed Image of Rutin Underlying Intracellular Signaling Pathways in Human SW480 Colorectal Cancer Cells Based on miRNAs-lncRNAs-mRNAs-TFs Interactions. Journal of Cellular Physiology. 2019. Vol. 234:15570–15580

            39. Alorda-Clara M, Torrens-Mas M, Morla-Barcelo PM, Roca P, Sastre-Serra J, Pons DG, et al.. High Concentrations of Genistein Decrease Cell Viability Depending on Oxidative Stress and Inflammation in Colon Cancer Cell Lines. International Journal of Molecular Sciences. 2022. Vol. 23:7526

            40. Pintova S, Dharmupari S, Moshier E, Zubizarreta N, Ang C, Holcombe RF. Genistein Combined with FOLFOX or FOLFOX-Bevacizumab for the Treatment of Metastatic Colorectal Cancer: Phase I/II Pilot Study. Cancer Chemotherapy and Pharmacology. 2019. Vol. 84:591–598

            41. Ko KP, Yeo Y, Yoon JH, Kim CS, Tokudome S, Ngoan LT, et al.. Plasma Phytoestrogens Concentration and Risk of Colorectal Cancer in Two Different Asian Populations. Clinical Nutrition. 2018. Vol. 37:1675–1682

            42. Liu X, Lan Y, Zhang L, Ye X, Shen Q, Mo G, et al.. Genistein Exerts Anti-Colorectal Cancer Actions: Clinical Reports, Computational and Validated Findings. Aging. 2023. Vol. 15:3678–3689

            43. Sun X, Zhou L, Wang Y, Deng G, Cao X, Ke B, et al.. Single-Cell Analyses Reveal Cannabidiol Rewires Tumor Microenvironment via Inhibiting Alternative Activation of Macrophage and Synergizes with Anti-PD-1 in Colon Cancer. J Pharm Anal. 2023. Vol. 13:726–744

            44. Kim NY, Mohan CD, Sethi G, Ahn KS. Cannabidiol Activates MAPK Pathway to Induce Apoptosis, Paraptosis, and Autophagy in Colorectal Cancer Cells. Journal of Cellular Biochemistry. 2024. Vol. 125:e30537

            45. Wang F, Dezfouli AB, Khosravi M, Sievert W, Stangl S, Schwab M, et al.. Cannabidiol-Induced Crosstalk of Apoptosis and Macroautophagy in Colorectal Cancer Cells Involves p53 and Hsp70. Cell Death Discovery. 2023. Vol. 9:286

            46. Kwon IS, Hwang YN, Park JH, Na HH, Kwon TH, Park JS, et al.. Metallothionein Family Proteins as Regulators of Zinc Ions Synergistically Enhance the Anticancer Effect of Cannabidiol in Human Colorectal Cancer Cells. International Journal of Molecular Sciences. 2023. Vol. 24:16621

            47. Lee HS, Tamia G, Song HJ, Amarakoon D, Wei CI, Lee SH. Cannabidiol Exerts Anti-Proliferative Activity via a Cannabinoid Receptor 2-Dependent Mechanism in Human Colorectal Cancer Cells. International Immunopharmacology. 2022. Vol. 108:108865

            48. Henning S, Niu Y, Lee N, Thames G, Minutti R, Wang H, et al.. Bioavailability and Antioxidant Activity of Tea Flavanols After Consumption of Green Tea, Black Tea, or a Green Tea Extract Supplement. The American Journal of Clinical Nutrition. 2004. Vol. 80:1558–1564

            49. Shin CM, Lee DH, Seo AY, Lee HJ, Kim SB, Son WC, et al.. Green Tea Extracts for the Prevention of Metachronous Colorectal Polyps Among Patients who Underwent Endoscopic Removal of Colorectal Adenomas: A Randomized Clinical Trial. Clinical Nutrition. 2018. Vol. 37:452–458

            50. Seufferlein T, Ettrich TJ, Menzler S, Messmann H, Kleber G, Zipprich A, et al.. Green Tea Extract to Prevent Colorectal Adenomas, Results of a Randomized, Placebo-Controlled Clinical Trial. The American Journal of Gastroenterology. 2022. Vol. 117:884–894

            51. Iram S, Rahman S, Ali S, Kim J. Tetranectin Targeting by Epigallocatechin Gallate Suppresses Colon Cancer Cell Proliferation. International Journal of Biological Macromolecules. 2022. Vol. 209:211–219

            52. Kwon OS, Jung JH, Shin EA, Park JE, Park WY, Kim SH. Epigallocatechin-3-Gallate Induces Apoptosis as a TRAIL Sensitizer via Activation of Caspase 8 and Death Receptor 5 in Human Colon Cancer Cells. Biomedicines. 2020. Vol. 8:84

            53. Ha T, Lee J, Lou Z, Lee BS, Kim CH, Lee SH. Identification of Epithelial-Specific ETS-1 (ESE-1) as a Tumor Suppressor and Molecular Target of Green Tea Compound, EGCG. Molecular Carcinogenesis. 2019. Vol. 58:922–932

            54. Zhu W, Oteiza PI. NADPH Oxidase 1: A Target in the Capacity of Dimeric ECG and EGCG Procyanidins to Inhibit Colorectal Cancer Cell Invasion. Redox Biology. 2023. Vol. 65:102827

            55. Wang Y, Jin SS, Li DT, Jiang XC, Afrasiyab , Khalid A, et al.. Improving the Anti-Tumor Effect of EGCG in Colorectal Cancer Cells by Blocking EGCG-Induced YAP Activation. American Journal of Cancer Research. 2023. Vol. 13:1407–1424

            56. Qu J, Liu Q, You G, Ye L, Jin Y, Kong L, et al.. Advances in Ameliorating Inflammatory Diseases and Cancers by Andrographolide: Pharmacokinetics, Pharmacodynamics, and Perspective. Medicinal Research Reviews. 2022. Vol. 42:1147–1178

            57. Liu Z, Wu X, Dai K, Li R, Zhang J, Sheng D, et al.. The New Andrographolide Derivative AGS-30 Induces Apoptosis in Human Colon Cancer Cells by Activating a ROS-Dependent JNK Signalling Pathway. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2022. Vol. 94:153824

            58. Reabroi S, Chairoungdua A, Saeeng R, Kasemsuk T, Saengsawang W, Zhu W, et al.. A Silyl Andrographolide Analogue Suppresses Wnt/beta-Catenin Signaling Pathway in Colon Cancer. Biomedicine & Pharmacotherapy. 2018. Vol. 101:414–421

            59. Li J, Li F, Tang F, Zhang J, Li R, Sheng D, et al.. AGS-30, An Andrographolide Derivative, Suppresses Tumor Angiogenesis and Growth In Vitro and In Vivo. Biochemical Pharmacology. 2020. Vol. 171:113694

            60. Banerjee V, Sharda N, Huse J, Singh D, Sokolov D, Czinn SJ, et al.. Synergistic Potential of Dual Andrographolide and Melatonin Targeting of Metastatic Colon Cancer Cells: Using the Chou-Talalay Combination Index Method. European Journal of Pharmacology. 2021. Vol. 897:173919

            61. Sokolov D, Sharda N, Giri B, Hassan MS, Singh D, Tarasiewicz A, et al.. Melatonin and Andrographolide Synergize to Inhibit the Colospheroid Phenotype by Targeting Wnt/beta-Catenin Signaling. Journal of Pineal Research. 2022. Vol. 73:e12808

            62. Wang W, Guo W, Li L, Fu Z, Liu W, Gao J, et al.. Andrographolide Reversed 5-FU Resistance in Human Colorectal Cancer by Elevating BAX Expression. Biochemical Pharmacology. 2016. Vol. 121:8–17

            63. Liu W, Fan T, Li M, Zhang G, Guo W, Yang X, et al.. Andrographolide Potentiates PD-1 Blockade Immunotherapy by Inhibiting COX2-Mediated PGE2 Release. International Immunopharmacology. 2020. Vol. 81:106206

            64. Gao J, Wu Z, Zhao M, Zhang R, Li M, Sun D, et al.. Allosteric Inhibition Reveals SHP2-Mediated Tumor Immunosuppression in Colon Cancer by Single-Cell Transcriptomics. Acta Pharmaceutica Sinica B. 2022. Vol. 12:149–166

            65. Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al.. The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2022. Vol. 17:362–387

            66. Yang F, Wang F, Liu Y, Wang S, Li X, Huang Y, et al.. Sulforaphane Induces Autophagy by Inhibition of HDAC6-Mediated PTEN Activation in Triple Negative Breast Cancer Cells. Life Sciences. 2018. Vol. 213:149–157

            67. Zheng Z, Lin K, Hu Y, Zhou Y, Ding X, Wang Y, et al.. Sulforaphane Metabolites Inhibit Migration and Invasion via Microtubule-Mediated Claudins Dysfunction or Inhibition of Autolysosome Formation in Human Non-Small Cell Lung Cancer Cells. Cell Death & Disease. 2019. Vol. 10:259

            68. Yan Y, Zhou Y, Li J, Zheng Z, Hu Y, Li L, et al.. Sulforaphane Downregulated Fatty Acid Synthase and Inhibited Microtubule-Mediated Mitophagy Leading to Apoptosis. Cell Death & Disease. 2021. Vol. 12:917

            69. Hu Y, Zhou Y, Yang G, Wang Y, Zheng Z, Li J, et al.. Sulforaphane-N-Acetyl-Cysteine Inhibited Autophagy Leading to Apoptosis via Hsp70-Mediated Microtubule Disruption. Cancer Letters. 2018. Vol. 431:85–95

            70. Xie C, Zhu J, Jiang Y, Chen J, Wang X, Geng S, et al.. Sulforaphane Inhibits the Acquisition of Tobacco Smoke-Induced Lung Cancer Stem Cell-Like Properties via the IL-6/DeltaNp63alpha/Notch Axis. Theranostics. 2019. Vol. 9:4827–4840

            71. Rakariyatham K, Yang X, Gao Z, Song M, Han Y, Chen X, et al.. Synergistic Chemopreventive Effect of Allyl Isothiocyanate and Sulforaphane on Non-Small Cell Lung Carcinoma Cells. Food & Function. 2019. Vol. 10:893–902

            72. Wang Y, Zhou Y, Zheng Z, Li J, Yan Y, Wu W. Sulforaphane Metabolites Reduce Resistance to Paclitaxel via Microtubule Disruption. Cell Death & Disease. 2018. Vol. 9:1134

            73. Wang TH, Chen CC, Huang KY, Shih YM, Chen CY. High Levels of EGFR Prevent Sulforaphane-Induced Reactive Oxygen Species-Mediated Apoptosis in Non-Small-Cell Lung Cancer Cells. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2019. Vol. 64:152926

            74. Fan J, Wu M, Wang J, Ren D, Zhao J, Yang G. 1,7-Bis(4-Hydroxyphenyl)-1,4-Heptadien-3-One Induces Lung Cancer Cell Apoptosis via the PI3K/Akt and ERK1/2 Pathways. Journal of Cellular Physiology. 2019. Vol. 234:6336–6349

            75. Chang M, Shang M, Yuan F, Guo W, Wang C. EF24 Exerts Cytotoxicity Against NSCLC via Inducing ROS Accumulation. Cancer Cell International. 2021. Vol. 21:531

            76. Wan Mohd Tajuddin WNB, Abas F, Othman I, Naidu R. Molecular Mechanisms of Antiproliferative and Apoptosis Activity by 1,5-Bis(4-Hydroxy-3-Methoxyphenyl)1,4-Pentadiene-3-One (MS13) on Human Non-Small Cell Lung Cancer Cells. International Journal of Molecular Sciences. 2021. Vol. 22:7424

            77. Liu X, Cui H, Li M, Chai Z, Wang H, Jin X, et al.. Tumor Killing by a Dietary Curcumin Mono-Carbonyl Analog that Works as a Selective ROS Generator via TrxR Inhibition. European Journal of Medicinal Chemistry. 2023. Vol. 250:115191

            78. Xie C, Zhu J, Yang X, Huang C, Zhou L, Meng Z, et al.. TAp63alpha Is Involved in Tobacco Smoke-Induced Lung Cancer EMT and the Anti-Cancer Activity of Curcumin via miR-19 Transcriptional Suppression. Frontiers in Cell and Developmental Biology. 2021. Vol. 9:645402

            79. Wu MF, Huang YH, Chiu LY, Cherng SH, Sheu GT, Yang TY. Curcumin Induces Apoptosis of Chemoresistant Lung Cancer Cells via ROS-Regulated p38 MAPK Phosphorylation. International Journal of Molecular Sciences. 2022. Vol. 23:8248

            80. Saddiq AA, El-Far AH, Mohamed Abdullah SA, Godugu K, Almaghrabi OA, Mousa SA. Curcumin, Thymoquinone, and 3, 3’-Diindolylmethane Combinations Attenuate Lung and Liver Cancers Progression. Frontiers in Pharmacology. 2022. Vol. 13:936996

            81. Hong X, Hsieh MT, Tseng TY, Lin HY, Chang HC, Yau ST, et al.. Diarylheptanoid 35d Overcomes EGFR TKI Resistance by Inducing hsp70-Mediated Lysosomal Degradation of EGFR in EGFR-Mutant Lung Adenocarcinoma. The Journal of Biological Chemistry. 2023. Vol. 299:104814

            82. Hsieh MT, Lee PC, Chiang YT, Lin HY, Lee DY. The Effects of a Curcumin Derivative and Osimertinib on Fatty Acyl Metabolism and Mitochondrial Functions in HCC827 Cells and Tumors. International Journal of Molecular Sciences. 2023. Vol. 24:12190

            83. Zhu W, Zhao Y, Zhang S, Li X, Xing L, Zhao H, et al.. Evaluation of Epigallocatechin-3-Gallate as a Radioprotective Agent During Radiotherapy of Lung Cancer Patients: A 5-Year Survival Analysis of a Phase 2 Study. Frontiers in Oncology. 2021. Vol. 11:686950

            84. Zhao H, Jia L, Chen G, Li X, Meng X, Zhao X, et al.. A Prospective, Three-Arm, Randomized Trial of EGCG for Preventing Radiation-Induced Esophagitis in Lung Cancer Patients Receiving Radiotherapy. Radiotherapy and Oncology: Journal of the European Society for Therapeutic Radiology and Oncology. 2019. Vol. 137:186–191

            85. Jiang P, Chen A, Wu X, Zhou M, Ul Haq I, Mariyam Z, et al.. NEAT1 Acts as an Inducer of Cancer Stem Cell-Like Phenotypes in NSCLC by Inhibiting EGCG-Upregulated CTR1. Journal of Cellular Physiology. 2018. Vol. 233:4852–4863

            86. Jiang P, Xu C, Chen L, Chen A, Wu X, Zhou M, et al.. Epigallocatechin-3-Gallate Inhibited Cancer Stem Cell-Like Properties by Targeting hsa-mir-485-5p/RXRalpha in Lung Cancer. Journal of Cellular Biochemistry. 2018. Vol. 119:8623–8635

            87. Jiang P, Xu C, Zhang P, Ren J, Mageed F, Wu X, et al.. Epigallocatechin-3-Gallate Inhibits Self-Renewal Ability of Lung Cancer Stem-Like Cells through Inhibition of CLOCK. International Journal of Molecular Medicine. 2020. Vol. 46:2216–2224

            88. Jiang P, Xu C, Chen L, Chen A, Wu X, Zhou M, et al.. EGCG Inhibits CSC-Like Properties through Targeting miR-485/CD44 Axis in A549-Cisplatin Resistant Cells. Molecular Carcinogenesis. 2018. Vol. 57:1835–1844

            89. Minnelli C, Cianfruglia L, Laudadio E, Mobbili G, Galeazzi R, Armeni T. Effect of Epigallocatechin-3-Gallate on EGFR Signaling and Migration in Non-Small Cell Lung Cancer. International Journal of Molecular Sciences. 2021. Vol. 22:11833

            90. Sun XL, Xiang ZM, Xie YR, Zhang N, Wang LX, Wu YL, et al.. Dimeric-(-)-Epigallocatechin-3-Gallate Inhibits the Proliferation of Lung Cancer Cells by Inhibiting the EGFR Signaling Pathway. Chemico-Biological Interactions. 2022. Vol. 365:110084

            91. Li F, Hao S, Gao J, Jiang P. EGCG Alleviates Obesity-Exacerbated Lung Cancer Progression by STAT1/SLC7A11 Pathway and Gut Microbiota. The Journal of Nutritional Biochemistry. 2023. Vol. 120:109416

            92. Zhou Y, Huang S, Guo Y, Ran M, Shan W, Chen WH, et al.. Epigallocatechin Gallate Circumvents Drug-Induced Resistance in Non-Small-Cell Lung Cancer by Modulating Glucose Metabolism and AMPK/AKT/MAPK axis. Phytotherapy Research: PTR. 2023. Vol. 37:5837–5853

            93. Wang L, Du H, Chen P. Chlorogenic Acid Inhibits the Proliferation of Human Lung Cancer A549 Cell Lines by Targeting Annexin A2 In Vitro and In Vivo. Biomedicine & Pharmacotherapy. 2020. Vol. 131:110673

            94. Huang S, Wang LL, Xue NN, Li C, Guo HH, Ren TK, et al.. Chlorogenic Acid Effectively Treats Cancers through Induction of Cancer Cell Differentiation. Theranostics. 2019. Vol. 9:6745–6763

            95. Cittadini MC, Repossi G, Albrecht C, Di Paola Naranjo R, Miranda AR, de Pascual-Teresa S, et al.. Effects of Bioavailable Phenolic Compounds from Ilex Paraguariensis on the Brain of Mice with Lung Adenocarcinoma. Phytotherapy Research: PTR. 2019. Vol. 33:1142–1149

            96. Shimazu T, Inoue M, Sasazuki S, Iwasaki M, Sawada N, Yamaji T, et al.. Isoflavone Intake and Risk of Lung Cancer: A Prospective Cohort Study in Japan. The American Journal of Clinical Nutrition. 2010. Vol. 91:722–728

            97. Chei CL, Sawada N, Khankari NK, Iwasaki M, Yamaji T, Cai H, et al.. Isoflavone and Soy Food Intake and Risk of Lung Cancer in Never Smokers: Report from Prospective Studies in Japan and China. European Journal of Nutrition. 2022. Vol. 62:125–137

            98. Nachvak SM, Moradi S, Anjom-Shoae J, Rahmani J, Nasiri M, Maleki V, et al.. Soy, Soy Isoflavones, and Protein Intake in Relation to Mortality from All Causes, Cancers, and Cardiovascular Diseases: A Systematic Review and Dose-Response Meta-Analysis of Prospective Cohort Studies. Journal of the Academy of Nutrition and Dietetics. 2019. Vol. 119:1483–1500 e17

            99. Fan Y, Wang M, Li Z, Jiang H, Shi J, Shi X, et al.. Intake of Soy, Soy Isoflavones and Soy Protein and Risk of Cancer Incidence and Mortality. Frontiers in Nutrition. 2022. Vol. 9:847421

            100. Yang WS, Va P, Wong MY, Zhang HL, Xiang YB. Soy Intake is Associated with Lower Lung Cancer Risk: Results from a Meta-Analysis of Epidemiologic Studies. The American Journal of Clinical Nutrition. 2011. Vol. 94:1575–1583

            101. Simone CB 2nd, Serebrenik AA, Gore EM, Mohindra P, Brown SL, Wang D, et al.. Multicenter Phase 1b/2a Clinical Trial of Radioprotectant BIO 300 Oral Suspension for Patients With Non-Small Cell Lung Cancer Receiving Concurrent Chemoradiotherapy. International Journal of Radiation Oncology, Biology, Physics. 2024. Vol. 118:404–414

            102. Yu Y, Xing Y, Zhang Q, Zhang Q, Huang S, Li X, et al.. Soy Isoflavone Genistein Inhibits hsa_circ_0031250/miR-873-5p/FOXM1 Axis to Suppress Non-Small-Cell Lung Cancer Progression. IUBMB Life. 2021. Vol. 73:92–107

            103. Chan L, Pang Y, Wang Y, Zhu D, Taledaohan A, Jia Y, et al.. Genistein-Induced Mitochondrial Dysfunction and FOXO3a/PUMA Expression in Non-Small Lung Cancer Cells. Pharmaceutical Biology. 2022. Vol. 60:1876–1883

            104. Suraweera TL, Merlin JPJ, Dellaire G, Xu Z, Rupasinghe HPV. Genistein and Procyanidin B2 Reduce Carcinogen-Induced Reactive Oxygen Species and DNA Damage through the Activation of Nrf2/ARE Cell Signaling in Bronchial Epithelial Cells In Vitro. International Journal of Molecular Sciences. 2023. Vol. 24:3676

            105. Zeng Y, Ma J, Xu L, Wu D. Natural Product Gossypol and its Derivatives in Precision Cancer Medicine. Current Medicinal Chemistry. 2019. Vol. 26:1849–1873

            106. Wang Y, Li X, Zhang L, Li M, Dai N, Luo H, et al.. A Randomized, Double-Blind, Placebo-Controlled Study of B-Cell Lymphoma 2 Homology 3 Mimetic Gossypol Combined with Docetaxel and Cisplatin for Advanced Non-Small Cell Lung Cancer with High Expression of Apurinic/Apyrimidinic Endonuclease 1. Investigational New Drugs. 2020. Vol. 38:1862–1871

            107. Zhou W, Wang W, Liang Y, Jiang R, Qiu F, Shao X, et al.. The RNA-Binding Protein LRPPRC Promotes Resistance to CDK4/6 Inhibition in Lung Cancer. Nature Communications. 2023. Vol. 14:4212

            108. Hussain Y, Meena A, Sinha RA. Gossypol Synergises Antiproliferative Effect of Sorafenib in Metastatic Lung Cancer Cells Following Chou-Talalay Algorithm. Toxicol In Vitro. 2023. Vol. 93:105666

            109. Xu J, Zhu GY, Cao D, Pan H, Li YW. Gossypol Overcomes EGFR-TKIs Resistance in Non-Small Cell Lung Cancer Cells by Targeting YAP/TAZ and EGFR(L858R/T790M). Biomedicine & Pharmacotherapy. 2019. Vol. 115:108860

            110. Bar-Sela G, Wollner M, Hammer L, Agbarya A, Dudnik E, Haim N. Mistletoe as Complementary Treatment in Patients with Advanced Non-Small-Cell Lung Cancer Treated with Carboplatin-Based Combinations: A Randomised Phase II Study. European Journal of Cancer. 2013. Vol. 49:1058–1064

            111. Lee YG, Jung I, Koo DH, Kang DY, Oh TY, Oh S, et al.. Efficacy and Safety of Viscum Album Extract (Helixor-M) to Treat Malignant Pleural Effusion in Patients with Lung Cancer. Supportive Care in Cancer: Official Journal of the Multinational Association of Supportive Care in Cancer. 2019. Vol. 27:1945–1949

            112. Schad F, Steinmann D, Oei SL, Thronicke A, Grah C. Evaluation of Quality of Life in Lung Cancer Patients Receiving Radiation and Viscum Album L.: A Real-World Data Study. Radiation Oncology. 2023. Vol. 18:47

            113. Schad F, Thronicke A, Steele ML, Merkle A, Matthes B, Grah C, et al.. Overall Survival of Stage IV Non-Small Cell Lung Cancer Patients Treated with Viscum Album L. in Addition to Chemotherapy, a Real-World Observational Multicenter Analysis. PloS One. 2018. Vol. 13:e0203058

            114. Shatat MA, Gauthier B, Yoon S, Yuan E, Yang P, Narla G, et al.. Mistletoe Lectin Inhibits Growth of Myc-Amplified Small-Cell Lung Cancer. Cancer Medicine. 2023. Vol. 12:8378–8387

            115. Barnard ME, Boeke CE, Tamimi RM. Established Breast Cancer Risk Factors and Risk of Intrinsic Tumor Subtypes. Biochimica et Biophysica Acta. 2015. Vol. 1856:73–85

            116. Sailo BL, Liu L, Chauhan S, Girisa S, Hegde M, Liang L, et al.. Harnessing Sulforaphane Potential as a Chemosensitizing Agent: A Comprehensive Review. Cancers. 2024. Vol. 16:244

            117. Shoaib S, Khan FB, Alsharif MA, Malik MS, Ahmed SA, Jamous YF, et al.. Reviewing the Prospective Pharmacological Potential of Isothiocyanates in Fight against Female-Specific Cancers. Cancers. 2023. Vol. 15:2390

            118. Zhang Z, Atwell LL, Farris PE, Ho E, Shannon J. Associations between Cruciferous Vegetable Intake and Selected Biomarkers Among Women Scheduled for Breast Biopsies. Public Health Nutrition. 2016. Vol. 19:1288–1295

            119. Wang Z, Tu C, Pratt R, Khoury T, Qu J, Fahey JW, et al.. A Presurgical-Window Intervention Trial of Isothiocyanate-Rich Broccoli Sprout Extract in Patients with Breast Cancer. Molecular Nutrition & Food Research. 2022. Vol. 66:e2101094

            120. Cao S, Hu S, Jiang P, Zhang Z, Li L, Wu Q. Effects of Sulforaphane on Breast Cancer Based on Metabolome and Microbiome. Food Science & Nutrition. 2023. Vol. 11:2277–2287

            121. Zhang Y, Lu Q, Li N, Xu M, Miyamoto T, Liu J. Sulforaphane Suppresses Metastasis of Triple-Negative Breast Cancer Cells by Targeting the RAF/MEK/ERK Pathway. NPJ Breast Cancer. 2022. Vol. 8:40

            122. Rong Y, Huang L, Yi K, Chen H, Liu S, Zhang W, et al.. Co-Administration of Sulforaphane and Doxorubicin Attenuates Breast Cancer Growth by Preventing the Accumulation of Myeloid-Derived Suppressor Cells. Cancer Letters. 2020. Vol. 493:189–196

            123. Li S, Wu H, Chen M, Tollefsbol TO. Paternal Combined Botanicals Contribute to the Prevention of Estrogen Receptor-Negative Mammary Cancer in Transgenic Mice. The Journal of Nutrition. 2023. Vol. 153:1959–1973

            124. Farghadani R, Naidu R. Curcumin as an Enhancer of Therapeutic Efficiency of Chemotherapy Drugs in Breast Cancer. International Journal of Molecular Sciences. 2022. Vol. 23:2144

            125. Barcelos KA, Mendonca CR, Noll M, Botelho AF, Francischini CRD, Silva MAM. Antitumor Properties of Curcumin in Breast Cancer Based on Preclinical Studies: A Systematic Review. Cancers. 2022. Vol. 14:2165

            126. Saghatelyan T, Tananyan A, Janoyan N, Tadevosyan A, Petrosyan H, Hovhannisyan A, et al.. Efficacy and Safety of Curcumin in Combination with Paclitaxel in Patients with Advanced, Metastatic Breast Cancer: A Comparative, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2020. Vol. 70:153218

            127. Kunihiro AG, Brickey JA, Frye JB, Cheng JN, Luis PB, Schneider C, et al.. Curcumin Inhibition of TGFbeta Signaling in Bone Metastatic Breast Cancer Cells and the Possible Role of Oxidative Metabolites. The Journal of Nutritional Biochemistry. 2022. Vol. 99:108842

            128. Li M, Guo T, Lin J, Huang X, Ke Q, Wu Y, et al.. Curcumin Inhibits the Invasion and Metastasis of Triple Negative Breast Cancer via Hedgehog/Gli1 Signaling Pathway. Journal of Ethnopharmacology. 2022. Vol. 283:114689

            129. Duan Y, Chen HL, Ling M, Zhang S, Ma FX, Zhang HC, et al.. The Curcumin Analog EF24 Inhibits Proliferation and Invasion of Triple-Negative Breast Cancer Cells by Targeting the Long Noncoding RNA HCG11/Sp1 Axis. Molecular and Cellular Biology. 2022. Vol. 42:e0016321

            130. Jang BY, Shin MK, Han DH, Sung JS. Curcumin Disrupts a Positive Feedback Loop between ADMSCs and Cancer Cells in the Breast Tumor Microenvironment via the CXCL12/CXCR4 Axis. Pharmaceutics. 2023. Vol. 15:2627

            131. Almalki E, Al-Amri A, Alrashed R, Al-Zharani M, Semlali A. The Curcumin Analog PAC Is a Potential Solution for the Treatment of Triple-Negative Breast Cancer by Modulating the Gene Expression of DNA Repair Pathways. International Journal of Molecular Sciences. 2023. Vol. 24:9649

            132. Nayak D, Paul S, Das C, Bhal S, Kundu CN. Quinacrine and Curcumin in Combination Decreased the Breast Cancer Angiogenesis by Modulating ABCG2 via VEGF A. Journal of Cell Communication and Signaling. 2023. Vol. 17:609–626

            133. Petrova L, Gergov N, Stoup M, Zapryanova S, Van Damme EJM, Lebègue N, et al.. Jacalin-Curcumin Complex Sensitizes the Breast Cancer MDA-MB-231 Cell Line. International Journal of Molecular Sciences. 2023. Vol. 24:17399

            134. Afshari H, Noori S, Zarghi A. Curcumin Potentiates the Anti-Inflammatory Effects of Tehranolide by Modulating the STAT3/NF-kappaB Signaling Pathway in Breast and Ovarian Cancer Cell Lines. Inflammopharmacology. 2023. Vol. 31:2541–2555

            135. Wang R, Yu H, Chen P, Yuan T, Zhang J. Integrated Transcriptome and Molecular Docking to Identify the Hub Superimposed Attenuation Targets of Curcumin in Breast Cancer Cells. International Journal of Molecular Sciences. 2023. Vol. 24:12479

            136. Samavat H, Ursin G, Emory TH, Lee E, Wang R, Torkelson CJ, et al.. A Randomized Controlled Trial of Green Tea Extract Supplementation and Mammographic Density in Postmenopausal Women at Increased Risk of Breast Cancer. Cancer Prevention Research. 2017. Vol. 10:710–718

            137. Samavat H, Wu AH, Ursin G, Torkelson CJ, Wang R, Yu MC, et al.. Green Tea Catechin Extract Supplementation Does Not Influence Circulating Sex Hormones and Insulin-Like Growth Factor Axis Proteins in a Randomized Controlled Trial of Postmenopausal Women at High Risk of Breast Cancer. The Journal of Nutrition. 2019. Vol. 149:619–627

            138. Zhao H, Zhu W, Zhao X, Li X, Zhou Z, Meng X, et al.. Efficacy of Epigallocatechin-3-Gallate in Preventing Dermatitis in Patients With Breast Cancer Receiving Postoperative Radiotherapy: A Double-Blind, Placebo-Controlled, Phase 2 Randomized Clinical Trial. JAMA Dermatology. 2022. Vol. 158:779–786

            139. Xu P, Yan F, Zhao Y, Chen X, Sun S, Wang Y, et al.. Green Tea Polyphenol EGCG Attenuates MDSCs-Mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine Breast Cancer Model. Nutrients. 2020. Vol. 12:1042

            140. Lee WJ, Cheng TC, Yen Y, Fang CL, Liao YC, Kuo CC, et al.. Tea Polyphenol Epigallocatechin-3-Gallate Inhibits Cell Proliferation in a Patient-Derived Triple-Negative Breast Cancer Xenograft Mouse Model via Inhibition of Proline-Dehydrogenase-Induced Effects. Journal of Food and Drug Analysis. 2021. Vol. 29:113–127

            141. Gonzalez Suarez N, Fernandez-Marrero Y, Torabidastgerdooei S, Annabi B. EGCG Prevents the Onset of an Inflammatory and Cancer-Associated Adipocyte-like Phenotype in Adipose-Derived Mesenchymal Stem/Stromal Cells in Response to the Triple-Negative Breast Cancer Secretome. Nutrients. 2022. Vol. 14:1099

            142. Xie L, Yi J, Song Y, Zhao M, Fan L, Zhao L. Suppression of GOLM1 by EGCG through HGF/HGFR/AKT/GSK-3beta/beta-Catenin/c-Myc Signaling Pathway Inhibits Cell Migration of MDA-MB-231. Food and Chemical Toxicology: An International Journal Published for the British Industrial Biological Research Association. 2021. Vol. 157:112574

            143. Santos RA, Andrade EDS, Monteiro M, Fialho E, Silva JL, Daleprane JB, et al.. Green Tea (Camellia Sinensis) Extract Induces p53-Mediated Cytotoxicity and Inhibits Migration of Breast Cancer Cells. Foods. 2021. Vol. 10:3154

            144. Guo Y, Cui Y, Li Y, Jin X, Wang D, Lei M, et al.. Cytoplasmic YAP1-Mediated ESCRT-III Assembly Promotes Autophagic Cell Death and is Ubiquitinated by NEDD4L in Breast Cancer. Cancer Commun (Lond). 2023. Vol. 43:582–612

            145. Zhang T, Li N, Wang R, Sun Y, He X, Lu X, et al.. Enhanced Therapeutic Efficacy of Doxorubicin Against Multidrug-Resistant Breast Cancer with Reduced Cardiotoxicity. Drug Delivery. 2023. Vol. 30:2189118

            146. Pop EA, Fischer LM, Coan AD, Gitzinger M, Nakamura J, Zeisel SH. Effects of a High Daily dose of Soy Isoflavones on DNA Damage, Apoptosis, and Estrogenic Outcomes in Healthy Postmenopausal Women: A Phase I Clinical Trial. Menopause. 2008. Vol. 15:684–692

            147. Wu AH, Spicer D, Garcia A, Tseng CC, Hovanessian-Larsen L, Sheth P, et al.. Double-Blind Randomized 12-Month Soy Intervention Had No Effects on Breast MRI Fibroglandular Tissue Density or Mammographic Density. Cancer Prevention Research. 2015. Vol. 8:942–951

            148. Sharma M, Arora I, Chen M, Wu H, Crowley MR, Tollefsbol TO, et al.. Therapeutic Effects of Dietary Soybean Genistein on Triple-Negative Breast Cancer via Regulation of Epigenetic Mechanisms. Nutrients. 2021. Vol. 13:3944

            149. Bezerra PHA, Amaral C, Almeida CF, Correia-da-Silva G, Torqueti MR, Teixeira N. In Vitro Effects of Combining Genistein with Aromatase Inhibitors: Concerns Regarding Its Consumption during Breast Cancer Treatment. Molecules. 2023. Vol. 28:4893

            150. Hu C, Wang M, Hu M, Ma S, Yang B, Xiao W, et al.. Genistein Induces Endocrine Resistance in Human Breast Cancer by Suppressing H3K27 Trimethylation. Endocrine-Related Cancer. 2022. Vol. 30:e220191

            151. Pawlicka MA, Zmorzynski S, Popek-Marciniec S, Filip AA. The Effects of Genistein at Different Concentrations on MCF-7 Breast Cancer Cells and BJ Dermal Fibroblasts. International Journal of Molecular Sciences. 2022. Vol. 23:12360

            152. Jo MJ, Kim BG, Kim WY, Lee DH, Yun HK, Jeong S, et al.. Cannabidiol Suppresses Angiogenesis and Stemness of Breast Cancer Cells by Downregulation of Hypoxia-Inducible Factors-1alpha. Cancers. 2021. Vol. 13:5667

            153. Surapaneni SK, Patel N, Sun L, Kommineni N, Kalvala AK, Gebeyehu A, et al.. Anticancer and Chemosensitization Effects of Cannabidiol in 2D and 3D Cultures of TNBC: Involvement of GADD45alpha, Integrin-alpha5, -beta5, -beta1, and Autophagy. Drug Delivery and Translational Research. 2022. Vol. 12:2762–2777

            154. Mokoena D, George BP, Abrahamse H. Cannabidiol Combination Enhances Photodynamic Therapy Effects on MCF-7 Breast Cancer Cells. Cells. 2024. Vol. 13:187

            155. Almeida CF, Teixeira N, Valente MJ, Vinggaard AM, Correia-da-Silva G, Amaral C. Cannabidiol as a Promising Adjuvant Therapy for Estrogen Receptor-Positive Breast Tumors: Unveiling Its Benefits with Aromatase Inhibitors. Cancers. 2023. Vol. 15:2517

            156. Proto MC, Fiore D, Bifulco M, Gazzerro P. Rimonabant and Cannabidiol Rewrite the Interactions between Breast Cancer Cells and Tumor Microenvironment. International Journal of Molecular Sciences. 2023. Vol. 24:13427

            157. Garcia-Morales L, Mendoza-Rodriguez MG, Tapia Ramirez J, Meza I. CBD Inhibits In Vivo Development of Human Breast Cancer Tumors. International Journal of Molecular Sciences. 2023. Vol. 24:13235

            158. D’Aloia A, Ceriani M, Tisi R, Stucchi S, Sacco E, Costa B. Cannabidiol Antiproliferative Effect in Triple-Negative Breast Cancer MDA-MB-231 Cells Is Modulated by Its Physical State and by IGF-1. International Journal of Molecular Sciences. 2022. Vol. 23:7145

            159. Idriss M, Hodroj MH, Fakhoury R, Rizk S. Beta-Tocotrienol Exhibits More Cytotoxic Effects than Gamma-Tocotrienol on Breast Cancer Cells by Promoting Apoptosis via a P53-Independent PI3-Kinase Dependent Pathway. Biomolecules. 2020. Vol. 10:577

            160. Kjaer IM, Kahns S, Timm S, Andersen RF, Madsen JS, Jakobsen EH, et al.. Phase II trial of Delta-Tocotrienol in Neoadjuvant Breast Cancer with Evaluation of Treatment Response Using ctDNA. Scientific Reports. 2023. Vol. 13:8419

            161. Nesaretnam K, Selvaduray KR, Abdul Razak G, Veerasenan SD, Gomez PA. Effectiveness of Tocotrienol-Rich Fraction Combined with Tamoxifen in the Management of Women with Early Breast Cancer: A Pilot Clinical Trial. Breast Cancer Research: BCR. 2010. Vol. 12:R81

            162. Ramdas P, Radhakrishnan AK, Abdu Sani AA, Kumari M, Anandha Rao JS, Abdul-Rahman PS. Advancing the Role of Gamma-Tocotrienol as Proteasomes Inhibitor: A Quantitative Proteomic Analysis of MDA-MB-231 Human Breast Cancer Cells. Biomolecules. 2019. Vol. 10:19

            163. Ding Y, Fan J, Fan Z, Zhang K. gamma-Tocotrienol Reverses Multidrug Resistance of Breast Cancer Cells through the Regulation of the gamma-Tocotrienol-NF-kappaB-P-gp Axis. The Journal of Steroid Biochemistry and Molecular Biology. 2021. Vol. 209:105835

            164. Pizato N, Kiffer L, Luzete BC, Assumpcao JAF, Correa LH, Melo HAB, et al.. Omega 3-DHA and Delta-Tocotrienol Modulate Lipid Droplet Biogenesis and Lipophagy in Breast Cancer Cells: The Impact in Cancer Aggressiveness. Nutrients. 2019. Vol. 11:1199

            165. Subramaiam H, Chu WL, Radhakrishnan AK, Chakravarthi S, Selvaduray KR, Kok YY. Evaluating Anticancer and Immunomodulatory Effects of Spirulina (Arthrospira) Platensis and Gamma-Tocotrienol Supplementation in a Syngeneic Mouse Model of Breast Cancer. Nutrients. 2021. Vol. 13:2320

            166. Wen L, Liu L, Wen L, Yu T, Wei F. Artesunate Promotes G2/M Cell Cycle Arrest in MCF7 Breast Cancer Cells through ATM Activation. Breast Cancer. 2018. Vol. 25:681–686

            167. von Hagens C, Walter-Sack I, Goeckenjan M, Storch-Hagenlocher B, Sertel S, Elsässer M, et al.. Long-Term Add-on Therapy (Compassionate Use) with Oral Artesunate in Patients with Metastatic Breast Cancer after Participating in a Phase I Study (ARTIC M33/2). Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2019. Vol. 54:140–148

            168. Saeed MEM, Drif AI, Efferth T. Biomarker Profiling Revealed Carcinoembryonic Antigen as a Target of Artesunate in a Ductal Breast Cancer Patient. Anticancer Research. 2022. Vol. 42:3483–3494

            169. Yao Y, Guo Q, Cao Y, Qiu Y, Tan R, Yu Z, et al.. Artemisinin Derivatives Inactivate Cancer-Associated Fibroblasts through Suppressing TGF-beta Signaling in Breast Cancer. Journal of Experimental & Clinical Cancer Research: CR. 2018. Vol. 37:282

            170. Terragno M, Vetrova A, Semenov O, Sayan AE, Kriajevska M, Tulchinsky E. Mesenchymal-Epithelial Transition and AXL Inhibitor TP-0903 Sensitise Triple-Negative Breast Cancer Cells to the Antimalarial Compound, Artesunate. Scientific Reports. 2024. Vol. 14:425

            171. Duarte D, Nunes M, Ricardo S, Vale N. Combination of Antimalarial and CNS Drugs with Antineoplastic Agents in MCF-7 Breast and HT-29 Colon Cancer Cells: Biosafety Evaluation and Mechanism of Action. Biomolecules. 2022. Vol. 12:1490

            172. Li Z, Zhu YT, Xiang M, Qiu JL, Luo SQ, Lin F. Enhanced Lysosomal Function is Critical for Paclitaxel Resistance in Cancer Cells: Reversed by Artesunate. Acta Pharmacologica Sinica. 2021. Vol. 42:624–632

            173. Li W, Zhao L, Li Y, Zhai Z. Artesunate Attenuates Proliferation of Epithelial Cells by Downregulating the NF-kappaB and AKT Signaling Pathways in Benign Mammary Gland Hyperplasia Rats. Annals of Translational Medicine. 2021. Vol. 9:848

            174. Steigenberger C, Schnell-Inderst P, Flatscher-Thoni M, Plank LM, Siebert U. Patient’ and Social Aspects Related to Complementary Mistletoe Therapy in Patients with Breast Cancer: A Systematic Review Commissioned by the German Agency for Health Technology Assessment. European Journal of Oncology Nursing. 2023. Vol. 65:102338

            175. Stocker A, Mehnert-Theuerkauf A, Hinz A, Ernst J. Utilization of Complementary and Alternative Medicine (CAM) by Women with Breast Cancer or Gynecological Cancer. PloS One. 2023. Vol. 18:e0285718

            176. Staupe H, Buentzel J, Keinki C, Buentzel J, Huebner J. Systematic Analysis of Mistletoe Prescriptions in Clinical Studies. Journal of Cancer Research and Clinical Oncology. 2023. Vol. 149:5559–5571

            177. Oei SL, Thronicke A, Kroz M, von Trott P, Schad F, Matthes H. Impact of Oncological Therapy and Viscum album L Treatment on Cancer-Related Fatigue and Internal Coherence in Nonmetastasized Breast Cancer Patients. Integrative Cancer Therapies. 2020. Vol. 19:1534735420917211

            178. Loef M, Paepke D, Walach H. Quality of Life in Breast Cancer Patients Treated With Mistletoe Extracts: A Systematic Review and Meta-Analysis. Integrative Cancer Therapies. 2023. Vol. 22:15347354231198074

            179. Schad F, Thronicke A. Safety of Combined Targeted and Helixor((R)) Viscum album L. Therapy in Breast and Gynecological Cancer Patients, a Real-World Data Study. International Journal of Environmental Research and Public Health. 2023. Vol. 20:2565

            180. Pelzer F, Troger W, Nat DR. Complementary Treatment with Mistletoe Extracts During Chemotherapy: Safety, Neutropenia, Fever, and Quality of Life Assessed in a Randomized Study. Journal of Alternative and Complementary Medicine. 2018. Vol. 24:954–961

            181. Kwon YS, Chun SY, Kim MK, Nan HY, Lee C, Kim S. Mistletoe Extract Targets the STAT3-FOXM1 Pathway to Induce Apoptosis and Inhibits Metastasis in Breast Cancer Cells. The American Journal of Chinese Medicine. 2021. Vol. 49:487–504

            182. Kozlowski HM, Pawlikowska M, Sobocinska J, Jedrzejewski T, Dzialuk A, Wrotek S. Distinct Modulatory Effects of Fever-Range Hyperthermia on the Response of Breast Cancer Cells and Macrophages to Mistletoe (Viscum album L.) Extract. Pharmaceuticals. 2021. Vol. 14:551

            183. De Jaeghere EA, Tuyaerts S, Van Nuffel AMT, Belmans A, Bogaerts K, Baiden-Amissah R, et al.. Pembrolizumab, Radiotherapy, and an Immunomodulatory Five-Drug Cocktail in Pretreated Patients with Persistent, Recurrent, or Metastatic Cervical or Endometrial Carcinoma: Results of the Phase II PRIMMO Study. Cancer Immunology, Immunotherapy: CII. 2023. Vol. 72:475–491

            184. Patino-Morales CC, Soto-Reyes E, Arechaga-Ocampo E, Ortiz-Sanchez E, Antonio-Vejar V, Pedraza-Chaverri J, et al.. Curcumin Stabilizes p53 by Interaction with NAD(P)H:quinone Oxidoreductase 1 in Tumor-Derived Cell Lines. Redox Biology. 2020. Vol. 28:101320

            185. Wang T, Wu X, Al Rudaisat M, Song Y, Cheng H. Curcumin Induces G2/M Arrest and Triggers Autophagy, ROS Generation and Cell Senescence in Cervical Cancer Cells. Journal of Cancer. 2020. Vol. 11:6704–6715

            186. Zhao X, Zhang R, Song Z, Yang K, He H, Jin L, et al.. Curcumin Suppressed the Proliferation and Apoptosis of HPV-Positive Cervical Cancer Cells by Directly Targeting the E6 Protein. Phytotherapy Research: PTR. 2023.

            187. Lee CY, Hsiao YH, Chen PN, Wu HH, Lu CY, Yang SF, et al.. CLEFMA Induces Intrinsic and Extrinsic Apoptotic Pathways through ERK1/2 and p38 Signalling in Uterine Cervical Cancer Cells. Journal of Cellular and Molecular Medicine. 2023. Vol. 27:446–455

            188. Lee CY, Ho YC, Lin CW, Hsin MC, Wang PH, Tang YC, et al.. EF-24 Inhibits TPA-Induced Cellular Migration and MMP-9 Expression through the p38 Signaling Pathway in Cervical Cancer Cells. Environmental Toxicology. 2023. Vol. 38:451–459

            189. Mohanty S, Huang J, Basu A. Enhancement of Cisplatin Sensitivity of Cisplatin-Resistant Human Cervical Carcinoma Cells by Bryostatin 1. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2005. Vol. 11:6730–6737

            190. Armstrong DK, Blessing JA, Rader J, Sorosky JI; Gynecologic Oncology Group S. A Randomized Phase II Evaluation of Bryostatin-1 (NSC #339555) in Persistent or Recurrent Squamous Cell Carcinoma of the Cervix: A Gynecologic Oncology Group Study. Investigational New Drugs. 2003. Vol. 21:453–457

            191. Nezhat F, Wadler S, Muggia F, Mandeli J, Goldberg G, Rahaman J, et al.. Phase II Trial of the Combination of Bryostatin-1 and Cisplatin in Advanced or Recurrent Carcinoma of the Cervix: A New York Gynecologic Oncology Group Study. Gynecologic Oncology. 2004. Vol. 93:144–148

            192. Garcia FA, Cornelison T, Nuno T, Greenspan DL, Byron JW, Hsu CH, et al.. Results of a Phase II Randomized, Double-Blind, Placebo-Controlled Trial of Polyphenon E in Women with Persistent High-Risk HPV Infection and Low-Grade Cervical Intraepithelial Neoplasia. Gynecologic Oncology. 2014. Vol. 132:377–382

            193. Dikshit A, Hales K, Hales DB. Whole Flaxseed Diet Alters Estrogen Metabolism to Promote 2-Methoxtestradiol-Induced Apoptosis in Hen Ovarian Cancer. The Journal of Nutritional Biochemistry. 2017. Vol. 42:117–125

            194. Pal P, Hales K, Petrik J, Hales DB. Pro-Apoptotic and Anti-Angiogenic Actions of 2-Methoxyestradiol and Docosahexaenoic Acid, the Biologically Derived Active Compounds from Flaxseed Diet, in Preventing Ovarian Cancer. Journal of Ovarian Research. 2019. Vol. 12:49

            195. Weston WC, Hales KH, Hales DB. Flaxseed Increases Animal Lifespan and Reduces Ovarian Cancer Severity by Toxically Augmenting One-Carbon Metabolism. Molecules. 2021. Vol. 26:5674

            196. Morgan RJ Jr, Leong L, Chow W, Gandara D, Frankel P, Garcia A, et al.. Phase II Trial of Bryostatin-1 in Combination with Cisplatin in Patients with Recurrent or Persistent Epithelial Ovarian Cancer: A California Cancer Consortium Study. Investigational New Drugs. 2012. Vol. 30:723–728

            197. Tuyaerts S, Rombauts K, Everaert T, Van Nuffel AMT, Amant F. A Phase 2 Study to Assess the Immunomodulatory Capacity of a Lecithin-Based Delivery System of Curcumin in Endometrial Cancer. Frontiers in Nutrition. 2018. Vol. 5:138

            198. Sirohi VK, Popli P, Sankhwar P, Kaushal JB, Gupta K, Manohar M, et al.. Curcumin Exhibits Anti-Tumor Effect and Attenuates Cellular Migration via Slit-2 Mediated Down-Regulation of SDF-1 and CXCR4 in Endometrial Adenocarcinoma Cells. The Journal of Nutritional Biochemistry. 2017. Vol. 44:60–70

            199. Zhang MJ, Shi M, Yu Y, Wang H, Ou R, Ge RS. CP41, a Novel Curcumin Analogue, Induces Apoptosis in Endometrial Cancer Cells by Activating the H3F3A/Proteasome-MAPK Signaling Pathway and Enhancing Oxidative Stress. Life Sciences. 2024. Vol. 338:122406

            200. Ollberding NJ, Lim U, Wilkens LR, Setiawan VW, Shvetsov YB, Henderson BE, et al.. Legume, Soy, Tofu, and Isoflavone Intake and Endometrial Cancer Risk in Postmenopausal Women in the Multiethnic Cohort Study. Journal of the National Cancer Institute. 2012. Vol. 104:67–76

            201. Yoriki K, Mori T, Aoyama K, Tarumi Y, Kataoka H, Kokabu T, et al.. Genistein Induces Long-Term Expression of Progesterone Receptor Regardless of Estrogen Receptor Status and Improves the Prognosis of Endometrial Cancer Patients. Scientific Reports. 2022. Vol. 12:10303

            202. Golabek-Grenda A, Kaczmarek M, Juzwa W, Olejnik A. Natural Resveratrol Analogs Differentially Target Endometriotic Cells into Apoptosis Pathways. Scientific Reports. 2023. Vol. 13:11468

            203. Wang YL, Shen Y, Xu JP, Han K, Zhou Y, Yang S, et al.. Pterostilbene Suppresses Human Endometrial Cancer Cells In Vitro by Down-Regulating miR-663b. Acta Pharmacologica Sinica. 2017. Vol. 38:1394–1400

            204. Motzer RJ, Jonasch E, Agarwal N, Alva A, Baine M, Beckermann K, et al.. Kidney Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network: JNCCN. 2022. Vol. 20:71–90

            205. Owens B. Kidney Cancer. Nature. 2016. Vol. 537:S97

            206. Li E, Zhang T, Sun X, Li Y, Geng H, Yu D, et al.. Sonic Hedgehog Pathway Mediates Genistein Inhibition of Renal Cancer Stem Cells. Oncology Letters. 2019. Vol. 18:3081–3091

            207. Buonerba C, De Placido P, Bruzzese D, Pagliuca M, Ungaro P, Bosso D, et al.. Isoquercetin as an Adjunct Therapy in Patients With Kidney Cancer Receiving First-Line Sunitinib (QUASAR): Results of a Phase I Trial. Frontiers in Pharmacology. 2018. Vol. 9:189

            208. Wu P, Wang X, Ma Y, Xu X, Liu W, Sheng Z, et al.. (3E,5E)-3,5-Bis(Pyridin-3-Methylene)-Tetrahydrothiopyran-4-One Enhances the Inhibitory Effect of Gemcitabine on Pancreatic Cancer Cells. Bioorganic Chemistry. 2020. Vol. 101:104022

            209. Delitto D, Wallet SM, Hughes SJ. Targeting Tumor Tolerance: A New Hope for Pancreatic Cancer Therapy? Pharmacology & Therapeutics. 2016. Vol. 166:9–29

            210. Pastorelli D, Fabricio ASC, Giovanis P, D’Ippolito S, Fiduccia P, Soldà C, et al.. Phytosome Complex of Curcumin as Complementary Therapy of Advanced Pancreatic Cancer Improves Safety and Efficacy of Gemcitabine: Results of a Prospective Phase II Trial. Pharmacological Research. 2018. Vol. 132:72–79

            211. Liu P, Ying Q, Liu H, Yu SQ, Bu LP, Shao L, et al.. Curcumin Enhances Anti-Cancer Efficacy of Either Gemcitabine or Docetaxel on Pancreatic Cancer Cells. Oncology Reports. 2020. Vol. 44:1393–1402

            212. Chen H, Jiang Y, Liu R, Deng J, Chen Q, Chen L, et al.. Curcumin Derivative C66 Suppresses Pancreatic Cancer Progression through the Inhibition of JNK-Mediated Inflammation. Molecules. 2022. Vol. 27:3076

            213. Zhai LL, Li WB, Chen LJ, Wang W, Ju TF, Yin DL. Curcumin Inhibits the Invasion and Migration of Pancreatic Cancer Cells by Upregulating TFPI-2 to Regulate ERK- and JNK-Mediated Epithelial-Mesenchymal Transition. European Journal of Nutrition. 2024. Vol. 63:639–651

            214. Malhotra L, Sharma S, Hariprasad G, Dhingra R, Mishra V, Sharma RS, et al.. Mechanism of Apoptosis Activation by Curcumin Rescued Mutant p53Y220C in Human Pancreatic Cancer. Biochimica et Biophysica Acta Molecular Cell Research. 2022. Vol. 1869:119343

            215. Malhotra L, Goyal HKV, Jhuria S, Dev K, Kumar S, Kumar M, et al.. Curcumin Rescue p53Y220C in BxPC-3 Pancreatic Adenocarcinomas Cell Line: Evidence-Based on Computational, Biophysical, and In Vivo Studies. Biochimica et Biophysica Acta General Subjects. 2021. Vol. 1865:129807

            216. Lohr JM, Karimi M, Omazic B, Kartalis N, Verbeke CS, Berkenstam A, et al.. A Phase I dose Escalation Trial of AXP107-11, a Novel Multi-Component Crystalline form of Genistein, in Combination with Gemcitabine in Chemotherapy-Naive Patients with Unresectable Pancreatic Cancer. Pancreatology: Official Journal of the International Association of Pancreatology. 2016. Vol. 16:640–645

            217. Mesmar F, Dai B, Ibrahim A, Hases L, Jafferali MH, Jose Augustine J, et al.. Clinical Candidate and Genistein Analogue AXP107-11 has Chemoenhancing Functions in Pancreatic Adenocarcinoma through G Protein-Coupled Estrogen Receptor Signaling. Cancer Medicine. 2019. Vol. 8:7705–7719

            218. Li T, Kuang T, Yang Z, Zhang Q, Zhang W, Fan Y. Co-Treatment with Everolimus, an mTOR-Specific Antagonist, or Downregulation of ELK1 Enhances the Sensitivity of Pancreatic Cancer Cells to Genistein. Frontiers in Cell and Developmental Biology. 2021. Vol. 9:633035

            219. Bi YL, Min M, Shen W, Liu Y. Genistein Induced Anticancer Effects on Pancreatic Cancer Cell Lines Involves Mitochondrial Apoptosis, G(0)/G(1)Cell Cycle Arrest and Regulation of STAT3 Signalling Pathway. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. 2018. Vol. 39:10–16

            220. Yang Y, Huynh N, Dumesny C, Wang K, He H, Nikfarjam M. Cannabinoids Inhibited Pancreatic Cancer via P-21 Activated Kinase 1 Mediated Pathway. International Journal of Molecular Sciences. 2020. Vol. 21:8035

            221. Alfonzetti T, Moreau M, Yasmin-Karim S, Ngwa W, Avery S, Goia D. Phytoradiotherapy to Enhance Cancer Treatment Outcomes with Cannabidiol, Bitter Melon Juice, and Plant Hemoglobin. Frontiers in Oncology. 2022. Vol. 12:1085686

            222. Luongo M, Marinelli O, Zeppa L, Aguzzi C, Morelli MB, Amantini C, et al.. Cannabidiol and Oxygen-Ozone Combination Induce Cytotoxicity in Human Pancreatic Ductal Adenocarcinoma Cell Lines. Cancers. 2020. Vol. 12:2774

            223. Garofano F, Sharma A, Abken H, Gonzalez-Carmona MA, Schmidt-Wolf IGH. A Low Dose of Pure Cannabidiol Is Sufficient to Stimulate the Cytotoxic Function of CIK Cells without Exerting the Downstream Mediators in Pancreatic Cancer Cells. International Journal of Molecular Sciences. 2022. Vol. 23:3783

            224. Bilim V, Kuroki H, Shirono Y, Murata M, Hiruma K, Tomita Y. Advanced Bladder Cancer: Changing the Treatment Landscape. Journal of Personalized Medicine. 2022. Vol. 12:1745

            225. Huang L, He C, Zheng S, Wu C, Ren M, Shan Y. AKT1/HK2 Axis-Mediated Glucose Metabolism: A Novel Therapeutic Target of Sulforaphane in Bladder Cancer. Molecular Nutrition & Food Research. 2022. Vol. 66:e2100738

            226. Wang F, Liu P, An H, Zhang Y. Sulforaphane Suppresses the Viability and Metastasis, and Promotes the Apoptosis of Bladder Cancer Cells by Inhibiting the Expression of FAT-1. International Journal of Molecular Medicine. 2020. Vol. 46:1085–1095

            227. Justin S, Rutz J, Maxeiner S, Chun FK, Juengel E, Blaheta RA. Bladder Cancer Metastasis Induced by Chronic Everolimus Application Can Be Counteracted by Sulforaphane In Vitro. International Journal of Molecular Sciences. 2020. Vol. 21:5582

            228. Justin S, Rutz J, Maxeiner S, Chun FK, Juengel E, Blaheta RA. Chronic Sulforaphane Administration Inhibits Resistance to the mTOR-Inhibitor Everolimus in Bladder Cancer Cells. International Journal of Molecular Sciences. 2020. Vol. 21:4026

            229. Xie H, Rutz J, Maxeiner S, Grein T, Thomas A, Juengel E, et al.. Plant-Derived Sulforaphane Suppresses Growth and Proliferation of Drug-Sensitive and Drug-Resistant Bladder Cancer Cell Lines In Vitro. Cancers. 2022. Vol. 14:4682

            230. Zhou Q, Jin P, Liu J, Li S, Liu W, Xi S. HER2 Overexpression Triggers the IL-8 to Promote Arsenic-Induced EMT and Stem Cell-Like Phenotypes in Human Bladder Epithelial Cells. Ecotoxicology and Environmental Safety. 2021. Vol. 208:111693

            231. Guo W, Wang H, Li C. Signal Pathways of Melanoma and Targeted Therapy. Signal Transduction and Targeted Therapy. 2021. Vol. 6:424

            232. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA: A Cancer Journal for Clinicians. 2021. Vol. 71:7–33

            233. Tahata S, Singh SV, Lin Y, Hahm ER, Beumer JH, Christner SM, et al.. Evaluation of Biodistribution of Sulforaphane after Administration of Oral Broccoli Sprout Extract in Melanoma Patients with Multiple Atypical Nevi. Cancer Prevention Research. 2018. Vol. 11:429–438

            234. Eom YS, Shah FH, Kim SJ. Sulforaphane Induces Cell Differentiation, Melanogenesis and also Inhibit the Proliferation of Melanoma Cells. European Journal of Pharmacology. 2022. Vol. 921:174894

            235. Arcidiacono P, Stabile AM, Ragonese F, Pistilli A, Calvieri S, Bottoni U, et al.. Anticarcinogenic Activities of Sulforaphane are Influenced by Nerve Growth Factor in Human Melanoma A375 Cells. Food and Chemical Toxicology: An International Journal Published for the British Industrial Biological Research Association. 2018. Vol. 113:154–161

            236. Morceau F, Chateauvieux S, Orsini M, Trecul A, Dicato M, Diederich M. Natural Compounds and Pharmaceuticals Reprogram Leukemia Cell Differentiation Pathways. Biotechnology Advances. 2015. Vol. 33:785–797

            237. Olivas-Aguirre M, Torres-Lopez L, Pottosin I, Dobrovinskaya O. Phenolic Compounds Cannabidiol, Curcumin and Quercetin Cause Mitochondrial Dysfunction and Suppress Acute Lymphoblastic Leukemia Cells. International Journal of Molecular Sciences. 2020. Vol. 22:204

            238. Olivas-Aguirre M, Perez-Chavez J, Torres-Lopez L, Hernandez-Cruz A, Pottosin I, Dobrovinskaya O. Dexamethasone-Induced Fatty Acid Oxidation and Autophagy/Mitophagy Are Essential for T-ALL Glucocorticoid Resistance. Cancers. 2023. Vol. 15:445

            239. Koszalka P, Stasilojc G, Miekus-Purwin N, Niedzwiecki M, Purwin M, Grabowski S, et al.. The Cooperative Anti-Neoplastic Activity of Polyphenolic Phytochemicals on Human T-Cell Acute Lymphoblastic Leukemia Cell Line MOLT-4 In Vitro. International Journal of Molecular Sciences. 2022. Vol. 23:4753

            240. Liu JM, Li M, Luo W, Sun HB. Curcumin Attenuates Adriamycin-Resistance of Acute Myeloid Leukemia by Inhibiting the lncRNA HOTAIR/miR-20a-5p/WT1 Axis. Laboratory Investigation; A Journal of Technical Methods and Pathology. 2021. Vol. 101:1308–1317

            241. Gaur T, Ali A, Sharma D, Gupta SK, Gota V, Bagal B, et al.. Mitocurcumin Utilizes Oxidative Stress to Upregulate JNK/p38 Signaling and Overcomes Cytarabine Resistance in Acute Myeloid Leukemia. Cell Signal. 2024. Vol. 114:111004

            242. Bistue-Rovira A, Rico LG, Bardina J, Junca J, Granada I, Bradford JA, et al.. Persistence of Chronic Lymphocytic Leukemia Stem-like Populations under Simultaneous In Vitro Treatment with Curcumin, Fludarabine, and Ibrutinib: Implications for Therapy Resistance. International Journal of Molecular Sciences. 2024. Vol. 25:1994

            243. Feriotto G, Marchetti P, Rondanin R, Tagliati F, Aguzzi S, Beninati S, et al.. Cytotoxicity of Isoxazole Curcumin Analogs on Chronic Myeloid Leukemia-Derived K562 Cell Lines Sensitive and Resistant to Imatinib. International Journal of Molecular Sciences. 2023. Vol. 24:2356

            244. Hsiao PC, Chang JH, Lee WJ, Ku CC, Tsai MY, Yang SF, et al.. The Curcumin Analogue, EF-24, Triggers p38 MAPK-Mediated Apoptotic Cell Death via Inducing PP2A-Modulated ERK Deactivation in Human Acute Myeloid Leukemia Cells. Cancers. 2020. Vol. 12:2163

            245. Fan YJ, Zhou YX, Zhang LR, Lin QF, Gao PZ, Cai F, et al.. C1206, a Novel Curcumin Derivative, Potently Inhibits Hsp90 and Human Chronic Myeloid Leukemia Cells In Vitro. Acta Pharmacologica Sinica. 2018. Vol. 39:649–658

            246. Liu B, Shen Y, Huang H, Croce KD, Wu M, Fan Y, et al.. Curcumin Derivative C212 Inhibits Hsp90 and Eliminates both Growing and Quiescent Leukemia Cells in Deep Dormancy. Cell Communication and Signaling: CCS. 2020. Vol. 18:159

            247. Sawesi S, Malkaram SA, Abd Elmageed ZY, Fandy TE. Modulation of the Activity of Histone Lysine Methyltransferases and Demethylases by Curcumin Analog in Leukaemia Cells. Journal of Cellular and Molecular Medicine. 2022. Vol. 26:5624–5633

            248. Mugnaini EN, Ghosh N. Lymphoma. Primary Care. 2016. Vol. 43:661–675

            249. Zhao Q, Guan J, Qin Y, Ren P, Zhang Z, Lv J, et al.. Curcumin Sensitizes Lymphoma Cells to DNA Damage Agents through Regulating Rad51-Dependent Homologous Recombination. Biomedicine & Pharmacotherapy. 2018. Vol. 97:115–119

            250. Zhang Y, Xiang J, Zhu N, Ge H, Sheng X, Deng S, et al.. Curcumin in Combination with Homoharringtonine Suppresses Lymphoma Cell Growth by Inhibiting the TGF-beta/Smad3 Signaling Pathway. Aging. 2021. Vol. 13:18757–18768

            251. Ge X, Du Y, Chen J, Zhu N, Yao J, Zhang X, et al.. Herbal NF-kappaB Inhibitors Sensitize Rituximab-Resistant B Lymphoma Cells to Complement-Mediated Cytolysis. Frontiers in Oncology. 2021. Vol. 11:751904

            252. Kovacs Z, Guttman A. Utilization of Analytical Omics Tools in the Molecular Diagnostics of Multiple Myeloma. Current Molecular Medicine. 2018. Vol. 18:260–272

            253. Santosa D, Suharti C, Riwanto I, Dharmana E, Pangarsa EA, Setiawan B, et al.. Curcumin as Adjuvant Therapy to Improve Remission in Myeloma Patients: A Pilot Randomized Clinical Trial. Caspian Journal of Internal Medicine. 2022. Vol. 13:375–384

            254. Booth L, Roberts JL, West C, Dent P. GZ17-6.02 Interacts with Proteasome Inhibitors to Kill Multiple Myeloma Cells. Oncotarget. 2024. Vol. 15:159–174

            255. Chen J, Ruijtenbeek R, Garssen J, Redegeld FA. Esterified Derivatives of DHA and EPA Increase Bortezomib Cytotoxicity in Human Multiple Myeloma Cells. European Journal of Pharmacology. 2022. Vol. 922:174883

            256. Chen J, Garssen J, Redegeld F. The Efficacy of Bortezomib in Human Multiple Myeloma Cells is Enhanced by Combination with Omega-3 Fatty Acids DHA and EPA: Timing is Essential. Clinical Nutrition. 2021. Vol. 40:1942–1953

            257. Chen J, Zaal EA, Berkers CR, Ruijtenbeek R, Garssen J, Redegeld FA. Omega-3 Fatty Acids DHA and EPA Reduce Bortezomib Resistance in Multiple Myeloma Cells by Promoting Glutathione Degradation. Cells. 2021. Vol. 10:2287

            258. Mekkawy SA, Abdalla MS, Omran MM, Hassan NM, Abdelfattah R, Abdel-Salam IM. Cancer Stem Cells as a Prognostic Biomarker and Therapeutic Target Using Curcumin/Piperine Extract for Multiple Myeloma. Asian Pacific Journal of Cancer Prevention: APJCP. 2022. Vol. 23:3507–3515

            259. Garofano F, Schmidt-Wolf IGH. High Expression of Cannabinoid Receptor 2 on Cytokine-Induced Killer Cells and Multiple Myeloma Cells. International Journal of Molecular Sciences. 2020. Vol. 21:3800

            260. Chen Q, Shu C, Laurence AD, Chen Y, Peng BG, Zhen ZJ, et al.. Effect of Huaier Granule on Recurrence after Curative Resection of HCC. A Multicentre, Randomised Clinical Trial. Gut. 2018. Vol. 67:2006–2016

            261. Luo S, Hu H. Huaier Granule Prolongs Overall Survival after Curative Resection of Hepatocarcinoma Carcinoma: A Propensity Score Analysis. Journal of Ethnopharmacology. 2023. Vol. 301:115774

            262. Shi K, Bi Y, Zeng X, Wang X. Effects of Adjuvant Huaier Granule Therapy on Survival Rate of Patients with Hepatocellular Carcinoma. Frontiers in Pharmacology. 2023. Vol. 14:1163304

            263. Zhou L, Pan LC, Zheng YG, Du GS, Fu XQ, Zhu ZD, et al.. Novel Strategy of Sirolimus Plus Thymalfasin and Huaier Granule on Tumor Recurrence of Hepatocellular Carcinoma Beyond the UCSF Criteria Following Liver Transplantation: A Single Center Experience. Oncology Letters. 2018. Vol. 16:4407–4417

            264. Gullo I, Grillo F, Mastracci L, Vanoli A, Carneiro F, Saragoni L, et al.. Precancerous Lesions of the Stomach, Gastric Cancer and Hereditary Gastric Cancer Syndromes. Pathologica. 2020. Vol. 112:166–185

            265. Zhu Y, Ouyang Z, Du H, Wang M, Wang J, Sun H, et al.. New Opportunities and Challenges of Natural Products Research: When Target Identification Meets Single-Cell Multiomics. Acta Pharmaceutica Sinica B. 2022. Vol. 12:4011–4039

            266. Li T, Pan J, Chen H, Fang Y, Sun Y. CXCR6-Based Immunotherapy in Autoimmune, Cancer and Inflammatory Infliction. Acta Pharmaceutica Sinica B. 2022. Vol. 12:3255–3262

            267. Wang ZY, Chen Z. Acute Promyelocytic Leukemia: From Highly Fatal to Highly Curable. Blood. 2008. Vol. 111:2505–2515

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            31 May 2024
            : 3
            : 2
            : 163-206
            Affiliations
            [a ]Zhejiang Hospital, Hangzhou 310013, China
            [b ]Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
            [c ]State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing 210023, China
            [d ]The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
            Author notes
            *Correspondence: yangsun@ 123456nju.edu.cn , Tel./Fax: +86-25-89687620, (Y. Sun); zjszyyyy@ 123456163.com , Tel./Fax: +0571-87072976 (W. Zhou)

            1These authors contributed equally to this work.

            Article
            10.15212/AMM-2023-0050
            0a6a9ac8-c0fb-47e1-970a-ad8b236486d2
            Copyright © 2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 21 December 2023
            : 19 March 2024
            : 30 April 2024
            Page count
            Figures: 2, Tables: 6, References: 267, Pages: 44
            Funding
            Funded by: National Key Research and Development Plan
            Award ID: 2022YFC3500202
            Funded by: Program A for Outstanding PhD candidate of Nanjing University
            Award ID: 202202A004
            Funded by: Zhejiang Provincial Natural Science Foundation of China
            Award ID: LYY22H310004
            Funded by: Zhejiang Medical and Health Science and Technology Plan Project of China
            Award ID: 2024KY1205
            Funded by: Science and Technology in Zhejiang Province Chinese Medicine Program
            Award ID: 2024ZL219
            Funded by: Pharmaceutical Association Hospital Pharmacy Special Scientific Researches
            Award ID: 2020ZYY18
            Funded by: Pharmaceutical Association Hospital Pharmacy Special Scientific Researches
            Award ID: 2022ZYY08
            Funded by: Key Projects of Nanjing Medical Science and Technology
            Award ID: ZKX21015
            This research was funded by the National Key Research and Development Plan (No. 2022YFC3500202), Program A for Outstanding PhD candidate of Nanjing University (No. 202202A004), Joint Funds of the Zhejiang Provincial Natural Science Foundation of China (No. LYY22H310004), Zhejiang Medical and Health Science and Technology Plan Project of China (No. 2024KY1205), Science and Technology in Zhejiang Province Chinese Medicine Program (No. 2024ZL219), Zhejiang Grants Funded by Pharmaceutical Association Hospital Pharmacy Special Scientific Researches (No. 2020ZYY18, No. 2022ZYY08), and Key Projects of Nanjing Medical Science and Technology (No. ZKX21015).
            Categories

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            cancer,clinical trial,Natural products

            Comments

            Comment on this article