Research into overcoming drug resistance in lung cancer treatment using CRISPR-Cas9 technology: a narrative review
Review Article

Research into overcoming drug resistance in lung cancer treatment using CRISPR-Cas9 technology: a narrative review

Bin Liu1#, Ziyu Wang1#, Meng Gu1#, Jinghui Wang1,2, Jinjing Tan1 ORCID logo

1Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China; 2Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Jinjing Tan, PhD. Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, No. 9 Beiguan Street, Beijing 101149, China. Email: jinjingtan@ccmu.edu.cn; Jinghui Wang, MD. Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, No. 9 Beiguan Street, Beijing 101149, China; Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China. Email: jinghuiwang2006@163.com.

Background and Objective: Lung cancer remains a leading cause of cancer-related mortality globally, with drug resistance posing a significant challenge to effective treatment. The advent of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9) technology offers a novel and precise gene-editing technology for targeting and negating drug resistance mechanisms in lung cancer. This review summarizes the research progress in the use of CRISPR-Cas9 technology for investigating and managing drug resistance in lung cancer treatment.

Methods: A literature search was conducted using the Web of Science and PubMed databases, with the following keywords: [CRISPR-Cas9], [lung cancer], [drug resistance], [gene editing], and [gene therapy]. The search was limited to articles published in English from 2002 to September 2023. From the search results, studies that utilized CRISPR-Cas9 technology in the context of lung cancer drug resistance were selected for further analysis and summarize.

Key Content and Findings: CRISPR-Cas9 technology enables precise DNA-sequence editing, allowing for the targeted addition, deletion, or modification of genes. It has been applied to investigate drug resistance in lung cancer by focusing on key genes such as epidermal growth factor receptor (EGFR), Kirsten rat sarcoma viral oncogene homolog (KRAS), tumor protein 53 (TP53), and B-cell lymphoma/leukemia-2 (BCL2), among others. The technology has shown potential in inhibiting tumor growth, repairing mutations, and enhancing the sensitivity of cancer cells to chemotherapy. Additionally, CRISPR-Cas9 has been used to identify novel key genes and molecular mechanisms contributing to drug resistance, offering new avenues for therapeutic intervention. The review also highlights the use of CRISPR-Cas9 in targeting immune escape mechanisms and the development of strategies to improve drug sensitivity.

Conclusions: The CRISPR-Cas9 technology holds great promise for advancing lung cancer treatment, particularly in addressing drug resistance. The ability to precisely target and edit genes involved in resistance pathways offers a powerful tool for developing more effective and personalized therapies. While challenges remain in terms of delivery, safety, and ethical considerations, ongoing research and technological refinements are expected to further enhance the role of CRISPR-Cas9 in improving patient outcomes in lung cancer treatment.

Keywords: Clustered regularly interspaced short palindromic repeats and clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9); lung cancer; drug resistance; gene editing; gene therapy


Submitted Jul 10, 2024. Accepted for publication Aug 14, 2024. Published online Aug 28, 2024.

doi: 10.21037/tlcr-24-592


Introduction

Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9) is a gene-editing technique that enables the precise cutting of DNA sequences, allowing for the addition, deletion, or modification of genes. Essentially, it employs a DNA sequence known as CRISPR-Cas9 to interact with target genes, forming an accurate gene-editing tool (1). CRISPR sequences are DNA sequences found in bacteria and archaea, which protect bacteria from viral infections by recognizing and cutting the DNA of invading viruses. Cas9 is an auxiliary protein of the CRISPR sequence, responsible for identifying and cutting DNA sequences that match the CRISPR sequence (2). By combining the Cas9 protein with the CRISPR sequence, precise cuts can be made to specific DNA sequences (3). CRISPR-Cas9 technology is advancing swiftly and has achieved significant milestones in various domains. For instance, a 2022 clinical study employed CRISPR-Cas9 to genetically alter donor T cells, aiming to treat pediatric patients with drug-resistant leukemia who had no remaining therapeutic options. This phase I clinical trial marked the inaugural use of “universal” CRISPR-engineered T cells in humans, signifying a pivotal evolution in deploying gene-edited cells against cancer. The researchers involved in this trial innovated and employed a novel class of precision universal genome-edited T cells (4). Furthermore, CRISPR-Cas9’s potential has been rigorously explored in the realm of chimeric antigen receptor (CAR) T-cell (CAR-T) therapy. A notable study introduced an enhanced gene-editing system capable of augmenting the efficiency of CAR-T therapies and pinpointed a low-toxicity, high-efficiency, virus-independent CAR-T manufacturing approach. The groundbreaking gene-editing technique involves tethering a modified Cas9 enzyme to a single-stranded DNA template by integrating a brief overhanging segment of double-stranded DNA (dsDNA) on both extremities. This not only acts as a blueprint for homology-directed repair but also houses the less hazardous Cas9 target sequence. In comparison with the Cas9 target sequence of dsDNA, the study using a hybrid ssDNA observed an average augmentation in knock-in efficiency and a two- to three-time greater yield. Impressively, with the application of the new Cas9-guided single-stranded templates, approximately half of the T cells adopted the new genes and were thus transformed into CAR-Ts (5). While continual enhancements in CRISPR-Cas9 technology are of paramount importance, its application in addressing existing clinical challenges holds equal significance. For instance, CRISPR-Cas9 can be employed to modulate gene expression, enabling insights into gene roles in disease progression. Alternatively, it can be used to rectify gene mutations, potentially altering the trajectory of mutation-induced diseases. In lung cancer research, CRISPR-Cas9 plays an instrumental role, with drug resistance studies being a focal area of investigation.

Lung cancer is a highly malignant tumor that ranks among the leading causes of global mortality, with a 5-year survival rate that is often less than 20% for advanced-stage disease (6,7). It includes non-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). The conventional treatment approach involves surgery, which should be the first line of action when possible (8,9). Other treatment conducted with drugs including chemotherapy, targeted therapy, and immunotherapy, are usually administered in patients with inoperable lung cancer or after surgery. Novel adjuvant therapies have emerged, which involve the use of corresponding antitumor drugs before surgery (10). These treatments aim to control tumor lesions prior to surgery and eliminate potential residual tumor cells. However, drug resistance remains a significant challenge in chemotherapy, targeted therapy, and immunotherapy, which are the focus of recent research (11-14). The emergence of drug resistance severely limits the clinical efficacy of lung cancer treatments (Figure 1) (15). The development of treatment resistance in cancer patients is a complex process driven by multiple reasons such as genetic heterogeneity, tumor microenvironment interactions, cancer stem cells, metabolic reprogramming, epigenetic alterations, and selective pressure from prolonged therapies. These diverse mechanisms enable tumor cells to evade the effects of therapeutic agents, presenting a major challenge in cancer management that requires a deeper understanding to develop more effective strategies. To overcome the challenge of treatment resistance in cancer, various therapeutic strategies have been explored. These include targeting multiple signaling pathways simultaneously, leveraging combination therapies to prevent the emergence of resistant clones, developing novel drug classes that can overcome specific resistance mechanisms, utilizing immunotherapies to harness the body’s immune system, and employing liquid biopsies to monitor the evolution of resistance, which all leads to personalized treatment.

Figure 1 The main treatment methods of lung cancer and the risk of drug resistance. CRISPR-Cas9, CRISPR and CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeats.

Before the advent of CRISPR-Cas9 technology, researchers had been working to improve drug resistance by altering by-pass signaling of the treatment target, which fail to work as usual, but the results were not ideal. CRISPR-Cas9 technology has emerged as a promising approach to address the challenge of cancer drug resistance. By precisely editing the genome of tumor cells, CRISPR-Cas9 can disrupt key resistance-conferring genes or sensitize cancer cells to existing therapies. This technology allows for targeted manipulation of genetic and epigenetic factors driving resistance, enhancing the efficacy of cancer treatments. This technique also allows researchers to study genetic variation and drug resistance mechanisms in lung cancer cells with persistent effects, enabling the development of more precise treatments. In this review, we summarize the progress in CRISPR-Cas9 research related to drug resistance in lung cancer. Our aim is to provide a valuable reference for the further development of CRISPR-Cas9-based gene intervention systems, which may ultimately improve the management of drug resistance in patients with lung cancer. We present this article in accordance with the Narrative Review reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-592/rc).


Methods

Literature selection

The word “CRISPR” first appeared as a name for a technology in 2002; therefore, in this review, we only searched for literature published between 2002 and September 2023. The database searched was Web of Science and PubMed, and the language of the literature was specified as English. The types of literature searched were research articles and reviews. Keywords searched include but were not limited to “CRISPR-Cas9”, “lung cancer”, “drug resistance”, “gene editing”, and “gene therapy”. The references from all the retrieved articles were manually read and are listed in the list of references. The search strategy is summarized in Table 1.

Table 1

Summary of the search strategy

Items Specification
Date of search September 6, 2023
Databases and other sources searched Web of Science, PubMed
Search terms used ((TS=(CRISPR-Cas9)) OR TS=(gene therapy)) OR TS=(lung cancer) AND TS=(drug resistance) AND TS=(gene editing)
Timeframe From January 1, 2002, to September 6, 2023
Inclusion criteria Article or review in English
Selection process Double-blind search, aggregated together for inclusion in the review

CRISPR-Cas9, CRISPR and CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeats; TKI, tyrosine kinase inhibitor.

Use of CRISPR-Cas9 in ameliorating drug resistance in lung cancer

This section discusses four main topics: CRISPR-Cas9 technology’s role in targeting gene expression or mutations related to lung cancer drug resistance, its application in addressing lung cancer’s drug-resistant signaling pathways, its interaction with immune-associated mechanisms in lung cancer drug resistance, and its potential for identifying the key genes that contribute to drug resistance in lung cancer.

Investigating drug resistance in lung cancer with CRISPR-Cas9-targeted genes

CRISPR-Cas9 can accurately target the critical genes of lung cancer cells, such as tumor-suppressor genes and proliferation-related genes, to inhibit tumor growth and spread (16,17). Several crucial genes involved in regulating drug resistance in lung cancer have been identified, among which (I) epidermal growth factor receptor (EGFR); (II) Kirsten rat sarcoma viral oncogene homolog (KRAS); (III) tumor protein 53 (TP53); (IV) B-cell lymphoma/leukemia-2 (BCL2); (V) phosphoinositol 3-kinase α (PIK3CA); (VI) anaplastic lymphoma kinase (ALK); and (VII) mesenchymal-epithelial transition (MET), are discussed in detail below.

  • EGFR is one of the most common mutated genes in lung cancer, with mutations leading to EGFR overactivation, thus promoting tumor growth and metastasis (18). EGFR mutations are closely associated with sensitivity to targeted therapy, but resistance can also develop during treatment (14,19). EGFR can guide the use of multiple targeted drugs, and CRISPR-Cas9 technology has been used to achieve EGFR gene expression inhibition and mutation repair. Researchers have designed specific RNA sequences using CRISPR-Cas9 technology to precisely cut the DNA sequence of the EGFR gene, thereby inhibiting its expression (20). This method has been demonstrated in both in vitro and in vivo experiments and has been shown to inhibit the growth of lung cancer cells (21). Additionally, CRISPR-Cas9 technology can be used to design specific RNA sequences that target Cas9 at the EGFR gene’s mutation site, after which a DNA repair template can be used to repair the mutation (22). The repair template, which can be single-stranded DNA or dsDNA, contains the normal EGFR gene sequence. The repair template works alongside Cas9 to restore the mutant site to the normal EGFR gene sequence, thus reinstating its normal function (23,24).
  • KRAS is another frequently mutated gene, with mutations causing excessive KRAS activation that promotes tumor growth and metastasis. KRAS mutations are found in approximately 25% of those with NSCLC, which is a common type of lung cancer (25). Oncogenic mutations in KRAS typically occur at hotspots in the protein (e.g., codons 12, 13, and 61), inducing protumorigenic signaling via downstream effector pathways by raising steady-state levels of KRAS proteins in the guanosine-5'-triphosphate (GTP)-bound state. A few examples of these pathways include mitogen-activated protein kinase (MTK) and phosphatidylinositol 3-kinase (PI3K) (26,27). It has been reported that CRISPR can be used to perform a positive-selection deep mutational scanning screen for mutations in KRAS G12C that confer resistance to KRAS G12C inhibition in Ba/F3 cells cultured for 7 days in the absence of interleukin-3 and in the presence of one of two KRAS G12C inhibitors, MRTX1257, or sotorasib. CRISPR-Cas9 may have the same effect as KRAS G12C inhibitors in countering drug resistance (28-30).
  • TP53 is a tumor-suppressor gene. Mutations result in the loss of TP53 function, promoting tumor growth and metastasis (31). Several studies have investigated the use of CRISPR-Cas9 to knock down TP53 in lung cancer cells (32-34). One study demonstrated the existence of an autophagic switch that occurs in response to the antitumoral drug cisplatin, which can affect the sensitivity of cancer cells to this treatment (35). CRISPR-Cas9 was also used to generate a set of isogenic NSCLC cell lines expressing wild-type p53 (H460wt) and a knockout of p53 (H460crp53) in order to determine how cisplatin sensitivity and autophagic function are affected by p53 status (36).
  • BCL2 has been identified as a contributing factor to the development of resistance against EGFR-tyrosine kinase inhibitors (TKIs) in NSCLC (37). A study reports low levels of BCL2 expression detected in NSCLC samples collected prior to resistance development and, conversely, report highly levels detected subsequent to the emergence of resistance (38). This suggests that upregulation of BCL2 may be a latent driver for resistance exploitation and that targeting BCL2 may be a useful strategy in treating NSCLC with acquired resistance (39). There have been no studies that have employed CRISPR to knock out BCL2 in lung cancer. However, for other cancer types, such as prostate or liver cancer, CRISPR-Cas9 has been used to successfully interfere with BCL2 expression in vitro and in vivo and to block the resistance caused by BCL2 expression (40,41).
  • PIK3CA is a signaling pathway gene, whose mutations cause the overactivation of PIK3CA, thus promoting tumor growth and metastasis (42). There are currently no studies that have specifically focused on using CRISPR-Cas9 to target PIK3CA in lung cancer. However, similar studies have been conducted in breast cancer. CRISPR-Cas9-based single-guided RNA (sgRNA) knockout screens were carried out on MCF7 and T47D estrogen receptor (ER)+ breast cancer cell lines mutated in PIK3CA to investigate possible mediators of inhibitor response (43). In one study, the researchers used CRISPR/Cas9 technology to knock out the PIK3CA gene in these cell lines and then treated a group with dimethyl sulfoxide and another group with PI3Kα inhibitor (alpelisib or taselisib) for 20 days. The genomic DNA of each group was extracted, and the sgRNA sequences were amplified via polymerase chain reaction and then retrieved via next-generation sequencing. The results of this study suggest that genomic alterations involving the genes that potentially affect mechanistic target of rapamycin (mTOR) activity are present in a significant proportion of primary and metastatic PIK3CA-mutant breast tumors and may play a role in limiting the sensitivity to PI3Kα inhibition. However, this study did not specifically arrive at a conclusion regarding the effects of the CRISPR knockout of PIK3CA on the sensitivity to PI3Kα inhibitors (44).
  • ALK is a mutated gene that causes overactivation of ALK, promoting tumor growth and metastasis (45). Oncogenic ALK gene rearrangements occur in a small percentage of patients with NSCLC and result in the overexpression and ligand-independent activation of ALK (46). The nature of the fusion partner determines the degree of activation (47). Although echinoderm microtubule-associated protein-like 4 (EML4) is the most common fusion partner, multiple other fusion partners have been reported (48,49). Five ALK inhibitors have been approved by the United States Food and Drug Administration (FDA) for use in treating NSCLC (alectinib, brigatinib, ceritinib, lorlatinib, and crizotinib) (50). Although these inhibitors have shown efficacy, resistance mechanisms to them exist and are an active area of research. ALK gene rearrangements can lead to drug resistance in NSCLC (51). Resistance mechanisms mediated by ALK include ALK amplification, copy number gain, and mutations of the ALK kinase domain. Depending on the resistance mechanism, high-potency, second-generation ALK-TKIs can be used (to target ALK-dependent resistance); otherwise, agents that target multiple pathways in addition to ALK (to target ALK-independent resistance) can be used (52). In other type of cancer, neuroblastoma, researchers used CRISPR-Cas9-based screening to identify genes associated with ALK inhibitor resistance in neuroblastoma cell lines. One study has found that mutations in ALK signaling pathways confer resistance to ALK inhibitors in neuroblastoma cells, resulting in collateral vulnerability (53).
  • MET is a signaling pathway gene, with mutations leading to overactivation of MET, thud promoting tumor growth and metastasis. MET mutations are closely associated with resistance to targeted therapies. CRISPR-Cas9’s regulation of these genes primarily involves expression regulation and mutation repair. Some studies have shown that the activation of MET negatively affects the effectiveness of TKIs in NSCLC due to crosstalk between MET and receptor tyrosine kinase (RTK) signaling pathways which starts with the activation of rat sarcoma virus (Ras) protein, a small G protein anchored to the plasma membrane(54,55).

In the evasion of cell death induced by EGFR-TKIs, MET amplification promotes downstream signal transduction through bypass activation. Therefore, MET amplification is an important mechanism of EGFR-TKI resistance, with a prevalence of 5–21% observed after first- or second-line EGFR-TKI resistance, ~5% after first-line therapy, and ~19% after later-line osimertinib resistance (56). Another study found that MET alterations are a recurring and actionable resistance mechanism in ALK-positive lung cancer. This suggests that targeting both ALK and MET pathways may be an effective treatment strategy for patients with this type of cancer. In one report, two patients with ALK-positive lung cancer and acquired MET alterations achieved rapid clinical and radiographic response to ALK-MET combination therapy. Therefore, the study provides rationale for pursuing ALK-MET combination therapy in clinic (57). Studies have also demonstrated that CRISPR-Cas9 can effectively interfere with MET both in vivo and in vitro (58,59). Therefore, it is believed that the CRISPR-Cas9 function of ALK gene editing, when combined with CRISPR-Cas9’s simultaneous targeting of MET and ALK expression or mutation, may have a role in regulating drug resistance in NSCLC.

CRISPR-Cas9 targeting signaling pathways related to drug resistance in lung cancer

The complex process of tumor drug resistance often transcends the function of single genes, making the study of relevant signaling pathways particularly crucial (60). CRISPR-Cas9 may influence the activity of the signaling pathways related to lung cancer drug resistance by regulating key nodes, thereby improving drug sensitivity (Figure 2). The currently known signaling pathways related to drug resistance in lung cancer include (I) the PI3K/protein kinase B (Akt)/mTOR signaling pathway; (II) Wnt/β-catenin signaling pathway; (III) Notch signaling pathway; (IV) Hedgehog signaling pathway; (V) nuclear factor-κB (NF-κB) signaling pathway; (VI) adenosine triphosphate (ATP)-binding cassette transporter signaling pathway; (VII) DNA repair signaling pathway; and (VIII) rat sarcoma virus (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway. (I) The PI3K/Akt/mTOR signaling pathway is essential for cell growth and survival and is involved in a variety of cellular processes, including cell proliferation, apoptosis, metabolism, and angiogenesis (61). (II) The Wnt/β-catenin signaling pathway is an important pathway for embryonic development and tissue regeneration. It has been implicated in numerous cellular processes, including cell proliferation, differentiation, and epithelial-mesenchymal transition (EMT) (62). (III) The Notch signaling pathway is a vital cell fate-determining pathway and involved in various cellular processes, including cell proliferation, differentiation, and cancer cell stemness (63). (IV) The Hedgehog signaling pathway is another critical cell fate-determining pathway and participates in numerous cellular processes, including cell proliferation, differentiation, and apoptosis (64). (V) The NF-κB signaling pathway, an essential inflammatory and immune response pathway, is involved in various cellular processes, including cell proliferation, apoptosis, inflammation, and immune response (65). (VI) The ABC transporter signaling pathway, includes ABC transporters, which are a class of transmembrane proteins that transport chemotherapy drugs from the intracellular to extracellular space, thus reducing drug accumulation in cells and decreasing drug efficacy (66). (VII) The DNA repair signaling pathway is closely linked to DNA repair, which is a vital repair mechanism for cells to ameliorate the damage to DNA caused by chemotherapy drugs, thereby reducing drug efficacy (67). (VIII) The RAS/RAF/MEK/ERK signaling pathway, which is critical to cell growth and survival pathway, is implicated in various cellular processes, including cell proliferation, apoptosis, metabolism, and angiogenesis. Abnormal activation of the RAS/RAF/MEK/ERK signaling pathway is closely related to the development of various tumors and is an essential mechanism of drug resistance in tumor-targeted therapy (68).

Figure 2 Use of CRISPR-Cas9 technology for the editing of tumor driver genes and its effect on the biological behavior of tumor cells. EGFR, epidermal growth factor receptor; KRAS, Kirsten rat sarcoma viral oncogene homolog; TP53, tumor protein 53; BCL2, B-cell lymphoma/leukemia-2; ALK, anaplastic lymphoma kinase; CRISPR-Cas9, CRISPR and CRISPR-associated protein 9; CRISPR, clustered regularly interspaced short palindromic repeats.

CRISPR-Cas9 reshapes biological function by making different adjustments at multiple points within the signaling pathway. For example, in the PI3K/Akt/mTOR signaling pathway, CRISPR-Cas9 can target several genes, such as PIK3CA, AKT1, and MTOR. Knockout of the PIK3CA gene can inhibit PI3K signaling pathway activity by altering key gene regulations, thus affecting cell proliferation and growth in breast cancer. Moreover, by knocking out the AKT1 gene in prostate cancer, the expression level of Akt protein can be reduced, impacting the downstream signaling pathway activity of Akt, another essential molecule in this signaling pathway. Simultaneously, MTOR gene knockout can decrease the phosphorylation level of mTOR protein, affecting the activity of the mTOR signaling pathway at the level of protein posttranslational modification (69,70).

CRISPR-Cas9 targeting immune escape in lung cancer resistance

Immune escape refers to the phenomenon in which certain pathogens or tumor cells evade the host immune system’s attack and clearance through various means, resulting in disease persistence or recurrence (71). The targeting of immune escape by CRISPR-Cas9 in lung cancer resistance is primarily based on the regulation of key genes involved in the process (72). The key genes of immune escape can be categorized as follows: (I) immune recognition genes, which includes T-cell receptor genes, B cell receptor genes, MHC molecular genes, and others, all of which play a critical role in antigen recognition and identification within the immune system (73); (II) immune signal transduction genes, which encompasses those related to the JAK-STAT signaling pathway, NF-κB signaling pathway, and PI3K-Akt signaling pathway, among others, and which are essential for signal transduction and immune response regulation within the immune system (74); (III) immune regulatory genes, which include immunosuppressive factor genes and immunostimulatory factor genes, which play a significant role in immune response regulation within the immune system (75); (IV) immune effector genes, comprising cytotoxin genes, chemokine genes, and cytokine genes, which are crucial for pathogen killing, immune cell attraction, and immune response regulation within the immune system (76); and (V) immune escape genes, which include tumor mutation genes, pathogen mutation genes, and immunosuppressive factor genes, which play a vital role in immune system evasion.

The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway is discussed as an example here. JAK-STAT signaling involves five key proteins: JAK protein, STAT protein, suppressor of cytokine signaling (SOCS) protein, cytokine receptor, and protein inhibitors of activated STAT (PIAS) protein. The JAK and STAT proteins are involved in the phosphorylation and dephosphorylation processes, while the SOCS and PIAS proteins serve as negative regulatory factors of JAK-STAT (77). The cytokine receptor is responsible for binding with cytokines and recruiting JAK and STAT proteins to regulate immune cell differentiation and function (78). CRISPR-Cas9 technology is capable of a variety of functions, such as knockout, addition, repair, and modification. Previous studies have demonstrated that CRISPR-Cas9 can significantly regulate the activity of the JAK-STAT pathway (79-81). Additionally, researchers have used CRISPR-Cas9 technology to target immune escape mechanisms in lung cancer cells, such as those involving programmed death ligand 1 (PD-L1) and transforming growth factor-β (TGF-β) (82). By cutting these genes and reducing their expression levels, the immune cells’ ability to attack lung cancer cells is enhanced. Moreover, novel immune cells have been developed, including CAR-Ts and natural killer cells, which more effectively target lung cancer cells (83).

Application of CRISPR-Cas9 in identifying the key genes and molecular mechanisms of lung cancer drug resistance

In addition to being applied in the knockout of the genes known to be related to resistance, CRISPR-Cas9 has been used as a powerful tool for exploring novel key genes and molecular mechanisms of drug resistance in lung cancer. By using CRISPR-Cas9 to knock out specific genes in lung cancer cells, researchers can identify which genes are integral to drug resistance and which genes can be targeted to overcome drug resistance (84,85). For instance, a study published in the prestigious journal Cancer Research in 2017 employed CRISPR-Cas9 to knock out the gene for the myeloid cell leukemia-1 (MCL-1) protein in lung cancer cells (86). The authors of this paper discovered that MCL-1 plays a critical role in mediating resistance to chemotherapy drugs such as cisplatin and etoposide. By targeting MCL-1 using CRISPR-Cas9 or other drugs, they were able to sensitize lung cancer cells to chemotherapy. Another study published in the esteemed journal Nature Communications in 2018 used CRISPR-Cas9 to knock out the gene for the Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1 (KEAP1) protein in lung cancer cells (87). KEAP1 is a negative regulator of the nuclear factor erythroid-2-related factor 2 (NRF2) pathway, which is often overexpressed in lung cancer and contributes to drug resistance. It is an E3 ubiquitin ligase, a critical regulator that targets NRF2 for ubiquitylation and subsequent degradation (88). The researchers found that knocking out KEAP1 increased the sensitivity of lung cancer cells to chemotherapy drugs such as cisplatin and paclitaxel. These studies highlight the potential of CRISPR-Cas9 to identify key genes and molecular mechanisms of drug resistance in lung cancer and to aid in the development of new therapeutic strategies to overcome drug resistance. However, further research is required before the intricate molecular mechanisms of drug resistance in lung cancer can be more fully comprehended and to develop safe and effective methods for delivering CRISPR-Cas9 to lung cancer cells in humans.

As previously mentioned, NRF2, which is encoded by gene NFE2L2, is a transcription factor that plays a pivotal role in regulating the antioxidant response in cells. In lung cancer, NRF2 is frequently overexpressed, leading to increased resistance to chemotherapy and radiation therapy (89-93). The use of CRISPR-Cas9 to knock out NRF2 in lung cancer cells has been shown to increase the sensitivity of these cells to chemotherapy and radiation therapy. Several studies have investigated the efficacy of CRISPR-Cas9 in knocking out NRF2 in lung cancer cells. For instance, according to a study published in 2018, knocking out NRF2 in lung cancer cells increased their sensitivity to chemotherapy (94). In another study published in 2019, CRISPR-Cas9 was used to knock out NRF2 in lung cancer cells and reduce tumor formation in mice (95). Schlafen 11 (SLFN11) is a gene involved in DNA repair and replication. Recent studies have shown that SLFN11 expression is associated with lung cancer sensitivity to chemotherapy and immunotherapy. Knocking out SLFN11 using CRISPR-Cas9 in lung cancer cells has been shown to reduce their sensitivity to chemotherapy and immunotherapy (96). One study demonstrated that SLFN11 can sensitize tumor cells to interferon-γ (IFN-γ)-mediated T-cell killing (97), while another study found that SLFN11 is responsible for platinum treatments activated immune-related pathways in high-grade serous ovarian cancer (98). In SCLC, poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor was found capable of regulating SLFN11 expression and demonstrated therapeutic synergy with temozolomide (96). In line with this reasoning, CRISPR-Cas9 may have a similar function in lung cancer.

Aurora B is a protein that plays a crucial role in cell division and is often overexpressed in lung cancer cells. Recent studies have investigated the use of CRISPR-Cas9 in targeting Aurora B in lung cancer cells as a potential therapeutic strategy. This research indicates that using CRISPR-Cas9 to knock out Aurora B in lung cancer cells reduces their ability to form tumors in mice (99). Another study demonstrated that using CRISPR-Cas9 to knock out Aurora B in lung cancer cells increases their sensitivity to chemotherapy drugs such as paclitaxel (100). Polybromo-1 (PBRM1) is a gene that plays a crucial role in regulating gene expression and is often mutated in lung cancer. Recent research has investigated the use of CRISPR-Cas9 to knock out PBRM1 in lung cancer cells as a potential therapeutic strategy. One study reported that knocking out PBRM1 using CRISPR-Cas9 in lung cancer cells reduced their ability to form tumors in mice (101). Another study revealed that knocking out PBRM1 using CRISPR-Cas9 in lung cancer cells increased their sensitivity to chemotherapy drugs such as cisplatin and gemcitabine (101).

Glycan-related genes play a pivotal role in the complex mechanisms underlying multidrug resistance (MDR) in cancer. Altered glycosylation patterns, facilitated by aberrant expression or mutations in genes involved in glycan biosynthesis and remodeling, can significantly influence drug transport, cell signaling, and adhesion - all of which contribute to the development of MDR. For instance, the upregulation of glycoprotein drug transporters, such as P-glycoprotein, is a well-characterized mechanism that enables cancer cells to actively efflux chemotherapeutic agents (102). Furthermore, glycans can modulate key signaling cascades linked to MDR, including the PI3K/Akt, MAPK, and Wnt pathways, promoting cell survival and proliferation (103,104). In this context, the emergence of CRISPR-Cas9 technology holds great promise for targeting glycan-related genes to overcome MDR. One study demonstrated that via CRISPR-Cas9 editing, deacetylated glycan sialic acid (Sia) regulated a MDR pathway in colon and lung cancer and disrupt drug efflux and alter glycosylation patterns (105). Additionally, combinatorial CRISPR approaches targeting multiple glycan-associated genes may provide a more comprehensive strategy to address the complexity of MDR mechanisms, thereby resensitizing resistant cancer cells and improving treatment outcomes.

Future research directions in the use of CRISPR-Cas9 technology to counter treatment resistance in lung cancer

Current advancements in CRISPR-Cas9 technology primarily center around enhancing editing efficacy and safety while fostering novel applications. This emphasis on the enhancement of editing efficacy can be categorized into two domains: the inception of new editing instruments and refinement of established research tools. Notably, considerable strides have been made within this research dimension. Comprehensive analyses of viral genomes have unearthed a plethora of potential CRISPR-based genome-editing mechanisms. One groundbreaking study completed a thorough examination of the CRISPR-Cas system in viruses, particularly phages, that plague bacteria and archaea. Astonishingly, the research identified approximately 6,000 phages equipped with CRISPR-Cas systems, spanning every acknowledged variant of the CRISPR-Cas framework. These systems represent promising tools for potential gene-editing applications (106). A case in point is the recently identified CRISPR-Cas12n (107). This system is a derivative of the V-U4-type CRISPR-Cas, closely related to TnpB—the transposition-linked nuclease and the putative progenitor of V-type CRISPR frameworks, and may be an early evolutionary bridge in TnpB’s transition into other V-type Cas nucleases.

Concerning specific diseases, specialized gene-editing tools promise enhanced outcomes. In a seminal study, researchers dissected DNA sequences of the fetal hemoglobin gene posttherapeutic editing using both CRISPR-Cas9 and base editors. Their findings indicated that Cas9 editing frequently yielded multifaceted, unsupervised editing across varied DNA sequences. Consequently, they innovated base editing which, as the results suggested, augmented fetal hemoglobin expression more effectively than did CRISPR-Cas9. Furthermore, the study demonstrated that the diversity of insertion and deletions instigated by CRISPR-Cas9 could spawn unanticipated phenotypic fluctuations. Such challenges might be circumvented through base editing, offering a potentially safer and more efficient therapeutic strategy for ailments such as sickle cell disease and β-thalassemia (108). Concurrently, the pioneering team behind the CRISPR-Cas9 technology has persisted in refining this gene editing approach. Their recent revelation is Fanzor, a novel RNA-steered DNA-cleaving enzyme observed in eukaryotes. Notably, this nascent CRISPR-like system is reconfigurable, allowing it to edit the human genome. Furthermore, the compact nature of the Fanzor system renders it more amenable to cell and tissue delivery as compared to the traditional CRISPR-Cas mechanisms. Additionally, lacking paracrine cleavage activity, Fanzor allows for greater precision in genome editing. The implications of this seminal work suggest that RNA-induced DNA cleavage might be ubiquitous across organisms (109).

Drawing from these investigations, we can speculate that deploying CRISPR-Cas9 in lung cancer drug resistance research may lead innovations in the following domains: (I) improvement of sgRNA design; (II) refinement of the Cas9 structure, active center, and specificity; (III) enhancement of vectors and delivery modalities, including the harnessing of complementary techniques such as extracellular vesicle delivery; and (IV) the customization of technology to align with specific tumor attributes or those of drug-resistant tumor cells.

As CRISPR-Cas9 technology continues to advance at a rapid pace, an increasing volume of research is being published on the addressing safety concerns pertaining to its use in humans. A recent study noted that CRISPR/Cas9 gene editing can induce cytotoxicity and genomic instability depending on the target sequence sites within the human genome. This undesirable effect operates via the pivotal tumor suppressor protein p53 and is influenced by various epigenetic factors in the DNA sequence proximate to the edited site and its surrounding area. Through computational methods, the authors of this study scrutinized the widely used CRISPR libraries tailored for human cells and identified 3,300 target sequence sites exhibiting pronounced toxic effects. They further revealed that nearly 15% of human genes involve at least one editing site associated with toxicity (110).

Other studies underscored the risks intrinsic to CRISPR therapies, emphasizing the potential for damage to the genome (111,112). Despite CRISPR genome editing demonstrating remarkable efficacy, its safety profile is not uniformly positive. At times, fragmented chromosomes fail to restore their functionality, undermining genome stability, which may lead to adverse oncogenic effects over time. CRISPR therapy has been broadly applied to address an array of conditions, from cancers to genetic syndromes; however, evaluations of its impact on immune system white blood cells indicates that up to 10% of treated cells exhibit a significant loss of cytogenetic material. This can in turn induce genomic instability, elevating cancer risks. The inaugural CRISPR clinical trial, conducted by the University of Pennsylvania in 2020, employed CRISPR technology on T cells from a donor. The T cells were engineered to express a receptor targeting cancer cells and to concurrently disrupt genes encoding the native receptor to avert autoimmunity (113). Another study discussed whether the potential advantages of CRISPR therapy might be overshadowed by the inherent risks of genomic cleavage (114). This speculation was predicated on the notion that damaged DNA may not invariably undergo successful repair, leading to the loss of significant chromosomal segments or even entire chromosomes. Such chromosomal aberrations can render the genome unstable and in a state reminiscent to that observed in cancer cells. In extreme scenarios, CRISPR therapy might inadvertently catalyze cellular carcinogenesis (111).

Nonetheless, in the specific context of lung cancer drug resistance research, our stance is that the immediate priority may lean toward extending patient survival and elevating the quality of treatment. Decisions on genomic implications may be deferred pending a more comprehensive understanding provided by ongoing research.


Conclusions

CRISPR-Cas9 was first reported to have the ability to reprogram DNA in 2012 (115). Over the past decade, CRISPR-Cas9 technology has achieved several milestones: it is the subject of a Nobel Prize for chemistry, and the first drug developed based on CRISPR/Cas9 technology will be approved this year for the treatment of sickle cell and other diseases (116). This review outlines the progress in the research on the treatment of drug resistance in lung cancer using CRISPR-Cas9 technology. The basic principle of CRISPR-Cas9 technology and its application in lung cancer treatment are discussed, specifically the work to overcome drug resistance in lung cancer via CRISPR-Cas9-targeted genes. The purpose of this review is to serve as a valuable reference for the further development of CRISPR-Cas9-based gene intervention systems, which may ultimately improve the management of drug resistance in patients with lung cancer.


Acknowledgments

We would like to thank the operators of the home-for-research website for the use of their platform in writing this review.

Funding: This work was supported by the Beijing Municipal Public Welfare Development and Reform Pilot Project for Medical Research Institutes (No. JYY2023-14) and the “Yun He” Talent Project of Tongzhou District (No. YHLD2019039).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-592/rc

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-592/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-592/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Ma Y, Zhang L, Huang X. Genome modification by CRISPR/Cas9. FEBS J 2014;281:5186-93. [Crossref] [PubMed]
  2. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 2014;346:1258096. [Crossref] [PubMed]
  3. Zhang D, Zhang Z, Unver T, et al. CRISPR/Cas: A powerful tool for gene function study and crop improvement. J Adv Res 2021;29:207-21. [Crossref] [PubMed]
  4. Ottaviano G, Georgiadis C, Gkazi SA, et al. Phase 1 clinical trial of CRISPR-engineered CAR19 universal T cells for treatment of children with refractory B cell leukemia. Sci Transl Med 2022;14:eabq3010. [Crossref] [PubMed]
  5. Shy BR, Vykunta VS, Ha A, et al. High-yield genome engineering in primary cells using a hybrid ssDNA repair template and small-molecule cocktails. Nat Biotechnol 2023;41:521-31. [Crossref] [PubMed]
  6. Wang SW, Gao C, Zheng YM, et al. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol Cancer 2022;21:57. [Crossref] [PubMed]
  7. Zhan T, Rindtorff N, Betge J, et al. CRISPR/Cas9 for cancer research and therapy. Semin Cancer Biol 2019;55:106-19. [Crossref] [PubMed]
  8. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7-33. [Crossref] [PubMed]
  9. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin 2016;66:115-32. [Crossref] [PubMed]
  10. Wang M, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med 2021;27:1345-56. [Crossref] [PubMed]
  11. Reck M, Remon J, Hellmann MD. First-Line Immunotherapy for Non-Small-Cell Lung Cancer. J Clin Oncol 2022;40:586-97. [Crossref] [PubMed]
  12. Tan AC, Tan DSW. Targeted Therapies for Lung Cancer Patients With Oncogenic Driver Molecular Alterations. J Clin Oncol 2022;40:611-25. [Crossref] [PubMed]
  13. Duma N, Santana-Davila R, Molina JR. Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clin Proc 2019;94:1623-40. [Crossref] [PubMed]
  14. Hirsch FR, Scagliotti GV, Mulshine JL, et al. Lung cancer: current therapies and new targeted treatments. Lancet 2017;389:299-311. [Crossref] [PubMed]
  15. Liu WJ, Du Y, Wen R, et al. Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther 2020;206:107438. [Crossref] [PubMed]
  16. Wang H, La Russa M, Qi LS. CRISPR/Cas9 in Genome Editing and Beyond. Annu Rev Biochem 2016;85:227-64. [Crossref] [PubMed]
  17. Chiou SH, Winters IP, Wang J, et al. Pancreatic cancer modeling using retrograde viral vector delivery and in vivo CRISPR/Cas9-mediated somatic genome editing. Genes Dev 2015;29:1576-85. [Crossref] [PubMed]
  18. Lynch TJ, Bell DW, Sordella R, 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. [Crossref] [PubMed]
  19. Liu Q, Yu S, Zhao W, et al. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018;17:53. [Crossref] [PubMed]
  20. Jin H, Shi Y, Lv Y, et al. EGFR activation limits the response of liver cancer to lenvatinib. Nature 2021;595:730-4. [Crossref] [PubMed]
  21. Tang H, Shrager JB. CRISPR/Cas-mediated genome editing to treat EGFR-mutant lung cancer: a personalized molecular surgical therapy. EMBO Mol Med 2016;8:83-5. [Crossref] [PubMed]
  22. Wang CS, Chang CH, Tzeng TY, et al. Gene-editing by CRISPR-Cas9 in combination with anthracycline therapy via tumor microenvironment-switchable, EGFR-targeted, and nucleus-directed nanoparticles for head and neck cancer suppression. Nanoscale Horiz 2021;6:729-43. [Crossref] [PubMed]
  23. Yoon AR, Jung BK, Choi E, et al. CRISPR-Cas12a with an oAd Induces Precise and Cancer-Specific Genomic Reprogramming of EGFR and Efficient Tumor Regression. Mol Ther 2020;28:2286-96. [Crossref] [PubMed]
  24. Park MY, Jung MH, Eo EY, et al. Generation of lung cancer cell lines harboring EGFR T790M mutation by CRISPR/Cas9-mediated genome editing. Oncotarget 2017;8:36331-8. [Crossref] [PubMed]
  25. Reck M, Carbone DP, Garassino M, et al. Targeting KRAS in non-small-cell lung cancer: recent progress and new approaches. Ann Oncol 2021;32:1101-10. [Crossref] [PubMed]
  26. Zhu G, Pei L, Xia H, et al. Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol Cancer 2021;20:143. [Crossref] [PubMed]
  27. Tomasini P, Walia P, Labbe C, et al. Targeting the KRAS Pathway in Non-Small Cell Lung Cancer. Oncologist 2016;21:1450-60. [Crossref] [PubMed]
  28. Hallin J, Bowcut V, Calinisan A, et al. Anti-tumor efficacy of a potent and selective non-covalent KRAS(G12D) inhibitor. Nat Med 2022;28:2171-82. [Crossref] [PubMed]
  29. Bender G, Fahrioglu Yamaci R, Taneri B. CRISPR and KRAS: a match yet to be made. J Biomed Sci 2021;28:77. [Crossref] [PubMed]
  30. Awad MM, Liu S, Rybkin II, et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N Engl J Med 2021;384:2382-93. [Crossref] [PubMed]
  31. Jiao XD, Qin BD, You P, et al. The prognostic value of TP53 and its correlation with EGFR mutation in advanced non-small cell lung cancer, an analysis based on cBioPortal data base. Lung Cancer 2018;123:70-5. [Crossref] [PubMed]
  32. Haapaniemi E, Botla S, Persson J, et al. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat Med 2018;24:927-30. [Crossref] [PubMed]
  33. Hou J, Cao X, Cheng Y, et al. Roles of TP53 gene in the development of resistance to PI3K inhibitor resistances in CRISPR-Cas9-edited lung adenocarcinoma cells. Cell Biol Toxicol 2020;36:481-92. [Crossref] [PubMed]
  34. Mirgayazova R, Khadiullina R, Chasov V, et al. Therapeutic Editing of the TP53 Gene: Is CRISPR/Cas9 an Option? Genes (Basel) 2020;11:704. [Crossref] [PubMed]
  35. Patel NH, Xu J, Saleh T, et al. Influence of nonprotective autophagy and the autophagic switch on sensitivity to cisplatin in non-small cell lung cancer cells. Biochem Pharmacol 2020;175:113896. [Crossref] [PubMed]
  36. Peng DH, Rodriguez BL, Diao L, et al. Th17 cells contribute to combination MEK inhibitor and anti-PD-L1 therapy resistance in KRAS/p53 mutant lung cancers. Nat Commun 2021;12:2606. [Crossref] [PubMed]
  37. Hasan A, Haque E, Hameed R, et al. Hsp90 inhibitor gedunin causes apoptosis in A549 lung cancer cells by disrupting Hsp90:Beclin-1:Bcl-2 interaction and downregulating autophagy. Life Sci 2020;256:118000. [Crossref] [PubMed]
  38. Liu Z, Gao W. Synergistic effects of Bcl-2 inhibitors with AZD9291 on overcoming the acquired resistance of AZD9291 in H1975 cells. Arch Toxicol 2020;94:3125-36. [Crossref] [PubMed]
  39. Cheong HT, Xu F, Choy CT, et al. Upregulation of Bcl2 in NSCLC with acquired resistance to EGFR-TKI. Oncol Lett 2018;15:901-7. [Crossref] [PubMed]
  40. Thomalla D, Beckmann L, Grimm C, et al. Deregulation and epigenetic modification of BCL2-family genes cause resistance to venetoclax in hematologic malignancies. Blood 2022;140:2113-26. [Crossref] [PubMed]
  41. Rodríguez Y, Unno K, Truica MI, et al. A Genome-Wide CRISPR Activation Screen Identifies PRRX2 as a Regulator of Enzalutamide Resistance in Prostate Cancer. Cancer Res 2022;82:2110-23. [Crossref] [PubMed]
  42. Tan AC. Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer (NSCLC). Thorac Cancer 2020;11:511-8. [Crossref] [PubMed]
  43. Ros S, Wright AJ, D'Santos P, et al. Metabolic Imaging Detects Resistance to PI3Kα Inhibition Mediated by Persistent FOXM1 Expression in ER+ Breast Cancer. Cancer Cell 2020;38:516-33.e9. [Crossref] [PubMed]
  44. Cai Y, Xu G, Wu F, et al. Genomic Alterations in PIK3CA-Mutated Breast Cancer Result in mTORC1 Activation and Limit the Sensitivity to PI3Kα Inhibitors. Cancer Res 2021;81:2470-80. [Crossref] [PubMed]
  45. Du X, Shao Y, Qin HF, et al. ALK-rearrangement in non-small-cell lung cancer (NSCLC). Thorac Cancer 2018;9:423-30. [Crossref] [PubMed]
  46. Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med 2012;18:378-81. [Crossref] [PubMed]
  47. Xiang Y, Zhang S, Fang X, et al. Therapeutic Advances of Rare ALK Fusions in Non-Small Cell Lung Cancer. Curr Oncol 2022;29:7816-31. [Crossref] [PubMed]
  48. Mao W, Chen R, Zhang J, et al. TMED2-ALK, a Novel ALK Fusion Gene Identified in a Patient With Lung Adenocarcinoma. J Thorac Oncol 2020;15:e37-9. [Crossref] [PubMed]
  49. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561-6. [Crossref] [PubMed]
  50. Rijavec E, Biello F, Indini A, et al. Current Insights on the Treatment of Anaplastic Lymphoma Kinase-Positive Metastatic Non-Small Cell Lung Cancer: Focus on Brigatinib. Clin Pharmacol 2022;14:1-9. [Crossref] [PubMed]
  51. Pan Y, Deng C, Qiu Z, et al. The Resistance Mechanisms and Treatment Strategies for ALK-Rearranged Non-Small Cell Lung Cancer. Front Oncol 2021;11:713530. [Crossref] [PubMed]
  52. Rotow J, Bivona TG. Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer 2017;17:637-58. [Crossref] [PubMed]
  53. Berlak M, Tucker E, Dorel M, et al. Mutations in ALK signaling pathways conferring resistance to ALK inhibitor treatment lead to collateral vulnerabilities in neuroblastoma cells. Mol Cancer 2022;21:126. [Crossref] [PubMed]
  54. Coleman N, Hong L, Zhang J, et al. 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. [Crossref] [PubMed]
  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. [Crossref] [PubMed]
  56. Dong Y, Xu J, Sun B, et al. MET-Targeted Therapies and Clinical Outcomes: A Systematic Literature Review. Mol Diagn Ther 2022;26:203-27. [Crossref] [PubMed]
  57. Dagogo-Jack I, Yoda S, Lennerz JK, et al. MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer. Clin Cancer Res 2020;26:2535-45. [Crossref] [PubMed]
  58. Zhang Y, Nguyen TTT, Shang E, et al. MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma. Cancer Res 2020;80:30-43. [Crossref] [PubMed]
  59. Togashi Y, Mizuuchi H, Tomida S, et al. MET gene exon 14 deletion created using the CRISPR/Cas9 system enhances cellular growth and sensitivity to a MET inhibitor. Lung Cancer 2015;90:590-7. [Crossref] [PubMed]
  60. Kholodenko BN, Rauch N, Kolch W, et al. A systematic analysis of signaling reactivation and drug resistance. Cell Rep 2021;35:109157. [Crossref] [PubMed]
  61. Fumarola C, Bonelli MA, Petronini PG, et al. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem Pharmacol 2014;90:197-207. [Crossref] [PubMed]
  62. Liu L, Zhu H, Liao Y, et al. Inhibition of Wnt/β-catenin pathway reverses multi-drug resistance and EMT in Oct4+/Nanog+ NSCLC cells. Biomed Pharmacother 2020;127:110225. [Crossref] [PubMed]
  63. Takahashi H, Sakakibara-Konishi J, Furuta M, et al. Notch pathway regulates osimertinib drug-tolerant persistence in EGFR-mutated non-small-cell lung cancer. Cancer Sci 2023;114:1635-50. [Crossref] [PubMed]
  64. Giroux-Leprieur E, Costantini A, Ding VW, et al. Hedgehog Signaling in Lung Cancer: From Oncogenesis to Cancer Treatment Resistance. Int J Mol Sci 2018;19:2835. [Crossref] [PubMed]
  65. Xiao L, Lan X, Shi X, et al. Cytoplasmic RAP1 mediates cisplatin resistance of non-small cell lung cancer. Cell Death Dis 2017;8:e2803. [Crossref] [PubMed]
  66. Gu J, Huang W, Wang X, et al. Hsa-miR-3178/RhoB/PI3K/Akt, a novel signaling pathway regulates ABC transporters to reverse gemcitabine resistance in pancreatic cancer. Mol Cancer 2022;21:112. [Crossref] [PubMed]
  67. Li LY, Guan YD, Chen XS, et al. DNA Repair Pathways in Cancer Therapy and Resistance. Front Pharmacol 2020;11:629266. [Crossref] [PubMed]
  68. Abrams SL, Steelman LS, Shelton JG, et al. The Raf/MEK/ERK pathway can govern drug resistance, apoptosis and sensitivity to targeted therapy. Cell Cycle 2010;9:1781-91. [Crossref] [PubMed]
  69. Akbari Kordkheyli V, Rashidi M, Shokri Y, et al. CRISPER/CAS System, a Novel Tool of Targeted Therapy of Drug-Resistant Lung Cancer. Adv Pharm Bull 2022;12:262-73. [Crossref] [PubMed]
  70. Huang L, Liao Z, Liu Z, et al. Application and Prospect of CRISPR/Cas9 Technology in Reversing Drug Resistance of Non-Small Cell Lung Cancer. Front Pharmacol 2022;13:900825. [Crossref] [PubMed]
  71. Anichini A, Perotti VE, Sgambelluri F, et al. Immune Escape Mechanisms in Non Small Cell Lung Cancer. Cancers (Basel) 2020;12:3605. [Crossref] [PubMed]
  72. Liu Z, Shi M, Ren Y, et al. Recent advances and applications of CRISPR-Cas9 in cancer immunotherapy. Mol Cancer 2023;22:35. [Crossref] [PubMed]
  73. Xu Y, Chen C, Guo Y, et al. Effect of CRISPR/Cas9-Edited PD-1/PD-L1 on Tumor Immunity and Immunotherapy. Front Immunol 2022;13:848327.
  74. Wang Y, Mohseni M, Grauel A, et al. SHP2 blockade enhances anti-tumor immunity via tumor cell intrinsic and extrinsic mechanisms. Sci Rep 2021;11:1399. [Crossref] [PubMed]
  75. Imaizumi T, Kumagai M, Sasaki N, et al. Interferon-gamma stimulates the expression of galectin-9 in cultured human endothelial cells. J Leukoc Biol 2002;72:486-91.
  76. Zhang X, Jin X, Sun R, et al. Gene knockout in cellular immunotherapy: Application and limitations. Cancer Lett 2022;540:215736. [Crossref] [PubMed]
  77. Hu X, Li J, Fu M, et al. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 2021;6:402. [Crossref] [PubMed]
  78. Gotthardt D, Trifinopoulos J, Sexl V, et al. JAK/STAT Cytokine Signaling at the Crossroad of NK Cell Development and Maturation. Front Immunol 2019;10:2590. [Crossref] [PubMed]
  79. Chen BR, Deshpande A, Barbosa K, et al. A JAK/STAT-mediated inflammatory signaling cascade drives oncogenesis in AF10-rearranged AML. Blood 2021;137:3403-15. [Crossref] [PubMed]
  80. Rah B, Rather RA, Bhat GR, et al. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022;13:821344. [Crossref] [PubMed]
  81. Wang L, Yukselten Y, Nuwagaba J, et al. JAK/STAT signaling pathway affects CCR5 expression in human CD4(+) T cells. Sci Adv 2024;10:eadl0368. [Crossref] [PubMed]
  82. Tang N, Cheng C, Zhang X, et al. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020;5:e133977. [Crossref] [PubMed]
  83. Azangou-Khyavy M, Ghasemi M, Khanali J, et al. CRISPR/Cas: From Tumor Gene Editing to T Cell-Based Immunotherapy of Cancer. Front Immunol 2020;11:2062. [Crossref] [PubMed]
  84. Chan YT, Lu Y, Wu J, et al. CRISPR-Cas9 library screening approach for anti-cancer drug discovery: overview and perspectives. Theranostics 2022;12:3329-44. [Crossref] [PubMed]
  85. Wei L, Lee D, Law CT, et al. Genome-wide CRISPR/Cas9 library screening identified PHGDH as a critical driver for Sorafenib resistance in HCC. Nat Commun 2019;10:4681. [Crossref] [PubMed]
  86. Lin S, Larrue C, Scheidegger NK, et al. An In Vivo CRISPR Screening Platform for Prioritizing Therapeutic Targets in AML. Cancer Discov 2022;12:432-49. [Crossref] [PubMed]
  87. Olagnier D, Brandtoft AM, Gunderstofte C, et al. Nrf2 negatively regulates STING indicating a link between antiviral sensing and metabolic reprogramming. Nat Commun 2018;9:3506. [Crossref] [PubMed]
  88. Tao S, Liu P, Luo G, et al. p97 Negatively Regulates NRF2 by Extracting Ubiquitylated NRF2 from the KEAP1-CUL3 E3 Complex. Mol Cell Biol 2017;37:e00660-16. [Crossref] [PubMed]
  89. Xie Y, Feng SL, He F, et al. Down-regulating Nrf2 by tangeretin reverses multiple drug resistance to both chemotherapy and EGFR tyrosine kinase inhibitors in lung cancer. Pharmacol Res 2022;186:106514. [Crossref] [PubMed]
  90. Sánchez-Ortega M, Carrera AC, Garrido A. Role of NRF2 in Lung Cancer. Cells 2021;10:1879. [Crossref] [PubMed]
  91. Wang XJ, Sun Z, Villeneuve NF, et al. Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis 2008;29:1235-43. [Crossref] [PubMed]
  92. Qian Z, Zhou T, Gurguis CI, et al. Nuclear factor, erythroid 2-like 2-associated molecular signature predicts lung cancer survival. Sci Rep 2015;5:16889. [Crossref] [PubMed]
  93. Tao S, Rojo de la Vega M, Chapman E, et al. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol Carcinog 2018;57:182-92. [Crossref] [PubMed]
  94. Bialk P, Wang Y, Banas K, et al. Functional Gene Knockout of NRF2 Increases Chemosensitivity of Human Lung Cancer A549 Cells In Vitro and in a Xenograft Mouse Model. Mol Ther Oncolytics 2018;11:75-89. [Crossref] [PubMed]
  95. Binkley MS, Jeon YJ, Nesselbush M, et al. KEAP1/NFE2L2 Mutations Predict Lung Cancer Radiation Resistance That Can Be Targeted by Glutaminase Inhibition. Cancer Discov 2020;10:1826-41. [Crossref] [PubMed]
  96. Lok BH, Gardner EE, Schneeberger VE, et al. PARP Inhibitor Activity Correlates with SLFN11 Expression and Demonstrates Synergy with Temozolomide in Small Cell Lung Cancer. Clin Cancer Res 2017;23:523-35. [Crossref] [PubMed]
  97. Mezzadra R, de Bruijn M, Jae LT, et al. SLFN11 can sensitize tumor cells towards IFN-γ-mediated T cell killing. PLoS One 2019;14:e0212053. [Crossref] [PubMed]
  98. Winkler C, King M, Berthe J, et al. SLFN11 captures cancer-immunity interactions associated with platinum sensitivity in high-grade serous ovarian cancer. JCI Insight 2021;6:e146098. [Crossref] [PubMed]
  99. Yu J, Zhou J, Xu F, et al. High expression of Aurora-B is correlated with poor prognosis and drug resistance in non-small cell lung cancer. Int J Biol Markers 2018;33:215-21. [Crossref] [PubMed]
  100. Al-Khafaji AS, Davies MP, Risk JM, et al. Aurora B expression modulates paclitaxel response in non-small cell lung cancer. Br J Cancer 2017;116:592-9. [Crossref] [PubMed]
  101. Liao S, Davoli T, Leng Y, et al. A genetic interaction analysis identifies cancer drivers that modify EGFR dependency. Genes Dev 2017;31:184-96. [Crossref] [PubMed]
  102. Karthika C, Sureshkumar R, Zehravi M, et al. Multidrug Resistance of Cancer Cells and the Vital Role of P-Glycoprotein. Life (Basel) 2022;12:897. [Crossref] [PubMed]
  103. Liu R, Chen Y, Liu G, et al. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis 2020;11:797. [Crossref] [PubMed]
  104. Wang Y, Chen H. Protein glycosylation alterations in hepatocellular carcinoma: function and clinical implications. Oncogene 2023;42:1970-9. [Crossref] [PubMed]
  105. Tuffour I, Amuzu S, Bayoumi H, et al. Early in vitro evidence indicates that deacetylated sialic acids modulate multi-drug resistance in colon and lung cancers via breast cancer resistance protein. Front Oncol 2023;13:1145333. [Crossref] [PubMed]
  106. Al-Shayeb B, Skopintsev P, Soczek KM, et al. Diverse virus-encoded CRISPR-Cas systems include streamlined genome editors. Cell 2022;185:4574-4586.e16. [Crossref] [PubMed]
  107. Chen W, Ma J, Wu Z, et al. Cas12n nucleases, early evolutionary intermediates of type V CRISPR, comprise a distinct family of miniature genome editors. Mol Cell 2023;83:2768-2780.e6. [Crossref] [PubMed]
  108. Mayuranathan T, Newby GA, Feng R, et al. Potent and uniform fetal hemoglobin induction via base editing. Nat Genet 2023;55:1210-20. [Crossref] [PubMed]
  109. Saito M, Xu P, Faure G, et al. Fanzor is a eukaryotic programmable RNA-guided endonuclease. Nature 2023;620:660-8. [Crossref] [PubMed]
  110. Werder RB, Liu T, Abo KM, et al. CRISPR interference interrogation of COPD GWAS genes reveals the functional significance of desmoplakin in iPSC-derived alveolar epithelial cells. Sci Adv 2022;8:eabo6566. [Crossref] [PubMed]
  111. Nahmad AD, Reuveni E, Goldschmidt E, et al. Frequent aneuploidy in primary human T cells after CRISPR-Cas9 cleavage. Nat Biotechnol 2022;40:1807-13. [Crossref] [PubMed]
  112. van Overbeek M, Capurso D, Carter MM, et al. DNA Repair Profiling Reveals Nonrandom Outcomes at Cas9-Mediated Breaks. Mol Cell 2016;63:633-46. [Crossref] [PubMed]
  113. Stadtmauer EA, Fraietta JA, Davis MM, et al. CRISPR-engineered T cells in patients with refractory cancer. Science 2020;367:eaba7365. [Crossref] [PubMed]
  114. Uddin F, Rudin CM, Sen T. CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future. Front Oncol 2020;10:1387. [Crossref] [PubMed]
  115. Gostimskaya I. CRISPR-Cas9: A History of Its Discovery and Ethical Considerations of Its Use in Genome Editing. Biochemistry (Mosc) 2022;87:777-88. [Crossref] [PubMed]
  116. Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. N Engl J Med 2021;384:252-60. [Crossref] [PubMed]

(English Language Editor: J. Gray)

Cite this article as: Liu B, Wang Z, Gu M, Wang J, Tan J. Research into overcoming drug resistance in lung cancer treatment using CRISPR-Cas9 technology: a narrative review. Transl Lung Cancer Res 2024;13(8):2067-2081. doi: 10.21037/tlcr-24-592

Download Citation