This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Lung cancer is a heterogeneous group of tumors, consisting of approximately 80 histologic, genetic, and molecularly defined subtypes, and non-small-cell lung cancer (NSCLC) accounts for approximately 85% of all these subtypes [1, 2]. WHO has classified NSCLC into 3 main types, that is, adenocarcinoma, squamous cell carcinoma, and large cell cancer, with adenocarcinoma (ADC, approximately 40–50% of cases), squamous cell carcinoma (SqCC, approximately 20–30% of cases), and large cell cancer (LCC, approximately 5–10% of cases) comprising the predominant histological subtypes of NSCLC [3, 4]. Genetic alterations, tobacco use (smoking and secondhand smoking), and environmental exposures, including air pollution, infection, and occupational exposures, have been demonstrated to be associated with the tumorigenesis of NSCLC. Based on the stage, histology, gene mutations, and patient’s condition, the treatments of NSCLC usually include the surgery, radiotherapy, chemotherapy, molecularly targeted therapy, and immunotherapy [5, 6]. However, NSCLC may reoccur and metastasize when there is a high risk that patients may harbor minimal residual [7]. Traditional cytotoxic chemotherapy and radiation therapy are the most common clinical treatment strategies but are always associated with many side effects [8]. Acquired drug resistance of NSCLC cells is always related to molecularly targeted therapy, and there are most toxicities as immune-related adverse events (irAEs), which resulted from excessive immunity against normal organs in clinical trials for immunotherapy [9–12]. Thus, it is necessary to find alternative therapeutic agents for NSCLC. Consequently, with more complicated elemental structures and abundant biological activities, TCM products primarily including TCM herbals or formulas have gradually become a huge resource base upon the development of anticancer drugs due to their intrinsic advantages [13–16].
TCM products have been used for the treatment of various diseases over thousands of year in eastern Asia [17, 18]. Since there is more recognition of these diseases or disorders, an extensive number of TCM products have been identified, and many useful drugs have been developed from these products, which have played wide roles in rejuvenation, cytoprotection, anti-inflammatory, anti-infection, antioxidation, anticancer, etc. [19–25]. Currently, TCM products and their biological activities are a subject of great interest in the pharmaceutical, health food, and cosmetics industries, and numbers of scientific studies in these field are increasing rapidly [26]. And a plenty of TCM products (all of extracts, formulas, and isolated single molecules) and their analogs and metabolites may serve as the shortcut or key resources of new drug candidates for anticancer therapy. Recent studies indicated that TCM products can be used as anticancer therapy with minimum side effects and can also be used as potential MDR modulators to overcome drug resistance [27–30]. In addition, many preclinical studies on NSCLC showed positive effects of TCM products including flavonoids, diterpenoids, triterpenoids, steroids, macrodiolides, and phenolics [31–35], so there is an urgent need for NSCLC animal models suitable for development of TCM products, which will shed light on the diversity of new drug discovery for NSCLC.
During the last decades, there has been a great effort in developing preclinical animal models for NSCLC, including cell lines-derived xenografts (CDX), genetically engineered mouse models (GEMM), and patient-derived xenografts (PDX). These models have been used to accelerate our understanding of NSCLC biology and pathogenesis [36, 37]. Because preliminarily preclinical toxicity and efficacy study in animal models are an important regulatory requirement in drug development to assess the safety and activity of the test TCM products prior to clinical evaluation, in this review, we focus on the role and development of kinds of mouse models in antitumor research and summarize that the efficacy and safety of TCM products for the treatment of NSCLC in mouse models. What deserves more attention is that we should take full advantage of mouse models to reexamine the important value of TCM products in NSCLC from a new perspective. Furthermore, the lack of systemic evaluation of pharmacology and toxicology using modern methods and the not-well-identified material base of TCM extracts or formulas are the major reasons why TCM extracts or formulas are not accepted as drugs by the western world [18]; thus, we also should seek for some kinds of mouse models, which are more suitable for development of TCM extracts or formulas when compared to synthetic chemical drugs.
2. Classification of Animal Models
The use of preclinical models is a core component in every aspect of translational cancer research ranging from the biological understanding of the disease to the development of new treatments, so insufficiencies in preclinical animal models are key factors in the high failure rates of oncology drug discovery and development [38]. An ideal animal model should allow the development of local tumors, it should replicate all stages of cancer progression, and finally, it should permit assessment of new therapeutic strategies. Since 1970s, screening of new drugs was performed in rapidly growing mouse models. Over the last 40–50 years, xenografts developed by growing cell lines or from fresh primary or metastatic human cancer tissues subcutaneously or orthotopically in immunodeficient mice are the most commonly used in vivo platform in preclinical drug development [39]. The classical models for cancer drug screening include CDXs, PDXs, cell line- or primary tumor-derived homografts in syngeneic mice, orthotopic patient-derived or cell line-derived xenografts, HIS, and GEMMs [40]. The flowchart of various models establishment is shown in Figure 1.
[figure(s) omitted; refer to PDF]
2.1. Cell-Derived Xenograft (CDX) Model
CDX mouse models are developed by implanting cell lines in mice to effectively grow tumors, which emerge from xenograft transplantations and are a mosaic of human cancer cells and mouse stromal cells. Furthermore, in order for human tumors to grow in mice, the host must be immunocompromised to prevent immune rejection [37]. There are several advantages with CDXs including relatively cost-effective and good reproducibility, high tumor-take rates, short turn-around time, and maintenance of molecular characteristics from the original cell line; thus, CDXs remain invaluable for large-scale drug screening, biomarker discovery, and PK/PD analysis of new drugs, as well as exploring drug resistance mechanisms [41, 42]. Presently, over 200 cell lines for NSCLC have been reported, and many NSCLC CDX mouse models were utilized to examine the effects of TCM products [43]. However, some problems still exist in CDX models: (i) CDX models cannot reconstitute complex human cancer elements such as the molecular heterogeneity of tumor cells and the tumor microenvironment, so approximately 88% of new drugs identified by CDXs fail in clinical research [44]. (ii) CDX models lack fully functional murine immune systems [45]. Therefore, actually, till now, cell-derived xenograft (CDX) models are mainly utilized to evaluate pharmacology, efficacy, and safety profiles of TCM monomers and synthetic chemical drugs at the early stage of preclinical research.
2.2. Patient-Derived Xenograft (PDX) Model
Patient-derived xenografts (PDX) are based on transferring primary tumors directly from the patient into an immunodeficient mouse. To establish patient-derived xenograft (PDX) model, patient tumors must be obtained freshly from surgery, mechanically or chemically digested at once, and transplanted in immunodeficient mouse while saving a small portion as a primary stock [46]. Patient‐derived xenograft models are considered superior to cell line‐derived xenograft (CDX) models in retaining the histological characteristics of patient tumors, such as tumor heterogeneity and tumor microenvironment, at least at early passages [47]. Along with the advancement of molecular and genomic analyses, researchers have focused on more details in individual cancer genes and the tumor microenvironment; thus, PDX models are more suitable for use in experiments exploring the sensitivity of drugs, which is more similar to clinical drug responsiveness, the molecular mechanisms of tumor progression, drugs resistance, PDX biobanks, and precision medicine by which the most effective drugs can be identified in individual PDX model [48, 49]. However, there are some limitations that PDX models are expensive with a low transplantation success rate and require a long tumor-take time [50]. Currently, the number of NSCLC PDX is increasing rapidly; it will be beneficial for a better understanding of the mechanisms by which NSCLC progresses and develops resistance to certain drugs especially for targeted small molecules including TCM monomers because the major challenge in the treatment of NSCLC patients is intrinsic or acquired resistance to chemotherapy [51]
2.3. Syngeneic Model
Syngeneic murine models are to implant murine-derived cancer lines or tissues on homozygouslyinbred murine strains. Herein, murine-derived cancer lines have either been derived from chemical-induced tumors or tumors that spontaneously generate in the same strain [50]. The advantage of syngeneic models is that the transplanted cancer lines or tissues, the tumor microenvironment, and the host are from the same species. This is extremely important when considering the close interaction between tumor and host characterized by the process of metastasis. Furthermore, syngeneic models are both time- and cost-effective and reproducible [52]. The disadvantage of syngeneic models is that their model systems are short of many important characteristics of human tumors. For instance, they are generally derived from homozygously inbred mice or rats and, therefore, lack the genetic complexity of human tumors. This allows for evaluation of toxicities, including on-target/off-tumor side effects, and the immunosuppressive microenvironments [53]. Now, due to the growing worldwide enthusiasm in cancer immunotherapy, syngeneic murine models are widely used tools for studying tumor immunity and immunotherapy response because there is a fully functional murine immune system [51, 54, 55].
2.4. Orthotopic Model
Orthotopic models were established by directly implanting tumor cells or fresh tumor tissues into mice in the anatomical location from which they were originally derived. As a result, the use of orthotopic implantation has resulted in tumor models resembling human cancer with respect to the biology and tumor microenvironment [56, 57]. Therefore, orthotopic models allow the evaluation of tumor behavior in an organ-specific microenvironment. In addition, because the lack of metastasis and altered drug responses have been reported in commonly used subcutaneous models, the orthotopic models enhance the possibility of distant metastatic spread in a superior manner and allow to elucidate the role of certain molecular pathways by direct comparison of the metastatic properties of a given cell with their genetically engineered counterpart. The additional transfection of these cells with a reporter, like GFP, DsRed, or luciferase, allows monitoring of tumor progress by in vivo imaging [58, 59], which makes orthotopic models more valuable, but challenging to construct and measure the tumor size. The 90% death of non-small-cell lung cancer (NSCLC) patients is primarily due to metastases, which are poorly amenable to therapeutic intervention [60]. Many studies have shown orthotopic engraftment of human NSCLC tumors in mice for the studies of metastasis [61–65]. Therefore, the remarkable advantages of orthotopic transplantation facilitate NSCLC cancer growth in a more natural environment, which can mimic an eventual onset and spreading of metastases similar to those found in men.
2.5. Humanized Mouse Models (HIS)
Humanized mouse models, which are engrafted with functional human cells and tissues, including human peripheral blood mononuclear cells (PBMCs), CD34+ human hematopoietic cells, or bone marrow-liver-thymus (BLT) tissue, are widely used for the biomedical research [66]. The NSG, NCG, and NOG mouse strains are suitable for creating humanized mice because they lack T-cell subsets, B cells, and natural killer (NK) cells. For example, Hu-PBL-NSG mouse model was established by engrafting human peripheral blood mononuclear cells (PBMCs) into NOD/SCID/IL-2Rγ−/− (NSG) mice. Then, an ALK positive cell line, H3122 (EML4-ALK variant 1), or a patient-derived tumor from EML4-ALK-rearranged (EML4-ALK variant 1) NSCLC patient was transplanted in NSG mice [48]. When engrafted with human peripheral blood mononuclear cells, tumor tissues, and immune systems, the humanized mice could recapitulate the biological responses faithfully. The humanized mouse models are now being widely used to study many human biological responses and diseases and are increasingly utilized as preclinical tools for evaluation of drugs and for identifying underlying mechanisms in the aspect of tumor immunotherapy. The recent clinical successes of immune checkpoint blockade and chimeric antigen receptor T cell therapies inspired a turning interest in cancer immunotherapy. Humanized mouse models could be a powerful animal model for testing efficacy of immunotherapeutic agents including TCM products. However, many humanized mice have a short experimental window because of the development of graft-versus-host disease (GVHD), usually within 4–8 weeks [51, 52]. Additionally, not all immune cell populations can be reconstituted in this system [67].
2.6. Genetically Engineered Mouse Models (GEMMs)
GEMMs of cancer represent a diverse collection of genetically modified mice, which are engineered to carry cloned oncogenes or lack tumor-suppressing genes and allow the investigation of human disease-associated genetic abnormalities in vivo. Such models can be divided into two forms: Germ-Line genetically engineered models, which develop cancers in an unintended (spontaneous) fashion, and conditional genetically engineered models, which provide regulated control of tumor onset utilizing tissue-specific, ligand-regulated, and/or viral-based technologies [53]. Presently, GEMMs for most of the common NSCLC driver mutations have been generated, including KRAS (KrasLSL − G12D/+, KrasLSL − G12D/ + /Tp53mutants, KrasLSL − G12D/+/Lkb1F/F, Lkb1F/−, Lkb1F/+ or Lkb1+/−, KrasLSL − G12D/+/Cdkn2aF/F or −/−, Tet-op Kras4bG12D/CCSP-rtTA), epidermal growth factor receptor (EGFR) (Tet-op EGFRL858R/CCSP-rtTA, Tet-op EGFRL858R + T790M/CCSP-rtTA, Tet-op EGFR∆E746 − A750 + T790M/CCSP-rtTA, Tet-op EGFRT790M/CCSP-rtTA), phosphoinositide-3-kinase catalytic alpha polypeptide (PIK3CA) (Tet-op PIK3CAH1047R/CCSP-rtTA) and echinoderm microtubule-associatedprotein-like 4 (EML4)–anaplastic lymphoma kinase (ALK)[ LSL-EML4-ALKL1196M/+ or F1174L/+, Tet-op EML4-ALK (Variant 1)/CCSP-rtTA, SPC-EML4-ALK (variant 1)] [68–70]. However, there are several disadvantages to GEMMs, including the relatively long period needed for GEMMs to develop tumor and the unpredictable nature of tumor development with regard to frequency and latency of tumorgenesis [71], which can become a drawback for preclinical evaluation of test articles including TCM.
3. Animal Models for Analysis and Testing of TCM Products as Chemotherapy Agents
The application of animal models has contributed remarkably to drug development. Mouse models are the most extensively used in preclinical NSCLC studies for in vivo analysis of tumor response to chemotherapy, which is defıned as a cytotoxic and systemic therapy that disrupts basic cellular processes such as proliferation, metastasis, maintenance, apoptosis, and angiogenesis, not just those with oncogenic drivers. Traditional Chinese Medicine products have been proved effective in cancer therapy by preventing recurrence and development of multidrug resistance with tolerable side effect and alleviated detrimental symptoms caused by standard chemotherapy. Many studies that included all NSCLC subtypes have successfully evaluated TCM products in mouse models including CDX, PDX, and orthotopic and GEMM models, and these TCM products have cytotoxic properties with many different mechanisms of action, such as the inhibition of tumor cell growth, the induction of apoptosis, DNA damage, and the inhibition of topoisomerases I and II (see Table 1).
Table 1
List of TCM products as chemotherapy agents reported anticancer effects in NSCLC models.
TCM products | Origin | NSCLC models | Underlying mechanisms |
Taxanes including paclitaxel and docetaxel | Paclitaxel derived from the bark of the Pacific yew, docetaxel is a semi-synthetic taxane derived from extracts of the European yew tree Taxus baccata | A549 (Adenocarcinoma), H1299 (Lack expression of p53 protein) | Tubulin inhibitor, which interferes with the normal function of cellular microtubule growth by binding to the β-subunits of the tubulin and locking the microtubules preventing further cell division [33, 72, 73]. |
Vincaleucoblastine including vindesine and vinorelbine | Vindesine is a vinca-alkaloid derived from vinblastine, vinorelbine is a semisynthetic vinca-alkaloid | Lu-65 (Lung large cell carcinoma) and PC-12 (Adenocarcinoma); the EGFR-wild-type A549 and the EGFR-mutated (exon 21 L858R/exon 20 T790M) H1975 [74] | Tubulin inhibitor, the mechanism of action is the inhibition of tubulin polymerization into microtubules. |
Evodiamine | A novel alkaloid, isolated from the fruit of Tetradium | H1975 (Adenocarcinoma) | NF-kB inhibitor, by downregulation of MUC1-C [75–77]. |
Honokiol | Purified from the magnolia tree | PC‐9/AR (EGFR _19del), PC‐9/3M [(EGFR-19del, T790M, C797S (cis)], A549,H1299, acquired resistance to cetuximab in NSCLC patient-derived xenograft (PDX) model | By overcoming Osim acquired resistance [78], being associated with induction of apoptotic cell death and inhibition of class I HDACs proteins and HDAC activity [79, 80]. |
Sinomenine | Purified from the roots of the plant Sinomenium acutum | HCC827 (Adenocarcinoma), H1975 | By reducing HK2-mediated glycolysis [81]. |
Formononetin | Originated mainly from red clovers and the Chinese herb Astragalus membranaceus | HCC827, H3255 (Adenocarcinoma), H1975, A549, H1299 [82] | By inducing cell cycle arrest, apoptosis, antiangiogenesis, and metastasis [83]. |
Quercetin | As the active compound from Yang-Yin-Qing-Fei-Tang (YYQFT), which is a well-known traditional Chinese medicine | A549, HCC827 | Through induction of apoptosis or autophagy, enhancement of chemosensitivity, and modulation of cancer stemness [84, 85]. |
Isoharringtonine (IHT) | As an alkaloid extracted from the leaves of Cephalotaxus koreana Nakai. | A549 | Inhibition of the growth of tumor spheroids, induced apoptotic cell death via the Intrinsic pathway [86]. |
MTE (M. Tenacissima extract) | Extracted from Marsdenia tenacissima (Roxb.) Wight & Arn | H1975 | Inhibiting ABCG2 activity, by elevating production of NO, in a PKA-dependent manner [87, 88]. |
Chlorella sorokiniana | Extracted from Chlorella | CL1-5 (Adenocarcinoma) | Downregulation of Bcl-2, XIAP and survival [89]. |
Yuanhuadine (YD; purity >98.5%) | Isolated from a CHCl3-soluble fraction of the flowers of Daphne genkwa | Acquired Gefitinib-resistantpatient-derived xenograft (PDX) model | By complete suppression of the AXL activation [90]. |
Feiji Recipe | As a classical herbal recipe | 2LL-EGFP-Ido orthotopic xenografts | By reducing IDO expression [91]. |
Cyclopamine tartrate (CycT), an improved analogue of Cyclopamine | Cyclopamine produced by Veratrum californicum being a rich source of steroidal alkaloids and growing in high mountain meadows | Orthotopically implanted H1299-luc NSCLC tumor xenografts | Inhibitor of the hedgehog (hh) signaling pathway [92–94]. |
Baicalein | Derived from the root of Scutellaria | A549 orthotopic xenografts | By inhibition of Id1 expression [89]. |
Yi-Fei-Jie-Du-Tang (YFJDT) | As a traditional Chinese medicine formula | A549 | Through upregulating FAT4, promoting autophagy [16]. |
Theabrownin | Derived from the green tea | H1299 (p53-deficient) and A549 (p53-wild type) | By the activation of MAPK/JNK signaling pathway [95]. |
Ze-Qi-Tang formula | As a traditional Chinese medicine | LLC orthotopic mouse model | By inducing apoptosis via STAT3/S100A9/Bcl-2/caspase-3 signaling [96]. |
BL02 formula | Consists of two herbs: Gentiana rhodantha Franch. Ex Hemsl and Gerbera anandria | HCC827 and A549 orthotopic xenografts | Inhibition of Rap1/cdc42 signaling [97]. |
Nevertheless, most of these TCM products, usually acting as isolated single molecules, which are similar to synthetic chemical drugs [18], may also increase the risk of TCM molecules induced adverse effects. Simultaneously, long history has proved TCM herbals or formulas to be of benefit over the risk of induced adverse effects, but it is noticed that TCM herbals or formulas mainly play a supporting role in the main treatment.
4. Animal Models for Analysis and Testing of TCM Products as Targeted Therapy Agents
There is now a better understanding of the role of driver mutations in NSCLC and how to target these mutations in treatment of NSCLC, for example, epidermal growth factor receptor (EGFR) mutation, ROS1-rearranged, BRAFV600E mutation, KRAS mutation, and anaplastic lymphoma kinase (ALK) fusion oncogene [98]. Although currently applicable to only a minority of patients with NSCLC, this development has resulted in prominent successes that have led to approved molecularly specific, biomarker-defined indications for targeted therapies. However, most of the available anticancer agents are designed to target specific driver mutations by altering the activity of involved transporters and genes. As cancer cells demonstrate complex cellular machinery, the regeneration of cancer tissues and drug resistance towards the therapy has been the main barrier in cancer treatment [99]. A plenty of TCM products, as a valuable and huge resource pool, exhibited multitargeted characteristics of antioxidant, antimitotic, anti-inflammatory, antibiotic, and anticancer activity. Therefore, this fact encourages the researchers to explore the multitargeted use of TCM products especially for monomers to overcome the shortcomings and seek alternative and safer treatment strategies. Many preclinical studies included the fact that TCM monomers were used in NSCLC treatment for multitargeted approach (see Table 2).
Table 2
List of TCM products as targeted therapy agents reported anticancer effects in NSCLC models.
Natural products | Origin | NSCLC models | Underlying mechanisms |
Tanshinone IIA | Isolated from the rhizome of the Chinese herb Salvia miltiorrhiza Bunge (danshen) | HCC827/gefitinib _resistant, A549 [100] | VEGFR inhibitor, via regulation of VEGFR/Akt pathway [101], suppression of EGFR signaling [102, 103] and downregulation of the expression levels of Bcl‐2, Caspase‐3, p‐Akt, and p‐PI3K proteins [104] |
Parthenolide | Originated from the medicinal plant feverfew (Tanacetum parthenium) | H1975, H460, A549 | By targeting EGFR through downregulation of ERK and AKT expression [105] |
Hydroxygenkwanin (HGK) | One of the active flavonoids extracted from the flower buds of Daphne genkwa Sieb.et Zucc. | H1975 | By inducing the proteasome-mediated degradation of EGFR, thus inhibiting EGFR-downstream signaling and inducing apoptosis [106] |
Deguelin | Derived from leguminous plants | HCC827, H1975, A549 and H3255 | By inhibiting EGFR signaling and promoting GSK3β/FBW7-mediated Mcl-1 destabilization. [107, 108] |
Baicalein | Derived from the root of Scutellaria baicalensis | H1299 | Binding to MAP4K3 and degradation of MAP4K3 [109] |
Diaporine A (D261) | Derived from the culture broth of endophytic fungus Diaporthe sp. 3lp-10 | NCI–H460 | By regulating miR-99a/mTOR signaling [110] |
Thevebioside | An active ingredient from the seed of Thevetia peruviana (pers) K. Schum (TPKS). | A549 | By inhibiting SRC mediated IGF-1R–PI3K-AKT signaling [111]. |
Compounds of tanshinone (CTN) | Extracted from Salvia miltiorrhiza Bunge roots | GLC-82 (Adenocarcinoma) | By inducing apoptosis through the mitochondrial pathway of apoptosis and PTEN-mediated inhibition of PI3K/Akt pathway [112] |
Toona sinensis leaf extract | Extracted from Toona sinensis leaves | H441 (Adenocarcinoma), H520 (squamous cell carcinoma) and H661 (Large cell cancer) | Downregulation of cyclin D1 and CDK4 by increasing the expression of cyclin-dependent inhibitor p27 [89] |
5. Animal Models for Analysis and Testing of TCM Products as Immunotherapy Agents
Lung cancer has traditionally been considered as a “nonimmunogenic” malignancy. However, the recent advancement about using immunotherapy to treat NSCLC has been driven by remarkable results from clinical studies evaluating antibodies to programmed death receptor 1 (PD-1) and programmed death ligand 1 (PD-L1). Immunotherapy is generally well tolerated and has demonstrated a survival benefit in patients with advanced NSCLC. On the one hand, many immunotherapeutic agents, including monoclonal antibodies and small molecules, have shown significant antitumor activity in NSCLC humanized mouse models in vivo. Therefore, without doubt, humanized mouse models could be an extremely important animal model for pursuing novel immunotherapy treatment strategies in advanced NSCLC. The evidence is that many of TCM products including Polyphenols (e.g., Curcumin, Resveratrol), Cardiotonic Steroids (e.g., Digoxin and Bufalin), Terpenoids (e.g., Paclitaxel, Artemisinin, and Triptolide), Polysaccharides (e.g., Lentinan), Saponins, and Capsaicin have potential immunomodulatory effects [113], so increasing the focus has been driven toward finding novel potential modulators of tumor immunotherapy from TCM products. Maybe these TCM products combined with immune checkpoint inhibitor can be further studied in humanized mouse models and exert their advantages in immunotherapy for NSCLC, with synergistic effects and reduced toxicity [114]. On the other hand, syngeneic mouse models used for preclinical studies including intact immune responses also can provide a closer correlation with human cancer, especially for TCM products, like herbal medicines and monomeric compound, having great potential for targeting immune modulators [115–118]. In addition, TCM formulas have made great breakthroughs in the treatment of NSCLC by regulating the body to suppress tumors on the whole [8]. Meanwhile, a number of studies have shown that TCM formulas suppress the proliferation of cancer cells through various mechanisms especially by regulating immune function. The efficacy and mechanism of TCM formulas in the therapy of NSCLS in syngeneic mouse models are summarized as follows (see Table 3).
Table 3
List of TCM formulas as targeted therapy agents reported anticancer effects in NSCLC models.
Formulas | Origin | NSCLC models | Underlying mechanisms |
Bu-Fei decoction | Consisting of six herbal Chinese medicines including Codonopsis pilosula, Schisandra chinensis, Rehmannia glutinosa, Astragalus, Aster and Cortex Mori. | H1975, A549 | Partially via IL-10 and PD-L1 regulation [119]. |
Yangyinwenyang (YYWY) | Consisting of Paris polyphylla var, yunnanensis, Gynostemma pentaphyllum, Ophiopogon intermedius D. Don, and Trigonella foenum-graecum L | The Lewis lung cancer cells in C57BL/6 female mice | Through facilitating the mature DCs to activate the proliferation and differentiation of T cells [120]. |
Hedyotis diffusa willd | Hedyotis diffusa Willd extract powder | The Lewis lung cancer cells in C57BL/6 female mice | May activate immunity, achieve anti-inflammatory, antiproliferative, and antimigration therapeutic effects by regulating multiple pathways. |
Yu-ping-Feng (YPF) | Composed of Astragali Radix (huangqi), Atractylodis Macrocephalae Rhizoma (Baizhu), and Saposhnikoviae Radix (Fangfeng) | The Lewis lung cancer cells in C57BL/6 female mice | Downregulated the expression of TGF-β, indoleamine 2, 3-dioxygenase, and IL-10 in tumor microenvironment and had a NK cell-dependent inhibitory effect on Lewis lung cancer [121]. |
6. Outlook and Challenges
Humans have been using TCM products to treat diseases for thousands of years [122]. TCM can regulate the proliferation, adhesion, apoptosis, and tumor migration and inhibit tumor angiogenesis and change immune system. Actually, the mechanism of action of many TCM products is still unclear, but the therapeutic effects of TCM products on various diseases and immune regulation have been proved [123]. Although many TCM products have been reported on antitumor and immune modulatory effects, the research and application of TCM are still limited. The current researches are mainly focused on the TCM isolated single molecules, following the evaluation system of chemical drugs. However, TCM is usually used in the form of formulas rather than individual purified molecules. The composition of many TCM formulas, usually administered orally, is extraordinarily complex and metabolized to more complex metabolites in the body. Therefore, it is challenging to evaluate the efficacy of TCM by in vitro experiments, including cell based assay.
The in vivo models can mimic the metabolism and distribution of drugs in the body; therefore, they are more suitable for evaluating the antitumor effect of TCM. However, there are limited reports about the application of TCM formulas, compared to the chemical drug or antibodies, in animal models, and most of the reported models are CDXs and syngeneic mouse models. As an important tool in antitumor research, mouse models have always been the gold standard to evaluate the antitumor activity of drugs [124]. In view of the fact that TCM formulas usually act on multiple targets and signal pathways and may have the function of regulating immunity, animal models with complete immune system function are more suitable for the efficacy study of TCM formulations. Both syngeneic mouse models and humanized models have fully functional immune system and are powerful tools to evaluate the anticancer efficacy of TCM objectively. Considering the cost and difficulty of modeling, it is suggested that syngeneic mouse models could be utilized for testing the antitumor activities of TCM formulas in the early research, and humanized models could be used in further research.
The high incidence of NSCLC and the high rate of recurrence and metastasis in postoperative patients result in the urgent demand for more kinds of effective drugs. At present, targeted drugs and immune drugs have provided more treatment options and helped prolong the lives for cancer patients. But there are still unsolved problems in toxicity, effectiveness, and drug resistance of antitumor drugs. As a massive resource pool, TCM products provide powerful resources for finding new drugs for NSCLC patients. However, due to the lack of the support of large-scale preclinical and clinical research based on the concept of evidence-based medicine, TCM can only be regarded as an adjuvant therapy in the standard treatment of NSCLC. Until now, there is no animal model developed specifically for the TCM research, and none of the available models are perfectly matching the mechanism or characters of TCM, especially TCM formulas. Although syngeneic mouse and humanized models can be used in TCM studies, the establishment of more suitable models would be a valuable and promising field of research. It is believed that, with the development of research tool and deeper exploration of TCM products, more efficacy studies on mouse tumor models will be carried out, facilitating the development of antitumor drugs.
Authors’ Contributions
Hongkui Chen and Min Zheng contributed equally to this work.
Acknowledgments
This work was supported by grants from the Program of Study on the Anti-Gout Mechanism of Fu Tea, A Traditional Chinese Fermented Tea (2021LK109).
[1] V. M. L. de Sousa, L. Carvalho, "Heterogeneity in lung cancer," Pathobiology, vol. 85 no. 1-2, pp. 96-107, DOI: 10.1159/000487440, 2018.
[2] A. G. Nicholson, M. S. Tsao, M. B. Beasley, A. C. Borczuk, E. Brambilla, W. A. Cooper, S. Dacic, D. Jain, K. M. Kerr, S. Lantuejoul, M. Noguchi, M. Papotti, N. Rekhtman, G. Scagliotti, P. van Schil, L. Sholl, Y. Yatabe, A. Yoshida, W. D. Travis, "The 2021 WHO classification of lung tumors: impact of advances since 2015," Journal of Thoracic Oncology, vol. 17 no. 3, pp. 362-387, DOI: 10.1016/j.jtho.2021.11.003, 2022.
[3] L. Osmani, F. Askin, E. Gabrielson, Q. K. Li, "Current WHO guidelines and the critical role of immunohistochemical markers in the subclassification of non-small cell lung carcinoma (NSCLC): moving from targeted therapy to immunotherapy," Seminars in Cancer Biology, vol. 52 no. Pt 1, pp. 103-109, DOI: 10.1016/j.semcancer.2017.11.019, 2018.
[4] N. Duma, R. Santana-Davila, J. R. Molina, "Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment," Mayo Clinic Proceedings, vol. 94 no. 8, pp. 1623-1640, DOI: 10.1016/j.mayocp.2019.01.013, 2019.
[5] Z. Li, Z. Feiyue, L. Gaofeng, "Traditional Chinese medicine and lung cancer-from theory to practice," Biomedicine & Pharmacotherapy, vol. 137,DOI: 10.1016/j.biopha.2021.111381, 2021.
[6] M. Alexander, S. Y. Kim, H. Cheng, "Update 2020: management of non-small cell lung cancer," Lung, vol. 198 no. 6, pp. 897-907, DOI: 10.1007/s00408-020-00407-5, 2020.
[7] B. Qiu, W. Guo, F. Zhang, F. Lv, Y. Ji, Y. Peng, X. Chen, H. Bao, Y. Xu, Y. Shao, F. Tan, Q. Xue, S. Gao, J. He, "Dynamic recurrence risk and adjuvant chemotherapy benefit prediction by ctDNA in resected NSCLC," Nature Communications, vol. 12 no. 1,DOI: 10.1038/s41467-021-27022-z, 2021.
[8] X. L. Su, J. W. Wang, H. Che, C. F. Wang, H. Jiang, X. Lei, W. Zhao, H. X. Kuang, Q. H. Wang, "Clinical application and mechanism of traditional Chinese medicine in treatment of lung cancer," Chinese Medical Journal, vol. 133 no. 24, pp. 2987-2997, DOI: 10.1097/cm9.0000000000001141, 2020.
[9] D. Westover, J. Zugazagoitia, B. Cho, C. Lovly, L. Paz-Ares, "Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors," Annals of Oncology, vol. 29 no. suppl_1, pp. i10-i19, DOI: 10.1093/annonc/mdx703, 2018.
[10] A. Roys, X. Chang, Y. Liu, X. Xu, Y. Wu, D. Zuo, "Resistance mechanisms and potent-targeted therapies of ROS1-positive lung cancer," Cancer Chemotherapy and Pharmacology, vol. 84 no. 4, pp. 679-688, DOI: 10.1007/s00280-019-03902-6, 2019.
[11] M. Ramos-Casals, J. R. Brahmer, M. K. Callahan, A. Flores-Chavez, N. Keegan, M. A. Khamashta, O. Lambotte, X. Mariette, A. Prat, M. E. Suarez-Almazor, "Immune-related adverse events of checkpoint inhibitors," Nature Reviews Disease Primers, vol. 6 no. 1,DOI: 10.1038/s41572-020-0160-6, 2020.
[12] T. C. Lam, K. Tsang, H. Choi, V. Lee, K. Lam, C. Chiang, T. So, W. Chan, S. Nyaw, F. Lim, J. Lau, J. Chik, F. Kong, A. Lee, "Combination atezolizumab, bevacizumab, pemetrexed and carboplatin for metastatic EGFR mutated NSCLC after TKI failure," Lung Cancer, vol. 159, pp. 18-26, DOI: 10.1016/j.lungcan.2021.07.004, 2021.
[13] J. An, H. Kim, K. M. Yang, "An aqueous extract of a bifidobacterium species induces apoptosis and inhibits invasiveness of non-small cell lung cancer cells," Journal of Microbiology and Biotechnology, vol. 30 no. 6, pp. 885-893, DOI: 10.4014/jmb.1912.12054, 2020.
[14] X. W. Zhang, W. Liu, H. L. Jiang, B. Mao, "Chinese herbal medicine for advanced non-small-cell lung cancer: a systematic review and meta-analysis," The American Journal of Chinese Medicine, vol. 46 no. 5, pp. 923-952, DOI: 10.1142/s0192415x18500490, 2018.
[15] E. Catanzaro, G. Greco, L. Potenza, C. Calcabrini, C. Fimognari, "Natural products to fight cancer: a focus on juglans regia," Toxins, vol. 10 no. 11,DOI: 10.3390/toxins10110469, 2018.
[16] S. Wang, Z. Wang, Y. Wu, C. Hou, X. Dai, Q. Wang, Y. Wu, C. Qian, X. Zhang, "The TCM prescription Yi-Fei-Jie-Du-Tang inhibit invasive migration and EMT of lung cancer cells by activating autophagy," Evidence-based Complementary and Alternative Medicine, vol. 2022,DOI: 10.1155/2022/9160616, 2022.
[17] S. Dutta, S. Mahalanobish, S. Saha, S. Ghosh, P. C. Sil, "Natural products: an upcoming therapeutic approach to cancer," Food and Chemical Toxicology, vol. 128, pp. 240-255, DOI: 10.1016/j.fct.2019.04.012, 2019.
[18] R. Liu, X. Li, N. Huang, M. Fan, R. Sun, "Toxicity of traditional Chinese medicine herbal and mineral products," Advances in Pharmacology, vol. 87, pp. 301-346, DOI: 10.1016/bs.apha.2019.08.001, 2020.
[19] B. Chopra, A. K. Dhingra, "Natural products: a lead for drug discovery and development," Phytotherapy Research, vol. 35 no. 9, pp. 4660-4702, DOI: 10.1002/ptr.7099, 2021.
[20] M. Hemagirri, S. Sasidharan, "In vitro antiaging activity of polyphenol rich Polyalthia longifolia (Annonaceae) leaf extract in Saccharomyces cerevisiae BY611 yeast cells," Journal of Ethnopharmacology, vol. 290,DOI: 10.1016/j.jep.2022.115110, 2022.
[21] S. W. Shin, Y. S. Hwang, "Anti-periodontitis effect of ethanol extracts of Alpinia katsumadai seeds," Nutrients, vol. 14 no. 1,DOI: 10.3390/nu14010136, 2021.
[22] M. A. Shibu, Y. J. Lin, C. Y. Chiang, C. Y. Lu, D. Goswami, N. Sundhar, S. Agarwal, M. N. Islam, P. Y. Lin, S. Z. Lin, T. J. Ho, W. T. Tsai, W. W. Kuo, C. Y. Huang, "Novel anti-aging herbal formulation Jing Si displays pleiotropic effects against aging associated disorders," Biomedicine & Pharmacotherapy, vol. 146,DOI: 10.1016/j.biopha.2021.112427, 2022.
[23] C. C. M. d. Jesus, M. Hd Araujo, T. L. B. V. Simao, E. B. Lasunskaia, T. Barth, M. F. Muzitano, S. C. Pinto, "Natural products from Vitex polygama and their antimycobacterial and anti-inflammatory activity," Natural Product Research, vol. 36 no. 5, pp. 1337-1341, DOI: 10.1080/14786419.2020.1868461, 2022.
[24] Y. O. Ali Abdalla, B. Subramaniam, S. Nyamathulla, N. Shamsuddin, N. M. Arshad, K. S. Mun, K. Awang, N. H. Nagoor, "Natural products for cancer therapy: a review of their mechanism of actions and toxicity in the past decade," The Journal of Tropical Medicine, vol. 2022,DOI: 10.1155/2022/5794350, 2022.
[25] M. Yuan, G. Zhang, W. Bai, X. Han, C. Li, S. Bian, "The role of bioactive compounds in natural products extracted from plants in cancer treatment and their mechanisms related to anticancer effects," Oxidative Medicine and Cellular Longevity, vol. 2022,DOI: 10.1155/2022/1429869, 2022.
[26] H. M. Ekiert, A. Szopa, "Biological activities of natural products," Molecules, vol. 25 no. 23,DOI: 10.3390/molecules25235769, 2020.
[27] Q. Cui, D. H. Yang, Z. S. Chen, "Special issue: natural products: anticancer and beyond," Molecules, vol. 23 no. 6,DOI: 10.3390/molecules23061246, 2018.
[28] A. Kumar, V. Jaitak, "Natural products as multidrug resistance modulators in cancer," European Journal of Medicinal Chemistry, vol. 176, pp. 268-291, DOI: 10.1016/j.ejmech.2019.05.027, 2019.
[29] Y. Guo, S. Liu, F. Luo, D. Tang, T. Yang, X. Yang, Y. Xie, "A nanosized codelivery system based on intracellular stimuli-triggereddual-drug release for multilevel chemotherapy amplification in drug-resistant breast cancer," Pharmaceutics, vol. 14 no. 2,DOI: 10.3390/pharmaceutics14020422, 2022.
[30] L. Song, X. Hu, X. Ren, J. Liu, X. Liu, "Antibacterial modes of herbal flavonoids combat resistant bacteria," Frontiers in Pharmacology, vol. 13,DOI: 10.3389/fphar.2022.873374, 2022.
[31] L. L. Huang, M. Tang, Q. Q. Du, C. X. Liu, C. Yan, J. L. Yang, Y. Li, "The effects and mechanisms of a biosynthetic ginsenoside 3 β , 12 β -Di-O-Glc-PPD on non-small cell lung cancer," OncoTargets and Therapy, vol. 12, pp. 7375-7385, DOI: 10.2147/ott.s217039, 2019.
[32] X. Zhou, B. Liu, Q. Ning, Z. Xia, R. Zhong, L. Zhang, L. Wu, "Combination of Huanglian Jiedu Decoction and erlotinib delays growth and improves sensitivity of EGFRmutated NSCLC cells in vitro and in vivo via STAT3/Bcl2 signaling," Oncology Reports, vol. 45 no. 1, pp. 217-229, DOI: 10.3892/or.2020.7848, 2020.
[33] M. S. Jeong, K. W. Lee, Y. J. Choi, Y. G. Kim, H. H. Hwang, S. Y. Lee, S. E. Jung, S. A. Park, J. H. Lee, Y. J. Joo, S. G. Cho, S. G. Ko, "Synergistic antitumor activity of SH003 and docetaxel via EGFR signaling inhibition in non-small cell lung cancer," International Journal of Molecular Sciences, vol. 22 no. 16,DOI: 10.3390/ijms22168405, 2021.
[34] L. Kui, M. Li, X. Yang, L. Yang, Q. Kong, Y. Pan, Z. Xu, S. Wang, D. Mo, Y. Dong, Y. Liu, J. Miao, "High-throughput screen of natural compounds and biomarkers for NSCLC treatment by differential expression and weighted gene coexpression network analysis (WGCNA)," BioMed Research International, vol. 2021,DOI: 10.1155/2021/5955343, 2021.
[35] L. Zhang, B. Liang, H. Xu, Y. Gong, W. Hu, Z. Jin, X. Wu, X. Chen, M. Li, L. Shi, Y. Shi, Y. Wang, L. Yang, "Cinobufagin induces FOXO1-regulated apoptosis, proliferation, migration, and invasion by inhibiting G9a in non-small-cell lung cancer A549 cells," Journal of Ethnopharmacology, vol. 291,DOI: 10.1016/j.jep.2022.115095, 2022.
[36] R. Shi, N. Radulovich, C. Ng, N. Liu, H. Notsuda, M. Cabanero, S. N. Martins-Filho, V. Raghavan, Q. Li, A. S. Mer, J. C. Rosen, M. Li, Y. H. Wang, L. Tamblyn, N. A. Pham, B. Haibe-Kains, G. Liu, N. Moghal, M. S. Tsao, "Organoid cultures as preclinical models of non-small cell lung cancer," Clinical Cancer Research, vol. 26 no. 5, pp. 1162-1174, DOI: 10.1158/1078-0432.ccr-19-1376, 2020.
[37] K. N. Suvilesh, Y. I. Nussbaum, V. Radhakrishnan, Y. Manjunath, D. M. Avella, K. F. Staveley-O’Carroll, E. T. Kimchi, A. A. Chaudhuri, C. R. Shyu, G. Li, K. Pantel, W. C. Warren, J. B. Mitchem, J. T. Kaifi, "Tumorigenic circulating tumor cells from xenograft mouse models of non-metastatic NSCLC patients reveal distinct single cell heterogeneity and drug responses," Molecular Cancer, vol. 21 no. 1,DOI: 10.1186/s12943-022-01553-5, 2022.
[38] A. Cornett, H. K. Athwal, E. Hill, G. Murphy, K. Yeoh, C. A. Moskaluk, R. L. Witt, N. J. D’Silva, S. Agarwal, I. M. Lombaert, "Serial patient-derived orthotopic xenografting of adenoid cystic carcinomas recapitulates stable expression of phenotypic alterations and innervation," EBioMedicine, vol. 41, pp. 175-184, DOI: 10.1016/j.ebiom.2019.02.011, 2019.
[39] Q. E. Yang, "Human cancer xenografts in immunocompromised mice provide an advanced genuine tumor model for research and drug development-a revisit of murine models for human cancers," Biochimica et Biophysica Acta (BBA)—General Subjects, vol. 1865 no. 8,DOI: 10.1016/j.bbagen.2021.129929, 2021.
[40] S. Guo, X. Jiang, B. Mao, Q. X. Li, "The design, analysis and application of mouse clinical trials in oncology drug development," BMC Cancer, vol. 19 no. 1,DOI: 10.1186/s12885-019-5907-7, 2019.
[41] Y. Xie, W. Shangguan, Z. Chen, Z. Zheng, Y. Chen, Q. Zhong, Y. Zhang, J. Yang, D. Zhu, W. Xie, "Establishment of sunitinib-resistant xenograft model of renal cell carcinoma and the identification of drug-resistant hub genes and pathways," Drug Design, Development and Therapy, vol. 15, pp. 5061-5074, DOI: 10.2147/dddt.s343718, 2021.
[42] R. Sharma, N. Martins, "Telomeres, DNA damage and ageing: potential leads from ayurvedic rasayana (anti-ageing) drugs," Journal of Clinical Medicine, vol. 9 no. 8,DOI: 10.3390/jcm9082544, 2020.
[43] A. P. Singh, D. Adrianzen Herrera, Y. Zhang, R. Perez-Soler, H. Cheng, "Mouse models in squamous cell lung cancer: impact for drug discovery," Expert Opinion on Drug Discovery, vol. 13 no. 4, pp. 347-358, DOI: 10.1080/17460441.2018.1437137, 2018.
[44] J. Chen, Y. Jin, S. Li, C. Qiao, X. Peng, Y. Li, Y. Gu, W. Wang, Y. You, J. Yin, Y. Shan, Y. X. Wang, M. Qin, H. Li, Y. Cai, Y. Dong, S. Peng, L. Pan, "Patient-derived xenografts are a reliable preclinical model for the personalized treatment of epithelial ovarian cancer," Frontiers in Oncology, vol. 11,DOI: 10.3389/fonc.2021.744256, 2021.
[45] S. Bisht, G. Feldmann, "Animal models for modeling pancreatic cancer and novel drug discovery," Expert Opinion on Drug Discovery, vol. 14 no. 2, pp. 127-142, DOI: 10.1080/17460441.2019.1566319, 2019.
[46] T. Tanaka, R. Nishie, S. Ueda, S. Miyamoto, S. Hashida, H. Konishi, S. Terada, Y. Kogata, H. Sasaki, S. Tsunetoh, K. Taniguchi, K. Komura, M. Ohmichi, "Patient-derived xenograft models in cervical cancer: a systematic review," International Journal of Molecular Sciences, vol. 22 no. 17,DOI: 10.3390/ijms22179369, 2021.
[47] B. Pan, X. Wei, X. Xu, "Patient-derived xenograft models in hepatopancreatobiliary cancer," Cancer Cell International, vol. 22 no. 1,DOI: 10.1186/s12935-022-02454-9, 2022.
[48] H. Y. Shin, E. Lee, W. Yang, H. S. Kim, D. Chung, H. Cho, J. H. Kim, "Identification of prognostic markers of gynecologic cancers utilizing patient-derived xenograft mouse models," Cancers, vol. 14 no. 3,DOI: 10.3390/cancers14030829, 2022.
[49] S. Abdolahi, Z. Ghazvinian, S. Muhammadnejad, M. Saleh, H. Asadzadeh Aghdaei, K. Baghaei, "Patient-derived xenograft (PDX) models, applications and challenges in cancer research," Journal of Translational Medicine, vol. 20 no. 1,DOI: 10.1186/s12967-022-03405-8, 2022.
[50] J. Qu, F. S. Kalyani, L. Liu, T. Cheng, L. Chen, "Tumor organoids: synergistic applications, current challenges, and future prospects in cancer therapy," Cancer Communications, vol. 41 no. 12, pp. 1331-1353, DOI: 10.1002/cac2.12224, 2021.
[51] Z. Zeng, C. J. Wong, L. Yang, N. Ouardaoui, D. Li, W. Zhang, S. Gu, Y. Zhang, Y. Liu, X. Wang, J. Fu, L. Zhou, B. Zhang, S. Kim, K. Yates, M. Brown, G. Freeman, R. Uppaluri, R. Manguso, X. Liu, "TISMO: syngeneic mouse tumor database to model tumor immunity and immunotherapy response," Nucleic Acids Research, vol. 50 no. D1, pp. D1391-D1397, DOI: 10.1093/nar/gkab804, 2022.
[52] T. Constantin, M. Pavalean, Ş. Bucur, M. Constantin, A. Nicolescu, I. Pacu, V. Madan, "Animal models in bladder cancer," Biomedicines, vol. 9 no. 12,DOI: 10.3390/biomedicines9121762, 2021.
[53] J. T. Zoine, S. E. Moore, M. P. Velasquez, "Leukemia’s next top model? syngeneic models to advance adoptive cellular therapy," Frontiers in Immunology, vol. 13,DOI: 10.3389/fimmu.2022.867103, 2022.
[54] V. Letchuman, L. Ampie, A. H. Shah, D. A. Brown, J. D. Heiss, P. Chittiboina, "Syngeneic murine glioblastoma models: reactionary immune changes and immunotherapy intervention outcomes," Neurosurgical Focus, vol. 52 no. 2,DOI: 10.3171/2021.11.focus21556, 2022.
[55] H. Li, Z. Liu, L. Liu, H. Zhang, C. Han, L. Girard, H. Park, A. Zhang, C. Dong, J. Ye, A. Rayford, M. Peyton, X. Li, K. Avila, X. Cao, S. Hu, M. M. Alam, E. A. Akbay, L. M. Solis, C. Behrens, S. Hernandez-Ruiz, W. Lu, I. Wistuba, J. V. Heymach, M. Chisamore, D. Micklem, H. Gabra, G. Gausdal, J. B. Lorens, B. Li, Y. X. Fu, J. D. Minna, R. A. Brekken, "AXL targeting restores PD-1 blockade sensitivity of STK11/LKB1 mutant NSCLC through expansion of TCF1 + CD8 T cells," Cell Reports Medicine, vol. 3 no. 3,DOI: 10.1016/j.xcrm.2022.100554, 2022.
[56] T. Higuchi, K. Igarashi, N. Yamamoto, K. Hayashi, H. Kimura, S. Miwa, M. Bouvet, H. Tsuchiya, R. M. Hoffman, "Osteosarcoma patient-derived orthotopic xenograft (PDOX) models used to identify novel and effective therapeutics: a review," Anticancer Research, vol. 41 no. 12, pp. 5865-5871, DOI: 10.21873/anticanres.15406, 2021.
[57] E. Magnotti, W. A. Marasco, "The latest animal models of ovarian cancer for novel drug discovery," Expert Opinion on Drug Discovery, vol. 13 no. 3, pp. 249-257, DOI: 10.1080/17460441.2018.1426567, 2018.
[58] R. C. Oliveira, A. M. Abrantes, J. G. Tralhao, M. F. Botelho, "The role of mouse models in colorectal cancer research-The need and the importance of the orthotopic models," Animal Models and Experimental Medicine, vol. 3 no. 1,DOI: 10.1002/ame2.12102, 2020.
[59] F. Burtin, C. S. Mullins, M. Linnebacher, "Mouse models of colorectal cancer: past, present and future perspectives," World Journal of Gastroenterology, vol. 26 no. 13, pp. 1394-1426, DOI: 10.3748/wjg.v26.i13.1394, 2020.
[60] U. H. Weidle, F. Birzele, A. Nopora, "MicroRNAs as potential targets for therapeutic intervention with metastasis of non-small cell lung cancer," Cancer Genomics & Proteomics, vol. 16 no. 2, pp. 99-119, DOI: 10.21873/cgp.20116, 2019.
[61] C. Li, J. Zhang, X. Yang, C. Hu, T. Chu, R. Zhong, Y. Shen, F. Hu, F. Pan, J. Xu, J. Lu, X. Zheng, H. Zhang, W. Nie, B. Han, X. Zhang, "hsa_circ_0003222 accelerates stemness and progression of non-small cell lung cancer by sponging miR-527," Cell Death & Disease, vol. 12 no. 9,DOI: 10.1038/s41419-021-04095-8, 2021.
[62] D. H. He, Y. Chen, Y. Zhou, S. Zhang, M. Hong, X. Yu, S. Wei, X. Fan, S. Li, Q. Wang, Y. Lu, Y. Liu, "Phytochemical library screening reveals betulinic acid as a novel Skp2-SCF E3 ligase inhibitor in non-small cell lung cancer," Cancer Science, vol. 112 no. 8, pp. 3218-3232, DOI: 10.1111/cas.15005, 2021.
[63] M. Friese-Hamim, A. Clark, D. Perrin, L. Crowley, C. Reusch, O. Bogatyrova, H. Zhang, T. Crandall, J. Lin, J. Ma, D. Bachner, J. Schmidt, M. Schaefer, C. Stroh, "Brain penetration and efficacy of tepotinib in orthotopic patient-derived xenograft models of MET-drivennon-small cell lung cancer brain metastases," Lung Cancer, vol. 163, pp. 77-86, DOI: 10.1016/j.lungcan.2021.11.020, 2022.
[64] U. Jarry, M. Bostoen, R. Pineau, L. Chaillot, V. Mennessier, P. Montagne, E. Motte, M. Gournay, A. Le Goff, T. Guillaudeux, R. Pedeux, "Orthotopic model of lung cancer: isolation of bone micro-metastases after tumor escape from Osimertinib treatment," BMC Cancer, vol. 21 no. 1,DOI: 10.1186/s12885-021-08205-9, 2021.
[65] D. H. Shin, J. Y. Jo, S. H. Kim, M. Choi, C. Han, B. K. Choi, S. S. Kim, "Midkine is a potential therapeutic target of tumorigenesis, angiogenesis, and metastasis in non-small cell lung cancer," Cancers, vol. 12 no. 9,DOI: 10.3390/cancers12092402, 2020.
[66] T. Tanaka, R. Nishie, S. Ueda, S. Miyamoto, S. Hashida, H. Konishi, S. Terada, Y. Kogata, H. Sasaki, S. Tsunetoh, K. Taniguchi, K. Komura, M. Ohmichi, "Endometrial cancer patient-derived xenograft models: a systematic review," Journal of Clinical Medicine, vol. 11 no. 9,DOI: 10.3390/jcm11092606, 2022.
[67] C. Yuan, X. Zhao, D. Wangmo, D. Alshareef, T. J. Gates, S. Subramanian, "Tumor models to assess immune response and tumor-microbiome interactions in colorectal cancer," Pharmacology & Therapeutics, vol. 231,DOI: 10.1016/j.pharmthera.2021.107981, 2022.
[68] Z. Chen, C. M. Fillmore, P. S. Hammerman, C. F. Kim, K. K. Wong, "Non-small-cell lung cancers: a heterogeneous set of diseases," Nature Reviews Cancer, vol. 14 no. 8, pp. 535-546, DOI: 10.1038/nrc3775, 2014.
[69] S. A. Hayes, A. L. Hudson, S. J. Clarke, M. P. Molloy, V. M. Howell, "From mice to men: GEMMs as trial patients for new NSCLC therapies," Seminars in Cell & Developmental Biology, vol. 27, pp. 118-127, DOI: 10.1016/j.semcdb.2014.04.002, 2014.
[70] H. Deng, Y. Gao, V. Trappetti, D. Hertig, D. Karatkevich, T. Losmanova, C. Urzi, H. Ge, G. A. Geest, R. Bruggmann, V. Djonov, J. M. Nuoffer, P. Vermathen, N. Zamboni, C. Riether, A. Ochsenbein, R. W. Peng, G. J. Kocher, R. A. Schmid, P. Dorn, T. M. Marti, "Targeting lactate dehydrogenase B-dependent mitochondrial metabolism affects tumor initiating cells and inhibits tumorigenesis of non-small cell lung cancer by inducing mtDNA damage," Cellular and Molecular Life Sciences, vol. 79 no. 8,DOI: 10.1007/s00018-022-04453-5, 2022.
[71] M. DuPage, T. Jacks, "Genetically engineered mouse models of cancer reveal new insights about the antitumor immune response," Current Opinion in Immunology, vol. 25 no. 2, pp. 192-199, DOI: 10.1016/j.coi.2013.02.005, 2013.
[72] R. Kandimalla, F. Aqil, S. S. Alhakeem, J. Jeyabalan, N. Tyagi, A. Agrawal, J. Yan, W. Spencer, S. Bondada, R. C. Gupta, "Targeted oral delivery of paclitaxel using colostrum-derived exosomes," Cancers, vol. 13 no. 15,DOI: 10.3390/cancers13153700, 2021.
[73] X. Q. Li, J. Ren, Y. Wang, J. Y. Su, Y. M. Zhu, C. G. Chen, W. G. Long, Q. Jiang, J. Li, "Synergistic killing effect of paclitaxel and honokiol in non-small cell lung cancer cells through paraptosis induction," Cellular Oncology, vol. 44 no. 1, pp. 135-150, DOI: 10.1007/s13402-020-00557-x, 2021.
[74] M. G. Dal Bello, A. Alama, G. Barletta, S. Coco, A. Truini, I. Vanni, S. Boccardo, C. Genova, E. Rijavec, F. Biello, G. Bottoni, G. Sambuceti, F. Grossi, "Sequential use of vinorelbine followed by gefitinib enhances the antitumor effect in NSCLC cell lines poorly responsive to reversible EGFR tyrosine kinase inhibitors," International Journal of Cancer, vol. 137 no. 12, pp. 2947-2958, DOI: 10.1002/ijc.29647, 2015.
[75] Z. B. Jiang, J. M. Huang, Y. J. Xie, Y. Z. Zhang, C. Chang, H. L. Lai, W. Wang, X. J. Yao, X. X. Fan, Q. B. Wu, C. Xie, M. F. Wang, E. L. H. Leung, "Evodiamine suppresses non-small cell lung cancer by elevating CD8 + T cells and downregulating the MUC1-C/PD-L1 axis," Journal of Experimental & Clinical Cancer Research, vol. 39 no. 1,DOI: 10.1186/s13046-020-01741-5, 2020.
[76] T. Su, X. Yang, J. H. Deng, Q. J. Huang, S. C. Huang, Y. M. Zhang, H. M. Zheng, Y. Wang, L. L. Lu, Z. Q. Liu, "Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo," Frontiers in Pharmacology, vol. 9,DOI: 10.3389/fphar.2018.00434, 2018.
[77] X. Yang, Y. Zhang, Y. Huang, Y. Wang, X. Qi, T. Su, L. Lu, "Evodiamine suppresses Notch3 signaling in lung tumorigenesis via direct binding to gamma-secretases," Phytomedicine, vol. 68,DOI: 10.1016/j.phymed.2020.153176, 2020.
[78] J. Zhang, Y. Zhang, W. Shen, R. Fu, Z. Ding, Y. Zhen, Y. Wan, "Cytological effects of honokiol treatment and its potential mechanism of action in non-small cell lung cancer," Biomedicine & Pharmacotherapy, vol. 117,DOI: 10.1016/j.biopha.2019.109058, 2019.
[79] H. E. Pearson, M. Iida, R. A. Orbuch, N. K. McDaniel, K. P. Nickel, R. J. Kimple, J. L. Arbiser, D. L. Wheeler, "Overcoming resistance to cetuximab with honokiol, a small-molecule polyphenol," Molecular Cancer Therapeutics, vol. 17 no. 1, pp. 204-214, DOI: 10.1158/1535-7163.mct-17-0384, 2018.
[80] T. Singh, R. Prasad, S. K. Katiyar, "Inhibition of class I histone deacetylases in non-small cell lung cancer by honokiol leads to suppression of cancer cell growth and induction of cell death in vitro and in vivo," Epigenetics, vol. 8 no. 1, pp. 54-65, DOI: 10.4161/epi.23078, 2013.
[81] W. Liu, X. Yu, L. Zhou, J. Li, M. Li, W. Li, F. Gao, "Sinomenine inhibits non-small cell lung cancer via downregulation of hexokinases II-mediated aerobic glycolysis," OncoTargets and Therapy, vol. 13, pp. 3209-3221, DOI: 10.2147/ott.s243212, 2020.
[82] X. Yu, F. Gao, W. Li, L. Zhou, W. Liu, M. Li, "Formononetin inhibits tumor growth by suppression of EGFR-Akt-Mcl-1 axis in non-small cell lung cancer," Journal of Experimental & Clinical Cancer Research, vol. 39 no. 1,DOI: 10.1186/s13046-020-01566-2, 2020.
[83] K. C. Tay, L. T. H. Tan, C. K. Chan, S. L. Hong, K. G. Chan, W. H. Yap, P. Pusparajah, L. H. Lee, B. H. Goh, "Formononetin: a review of its anticancer potentials and mechanisms," Frontiers in Pharmacology, vol. 10,DOI: 10.3389/fphar.2019.00820, 2019.
[84] H. Li, L. Tan, J. W. Zhang, H. Chen, B. Liang, T. Qiu, Q. S. Li, M. Cai, Q. H. Zhang, "Quercetin is the active component of Yang-Yin-Qing-Fei-Tang to induce apoptosis in non-small cell lung cancer," The American Journal of Chinese Medicine, vol. 47 no. 4, pp. 879-893, DOI: 10.1142/s0192415x19500460, 2019.
[85] J. H. Chang, S. L. Lai, W. S. Chen, W. Y. Hung, J. M. Chow, M. Hsiao, W. J. Lee, M. H. Chien, "Quercetin suppresses the metastatic ability of lung cancer through inhibiting snail-dependent Akt activation and snail-independent ADAM9 expression pathways," Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, vol. 1864 no. 10, pp. 1746-1758, DOI: 10.1016/j.bbamcr.2017.06.017, 2017.
[86] J. H. Lee, S. Y. Park, W. Hwang, J. Y. Sung, M. L. Cho, J. Shim, Y. N. Kim, K. Yoon, "Isoharringtonine induces apoptosis of non-small cell lung cancer cells in tumorspheroids via the intrinsic pathway," Biomolecules, vol. 10 no. 11,DOI: 10.3390/biom10111521, 2020.
[87] Z. Li, H. Hao, W. Tian, Y. Jiao, X. Deng, S. Han, J. Han, "Nitric oxide, a communicator between tumor cells and endothelial cells, mediates the anti-tumor effects of Marsdenia Tenacissima extract (MTE)," Journal of Ethnopharmacology, vol. 250,DOI: 10.1016/j.jep.2019.112524, 2020.
[88] C. Zhao, H. Hao, H. Zhao, W. Ren, Y. Jiao, G. An, H. Sun, S. Han, P. Li, "Marsdenia tenacissima extract promotes gefitinib accumulation in tumor tissues of lung cancer xenograft mice via inhibiting ABCG2 activity," Journal of Ethnopharmacology, vol. 255,DOI: 10.1016/j.jep.2020.112770, 2020.
[89] Z. Zhao, B. Liu, J. Sun, L. Lu, L. Liu, J. Qiu, Q. Li, C. Yan, S. Jiang, N. Mohammadtursun, W. Ma, M. Li, J. Dong, W. Gong, "Baicalein inhibits orthotopic human non-small cell lung cancer xenografts via src/id1 pathway," Evidence-based Complementary and Alternative Medicine, vol. 2019,DOI: 10.1155/2019/9806062, 2019.
[90] I. Jeong, J. Song, S. Y. Bae, S. K. Lee, "Overcoming the intrinsic gefitinib-resistance via downregulation of AXL in non-small cell lung cancer," Journal of Cancer Prevention, vol. 24 no. 4, pp. 217-223, DOI: 10.15430/jcp.2019.24.4.217, 2019.
[91] B. Luo, Z. Que, Z. Zhou, Q. Wang, C. Dong, Y. Jiang, B. Hu, H. Shi, Y. Jin, J. Liu, H. Li, L. Wang, J. Tian, "Feiji recipe inhibits the growth of lung cancer by modulating T-cell immunity through indoleamine-2, 3-dioxygenase pathway in an orthotopic implantation model," Journal of Integrative Medicine, vol. 16 no. 4, pp. 283-289, DOI: 10.1016/j.joim.2018.04.008, 2018.
[92] S. P. Kalainayakan, P. Ghosh, S. Dey, K. E. Fitzgerald, S. Sohoni, P. C. Konduri, M. Garrossian, L. Liu, L. Zhang, "Cyclopamine tartrate, a modulator of hedgehog signaling and mitochondrial respiration, effectively arrests lung tumor growth and progression," Scientific Reports, vol. 9 no. 1,DOI: 10.1038/s41598-018-38345-1, 2019.
[93] M. W. Turner, R. Cruz, J. Mattos, N. Baughman, J. Elwell, J. Fothergill, A. Nielsen, J. Brookhouse, A. Bartlett, P. Malek, X. Pu, M. D. King, O. M. McDougal, "Cyclopamine bioactivity by extraction method from Veratrum californicum," Bioorganic & Medicinal Chemistry, vol. 24 no. 16, pp. 3752-3757, DOI: 10.1016/j.bmc.2016.06.017, 2016.
[94] M. W. Turner, M. Rossi, V. Campfield, J. French, E. Hunt, E. Wade, O. M. McDougal, "Steroidal alkaloid variation in Veratrum californicum as determined by modern methods of analytical analysis," Fitoterapia, vol. 137,DOI: 10.1016/j.fitote.2019.104281, 2019.
[95] X. Xiao, L. Guo, W. Dai, B. Yan, J. Zhang, Q. Yuan, L. Zhou, L. Shan, T. Efferth, "Green tea-derived theabrownin suppresses human non-small cell lung carcinoma in xenograft model through activation of not only p53 signaling but also MAPK/JNK signaling pathway," Journal of Ethnopharmacology, vol. 291,DOI: 10.1016/j.jep.2022.115167, 2022.
[96] Z. H. Xu, Yz Zhu, L. Su, X. Tang, C. Yao, X. Jiao, Y. Hou, X. Chen, L. Wei, W. Wang, J. Wang, C. Gong, X. Zhu, F. Zhang, S. Zhu, C. Zou, "Ze-Qi-Tang formula induces granulocytic myeloid-derived suppressor cell apoptosis via STAT3/S100a9/bcl-2/caspase-3 signaling to prolong the survival of mice with orthotopic lung cancer," Mediators of Inflammation, vol. 2021,DOI: 10.1155/2021/8856326, 2021.
[97] H. Y. Tan, V. W. T. Ho, Y. T. Chan, C. Zhang, N. Wang, W. Xia, Y. Feng, "Combination of Gentiana rhodantha and Gerbera anandria in the BL02 formula as therapeutics to non-small cell lung carcinoma acting via Rap1/cdc42 signaling: a transcriptomics/bio-informatics biological validation approach," Pharmacological Research, vol. 155,DOI: 10.1016/j.phrs.2019.104415, 2020.
[98] D. R. Camidge, R. C. Doebele, K. M. Kerr, "Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC," Nature Reviews Clinical Oncology, vol. 16 no. 6, pp. 341-355, DOI: 10.1038/s41571-019-0173-9, 2019.
[99] M. S. Kumar, K. M. Adki, "Marine natural products for multi-targeted cancer treatment: a future insight," Biomedicine & Pharmacotherapy, vol. 105, pp. 233-245, DOI: 10.1016/j.biopha.2018.05.142, 2018.
[100] F. Liu, G. Yu, G. Wang, H. Liu, X. Wu, Q. Wang, M. Liu, K. Liao, M. Wu, X. Cheng, H. Hao, "An NQO1-initiated and p53-independent apoptotic pathway determines the anti-tumor effect of tanshinone IIA against non-small cell lung cancer," PLoS One, vol. 7 no. 7,DOI: 10.1371/journal.pone.0042138, 2012.
[101] R. Wang, Z. Luo, H. Zhang, T. Wang, "Tanshinone IIA reverses gefitinib-resistance in human non-small-cell lung cancer via regulation of VEGFR/akt pathway," OncoTargets and Therapy, vol. 12, pp. 9355-9365, DOI: 10.2147/ott.s221228, 2019.
[102] F. Gao, M. Li, W. Liu, W. Li, "Inhibition of EGFR signaling and activation of mitochondrial apoptosis contribute to tanshinone IIA-mediated tumor suppression in non-small cell lung cancer cells," OncoTargets and Therapy, vol. 13, pp. 2757-2769, DOI: 10.2147/ott.s246606, 2020.
[103] J. Xie, J. Liu, H. Liu, S. Liang, M. Lin, Y. Gu, T. Liu, D. Wang, H. Ge, Sl Mo, "The antitumor effect of tanshinone IIA on anti-proliferation and decreasing VEGF/VEGFR2 expression on the human non-small cell lung cancer A549 cell line," Acta Pharmaceutica Sinica B, vol. 5 no. 6, pp. 554-563, DOI: 10.1016/j.apsb.2015.07.008, 2015.
[104] X. Z. Liao, Y. Gao, S. Huang, Z. Chen, L. Sun, J. Liu, H. Chen, L. Yu, J. Zhang, L. Lin, "Tanshinone IIA combined with cisplatin synergistically inhibits non-small-cell lung cancer in vitro and in vivo via down-regulating the phosphatidylinositol 3-kinase/Akt signalling pathway," Phytotherapy Research, vol. 33 no. 9, pp. 2298-2309, DOI: 10.1002/ptr.6392, 2019.
[105] X. Li, R. Huang, M. Li, Z. Zhu, Z. Chen, L. Cui, H. Luo, L. Luo, "Parthenolide inhibits the growth of non-small cell lung cancer by targeting epidermal growth factor receptor," Cancer Cell International, vol. 20 no. 1,DOI: 10.1186/s12935-020-01658-1, 2020.
[106] Y. L. Leu, T. H. Wang, C. C. Wu, K. Y. Huang, Y. W. Jiang, Y. C. Hsu, C. Y. Chen, "Hydroxygenkwanin suppresses non-small cell lung cancer progression by enhancing EGFR degradation," Molecules, vol. 25 no. 4,DOI: 10.3390/molecules25040941, 2020.
[107] F. Gao, X. Yu, M. Li, L. Zhou, W. Liu, W. Li, H Liu, "Deguelin suppresses non-small cell lung cancer by inhibiting EGFR signaling and promoting GSK3 β /FBW7-mediated Mcl-1 destabilization," Cell Death & Disease, vol. 11 no. 2,DOI: 10.1038/s41419-020-2344-0, 2020.
[108] W. Kang, X. Zheng, P. Wang, S. Guo, "Deguelin exerts anticancer activity of human gastric cancer MGC-803 and MKN-45 cells in vitro," International Journal of Molecular Medicine, vol. 41 no. 6, pp. 3157-3166, DOI: 10.3892/ijmm.2018.3532, 2018.
[109] J. Li, L. Yan, J. Luo, L. Tong, Y. Gao, W. Feng, F. Wang, W. Cui, S. Li, Z. Sun, "Baicalein suppresses growth of non-small cell lung carcinoma by targeting MAP4K3," Biomedicine & Pharmacotherapy, vol. 133, 2021.
[110] Y. Song, H. Dou, P. Wang, S. Zhao, T. Wang, W. Gong, J. Zhao, E. Li, R. Tan, Y. Hou, "A novel small-molecule compound diaporine A inhibits non-small cell lung cancer growth by regulating miR-99a/mTOR signaling," Cancer Biology & Therapy, vol. 15 no. 10, pp. 1423-1430, DOI: 10.4161/cbt.29925, 2014.
[111] C. Yao, L. Su, F. Zhang, X. Zhu, Y. Zhu, L. Wei, X. Jiao, Y. Hou, X. Chen, W. Wang, J. Wang, X. Zhu, C. Zou, S. Zhu, Z. Xu, "Thevebioside, the active ingredient of traditional Chinese medicine, promotes ubiquitin-mediated SRC-3 degradation to induce NSCLC cells apoptosis," Cancer Letters, vol. 493, pp. 167-177, DOI: 10.1016/j.canlet.2020.08.011, 2020.
[112] Y. T. Ye, W. Zhong, P. Sun, D. Wang, C. Wang, L. M. Hu, J. Q. Qian, "Apoptosis induced by the methanol extract of Salvia miltiorrhiza Bunge in non-small cell lung cancer through PTEN-mediated inhibition of PI3K/Akt pathway," Journal of Ethnopharmacology, vol. 200, pp. 107-116, DOI: 10.1016/j.jep.2016.12.051, 2017.
[113] L. J. Deng, M. Qi, N. Li, Y. Lei, D. Zhang, J. Chen, "Natural products and their derivatives: promising modulators of tumor immunotherapy," Journal of Leukocyte Biology, vol. 108 no. 2, pp. 493-508, DOI: 10.1002/jlb.3mr0320-444r, 2020.
[114] J. Li, S. Wang, N. Wang, Z. Wang, "Research advances of traditional Chinese medicine in cancer immunotherapy," Chinese Medicine and Culture, vol. 3 no. 4, pp. 245-253, DOI: 10.4103/cmac.cmac_42_20, 2020.
[115] K. P. Terracina, T. Aoyagi, W. C. Huang, M. Nagahashi, A. Yamada, K. Aoki, K. Takabe, "Development of a metastatic murine colon cancer model," Journal of Surgical Research, vol. 199 no. 1, pp. 106-114, DOI: 10.1016/j.jss.2015.04.030, 2015.
[116] A. Orillion, A. Hashimoto, N. Damayanti, L. Shen, R. Adelaiye-Ogala, S. Arisa, S. Chintala, P. Ordentlich, C. Kao, B. Elzey, D. Gabrilovich, R. Pili, "Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma," Clinical Cancer Research, vol. 23 no. 17, pp. 5187-5201, DOI: 10.1158/1078-0432.ccr-17-0741, 2017.
[117] H. Li, N. Huang, W. Zhu, J. Wu, X. Yang, W. Teng, J. Tian, Z. Fang, Y. Luo, M. Chen, Y. Li, "Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2," BMC Cancer, vol. 18 no. 1,DOI: 10.1186/s12885-018-4299-4, 2018.
[118] N. Zhang, Y. Dou, L. Liu, X. Zhang, X. Liu, Q. Zeng, Y. Liu, M. Yin, X. Liu, H. Deng, D. Song, "SA-49, a novel aloperine derivative, induces MITF-dependent lysosomal degradation of PD-L1," EBioMedicine, vol. 40, pp. 151-162, DOI: 10.1016/j.ebiom.2019.01.054, 2019.
[119] L. Pang, S. Han, Y. Jiao, S. Jiang, X. He, P. Li, "Bu Fei decoction attenuates the tumor associated macrophage stimulated proliferation, migration, invasion and immunosuppression of non-small cell lung cancer, partially via IL-10 and PD-L1 regulation," International Journal of Oncology, vol. 51 no. 1, pp. 25-38, DOI: 10.3892/ijo.2017.4014, 2017.
[120] B. Zhao, X. Hui, L. Jiao, L. Bi, L. Wang, P. Huang, W. Yang, Y. Yin, S. Jin, C. Wang, X. Zhang, L. Xu, "A TCM formula YYWY inhibits tumor growth in non-small cell lung cancer and enhances immune-response through facilitating the maturation of dendritic cells," Frontiers in Pharmacology, vol. 11,DOI: 10.3389/fphar.2020.00798, 2020.
[121] Y. Luo, J. Wu, X. Zhu, C. Gong, C. Yao, Z. Ni, L. Wang, L. Ni, Y. Li, S. Zhu, "NK cell-dependent growth inhibition of lewis lung cancer by Yu-Ping-Feng, an ancient Chinese herbal formula," Mediators Inflamm, vol. 2016,DOI: 10.1155/2016/3541283, 2016.
[122] D. Karati, R. Varghese, K. R. Mahadik, R. Sharma, D. Kumar, "Plant bioactives in the treatment of inflammation of skeletal muscles: a molecular perspective," Evidence-based Complementary and Alternative Medicine, vol. 2022,DOI: 10.1155/2022/4295802, 2022.
[123] H. Li, W. Wei, H. Xu, "Drug discovery is an eternal challenge for the biomedical sciences," Acta Materia Medica, vol. 1 no. 1,DOI: 10.15212/AMM-2022-1001, 2021.
[124] D. Shah, M. Gandhi, A. Kumar, N. Cruz-Martins, R. Sharma, S. Nair, "Current insights into epigenetics, noncoding RNA interactome and clinical pharmacokinetics of dietary polyphenols in cancer chemoprevention," Critical Reviews in Food Science and Nutrition, vol. no. 3,DOI: 10.1080/10408398.2021.1968786, 2021.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2022 Hongkui Chen et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
Non-small-cell lung cancer (NSCLC) is known as one of the most lethal cancers, causing more than 1 million deaths annually worldwide. Therefore, the development of novel therapeutic drugs for NSCLC has become an urgent need. Herein, various mouse models provide great convenience not only for researchers but also for the development of antitumor drug. Meanwhile, TCM, as a valuable and largely untapped resource pool for modern medicine, provides research resources for the treatment of various diseases. Until now, cell-derived xenograft (CDX) model, patient-derived xenograft (PDX) model, syngeneic model, orthotopic model, humanized mouse model (HIS), and genetically engineered mouse models (GEMMs) have been reported in TCM evaluation. This review shows the role and current status of kinds of mouse models in antitumor research and summarizes the application progress of TCM including extracts, formulas, and isolated single molecules for NSCLC therapy in various mouse models; more importantly, it provides a theoretical exploration of what kind of mouse models is ideal for TCM efficacy evaluation in future. However, there are still huge challenges and limitations in the development of mouse models specifically for the TCM research, and none of the available models are perfectly matching the characteristics of TCM, which suppress the tumor growth through various mechanisms, especially by regulating immune function. Nevertheless, with fully functional immune system existing in syngeneic model and humanized mouse model (HIS), it is still suggested that these two models are more suitable for development of TCM especially for TCM extracts or formulas. Moreover, continued efforts are needed to generate more reliable mouse models to test TCM formulas in future research.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details




1 Shanghai Lidebiotech Co. Ltd., Shanghai 201203, China
2 School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
3 School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China