A clinician’s perspective: what tumor-organoid researchers ought to know

Article information

Organoid. 2025;5.e1
Publication date (electronic) : 2025 January 25
doi : https://doi.org/10.51335/organoid.2025.5.e1
Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
Correspondence to: Jeong Uk Lim Division of Pulmonary, Critical Care and Allergy, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10 63(yuksam)-ro, Yeongdeungpo-gu, Seoul 07345, Korea E-mail: cracovian@catholic.ac.kr
Received 2025 January 15; Revised 2025 January 17; Accepted 2025 January 20.

Abstract

Patient-derived tumor organoids (PDTOs) provide powerful platforms for modeling human tumors, offering insights into cancer biology and personalized therapy development. Incorporating clinical knowledge—such as cancer staging, molecular profiling, and treatment outcomes—enhances the relevance of PDTO research. Lung cancer staging systems guide tumor sample selection, thereby influencing organoid growth and treatment response studies. Clinical endpoints and molecular characteristics further align organoid experiments with real-world therapeutic challenges and patient care. Despite their potential, PDTO research faces challenges in replicating tumor heterogeneity and the tumor microenvironment, both of which are critical for studying drug resistance and treatment efficacy. Addressing these complexities requires close collaboration between researchers and clinicians to ensure that organoid models accurately reflect the clinical course of the disease. This review provides information on integrating clinical insights into PDTO research, focusing on aspects such as sample selection, clinical endpoints, and the implications of tumor staging. By bridging the gap between basic organoid research and clinical oncology, the review aims to guide researchers in enhancing the translational value of their work. A clear understanding of these clinical aspects can support the development of more robust organoid models, ultimately advancing cancer treatment strategies and improving patient outcomes.

Introduction

Patient-derived tumor organoids (PDTOs) serve as an innovative and impactful model for advancing cancer research and therapeutic strategies. Owing to their structural and functional similarities to human tumors, PDTOs have gained considerable interest for applications in cancer modeling, personalized treatment, drug evaluation, tumor tissue preservation, investigation of tumorigenesis, and various translational research studies [15]. Furthermore, PDTOs are being explored as platforms for clinical applications—including biomarker identification and anticancer drug screening—to improve the effectiveness of personalized therapy [6,7].

Although the use of organoids holds immense potential, effective translation to clinical applications requires a robust understanding of real-world clinical priorities. PDTOs are invaluable for bridging the gap between laboratory models and clinical practice because they closely replicate the genetic and phenotypic characteristics of the original tumors. As such, they have become effective tools for drug screening and biomarker discovery. Studies have demonstrated that PDTOs can predict patient-specific responses to treatments, thereby enhancing personalized medicine approaches. For instance, research has shown that organoids derived from colorectal cancer patients can forecast individual responses to chemotherapy, aiding in the selection of effective treatment regimens [8]. Additionally, PDTOs have been utilized to assess the efficacy of oncolytic adenoviruses, providing insights into personalized virotherapy strategies [9].

Integrating clinical information—such as cancer staging, clinical endpoints (overall survival [OS] and progression-free survival [PFS]), methods for obtaining viable tumor specimens, and the implications of mutation studies and programmed cell death ligand 1 (PD-L1) expression—is crucial for the effective acquisition and interpretation of PDTOs. This comprehensive understanding enhances the translational potential of organoid research [10].

This review aims to offer tumor-organoid researchers insights into the clinical oncology landscape, with a particular focus on lung cancer.

Materials and methods

Ethics statement: This study was a literature review of previously published studies and was therefore exempt from institutional review board approval.

This narrative review was conducted to provide a comprehensive understanding of PDTOs in the context of lung cancer research and clinical applications. References were identified through a systematic search of databases including PubMed, Scopus, and Web of Science. The search employed keywords such as “patient-derived tumor organoids,” “lung cancer,” "cancer staging,” “clinical endpoints,” “mutation profiling,” “biomarkers,” “drug resistance,” and “immunotherapy.” Boolean operators such as “AND,” “OR,” and “NOT” were used to refine search results and ensure their relevance to the review’s objectives.

Articles were selected based on their focus on PDTOs in cancer modeling, translational applications, and clinical integration. Priority was given to studies published in high-impact, peer-reviewed journals between 2015 and 2024.

Lung cancer pathologic types

When samples are acquired and examined under a microscope, professional hospital pathologists diagnose whether the tissue is cancerous. Once lung cancer is confirmed, pathologic typing is performed. Approximately 15% to 20% of lung cancers are diagnosed as small-cell lung cancer (SCLC), which exhibits neuroendocrine features and expresses specific markers characteristic of SCLC. This type is distinct for its high proliferation rate and aggressive metastatic potential. Despite an initially high response to chemotherapy, patients diagnosed with SCLC often have a poor prognosis [11].

Non-small cell lung cancer (NSCLC), which includes all lung cancers not classified as SCLC, accounts for approximately 80% to 85% of cases. Among the NSCLC subtypes, adenocarcinoma represents about 55% to 60% of cases, followed by squamous cell carcinoma at 30% to 35%. Other subtypes include unspecified NSCLC, large cell neuroendocrine carcinoma, and other less common types. Adenocarcinoma and squamous cell carcinoma together represent the majority of NSCLC cases [12]. Notably, squamous cell carcinoma is more strongly associated with smoking than other NSCLC subtypes and is commonly found in the central regions of the lung.

Differentiating pathologic types is important for two reasons: (1) different subtypes exhibit distinct clinical outcomes, and (2) treatment modalities vary based on the pathologic subtype.

Lung cancer staging and its clinical importance

1. Overview of lung cancer staging

Lung cancer is the most frequently diagnosed cancer worldwide and remains a leading cause of global mortality [13]. Histologically, lung cancer is categorized into SCLC and NSCLC. SCLC constitutes approximately 15% to 20% of cases, while NSCLC accounts for the remaining 80% to 85% [14,15].

Staging lung cancer is essential for assessing disease progression, predicting prognosis, and determining optimal treatment strategies [16]. The tumor, node, and metastasis (TNM) classification system plays a key role in evaluating disease burden, as it assesses the size of the primary tumor, the involvement of mediastinal lymph nodes, and the extent of metastatic spread. Treatment strategies are determined based on the cancer stage [17]. The ninth edition of the TNM Classification, developed by the International Association for the Study of Lung Cancer, is the most recent version used for lung cancer staging [18]. This system, derived from an extensive global dataset, provides detailed criteria for evaluating the extent of the disease.

The TNM system comprises three key components: (1) T (Tumor): Represents the size and extent of the primary tumor; (2) N (Node): Indicates whether the cancer has involved mediastinal lymph nodes; (3) M (Metastasis): Describes whether the cancer has spread to distant organs in the body.

The system uses letters and numbers to provide detailed information about the disease. For example, designations like T1N0MX or T3N1M0 indicate different aspects of the cancer’s characteristics [19]. The combined TNM stages are then used to classify cancer from stage I to stage IV. A higher number following T or N indicates a greater disease burden and is more likely to correspond to an advanced stage. If intrathoracic lesions, such as contralateral nodules or malignant pleural or pericardial effusions, are present (and no extrathoracic metastatic lesions exist), the M stage is classified as M1a. For patients with extrathoracic metastatic lesions (located outside the chest region), the classification will be M1b, M1c1, or M1c2, depending on the number and extent of the lesions. All metastatic lesions, whether intrathoracic or extrathoracic, are classified as stage IV [20].

A staging system categorizes cancer patients from stage I (early, localized cancer) to stage IV (advanced cancer that has spread to distant areas). This classification assists healthcare providers in developing tailored treatment plans, optimizing patient care, and improving outcomes by aligning interventions with the cancer's progression stage. Lung cancer management employs multiple treatment approaches, including surgery, radiotherapy, and various anticancer therapies. The choice of treatment depends on factors such as the cancer stage, histological subtype, and the patient’s overall health status [21]. For early-stage cases (stages I or II), curative interventions such as surgery or stereotactic radiotherapy (for patients who are not surgical candidates) are commonly used. In contrast, advanced or metastatic lung cancers are typically managed with palliative chemotherapy and/or radiotherapy, as surgical options are generally not feasible [21].

For SCLC, although the TNM staging system is used, treatment decisions are primarily based on a unique staging system that classifies the disease as either “limited stage” (where all lesions are confined to a single radiation field with no metastases) or “extensive stage” (where lesions extend beyond a single radiation field and are often accompanied by metastatic lesions) [22].

2. Why staging matters for organoid research

Lung cancer staging is essential not only for patient treatment but also for researchers studying tumor organoids. Some studies have incorporated tumor burden—reflected by lung cancer staging at diagnosis—as one of the indices for predicting prognosis based on PDTOs [23].

Localized tumors (stage I–II): These cancers are usually small and confined to the lung, and they can often be removed surgically. For these stages, the priority is predicting early disease recurrence after complete resection. The five-year recurrence rate can be as high as 45% in stage 1b and 62% in stage II. Patients in these stages often receive adjuvant chemotherapy to reduce the risk of recurrence following surgery [24]. PDTOs can serve as a platform for selecting the most effective chemotherapy regimens.

Advanced tumors (stage III–IV): These cancers have spread further, either to nearby lymph nodes or to distant parts of the body. Although stage IIIA—an earlier stage within the advanced group—may be treated with complete surgical resection, most patients with stage III or IV disease typically receive non-surgical treatments, such as traditional chemotherapy, targeted therapy, immunotherapy, radiotherapy, or combinations thereof. These tumors are generally more complex, with higher mutation rates and greater resistance to treatment.

By understanding the stage of the cancer, researchers can select appropriate samples to create organoids that accurately represent the disease. This is important for testing drugs and discovering new treatments tailored to specific stages of lung cancer.

Cancer specimen acquisition and its challenges

1. Obtaining tumor samples from patients

Clinically, obtaining high-quality tumor specimens is essential for diagnostic and therapeutic decisions. Methods include biopsies, fine-needle aspirations, and surgical resections, depending on the tumor’s location, stage, and the patient’s condition. Researchers working with organoids should understand that each method yields samples with different viability, cellular diversity, and size. For example, surgical resections can provide large, viable samples suitable for organoid culture, while needle biopsies may yield smaller specimens with limited cellular diversity.

It is crucial to consider the timing of sample collection. In lung cancer, most samples are obtained at the time of diagnosis. Samples collected after complete resection are particularly valuable because they closely resemble the actual human tumor and are typically free from chemotherapy exposure, thus representing the patient’s native tumor microenvironment. Most diagnostic samples are obtained via percutaneous needle biopsy or aspiration, which involves extracting a small piece of tumor tissue by inserting a needle into the lung tumor. Alternatively, samples may be acquired through surgery, typically performed to obtain a larger specimen for diagnostic or curative purposes. Additionally, malignant pleural effusion samples can be collected, offering a relatively easy and safe option, as procedures like pigtail catheter insertion or thoracotomy provide access to a substantial number of tumor cells [25]. Tumor samples can also be obtained through bronchoscopy or endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA). During bronchoscopy, clinicians access the patient’s airways to collect tumor tissue under direct visualization [26]. EBUS-TBNA allows for the collection of tissue from mediastinal or hilar lymph nodes using ultrasound guidance via a bronchoscope, providing a minimally invasive method for sampling tumors or lymph nodes in the mediastinum [27,28].

Recent studies have shown that malignant pleural effusions are a reliable source of tumor cells for generating tumor organoids, possibly due to their high cancer cell purity. It is important to note the site of tumor sample collection.

The situation differs when samples are collected after chemotherapy has been initiated. In terms of understanding a patient’s clinical course, the response to actual drugs holds greater significance compared to samples collected at baseline. Two factors must be considered when interpreting PDTOs growth and drug responses: whether the samples were collected while the patient was responding well to anticancer treatment and whether they were influenced by the effects of the ongoing chemotherapy regimen.

Therefore, researchers working with PDTOs should have a clear understanding of the patient’s clinical endpoints and engage in discussions with the clinicians managing the patients.

2. Challenges in sample collection

In lung cancer, collecting representative samples can be challenging due to the tumor's location and heterogeneity. EBUS-TBNA is one technique used to access mediastinal lymph nodes in cases of metastasis. Understanding these techniques aids organoid researchers in collaborating with clinicians to obtain samples that best represent the tumor’s heterogeneity, which is important for studying drug resistance and mutation evolution.

Tumor tissues obtained from patients are processed for dissociation to isolate patient-derived cancer cells using a combination of mechanical and chemical dissociation techniques [29]. A notable issue is the presence of non-cancerous cells in the sample. During the process of generating organoids from tumor tissues, normal cells adjacent to tumor cells may proliferate more rapidly than cancer cells, resulting in contamination of lung cancer organoids and hindering their development and application. A study investigating the purity of lung adenocarcinoma organoids found that 58% of the samples were contaminated by healthy airway cells [30]. Therefore, obtaining samples with a high proportion of cancer cells is important.

Clinical endpoints: OS, PFS, and the objective response rate

1. OS

OS, defined as the time from treatment initiation to death from any cause, remains the gold standard for evaluating cancer treatment efficacy. From a clinician’s perspective, one of the primary goals in managing lung cancer patients is to prolong OS as much as possible [31]. Although OS is a clear and easily interpretable endpoint, it is less applicable to slowly progressive diseases or patients with a long expected survival [31].

2. PFS

PFS measures the duration during and after treatment that a patient lives without disease progression. In clinical trials, comparing the PFS of different treatment groups helps determine which treatment is most effective in delaying disease progression. According to the Response Evaluation Criteria in Solid Tumors (RECIST), disease progression is defined as a 20% increase in the sum of the diameters of target lesions [32].

3. Objective response rate

The objective response rate (ORR), which is the percentage of patients who experience a partial or complete response to treatment, provides immediate feedback on treatment efficacy. The ORR, defined by RECIST criteria, is one of the most frequently used endpoints [33].

After undergoing chemotherapy, patients typically receive computed tomography and/or magnetic resonance imaging scans every 2 to 3 months to monitor target lesions, which are specific areas tracked to assess treatment response. For lung cancer patients, these target lesions usually include the primary lung tumor and notable metastatic sites. A significant reduction in the size of these lesions indicates regression. Depending on the extent of the reduction, patients are categorized as having either a “partial response” or a “complete response.” If patients achieve either response at least once during treatment, they are considered to have demonstrated an Objective response [34]. Evaluating whether patients’ actual treatment responses align with the drug responses observed in PDTO models is important for assessing the translational relevance of organoid-based research.

Mutation studies in lung cancer: EGFR, KRAS, and ALK

1. Relevance of mutation profiling

One of the potential strengths of PDTOs in oncology is their ability to assess the efficacy of various drugs on a patient’s tumor cells prior to clinical administration, thereby facilitating more precise and effective treatment [35]. Understanding key genetic alterations in patients is crucial for this process, as targeted therapies are designed based on these genetic features [3640].

Mutation profiling is essential in lung cancer treatment because specific genetic alterations serve as therapeutic targets. Approximately 15% to 30% of NSCLC cases harbor Epidermal Growth Factor Receptor (EGFR) mutations, while Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are present in about 25% of cases in Western populations. Although less frequent, ALK rearrangements are highly actionable [41].

Identifying these mutations enables the use of targeted therapies, which have significantly improved patient outcomes. For example, cancers with EGFR mutations can be treated with tyrosine kinase inhibitors (TKIs), leading to increased survival rates. Similarly, patients with ALK rearrangements benefit from ALK inhibitors, which have shown efficacy in halting disease progression [42]. Key genetic alterations can be identified through single-gene analysis methods or next-generation sequencing, which allows the detection of a panel of critical mutations in cancer tissue or patients’ blood samples [43].

Therefore, comprehensive genetic profiling is crucial for personalized treatment strategies in NSCLC, as it allows for the selection of appropriate targeted therapies based on individual tumor genetics. This approach not only improves therapeutic efficacy but also minimizes unnecessary side effects by avoiding ineffective treatments.

2. EGFR mutations

EGFR mutations are present in approximately 40% to 59% of NSCLC cases in Asian patients and 5% to 19.4% of NSCLC cases in Caucasian patients [44]. Certain genetic alterations in EGFR are amenable to treatment with TKIs. Common EGFR mutations in NSCLC that are targetable by TKIs include deletions in exon 19 and the single amino acid substitution L858R in exon 21 [45]. First-generation TKIs, such as erlotinib and gefitinib [46], were followed by second-generation TKIs like afatinib and dacomitinib [47,48]. Currently, third-generation TKIs, including osimertinib and lazertinib, are the standard of care for first-line treatment of common EGFR mutations [37,49].

Although EGFR mutations initially respond well to TKIs, resistance eventually develops due to additional mutations. Despite TKIs blocking cancer cell proliferation pathways, cancer cells adapt by activating alternative pathways or altering the target proteins to which TKIs bind, ultimately reducing drug efficacy. Numerous clinically significant resistance mutations exist, some of which will be discussed later.

Upon progression on osimertinib, approximately 15% of tumors develop on-target mutations [50], with EGFR C797X in exon 20 being the most prevalent. This mutation interferes with the covalent binding of osimertinib to the EGFR kinase domain. Other notable acquired mutations include L718Q/V, G719A, and G724S in exon 18 [51].

Another notable mechanism of resistance involves MET-dependent resistance, often activated by the formation of homodimers or through trans-activation by other tyrosine kinase receptors. MET amplification contributes to resistance in approximately 50% to 60% of cases treated with first- and second-generation EGFR TKIs [52,53], and in 15% to 19% of cases involving third-generation EGFR TKIs [54]. In EGFR-mutant NSCLC, MET amplification leads to resistance by persistently activating key downstream signaling pathways—such as MAPK, STAT, and PI3K/AKT—which continue to drive tumor cell proliferation and survival independent of EGFR signaling [52,55]. Overcoming this resistance requires concurrent targeting of both EGFR and MET receptors, highlighting the potential utility of anti‑MET agents in combination with EGFR TKIs to achieve a more effective antitumor response [56].

Another notable mutation involves the human epidermal growth factor receptor 2 (HER2), a tyrosine kinase receptor encoded by the ERBB2 gene [57]. HER2 amplification is observed in 5% of patients who develop resistance to second-line osimertinib and in 2% of cases using first-line osimertinib [58,59].

RET fusions have also been reported as an acquired resistance mechanism after EGFR TKI treatment [60,61]. In fact, RET fusions are more frequently linked to acquired resistance to third-generation EGFR TKIs compared to earlier generations [62]. BRAF V600E-mediated osimertinib resistance is also notable [63,64]. Recent studies indicate that the activation of the PI3K/AKT/mTOR signaling pathway contributes to the aggressiveness of lung cancer [65].

Because the majority of NSCLC patients have targetable EGFR mutations and many eventually develop resistance to EGFR TKIs, there is a significant unmet need for detecting resistance-related genetic alterations and identifying effective subsequent treatments to overcome this challenge.

3. ALK mutations

ALK gene fusion was first recognized for its clinical relevance in lung cancer in 2007 [66]. Patients with ALK rearrangements represent approximately 2–5% of all NSCLC cases [67]. These rearrangements, often caused by an inversion of the ALK gene with the echinoderm microtubule-associated protein-like 4 (EML4) gene, render patients eligible for treatment with ALK TKIs [6870].

4. KRAS

The KRAS gene, part of the RAS oncogene family, is mutated in approximately 25% to 35% of newly diagnosed NSCLC cases [71]. Among these mutations, G12C is the most prevalent, accounting for roughly 40% of cases, followed by G12V (21%), G12D (17%), and G12A (10%) [72].

One of the most notable KRAS mutations is KRAS G12C. Based on the results of the CodeBreak 100 trial, sotorasib—a KRAS G12C inhibitor—demonstrated an ORR of 36% in patients with KRAS G12C-mutated NSCLC and has been approved for clinical use [73,74]. Similarly, adagrasib, another KRAS G12C inhibitor, has received United States Food and Drug Administration approval for the treatment of NSCLC patients who have received at least one prior systemic therapy [41,75]. Despite the introduction of these groundbreaking KRAS inhibitors, resistance inevitably develops, as with EGFR TKIs. Primary resistance mechanisms have been linked to loss-of-function mutations in KEAP1, SMARCA4, CDKN2A, and STK11. These mutations are significantly enriched in patients with KRAS G12C-mutated NSCLC who experience early progression (within 3 months without achieving an objective response) following treatment with sotorasib or adagrasib. Interestingly, STK11 loss did not adversely affect treatment outcomes in patients with KEAP1 wild-type tumors, suggesting that the negative effects of STK11 mutations in patients treated with G12C inhibitors are largely driven by concurrent KEAP1 mutations [76]. There is an unmet need to identify key resistance mechanisms and develop effective next-line treatments for patients who experience progression after KRAS inhibitor therapy.

Understanding PD-L1 as a biomarker

Molecular markers play a crucial role in lung cancer immunotherapy, with proteins such as programmed cell death protein 1 (PD-1) and its ligand PD-L1 serving as key predictors of response to immunotherapeutic agents [77,78]. PD-1 is a critical immune checkpoint that enables cancer cells to evade immune responses by interacting with PD-L1 expressed on tumor cells, thereby inhibiting the tumor-killing activity of CD8 T cells [79].

Immune checkpoint inhibitors (ICIs) are a mainstay modality in the treatment of metastatic NSCLC. Used either alone or in combination with platinum-based chemotherapy, these therapies have demonstrated significant benefits—including prolonged OS and PFS—in patients with locally advanced or metastatic NSCLC [80]. PD-L1 expression serves as a predictive marker for response to ICIs such as pembrolizumab and nivolumab in NSCLC [81]. For organoid researchers, incorporating PD-L1 expression into immunotherapy model development is crucial, as it can influence the models’ responsiveness to immune-based treatments.

Integrating tumor-organoid models into patient care

Clinically relevant tumor-organoid models must reflect patient-specific characteristics, such as mutation profiles, histological features, and response patterns. By correlating organoid responses with clinical outcomes like OS, PFS, and ORR, researchers can validate the predictive potential of organoids. Longitudinal studies that track changes in organoid models over time could simulate a patient’s clinical course, providing insights into disease progression and treatment responses.

Collaboration between clinicians and researchers is crucial for optimizing the utility of organoid models. Clinicians can provide vital information regarding patient selection criteria, sample acquisition methods, and clinical endpoints. Conversely, organoid researchers can offer clinicians personalized insights into patient-specific treatment responses. This collaboration supports clinicians by providing a more accurate depiction of the patient’s tumor microenvironment and by identifying potentially effective treatment modalities. The checklist of key clinical points to consider when processing PDTOs is provided in Table 1.

Checklist of clinical features for lung cancer PDTOs

Future directions and challenges

Advances in technology have facilitated the development and application of targeted therapies and immunotherapies for lung cancer treatment. The combination of immunotherapy with chemotherapy is expected to heighten the demand for personalized drug regimens [82]. Consequently, the importance of drug efficacy prediction analyses is likely to expand. Lung cancer organoid models offer a valuable platform for assessing the effectiveness of various anticancer agents, including immunotherapies, and for evaluating combinations of multiple drugs. Additionally, these models serve as predictive tools for the efficacy of next-generation anticancer drugs currently in development.

One challenge in utilizing PDTOs for personalized lung cancer treatment is ensuring their effective and consistent application, with tumor heterogeneity presenting a potential limitation. Single biopsies may not capture the full genetic diversity of a tumor. Employing multi-region sampling or integrating circulating tumor DNA into organoid cultures could enhance model accuracy. By representing multiple tumor sites, organoids could become more robust predictors of patient response, particularly for metastasized and heterogeneous tumors like advanced NSCLC.

Furthermore, there is an unmet need to replicate a tumor microenvironment that is suitable for immunotherapy drug testing. To address this, co-culturing immune cells is essential. However, accurately simulating a tumor-immune microenvironment that closely resembles that of the patient who provided the cancer tissue remains a significant challenge.

Conclusion

This review highlights the essential clinical considerations that tumor-organoid researchers should understand to bridge the gap between laboratory models and patient care. By integrating knowledge of lung cancer staging, specimen acquisition, clinical endpoints, mutation studies, and immunotherapy markers, researchers can increase the translational value of organoid models. Collaborative efforts between clinicians and researchers, combined with advancements in organoid technology, have the potential to transform the role of organoids in precision oncology. As research progresses, tumor organoids could facilitate a personalized approach to cancer treatment, supporting clinical decision-making and improving patient outcomes.

Notes

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This research was supported by a grant (RS-2024-00332142) from ministry of food and drug safety.

Data availability

Not applicable.

References

1. Xu H, Lyu X, Yi M, Zhao W, Song Y, Wu K. Organoid technology and applications in cancer research. J Hematol Oncol 2018;11:116. 10.1186/s13045-018-0662-9. 30219074.
2. Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat Protoc 2020;15:3380–9. 10.1038/s41596-020-0379-4. 32929210.
3. Xu R, Zhou X, Wang S, Trinkle C. Tumor organoid models in precision medicine and investigating cancer-stromal interactions. Pharmacol Ther 2021;218:107668. 10.1016/j.pharmthera.2020.107668. 32853629.
4. Aboulkheyr Es H, Montazeri L, Aref AR, Vosough M, Baharvand H. Personalized cancer medicine: an organoid approach. Trends Biotechnol 2018;36:358–71. 10.1016/j.tibtech.2017.12.005. 29366522.
5. Kondo J, Inoue M. Application of cancer organoid model for drug screening and personalized therapy. Cells 2019;8:470. 10.3390/cells8050470. 31108870.
6. Yang Y, Kong Y, Cui J, Hou Y, Gu Z, Ma C. Advances and applications of cancer organoids in drug screening and personalized medicine. Stem Cell Rev Rep 2024;20:1213–26. 10.1007/s12015-024-10714-6. 38532032.
7. Xu H, Jiao D, Liu A, Wu K. Tumor organoids: applications in cancer modeling and potentials in precision medicine. J Hematol Oncol 2022;15:58. 10.1186/s13045-022-01278-4. 35551634.
8. Chitrangi S, Vaity P, Jamdar A, Bhatt S. Patient-derived organoids for precision oncology: a platform to facilitate clinical decision making. BMC Cancer 2023;23:689. 10.1186/s12885-023-11078-9. 37479967.
9. Wang J, Chen C, Wang L, Xie M, Ge X, Wu S, et al. Patient-derived tumor organoids: new progress and opportunities to facilitate precision cancer immunotherapy. Front Oncol 2022;12:872531. 10.3389/fonc.2022.872531. 35449581.
10. Zeng X, Ma Q, Li XK, You LT, Li J, Fu X, et al. Patient-derived organoids of lung cancer based on organoids-on-a-chip: enhancing clinical and translational applications. Front Bioeng Biotechnol 2023;11:1205157. 10.3389/fbioe.2023.1205157. 37304140.
11. Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Primers 2021;7:3. 10.1038/s41572-020-00235-0. 33446664.
12. Zappa C, Mousa SA. Non-small cell lung cancer: current treatment and future advances. Transl Lung Cancer Res 2016;5:288–300. 10.21037/tlcr.2016.06.07. 27413711.
13. Sung CW, Lu TC, Fang CC, Lin JY, Yeh HF, Huang CH, et al. Factors associated with a high-risk return visit to the emergency department: a case-crossover study. Eur J Emerg Med 2021;28:394–401. 10.1097/mej.0000000000000851. 34191766.
14. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JH, Beasley MB, 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 2015;10:1243–60. 10.3410/f.725757738.793546406. 26291008.
15. Osmani L, Askin F, Gabrielson E, Li QK. Current WHO guidelines and the critical role of immunohistochemical markers in the subclassification of non-small cell lung carcinoma (NSCLC): moving from targeted therapy to immunotherapy. Semin Cancer Biol 2018;52(Pt 1):103–9. 10.1016/j.semcancer.2017.11.019. 29183778.
16. Liam CK, Liam YS, Poh ME, Wong CK. Accuracy of lung cancer staging in the multidisciplinary team setting. Transl Lung Cancer Res 2020;9:1654–66. 10.21037/tlcr.2019.11.28. 32953539.
17. Lababede O, Meziane MA. The eighth edition of TNM staging of lung cancer: reference chart and diagrams. Oncologist 2018;23:844–8. 10.1634/theoncologist.2017-0659. 29650687.
18. Detterbeck FC, Woodard GA, Bader AS, Dacic S, Grant MJ, Park HS, et al. The proposed ninth edition TNM classification of lung cancer. Chest 2024;166:882–95. 10.1016/j.chest.2024.05.026. 38885896.
19. American Cancer Society. Cancer staging [Internet]. American Cancer Society; 2024 [cited 2025 Jan 4]. Available from: https://www.cancer.org/cancer/diagnosis-staging/staging.html.
20. Memo in Oncology. Proposals for the 9th edition of the TNM classification [Internet]. Memo in Oncology; 2023 [cited 2025 Jan 12]. Available from: https://memoinoncology.com/wclc-2023/proposals-for-the-9th-edition-of-the-tnm-classification/.
21. Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. Non-small cell lung cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022;20:497–530. 35545176.
22. Kalemkerian GP, Akerley W, Bogner P, Borghaei H, Chow LQ, Downey RJ, et al. Small cell lung cancer. J Natl Compr Canc Netw 2013;11:78–98. 23307984.
23. Lee SY, Cho HJ, Choi J, Ku B, Moon SW, Moon MH, et al. Cancer organoid-based diagnosis reactivity prediction (CODRP) index-based anticancer drug sensitivity test in ALK-rearrangement positive non-small cell lung cancer (NSCLC). J Exp Clin Cancer Res 2023;42:309. 10.1186/s13046-023-02899-4. 37993887.
24. Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd FA, Stephens RJ, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol 2008;26:3552–9. 10.1200/jco.2007.13.9030. 18506026.
25. American Thoracic Society. Management of malignant pleural effusions. Am J Respir Crit Care Med 2000;162:1987–2001. 10.1164/ajrccm.162.5.ats8-00. 11069845.
26. Herth FJ, Eberhardt R, Ernst A. The future of bronchoscopy in diagnosing, staging and treatment of lung cancer. Respiration 2006;73:399–409. 10.1159/000093369. 16775411.
27. Yamamoto S, Nakayama M. Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA): revolutionizing the landscape of lung disease diagnostics. J Med Ultrason (2001) 2024;51:245–51. 10.1007/s10396-023-01391-y. 38114815.
28. Medford AR, Bennett JA, Free CM, Agrawal S. Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA): applications in chest disease. Respirology 2010;15:71–9. 10.1111/j.1440-1843.2009.01652.x. 19895387.
29. Lee E, Lee SY, Seong YJ, Ku B, Cho HJ, Kim K, et al. Lung cancer organoid-based drug evaluation models and new drug development application trends. Transl Lung Cancer Res 2024;13:3741–63. 10.21037/tlcr-24-603. 39830742.
30. Dijkstra KK, Monkhorst K, Schipper LJ, Hartemink KJ, Smit EF, Kaing S, et al. Challenges in establishing pure lung cancer organoids limit their utility for personalized medicine. Cell Rep 2020;31:107588. 10.1016/j.celrep.2020.107588. 32375033.
31. Delgado A, Guddati AK. Clinical endpoints in oncology: a primer. Am J Cancer Res 2021;11:1121–31. 33948349.
32. Villaruz LC, Socinski MA. The clinical viewpoint: definitions, limitations of RECIST, practical considerations of measurement. Clin Cancer Res 2013;19:2629–36. 10.1158/1078-0432.ccr-12-2935. 23669423.
33. Zettler ME, Lee CH, Gajra A, Feinberg BA. Assessment of objective response rate (ORR) by investigator versus blinded independent central review in pivotal trials of drugs approved for solid tumor indications. J Clin Oncol 2021;39(15_suppl):e13570. 10.1200/JCO.2021.39.15_suppl.e13570.
34. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026. 19097774.
35. Swain B, Maddi S. Patient-derived tumor organoids: generation and applications in disease modeling and personalized therapy. Nat Cell Sci 2024;2:185–197. 10.61474/ncs.2024.00008.
36. Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med 2020;382:41–50. 10.1056/nejmoa1913662. 31751012.
37. Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med 2021;384:2371–81. 34096690.
38. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med 2017;377:829–38. 10.1056/nejmoa1704795. 28586279.
39. Paik PK, Felip E, Veillon R, Sakai H, Cortot AB, Garassino MC, et al. Tepotinib in non-small-cell lung cancer with MET Exon 14 skipping mutations. N Engl J Med 2020;383:931–43. 32469185.
40. Jänne PA, Riely GJ, Gadgeel SM, Heist RS, Ou SI, Pacheco JM, et al. Adagrasib in non-small-cell lung cancer harboring a KRASG12C mutation. N Engl J Med 2022;387:120–31. 35658005.
41. Grosse A, Grosse C, Rechsteiner M, Soltermann A. Analysis of the frequency of oncogenic driver mutations and correlation with clinicopathological characteristics in patients with lung adenocarcinoma from Northeastern Switzerland. Diagn Pathol 2019;14:18. 10.1186/s13000-019-0789-1. 30744664.
42. Araghi M, Mannani R, Heidarnejad Maleki A, Hamidi A, Rostami S, Safa SH, et al. Recent advances in non-small cell lung cancer targeted therapy; an update review. Cancer Cell Int 2023;23:162. 10.1186/s12935-023-02990-y. 37568193.
43. Hussen BM, Abdullah ST, Salihi A, Sabir DK, Sidiq KR, Rasul MF, et al. The emerging roles of NGS in clinical oncology and personalized medicine. Pathol Res Pract 2022;230:153760. 10.1016/j.prp.2022.153760. 35033746.
44. Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol 2022;17:362–87. 10.1016/j.jtho.2021.11.003. 34808341.
45. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129–39. 10.3410/f.1018878.214468. 15118073.
46. Lee CK, Davies L, Wu YL, Mitsudomi T, Inoue A, Rosell R, et al. Gefitinib or Erlotinib vs Chemotherapy for EGFR Mutation-Positive Lung Cancer: Individual Patient Data Meta-Analysis of Overall Survival. J Natl Cancer Inst 2017;109:djw279. 10.1093/jnci/djw279. 28376144.
47. Yang JC, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141–51. 10.1016/s1470-2045(14)71173-8. 25589191.
48. Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Updated overall survival in a randomized study comparing dacomitinib with gefitinib as first-line treatment in patients with advanced non-small-cell lung cancer and EGFR-activating mutations. Drugs 2021;81:257–66. 10.1007/s40265-020-01441-6. 33331989.
49. Cho BC, Ahn MJ, Kang JH, Soo RA, Reungwetwattana T, Yang JC, et al. Lazertinib versus gefitinib as first-line treatment in patients with EGFR-mutated advanced non-small-cell lung cancer: results from LASER301. J Clin Oncol 2023;41:4208–17. 10.1200/jco.23.00515. 37379502.
50. Chmielecki J, Gray JE, Cheng Y, Ohe Y, Imamura F, Cho BC, et al. Candidate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat Commun 2023;14:1070. 10.1038/s41467-023-35961-y. 36849494.
51. Rotow JK, Lee JK, Madison RW, Oxnard GR, Jänne PA, Schrock AB. Real-world genomic profile of EGFR second-site mutations and other osimertinib resistance mechanisms and clinical landscape of NSCLC post-osimertinib. J Thorac Oncol 2024;19:227–39. 10.1016/j.jtho.2023.09.1453. 37806383.
52. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43. 10.3410/f.1084903.539084. 17463250.
53. Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A 2007;104:20932–7. 10.1073/pnas.0710370104. 18093943.
54. Papadimitrakopoulou VA, Wu YL, Han JY, Ahn MJ, Ramalingam SS, John T, et al. Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann Oncol 2018;29(Suppl 8):viii741. 10.1093/annonc/mdy424.064.
55. Zhang Z, Yang S, Wang Q. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark Res 2019;7:27. 10.1186/s40364-019-0179-6. 31832192.
56. Coleman N, Hong L, Zhang J, Heymach J, Hong D, Le X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021;6:100319. 10.1016/j.esmoop.2021.100319. 34837746.
57. Bertoli E, De Carlo E, Del Conte A, Stanzione B, Revelant A, Fassetta K, et al. Acquired resistance to osimertinib in EGFR-mutated non-small cell lung cancer: how do we overcome it? Int J Mol Sci 2022;23:6936. 10.3390/ijms23136936. 35805940.
58. Chmielecki J, Mok T, Wu YL, Han JY, Ahn MJ, Ramalingam SS, et al. Analysis of acquired resistance mechanisms to osimertinib in patients with EGFR-mutated advanced non-small cell lung cancer from the AURA3 trial. Nat Commun 2023;14:1071. 10.1038/s41467-023-35962-x. 36849516.
59. Ramalingam SS, Cheng Y, Zhou C, Ohe Y, Imamura F, Cho BC, et al. Mechanisms of acquired resistance to first-line osimertinib: preliminary data from the phase III FLAURA study. Ann Oncol 2018;29(Suppl 8):viii740. 10.1093/annonc/mdy424.063.
60. Bing Z, Wang W, Wang D, Ma T. Identification of RET fusion as mechanisms of resistance to EGFR tyrosine-kinase inhibitors. J Clin Oncol 2021;39(15_suppl):e21074. https://doi.org/10.1200/JCO.2021.39.15_suppl.e21074. 10.1200/JCO.2021.39.15_suppl.e21074.
61. Yao Y, Zhang M, Liu X, Zhao J, Cheng X, Zeng A, et al. RET fusion in first/third-generation EGFR-TKIs resistance in advanced non-small cell lung cancer. J Clin Oncol 2019;37(15_suppl):e20634. 10.1200/JCO.2019.37.15_suppl.e20634.
62. Wang C, Zhang Z, Sun Y, Wang S, Wu M, Ou Q, et al. RET fusions as primary oncogenic drivers and secondary acquired resistance to EGFR tyrosine kinase inhibitors in patients with non-small-cell lung cancer. J Transl Med 2022;20:390. 10.1186/s12967-022-03593-3. 36059009.
63. Xie Z, Gu Y, Xie X, Lin X, Ouyang M, Qin Y, et al. Lung adenocarcinoma harboring concomitant EGFR mutations and BRAF V600E responds to a combination of osimertinib and vemurafenib to overcome osimertinib resistance. Clin Lung Cancer 2021;22e390. 10.1016/j.cllc.2020.06.008. 32693944.
64. Meng P, Koopman B, Kok K, Ter Elst A, Schuuring E, van Kempen LC, et al. Combined osimertinib, dabrafenib and trametinib treatment for advanced non-small-cell lung cancer patients with an osimertinib-induced BRAF V600E mutation. Lung Cancer 2020;146:358–61. 10.1016/j.lungcan.2020.05.036. 32534795.
65. Pothongsrisit S, Pongrakhananon V. Targeting the PI3K/AKT/mTOR signaling pathway in lung cancer: an update regarding potential drugs and natural products. Molecules 2021;26:4100. 10.3390/molecules26134100. 34279440.
66. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–6. 10.1038/nature05945. 17625570.
67. Tan AC, Tan DS. Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. J Clin Oncol 2022;40:611–25. 10.1200/jco.21.01626. 34985916.
68. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: precision medicine takes on drug resistance. Cancer Discov 2017;7:137–55. 10.1158/2159-8290.cd-16-1123. 28122866.
69. Wu YL, Dziadziuszko R, Ahn JS, Barlesi F, Nishio M, Lee DH, et al. Alectinib in resected ALK-positive non-small-cell lung cancer. N Engl J Med 2024;390:1265–76. 10.1056/nejmoa2310532. 38598794.
70. Camidge DR, Kim HR, Ahn MJ, Yang JC, Han JY, Lee JS, et al. Brigatinib versus crizotinib in ALK-positive non-small-cell lung cancer. N Engl J Med 2018;379:2027–39. 10.3410/f.734065835.793552500. 30280657.
71. Kempf E, Rousseau B, Besse B, Paz-Ares L. KRAS oncogene in lung cancer: focus on molecularly driven clinical trials. Eur Respir Rev 2016;25:71–6. 10.1183/16000617.0071-2015. 26929424.
72. Dogan S, Shen R, Ang DC, Johnson ML, D'Angelo SP, Paik PK, et al. Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant cancers. Clin Cancer Res 2012;18:6169–77. 10.1158/1078-0432.ccr-11-3265. 23014527.
73. de Langen AJ, Johnson ML, Mazieres J, Dingemans AC, Mountzios G, Pless M, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: a randomised, open-label, phase 3 trial. Lancet 2023;401:733–46. 10.1016/s0140-6736(23)00221-0. 36764316.
74. European Medicines Agency. Lumykras [Internet]. European Medicines Agency; 2024 [cited 2025 Jan 15]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/lumykras.
75. Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical acquired resistance to KRASG12C inhibition through a novel KRAS Switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation. Cancer Discov 2021;11:1913–22. 10.1158/2159-8290.cd-21-0365. 33824136.
76. Di Federico A, Ricciotti I, Favorito V, Michelina SV, Scaparone P, Metro G, et al. Resistance to KRAS G12C inhibition in non-small cell lung cancer. Curr Oncol Rep 2023;25:1017–29. 10.1007/s11912-023-01436-y. 37378881.
77. Tsoukalas N, Kiakou M, Tsapakidis K, Tolia M, Aravantinou-Fatorou E, Baxevanos P, et al. PD-1 and PD-L1 as immunotherapy targets and biomarkers in non-small cell lung cancer. J BUON 2019;24:883–8. 31424637.
78. Reck M, Remon J, Hellmann MD. First-line immunotherapy for non-small-cell lung cancer. J Clin Oncol 2022;40:586–97. 10.1200/jco.21.01497. 34985920.
79. Zarour HM. Reversing T-cell dysfunction and exhaustion in cancer. Clin Cancer Res 2016;22:1856–64. 10.1158/1078-0432.ccr-15-1849. 27084739.
80. Lim JU, Yeo CD. Update on adjuvant therapy in completely resected NSCLC patients. Thorac Cancer 2022;13:277–83. 10.1111/1759-7714.14277. 34898012.
81. Incorvaia L, Fanale D, Badalamenti G, Barraco N, Bono M, Corsini LR, et al. Programmed death ligand 1 (PD-L1) as a predictive biomarker for pembrolizumab therapy in patients with advanced non-small-cell lung cancer (NSCLC). Adv Ther 2019;36:2600–17. 10.1007/s12325-019-01057-7. 31432460.
82. Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, et al. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Ther 2021;6:201. 10.1038/s41392-021-00572-w. 34054126.

Article information Continued

Table 1.

Checklist of clinical features for lung cancer PDTOs

Step Checklist item
Patient and sample selection - Confirm lung cancer histological subtype (e.g., adenocarcinoma, squamous cell carcinoma)
- Include detailed patient staging information (TNM classification)
- Confirm EGFR, ALK, ROS-1, KRAS, or other mutation status to mimic targeted treatment scenarios
- Collect clinical data on PD-1/PD-L1 expression for immunotherapy relevance
- Ensure ethical compliance and patient consent for sample use
Sample collection and handling - Check which samples are used to acquire patient-derived tumors
- Minimize sample handling time to preserve cell integrity
- Check whether samples include abundant non-cancerous cells
- Use optimized preservation media to maintain sample viability during transport
Organoid culture and maintenance - Select culture media and conditions specific to lung tissue requirements
- Consider co-culture with immune cells to study the immune response in organoids
- Validate growth and morphology periodically to ensure that patient-derived characteristics are maintained
Genetic and molecular analysis - Verify key genetic or molecular alteration profiles of the patients who provide samples, and confirm the timing of the tests (before or after organoid sample collection).
- For PDTOs, consider performing tests for key genetic alterations to determine whether the patient’s tumor characteristics are replicated.
- Compare organoid genetic alterations with the primary tumor.

PDTO, patient-derived tumor organoid; TNM, tumor-node-metastasis classification; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; KRAS, Kirsten rat sarcoma viral oncogene homolog; PD-1, programmed cell death protein 1; PD-L1, programmed cell death ligand 1.