Next Article in Journal
Impact of the Number of Positive Pelvic Lymph Nodes on Risk of Para-Aortic Recurrence in Patients with Clinically Early Cervical Cancer Treated by a Radical Hysterectomy and Pelvic Lymphadenectomy
Next Article in Special Issue
Correction: Dekker, S.E.; Deng, L. Clinical Advances and Challenges in Targeting KRAS Mutations in Non-Small Cell Lung Cancer. Cancers 2024, 16, 3885
Previous Article in Journal
Prospects for Narrow Band Imaging Magnification Endoscopy in Oral Lesions: Recommendations from Oral and Maxillofacial Surgeons and a Gastroenterologist
Previous Article in Special Issue
Clinical Advances and Challenges in Targeting KRAS Mutations in Non-Small Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Preclinical Models for Functional Precision Lung Cancer Research

by
Jie-Zeng Yu
1,†,
Zsofia Kiss
1,†,
Weijie Ma
1,2,
Ruqiang Liang
1 and
Tianhong Li
1,3,*
1
Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
2
Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
3
Medical Service, Hematology/Oncology, Veterans Affairs Northern California Health Care System, Mather, CA 10535, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(1), 22; https://doi.org/10.3390/cancers17010022
Submission received: 15 November 2024 / Revised: 20 December 2024 / Accepted: 23 December 2024 / Published: 25 December 2024

Simple Summary

The landscape of lung cancer diagnosis and treatment has transformed over the past two decades, ushering in the era of precision medicine for lung cancer. In systemic therapy alone, 43 drugs have gained FDA approval since 2020, encompassing chemotherapy, molecularly targeted therapies, immunotherapy, and antibody–drug conjugates. These therapies have significantly improved patient survival and quality of life. Improved preclinical models have been crucial in driving these advancements. Increasingly, the impact of tumor heterogeneity, along with its interplay with the tumor microenvironment and immune system, has been recognized as critical in influencing responses to these agents. This review provides an overview of the key characteristics, advantages, and limitations of current in vitro and in vivo models used in functional precision lung cancer research of systemic therapeutics.

Abstract

Patient-centered precision oncology strives to deliver individualized cancer care. In lung cancer, preclinical models and technological innovations have become critical in advancing this approach. Preclinical models enable deeper insights into tumor biology and enhance the selection of appropriate systemic therapies across chemotherapy, targeted therapies, immunotherapies, antibody–drug conjugates, and emerging investigational treatments. While traditional human lung cancer cell lines offer a basic framework for cancer research, they often lack the tumor heterogeneity and intricate tumor–stromal interactions necessary to accurately predict patient-specific clinical outcomes. Patient-derived xenografts (PDXs), however, retain the original tumor’s histopathology and genetic features, providing a more reliable model for predicting responses to systemic therapeutics, especially molecularly targeted therapies. For studying immunotherapies and antibody–drug conjugates, humanized PDX mouse models, syngeneic mouse models, and genetically engineered mouse models (GEMMs) are increasingly utilized. Despite their value, these in vivo models are costly, labor-intensive, and time-consuming. Recently, patient-derived lung cancer organoids (LCOs) have emerged as a promising in vitro tool for functional precision oncology studies. These LCOs demonstrate high success rates in growth and maintenance, accurately represent the histology and genomics of the original tumors and exhibit strong correlations with clinical treatment responses. Further supported by advancements in imaging, spatial and single-cell transcriptomics, proteomics, and artificial intelligence, these preclinical models are reshaping the landscape of drug development and functional precision lung cancer research. This integrated approach holds the potential to deliver increasingly accurate, personalized treatment strategies, ultimately enhancing patient outcomes in lung cancer.

1. Introduction

Lung cancer is the leading cause of cancer-related death worldwide [1]. Fortunately, the management of lung cancer has significantly evolved over the last decade, as reviewed in this special issue [2,3,4,5,6,7]. Lung cancer is a heterogeneous disease, and its clinical management depends on histopathological diagnosis, staging, and molecular and immune biomarker findings [8,9]. Non-small cell lung cancer (NSCLC) accounts for over 80% of lung cancer cases, followed by small cell lung cancer (SCLC), which makes up 10–15% [5]. It is reported that approximately 40% of newly diagnosed lung cancer patients exhibit metastasis disease [10]. Based on tumor origin, NSCLC is further classified into lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), neuroendocrine tumors (~5%), sarcomas, and SMARCA4-deficient subtypes [11,12,13].
Precision oncology represents a paradigm shift toward a more individualized approach in cancer care, tailoring treatments based on the unique characteristics of each patient and their disease, especially for late-stage cancer patients [14]. NSCLC has become a prominent example of precision medicine among solid tumor cancers. All patients with NSCLC undergo immune biomarker PD-L1 immunohistochemistry (IHC) and tumor genomic profiling to guide the selection of first-line and subsequent systemic treatments, including molecularly targeted therapy or immune checkpoint inhibitors (ICIs), either alone or in combination with chemotherapy [7]. However, despite initial responses, resistance inevitably develops. The advances in cancer treatment would not have been possible without preclinical experimental models and technological improvements that enable the investigation of various aspects of disease initiation, progression, and tumor responses. Furthermore, patient-derived tumor models are essential for elucidating the mechanisms of drug resistance, evaluating novel drug efficacy with or without current treatments, and identifying biomarkers of response for patient stratification to inform future personalized therapies. This comprehensive review aims to summarize the different in vitro and in vivo models (Figure 1), highlighting their advantages, limitations, and applications in advancing personalized medicine.

2. In Vitro Model

2.1. Human Lung Cancer Cell Line Models

Human lung cancer cell lines are one of the earliest preclinical models and have been utilized widely to gain a plethora of information on cancer initiation, progression, and metastasis on the molecular and cellular levels. The use of these cancer cell lines significantly advanced our understanding the molecular biology of lung cancer. These cancer cell lines have shown the ability to maintain the expression of the “hallmarks of cancer” [15] (except for angiogenesis that requires the presence of stromal tissues). These include driver mutations that have been identified and are essential for the malignant phenotype. The identification of these driver mutations would have been impossible without the use of these cancer cell lines where one can knockout or overexpress certain oncogenes or tumor suppressors to understand their function and role in the development and maintenance of cancer. For example, in vitro lung cancer cell lines allowed the identification of TP53 mutations. Their significance in lung cancer has provided an understanding on the relationship between copy number gains, other mutations, and mutant-allele-specific imbalance in cancers [16,17,18]. Identifying sites of allelic loss or gain in lung cancer cell lines that are more frequent was also crucial in understanding the common key players in lung cancer pathogenesis. For example, lung cancer cell lines were used to show that RB is crucial in small cell lung carcinoma (SCLC) [19]. RB, just like CDKN2A, acts as a cell cycle checkpoint. While CDKN2A is known to be inactivated in many tumors, interestingly, inactivating point mutations in RB are mainly limited to SCLC and bladder cancers. In addition, the concept that there was a mutually exclusive RB-cyclin–CDKN2A tumor suppressor pathway and that this pathway can be inactivated by either mutational or epigenetic alterations in various human cancers was discovered using lung cancer cell lines [20,21].
Recent WHO clarification defines four molecular subtypes of small cell lung carcinoma (SCLC), which has therapeutic implications for emerging precision oncology treatment [22]. These subtypes were defined based on the differential expression of four key transcriptional regulators: achaete-scute homolog 1 (ASCL1, also known as ASH1), neurogenic differentiation factor 1 (NeuroD1), yes-associated protein 1 (YAP1), and POU class 2 homeobox 3 (POU2F3) [22]. Where multiple transcription regulators are expressed, samples are classified by the expression level of the transcription regulator that has the greatest relative overall expression. Studies focusing on understanding the critical signaling pathways dominating in SCLC subtypes would allow the research community to develop more promising therapeutic targets. For example, recent data indicated that Delta-like protein 3 (DLL3), including an antibody–drug conjugate, a bispecific T cell engager, and a chimeric antigen receptor (CAR)-T cell construct, can target SCLC-A subtypes [23]. Unfortunately, despite many targeted agents that are effective in many hematological and solid tumors, their success in treating SCLC have been disappointing [24,25,26,27].
The use of these various NSCLC cell lines is far more common and has allowed researchers to identify a plethora of genes that have been shown to be important in lung cancer pathogenesis [28,29]. The gene TITF1, known to play a role as a master transcription factor required for the differentiation of the peripheral airway, has been shown to be frequently amplified in lung cancer tumors and cell lines and has been shown to be a lineage-specific oncogene [30,31]. Gaining an understanding that the activation mutation in the kinase domain of the epidermal growth factor receptor (EGFR) was in close correlation with the sensitivity/resistance to tyrosine kinase inhibitor-based drugs (TKIs) was achieved via studies conducted using lung cancer cell lines [32]. Subsequently, almost all important biological characterization of intrinsic and acquired resistance mechanisms in the EGFR mutations was performed using cell lines [33,34].
In summary, without these cancer cell lines, the findings we gained and successfully “translated” to clinical applications would have been severely hampered. These cell lines are relatively inexpensive, scalable, and widely available [35,36]. Over 300 NSCLC cell lines are available in cell line collections [37,38]. Most NSCLC cell lines are derived from LUAD, and fewer LUSC cell lines are available due to the difficulty of culturing them. Thus, although sequencing efforts have allowed the identification of mutational status for many NSCLCs, unfortunately, LUSC cell lines tend to be less well characterized. Fortunately, these NSCLC cell lines have been shown to maintain some of the fundamental features of the tumors from which they were derived. Table 1 summarizes common human lung cancer cell lines and their genetic mutations. For example, Cross et al. used the EGFR-mutant lung cancer cell lines PC9, H1975, H1650, and H3255 to validate the function of EGFR inhibitor osimertinib, and their results indicated effective inhibition of EGFR phosphorylation [39]. Unfortunately, however, due to their extensive use and passaging from the time they were established, they do show differences in their genetic characterization of the parental tumor [40], thus limiting the reflection of the clinical response in drug screenings in a clinical trial setting.

2.2. Primary Cell Cultures

Due to the concern for potential loss or alteration in some of the genomic characters of the immortalized human lung cancer cell lines during passages, patient-derived primary cell cultures have been extensively studied during the past decade. These patient-derived primary cell cultures better preserve the tumor genomic profiles due to the relative recency of their establishment [65]. Moreover, primary cell cultures have a great advantage over immortalized cell lines for understanding the cellular interaction between various cell types including epithelial cells, fibroblast cells and immune cells. For example, Beverly et al. were able to establish an in vitro tumor microenvironment (TME) system using primary cell culture that allowed them to rapidly expand stromal progenitors from resected patient lung tumor specimens and showed that these progenitor populations retained the expression of pluripotency markers and secreted factors that are associated with cancer progression and tumor growth [66]. This model provides a valid tool to understanding the biological functions of these progenitor cell populations and develop strategies to inhibit the tumor growth and metastasis.
Another advantage to primary cell cultures is in the aspect of drug discovery and toxicity studies. Evaluating the efficacy and/or toxicity of candidate drugs or their therapeutic potential using primary cell cultures may provide a reflection closer to the predictive biological response in patients with identical or closely related disease conditions. As such, Benet et al. showed that using patient-derived primary cell culture combined with an immunofluorescence-based functional assay can effectively quantify tumor cell response to targeted therapy in mixed cell cultures [67]. An interesting study was conducted by Dandachi and her colleagues, where they obtained a primary cell culture from a patient with primary resistant LUAD. They identified two biologically profoundly distinct adenocarcinoma cell subpopulations. One they cultured in a spheroid culture system, while the other was only able to grow and proliferate under adherent conditions. They characterized these subpopulations and learned that the subpopulation from the spheroid culture system was strongly associated with the epithelial phenotype and expressed many of the cancer stem cell (CSC) markers, such as ALDH1 and CD133, while the adherent subpopulation was of a mesenchymal feature and did not express CSC markers. When testing this subpopulation with cisplatin, they demonstrated that the subpopulation from the spheroid culture was significantly more resistant to cisplatin compared to the adherent subpopulation. This study indicated that the primary LUAD cell culture was derived from a patient with resistant disease with an epithelial aggressive subpopulation of cells associated with stem cell features and resistance to therapy [68]. In summary, primary cell cultures can be invaluable because they more closely resemble the properties of cells in tissue and TME as well as allow analysis of cells from specific patient populations.
One frequent challenge when working with primary cell cultures is the lack of indefinite proliferative capacity, which can limit their utility in long-term culture of these cells. However, recent advances in cell culture techniques and the development of specialized culture media, scaffolds, and growth factors have made it possible to extend the lifespan of primary cells. As such, Odintsov and his colleagues established two cell line models from NRG1-rearranged (Neuregulin-1) lung adenocarcinoma samples and showed via transcriptome analysis the activation of the mTOR pathway in these NRG1 fusion-bearing samples. These cell lines were also more sensitive to mTOR pathway inhibitors (such as rapamycin), indicating potential therapeutic significance for patients for whom ERBB-directed therapy fails [69]. Studies indicate the potential advantage of primary cell cultures over established human lung cancer cell lines; however, the success rate in establishing patient-derived primary cell cultures is relatively low [67,70].

2.3. Conditionally Reprogrammed Cell

Conditional reprogramming (CR) is a type of cell culture that involves culturing the cells from patient samples with irradiated mouse cells in the presence of a Rho-kinase inhibitor (ROCK). The most common Rho kinase inhibitor is Y27632, and the most common irradiated mouse cell line used is Swiss 3 T3-J2 mouse fibroblasts [71]. Cells can quickly develop a stem-like character with high proliferation potential, retaining the original karyotypes under this specific condition [72]. This type of culture system (CR) is widely used for cells of epithelial origin.
One of the key advantages of the CR system is that it allows researchers (both basic and preclinical researchers) to describe the biological characteristics of cancer and explore and identify the relevant mechanism for drug resistance, maintenance, and establishment of tumors. It faithfully mirrors primary cancerous cells and can delineate the cellular, molecular, and genetic characterizations of cancer malignancy. As such, the CR system was used to establish primary cultures from NSCLC, and drug-response profiling was used to identify the histopathological subtypes of NSCLC-selective signal plasticity and associated therapeutic weaknesses [73]. For example, Brodovsky et al. established human ovarian and lung patient-derived xenograft (PDX) tumors into conditionally reprogrammed cell lines (CR-PDX). When comparing the genetic profile and histology of this CR-PDX to the parental tumors, they showed that CR-PDX maintained the original characteristics of the parental tumors with high fidelity [74]. Several studies used CR cells to elucidate the molecular players that have been shown to be crucial in cancer malignancy and thus establish a better target to abolish tumor growth. For example, Beglyarova et al. showed that MYC-ERCC3 interaction is a critical survival signal for pancreatic ductal adenocarcinoma (PDAC) via the use of conditionally reprogrammed patient-derived pancreatic cancerous cells [73]. Likewise, Yuan et al. used the CR system to identify a unique mutation from a duplication of the promoter and oncogene regions in the HPV-11 viral genome that were responsible for aggressive clinical features [75]. Alamri et al. used the CR technology to identify a novel therapeutic target, a gene fusion called KRT14-KRT5, in mucoepidermoid carcinoma and other salivary-gland neoplasms [76]. One advantage of using these CR cell lines is that besides retaining the genetic characteristics of the parental tumors, they are amenable to gene manipulation and drug screening and can even be used for implantation into immunodeficient mouse models for in vivo experiments [77]. One of the most common ways to obtain tumors is via biopsy; however, it is an invasive method, and thus, there are more and more studies focusing on obtaining tumor cells in a non-invasive manner, such as collecting cancer cells from other body fluids such as blood or urine. Jiang et al. reported an overall successful rate of 83.3% in establishing CRC of bladder cancer from urine [78].

2.4. Cancer Spheroids and Organoids

While the establishment of lung cancer cell lines over the past few decades has allowed the advancement of lung cancer research, it has also brought many potential challenges. As such, while cell lines with distinctive EGFR (epidermal growth factor receptor) mutations show different sensitivity to TKIs (tyrosine kinase inhibitors) [79], unfortunately, they fail in clinical trials using experimental therapies [65]. The introduction of immunocompromised mouse colonies in research has allowed the establishment of PDX mouse models that allow not only the maintenance of patient-derived cancer cells that do not survive in vitro for prolonged periods of time but also the establishment of the complex microenvironment a tumor would typically grow in, including a nutrient- and oxygen-rich blood supply capable of removing toxins, the extracellular matrix, the presence of other cell types, and growth factors. It allows tumors to promote blood vessel formation and to metastasize—something that cannot yet be achieved in in vitro settings.
Unfortunately, the inherent variability from mouse to mouse in establishing tumors leads to difficulty in establishing a standardized protocol. Furthermore, the lack of competent immune system in the PDX models indicates that they may not accurately reflect disease progression and the therapeutic response observed in immune-competent humans. For example, studies focusing on ICI-based therapies or other immunotherapeutic cancer vaccine research using these PDX mouse models will not be feasible to assess the efficacy of these drug studies.
Patient-derived lung cancer spheroids were first established by Eramo et al. in 2008 [80]. They were able to create a personalized 3D model to use to generate xenografts that recapitulated the histology of the parental tumors [80]. From cell composition to tissue structure, they are similar to human tumors; organoids can self-renew and self-organization, which can simulate the microenvironment of human cancer [81]. With the establishment of spheroids, many laboratories have focused on using patient-derived lung cancer spheroids for both in vitro and in vivo studies for drug testing and various molecular analyses [82,83,84,85,86]. The attempt by Endo et al. to generate NSCLC spheroids from tumor tissues or pleural effusion using Matrigel marked a step closer to optimizing organoid culture conditions [87]. These advancements further stimulated researchers to attempt to generate lung 3D cultures in a semi-structured environment using human respiratory epithelial cells isolated from nasal polyps. This method showed the generation of tubular structures containing cuboidal-shaped polarized cells, ciliated cells, secretory cells, and undifferentiated cells with epithelial cells that can contract [88]. However, using lung cancer spheroids for patient prognosis and for the ability to be able to select the right treatment of choice confidently is limited.
These studies prepared the road for lung organoid development. Following the path, various laboratories successfully established the first generation of normal lung organoids using human induced pluripotent stem cells (iPSC) [89,90,91]. Soon after, organoids were derived from adult lung primary cells and embryonic lung epithelial cells [92,93]. All these further enhanced our understanding of the culturing conditions with many trials. Clevers et al. were the pioneers in establishing lung cancer organoids (LCOs). The unique characteristics of these LCOs were that they retained the histopathology and the mutation profile of the original tumors and were amenable for both small-scale drug-screening experiments and orthotopic transplantation [94]. One reoccurring problem and disadvantage observed during these advancements is the competitive growth of normal epithelial cells. One possibility of overcoming this phenomenon is using media that lack the necessary factors to grow normal lung organoids [95]. Other possibilities include hand-picking tumor organoids [96,97] and deriving cancer cells from extrapulmonary sources such as metastases [94,98,99,100], pleural effusions, or PDXs [99,100,101,102]. While the generation of LCOs from lung cancer metastases has been shown to have a higher rate of success than from intrapulmonary samples [96], biopsy samples tend to be very small in size. Malignant effusion samples have shown promise as a source for LCOs, but the number of studies is still very low [99,100,101]. However, Kim et al. in their study showed that the LCOs they generated from effusion samples did, indeed, reproduce the genetic features of advanced LUAD and that they were amenable for predictive studies [99].
The unique characteristics mentioned above of the tumor organoids allow both basic and translational research applications. Several studies in various cancer types have used organoids to study critical steps in cancer initiation and progression, such as self-renewal, drug resistance, heterogeneity oncogenic transformation, circulating tumor cells [103,104,105,106,107,108], and drug screening [109]. The first study using lung organoids to obtain transcriptional and proteomic profiles or normal epithelial progenitors as compared to early-stage lung cancer was performed by Dost et al., where they used organoids that were derived from human iPSC and murine lung epithelial cells to model LUAD development [110]. Their study provided a comprehensive molecular landscape of K-RAS-driven lung tumorigenesis and, furthermore, encouraged other researchers to carry out studies on LCOs to provide new insights on the role of specific genes in lung cancer and/or on the biological functions of lung cancer cells [102,111,112,113].
The establishment of the LCO technology allowed researchers to perform the first drug-testing experiments within the context of various genomic alterations [114]. Sachs et al. were among the first to show that LCOs are, indeed, amenable to drug screening while being able to retain driver mutations of parental tumors with differential responses to chemotherapeutics and targeted inhibitors such as erlotinib, gefitinib, crizotinib, and alpesilib [94]. Kim et al. were able to establish a biobank that contained 80 LCOs and recapitulated the histology and genetic features of major lung cancer subtypes and responded to targeted drugs respective to their genomic alterations [99]. Various laboratories followed the establishment of biobanks composed of LCOs and tested both monoclonal antibody-based as well as antibody–drug conjugates (ADCs), ICIs, and targeted inhibitors and have found that LCOs are amenable for drug screening and that their responses correlated with their genetic profile, which represents the parental tumors’ genetic profile with high fidelity [109,115,116]. Interestingly, Li Z. et al. showed that several drugs were effective on LCOs without the related mutation. While this clearly indicates that not 100% of organoids preserve the genetic profile, drug testing using LCOs correlated with the predictive drug response as shown previously by PDO-based clinical trials [117,118]. It also indicates that routine drug testing on LCOs may disclose patients that could unexpectedly benefit from certain targeted treatments that otherwise their genetic profiling would not indicate [119]. Single-cell RNA (scRNA) sequencing and quantitative imaging technology have been used to assess the spatial, genomic, and transcriptome analysis of organoids at the single-cell level, which holds tremendous value in precision oncology in lung cancer. Studying the heterogeneity and evolution of tumor organoids via scRNA sequencing indicated that the CD44-positive subpopulation is responsible for drug resistance by hyper-activating the Jak-STAT signaling pathway in a hepatobiliary tumor [118]. Interestingly, in another study, Wang et. showed that the organoids from lung cancer patients’ malignant serous effusion match both the pathological characteristics and genomic profiling with the original malignant serous effusion with only a few somatic alterations. In vitro drug screening on these organoids was tailored to individual patients and was shown to discriminate between clinically sensitive and resistant patients [120].

2.5. Co-Culture System of Patient-Derived Immune Cells and Patient-Derived Tumors

Recent studies have shown the importance of various cellular and molecular components in the TME in prognosis and treatment responses to immunotherapy. Co-culture of LCOs with the systemic adding of key components of these factors allows researchers to define its contributions using in vitro models. Monoclonal antibody-based therapies that target the inhibitory receptors expressed by immune cells (i.e., ICIs) have shown remarkable response rates in various solid tumor malignancies [121]. Developing preclinical models that allow one to investigate the TME and guide clinical precision therapy is gaining more importance [122]. ICIs have emerged as a groundbreaking advancement in treating various cancers. To name a few, antiprogrammed cell death protein 1 (PD-1) and anticytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have shown significant clinical efficacy in specific patient populations [123,124]. Many clinical trials indicate these therapies’ effectiveness [125] and, more importantly, have demonstrated exceptionally high efficacy in tumor types characterized by high mutational burden [126]. However, unfortunately, while these therapies focusing on ICIs show promise, their effectiveness, especially in solid tumors, is still very limited [127].
Studies have focused on establishing a co-culture system with peripheral blood mononuclear cells (PBMCs) and cancer cells. For example, Saraiva et al. could co-culture PBMCs and spheroids derived from the breast cancer cell line MDA-MB-230. They observed that the patient’s immune cells exhibited a wide range of antitumor responses and that this can be manipulated to improve their ability to lower the viability of tumor cells [128]. Co-culture of autologous tumor organoids and PBMCs is a great way to enrich tumor-reactive T cells from the PBMCs of patients. It can be used to assess the efficiency of killing matched tumor organoids. Dijkstra et al. showed in their study that co-culturing the autologous tumor organoids with PBMCs indeed can provide an unbiased strategy for the isolation of tumor-reactive T cells and can provide a means by which one can assess the sensitivity of tumor cells to T cell-mediated attack at the level of the individual patient [129]. Ma et al. used cell pellets from pleural effusions from patients that were characterized as oncogene-driven, based on if patients whose tumors had at least one driver oncogene (EGFR, MET exon 14 skip, or ErbB2 mutation, ALK, ROS1, or RET fusions), or non-oncogene-driven; these were used to co-culture with their respective autologous PBMCs to assess the blood biomarker profiles with respect to the effect of tyrosine kinase inhibitors (TKIs) and the responsiveness to ICIs [130]. The study revealed that ICI treatment activated additional immune cell types and that TKI treatment could either antagonize or enhance the effect of ICIs [130]. Table 2 summarizes the advantages, disadvantages, and key applications of these in vitro models.

3. In Vivo Models

The expansion of various in vitro models in cancer research has enabled us to advance in understanding basic tumor biology from initiation to metastasis and allowed the identification of the major molecular players that have an essential role in tumorigenesis. The sequencing of larger fragments ultimately allowed us to sequence the human genome along with the genomes of many other organisms and learn the genomic similarities between humans and mice, for example. These further encouraged researchers to establish mouse models for cancer research to understand tumor growth. All these led to the development of various mouse models from immunocompetent humanized animals that bear human tumors with a responsive immune system to learn the immune response and tumor interaction to immunocompromised animals that can tolerate cross-species cancer cells such as human cancer cells and nude mice strains that not only lack immune system but also are hairless, allowing one to be able to perform whole body imaging (Figure 2). This next section briefly summarizes the various mouse models that are frequently used and their potential in cancer research. Table 3 summarizes the advantages, disadvantages and key applications of these in vivo models.

3.1. Carcinogen-Induced Mouse Model

These models refer to particular cancer types developed in the animals via exposure to certain environmental risk factors, for example, carcinogenic chemicals, radiation, viruses of microbial flora, or physical stimuli [131]. One of the most significant advantages to these models is that they can simulate the tumor progression from the early stage, thus leading to the identification of general mechanisms of cellular alterations required for tumor formation. Additionally, they may be able to provide insights into tissue-specific features. In lung cancer studies, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced mouse models are a great source to study lung cancer development due to cigarette smoking. Other compounds such as polyaromatic hydrocarbons as benzo(a)pyrene (BaP), N-nitroso-tris-chloroethylurea (NTCU), diethylnitrosamine, and 2-methycholanthrene (MCA) are also widely used for establishing chemically induced lung cancer mouse models [132]. For example, Wang et al. used NTCU on eight different strains of mice via skin painting and found that in five strains (SWR/J, NIH Swiss, A/J, Balb/cJ, and FVB/J), this method allowed for the establishment of lung small cell carcinoma (SCC). However, this method failed to establish tumor models in the inbred strains AKR/J, 129/svJ, and C57BL/6J [133]. Another study focused on repeated intratracheal injections of MCA in BC3Fl and DBA/2 mice strains and were able to induce squamous cell lung carcinoma [134]. When orally administering BaP to Swiss albino mice at 50 mg/kg dosage, twice a week for four weeks, Rajendran et al. were able to induce lung tumor models [135]. A similar study also succeeded where the researchers administered BaP at 100 mg/kg intraperitoneally into A/J mice to assess the chemoprevention efficacy of deguelin and silibinin [136]. Taken together, many of the compounds above are a great source when establishing lung cancer mouse models to study the effects of cigarette smoking, tumor initiation and progression, and the potential effects of currently available chemotherapeutics to combat lung cancer.

3.2. Syngeneic Mouse Models

Syngeneic mouse models are powerful resources that allow the generation of tumors in immunocompetent mice. Their ability to model the genetics of human disease is limited, but gene editing strategies can be used to engineer clinically relevant mutations. Notably, most human tumors with driver gene(s) mutation also tend to be coupled with genomic rearrangements, which these models still fail to recapitulate. Despite these limitations, these models are still very powerful, as one can generate orthotopic tumors, and these tumors will have similar histological features as their human counterparts, thus allowing one to study both the evolution of a complex TME and the assessment of various therapeutics [137,138]. Furthermore, assessing the response to various immunotherapies places these mouse models at the forefront of other immunocompromised mouse models. It has allowed researchers to understand the resistance mechanism of certain immunotherapies, such as anti-PD-1 [139]. Several other studies highlight the importance of these models, from assessing combination therapy with checkpoint inhibitors to identifying genetic players that correlate with pathways that, if targeted in combination, can reduce tumor growth and prolong the overall survival of patients. For example, Ajona et al. established syngeneic mouse models for lung cancer and showed that inhibition of the C5a/C5aR1 and PD-1 signaling have synergistic antitumor effects [140]. Meraz and his colleagues delivered TUSC2 (tumor suppressor that encodes a multikinase inhibitor and has been shown to be lost in non-small cell lung carcinoma) systemically by nanovesicles, which was shown to mediate tumor regression. Since TUSC2 is known to regulate immune cells, Meraz and his team assessed the TUSC2 efficacy on antitumor immune response alone and in combination with anti-PD-1 in K-RAS mutant syngeneic mouse models. Their results indicate that while TUSC2 alone significantly reduced tumor growth and prolonged survival compared with anti-PD-1 only, when combined, this effect was significantly enhanced and correlated with an increase in circulating natural killer (NK) cells and CD8+ T cells and a decrease in regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and T cell checkpoint receptors PD-1, CTLA-4, and TIM-3 [141]. In summary, while it is not fully understood yet if syngeneic models truly reflect the human immune response, it is clear that these models are extremely useful in many aspects of cancer biology and allow researchers to obtain a plethora of functional assessments to pave the road for the development of novel therapeutics.

3.3. Transgenic/Genetically Engineered Mouse Models

The genetically engineered mouse model (GEMM) for cancers refers to an animal strain with manipulated genomic alterations, specifically the overexpression of an oncogene or the loss of a tumor suppressor gene function [142]. The GEMMs can be divided into transgenic and endogenous ones [143], allowing the investigation of the function of certain genes or pathways during tumor development or progression [144]. The ability to manipulate certain gene(s) in these models allow for further study to evaluate anticancer drug efficacy [145]. Furthermore, since these models are immunocompetent, they can also be invaluable for immune therapy assessments. For example, the K-RAS-LSL-G12D mouse model is an excellent source in lung cancer research as most lung cancer patients with a history of cigarette smoking are also known to have a mutation in the K-RAS gene. There are various ways to establish GEMs, most relying on engineered nucleases. These engineered nucleases are composed of sequence-specific DNA-binding domains fused to a non-specific DNA cleavage molecule [146,147]. The advantage of these chimeric nucleases is that they are able to execute genetic modifications in an efficient and precise manner by inducing targeted DNA double-strand breaks (DSBs) which then triggers the cellular DNA repair mechanism either via error-prone non-homologous end joining (NHEJ) or via homology-directed repair (HDR) to “repair” the cleavage [148]. Zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) were at the forefront where the DNA-binding domain was fused with the cleavage domain of the FokI endonuclease [149,150]. Briefly, a pair of these nucleases bind to opposite strands of adjacent sequences separated by a short spacer sequence where the target site is located [149,150]. The usage of these nucleases is still limited because their construction requires modular assembly technology for generating the DNA-binding domains. The next most effective and simplest engineered nucleases are the CRISPR/Cas9 system. This system contains the Cas9 nuclease and so-called single guide RNA (sgRNA) M [150,151]. The single guide RNA is usually about 20 nucleotides in length and is complementary to the target site, followed by a tri-nucleotide protospacer adjacent motif (PAM) in the genome. This allows the Cas9 nuclease to be recruited to the target sequence [149,151]. Because of its simplicity and precision, CRISPR/Cas9 is currently the most prominent tool for genome engineering. Rakhit et al. used a Cre-regulated genetically engineered mouse model for lung adenocarcinoma development driven by K-RAS G12D (K-RAS-LSL-G12D mouse model). They tracked the release of cell-free DNA vs. cell-tumor DNA (cfDNA/ctDNA) and compared this with the tumor burden that was captured by micro-computed tomography (CT). To monitor ctDNA, they developed a droplet digital PCR (ddPCR) assay so that they can discriminate the K-RAS-lox-GD12 allele from the K-RAS-LSL-G12D and the K-RAS WT (wild type) alleles. They showed that the micro-CT correlated with endpoint histology and detected pre-malignant tumors with a combined tumor volume of 7 mm3 or larger [152]. By this model, they validated that cfDNA/ctDNA levels can be used as an early detection method for lung cancer, which correlated with mouse micro-CT measurement results in a consecutive test.
In another study focusing on the initiation and maintenance of lung cancer, researchers established two doxycycline-inducible transgenic mouse models: one bearing a point mutation in the EGFR gene (substitution of arginine for leucine) at exon 21 (EGFR L858R), while the other expressing a deletion of exon 19 (EGFR DeltaL747-S752). Both are common mutations in most lung cancer patients. When induced with doxycycline, they could express these EGFR mutants that lead to the development of lung adenocarcinomas. Interestingly, two weeks after doxycycline indication, the EGFR L8585R mutants showed diffuse lung cancer that resembled human bronchioalveolar carcinoma in contrast to the mutant-deletion mice (EGFR DeltaL747-S752) that developed multifocal tumors embedded in normal lung parenchyma. When they withdrew doxycycline or treated with first-generation EGFR TKI erlotinib, they observed tumor regression, indicating that these mutations are indeed required for tumor maintenance [153]. It is worthwhile to note that secondary mutations arising in tumors after an antitumor treatment is a very common phenomenon, and Politi et al. showed that GEMMs can simulate this. This was demonstrated by using the point mutant mouse model (EGFR L858R) that started treatment with erlotinib. In the study, they showed that initially, tumors were sensitive to erlotinib, but after multiple cycles of drug treatment, they became resistant. They correlated this resistance to a secondary mutation arising in the EGFR T790M and MET amplification [154]. Using an EGFR L858R/T790M transgenic mouse model, K-K Wong’s team showed that second-generation EGFR TKI afatinib (BIBW2992) induced tumor regression in xenograft and transgenic lung cancer models [155]. MET amplification is a resistance mechanism common to third-generation EGFR TKI osimertinib. Maraver’s lab recapitulated this acquired molecular resistance mechanism by generating an EGFR/MET transgenic mouse model and showed the addition of MET inhibitors to osimertinib-induced tumor regression [156]. These important preclinical data support the clinical testing of these drugs that have gained FDA approvals.

3.4. Cell Line-Derived Xenografts (CDXs)

CDXs are widely used for preclinical drug efficacy tests, pharmacokinetic and pharmacodynamic (PK/PD) correlation, and combination therapy. Human tumor cell line xenografts establish tumors when implanted in immunodeficient mice. In this model, it is feasible to investigate the efficacy of novel agents in inhibiting tumor growth and development and the mechanisms of action. Although implanting human tumors directly into mice is gradually becoming popular in antitumor drug discovery due to their accurate recapitulation of patient tumor features, CDX models are still proven to be an efficient method for in vivo study in terms of their excellent repeatability. There are approximately 300 cell lines for human lung cancer, most of which belong to NSCLC, and only a few belong to SCLC. The use of CDXs is important in understanding drug effect with respect to tumor inhibition. For example, one research group established CDXs by suspending 2 × 106 A549 cells in 100 μL serum-free medium and inoculating subcutaneously into 4–5-week-old male BALB/C nude mice. It took about two weeks to reach a tumor volume of 100–120 mm3. Then they treated these mice with FEN1 inhibitor and cisplatin, respectively, or in combination. FEN1 is a major component of the base excision repair pathway for DNA repair systems and is important in maintaining genomic stability through DNA replication and repair. They showed that FEN1 is critical for the rapid proliferation of lung cancer cells, as when FEN1 is suppressed, a decrease in DNA replication and the accumulation of DNA damage were observed, leading to apoptosis.
How FEN1 was altered also dictated how the lung cancer cells responded to chemotherapeutic drugs. If they targeted FEN1 with a small-molecule inhibitor, they noticed the enhanced therapeutic effect of cisplatin [157]. In another study, Kim et al. used CDX models to identify the Hedgehog pathway transcription factor GLI1 as a critical driver for lung squamous cell carcinoma. Human lung cancer datasets indicated that GLI1 mRNA is highly expressed and correlates with poor prognosis. While inhibitors targeting the Hedgehog pathway did not seem to alter the expression of GLI1, interestingly, modulation of the PI3K/Akt axis allowed them to modulate GLI1 expression. When these tumors were grown in CDXs, they observed that tumor growth could be attenuated in the tumors that harbor amplification of PIK3CA (PI3K gene) by antagonizing GLI1 and PI3K, further supporting their regulatory function [158]. It is clear from these and other similar studies that CDXs are invaluable in assessing drug effects and can aid in identifying molecular players that may have a crucial role in cancer initiation and progression. One drawback of CDXs is that they rely on established human cell lines, which are mainly of Caucasian origin. Thus, our research is gaining an understanding of tumor characteristics and drug responses, which are narrowed down only to Caucasian populations, thus limiting the predictable response in other ethical groups. Furthermore, the tumors formed in CDXs lack the TME that would occur in a real setting.

3.5. Patient-Derived Xenografts (PDXs)

The introduction of immunocompromised mouse colonies in research has allowed the establishment of PDX mouse models that allowed both the maintenance of patient-derived cancer cells that do not survive in vitro for prolonged periods and also the establishment of the complex microenvironment a tumor would normally grow in, including a nutrient- and oxygen-rich blood supply capable of removing toxins, the extracellular matrix, the presence of other cell types, and growth factors. The PDX model inoculates a patient’s tumor tissue directly into immunodeficient mice by subcutaneous or orthotopic transplantation. The most commonly used strain for the establishment of PDX models is the NOD-scid IL2Rgammanull (NSG) mice that lack functional B and T cells and natural killer cell activity and are the most immunodeficient yet are physiologically durable and can establish consistent engraftment of human primary tumors; they are invaluable for in vivo drug testing [159]. This method retains tumor tissue heterogeneity because it is not artificially cultured; tumors’ biological characteristics remain more complete, so these models can better simulate the reality of tumor patients [160,161]. Several genomic databases provide abundant resources for PDX gene characterization (Table 4).
As PDXs have been increasingly used in precision oncology for lung cancer patients, it is crucial to standardize the development and genomic analysis tool to characterize PDX tumors and compare to donor patient tumors. Meehan TF et al. summarized a criterion to evaluate PDXs [165]. A PDX has three main applications. The first is basic research. Some scholars have studied the gene expression differences between successful and unsuccessful tumor tissue modeling and found that there are 163 abnormally expressed genes, and these genes are mainly concentrated in signal pathways such as cell cycle/mitosis and cell proliferation [166]. Several genomic data analysis workflows and guidelines have been reported as well. Genomic data analysis can help researchers discover key oncogenic mutation and new tumor biomarkers [167]. The second application is preclinical. Compared with the CDX model, the clinical relevance of the PDX model is up to 89–90% as PDXs can better simulate human responses to drugs and indicate a more reliable way to assess preclinical drug analysis. Furthermore, PDX models can evaluate molecular targeted therapy [168], chemotherapy [169,170], and ADCs [171]. Using PDXs, researchers found that EGFR-activating mutation PDX models were very sensitive to gefitinib, while KRAS mutation PDX models were not [172]. Our team recently utilized several PDX models to examine the effects of combined TKI and statins in TKI-resistant patients. Simvastatin demonstrated a powerful antitumor effect in tested LUAD cell lines and PDX tumors, irrespective of tumor genotypes [173]. Nitin Roper et al. found a osimertinib and savolitinib combination works best for osimertinib-resistant EGFR-mutant tumors with MET pathway activation in PDX models [174]. The third application is in the clinical phase, where cancer patients often develop drug resistance, leading to treatment failure. Developing an efficient method to test drug efficacy in a single dosage or combination is urgent. Since the PDX can largely retain the tumor biological characteristics in patients and is a reliable model when it comes to testing drug efficacy, it could be used to guide clinical treatment regimes. For example, a study reported that the PDX models of subrenal capsule xenografts showed consistent responses to the chemotherapy when correlating it with their clinical data [170]. Compared with other existing models, PDXs could preserve TME to a great extent; however, compared with the high success rate of CDX, the PDX success rate is significantly different in different tumors. The highest success rate when establishing a PDX is of acute lymphocytic leukemia, which is up to 64%. In contrast, the success rate of lung cancer PDX is lower [175]. In addition, the establishment of the PDX model is also relatively long. The first generation of tumors grown from tumor tissues taken from patients (P0 generation) and implanted in immunodeficient mice takes about 1–8 months to establish [176].
Many factors affect the success of a PDX. The most important factors include the quality and viability of tumor tissue [177]. Others include the degree of immune deficiency in transplanted mice, the interval between patients’ adjuvant chemotherapy, the implantation time of tissues, the method of tumor extraction, and the stage and type of tumor. As such, it was reported that the success rate of the PDX model of lung squamous cell carcinoma tissue is higher than that of lung adenocarcinoma [178]. The malignancy of the tumor tissue has a more significant effect on the modeling success rate. The higher the degree of malignancy, the higher the modeling success rate [179]. It is important to note that the method used to establish patient-derived xenografts (PDXs) from tumor tissue sections can impact the success rate of engraftment. Specifically, the tumor can be sectioned into smaller fragments, enzymatically digested, or physically manipulated into a single-cell suspension, each approach offering its own benefits and drawbacks. One advantage of using tumor fragments over a single-cell suspension is that fragments can retain cell–cell interactions and some tissue architecture of the original tumor, thereby better mimicking the TME. In contrast, the advantage of a single-cell suspension is that it allows scientists to collect an unbiased sample of the entire tumor rather than inadvertently selecting spatially enriched subclones during analysis or tumor passaging [180]. However, establishing a single-cell suspension requires tumors to be either enzymatically digested or mechanically chopped into fine pieces, which may cause the cells to undergo anoikis, thus affecting their viability and the success of engraftment. Another potential disadvantage of PDX models is that during tumor growth in the animal, mouse-derived stroma gradually replaces the human stroma after three to five passages, leading to a loss of the model’s original biological characteristics [181]. Additionally, PDX models require immunodeficient mice, which limits studies that could benefit from evaluating the effectiveness of immunotherapeutic agents.

3.6. Humanized Mouse Models

As immunotherapy becomes an increasingly important treatment paradigm, the clinical response to these therapies remains highly variable. We still lack a full understanding of their mechanisms of action and specific biomarkers of response. Consequently, there is an unmet need for in vivo models that can replicate the interactions between the human immune system and tumors. One of the major limitations of using the NSG mouse strain for PDX models is the absence of a competent immune system, meaning they may not accurately reflect disease progression or therapeutic responses that would occur in immune-competent humans. Over the past decade, significant progress has been made in developing preclinical models for evaluating cancer immunotherapies. Recently developed humanized mouse models provide a unique tool for assessing the antitumor response of the human immune system to checkpoint inhibitors [182]. Currently, there are two primary approaches for establishing humanized mouse models, both of which require sublethal irradiation before the transplantation of immune cells [182,183] (Table 5).
One of these methods uses human PBMCs (Hu-PBMCs) or tumor-infiltrating lymphocytes, while the other method requires human hematopoietic stem and progenitor cells (Hu-HPSCs) [184]. For the Hu-PBMC-derived model, first, one has to isolate PBMCs, which is usually performed using Ficoll—which is a hydrophilic polysaccharide that allows the plasma and lymphocytes, erythrocytes, and PBMCs to separate based on their densities following centrifugation. The isolated PBMCs are then transplanted into the previously irradiated mice via intravenous administration. The most significant advantage of this model is that it is relatively easy and takes about 4 weeks to establish. Studying T cell function in this model is of benefit as human CD3+ cells, including both CD4+ and CD8+ subsets, are the most abundant cell population that survives the engraftment process; however, FOXP3+CD25+CD127low regulatory T cells (Treg) are only detectable for the first 2–4 weeks post-injection before they become undetectable [185]. Human innate cell populations (such as myeloid and NK cells) survive for the first few days in the animals and become undetectable both in the circulation and tissues. Interestingly, CD19+ B cells are maintained at low levels in specific sites such as the sleep and bone marrow for several weeks. Moreover, human IgGs can be detected in the peripheral blood of these animals for their whole lifespan [186]. However, the drawback of this Hu-PBMC-derived model is that immune therapy studies can only be performed in a relatively short period due to the graft-versus-host disease that happens faster in this model than in the other models. Lin et al. established this type of mouse model in their study to determine the efficacy of PD-1/PD-L1 immunotherapies and have showed this PBMC-derived PDX model was an invaluable tool for their study [181]. They also established Hu-HPSC models by first reconstituting the human immune system by transferring human CD34+ hematopoietic stem and progenitor cells into the mice. Once established, they then transplanted human lung cancer cells [182]. While the establishment of the Hu-HPSC takes a longer time, it can also be used for long-term immune therapy surveillance studies. The advantage of this mouse model is that they have a more complete hematopoietic system that includes innate immune cells, adaptive immune cells, and even low numbers of red blood cells and platelets [187], and GVHD is uncommon. CD34+ HSPCs are most reliably obtained from umbilical cord blood (UCB) or peripheral cells mobilized in response to G-CSF (CSF3), fetal liver tissue, and bone marrow [188]. It is important to note that many parameters can affect the engraftment of CD34+ HSPCs into immunodeficient mice, for example, the genetic background of the strain, the age of the animal, the source of CD34+ HSPCs, the route of injection and the number of CD34+ HSPCs, and the regimen used [189,190,191,192,193]. In addition, studies indicate that the source of CD34+ HSPCs influences the functionality of human T cells developed in engrafted mice, for example, fetal CD34+ HSPCs give rise to T cells with greater immune tolerance versus those that received adult CD34+ HSPCs [194]. Several other limitations are important to consider. These include the incomplete development of mature human innate cell lineages (monocytes, macrophages, DCs, and NK cells), incomplete functionality of the human B cells, and the absence of human leukocyte antigen (HLA) expression essential for developing HLA-restricted T cells. Several efforts to improve humanized mouse models have focused on incorporating various primitive immune cells to better evaluate immune response dynamics, understand mechanisms of immune evasion, and assess both the efficacy and potential adverse effects of immunotherapies [195,196]. However, these models are limited by the availability of donor resources.

4. Conclusions

Despite substantial progress in developing various preclinical models for functional precision research, each model discussed herein has its own limitations. Figure 3 summarizes the clinical applications for key in vitro and in vivo lung cancer models described in this review. Traditional models often fail to capture the full spectrum of human immune responses, which can limit the predictability of clinical outcomes. The improvement of preclinical models that accurately translate human immunity remains a top priority in cancer research [197]. New models, such as humanized PDXs with reconstituted immune systems and LCO–immune cell co-cultures, have shown promise in bridging this gap. By better replicating human immunity, these models are crucial for the development of precision immunotherapies tailored to individual lung cancer patients, ultimately improving therapeutic efficacy and reducing adverse effects.

Author Contributions

Conceptualization, J.-Z.Y. and T.L.; methodology, J.-Z.Y. and T.L.; data curation, J.-Z.Y., Z.K. and T.L.; writing—original draft preparation, J.-Z.Y., Z.K., W.M. and T.L.; writing—review and editing Z.K., J.-Z.Y., W.M., R.L. and T.L.; visualization, J.-Z.Y. and T.L.; supervision, T.L. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge funding support in part from Merit Award (I01BX003895) and Lung Precision Oncology Program (L0014) from the Office of Research & Development (ORD) of the Department of Veterans Affairs (VA), University of California Lung Cancer Consortium (UCLCC), and Personalized Cancer Therapy Gift Fund (to T.L.).

Conflicts of Interest

T.L. receives research grants to conduct clinical trials as principal investigator at local institute from AbbVie, Amgen, Astellas, AstraZeneca, Bristol Myers Squibb (BMS), Chugai Pharma, Duality Biologics, Genentech/LaRoche, Jounce Therapeutics, LabyRx Immuno-Oncology, Merck, OncoC4/BioNTech, Novartis, RasCal Therapeutics, Tempus, Xilio Therapeutics. T.L. served in the steering committee for BMS.

Abbreviations

ALKanaplastic lymphoma kinase
BaPpolyaromatic hydrocarbons as benzo(a)pyrene
BRAFv-raf murine sarcoma viral oncogene homolog B
CDXcell line-derived xenograft
CRconditional reprogramming
CRCsconditional reprogramming cultures
CR-PDXconditionally reprogrammed cell lines
CTLA-4cytotoxic T-lymphocyte-associated protein 4
EGFRepidermal growth factor receptor
ErbB2erb-b2 receptor tyrosine kinase 2
FEN1flap endonuclease 1
GEMgenetically engineered model
GEMMsgenetically engineered mouse models
GLI1glioma-associated oncogene homolog 1
GVHDgraft-versus-host disease
GM-CSFgranulocyte–macrophage colony-stimulating factor
HER2human epidermal growth factor 2
HLAhuman leukocyte antigen
HPSChematopoietic stem and progenitor cell
Hu-HPSCshuman pluripotent stem cells
Hu-PBMCshuman peripheral blood mononuclear cells
ICIsimmune checkpoint inhibitors
IFinterfemoral
IPintraperitoneal
iPSCinduced pluripotent stem cells
IVintravenous
KRASKirsten rat sarcoma
LCOslung cancer organoids
LUADlung adenocarcinoma
LUSClung squamous cell carcinoma
MCAdiethylnitrosamine and 2-methycholanthrene
MDSCsmyeloid-derived suppressor cells
METmesenchymal epithelial transition
NKnatural killer
NNK4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone
NRASneuroblastoma RAS viral
NSCLCnon-small cell lung cancer
NSGnod-scid il2rgammanull
NTCUn-nitroso-tris-chloroethylurea
PDpharmacodynamic
PD-1programmed cell death protein 1
PDOspatient-derived organoids
PDXspatient-derived xenografts
PI3Kphosphatidylinositol 3-kinase
PKpharmacokinetic
PMBCperipheral blood mononuclear cell
RETrearranged during transfection
ROCKrho kinase inhibitor
ROS1c-ros oncogene 1
SCLCsmall cell lung cancer
scRNAsingle-cell RNA
TKIstyrosine kinase inhibitors
TMEtumor microenvironment
Tregsregulatory T cells
UCBumbilical cord blood

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Li, S.; de Camargo Correia, G.S.; Wang, J.; Manochakian, R.; Zhao, Y.; Lou, Y. Emerging Targeted Therapies in Advanced Non-Small-Cell Lung Cancer. Cancers 2023, 15, 2899. [Google Scholar] [CrossRef]
  3. Reyes, A.; Pharaon, R.; Mohanty, A.; Massarelli, E. Arising Novel Agents in Lung Cancer: Are Bispecifics and ADCs the New Paradigm? Cancers 2023, 15, 3162. [Google Scholar] [CrossRef]
  4. Patel, S.R.; Das, M. Small Cell Lung Cancer: Emerging Targets and Strategies for Precision Therapy. Cancers 2023, 15, 4016. [Google Scholar] [CrossRef] [PubMed]
  5. Mulvey, C.K. Emerging Precision Medicine Approaches for Lung Neuroendocrine Tumors. Cancers 2023, 15, 5575. [Google Scholar] [CrossRef]
  6. Yun, K.M.; Bazhenova, L. Emerging New Targets in Systemic Therapy for Malignant Pleural Mesothelioma. Cancers 2024, 16, 1252. [Google Scholar] [CrossRef]
  7. Li, T.; Ma, W.; Al-Obeidi, E. Evolving Precision First-Line Systemic Treatment for Patients with Unresectable Non-Small Cell Lung Cancer. Cancers 2024, 16, 2350. [Google Scholar] [CrossRef]
  8. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
  9. Lindeman, N.I.; Cagle, P.T.; Aisner, D.L.; Arcila, M.E.; Beasley, M.B.; Bernicker, E.H.; Colasacco, C.; Dacic, S.; Hirsch, F.R.; Kerr, K.; et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment with Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J. Mol. Diagn. 2018, 20, 129–159. [Google Scholar]
  10. Xie, T.; Qiu, B.M.; Luo, J.; Diao, Y.F.; Hu, L.W.; Liu, X.L.; Shen, Y. Distant metastasis patterns among lung cancer subtypes and impact of primary tumor resection on survival in metastatic lung cancer using SEER database. Sci. Rep. 2024, 14, 22445. [Google Scholar] [CrossRef]
  11. Chen, Z.; Fillmore, C.M.; Hammerman, P.S.; Kim, C.F.; Wong, K.K. Non-small-cell lung cancers: A heterogeneous set of diseases. Nat. Rev. Cancer 2014, 14, 535–546. [Google Scholar] [CrossRef] [PubMed]
  12. Ferone, G.; Lee, M.C.; Sage, J.; Berns, A. Cells of origin of lung cancers: Lessons from mouse studies. Genes Dev. 2020, 34, 1017–1032. [Google Scholar] [CrossRef] [PubMed]
  13. Tata, P.R.; Rajagopal, J. Plasticity in the lung: Making and breaking cell identity. Development 2017, 144, 755–766. [Google Scholar] [CrossRef]
  14. Collins, F.S.; Varmus, H. A new initiative on precision medicine. N. Engl. J. Med. 2015, 372, 793–795. [Google Scholar] [CrossRef] [PubMed]
  15. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  16. Gandhi, J.; Zhang, J.; Xie, Y.; Soh, J.; Shigematsu, H.; Zhang, W.; Yamamoto, H.; Peyton, M.; Girard, L.; Lockwood, W.W.; et al. Alterations in genes of the EGFR signaling pathway and their relationship to EGFR tyrosine kinase inhibitor sensitivity in lung cancer cell lines. PLoS ONE 2009, 4, e4576. [Google Scholar] [CrossRef] [PubMed]
  17. Singh, A.; Greninger, P.; Rhodes, D.; Koopman, L.; Violette, S.; Bardeesy, N.; Settleman, J. A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell 2009, 15, 489–500. [Google Scholar] [CrossRef]
  18. Soh, J.; Okumura, N.; Lockwood, W.W.; Yamamoto, H.; Shigematsu, H.; Zhang, W.; Chari, R.; Shames, D.S.; Tang, X.; MacAulay, C.; et al. Oncogene mutations, copy number gains and mutant allele specific imbalance (MASI) frequently occur together in tumor cells. PLoS ONE 2009, 4, e7464. [Google Scholar] [CrossRef] [PubMed]
  19. Harbour, J.W.; Lai, S.L.; Whang-Peng, J.; Gazdar, A.F.; Minna, J.D.; Kaye, F.J. Abnormalities in structure and expression of the human retinoblastoma gene in SCLC. Science 1988, 241, 353–357. [Google Scholar] [CrossRef]
  20. Otterson, G.A.; Kratzke, R.A.; Coxon, A.; Kim, Y.W.; Kaye, F.J. Absence of p16INK4 protein is restricted to the subset of lung cancer lines that retains wildtype RB. Oncogene 1994, 9, 3375–3378. [Google Scholar]
  21. FJ, K. B and cyclin dependent kinase pathways: Defining a distinction between RB and p16 loss in lung cancer. Oncogene 2002, 2145, 6908–6914. [Google Scholar]
  22. Rudin, C.M.; Poirier, J.T.; Byers, L.A.; Dive, C.; Dowlati, A.; George, J.; Heymach, J.V.; Johnson, J.E.; Lehman, J.M.; MacPherson, D.; et al. Author Correction: Molecular subtypes of small cell lung cancer: A synthesis of human and mouse model data. Nat. Rev. Cancer 2019, 19, 415. [Google Scholar] [CrossRef] [PubMed]
  23. Saunders, L.R.; Bankovich, A.J.; Anderson, W.C.; Aujay, M.A.; Bheddah, S.; Black, K.; Desai, R.; Escarpe, P.A.; Hampl, J.; Laysang, A.; et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl. Med. 2015, 7, 302ra136. [Google Scholar] [CrossRef]
  24. Mok, T.S.; Wu, Y.L.; Thongprasert, S.; Yang, C.H.; Chu, D.T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [Google Scholar] [CrossRef] [PubMed]
  25. Druker, B.J.; Guilhot, F.; O’Brien, S.G.; Gathmann, I.; Kantarjian, H.; Gattermann, N.; Deininger, M.W.; Silver, R.T.; Goldman, J.M.; Stone, R.M.; et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N. Engl. J. Med. 2006, 355, 2408–2417. [Google Scholar] [CrossRef]
  26. Shepherd, F.A.; Rodrigues Pereira, J.; Ciuleanu, T.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 2005, 353, 123–132. [Google Scholar] [CrossRef] [PubMed]
  27. Demetri, G.D.; von Mehren, M.; Blanke, C.D.; Van den Abbeele, A.D.; Eisenberg, B.; Roberts, P.J.; Heinrich, M.C.; Tuveson, D.A.; Singer, S.; Janicek, M.; et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Engl. J. Med. 2002, 347, 472–480. [Google Scholar] [CrossRef] [PubMed]
  28. Coe, B.P.; Lockwood, W.W.; Girard, L.; Chari, R.; Macaulay, C.; Lam, S.; Gazdar, A.F.; Minna, J.D.; Lam, W.L. Differential disruption of cell cycle pathways in small cell and non-small cell lung cancer. Br. J. Cancer 2006, 94, 1927–1935. [Google Scholar] [CrossRef] [PubMed]
  29. Lockwood, W.W.; Chari, R.; Coe, B.P.; Girard, L.; Macaulay, C.; Lam, S.; Gazdar, A.F.; Minna, J.D.; Lam, W.L. DNA amplification is a ubiquitous mechanism of oncogene activation in lung and other cancers. Oncogene 2008, 27, 4615–4624. [Google Scholar] [CrossRef] [PubMed]
  30. Kwei, K.A.; Kim, Y.H.; Girard, L.; Kao, J.; Pacyna-Gengelbach, M.; Salari, K.; Lee, J.; Choi, Y.L.; Sato, M.; Wang, P.; et al. Genomic profiling identifies TITF1 as a lineage-specific oncogene amplified in lung cancer. Oncogene 2008, 27, 3635–3640. [Google Scholar] [CrossRef] [PubMed]
  31. Tanaka, H.; Yanagisawa, K.; Shinjo, K.; Taguchi, A.; Maeno, K.; Tomida, S.; Shimada, Y.; Osada, H.; Kosaka, T.; Matsubara, H.; et al. Lineage-specific dependency of lung adenocarcinomas on the lung development regulator TTF-1. Cancer Res. 2007, 67, 6007–6011. [Google Scholar] [CrossRef] [PubMed]
  32. Paez, J.G.; Janne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef]
  33. Engelman, J.A.; Janne, P.A. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin. Cancer Res. 2008, 14, 2895–2899. [Google Scholar] [CrossRef] [PubMed]
  34. Gazdar, A.F. Activating and resistance mutations of EGFR in non-small-cell lung cancer: Role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 2009, 28 (Suppl. S1), S24–S31. [Google Scholar] [CrossRef]
  35. Geraghty, R.J.; Capes-Davis, A.; Davis, J.M.; Downward, J.; Freshney, R.I.; Knezevic, I.; Lovell-Badge, R.; Masters, J.R.; Meredith, J.; Stacey, G.N.; et al. Guidelines for the use of cell lines in biomedical research. Br. J. Cancer 2014, 111, 1021–1046. [Google Scholar] [CrossRef] [PubMed]
  36. Wilding, J.L.; Bodmer, W.F. Cancer cell lines for drug discovery and development. Cancer Res. 2014, 74, 2377–2384. [Google Scholar] [CrossRef] [PubMed]
  37. Gazdar, A.F.; Hirsch, F.R.; Minna, J.D. Correction: “From Mice to Men and Back: An Assessment of Preclinical Model Systems for the Study of Lung Cancers”. J. Thorac. Oncol. 2016, 11, e88–e89. [Google Scholar] [CrossRef] [PubMed]
  38. Gazdar, A.F.; Girard, L.; Lockwood, W.W.; Lam, W.L.; Minna, J.D. Lung cancer cell lines as tools for biomedical discovery and research. J. Natl. Cancer Inst. 2016, 102, 1310–1320. [Google Scholar] [CrossRef] [PubMed]
  39. Cross, D.A.; Ashton, S.E.; Ghiorghiu, S.; Eberlein, C.; Nebhan, C.A.; Spitzler, P.J.; Orme, J.P.; Finlay, M.R.; Ward, R.A.; Mellor, M.J.; et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014, 4, 1046–1061. [Google Scholar] [CrossRef] [PubMed]
  40. Wistuba, I.I.; Bryant, D.; Behrens, C.; Milchgrub, S.; Virmani, A.K.; Ashfaq, R.; Minna, J.D.; Gazdar, A.F. Comparison of features of human lung cancer cell lines and their corresponding tumors. Clin. Cancer Res. 1999, 5, 991–1000. [Google Scholar]
  41. Li, T.; Ling, Y.H.; Goldman, I.D.; Perez-Soler, R. Schedule-dependent cytotoxic synergism of pemetrexed and erlotinib in human non-small cell lung cancer cells. Clin. Cancer Res. 2007, 13, 3413–3422. [Google Scholar] [CrossRef]
  42. Osude, C.; Lin, L.; Patel, M.; Eckburg, A.; Berei, J.; Kuckovic, A.; Dube, N.; Rastogi, A.; Gautam, S.; Smith, T.J.; et al. Mediating EGFR-TKI Resistance by VEGF/VEGFR Autocrine Pathway in Non-Small Cell Lung Cancer. Cells 2022, 11, 1694. [Google Scholar] [CrossRef]
  43. Cragg, M.S.; Kuroda, J.; Puthalakath, H.; Huang, D.C.; Strasser, A. Gefitinib-induced killing of NSCLC cell lines expressing mutant EGFR requires BIM and can be enhanced by BH3 mimetics. PLoS Med. 2007, 4, 1681–1689; discussion 1690. [Google Scholar] [CrossRef]
  44. Chava, S.; Bugide, S.; Zhang, X.; Gupta, R.; Wajapeyee, N. Betacellulin promotes tumor development and EGFR mutant lung cancer growth by stimulating the EGFR pathway and suppressing apoptosis. iScience 2022, 25, 104211. [Google Scholar] [CrossRef]
  45. Forcella, M.; Oldani, M.; Epistolio, S.; Freguia, S.; Monti, E.; Fusi, P.; Frattini, M. Non-small cell lung cancer (NSCLC), EGFR downstream pathway activation and TKI targeted therapies sensitivity: Effect of the plasma membrane-associated NEU3. PLoS ONE 2017, 12, e0187289. [Google Scholar] [CrossRef]
  46. Wu, L.; Yu, Y.; Xu, L.; Wang, X.; Zhou, J.; Wang, Y. TROY Modulates Cancer Stem-Like Cell Properties and Gefitinib Resistance Through EMT Signaling in Non-Small Cell Lung Cancer. Front. Genet. 2022, 13, 881875. [Google Scholar] [CrossRef] [PubMed]
  47. Song, Y.A.; Ma, T.; Zhang, X.Y.; Cheng, X.S.; Olajuyin, A.M.; Sun, Z.F.; Zhang, X.J. Apatinib preferentially inhibits PC9 gefitinib-resistant cancer cells by inducing cell cycle arrest and inhibiting VEGFR signaling pathway. Cancer Cell Int. 2019, 19, 117. [Google Scholar] [CrossRef] [PubMed]
  48. Guerra, S.L.; Maertens, O.; Kuzmickas, R.; De Raedt, T.; Adeyemi, R.O.; Guild, C.J.; Guillemette, S.; Redig, A.J.; Chambers, E.S.; Xu, M.; et al. A Deregulated HOX Gene Axis Confers an Epigenetic Vulnerability in KRAS-Mutant Lung Cancers. Cancer Cell 2020, 37, 705–719.e6. [Google Scholar] [CrossRef] [PubMed]
  49. Sunaga, N.; Shames, D.S.; Girard, L.; Peyton, M.; Larsen, J.E.; Imai, H.; Soh, J.; Sato, M.; Yanagitani, N.; Kaira, K.; et al. Knockdown of oncogenic KRAS in non-small cell lung cancers suppresses tumor growth and sensitizes tumor cells to targeted therapy. Mol. Cancer Ther. 2011, 10, 336–346. [Google Scholar] [CrossRef]
  50. Blanco, R.; Iwakawa, R.; Tang, M.; Kohno, T.; Angulo, B.; Pio, R.; Montuenga, L.M.; Minna, J.D.; Yokota, J.; Sanchez-Cespedes, M. A gene-alteration profile of human lung cancer cell lines. Hum. Mutat. 2009, 30, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
  51. Fawdar, S.; Trotter, E.W.; Li, Y.; Stephenson, N.L.; Hanke, F.; Marusiak, A.A.; Edwards, Z.C.; Ientile, S.; Waszkowycz, B.; Miller, C.J.; et al. Targeted genetic dependency screen facilitates identification of actionable mutations in FGFR4, MAP3K9, and PAK5 in lung cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 12426–12431. [Google Scholar] [CrossRef]
  52. Song, Z.; Liu, F.; Zhang, J. Targeting NRAS(Q61K) mutant delays tumor growth and angiogenesis in non-small cell lung cancer. Am. J. Cancer Res. 2017, 7, 831–844. [Google Scholar]
  53. Lee, G.D.; Lee, S.E.; Oh, D.Y.; Yu, D.B.; Jeong, H.M.; Kim, J.; Hong, S.; Jung, H.S.; Oh, E.; Song, J.Y.; et al. MET Exon 14 Skipping Mutations in Lung Adenocarcinoma: Clinicopathologic Implications and Prognostic Values. J. Thorac. Oncol. 2017, 12, 1233–1246. [Google Scholar] [CrossRef]
  54. Koivunen, J.P.; Mermel, C.; Zejnullahu, K.; Murphy, C.; Lifshits, E.; Holmes, A.J.; Choi, H.G.; Kim, J.; Chiang, D.; Thomas, R.; et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin. Cancer Res. 2008, 14, 4275–4283. [Google Scholar] [CrossRef] [PubMed]
  55. Giannopoulou, E.; Nikolakopoulos, A.; Kotsirilou, D.; Lampropoulou, A.; Raftopoulou, S.; Papadimitriou, E.; Theocharis, A.D.; Makatsoris, T.; Fasseas, K.; Kalofonos, H.P. Epidermal growth factor receptor status and Notch inhibition in non-small cell lung cancer cells. J. Biomed. Sci. 2015, 22, 98. [Google Scholar] [CrossRef] [PubMed]
  56. Bamford, S.; Dawson, E.; Forbes, S.; Clements, J.; Pettett, R.; Dogan, A.; Flanagan, A.; Teague, J.; Futreal, P.A.; Stratton, M.R.; et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 2004, 91, 355–358. [Google Scholar] [CrossRef]
  57. Palanikumar, L.; Karpauskaite, L.; Al-Sayegh, M.; Chehade, I.; Alam, M.; Hassan, S.; Maity, D.; Ali, L.; Kalmouni, M.; Hunashal, Y.; et al. Protein mimetic amyloid inhibitor potently abrogates cancer-associated mutant p53 aggregation and restores tumor suppressor function. Nat. Commun. 2021, 12, 3962. [Google Scholar] [CrossRef] [PubMed]
  58. Ogoshi, Y.; Shien, K.; Yoshioka, T.; Torigoe, H.; Sato, H.; Sakaguchi, M.; Tomida, S.; Namba, K.; Kurihara, E.; Takahashi, Y.; et al. Anti-tumor effect of neratinib against lung cancer cells harboring HER2 oncogene alterations. Oncol. Lett. 2019, 17, 2729–2736. [Google Scholar] [CrossRef] [PubMed]
  59. Cole, S.P.; Pinkoski, M.J.; Bhardwaj, G.; Deeley, R.G. Elevated expression of annexin II (lipocortin II, p36) in a multidrug resistant small cell lung cancer cell line. Br. J. Cancer 1992, 65, 498–502. [Google Scholar] [CrossRef]
  60. Pettengill, O.S.; Sorenson, G.D.; Wurster-Hill, D.H.; Curphey, T.J.; Noll, W.W.; Cate, C.C.; Maurer, L.H. Isolation and growth characteristics of continuous cell lines from small-cell carcinoma of the lung. Cancer 1980, 45, 906–918. [Google Scholar] [CrossRef]
  61. Dutil, J.; Chen, Z.; Monteiro, A.N.; Teer, J.K.; Eschrich, S.A. An Interactive Resource to Probe Genetic Diversity and Estimated Ancestry in Cancer Cell Lines. Cancer Res. 2019, 79, 1263–1273. [Google Scholar] [CrossRef]
  62. Twentyman, P.R.; Wright, K.A.; Mistry, P.; Kelland, L.R.; Murrer, B.A. Sensitivity to novel platinum compounds of panels of human lung cancer cell lines with acquired and inherent resistance to cisplatin. Cancer Res. 1992, 52, 5674–5680. [Google Scholar] [PubMed]
  63. Fisher, E.R.; Paulson, J.D. A new in vitro cell line established from human large cell variant of oat cell lung cancer. Cancer Res. 1978, 38, 3830–3835. [Google Scholar]
  64. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  65. Huo, K.G.; D’Arcangelo, E.; Tsao, M.S. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Transl. Lung Cancer Res. 2020, 9, 2214–2232. [Google Scholar] [CrossRef] [PubMed]
  66. Saforo, D.; Omer, L.; Smolenkov, A.; Barve, A.; Casson, L.; Boyd, N.; Clark, G.; Siskind, L.; Beverly, L. Primary lung cancer samples cultured under microenvironment-mimetic conditions enrich for mesenchymal stem-like cells that promote metastasis. Sci. Rep. 2019, 9, 4177. [Google Scholar] [CrossRef] [PubMed]
  67. Kodack, D.P.; Farago, A.F.; Dastur, A.; Held, M.A.; Dardaei, L.; Friboulet, L.; von Flotow, F.; Damon, L.J.; Lee, D.; Parks, M.; et al. Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care. Cell Rep. 2017, 21, 3298–3309. [Google Scholar] [CrossRef] [PubMed]
  68. Tiran, V.; Lindenmann, J.; Brcic, L.; Heitzer, E.; Stanzer, S.; Tabrizi-Wizsy, N.G.; Stacher, E.; Stoeger, H.; Popper, H.H.; Balic, M.; et al. Primary patient-derived lung adenocarcinoma cell culture challenges the association of cancer stem cells with epithelial-to-mesenchymal transition. Sci. Rep. 2017, 7, 10040. [Google Scholar] [CrossRef] [PubMed]
  69. Odintsov, I.; Mattar, M.S.; Lui, A.J.; Offin, M.; Kurzatkowski, C.; Delasos, L.; Khodos, I.; Asher, M.; Daly, R.M.; Rekhtman, N.; et al. Novel Preclinical Patient-Derived Lung Cancer Models Reveal Inhibition of HER3 and MTOR Signaling as Therapeutic Strategies for NRG1 Fusion-Positive Cancers. J. Thorac. Oncol. 2021, 16, 1149–1165. [Google Scholar] [CrossRef] [PubMed]
  70. Crystal, A.S.; Shaw, A.T.; Sequist, L.V.; Friboulet, L.; Niederst, M.J.; Lockerman, E.L.; Frias, R.L.; Gainor, J.F.; Amzallag, A.; Greninger, P.; et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science 2014, 346, 1480–1486. [Google Scholar] [CrossRef] [PubMed]
  71. Liu, X.; Ory, V.; Chapman, S.; Yuan, H.; Albanese, C.; Kallakury, B.; Timofeeva, O.A.; Nealon, C.; Dakic, A.; Simic, V.; et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am. J. Pathol. 2012, 180, 599–607. [Google Scholar] [CrossRef]
  72. Liu, X.; Krawczyk, E.; Suprynowicz, F.A.; Palechor-Ceron, N.; Yuan, H.; Dakic, A.; Simic, V.; Zheng, Y.L.; Sripadhan, P.; Chen, C.; et al. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat. Protoc. 2017, 12, 439–451. [Google Scholar] [CrossRef]
  73. Talwelkar, S.S.; Nagaraj, A.S.; Devlin, J.R.; Hemmes, A.; Potdar, S.; Kiss, E.A.; Saharinen, P.; Salmenkivi, K.; Mayranpaa, M.I.; Wennerberg, K.; et al. Receptor Tyrosine Kinase Signaling Networks Define Sensitivity to ERBB Inhibition and Stratify Kras-Mutant Lung Cancers. Mol. Cancer Ther. 2019, 18, 1863–1874. [Google Scholar] [CrossRef]
  74. Borodovsky, A.; McQuiston, T.J.; Stetson, D.; Ahmed, A.; Whitston, D.; Zhang, J.; Grondine, M.; Lawson, D.; Challberg, S.S.; Zinda, M.; et al. Generation of stable PDX derived cell lines using conditional reprogramming. Mol. Cancer 2017, 16, 177. [Google Scholar] [CrossRef]
  75. Yuan, H.; Myers, S.; Wang, J.; Zhou, D.; Woo, J.A.; Kallakury, B.; Ju, A.; Bazylewicz, M.; Carter, Y.M.; Albanese, C.; et al. Use of reprogrammed cells to identify therapy for respiratory papillomatosis. N. Engl. J. Med. 2012, 367, 1220–1227. [Google Scholar] [CrossRef] [PubMed]
  76. Alamri, A.M.; Liu, X.; Blancato, J.K.; Haddad, B.R.; Wang, W.; Zhong, X.; Choudhary, S.; Krawczyk, E.; Kallakury, B.V.; Davidson, B.J.; et al. Expanding primary cells from mucoepidermoid and other salivary gland neoplasms for genetic and chemosensitivity testing. Dis. Model. Mech. 2018, 11, dmm031716. [Google Scholar] [CrossRef] [PubMed]
  77. Zhong, M.; Fu, L. Culture and application of conditionally reprogrammed primary tumor cells. Gastroenterol. Rep. 2020, 8, 224–233. [Google Scholar] [CrossRef]
  78. Jiang, S.; Wang, J.; Yang, C.; Tan, R.; Hou, J.; Shi, Y.; Zhang, H.; Ma, S.; Wang, J.; Zhang, M.; et al. Continuous culture of urine-derived bladder cancer cells for precision medicine. Protein Cell 2019, 10, 902–907. [Google Scholar] [CrossRef]
  79. Hirano, T.; Yasuda, H.; Tani, T.; Hamamoto, J.; Oashi, A.; Ishioka, K.; Arai, D.; Nukaga, S.; Miyawaki, M.; Kawada, I.; et al. In Vitro modeling to determine mutation specificity of EGFR tyrosine kinase inhibitors against clinically relevant EGFR mutants in non-small-cell lung cancer. Oncotarget 2015, 6, 38789–38803. [Google Scholar] [CrossRef]
  80. Eramo, A.; Lotti, F.; Sette, G.; Pilozzi, E.; Biffoni, M.; Di Virgilio, A.; Conticello, C.; Ruco, L.; Peschle, C.; De Maria, R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008, 15, 504–514. [Google Scholar] [CrossRef] [PubMed]
  81. Camp, J.G.; Sekine, K.; Gerber, T.; Loeffler-Wirth, H.; Binder, H.; Gac, M.; Kanton, S.; Kageyama, J.; Damm, G.; Seehofer, D.; et al. Multilineage communication regulates human liver bud development from pluripotency. Nature 2017, 546, 533–538. [Google Scholar] [CrossRef]
  82. Orienti, I.; Francescangeli, F.; de Angelis, M.L.; Fecchi, K.; Bongiorno-Borbone, L.; Signore, M.; Peschiaroli, A.; Boe, A.; Bruselles, A.; Costantino, A.; et al. A new bioavailable fenretinide formulation with antiproliferative, antimetabolic, and cytotoxic effects on solid tumors. Cell Death Dis. 2019, 10, 529. [Google Scholar] [CrossRef] [PubMed]
  83. Orienti, I.; Salvati, V.; Sette, G.; Zucchetti, M.; Bongiorno-Borbone, L.; Peschiaroli, A.; Zolla, L.; Francescangeli, F.; Ferrari, M.; Matteo, C.; et al. A novel oral micellar fenretinide formulation with enhanced bioavailability and antitumour activity against multiple tumours from cancer stem cells. J. Exp. Clin. Cancer Res. 2019, 38, 373. [Google Scholar] [CrossRef] [PubMed]
  84. Zeuner, A.; Francescangeli, F.; Contavalli, P.; Zapparelli, G.; Apuzzo, T.; Eramo, A.; Baiocchi, M.; De Angelis, M.L.; Biffoni, M.; Sette, G.; et al. Elimination of quiescent/slow-proliferating cancer stem cells by Bcl-XL inhibition in non-small cell lung cancer. Cell Death Differ. 2014, 21, 1877–1888. [Google Scholar] [CrossRef] [PubMed]
  85. Kamer, I.; Bab-Dinitz, E.; Zadok, O.; Ofek, E.; Gottfried, T.; Daniel-Meshulam, I.; Hout-Siloni, G.; Ben Nun, A.; Barshack, I.; Onn, A.; et al. Immunotherapy response modeling by ex-vivo organ culture for lung cancer. Cancer Immunol. Immunother. 2021, 70, 2223–2234. [Google Scholar] [CrossRef] [PubMed]
  86. Ivanova, E.; Kuraguchi, M.; Xu, M.; Portell, A.J.; Taus, L.; Diala, I.; Lalani, A.S.; Choi, J.; Chambers, E.S.; Li, S.; et al. Use of Ex Vivo Patient-Derived Tumor Organotypic Spheroids to Identify Combination Therapies for HER2 Mutant Non-Small Cell Lung Cancer. Clin. Cancer Res. 2020, 26, 2393–2403. [Google Scholar] [CrossRef]
  87. Endo, H.; Okami, J.; Okuyama, H.; Kumagai, T.; Uchida, J.; Kondo, J.; Takehara, T.; Nishizawa, Y.; Imamura, F.; Higashiyama, M.; et al. Spheroid culture of primary lung cancer cells with neuregulin 1/HER3 pathway activation. J. Thorac. Oncol. 2013, 8, 131–139. [Google Scholar] [CrossRef]
  88. Benali, R.; Tournier, J.M.; Chevillard, M.; Zahm, J.M.; Klossek, J.M.; Hinnrasky, J.; Gaillard, D.; Maquart, F.X.; Puchelle, E. Tubule formation by human surface respiratory epithelial cells cultured in a three-dimensional collagen lattice. Am. J. Physiol. 1993, 264, L183–L192. [Google Scholar] [CrossRef]
  89. Chen, Y.W.; Huang, S.X.; de Carvalho, A.; Ho, S.H.; Islam, M.N.; Volpi, S.; Notarangelo, L.D.; Ciancanelli, M.; Casanova, J.L.; Bhattacharya, J.; et al. A three-dimensional model of human lung development and disease from pluripotent stem cells. Nat. Cell Biol. 2017, 19, 542–549. [Google Scholar] [CrossRef]
  90. Dye, B.R.; Hill, D.R.; Ferguson, M.A.; Tsai, Y.H.; Nagy, M.S.; Dyal, R.; Wells, J.M.; Mayhew, C.N.; Nattiv, R.; Klein, O.D.; et al. In Vitro generation of human pluripotent stem cell derived lung organoids. eLife 2015, 4, e05098. [Google Scholar] [CrossRef]
  91. McCauley, K.B.; Hawkins, F.; Serra, M.; Thomas, D.C.; Jacob, A.; Kotton, D.N. Efficient Derivation of Functional Human Airway Epithelium from Pluripotent Stem Cells via Temporal Regulation of Wnt Signaling. Cell Stem Cell 2017, 20, 844–857.e6. [Google Scholar] [CrossRef] [PubMed]
  92. Tan, Q.; Choi, K.M.; Sicard, D.; Tschumperlin, D.J. Human airway organoid engineering as a step toward lung regeneration and disease modeling. Biomaterials 2017, 113, 118–132. [Google Scholar] [CrossRef]
  93. Nikolic, M.Z.; Caritg, O.; Jeng, Q.; Johnson, J.A.; Sun, D.; Howell, K.J.; Brady, J.L.; Laresgoiti, U.; Allen, G.; Butler, R.; et al. Human embryonic lung epithelial tips are multipotent progenitors that can be expanded in vitro as long-term self-renewing organoids. eLife 2017, 6, e26575. [Google Scholar] [CrossRef] [PubMed]
  94. Sachs, N.; Papaspyropoulos, A.; Zomer-van Ommen, D.D.; Heo, I.; Bottinger, L.; Klay, D.; Weeber, F.; Huelsz-Prince, G.; Iakobachvili, N.; Amatngalim, G.D.; et al. Long-term expanding human airway organoids for disease modeling. EMBO J. 2019, 38, e100300. [Google Scholar] [CrossRef] [PubMed]
  95. Hu, Y.; Sui, X.; Song, F.; Li, Y.; Li, K.; Chen, Z.; Yang, F.; Chen, X.; Zhang, Y.; Wang, X.; et al. Lung cancer organoids analyzed on microwell arrays predict drug responses of patients within a week. Nat. Commun. 2021, 12, 2581. [Google Scholar] [CrossRef]
  96. Dijkstra, K.K.; Monkhorst, K.; Schipper, L.J.; Hartemink, K.J.; Smit, E.F.; Kaing, S.; de Groot, R.; Wolkers, M.C.; Clevers, H.; Cuppen, E.; et al. Challenges in Establishing Pure Lung Cancer Organoids Limit Their Utility for Personalized Medicine. Util. Pers. Med. 2020, 31, 107588. [Google Scholar] [CrossRef]
  97. Li, Z.; Yu, L.; Chen, D.; Meng, Z.; Chen, W.; Huang, W. Protocol for generation of lung adenocarcinoma organoids from clinical samples. STAR Protoc. 2021, 2, 100239. [Google Scholar] [CrossRef] [PubMed]
  98. Finnberg, N.K.; Gokare, P.; Lev, A.; Grivennikov, S.I.; MacFarlane, A.W.t.; Campbell, K.S.; Winters, R.M.; Kaputa, K.; Farma, J.M.; Abbas, A.E.; et al. Application of 3D tumoroid systems to define immune and cytotoxic therapeutic responses based on tumoroid and tissue slice culture molecular signatures. Oncotarget 2017, 8, 66747–66757. [Google Scholar] [CrossRef] [PubMed]
  99. Kim, S.Y.; Kim, S.M.; Lim, S.; Lee, J.Y.; Choi, S.J.; Yang, S.D.; Yun, M.R.; Kim, C.G.; Gu, S.R.; Park, C.; et al. Modeling Clinical Responses to Targeted Therapies by Patient-Derived Organoids of Advanced Lung Adenocarcinoma. Clin. Cancer Res. 2021, 27, 4397–4409. [Google Scholar] [CrossRef] [PubMed]
  100. Yokota, E.; Iwai, M.; Yukawa, T.; Yoshida, M.; Naomoto, Y.; Haisa, M.; Monobe, Y.; Takigawa, N.; Guo, M.; Maeda, Y.; et al. Clinical application of a lung cancer organoid (tumoroid) culture system. NPJ Precis. Oncol. 2021, 5, 29. [Google Scholar] [CrossRef]
  101. Mazzocchi, A.; Dominijanni, A.; Soker, S. Pleural Effusion Aspirate for Use in 3D Lung Cancer Modeling and Chemotherapy Screening. Methods Mol. Biol. 2022, 2394, 471–483. [Google Scholar] [PubMed]
  102. Ma, X.; Yang, S.; Jiang, H.; Wang, Y.; Xiang, Z. Transcriptomic analysis of tumor tissues and organoids reveals the crucial genes regulating the proliferation of lung adenocarcinoma. J. Transl. Med. 2021, 19, 368. [Google Scholar] [CrossRef]
  103. Lo, Y.H.; Karlsson, K.; Kuo, C.J. Applications of Organoids for Cancer Biology and Precision Medicine. Nat. Cancer 2020, 1, 761–773. [Google Scholar] [CrossRef] [PubMed]
  104. De Angelis, M.L.; Francescangeli, F.; Nicolazzo, C.; Signore, M.; Giuliani, A.; Colace, L.; Boe, A.; Magri, V.; Baiocchi, M.; Ciardi, A.; et al. An organoid model of colorectal circulating tumor cells with stem cell features, hybrid EMT state and distinctive therapy response profile. J. Exp. Clin. Cancer Res. 2022, 41, 86. [Google Scholar] [CrossRef]
  105. Bolhaqueiro, A.C.F.; Ponsioen, B.; Bakker, B.; Klaasen, S.J.; Kucukkose, E.; van Jaarsveld, R.H.; Vivie, J.; Verlaan-Klink, I.; Hami, N.; Spierings, D.C.J.; et al. Ongoing chromosomal instability and karyotype evolution in human colorectal cancer organoids. Nat. Genet. 2019, 51, 824–834. [Google Scholar] [CrossRef]
  106. Drost, J.; van Boxtel, R.; Blokzijl, F.; Mizutani, T.; Sasaki, N.; Sasselli, V.; de Ligt, J.; Behjati, S.; Grolleman, J.E.; van Wezel, T.; et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science 2017, 358, 234–238. [Google Scholar] [CrossRef] [PubMed]
  107. Drost, J.; van Jaarsveld, R.H.; Ponsioen, B.; Zimberlin, C.; van Boxtel, R.; Buijs, A.; Sachs, N.; Overmeer, R.M.; Offerhaus, G.J.; Begthel, H.; et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 2015, 521, 43–47. [Google Scholar] [CrossRef]
  108. Li, X.; Nadauld, L.; Ootani, A.; Corney, D.C.; Pai, R.K.; Gevaert, O.; Cantrell, M.A.; Rack, P.G.; Neal, J.T.; Chan, C.W.; et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat. Med. 2014, 20, 769–777. [Google Scholar] [CrossRef] [PubMed]
  109. Shi, R.; Radulovich, N.; Ng, C.; Liu, N.; Notsuda, H.; Cabanero, M.; Martins-Filho, S.N.; Raghavan, V.; Li, Q.; Mer, A.S.; et al. Organoid Cultures as Preclinical Models of Non-Small Cell Lung Cancer. Clin. Cancer Res. 2021, 26, 1162–1174. [Google Scholar] [CrossRef]
  110. Dost, A.F.M.; Moye, A.L.; Vedaie, M.; Tran, L.M.; Fung, E.; Heinze, D.; Villacorta-Martin, C.; Huang, J.; Hekman, R.; Kwan, J.H.; et al. Organoids Model Transcriptional Hallmarks of Oncogenic KRAS Activation in Lung Epithelial Progenitor Cells. Cell Stem Cell 2020, 27, 663–678 e668. [Google Scholar] [CrossRef]
  111. Semba, T.; Sato, R.; Kasuga, A.; Suina, K.; Shibata, T.; Kohno, T.; Suzuki, M.; Saya, H.; Arima, Y. Lung Adenocarcinoma Mouse Models Based on Orthotopic Transplantation of Syngeneic Tumor-Initiating Cells Expressing EpCAM, SCA-1, and Ly6d. Cancers 2020, 12, 3805. [Google Scholar] [CrossRef] [PubMed]
  112. Na, F.; Pan, X.; Chen, J.; Chen, X.; Wang, M.; Chi, P.; You, L.; Zhang, L.; Zhong, A.; Zhao, L.; et al. KMT2C deficiency promotes small cell lung cancer metastasis through DNMT3A-mediated epigenetic reprogramming. Nat. Cancer 2023, 3, 753–767. [Google Scholar] [CrossRef] [PubMed]
  113. Nacarino-Palma, A.; Rejano-Gordillo, C.M.; Gonzalez-Rico, F.J.; Ordiales-Talavero, A.; Roman, A.C.; Cuadrado, M.; Bustelo, X.R.; Merino, J.M.; Fernandez-Salguero, P.M. Loss of Aryl Hydrocarbon Receptor Favors K-Ras(G12D)-Driven Non-Small Cell Lung Cancer. Cancers 2021, 13, 4071. [Google Scholar] [CrossRef]
  114. Wu, M.; Liao, Y.; Tang, L. Non-small cell lung cancer organoids: Advances and challenges in current applications. Chin. J. Cancer Res. 2024, 36, 455–473. [Google Scholar] [CrossRef]
  115. Takahashi, N.; Hoshi, H.; Higa, A.; Hiyama, G.; Tamura, H.; Ogawa, M.; Takagi, K.; Goda, K.; Okabe, N.; Muto, S.; et al. An In Vitro System for Evaluating Molecular Targeted Drugs Using Lung Patient-Derived Tumor Organoids. Cells 2019, 8, 481. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, J.H.; Chu, X.P.; Zhang, J.T.; Nie, Q.; Tang, W.F.; Su, J.; Yan, H.H.; Zheng, H.P.; Chen, Z.X.; Chen, X.; et al. Genomic characteristics and drug screening among organoids derived from non-small cell lung cancer patients. Thorat. Cancer 2020, 11, 2279–2290. [Google Scholar] [CrossRef] [PubMed]
  117. Ooft, S.N.; Weeber, F.; Dijkstra, K.K.; McLean, C.M.; Kaing, S.; van Werkhoven, E.; Schipper, L.; Hoes, L.; Vis, D.J.; van de Haar, J.; et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci. Transl. Med. 2019, 11, 2574. [Google Scholar] [CrossRef]
  118. Ooft, S.N.; Weeber, F.; Schipper, L.; Dijkstra, K.; McLean, C.; Kaing, S.; van de Haar, J.; Prevoo, W.; van Werkhoven, E.; Snaebjornsson, P.; et al. Prospective experimental treatment of colorectal cancer patients based on organoid drug responses. ESMO Open 2021, 6, 100103. [Google Scholar] [CrossRef]
  119. Li, Z.; Qian, Y.; Li, W.; Liu, L.; Yu, L.; Liu, X.; Wu, G.; Wang, Y.; Luo, W.; Fang, F.; et al. Human Lung Adenocarcinoma-Derived Organoid Models for Drug Screening. eScience 2020, 23, 101411. [Google Scholar] [CrossRef]
  120. Wang, H.M.; Zhang, C.Y.; Peng, K.C.; Chen, Z.X.; Su, J.W.; Li, Y.F.; Li, W.F.; Gao, Q.Y.; Zhang, S.L.; Chen, Y.Q.; et al. Using patient-derived organoids to predict locally advanced or metastatic lung cancer tumor response: A real-world study. Cell Rep. Med. 2023, 4, 100911. [Google Scholar] [CrossRef] [PubMed]
  121. Giraldo, N.A.; Taube, J.M. PD-L1 and Other Immunological Diagnosis Tools. In Oncoimmunology: A Practical Guide for Cancer Immunotherapy; Springer: Cham, Switzerland, 2018; pp. 371–385. [Google Scholar]
  122. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H.; et al. The clinical role of the TME in solid cancer. Br. J. Cancer 2019, 120, 45–53. [Google Scholar] [CrossRef] [PubMed]
  123. Toor, S.M.; Sasidharan Nair, V.; Decock, J.; Elkord, E. Immune checkpoints in the tumor microenvironment. Semin. Cancer Biol. 2020, 65, 1–12. [Google Scholar] [CrossRef] [PubMed]
  124. Hammers, H.J.; Plimack, E.R.; Infante, J.R.; Rini, B.I.; McDermott, D.F.; Lewis, L.D.; Voss, M.H.; Sharma, P.; Pal, S.K.; Razak, A.R.A.; et al. Safety and Efficacy of Nivolumab in Combination with Ipilimumab in Metastatic Renal Cell Carcinoma: The CheckMate 016 Study. J. Clin. Oncol. 2017, 35, 3851–3858. [Google Scholar] [CrossRef]
  125. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef]
  126. Cao, D.; Xu, H.; Xu, X.; Guo, T.; Ge, W. High tumor mutation burden predicts better efficacy of immunotherapy: A pooled analysis of 103078 cancer patients. Oncoimmunology 2019, 8, e1629258. [Google Scholar] [CrossRef]
  127. Menon, S.; Shin, S.; Dy, G. Advances in Cancer Immunotherapy in Solid Tumors. Cancers 2016, 8, 106. [Google Scholar] [CrossRef]
  128. Saraiva, D.P.; Matias, A.T.; Braga, S.; Jacinto, A.; Cabral, M.G. Establishment of a 3D Co-culture with MDA-MB-231 Breast Cancer Cell Line and Patient-Derived Immune Cells for Application in the Development of Immunotherapies. Front. Oncol. 2020, 10, 1543. [Google Scholar] [CrossRef] [PubMed]
  129. Dijkstra, K.K.; Cattaneo, C.M.; Weeber, F.; Chalabi, M.; van de Haar, J.; Fanchi, L.F.; Slagter, M.; van der Velden, D.L.; Kaing, S.; Kelderman, S.; et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell 2018, 174, 1586–1598.e12. [Google Scholar] [CrossRef]
  130. Ma, W.; Zeng, J.; Chen, S.; Lyu, Y.; Toomey, K.A.; Phan, C.T.; Yoneda, K.Y.; Li, T. Small molecule tyrosine kinase inhibitors modulated blood immune cell counts in patients with oncogene-driven NSCLC. Biomark. Res. 2021, 9, 69. [Google Scholar] [CrossRef]
  131. Lyman, G. Risk factors for cancer. Prim. Care 1992, 19, 465–479. [Google Scholar] [CrossRef]
  132. Kasala, E.R.; Bodduluru, L.N.; Barua, C.C.; Sriram, C.S.; Gogoi, R. Benzo(a)pyrene induced lung cancer: Role of dietary phytochemicals in chemoprevention. Pharmacol. Rep. 2015, 67, 996–1009. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, Y.; Zhang, Z.; Yan, Y.; Lemon, W.J.; LaRegina, M.; Morrison, C.; Lubet, R.; You, M. A chemically induced model for squamous cell carcinoma of the lung in mice: Histopathology and strain susceptibility. Cancer Res. 2004, 64, 1647–1654. [Google Scholar] [CrossRef] [PubMed]
  134. Nettesheim, P.; AS, H. nduction of squamous cell carcinoma in the respiratory tract of mice. J. Natl. Cancer Inst. 1971, 47, 697–701. [Google Scholar]
  135. Rajendran, P.; Ekambaram, G.; Sakthisekaran, D. Protective role of mangiferin against Benzo(a)pyrene induced lung carcinogenesis in experimental animals. Biol. Pharm. Bull. 2008, 31, 1053–1058. [Google Scholar] [CrossRef] [PubMed]
  136. Yan, Y.; Wang, Y.; Tan, Q.; Lubet, R.A.; You, M. Efficacy of deguelin and silibinin on benzo(a)pyrene-induced lung tumorigenesis in A/J mice. Neoplasia 2005, 7, 1053–1057. [Google Scholar] [CrossRef]
  137. Nolan, K.; Verzosa, G.; Cleaver, T.; Tippimanchai, D.; DePledge, L.N.; Wang, X.J.; Young, C.; Le, A.; Doebele, R.; Li, H.; et al. Development of syngeneic murine cell lines for use in immunocompetent orthotopic lung cancer models. Cancer Cell Int. 2020, 20, 417. [Google Scholar] [CrossRef] [PubMed]
  138. Schreiber, L.M.; Urbiola, C.; Das, K.; Spiesschaert, B.; Kimpel, J.; Heinemann, F.; Stierstorfer, B.; Muller, P.; Petersson, M.; Erlmann, P.; et al. The lytic activity of VSV-GP treatment dominates the therapeutic effects in a syngeneic model of lung cancer. Br. J. Cancer 2019, 121, 647–658. [Google Scholar] [CrossRef] [PubMed]
  139. Yan, X.; Wang, L.; Zhang, R.; Pu, X.; Wu, S.; Yu, L.; Meraz, I.M.; Zhang, X.; Wang, J.F.; Gibbons, D.L.; et al. Overcoming resistance to anti-PD immunotherapy in a syngeneic mouse lung cancer model using locoregional virotherapy. Oncoimmunology 2017, 7, e1376156. [Google Scholar] [CrossRef] [PubMed]
  140. Ajona, D.; Ortiz-Espinosa, S.; Moreno, H.; Lozano, T.; Pajares, M.J.; Agorreta, J.; Bertolo, C.; Lasarte, J.J.; Vicent, S.; Hoehlig, K.; et al. A Combined PD-1/C5a Blockade Synergistically Protects against Lung Cancer Growth and Metastasis. Cancer Discov. 2017, 7, 694–703. [Google Scholar] [CrossRef]
  141. Meraz, I.M.; Majidi, M.; Cao, X.; Lin, H.; Li, L.; Wang, J.; Baladandayuthapani, V.; Rice, D.; Sepesi, B.; Ji, L.; et al. TUSC2 Immunogene Therapy Synergizes with Anti-PD-1 through Enhanced Proliferation and Infiltration of Natural Killer Cells in Syngeneic Kras-Mutant Mouse Lung Cancer Models. Immunol. Res. 2018, 6, 163–177. [Google Scholar] [CrossRef] [PubMed]
  142. Doetschman, T. GI GEMs: Genetically engineered mouse models of gastrointestinal disease. Gastroenterology 2011, 140, 380–385 e382. [Google Scholar] [CrossRef] [PubMed]
  143. Frese, K.K.; Tuveson, D.A. Maximizing mouse cancer models. Nat. Rev. Cancer 2007, 7, 645–658. [Google Scholar] [CrossRef] [PubMed]
  144. Schaffer, B.E.; Park, K.S.; Yiu, G.; Conklin, J.F.; Lin, C.; Burkhart, D.L.; Karnezis, A.N.; Sweet-Cordero, E.A.; Sage, J. Loss of p130 accelerates tumor development in a mouse model for human small-cell lung carcinoma. Cancer Res. 2010, 70, 3877–3883. [Google Scholar] [CrossRef]
  145. Manchado, E.; Weissmueller, S.; Morris, J.P.; Chen, C.C.; Wullenkord, R.; Lujambio, A.; de Stanchina, E.; Poirier, J.T.; Gainor, J.F.; Corcoran, R.B.; et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature 2016, 534, 647–651. [Google Scholar] [CrossRef] [PubMed]
  146. Urnov, F.D.; Rebar, E.J.; Holmes, M.C.; Zhang, H.S.; Gregory, P.D. Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet. 2010, 11, 636–646. [Google Scholar] [CrossRef] [PubMed]
  147. Carroll, D. Genome engineering with zinc-finger nucleases. Genetics 2011, 188, 773–782. [Google Scholar] [CrossRef] [PubMed]
  148. Wyman, C.; Kanaar, R. DNA double-strand break repair: All’s well that ends well. Annu. Rev. Genet. 2006, 40, 363–383. [Google Scholar] [CrossRef]
  149. Gaj, T.; Gersbach, C.A.; Barbas, C.F., 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013, 31, 397–405. [Google Scholar] [CrossRef] [PubMed]
  150. Mashimo, T. Gene targeting technologies in rats: Zinc finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats. Dev. Growth Differ. 2014, 56, 46–52. [Google Scholar] [CrossRef]
  151. Wright, A.V.; Nuñez, J.K.; Doudna, J.A. Biology and applications of CRISPR systems: Harnessing nature’s toolbox for genome engineering. Cell 2016, 164, 29–44. [Google Scholar] [CrossRef] [PubMed]
  152. Rakhit, C.P.; Trigg, R.M.; Le Quesne, J.; Kelly, M.; Shaw, J.A.; Pritchard, C.; Martins, L.M. Early detection of pre-malignant lesions in a KRASG12D-driven mouse lung cancer model by monitoring circulating free DNA. Dis. Models Mech. 2019, 12, dmm036863. [Google Scholar] [CrossRef]
  153. Politi, K.; Zakowski, M.F.; Fan, P.D.; Schonfeld, E.A.; Pao, W.; Varmus, H.E. Lung adenocarcinomas induced in mice by mutant EGF receptors found in human lung cancers respond to a tyrosine kinase inhibitor or to down-regulation of the receptors. Genes Dev. 2006, 20, 1496–1510. [Google Scholar] [CrossRef]
  154. Politi, K.; Fan, P.D.; Shen, R.; Zakowski, M.; Varmus, H. Erlotinib resistance in mouse models of epidermal growth factor receptor-induced lung adenocarcinoma. Dis. Models Mech. 2010, 3, 111–119. [Google Scholar] [CrossRef]
  155. Li, D.; Ambrogio, L.; Shimamura, T.; Kubo, S.; Takahashi, M.; Chirieac, L.R.; Padera, R.F.; Shapiro, G.I.; Baum, A.; Himmelsbach, F.; et al. BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 2008, 27, 4702–4711. [Google Scholar] [CrossRef]
  156. Mancini, M.; Thomas, Q.D.; Bourdel, S.; Papon, L.; Bousquet, E.; Jalta, P.; La Monica, S.; Travert, C.; Alfieri, R.; Quantin, X.; et al. Generation and Characterization of a New Preclinical Mouse Model of EGFR-Driven Lung Cancer with MET-Induced Osimertinib Resistance. Cancers 2021, 13, 3441. [Google Scholar] [CrossRef]
  157. He, L.; Luo, L.; Zhu, H.; Yang, H.; Zhang, Y.; Wu, H.; Sun, H.; Jiang, F.; Kathera, C.S.; Liu, L.; et al. FEN1 promotes tumor progression and confers cisplatin resistance in non-small-cell lung cancer. Mol. Oncol. 2017, 11, 1302–1303. [Google Scholar] [CrossRef] [PubMed]
  158. Kasiri, S.; Shao, C.; Chen, B.; Wilson, A.N.; Yenerall, P.; Timmons, B.C.; Girard, L.; Tian, H.; Behrens, C.; Wistuba, I.I.; et al. GLI1 Blockade Potentiates the Antitumor Activity of PI3K Antagonists in Lung Squamous Cell Carcinoma. Cancer Res. 2017, 77, 4448–4459. [Google Scholar] [CrossRef]
  159. Lai, Y.; Wei, X.; Lin, S.; Qin, L.; Cheng, L.; Li, P. Current status and perspectives of patient-derived xenograft models in cancer research. J. Hematol. Oncol. 2017, 10, 106. [Google Scholar] [CrossRef] [PubMed]
  160. Fichtner, I.; Rolff, J.; Soong, R.; Hoffmann, J.; Hammer, S.; Sommer, A.; Becker, M.; Merk, J. Establishment of patient-derived non-small cell lung cancer xenografts as models for the identification of predictive biomarkers. Clin. Cancer Res. 2008, 14, 6456–6468. [Google Scholar] [CrossRef] [PubMed]
  161. Merk, J.; Rolff, J.; Becker, M.; Leschber, G.; Fichtner, I. Patient-derived xenografts of non-small-cell lung cancer: A pre-clinical model to evaluate adjuvant chemotherapy? Eur. J. Cardiothorac. Surg. 2009, 36, 454–459. [Google Scholar] [CrossRef]
  162. Blake, J.A.; Baldarelli, R.; Kadin, J.A.; Richardson, J.E.; Smith, C.L.; Bult, C.J.; Mouse Genome Database, G. Mouse Genome Database (MGD): Knowledgebase for mouse-human comparative biology. Nucleic Acids Res. 2021, 49, D981–D987. [Google Scholar] [CrossRef] [PubMed]
  163. Baldarelli, R.M.; Smith, C.M.; Finger, J.H.; Hayamizu, T.F.; McCright, I.J.; Xu, J.; Shaw, D.R.; Beal, J.S.; Blodgett, O.; Campbell, J.; et al. The mouse Gene Expression Database (GXD): 2021 update. Nucleic Acids Res. 2021, 49, D924–D931. [Google Scholar] [CrossRef]
  164. Begley, D.A.; Krupke, D.M.; Neuhauser, S.; Sundberg, J.; Bult, C.J. Abstract 1190: MMHCdb: A knowledgebase for the evolving landscape of mouse models of human cancer. Cancer Res. 2022, 82, 1190. [Google Scholar] [CrossRef]
  165. Meehan, T.F.; Conte, N.; Goldstein, T.; Inghirami, G.; Murakami, M.A.; Brabetz, S.; Gu, Z.; Wiser, J.A.; Dunn, P.; Begley, D.A.; et al. PDX-MI: Minimal Information for Patient-Derived Tumor Xenograft Models. Cancer Res. 2017, 77, e62–e66. [Google Scholar] [CrossRef]
  166. Stewart, E.L.; Mascaux, C.; Pham, N.A.; Sakashita, S.; Sykes, J.; Kim, L.; Yanagawa, N.; Allo, G.; Ishizawa, K.; Wang, D.; et al. Clinical Utility of Patient-Derived Xenografts to Determine Biomarkers of Prognosis and Map Resistance Pathways in EGFR-Mutant Lung Adenocarcinoma. J. Clin. Oncol. 2015, 33, 2472–2480. [Google Scholar] [CrossRef] [PubMed]
  167. Woo, X.Y.; Srivastava, A.; Graber, J.H.; Yadav, V.; Sarsani, V.K.; Simons, A.; Beane, G.; Grubb, S.; Ananda, G.; Liu, R.; et al. Genomic data analysis workflows for tumors from patient-derived xenografts (PDXs): Challenges and guidelines. BMC Med. Genom. 2019, 12, 92. [Google Scholar] [CrossRef] [PubMed]
  168. Jo, H.; Yagishita, S.; Hayashi, Y.; Ryu, S.; Suzuki, M.; Kohsaka, S.; Ueno, T.; Matsumoto, Y.; Horinouchi, H.; Ohe, Y.; et al. Comparative Study on the Efficacy and Exposure of Molecular Target Agents in Non-small Cell Lung Cancer PDX Models with Driver Genetic Alterations. Mol. Cancer Ther. 2022, 21, 359–370. [Google Scholar] [CrossRef]
  169. Rydberg Millrud, C.; Deronic, A.; Gronberg, C.; Jaensson Gyllenback, E.; von Wachenfeldt, K.; Forsberg, G.; Liberg, D. Blockade of IL-1alpha and IL-1beta signaling by the anti-IL1RAP antibody nadunolimab (CAN04) mediates synergistic anti-tumor efficacy with chemotherapy. Cancer Immunol. Immunother. 2023, 72, 667–678. [Google Scholar] [CrossRef] [PubMed]
  170. Dong, X.; Guan, J.; English, J.C.; Flint, J.; Yee, J.; Evans, K.; Murray, N.; Macaulay, C.; Ng, R.T.; Gout, P.W.; et al. Patient-derived first generation xenografts of non-small cell lung cancers: Promising tools for predicting drug responses for personalized chemotherapy. Clin. Cancer Res. 2010, 16, 1442–1451. [Google Scholar] [CrossRef]
  171. Wang, J.; Anderson, M.G.; Oleksijew, A.; Vaidya, K.S.; Boghaert, E.R.; Tucker, L.; Zhang, Q.; Han, E.K.; Palma, J.P.; Naumovski, L.; et al. ABBV-399, a c-Met Antibody-Drug Conjugate that Targets Both MET-Amplified and c-Met-Overexpressing Tumors, Irrespective of MET Pathway Dependence. Clin. Cancer Res. 2017, 23, 992–1000. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, X.C.; Zhang, J.; Li, M.; Huang, X.S.; Yang, X.N.; Zhong, W.Z.; Xie, L.; Zhang, L.; Zhou, M.; Gavine, P.; et al. Establishment of patient-derived non-small cell lung cancer xenograft models with genetic aberrations within EGFR, KRAS and FGFR1: Useful tools for preclinical studies of targeted therapies. J. Transl. Med. 2013, 11, 168. [Google Scholar] [CrossRef] [PubMed]
  173. Ma, W.; Wei, S.; Li, Q.; Zeng, J.; Xiao, W.; Zhou, C.; Yoneda, K.Y.; Zeki, A.A.; Li, T. Simvastatin Overcomes Resistance to Tyrosine Kinase Inhibitors in Patient-derived, Oncogene-driven Lung Adenocarcinoma Models. Mol. Cancer Ther. 2024, 23, 700–710. [Google Scholar] [CrossRef]
  174. Roper, N.; El Meskini, R.; Maity, T.; Atkinson, D.; Day, A.; Pate, N.; Cultraro, C.M.; Pack, S.; Zgonc, V.; Weaver Ohler, Z.; et al. Functional Heterogeneity in MET Pathway Activation in PDX Models of Osimertinib-resistant EGFR-driven Lung Cancer. Cancer Res. Commun. 2024, 4, 337–348. [Google Scholar] [CrossRef] [PubMed]
  175. Qin, B.; Jiao, X.; Yuan, L.; Liu, K.; Zang, Y. Advances in Patient Derived Tumor Xenograft (PDTX) Model from Lung Cancer. Zhongguo Fei Ai Za Zhi 2017, 20, 715–719. [Google Scholar]
  176. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  177. Gao, H.; Korn, J.M.; Ferretti, S.; Monahan, J.E.; Wang, Y.; Singh, M.; Zhang, C.; Schnell, C.; Yang, G.; Zhang, Y.; et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 2015, 21, 1318–1325. [Google Scholar] [CrossRef]
  178. Ilie, M.; Nunes, M.; Blot, L.; Hofman, V.; Long-Mira, E.; Butori, C.; Selva, E.; Merino-Trigo, A.; Venissac, N.; Mouroux, J.; et al. Setting up a wide panel of patient-derived tumor xenografts of non-small cell lung cancer by improving the preanalytical steps. Cancer Med. 2015, 4, 201–211. [Google Scholar] [CrossRef] [PubMed]
  179. Fu, S.; Zhao, J.; Bai, H.; Duan, J.; Wang, Z.; An, T.; Wang, J. High-fidelity of non-small cell lung cancer xenograft models derived from bronchoscopy-guided biopsies. Thorac. Cancer 2016, 7, 100–110. [Google Scholar] [CrossRef] [PubMed]
  180. Williams, S.A.; Anderson, W.C.; Santaguida, M.T.; Dylla, S.J. Patient-derived xenografts, the cancer stem cell paradigm, and cancer pathobiology in the 21st century. Lab. Investig. 2013, 93, 970–982. [Google Scholar] [CrossRef] [PubMed]
  181. Yoshida, G.J. Applications of patient-derived tumor xenograft models and tumor organoids. J. Hematol. Oncol. 2020, 13, 4. [Google Scholar] [CrossRef]
  182. Lin, S.; Huang, G.; Cheng, L.; Li, Z.; Xiao, Y.; Deng, Q.; Jiang, Y.; Li, B.; Lin, S.; Wang, S.; et al. Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy. MAbs 2018, 10, 1301–1311. [Google Scholar] [CrossRef]
  183. Shultz, L.D.; Brehm, M.A.; Garcia-Martinez, J.V.; Greiner, D.L. Humanized mice for immune system investigation: Progress, promise and challenges. Nat. Rev. Immunol. 2012, 12, 786–798. [Google Scholar] [CrossRef]
  184. Abdel-Aziz, A.K.; Saadeldin, M.K.; D’Amico, P.; Orecchioni, S.; Bertolini, F.; Curigliano, G.; Minucci, S. Preclinical models of breast cancer: Two-way shuttles for immune checkpoint inhibitors from and to patient bedside. Eur. J. Cancer 2019, 122, 22–41. [Google Scholar] [CrossRef]
  185. Brehm, M.A.e.a. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression. FASEB J. 2019, 33, 3137–3151. [Google Scholar] [CrossRef] [PubMed]
  186. Yanagawa, S.; Tahara, H.; Shirouzu, T.; Kawai, S.; Tanaka, Y.; Ide, K.; Akimoto, S.; Ohdan, H. Development of a humanized mouse model to analyze antibodies specific for human leukocyte antigen (HLA). PLoS ONE 2021, 16, e0236614. [Google Scholar] [CrossRef] [PubMed]
  187. Cheng, H.; Zheng, Z.; Cheng, T. New paradigms on hematopoietic stem cell differentiation. Protein Cell 2020, 11, 34–44. [Google Scholar] [CrossRef]
  188. Hess, N.J.; Lindner, P.N.; Vazquez, J.; Grindel, S.; Hudson, A.W.; Stanic, A.K.; Ikeda, A.; Hematti, P.; Gumperz, J.E. Different Human Immune Lineage Compositions Are Generated in Non-Conditioned NBSGW Mice Depending on HSPC Source. Front. Immunol. 2020, 11, 573406. [Google Scholar] [CrossRef]
  189. Verma, B.; Wesa, A. Establishment of Humanized Mice from Peripheral Blood Mononuclear Cells or Cord Blood CD34+ Hematopoietic Stem Cells for Immune-Oncology Studies Evaluating New Therapeutic Agents. Curr. Protoc. Pharmacol. 2020, 89, e77. [Google Scholar] [CrossRef] [PubMed]
  190. Pearson, T.; Greiner, D.L.; Shultz, L.D. Creation of “humanized” mice to study human immunity. Curr. Protoc. Immunol. 2008, 81, 15–21. [Google Scholar] [CrossRef]
  191. Hasgur, S.; Aryee, K.E.; Shultz, L.D.; Greiner, D.L.; Brehm, M.A. Generation of Immunodeficient Mice Bearing Human Immune Systems by the Engraftment of Hematopoietic Stem Cells. Methods Mol. Biol. 2016, 1438, 67–78. [Google Scholar] [PubMed]
  192. Brehm, M.A.; Cuthbert, A.; Yang, C.; Miller, D.M.; DiIorio, P.; Laning, J.; Burzenski, L.; Gott, B.; Foreman, O.; Kavirayani, A.; et al. Parameters for establishing humanized mouse models to study human immunity: Analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rgamma(null) mutation. Clin. Immunol. 2010, 135, 84–98. [Google Scholar] [CrossRef]
  193. Lepus, C.M.; Gibson, T.F.; Gerber, S.A.; Kawikova, I.; Szczepanik, M.; Hossain, J.; Ablamunits, V.; Kirkiles-Smith, N.; Herold, K.C.; Donis, R.O.; et al. Comparison of human fetal liver, umbilical cord blood, and adult blood hematopoietic stem cell engraftment in NOD-scid/gammac−/−, Balb/c-Rag1−/−gammac−/−, and C.B-17-scid/bg immunodeficient mice. Hum. Immunol. 2009, 70, 790–802. [Google Scholar] [CrossRef]
  194. Mold, J.E.; Venkatasubrahmanyam, S.; Burt, T.D.; Michaelsson, J.; Rivera, J.M.; Galkina, S.A.; Weinberg, K.; Stoddart, C.A.; McCune, J.M. Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science 2010, 330, 1695–1699. [Google Scholar] [CrossRef] [PubMed]
  195. Matas-Cespedes, A.; Brown, L.; Mahbubani, K.T.; Bareham, B.; Higgins, J.; Curran, M.; de Haan, L.; Lapointe, J.M.; Stebbings, R.; Saeb-Parsy, K. Use of human splenocytes in an innovative humanised mouse model for prediction of immunotherapy-induced cytokine release syndrome. Clin. Transl. Immunol. 2020, 9, e1202. [Google Scholar] [CrossRef] [PubMed]
  196. Garcia-Beltran, W.F.; Claiborne, D.T.; Maldini, C.R.; Phelps, M.; Vrbanac, V.; Karpel, M.E.; Krupp, K.L.; Power, K.A.; Boutwell, C.L.; Balazs, A.B.; et al. Innate Immune Reconstitution in Humanized Bone Marrow-Liver-Thymus (HuBLT) Mice Governs Adaptive Cellular Immune Function and Responses to HIV-1 Infection. Front. Immunol. 2021, 12, 667393. [Google Scholar] [CrossRef]
  197. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the various in vitro methods. Immortalized cell lines, primary cell cultures, and organoids established from lung cancer patients serve as essential tools in precision oncology research. Each model offers unique insights into lung cancer biology and treatment responses. Co-culture of patient-derived lung tumor cells with PBMCs isolated from the same patient’s whole blood provides a more physiologically relevant model by incorporating the patient’s immune cells, allowing for real-time study of immune–tumor interactions (by Figdraw.com, accessed on 25 August 2022). Abbreviations: CRCs, conditional reprogramming cultures; PBMC, peripheral blood mononuclear cell; ROCK, Rho kinase.
Figure 1. Schematic representation of the various in vitro methods. Immortalized cell lines, primary cell cultures, and organoids established from lung cancer patients serve as essential tools in precision oncology research. Each model offers unique insights into lung cancer biology and treatment responses. Co-culture of patient-derived lung tumor cells with PBMCs isolated from the same patient’s whole blood provides a more physiologically relevant model by incorporating the patient’s immune cells, allowing for real-time study of immune–tumor interactions (by Figdraw.com, accessed on 25 August 2022). Abbreviations: CRCs, conditional reprogramming cultures; PBMC, peripheral blood mononuclear cell; ROCK, Rho kinase.
Cancers 17 00022 g001
Figure 2. A schematic representation of the in vivo mouse models currently available for cancer research. Carcinogen-induced mouse models are induced to develop certain types of cancer by exposing them to certain environmental risk factors (carcinogenic chemicals, radiation, etc.). Syngeneic mouse models are immunocompetent animals that bear tumors of mouse origin. CDX models are immunodeficient mouse models, and tumors are implanted to assess drug function on a tumor. GEMM models are mouse strains that have been manipulated genetically either by the overexpression of an oncogene or by the loss of a tumor suppressor gene function. PDX models are immunocompromised animals implanted with tumors of human origin. Humanized PDX models can represent the human immune system to a certain extent along with tumors of human origin to study tumor–immune system interactions (by Figdraw.com; accessed on 27 August 2022).
Figure 2. A schematic representation of the in vivo mouse models currently available for cancer research. Carcinogen-induced mouse models are induced to develop certain types of cancer by exposing them to certain environmental risk factors (carcinogenic chemicals, radiation, etc.). Syngeneic mouse models are immunocompetent animals that bear tumors of mouse origin. CDX models are immunodeficient mouse models, and tumors are implanted to assess drug function on a tumor. GEMM models are mouse strains that have been manipulated genetically either by the overexpression of an oncogene or by the loss of a tumor suppressor gene function. PDX models are immunocompromised animals implanted with tumors of human origin. Humanized PDX models can represent the human immune system to a certain extent along with tumors of human origin to study tumor–immune system interactions (by Figdraw.com; accessed on 27 August 2022).
Cancers 17 00022 g002
Figure 3. Summary of key preclinical models for precision lung cancer research.
Figure 3. Summary of key preclinical models for precision lung cancer research.
Cancers 17 00022 g003
Table 1. Summary of the most commonly used human lung cancer cell lines.
Table 1. Summary of the most commonly used human lung cancer cell lines.
Cell LineHistologyDriver Oncogene(s)Other MutationsReference
H1975AdenocarcinomaEGFR L858R, T790MPIK3CA, TP53[41]
H3255AdenocarcinomaEGFR L858RTP53[42,43]
HCC2935AdenocarcinomaEGFR Exon19 del (E746-A750)TP53, APC[44]
HCC4006AdenocarcinomaEGFR Exon 19 del (L747-E749)TP53, PIK3CA[45]
H1650AdenocarcinomaEGFR Exon19 del (E746-A750), T790MTP53[43]
HCC827AdenocarcinomaEGFR Exon19 del (E746-A750)TP53[43,46]
PC9AdenocarcinomaEGFR Exon 19 del (E746-A750)TP53[47]
H1573AdenocarcinomaKRAS G12A, NRASQ61KPTPN1, TP53[48]
H23AdenocarcinomaKRAS G12CTP53, ATM, STK11[49]
H460Large cell carcinomaKRAS Q61HSTK11, PIK3CA, TP53[50]
A549AdenocarcinomaKRAS G12SSTK11, TP53[41]
H2122AdenocarcinomaEML4-ALK variant 3a/bTP53[51]
H358AdenocarcinomaKRAS G12CCTNNB1, TP53 deletion[49]
H1299AdenocarcinomaN-RAS Q61KTP53 deletion[49,52]
H596AdenocarcinomaMET exon 14 skippingPIK3CA, RB1, TP53[53]
H522AdenocarcinomaKRAS G12STP53[50]
H2228AdenocarcinomaEML4-ALK fusion v3
ALK-PTPN3
TP53[54]
H661Large cell carcinomaARHGAP35 K179* mutationCDKN2A, LASP1, TP53[55]
H2126Adenocarcinoma-SMARCA4, TP53[56]
H1437Adenocarcinoma-TP53[56]
H1563Adenocarcinoma-CDKN2A[56]
H661Large cell carcinoma-CDKN2A, LASP1, TP53[56]
H1770Carcinoma-TP53[57]
H2170Squamous cell carcinoma-RHOA, TP53[58]
H69PRSCLC-PIK3X, TP53, RB1[59]
DMS235SCLC- [60]
H2066SCLC-TP53[61]
COR-L279SCLC-EP300, TP53[62]
SHP-77SCLC-ABL1, KRAS, RAC1, TP53[63]
NCI-H727SCLC-PKD1L-TNS3 fusion,
KRAS, TP53
[64]
Note: Driver oncogenes are defined as EGFR: epidermal growth factor receptor; KRAS: Kirsten rat sarcoma; HER2: human epidermal growth factor 2; EML4-ALK: echinoderm microtubule-associated protein-like 4-Anaplastic lymphoma kinase; MET: mesenchymal epithelial transition; ROS: c-ROS oncogene 1; BRAF: v-raf murine sarcoma viral oncogene homolog B; NRAS: neuroblastoma RAS viral; SCLC: small cell lung carcinoma.
Table 2. Summary of in vitro models.
Table 2. Summary of in vitro models.
In Vitro Models AdvantagesDisadvantagesApplications
Human lung cancer cell line modelsInexpensive, scalable, and widely available.Limited representation of parent tumor heterogeneity; no TME.Study cancer at molecular and cellular levels.
Primary cell culturesCloser to the patient genomic profile.Low success rate in establishing patient-derived primary cell cultures.Study cancer at molecular and cellular levels.
Conditionally reprogrammed cellRemaining the original karyotypes.No TME.Study cancer at molecular and cellular levels.
Cancer spheroids and organoidsHighly mimic original histopathology of tumors, rapid and robust growth.Expensive, lack of TME and tumor heterogeneity.Study self-renewal, drug resistance, heterogeneity oncogenic transformation, and drug screening.
Co-culture system of patient-derived immune cells and patient-derived tumorsMimic the TME.Difficulty in reproducing results and interpreting results.Study the interaction between immune system and tumor cell.
Abbreviations: TME, tumor microenvironment.
Table 3. Summary of in vivo models.
Table 3. Summary of in vivo models.
In Vivo Models AdvantagesDisadvantagesApplications
Carcinogen-induced mouse modelsTumor formation time is similar to human tumor growth progress.1. Relatively long latency.
2. Uncontrollable experimental results.
3. Not popular anymore.
Study tumor morphology, histology, and molecular characteristics.
XenograftsCDXReadily available, over 300 models, cost-effective, easy to manipulate genetically, and widely used for high-throughput screening of drugs.1. Limited representation of tumor heterogeneity and microenvironment; results may not always translate to clinical outcomes.
2. Except the subcutaneous implantation, other transplant methods are technically difficult and need special technology to monitor the tumor growth.
Best suited for initial drug screening and mechanistic studies involving genetic and molecular pathways.
PDXRetain the genetic, histological, and morphological features of original patient’s tumors.1. Require immunocompromised mice, which is not suitable to evaluate immunotherapy.
2. Snapshot of patient tumor that cannot reflect the heterogeneous of whole tumor.
3. Orthotopic transplantation requires higher technical skill, has high cost, and needs in vivo imaging tool to monitor the tumor growth.
4. Takes 1–9 months to establish a model.
5. Mouse-derived cells gradually replaced human stromal cells.
Valuable for validating drug efficacy of chemotherapy and/or molecularly targeted therapy for individual patients and tailoring treatment plans for specific genetic profiles.
GEMMMimic genetic mutations observed in human cancers, allowing for the study of tumor initiation, progression, and therapy resistance within an immune-competent context.1. Time-consuming and expensive to develop; can lack the full range of human tumor heterogeneity.
2. CRISPR/Cas9 system can evaluate off-target activity.
1. Study the function of tumor gene mutations and mechanisms of drug resistance.
2. Evaluate the mutation effect of immunotherapy.
Syngeneic models1. Genetically identical to the host, enabling studies for immunotherapy and drug toxicity within a fully functional immune system.
2. Cost-effective compared to humanized models.
1. Tumor lines are typically murine in origin, which may not accurately reflect human tumor biology.
2. Adapt to mouse biology only, not sure if the outcome is suitable for the human immune system.
Useful for evaluating the interaction between tumor cells and immune cells, as well as testing immunotherapeutic agents.
Humanized PDX models1. Hu-PBMC model is fast to grow.
2. Incorporate a functional human immune system, allowing for long-term research and low or miner GVHD rate.
1. Complex and expensive to establish.
2. Variability in immune reconstitution can affect reproducibility (low reconstitution rates of NK and B cells).
3. Short study period due to mouse developing GVHD.
Ideal for studying ICIs, CAR-T cell therapies, ADC, and other immunomodulatory treatments.
Patient-derived LCOs1. Offer a high success rate in establishing cultures that maintain the histopathological and genomic fidelity of primary tumors.
2. Enable rapid drug screening and correlate well with clinical outcomes.
Organoids may not fully replicate the TME, including interactions with the immune system and stroma.Emerging as a crucial tool for personalized medicine, facilitating the testing of various treatment regimens and helping guide clinical decision-making.
Abbreviations: CDX, cell line derived xenograft; PDX, patients-derived xenograft; SCLC, small cell lung cancer; NSCLC non-small cell lung cancer; GEMM, genetically engineered mouse model; PBMC, peripheral blood mononuclear cell; IV, intravenous; IP, intraperitoneal; IF, interfemoral; HPSC, hematopoietic stem and progenitor cells; GVHD, graft-versus-host disease; NK, natural killer, TME, tumor microenvironment.
Table 4. Resources for PDX and mouse models.
Table 4. Resources for PDX and mouse models.
DatabaseWebsiteInformationReference
Mouse Genome Database (MGD)http://www.informatics.jax.org/Gene characterization, nomenclature, mapping, gene homologies among vertebrates, sequence links, phenotypes, allelic variants and mutants, and strain data[162]
Gene Expression Database (GXD)http://www.informatics.jax.org/expression.shtmlGene expression information from the laboratory mouse[163]
Mouse Models of Human Cancer database (MMHCdb)http://tumor.informatics.jax.org/mtbwi/index.doSpontaneous and induced tumors in mice including GEMM, PDX[164]
Abbreviations: GEMM: genetically engineered mouse model; PDX: patient-derived xenograft.
Table 5. Comparison of the different humanized mouse models.
Table 5. Comparison of the different humanized mouse models.
NameMethodAdvantagesDisadvantages
Humanized PBMC (hu-PBMC) Mouse ModelsTail vein injection of hu-PBMCs, which include lymphocytes (T, B cells and NK cells), neutrophils, and monocytes
  • Easy and fast to build
  • Cost-effective
  • Idea model for studying T cell immunity which could be maintained by cytokines or GM-CSF or IFNγ
  • Short term study model
  • Fast GVHD (3–9 weeks)
  • B cells reconstitute rates are low
Humanized CD34+ (hu-HPSC) Mouse ModelsTail vein injection of hu-HPSCs, which include hematopoietic stem cells (HSC) and hematopoietic progenitor cells
  • Multi-lineage engraftment
  • Robust T cell maturation
  • Idea long-term study model without GVHD
  • Longer time to build
  • More expensive
  • NK and B cell reconstitute rates are low
Knock-in Humanized Mouse ModelsKnock-in human gene to replace murine gene
  • Fully functional immune system
  • Long time to build
  • High cost
Human Fetal Bone, Liver, and Thymic Tissue (BLT) EngraftmentSubcapsular injection of HLA-matched fetal thymus or other immune organs into kidney capsule; more intact TME
  • Best mimic the human TME for immunotherapeutics
  • Limited resources from fetal thymus
  • Ethical report
Spleen Mononuclear Cell (SPMC) EngraftmentIntraperitoneal injection of single cells from donor splenic tissue; More B cells and TM cells than PBMCs
  • Prediction of CRS while minimizing GVHD
  • Difficult to build
  • Limited resources for deceased spleen donor
Abbreviations: CRS, cytokine release syndrome; GM-CSF, granulocyte–macrophage colony-stimulating factor; GVHD, graft-versus-host disease; HLA, human leukocyte antigen; TM, memory T cells; TME, tumor microenvironment.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yu, J.-Z.; Kiss, Z.; Ma, W.; Liang, R.; Li, T. Preclinical Models for Functional Precision Lung Cancer Research. Cancers 2025, 17, 22. https://doi.org/10.3390/cancers17010022

AMA Style

Yu J-Z, Kiss Z, Ma W, Liang R, Li T. Preclinical Models for Functional Precision Lung Cancer Research. Cancers. 2025; 17(1):22. https://doi.org/10.3390/cancers17010022

Chicago/Turabian Style

Yu, Jie-Zeng, Zsofia Kiss, Weijie Ma, Ruqiang Liang, and Tianhong Li. 2025. "Preclinical Models for Functional Precision Lung Cancer Research" Cancers 17, no. 1: 22. https://doi.org/10.3390/cancers17010022

APA Style

Yu, J.-Z., Kiss, Z., Ma, W., Liang, R., & Li, T. (2025). Preclinical Models for Functional Precision Lung Cancer Research. Cancers, 17(1), 22. https://doi.org/10.3390/cancers17010022

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

Article Metrics

Back to TopTop