Open Access

Recent advances in lung cancer organoid (tumoroid) research (Review)

  • Authors:
    • Qiang Zhang
    • Mingyang Zhang
  • View Affiliations

  • Published online on: August 1, 2024     https://doi.org/10.3892/etm.2024.12672
  • Article Number: 383
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is the most critical type of malignant tumor that threatens human health. Traditional preclinical models have certain defects; for example, they cannot accurately reflect the characteristics of lung cancer and their development is costly and time‑consuming. Through self‑organization, cancer stem cells (CSCs) generate cancer organoids that have a structure similar to that of lung cancer tissues, overcoming to some extent the aforementioned challenges, thus enabling them to have broader application prospects. Lung cancer organoid (LCO) development methods can be divided into three broad categories based on the source of cells, which include cell lines, patient‑derived xenografts and patient tumor tissue/pleural effusion. There are 17 different methods that have been described for the development of LCOs. These methods can be further merged into six categories based on the source of cells, the pre‑treatment method used, the composition of the medium and the culture scaffold. These categories are: i) CSCs induced by defined transcription factors; ii) suspension culture; iii) relative optimal culture medium; iv) suboptimal culture medium; v) mechanical digestion and suboptimal culture medium; and vi) hydrogel scaffold. In the current review, the advantages and disadvantages of each of the aforementioned methods are summarized, and references for supporting studies are cited.

1. Introduction

Lung cancer constitutes 11.4% of all newly diagnosed cases of cancer, and is the leading cause of cancer-associated mortality worldwide (1). Lung cancer is a heterogenous disease that is divided into two major categories based on histopathology: Small cell lung cancer (SCLC) and non-SCLC (NSCLC). NSCLC accounts for 85% of lung cancer cases, and has several subtypes, including lung adenocarcinoma (LUAD), lung squamous cell carcinoma and large cell lung carcinoma (2). SCLC is a type of neuroendocrine tumor that is classified into two subtypes: SCLC and combined SCLC (3,4). Importantly, in addition to different histopathological subtypes, the heterogeneity of lung cancer also refers to differences between patients with the same subtype, or differences among cells in the same tumor tissue (5-10). The heterogeneity of lung cancer affects clinical treatment, since patients with the same pathological type may have diverse reactions to the same treatment (11-14). To effectively treat lung cancer, it is necessary to explore the source of lung cancer heterogeneity, identify specific antitumor drugs and achieve personalized treatment for patients.

Conventional two-dimensional (2D) culture and patient-derived xenograft (PDX) models are useful tools that assist in understanding the mechanisms underlying the occurrence, development and heterogeneity of lung cancer. However, these tools have certain limitations. Culture methods, passage numbers and other unexpected factors may cause tumor cell lines to lose the phenotype and genotype of a primary tumor in a 2D culture model (15-17). Moreover, as 2D culture models lack extracellular matrix, stromal cells and immune cells, they cannot accurately simulate the tumor microenvironment (TME) and hence the conditions affecting cancers in vivo (18-21). Moreover, the defects of 2D culture models frequently cause antitumor drugs to show efficacies and toxicities in vivo that are different from those obtained in vitro during drug screening, which causes rapid drug screening to be challenging (22,23). Compared with 2D culture models, PDX models in which researchers transplant surgically resected tumor tissues into immunodeficient mice, are more accurate in representing the phenotype, genotype and TME of the parental tumor (24-28). However, the following issues can be observed: i) The proportion of transplantations that are successful in the establishment of PDX models is sometimes too low (29-31); ii) the process of successfully developing PDX models for drug screening is time consuming (32,33), and the condition of the patient often deteriorates during this period; iii) murine stromal cells gradually replace patient stromal cells, which changes the TME of the PDX model (34,35); and iv) the mechanism of interaction between tumor cells and immune cells in a PDX model is challenging to investigate because general PDX models lack immune cells (36,37).

The narrow limitations of conventional tumor cell lines and PDX models have driven researchers to investigate improved preclinical tumor models that preserve the characteristics of primary tumors to the largest extent. Such models are designed to be built in a short amount of time for rapid antitumor drug screening and expanded for the long-term investigation of cancer mechanisms and modification of treatment plans. Tumor organoids, also known as tumoroids (38), have thus emerged. Through self-organization, stem cells generate organoids that retain almost all the features of parental tissues (39,40). Numerous studies have demonstrated that tumor organoids have broad applications in different types of cancer (41-47). Researchers are using organoids to investigate lung cancer, with different studies describing various methods for the establishment of lung cancer organoids (LCOs) (48-80). These methods can be divided into three broad categories based on the source of cells used, namely cancer cell line-based LCOs, PDX-derived LCOs and patient-derived LCOs (Fig. 1). At present, 17 different methods for building LCOs have been described in the literature (Tables I and SI). These methods can be merged into six categories based on the source of cells, the pre-treatment method used, the composition of the medium and/or the culture scaffold: i) Cancer stem cells (CSCs) induced by defined transcription factors (48); ii) suspension culture method (52,66); iii) relative optimal culture medium with serum-free additive, amino acids, growth factor, stemness-related signaling pathway activators and apoptosis signaling pathway inhibitors (49-51,53,55,57,68,76,79,80); iv) suboptimal culture medium (relative optimal culture medium without stemness-associated signaling pathway activators) (71); v) mechanical digestion and suboptimal culture medium (70); and vi) hydrogel scaffold (69,78) (Fig. 1; Tables I, II, SI and SII). The current review presents the advantages and drawbacks of these methods, and cites references for relevant studies. The three broad categories of LCOs are discussed, along with the different methods used to establish LCOs.

Table I

Characteristics of methods for the establishment of lung cancer organoids.

Table I

Characteristics of methods for the establishment of lung cancer organoids.

First author/s, year/sType of cancerStageCell sourceSuccess rate of organoid establishmentSuccess rate of pure LCO establishment, %Success rate of recoveryDefinition of long-term culture, passagesNumber of studies(Refs.)
Ogawa et al, 2017NSCLC (LUAD)-A549 cell line--- 1(48)
Li et al, 2020NSCLCI-IIIPTT71.43% (10/14)-100% (10/10)-1(49)
Han et al, 2022NSCLC-PTT----1(50)
Wang et al, 2019NSCLC (LUAD)-PTT----1(51)
Zhang et al, 2021NSCLC (LUAD)IPTT----1(52)
Li et al, 2020 and 2021NSCLC (LUAD)I-IIIPTT80% (12/15)---2(53,54)
Shi et al, 2020; Liu et al, 2022NSCLC (LUSC, LUAD)IIIPTT/PDX88% (57/65)-->102(55,56)
Sachs et al, 2019; Dijkstra et al, 2018; Cattaneo et al, 2020; Dijkstra et al, 2020; Bie et al, 2021; Sándor et al, 2021; Kim et al, 2021; Padmanabhan et al, 2021; Yokota et al, 2021NSCLC-PTT/PE41-88%7-92.7->108(57-65)
Tamura et al, 2018; Takahashi et al, 2019NSCLC (LUSC, LASC)-PTT/PE    2(66-67)
Ma et al, 2021NSCLC (LUAD)-PTT/PE----1(68)
Mazzocchi et al, 2022NSCLC (LUAD)PE----1(69)
Hu et al, 2021NSCLC (LUAD, LUSC), SCLCIII-IVPTT79% (81/103)---1(70)
Liu et al, 2022; Kim et al, 2019; Jung et al, 2019; Chen et al, 2020; Chen et al, 2022; Peng et al, 2022NSCLC (LUAD, LCLC, LUSC, LASC), SCLC-PTT58-87%7170% (39/56)>10 (56,71-75)
Choi et al, 2021SCLC-PTT80% (8/10)--7-122(76,77)
Gmeiner et al, 2020SCLC-PDX----1(78)
Chen et al, 2022SCLC-PDX----1(79)
Redin et al, 2022SCLC-PDX----1(80)

[i] LASC, lung adenosquamous carcinoma; LCO, lung cancer organoid; LCLC, large cell lung carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; NSCLC, non-small cell lung cancer; PDX, patient-derived xenograft; PE, pleural effusion; PTT, patient tumor tissue; SCLC, small cell lung cancer.

Table II

Characteristics of the culture medium in different methods for the establishment of lung cancer organoids.

Table II

Characteristics of the culture medium in different methods for the establishment of lung cancer organoids.

 Culture medium 
  Stemness-related signaling pathway activatorsApoptosis signaling pathway inhibitors 
First author/s, yearMethodMediumSFAAmino acidGrowth factorWnt signalingBMP signalingTGF-β signalingRho-ROCK signalingp38 MAPK signaling(Refs.)
Ogawa et al, 2017IDMEM--FGF2, insulin, transferrin-----(48)a
Zhang et al, 2021IIDMEM/F12N2/B27-EGF, FGF10, FGF2, HGF-----(52)b
Tamura et al, 2018II FBIM002c (suspension culture)--------(66)
Li et al, 2020IIIAdDMEM/F12N2/B27GM, Gln, NAM, NACEGF, FGF10, FGF2, PGE2, gastrin 1Wnt3A, RSPO1NOGA83-01-SB202190(49)a
Han et al, 2022IIIAdDMEMB27GM, NAM, NACEGF, FGF10, FGF2, PGE2RSPONOGA83-01Y27632SB202190(50)
Wang et al, 2019IIIAdMEM/F12B27GM, NAC, NAMEGF, FGF10, FGF2, DHTRSPONOGA83-01Y27632SB202190(51)
Li et al, 2020IIIAdDMEM/F12B27GM, NAM, NACFGF10, FGF7RSPO1NOGA83-01Y27632SB202190(53)
Shi et al, 2020IIIAdDMEM/F12B27GM, NACEGF, FGF10, FGF4CHIR99021NOGA83-01Y27632-(55)
Sachs et al, 2019IIIAO medium; AdDMEM/F12B27GM, NAM, NACFGF10, FGF7RSPO1NOGA83-01Y27632SB202190(57)
Ma et al, 2021IIILUAD organoid\ culture medium (OmaStem®)d--------(68)
Choi et al, 2021IIIAdDMEM/F12N2/B27-FGF2, EGFWnt3A, RSPO1NOGA83-01Y27632-(76)
Chen et al, 2022IIIDMEM/F12 (PRF)B27Glutamine, NAC, NAMFGF10, EGF, FGF7RSPO3-A83-01Y27632SB202190(79)
Redin et al, 2022IIIDMEM/F12B27NAC, NAMEGF, FGF, gastrin 1RSPONOGA83-01Y27632-(80)e
Kim et al, 2019IVDMEM/F12N2/B27-FGF2, EGF---Y27632-(71)
Hu et al, 2021VDMEM/F12N2/B27GM, NAC, NAMEGF--A83-01Y27632SB202190(70)f
Mazzocchi et al, 2022VIRPMI-5--------(69)
Gmeiner et al, 2020VIDMEM--------(78)

[i] aCulture includes bovine serum albumin;

[ii] bculture includes inactivated fetal bovine serum;

[iii] cFBIM002 medium from the Fukushima Translational Research Project (Fukushima/Japan);

[iv] dOmaStem® Lung Cancer Medium (Human), OM14 (Guangzhou/China);

[v] eculture includes HyClone serum;

[vi] fculture includes forskolin and dexamethasone. adDMEM, advanced Dulbecco's modified Eagle's medium; AO, airway organoid; B27, B-27 supplement; BMP, bone morphogenetic protein; DHT, dihydrotestosterone; DMEM, Dulbecco's modified Eagle's medium; EGF, epidermal growth factor; F12, Ham's F 12 nutrient medium; FGF, fibroblast growth factor; Gln, L-glutamine; GM, Glutamax; HGF, hepatocyte growth factor; LUAD, lung adenocarcinoma; MAPK, mitogen-activated protein kinase; N2, N-2 supplement; NAC, N-acetyl-l-cysteine; NAM, nicotinamide; NOG, Noggin; PGE2, prostaglandin E 2; PRF, phenol red-free; Rho, Ras homology; ROCK, Rho-associated kinase; RPMI-5, RPMI 1640 medium with 5% fetal bovine serum; RSPO, R-spondin; SFA, serum free additive; TGF-β, transforming growth factor-β; Wnt, wingless and int-1.

2. Cancer cell line-based LCOs

Stem cells are indispensable for organoid generation (81). The ideal methodology for medical research is the use of CSCs from patient tissues to create lung tumoroids. However, ethical issues, success rates and the scarcity of specimens require consideration when using patient tissues. These factors limit the repeatability of studies to a certain extent, and may result in researchers being forced to seek alternative options for the transformation of cancer cell lines into CSCs. Researchers have used transformed CSCs to generate tumoroids, and various methods have been used to transform cancer cell lines into CSCs (82,83). In addition, researchers may be able to expand the CSC population by three-dimensional (3D) spheroid culture to acquire a sufficient number of CSCs for the establishment of tumoroids (84,85). Oshima et al (86) showed that colon cancer line cells can be successfully induced to form CSCs by the transfection of stem cell transcription factors, namely a combination of octamer-binding protein 3/4 (OCT3/4), SRY-box transcription factor 2 (SOX2) and Kruppel-like factor 4 (KLF4), known as OSK, into colon cancer cells (87). Subsequently, the authors attempted to induce lung cancer A549 cells into CSCs using this method, and colonies of OSK-A549 cells with chemoresistance, a delayed cell cycle, enhanced sphere formation ability and tumorigenicity were successfully obtained (48). Lung tumoroids were subsequently established by co-culturing the OSK-A549-colony cells with human umbilical vein endothelial cells and human mesenchymal stem cells (Fig. 1) (48). The results of hematoxylin and eosin staining and immunostaining showed that these tumoroids comprised distinctive cohesive cell nests that were analogous to lung cancer tissues (48). Gene expression analysis was conducted to explore changes in the gene expression profile during stem cell transformation (48). The results revealed that interleukin (IL)-6 was expressed at high levels in OSK-A549-colony cells compared with control cells, which increased the resistance of the OSK-A549-colony organoids to chemotherapy and facilitated the ability to construct lung tumoroids from them (48). IL-6 was also found to be expressed at high levels in the majority of patient tissue samples, independently from their gene mutation status and tumor staging (48). These findings suggest that IL-6 has the potential to become a novel therapeutic target for LUAD. This method is a feasible strategy for the generation of lung tumoroids and exploration of the mechanism of lung cancer development by driving lung CSC transformation. The use of cancer cell lines to generate LCOs has the following advantages: i) Cell lines are readily available; ii) there are few ethical issues concerning the use of cell lines to conduct medical research; and iii) repeat tests with the same cell lines can be carried out. However, there are also some disadvantages: i) Random gene mutations may be generated during long-term cell line 2D culture, leading to cell line diversity in terms of the gene profile in different laboratories; and ii) stromal, immune and nerve cells, and capillaries are absent from the tumoroid system, and consequently the interaction of cancer cells with the TME cannot be studied using this type of model. However, this is a common issue in all types of tumoroid models.

Despite their disadvantages, cancer cell line-based LCOs have certain practical utility. There are, however, some aspects of such LCOs that require further investigation: i) Whether lung cancer cells can be transformed into lung CSCs by transfection with other combinations of stem cell transcription factors such as OCT3/4, SOX2, KLF4, cellular myelocytomatosis oncogene and/or Nanog homeobox; ii) differences among stem cells induced by transfection with different combinations of stem cell transcription factors; iii) whether other lung cancer cell lines, especially SCLC cell lines, can also be transformed into lung CSCs and used to successfully establish tumoroids using this method,; and iv) whether this approach is suitable for use in drug screening. In addition, as some studies have reported that pyroptosis plays an important role in the occurrence and development of lung cancer (88-90), it is not yet known if cancer cell line-based LCOs can be used for researching the mechanism of drug-induced lung cancer cell pyroptosis. To broaden the application scope of cancer cell line-based LCOs, these unknown factors require elucidation.

3. PDX-derived LCOs

As PDXs effectively maintain the characteristics of primary tumors, including the phenotype, genetic profile and TME, they are widely used as preclinical models to explore the mechanisms of tumorigenesis and development, screen antitumor drugs and discover novel therapeutic methods (91-94). However, the high cost and time-consuming process of developing PDXs limit their usage (95,96). To use the full advantages of PDXs while avoiding their disadvantages, investigators have developed PDX-derived organoid models that have already been applied to multiple tumor types, and shown to preserve the genomic and transcriptomic profiles, protein markers and drug response of primary PDXs (94,97-99). These models have been used to study the pathogenesis of lung cancer and screen antitumor drugs (Fig. 1). A total of four methods to create PDX-derived LCOs are presented in the current review (Table I) (55,78-80). Only one of these was used to establish PDX-derived organoids of NSCLC (55), while the other three were used to generate PDX-derived organoids of SCLC (78-80).

Shi et al (55) reported a method for the creation of PDX-derived organoids of NSCLC using relatively optimal culture medium, with short- and long-term PDX-derived LCO cultures. The study found that the PDX-derived LCOs reflected the histological and cell lineage characteristics, and drug sensitivity of the parental PDXs to a large extent, in both short- and long-term culture. Even following the prolonged culture of LCOs, the mutations, copy number landscape and gene expression profiles of organoids and primary PDXs were comparable. In addition, short-term PDX-derived LCO culture was able to establish tumoroid models rapidly, and the drug sensitivity of the LCOs was consistent with that of the parental PDXs. These findings indicate that these LCOs have certain application prospects in drug screening.

Delayed diagnosis, high aggressiveness, susceptibility to relapse and poor prognosis are the basic characteristics of SCLC (4,100-102). Although SCLC is divided into four subtypes based on expression of the transcription factors achaete-scute homolog 1, neuronal differentiation 1, POU class 2 homeobox 3 and Yes1 associated transcriptional regulator (103), there is no specific and effective treatment for each subtype (104). Traditional preclinical models perform poorly in the exploration of novel markers and treatment methods for SCLC (105). Therefore, it is urgently necessary to develop more efficient preclinical models. Tumor organoids have unique advantages in that regard, and researchers have been attempting to establish SCLC organoids (71,72,77-80). Gmeiner et al (78), Chen et al (79) and Redin et al (80) have each reported methods for the culture of SCLC PDX-derived organoids to explore drug resistance mechanisms and screen specific antitumor drugs. The study by Gmeiner et al (78) indicated that overexpression of the transcription factor E2 promoter binding factor 1-3 caused the upregulation of thymidylate synthase and the increased malignancy of SCLC. To test the efficacy of thymidylate synthase inhibitors in the treatment of SCLC, the authors created SCLC PDX-derived organoid models and used them to demonstrate that SCLC is sensitive to CF10, a novel fluoropyrimidine polymer. Retinoic acid receptor-related orphan receptors (RORs), including RORα, RORβ and RORγ, participate in a variety of physiological and pathological reactions through ligand-dependent interactions with co-regulators (106). Chen et al (79) found that the high expression of RORγ improved SCLC cell growth and inhibited apoptosis, while the RORγ antagonists XY018 and GSK805 eliminated this effect in H446 and H1048 cells. These results were verified in SCLC PDX-derived organoids. Chemical library screening, and cellular thermal shift and surface plasmon resonance assays were used to identify N-hydroxyapiosporamide (N-hydap), as a potent and selective RORγ antagonist. N-hydap was more efficient at suppressing the growth and survival of cancer cells than GSK805 and XY018, which was confirmed using SCLC PDX-derived organoid models. The study by Chen et al (79) provides a new approach for the screening of targeted antitumor drugs. Redin et al (80) described another method for the generation of SCLC PDX-derived organoids, which they used to verify the curative effect of the YES1-specific inhibitor CH6953755 on SCLC. All three methods involve the use of newly established, not passaged, PDX-derived organoids to explore the mechanism of lung cancer development and screen antitumor drugs. The study findings indicate that short-term culture PDX-derived LCOs are reliable in the preclinical research of lung cancer.

The aforementioned four methods can be divided into two categories, those using relative optimal culture medium and those using a hydrogel scaffold. The methods developed by Shi et al (55), Chen et al (79) and Redin et al (80) are in the former category, while that developed by Gmeiner et al (78) is in the latter category. The details of these methods are presented in Table II.

These studies used four different digestion methods to dissociate tissues into single cells. Digestive strategies using the combination of Liberase TM, which comprises a combination of collagenase I, II and thermolysin, with TrypLE involve lower concentrations of enzymes with higher digestive efficiency compared with those that use a combination of collagenase II and TrypLE. Digestive methods using collagenase IV alone also have a higher digestive efficiency compared with those that using a combination of collagenase II and TrypLE. This may be due to collagenases I and IV having higher activity than collagenase II. Notably, treatment with TrypLE, a recombinant trypsin-like protease (107) used in two different digestive enzyme combinations, has been shown to result in a significantly higher cell viability compared with trypsin (108). Any pure collagenase is not able to effectively dissociate tissues into single cells (109). The combination of any collagenase and TrypLE may have a greater ability to generate single cells from tissues than either enzyme used alone.

Cell-Titer Glo reagents manufactured by Promega Corporation were used to measure the viability of organoids in the method reported by Chen et al (79). To avoid interference with the detection of fluorescence, phenol red-free Dulbecco's modified Eagle's medium/Ham's F 12 nutrient medium (DMEM/F12) was used.

Chen et al (79) added glutamine to the culture medium. Glutamine serves as a nitrogen source for the biosynthesis of a number of important substances, including nucleotides, nicotinamide adenine dinucleotide, the protein glycosylation precursor glucosamine-6-phosphate, and asparagine (110-112). Although mammalian cells have the ability to synthesize glutamine de novo, numerous types of cancers cannot grow and proliferate in an environment lacking exogenous glutamine (113). As DMEM/F12 contains glutamine, most methods that use DMEM/F12 to culture cells do not involve the addition of extra glutamine to the culture medium (52,71,80). More importantly, glutamine naturally breaks down to form ammonia (114), which is toxic for mammalian cell cultures (115). Therefore, excessive glutamine may be disadvantageous for cells. In one study, the authors replaced the glutamine in DMEM/F12 with L-alanyl-L-glutamine, known as Glutamax, and found it to be a suitable substitute because of its improved solubility and stability during cell culture (116).

CHIR99021 was added to the culture medium in the procedure described by Shi et al (55); this LCO culture method is the only approach discussed in the present review to include CHIR99021 in the culture medium. CHIR99021 activates the wingless and int-1 (Wnt) signaling pathway via the inhibition of glycogen synthase kinase 3β (117,118). Activation of the Wnt signaling pathway maintains the stemness of stem cells and organoid formation (119,120). CHIR99021 has been used to generate organoids from other types of cancer tissue, such as bladder cancer tissue (121). Shi et al (55) reported that their method of establishing LCOs has a higher success rate (88%) than other methods, which may be due to the addition of CHIR99021. However, this hypothesis requires verification by additional experiments.

Gmeiner et al (78) used HyStem-HP Hydrogel as a 3D scaffold for organoid culture. The type of 3D scaffold has numerous desirable features, including high transparency and cellular affinity, and being easy to standardize. These suggest it has application prospects in the field of organoid culture.

Redin et al (80) added 10% HyClone serum to the LCO culture medium. This is the only method covered in the present review that involves supplementation of the LCO culture medium with serum. Although some studies have demonstrated that the proper addition of fetal bovine serum (FBS) enhances organoid formation (122-124), FBS contains unknown components that might cause the organoid culture to fail (125). Therefore, most methods of organoid culture do not include FBS. Nonetheless, the role of FBS in organoid culture is worthy of exploration.

Redin et al (80) added gastrin I to their culture medium. Gastrin is an important growth factor for digestive system tumors (126), which can prolong the survival time of digestive system tumoroids (127-134), and is often added to the medium used to culture them. However, it is not clear whether gastrin addition is beneficial to lung tumoroid culture.

PDX-derived LCOs have certain advantages compared with cancer cell line-based or patient-derived LCOs: i) The phenotype, genetic profiles and heterogeneity of parental tumors are more effectively preserved in PDX-derived LCOs than in lung cancer cell line-based LCOs; ii) compared with patient samples, PDXs are easier to obtain and can undergo long-term expansion by passaging, which ensures the repeatability and sustainability of experiments; iii) the establishment of PDX-derived LCOs has lower requirements for ethical approval compared with patient-derived LCOs; and iv) PDX-derived LCOs are easier to develop than patient-derived LCOs (55). However, there is a clear disadvantage of PDX-derived LCOs, in addition to the defects common to all LCOs: PDX-derived LCOs may be contaminated by mouse cells during short-term culture, which is likely to affect the phenotypic identification of tumors, genotype analysis or antitumor drug screening. These factors may restrict their application.

4. Patient-derived LCOs

Patient-derived LCOs are ideal for researching the mechanism of initiation, development and drug resistance of lung cancer, and for exploring new biomarkers, antitumor drugs and treatment protocols. The samples used to create patient-derived LCOs mainly originate from cancer tissue, including that obtained during surgery or biopsy, or from the exfoliative tumor cells present in pleural effusion (PE) (Fig. 1). In the current review, 13 methods used to generate LCOs from patient samples are presented (Tables I, II, SI and SII) (49-53,55,57,66,68-71,76). These involve all six categories of methods used to establish organoids. The success rates of organoid and pure LCO establishment using these methods are variable (57,60), which might influence subsequent mechanistic research and drug screening. Therefore, it is necessary to compare and analyze the details of the methods investigated in these studies to select the ideal method for the culture of LCOs.

The relative optimal culture medium method is the most popular, and the most representative method among all those reported is that described by Sachs et al (57). Although different laboratories generated LCOs via similar methods, the success rate of organoid establishment ranged from 41 to 88%. The success rate range of pure LCO establishment was also diverse, ranging from 7 to 92.7% (60,63). Dijkstra et al (60) reported lower success rates of organoid establishment (41%) and LCO establishment (17%) compared with other studies (57,63). Patients with stage IV adenocarcinoma accounted for 78% of all patients in the study by Dijkstra et al suggesting that the low success rates might be due to the degree of tumor malignancy. Kim et al (63) used 77 malignant effusion samples, three brain metastasis samples, a single bone metastasis sample and two primary lung tumor samples from patients with advanced LUAD to successfully generate LCOs, and the success rates of organoid and pure LCO establishment were 83 and 92.7%, respectively. According to these results, it can be concluded that samples from malignant effusions or metastatic foci easily form pure cancer organoids. This may be due to the airway organoid (AO) culture medium being more suitable for normal airway epithelial cell growth and samples from malignant effusions or metastasis foci being less easily contaminated by normal epithelial cells than those from primary lung tissues. If normal epithelial cells are not removed during the pretreatment process, cancer cells are rapidly overtaken by normal epithelial cells during organoid culture, leading to failure of the cancer organoid culture (135,136). However, surgically resected tumor tissues are a prominent source of material for LCO culture. To make full use of these tissues, a number of researchers have sought to devise improved culture methods. Kim et al (71) developed a new method using LCO suboptimal medium free of Wnt3a, Noggin and A83-01 to culture lung tumoroids, which improved the growth of cancer organoids and inhibited that of normal epithelial organoids. When surgically resected tumor tissues were used to establish organoids, the success rates were 58-87% (71,73), which were comparable with the 41-88% success rates of the method described by Sachs et al (57). Moreover, a success rate of pure LCO establishment of 71% was observed (73), which is higher than the 17% reported by Dijkstra et al (60). Therefore, it is speculated that LCO suboptimal medium may be superior to AO medium in cancer organoid culture. It is noteworthy that that the pretreatment method used by Kim et al (71) differed from that used by Sachs et al (57). Sachs et al (57) used only collagenase to digest tissue. By contrast, Kim et al (71) used DNase and collagenase/dispase to isolate single lung cancer cells. Gohi et al (137) reported that digestion using a combination of collagenase and DNase is conducive to the maintenance of cell surface antigen integrity and cell activity. These findings are meaningful for subsequent cancer organoid culture. Therefore, the pretreatment method reported by Kim et al (71) may partially contribute to the high success rates of organoid establishment and pure LCO establishment that were obtained. The importance of pretreatment methods was supported by Hu et al (70), who found mechanical processing to be more beneficial for tumor organoid formation than enzymatic digestion, with the latter being beneficial for normal organoid formation. Moreover, their study revealed that a medium without R-spondin and Noggin is conducive to the establishment of pure LCOs. Overall, it may be easier to generate higher purity LCOs with an acceptable success rate by the use of mechanical digestion and the suboptimal culture medium method (Tables I; SI).

In addition to the success rates of organoid establishment and pure LCO establishment, researchers have evaluated the sustainability of LCOs, which includes the expansion and efficient reconstitution of cryopreserved LCOs. Short-term organoid culture is sufficient to perform drug screening for patients whose cancer tissue has been used to generate LCOs. However, the long-term expansion of tumor organoids and efficient reconstitution of cryopreserved organoids are necessary to provide sufficient tumor organoids for the establishment of LCO biobanks for use in long-term studies, such as those for antitumor drug discovery, the elucidation of drug resistance mechanisms and improvement of treatment protocols. Four methods of LCO long-term culture are covered in the present review (55,57,71,76). In terms of time, different definitions of tumoroid long-term culture have been proposed. However, as regards passage number, the definitions of tumoroid long-term culture are similar (>10 passages). The results of the study by Yokota et al (65) showed that the AO medium is a more robust tumor organoid culture medium than the media used by Kim et al (71) and Shi et al (55). While AO medium is suitable for the growth of all lung epithelial cells, some LCOs with particular mutations may also be long-term expanded in AO medium (65). The study of Yokota et al (65) revealed that activation of the Wnt/β-catenin pathway is a prerequisite for the maintenance of certain LCOs (TPM3-ROS1; TP53K120Sfs*3) in long-term culture. This was verified by Choi et al (76), who found that Wnt3A and R-spondin1 do not promote SCLC tumoroid formation but play important roles in the long-term culture of SCLC tumoroids. Therefore, if the specific mutations of lung cancer tissues are unknown, AO medium appears to be a suitable choice for lung tumoroid long-term culture. Nevertheless, the removal of activators of the Wnt/β-catenin pathway did not influence other lung tumoroids in long-term culture, for example LCOs (BRAFG469A; TP53T155P) (65). These results are consistent with those of Kim et al (71), who found that most LCOs could be long-term expanded in LCO suboptimal medium. In general, the efficient reconstitution of cryopreserved LCOs is essential for the establishment of LCO biobanks. The cell viability of LCOs before cryopreservation and the methods of cryopreservation used determine the success or failure of recovery. Although LCO suboptimal medium contains fewer reagents than relative optimal culture medium, Kim et al (71) reported that tumoroids cultured in the former medium had a high recovery success rate (70%) after freezing. This indicates that LCO suboptimal medium can effectively sustain cell viability and provide high recovery success rates. In a study by Shi et al (55), long-term culture was achieved for 15% of NSCLC organoids, and these organoids had good cell viability, with all of them being recoverable after >1 year of cryopreservation and continued passaging. Hu et al (70) chose tumoroids with fast growth rates and short generation intervals for cryopreservation. Of the five lung tumoroids used, four were successfully cryopreserved and thawed. These results showed that cell viability is a critical factor in the efficient reconstitution of cryopreserved LCOs. Therefore, it is necessary to screen for high-viability LCOs before cryopreservation.

Jung et al (72) developed a one-stop microfluidic device for use in tumoroid culture and drug screening. The microphysiological system allows the quantity of lung tumoroids and concentration of drugs to be controlled, which facilitates the standardization of drug screening. Tamura et al (66) reported a suspension culture method which they used to generate patient-derived tumor organoids for the Fukushima Translational Research Project (Fukushima-PDOs). However, a long culture time of 3-6 months was required to successfully establish Fukushima-PDOs, which is unsuitable for the rapid screening of drugs for patients who have provided cancer tissues. The Fukushima-PDOs were found to be able to expand long term, which may contribute to the establishment of LCO biobanks for use in long-term studies. As uniform cell sizes are important for high-throughput screening, the authors used CellPet FT to mince the tumoroids and obtain organoids of similar sizes. The study by Dijkstra et al (58) described how tumor-reactive T cells can be produced through the coculture of peripheral blood lymphocytes and tumor organoids. These T cells specifically kill tumoroids, and the method enables T-cell-based therapies and interactions between T cells and tumor cells to be researched in vitro.

In general, patient-derived LCOs are improved preclinical models is comparison with other traditional models for the following reasons: i) Patient-derived LCOs have excellent fidelity because they are directly structured with patient tumor tissue or PE; ii) patient-derived LCOs can be generated in weeks or even days, and short-term cultured organoids are able to predict the responses of patients to antitumor drugs. The integrated superhydrophobic microwell array chip (InSMAR-chip) shortens the time for drug screening to 1 week, thereby saving precious time for patients requiring treatment (70); and iii) the combination of patient-derived LCOs and microfluidic devices can standardize drug screening to help clinicians in the formulation of appropriate medication plans. Patient-derived LCOs also have certain limitations: i) As samples originate from patients with lung cancer, they are of high value and if the establishment of patient-derived LCO fails, it is challenging to compensate for the loss; ii) although patient-derived LCOs can be long-term expanded, immortality of patient-derived LCOs in vitro has not yet been achieved.

A number of issues remain to be resolved including: i) How the optimization of patient-derived LCO culture methods can be achieved to eliminate normal organic contamination and maintain long-term lung tumoroid culture and even achieve immortality; ii) how the standardization of patient-derived LCO culture methods can be accomplished; iii) how the creation of a co-culture system of patient-derived LCOs and the microenvironment can be realized; and iv) how the success rate of LCO establishment from patient biopsy samples can be enhanced.

5. Conclusions

LCOs derived from three major resources greatly promote preclinical research, with applications including the exploration of cancer mechanisms, searching for novel tumor biomarkers, screening of antitumor drugs and improving treatment plans. The three major types of lung tumoroids, which are cancer cell line-based LCOs, PDX-derived LCOs and patient-derived LCOs, are complementary, and the results of studies on the three major types of lung tumoroids have provided a more comprehensive understanding of the pathogenesis of lung cancer (48,50,52). Furthermore, there are six different categories of methods that can be used to establish LCOs. Appropriate models or methods can be selected based on the requirements of researchers. However, the present review puts forth several suggestions: i) AO medium is a more robust tumor organoid culture medium than other media used for the long-term culture of LCOs; ii) the mechanical digestion and suboptimal culture medium method is more conducive to the establishment of pure LCOs, while the mechanical dissociation method is beneficial for the passage of organoids; iii) malignant PE samples appear to have a high tendency to establish pure LCOs; and iv) the use of CellPet FT to process organoids is beneficial for standardization in high-throughput screening. The achievement of standardization is important in lung tumoroid culture. The combination of bioengineering technology, including microfluidic devices and the InSMAR-chip, and lung tumoroid culture has accelerated the standardization of lung tumoroid applications to some degree. It is hypothesized that with technical progress, lung tumoroids will have broader application prospects in the future.

Supplementary Material

Genetic background of samples and application area of methods for the establishment of lung cancer organoids.
Characteristics of specimen pre-treatment methods for the establishment of lung cancer organoids, scaffold and enzymes of passaging.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

QZ wrote the manuscript and prepared the figures. MZ conceived and designed the study. MZ revised the manuscript critically for important intellectual content. Both authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

2 

Wang M, Herbst RS and Boshoff C: Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 27:1345–1356. 2021.PubMed/NCBI View Article : Google Scholar

3 

Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, et al: The 2015 World Health Organization Classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 10:1243–1260. 2015.PubMed/NCBI View Article : Google Scholar

4 

Rudin CM, Brambilla E, Faivre-Finn C and Sage J: Small-cell lung cancer. Nat Rev Dis Primers. 7(3)2021.PubMed/NCBI View Article : Google Scholar

5 

Prabavathy D and Ramadoss N: Heterogeneity of small cell lung cancer stem cells. Adv Exp Med Biol. 1139:41–57. 2019.PubMed/NCBI View Article : Google Scholar

6 

Zhang Y, Chang L, Yang Y, Fang W, Guan Y, Wu A, Hong S, Zhou H, Chen G, Chen X, et al: Intratumor heterogeneity comparison among different subtypes of non-small-cell lung cancer through multi-region tissue and matched ctDNA sequencing. Mol Cancer. 18(7)2019.PubMed/NCBI View Article : Google Scholar

7 

de Sousa VML and Carvalho L: Heterogeneity in lung cancer. Pathobiology. 85:96–107. 2018.PubMed/NCBI View Article : Google Scholar

8 

Krohn A, Ahrens T, Yalcin A, Plönes T, Wehrle J, Taromi S, Wollner S, Follo M, Brabletz T, Mani SA, et al: Tumor cell heterogeneity in small cell lung cancer (SCLC): Phenotypical and functional differences associated with Epithelial-Mesenchymal Transition (EMT) and DNA methylation changes. PLoS One. 9(e100249)2014.PubMed/NCBI View Article : Google Scholar

9 

Liao H, Luo X, Huang Y, Yang X, Zheng Y, Qin X, Tan J, Shen P, Tian R, Cai W, et al: Mining the prognostic role of DNA methylation heterogeneity in lung adenocarcinoma. Dis Markers. 2022(9389372)2022.PubMed/NCBI View Article : Google Scholar

10 

Arora L, Kalia M, Dasgupta S, Singh N, Verma AK and Pal D: Development of a Multicellular 3D tumor model to study cellular heterogeneity and plasticity in NSCLC tumor microenvironment. Front Oncol. 12(881207)2022.PubMed/NCBI View Article : Google Scholar

11 

Wang Q, Li M, Yang M, Yang Y, Song F, Zhang W, Li X and Chen K: Analysis of immune-related signatures of lung adenocarcinoma identified two distinct subtypes: Implications for immune checkpoint blockade therapy. Aging (Albany NY). 12:3312–3339. 2020.PubMed/NCBI View Article : Google Scholar

12 

Liu LP, Lu L, Zhao QQ, Kou QJ, Jiang ZZ, Gui R, Luo YW and Zhao QY: Identification and validation of the pyroptosis-related molecular subtypes of lung adenocarcinoma by bioinformatics and machine learning. Front Cell Dev Biol. 9(756340)2021.PubMed/NCBI View Article : Google Scholar

13 

Kogure Y, Iwasawa S, Saka H, Hamamoto Y, Kada A, Hashimoto H, Atagi S, Takiguchi Y, Ebi N, Inoue A, et al: Efficacy and safety of carboplatin with nab-paclitaxel versus docetaxel in older patients with squamous non-small-cell lung cancer (CAPITAL): A randomised, multicentre, open-label, phase 3 trial. Lancet Healthy Longev. 2:e791–e800. 2021.PubMed/NCBI View Article : Google Scholar

14 

Spigel DR, Vicente D, Ciuleanu TE, Gettinger S, Peters S, Horn L, Audigier-Valette C, Pardo Aranda N, Juan-Vidal O, Cheng Y, et al: Second-line nivolumab in relapsed small-cell lung cancer: CheckMate 331(☆). Ann Oncol. 32:631–641. 2021.PubMed/NCBI View Article : Google Scholar

15 

Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, Pastorino S, Purow BW, Christopher N, Zhang W, et al: Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 9:391–403. 2006.PubMed/NCBI View Article : Google Scholar

16 

Muff R, Rath P, Ram Kumar RM, Husmann K, Born W, Baudis M and Fuchs B: Genomic instability of osteosarcoma cell lines in culture: Impact on the prediction of metastasis relevant genes. PLoS One. 10(e0125611)2015.PubMed/NCBI View Article : Google Scholar

17 

Kasai F, Hirayama N, Ozawa M, Iemura M and Kohara A: Changes of heterogeneous cell populations in the Ishikawa cell line during long-term culture: Proposal for an in vitro clonal evolution model of tumor cells. Genomics. 107:259–266. 2016.PubMed/NCBI View Article : Google Scholar

18 

Bahcecioglu G, Basara G, Ellis BW, Ren X and Zorlutuna P: Breast cancer models: Engineering the tumor microenvironment. Acta Biomater. 106:1–21. 2020.PubMed/NCBI View Article : Google Scholar

19 

Nolan JC, Frawley T, Tighe J, Soh H, Curtin C and Piskareva O: Preclinical models for neuroblastoma: Advances and challenges. Cancer Lett. 474:53–62. 2020.PubMed/NCBI View Article : Google Scholar

20 

Lee SW, Kwak HS, Kang MH, Park YY and Jeong GS: Fibroblast-associated tumour microenvironment induces vascular structure-networked tumouroid. Sci Rep. 8(2365)2018.PubMed/NCBI View Article : Google Scholar

21 

Salinas-Vera YM, Valdés J, Hidalgo-Miranda A, Cisneros-Villanueva M, Marchat LA, Nuñez-Olvera SI, Ramos-Payán R, Pérez-Plasencia C, Arriaga-Pizano LA, Prieto-Chávez JL, et al: Three-dimensional organotypic cultures reshape the microRNAs transcriptional program in breast cancer cells. Cancers (Basel). 14(2490)2022.PubMed/NCBI View Article : Google Scholar

22 

Jo Y, Choi N, Kim K, Koo HJ, Choi J and Kim HN: Chemoresistance of cancer cells: Requirements of tumor microenvironment-mimicking in vitro models in anti-cancer drug development. Theranostics. 8:5259–5275. 2018.PubMed/NCBI View Article : Google Scholar

23 

Shang M, Soon RH, Lim CT, Khoo BL and Han J: Microfluidic modelling of the tumor microenvironment for anti-cancer drug development. Lab Chip. 19:369–386. 2019.PubMed/NCBI View Article : Google Scholar

24 

Siolas D and Hannon GJ: Patient-derived tumor xenografts: Transforming clinical samples into mouse models. Cancer Res. 73:5315–5319. 2013.PubMed/NCBI View Article : Google Scholar

25 

Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, Wu R, Brahmbhatt S, Mo F, Jong L, et al: High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res. 74:1272–1283. 2014.PubMed/NCBI View Article : Google Scholar

26 

Xiao T, Li W, Wang X, Xu H, Yang J, Wu Q, Huang Y, Geradts J, Jiang P, Fei T, et al: Estrogen-regulated feedback loop limits the efficacy of estrogen receptor-targeted breast cancer therapy. Proc Natl Acad Sci USA. 115:7869–7878. 2018.PubMed/NCBI View Article : Google Scholar

27 

Yoshida GJ: Applications of patient-derived tumor xenograft models and tumor organoids. J Hematol Oncol. 13(4)2020.PubMed/NCBI View Article : Google Scholar

28 

Li Z, Zheng W, Wang H, Cheng Y, Fang Y, Wu F, Sun G, Sun G, Lv C and Hui B: Application of animal models in cancer research: Recent progress and future prospects. Cancer Manag Res. 13:2455–2475. 2021.PubMed/NCBI View Article : Google Scholar

29 

Kuwata T, Yanagihara K, Iino Y, Komatsu T, Ochiai A, Sekine S, Taniguchi H, Katai H, Kinoshita T and Ohtsu A: Establishment of novel gastric cancer patient-derived xenografts and cell lines: Pathological comparison between primary tumor, patient-derived, and cell-line derived xenografts. Cells. 8(585)2019.PubMed/NCBI View Article : Google Scholar

30 

Recondo G, Mahjoubi L, Maillard A, Loriot Y, Bigot L, Facchinetti F, Bahleda R, Gazzah A, Hollebecque A, Mezquita L, et al: Feasibility and first reports of the MATCH-R repeated biopsy trial at Gustave Roussy. NPJ Precis Oncol. 4(27)2020.PubMed/NCBI View Article : Google Scholar

31 

Heo EJ, Cho YJ, Cho WC, Hong JE, Jeon HK, Oh DY, Choi YL, Song SY, Choi JJ, Bae DS, et al: Patient-derived xenograft models of epithelial ovarian cancer for preclinical studies. Cancer Res Treat. 49:915–926. 2017.PubMed/NCBI View Article : Google Scholar

32 

Tucker ER, George S, Angelini P, Bruna A and Chesler L: The promise of Patient-derived preclinical models to accelerate the implementation of personalised medicine for children with neuroblastoma. J Pers Med. 11(248)2021.PubMed/NCBI View Article : Google Scholar

33 

Zhuang Y, Grainger JM, Vedell PT, Yu J, Moyer AM, Gao H, Fan XY, Qin S, Liu D, Kalari KR, et al: Establishment and characterization of immortalized human breast cancer cell lines from breast cancer patient-derived xenografts (PDX). NPJ Breast Cancer. 7(79)2021.PubMed/NCBI View Article : Google Scholar

34 

Martinez-Garcia R, Juan D, Rausell A, Muñoz M, Baños N, Menéndez C, Lopez-Casas PP, Rico D, Valencia A and Hidalgo M: Transcriptional dissection of pancreatic tumors engrafted in mice. Genome Med. 6(27)2014.PubMed/NCBI View Article : Google Scholar

35 

Hakuno SK, Michiels E, Kuhlemaijer EB, Rooman I, Hawinkels L and Slingerland M: Multicellular modelling of Difficult-to-Treat gastrointestinal cancers: Current possibilities and challenges. Int J Mol Sci. 23(3147)2022.PubMed/NCBI View Article : Google Scholar

36 

Jung J, Seol HS and Chang S: The generation and application of Patient-derived xenograft model for cancer research. Cancer Res Treat. 50:1–10. 2018.PubMed/NCBI View Article : Google Scholar

37 

Meraz IM, Majidi M, Meng F, Shao R, Ha MJ, Neri S, Fang B, Lin SH, Tinkey PT, Shpall EJ, et al: An Improved Patient-derived xenograft humanized mouse model for evaluation of lung cancer immune responses. Cancer Immunol Res. 7:1267–1279. 2019.PubMed/NCBI View Article : Google Scholar

38 

Ganesh K, Wu C, O'Rourke KP, Szeglin BC, Zheng Y, Sauvé CG, Adileh M, Wasserman I, Marco MR, Kim AS, et al: A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 25:1607–1614. 2019.PubMed/NCBI View Article : Google Scholar

39 

Xia X, Li F, He J, Aji R and Gao D: Organoid technology in cancer precision medicine. Cancer Lett. 457:20–27. 2019.PubMed/NCBI View Article : Google Scholar

40 

Brassard JA and Lutolf MP: Engineering stem cell Self-organization to Build better organoids. Cell Stem Cell. 24:860–876. 2019.PubMed/NCBI View Article : Google Scholar

41 

Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, Xia F, Fu G, Deng Y, Pan M, et al: Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 26:17–26.e6. 2020.PubMed/NCBI View Article : Google Scholar

42 

Lõhmussaar K, Oka R, Espejo Valle-Inclan J, Smits MHH, Wardak H, Korving J, Begthel H, Proost N, van de Ven M, Kranenburg OW, et al: Patient-derived organoids model cervical tissue dynamics and viral oncogenesis in cervical cancer. Cell Stem Cell. 28:1380–1396.e6. 2021.PubMed/NCBI View Article : Google Scholar

43 

Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, Chua CW, Barlow LJ, Kandoth C, Williams AB, et al: Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 173:515–528.e17. 2018.PubMed/NCBI View Article : Google Scholar

44 

Servant R, Garioni M, Vlajnic T, Blind M, Pueschel H, Müller DC, Zellweger T, Templeton AJ, Garofoli A, Maletti S, et al: Prostate cancer patient-derived organoids: Detailed outcome from a prospective cohort of 81 clinical specimens. J Pathol. 254:543–555. 2021.PubMed/NCBI View Article : Google Scholar

45 

Chen P, Zhang X, Ding R, Yang L, Lyu X, Zeng J, Lei JH, Wang L, Bi J, Shao N, et al: Patient-derived organoids can guide personalized-therapies for patients with advanced breast cancer. Adv Sci (Weinh). 8(e2101176)2021.PubMed/NCBI View Article : Google Scholar

46 

Seidlitz T and Stange DE: Gastrointestinal cancer organoids-applications in basic and translational cancer research. Exp Mol Med. 53:1459–1470. 2021.PubMed/NCBI View Article : Google Scholar

47 

Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarró LM, Bradshaw CR, Allen GE, Arnes-Benito R, Sidorova O, Gaspersz MP, et al: Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med. 23:1424–1435. 2017.PubMed/NCBI View Article : Google Scholar

48 

Ogawa H, Koyanagi-Aoi M, Otani K, Zen Y, Maniwa Y and Aoi T: Interleukin-6 blockade attenuates lung cancer tissue construction integrated by cancer stem cells. Sci Rep. 7(12317)2017.PubMed/NCBI View Article : Google Scholar

49 

Li YF, Gao Y, Liang BW, Cao XQ, Sun ZJ, Yu JH, Liu ZD and Han Y: Patient-derived organoids of non-small cells lung cancer and their application for drug screening. Neoplasma. 67:430–437. 2020.PubMed/NCBI View Article : Google Scholar

50 

Han Y, Lee T, He Y, Raman R, Irizarry A, Martin ML and Giaccone G: The regulation of CD73 in non-small cell lung cancer. Eur J Cancer. 170:91–102. 2022.PubMed/NCBI View Article : Google Scholar

51 

Wang Y, Jiang T, Qin Z, Jiang J, Wang Q, Yang S, Rivard C, Gao G, Ng TL, Tu MM, et al: HER2 exon 20 insertions in non-small-cell lung cancer are sensitive to the irreversible pan-HER receptor tyrosine kinase inhibitor pyrotinib. Ann Oncol. 30:447–455. 2019.PubMed/NCBI View Article : Google Scholar

52 

Zhang P, He B, Cai Q, Tu G, Peng X, Zhao Z, Peng W, Yu F, Wang M, Tao Y, et al: Decreased IL-6 and NK cells in Early-stage lung adenocarcinoma presenting as ground-glass opacity. Front Oncol. 11(705888)2021.PubMed/NCBI View Article : Google Scholar

53 

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. iScience. 23(101411)2020.PubMed/NCBI View Article : Google Scholar

54 

Li Z, Yu L, Chen D, Meng Z, Chen W and Huang W: Protocol for generation of lung adenocarcinoma organoids from clinical samples. STAR Protoc. 2(100239)2021.PubMed/NCBI View Article : Google Scholar

55 

Shi R, Radulovich N, Ng C, Liu N, Notsuda H, Cabanero M, Martins-Filho SN, Raghavan V, Li Q, Mer AS, et al: Organoid cultures as preclinical models of Non-small cell lung cancer. Clin Cancer Res. 26:1162–1174. 2020.PubMed/NCBI View Article : Google Scholar

56 

Liu T, Guo W, Luo K, Li L, Dong J, Liu M, Shi X, Wang Z, Zhang J, Yin J, et al: Smoke-induced SAV1 gene promoter hypermethylation disrupts YAP negative feedback and promotes malignant progression of non-small cell lung cancer. Int J Biol Sci. 18:4497–4512. 2022.PubMed/NCBI View Article : Google Scholar

57 

Sachs N, Papaspyropoulos A, Zomer-van Ommen DD, Heo I, Böttinger L, Klay D, Weeber F, Huelsz-Prince G, Iakobachvili N, Amatngalim GD, et al: Long-term expanding human airway organoids for disease modeling. EMBO J. 38(e100300)2019.PubMed/NCBI View Article : Google Scholar

58 

Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, Slagter M, van der Velden DL, Kaing S, Kelderman S, et al: Generation of Tumor-Reactive T cells by Co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 174:1586–1598.e12. 2018.PubMed/NCBI View Article : Google Scholar

59 

Cattaneo CM, Dijkstra KK, Fanchi LF, Kelderman S, Kaing S, van Rooij N, van den Brink S, Schumacher TN and Voest EE: Tumor organoid-T-cell coculture systems. Nat Protoc. 15:15–39. 2020.PubMed/NCBI View Article : Google Scholar

60 

Dijkstra KK, Monkhorst K, Schipper LJ, Hartemink KJ, Smit EF, Kaing S, de Groot R, Wolkers MC, Clevers H, Cuppen E, et al: Challenges in establishing pure lung cancer organoids limit their utility for personalized medicine. Cell Rep. 31(107588)2020.PubMed/NCBI View Article : Google Scholar

61 

Bie Y, Wang J, Xiong L, Wang D, Liao J, Zhang Y and Lin H: Lung adenocarcinoma organoids harboring EGFR 19Del and L643V double mutations respond to osimertinib and gefitinib: A case report. Medicine (Baltimore). 100(e24793)2021.PubMed/NCBI View Article : Google Scholar

62 

Sándor GO, Soós A, Lörincz P, Rojkó L, Harkó T, Bogyó L, Tölgyes T, Bursics A, Buzás EI, Moldvay J, et al: Wnt activity and cell proliferation are coupled to extracellular vesicle release in multiple organoid models. Front Cell Dev Biol. 9(670825)2021.PubMed/NCBI View Article : Google Scholar

63 

Kim SY, Kim SM, Lim S, Lee JY, Choi SJ, Yang SD, Yun MR, Kim CG, Gu SR, Park C, et al: Modeling clinical responses to targeted therapies by patient-derived organoids of advanced lung adenocarcinoma. Clin Cancer Res. 27:4397–4409. 2021.PubMed/NCBI View Article : Google Scholar

64 

Padmanabhan J, Saha B, Powell C, Mo Q, Perez BA and Chellappan S: Inhibitors targeting CDK9 show high efficacy against osimertinib and AMG510 resistant lung adenocarcinoma cells. Cancers (Basel). 13(3909)2021.PubMed/NCBI View Article : Google Scholar

65 

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. 5(29)2021.PubMed/NCBI View Article : Google Scholar

66 

Tamura H, Higa A, Hoshi H, Hiyama G, Takahashi N, Ryufuku M, Morisawa G, Yanagisawa Y, Ito E, Imai JI, et al: Evaluation of anticancer agents using patient-derived tumor organoids characteristically similar to source tissues. Oncol Rep. 40:635–646. 2018.PubMed/NCBI View Article : Google Scholar

67 

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. 8(481)2019.PubMed/NCBI View Article : Google Scholar

68 

Ma X, Yang S, Jiang H, Wang Y and Xiang Z: Transcriptomic analysis of tumor tissues and organoids reveals the crucial genes regulating the proliferation of lung adenocarcinoma. J Transl Med. 19(368)2021.PubMed/NCBI View Article : Google Scholar

69 

Mazzocchi A, Dominijanni A and Soker S: Pleural effusion aspirate for use in 3D lung cancer modeling and chemotherapy screening. Methods Mol Biol. 2394:471–483. 2022.PubMed/NCBI View Article : Google Scholar

70 

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. 12(2581)2021.PubMed/NCBI View Article : Google Scholar

71 

Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, Jung DJ, Shin TH, Jeong GS, Kim DK, et al: Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 10(3991)2019.PubMed/NCBI View Article : Google Scholar

72 

Jung DJ, Shin TH, Kim M, Sung CO, Jang SJ and Jeong GS: A one-stop microfluidic-based lung cancer organoid culture platform for testing drug sensitivity. Lab Chip. 19:2854–2865. 2019.PubMed/NCBI View Article : Google Scholar

73 

Chen JH, Chu XP, Zhang JT, Nie Q, Tang WF, Su J, Yan HH, Zheng HP, Chen ZX, Chen X, et al: Genomic characteristics and drug screening among organoids derived from non-small cell lung cancer patients. Thorac Cancer. 11:2279–2290. 2020.PubMed/NCBI View Article : Google Scholar

74 

Chen X, Liu Y, Wang Y, Wang C, Chen X, Xiong Y, Liu L, Yuan X, Tang H, Shu C, et al: CYP4F2-catalyzed metabolism of arachidonic acid promotes stromal cell-mediated immunosuppression in non-small cell lung cancer. Cancer Res. 82:4016–4030. 2022.PubMed/NCBI View Article : Google Scholar

75 

Peng KC, Su JW, Xie Z, Wang HM, Fang MM, Li WF, Chen YQ, Guan XH, Su J, Yan HH, et al: Clinical outcomes of EGFR+/METamp+ vs. EGFR+/METamp-untreated patients with advanced non-small cell lung cancer. Thorac Cancer. 13:1619–1630. 2022.PubMed/NCBI View Article : Google Scholar

76 

Choi SY, Cho YH, Kim DS, Ji W, Choi CM, Lee JC, Rho JK and Jeong GS: Establishment and long-term expansion of small cell lung cancer patient-derived tumor organoids. Int J Mol Sci. 22(1349)2021.PubMed/NCBI View Article : Google Scholar

77 

Choi YJ, Lee H, Kim DS, Kim DH, Kang MH, Cho YH, Choi CM, Yoo J, Lee KO, Choi EK, et al: Discovery of a novel CDK7 inhibitor YPN-005 in small cell lung cancer. Eur J Pharmacol. 907(174298)2021.PubMed/NCBI View Article : Google Scholar

78 

Gmeiner WH, Miller LD, Chou JW, Dominijanni A, Mutkus L, Marini F, Ruiz J, Dotson T, Thomas KW, Parks G, et al: Dysregulated pyrimidine biosynthesis contributes to 5-FU resistance in SCLC Patient-derived organoids but response to a novel polymeric fluoropyrimidine, CF10. Cancers (Basel). 12(788)2020.PubMed/NCBI View Article : Google Scholar

79 

Chen J, Hu Y, Zhang J, Wang Q, Wu X, Huang W, Wang Q, Cai G, Wang H, Ou T, et al: Therapeutic targeting RORγ with natural product N-hydroxyapiosporamide for small cell lung cancer by reprogramming neuroendocrine fate. Pharmacol Res. 178(106160)2022.PubMed/NCBI View Article : Google Scholar

80 

Redin E, Garrido-Martin EM, Valencia K, Redrado M, Solorzano JL, Carias R, Echepare M, Exposito F, Serrano D, Ferrer I, et al: YES1 is a druggable oncogenic target in Small Cell Lung Cancer. J Thorac Oncol. 17:1387–1403. 2022.PubMed/NCBI View Article : Google Scholar

81 

Lancaster MA and Knoblich JA: Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 345(1247125)2014.PubMed/NCBI View Article : Google Scholar

82 

Suzuka J, Tsuda M, Wang L, Kohsaka S, Kishida K, Semba S, Sugino H, Aburatani S, Frauenlob M, Kurokawa T, et al: Rapid reprogramming of tumour cells into cancer stem cells on double-network hydrogels. Nat Biomed Eng. 5:914–925. 2021.PubMed/NCBI View Article : Google Scholar

83 

Xu Z, Jia Y, Huang X, Feng N and Li Y: Rapid induction of pancreatic cancer cells to cancer stem cells via heterochromatin modulation. Cell Cycle. 17:1487–1495. 2018.PubMed/NCBI View Article : Google Scholar

84 

Ishiguro T, Ohata H, Sato A, Yamawaki K, Enomoto T and Okamoto K: Tumor-derived spheroids: Relevance to cancer stem cells and clinical applications. Cancer Sci. 108:283–289. 2017.PubMed/NCBI View Article : Google Scholar

85 

Weiswald LB, Bellet D and Dangles-Marie V: Spherical cancer models in tumor biology. Neoplasia. 17:1–15. 2015.PubMed/NCBI View Article : Google Scholar

86 

Oshima N, Yamada Y, Nagayama S, Kawada K, Hasegawa S, Okabe H, Sakai Y and Aoi T: Induction of cancer stem cell properties in colon cancer cells by defined factors. PLoS One. 9(e101735)2014.PubMed/NCBI View Article : Google Scholar

87 

Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Orlov YL, Zhang W, Jiang J, et al: Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell. 133:1106–1117. 2008.PubMed/NCBI View Article : Google Scholar

88 

Zhang CC, Li CG, Wang YF, Xu LH, He XH, Zeng QZ, Zeng CY, Mai FY, Hu B and Ouyang DY: Chemotherapeutic paclitaxel and cisplatin differentially induce pyroptosis in A549 lung cancer cells via caspase-3/GSDME activation. Apoptosis. 24:312–325. 2019.PubMed/NCBI View Article : Google Scholar

89 

Long K, Gu L, Li L, Zhang Z, Li E, Zhang Y, He L, Pan F, Guo Z and Hu Z: Small-molecule inhibition of APE1 induces apoptosis, pyroptosis, and necroptosis in non-small cell lung cancer. Cell Death Dis. 12(503)2021.PubMed/NCBI View Article : Google Scholar

90 

Song J, Sun Y, Cao H, Liu Z, Xi L, Dong C, Yang R and Shi Y: A novel pyroptosis-related lncRNA signature for prognostic prediction in patients with lung adenocarcinoma. Bioengineered. 12:5932–5949. 2021.PubMed/NCBI View Article : Google Scholar

91 

Coleman CN, Higgins GS, Brown JM, Baumann M, Kirsch DG, Willers H, Prasanna PG, Dewhirst MW, Bernhard EJ and Ahmed MM: Improving the predictive value of preclinical studies in support of radiotherapy clinical trials. Clin Cancer Res. 22:3138–3147. 2016.PubMed/NCBI View Article : Google Scholar

92 

Sereti E, Karagianellou T, Kotsoni I, Magouliotis D, Kamposioras K, Ulukaya E, Sakellaridis N, Zacharoulis D and Dimas K: Patient derived xenografts (PDX) for personalized treatment of pancreatic cancer: Emerging allies in the war on a devastating cancer? J Proteomics. 188:107–118. 2018.PubMed/NCBI View Article : Google Scholar

93 

Invrea F, Rovito R, Torchiaro E, Petti C, Isella C and Medico E: Patient-derived xenografts (PDXs) as model systems for human cancer. Curr Opin Biotechnol. 63:151–156. 2020.PubMed/NCBI View Article : Google Scholar

94 

Beshiri ML, Tice CM, Tran C, Nguyen HM, Sowalsky AG, Agarwal S, Jansson KH, Yang Q, McGowen KM, Yin J, et al: A PDX/Organoid biobank of advanced prostate cancers captures genomic and phenotypic heterogeneity for disease modeling and therapeutic screening. Clin Cancer Res. 24:4332–4345. 2018.PubMed/NCBI View Article : Google Scholar

95 

Fujii E, Kato A and Suzuki M: Patient-derived xenograft (PDX) models: Characteristics and points to consider for the process of establishment. J Toxicol Pathol. 33:153–160. 2020.PubMed/NCBI View Article : Google Scholar

96 

Abdolahi S, Ghazvinian Z, Muhammadnejad S, Saleh M, Asadzadeh Aghdaei H and Baghaei K: Patient-derived xenograft (PDX) models, applications and challenges in cancer research. J Transl Med. 20(206)2022.PubMed/NCBI View Article : Google Scholar

97 

Fong ELS, Toh TB, Lin QXX, Liu Z, Hooi L, Mohd Abdul Rashid MB, Benoukraf T, Chow EK, Huynh TH and Yu H: Generation of matched patient-derived xenograft in vitro-in vivo models using 3D macroporous hydrogels for the study of liver cancer. Biomaterials. 159:229–240. 2018.PubMed/NCBI View Article : Google Scholar

98 

Nelson SR, Zhang C, Roche S, O'Neill F, Swan N, Luo Y, Larkin A, Crown J and Walsh N: Modelling of pancreatic cancer biology: Transcriptomic signature for 3D PDX-derived organoids and primary cell line organoid development. Sci Rep. 10(2778)2020.PubMed/NCBI View Article : Google Scholar

99 

Romero-Calvo I, Weber CR, Ray M, Brown M, Kirby K, Nandi RK, Long TM, Sparrow SM, Ugolkov A, Qiang W, et al: Human organoids share structural and genetic features with primary pancreatic adenocarcinoma tumors. Mol Cancer Res. 17:70–83. 2019.PubMed/NCBI View Article : Google Scholar

100 

Chauhan AF and Liu SV: Small cell lung cancer: Advances in diagnosis and management. Semin Respir Crit Care Med. 41:435–446. 2020.PubMed/NCBI View Article : Google Scholar

101 

Wang Y, Zou S, Zhao Z, Liu P, Ke C and Xu S: New insights into small-cell lung cancer development and therapy. Cell Biol Int. 44:1564–1576. 2020.PubMed/NCBI View Article : Google Scholar

102 

Wang WZ, Shulman A, Amann JM, Carbone DP and Tsichlis PN: Small cell lung cancer: Subtypes and therapeutic implications. Semin Cancer Biol. 86:543–554. 2022.PubMed/NCBI View Article : Google Scholar

103 

Ireland AS, Micinski AM, Kastner DW, Guo B, Wait SJ, Spainhower KB, Conley CC, Chen OS, Guthrie MR, Soltero D, et al: MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell. 38:60–78.e12. 2020.PubMed/NCBI View Article : Google Scholar

104 

Kalemkerian GP, Loo BW, Akerley W, Attia A, Bassetti M, Boumber Y, Decker R, Dobelbower MC, Dowlati A, Downey RJ, et al: NCCN Guidelines Insights: Small cell lung cancer, version 2.2018. J Natl Compr Canc Netw. 16:1171–1182. 2018.PubMed/NCBI View Article : Google Scholar

105 

Drapkin BJ and Rudin CM: Advances in small-cell lung cancer (SCLC) translational research. Cold Spring Harb Perspect Med. 11(a038240)2021.PubMed/NCBI View Article : Google Scholar

106 

Fan J, Lv Z, Yang G, Liao TT, Xu J, Wu F, Huang Q, Guo M, Hu G, Zhou M, et al: Retinoic acid receptor-related orphan receptors: Critical roles in tumorigenesis. Front Immunol. 9(1187)2018.PubMed/NCBI View Article : Google Scholar

107 

Hogan S, O'Gara JP and O'Neill E: Novel treatment of staphylococcus aureus Device-related infections using fibrinolytic agents. Antimicrob Agents Chemother. 62:e02008–17. 2018.PubMed/NCBI View Article : Google Scholar

108 

Gobin CM, Menefee JN, Lattimore CC, Doty A and Fredenburg KM: Cell Dissociation enzymes affect Annexin V/Flow-cytometric apoptotic assay outcomes After miRNA-based transient transfection. Anticancer Res. 42:2819–2825. 2022.PubMed/NCBI View Article : Google Scholar

109 

Maruyama I, Yoshida C, Kobayashi M, Oyamada H and Momose K: Preparation of single smooth muscle cells from guinea pig taenia coli by combinations of purified collagenase and papain. J Pharmacol Methods. 18:151–161. 1987.PubMed/NCBI View Article : Google Scholar

110 

Wise DR and Thompson CB: Glutamine addiction: A new therapeutic target in cancer. Trends Biochem Sci. 35:427–433. 2010.PubMed/NCBI View Article : Google Scholar

111 

Richards NG and Schuster SM: Mechanistic issues in asparagine synthetase catalysis. Adv Enzymol Relat Areas Mol Biol. 72:145–198. 1998.PubMed/NCBI View Article : Google Scholar

112 

Wellen KE, Lu C, Mancuso A, Lemons JM, Ryczko M, Dennis JW, Rabinowitz JD, Coller HA and Thompson CB: The hexosamine biosynthetic pathway couples growth factor-induced glutamine uptake to glucose metabolism. Genes Dev. 24:2784–2799. 2010.PubMed/NCBI View Article : Google Scholar

113 

Zhang J, Pavlova NN and Thompson CB: Cancer cell metabolism: The essential role of the nonessential amino acid, glutamine. EMBO J. 36:1302–1315. 2017.PubMed/NCBI View Article : Google Scholar

114 

Heeneman S, Deutz NE and Buurman WA: The concentrations of glutamine and ammonia in commercially available cell culture media. J Immunol Methods. 166:85–91. 1993.PubMed/NCBI View Article : Google Scholar

115 

Schneider M, Marison IW and von Stockar U: The importance of ammonia in mammalian cell culture. J Biotechnol. 46:161–185. 1996.PubMed/NCBI View Article : Google Scholar

116 

Imamoto Y, Tanaka H, Takahashi K, Konno Y and Suzawa T: Advantages of AlaGln as an additive to cell culture medium: Use with anti-CD20 chimeric antibody-producing POTELLIGENT™ CHO cell lines. Cytotechnology. 65:135–143. 2013.PubMed/NCBI View Article : Google Scholar

117 

Yoshida Y, Soma T, Matsuzaki T and Kishimoto J: Wnt activator CHIR99021-stimulated human dermal papilla spheroids contribute to hair follicle formation and production of reconstituted follicle-enriched human skin. Biochem Biophys Res Commun. 516:599–605. 2019.PubMed/NCBI View Article : Google Scholar

118 

An WF, Germain AR, Bishop JA, Nag PP, Metkar S, Ketterman J, Walk M, Weiwer M, Liu X, Patnaik D, et al: Discovery of potent and highly selective inhibitors of GSK3b. In: Probe Reports from the NIH Molecular Libraries Program. National Center for Biotechnology Information (US), Bethesda (MD), 2010.

119 

Takahashi T and Shiraishi A: Stem cell signaling pathways in the small intestine. Int J Mol Sci. 21(2032)2020.PubMed/NCBI View Article : Google Scholar

120 

Vincan E, Schwab RHM, Flanagan DJ, Moselen JM, Tran BM, Barker N and Phesse TJ: The Central role of wnt signaling and organoid technology in personalizing anticancer therapy. Prog Mol Biol Transl Sci. 153:299–319. 2018.PubMed/NCBI View Article : Google Scholar

121 

Yoshida T, Singh AK, Bishai WR, McConkey DJ and Bivalacqua TJ: Organoid culture of bladder cancer cells. Investig Clin Urol. 59:149–151. 2018.PubMed/NCBI View Article : Google Scholar

122 

Djomehri SI, Burman B, Gonzalez ME, Takayama S and Kleer CG: A reproducible scaffold-free 3D organoid model to study neoplastic progression in breast cancer. J Cell Commun Signal. 13:129–143. 2019.PubMed/NCBI View Article : Google Scholar

123 

Ahn Y, An JH, Yang HJ, Lee DG, Kim J, Koh H, Park YH, Song BS, Sim BW, Lee HJ, et al: Human blood vessel organoids penetrate human cerebral organoids and form a Vessel-like system. Cells. 10(2036)2021.PubMed/NCBI View Article : Google Scholar

124 

Li Y, Wang R, Huang D, Ma X, Mo S, Guo Q, Fu G, Li Y, Xu X, Hu X, et al: A novel human colon signet-ring cell carcinoma organoid line: Establishment, characterization and application. Carcinogenesis. 41:993–1004. 2020.PubMed/NCBI View Article : Google Scholar

125 

Ma HC, Zhu YJ, Zhou R, Yu YY, Xiao ZZ and Zhang HB: Lung cancer organoids, a promising model still with long way to go. Crit Rev Oncol Hematol. 171(103610)2022.PubMed/NCBI View Article : Google Scholar

126 

Maddalo G, Spolverato Y, Rugge M and Farinati F: Gastrin: From pathophysiology to cancer prevention and treatment. Eur J Cancer Prev. 23:258–263. 2014.PubMed/NCBI View Article : Google Scholar

127 

Zheng B, Ko KP, Fang X, Wang X, Zhang J, Jun S, Kim BJ, Luo W, Kim MJ, Jung YS, et al: A new murine esophageal organoid culture method and organoid-based model of esophageal squamous cell neoplasia. iScience. 24(103440)2021.PubMed/NCBI View Article : Google Scholar

128 

Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, Dwinell MB, Hunt B, Evans DB, Gershan J, et al: Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. 18(335)2018.PubMed/NCBI View Article : Google Scholar

129 

Kawasaki K, Toshimitsu K, Matano M, Fujita M, Fujii M, Togasaki K, Ebisudani T, Shimokawa M, Takano A, Takahashi S, et al: An organoid biobank of neuroendocrine neoplasms enables genotype-phenotype mapping. Cell. 183:1420–1435.e21. 2020.PubMed/NCBI View Article : Google Scholar

130 

Shiota J, Samuelson LC and Razumilava N: Hepatobiliary organoids and their applications for studies of liver health and disease: Are We There Yet? Hepatology. 74:2251–2263. 2021.PubMed/NCBI View Article : Google Scholar

131 

Dong R, Zhang B and Zhang X: Liver organoids: An in vitro 3D model for liver cancer study. Cell Biosci. 12(152)2022.PubMed/NCBI View Article : Google Scholar

132 

Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, et al: Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology. 141:1762–1772. 2011.PubMed/NCBI View Article : Google Scholar

133 

Fatehullah A, Tan SH and Barker N: Organoids as an in vitro model of human development and disease. Nat Cell Biol. 18:246–254. 2016.PubMed/NCBI View Article : Google Scholar

134 

van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, van Houdt W, van Gorp J, Taylor-Weiner A, Kester L, et al: Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 161:933–945. 2015.PubMed/NCBI View Article : Google Scholar

135 

Karthaus WR, Iaquinta PJ, Drost J, Gracanin A, van Boxtel R, Wongvipat J, Dowling CM, Gao D, Begthel H, Sachs N, et al: Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell. 159:163–175. 2014.PubMed/NCBI View Article : Google Scholar

136 

Verissimo CS, Overmeer RM, Ponsioen B, Drost J, Mertens S, Verlaan-Klink I, Gerwen BV, van der Ven M, Wetering MV, Egan DA, et al: Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. Elife. 5(e18489)2016.PubMed/NCBI View Article : Google Scholar

137 

Gohi B, Liu XY, Zeng HY, Xu S, Ake KMH, Cao XJ, Zou KM and Namulondo S: Enhanced efficiency in isolation and expansion of hAMSCs via dual enzyme digestion and micro-carrier. Cell Biosci. 10(2)2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2024
Volume 28 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Q and Zhang M: Recent advances in lung cancer organoid (tumoroid) research (Review). Exp Ther Med 28: 383, 2024.
APA
Zhang, Q., & Zhang, M. (2024). Recent advances in lung cancer organoid (tumoroid) research (Review). Experimental and Therapeutic Medicine, 28, 383. https://doi.org/10.3892/etm.2024.12672
MLA
Zhang, Q., Zhang, M."Recent advances in lung cancer organoid (tumoroid) research (Review)". Experimental and Therapeutic Medicine 28.4 (2024): 383.
Chicago
Zhang, Q., Zhang, M."Recent advances in lung cancer organoid (tumoroid) research (Review)". Experimental and Therapeutic Medicine 28, no. 4 (2024): 383. https://doi.org/10.3892/etm.2024.12672