Advances of cell therapy in lung cancer: a narrative review
Review Article

Advances of cell therapy in lung cancer: a narrative review

Lu Lv^, Wenyan Chen^, Na Chen^, Enhai Cui^

Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China

Contributions: (I) Conception and design: L Lv, E Cui; (II) Administrative support: E Cui; (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.

^ORCID: Lu Lv, 0000-0001-5966-3464; Wenyan Chen, 0000-0002-4381-0711; Na Chen, 0009-0003-7214-2964; Enhai Cui, 0000-0002-8808-8793.

Correspondence to: Enhai Cui, BM. Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 1558 Third Ring North Road, Huzhou 313000, China. Email: kjkceh@126.com.

Background and Objective: Lung cancer is the second most prevalent malignancy and has the highest death rate. The main approaches for lung cancer treatment include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, the treatments of the disease need to be further improved. An increasing number of scientific investigations indicated cell therapy to be a successful new treatment for lung cancer. Cell therapy can improve the host’s immunity to disease and can compensate for the shortcomings in the therapeutic effects of traditional treatments, particularly in the case of cancer treatment. However, due to its recent development, its clinical efficacy still needs to be further examined. In order to provide an updated source on cell therapy for lung cancer, this paper summarizes the clinical use of chimeric antigen receptor T cells (CAR-Ts), stem cells, cytokine-induced killer cells (CIKs), and tumor-infiltrating lymphocytes (TILs) and discusses recent clinical advancements.

Methods: We performed a search of the PubMed database on March 28, 2023, and again on June 10, 2023. A review of retrieved literature related to cell therapy and treatments for lung cancer was completed.

Key Content and Findings: Cell therapy has been applied in clinical studies on the treatment of disorders of the hematologic system, digestive system, respiratory system, and other systems. CAR-T therapy has been successfully used in the treatment of B-cell malignancies, which suggests that cell therapy has broad prospects in the treatment of malignant tumors. CAR-T, stem cells, CIKs, and TILs exert antitumor activity and can recognize and could be used to treat lung cancer.

Conclusions: Cell therapy represents a novel solution in the treatment of lung cancer. Cell therapy, when combined with traditional therapies, can compensate for the shortcomings of these methods. Further research is needed to reduce the occurrence of adverse reactions and provide a more effective approach in treating lung cancer.

Keywords: Lung cancer; chimeric antigen receptor T cells (CAR-Ts); stem cells; cytokine-induced killer cells (CIKs); tumor-infiltrating lymphocytes (TILs)


Submitted Jun 27, 2023. Accepted for publication Oct 20, 2023. Published online Dec 15, 2023.

doi: 10.21037/jtd-23-1015


Introduction

According to the latest statistics from the American Cancer Society, there were 19.3 million new cases of cancer and nearly 10 million cancer-related deaths globally in 2020. Lung cancer, which accounts for around 11.4% of cancer diagnoses and 18.0% of fatalities, is the second most prevalent malignancy and has the highest death rate (1). In China, the age-standardized mortality rate of lung cancer increased from 1973 to 2015 (2), and the lung cancer incidence and mortality rate in China were 59.89/100,000 and 47.51/100,000 in 2016, respectively, according to data from the National Cancer Center (3), with patients having a poor prognosis.

At present, the main approaches for lung cancer treatment include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Surgery is the first choice for early lung cancer. The 5-year survival rate of patients with stage IA–IIB lung cancer after surgery can reach 53–92%. However, the risk of surgery is high, and surgery has certain requirements regarding the tumor-node-metastasis (TNM) classification of patients (4). Chemotherapy can effectively improve the cancer cure rate and slow the growth and metastasis of tumor. However, it has strong toxicity and numerous adverse effects and cannot completely eliminate cancer cells. Many patients of advanced age relapse due to drug resistance, and some others are not sensitive to chemotherapy (5). Radiotherapy is frequently used as an auxiliary treatment method to operation, but it can lead to serious adverse reactions such as decreased immunity and radiation pneumonitis (6). Targeted therapy is used in the treatment of gene mutation-related lung cancer, which can specifically kill tumor cells. However, it has certain drawbacks, such as drug resistance and a narrow range of applications. Immunotherapies, especially immune checkpoint inhibitors (ICIs) and adoptive cell therapy (ACT), have transformed the treatment landscape for a variety of solid malignancies. Yet, immunotherapy’s unique toxicity needs to be examined further (7). From the year 2000 to 2014, the age-standardized 5-year net survival of lung cancer ranged from 10% to 20% in most countries, and thus treatments for this disease need to be further improved (8).

In 2018, the results of the first-in-human neural stem cell transplantation for the treatment of chronic spinal cord injury were published. After treatment, three of the four participants showed significant improvement in disease symptoms, with no complications (9). Cell therapy has been investigated in clinical studies as a treatment for disorders of the hematologic system, digestive system, respiratory system, and other systems. The majority of cell therapies are still in the early stages of development, but hematopoietic stem cell (HSC) transplantation is a proven method for treating hematological illnesses. Chimeric antigen receptor T cells (CAR-Ts) have been successfully used in the treatment of B-cell malignancies, resulting in a complete remission (CR) rate of up to 90% in patients with acute lymphoblastic leukemia, which suggests that cell therapy has good potential in the treatment of malignant tumors (10). As a novel therapeutic approach, cell therapy has been gradually applied to the treatment of lung cancer over recent years, and a degree of research progress has been made. In this paper, the theoretical basis, compelling evidence, research status, development potential, and current challenges of cell therapy for lung cancer are all thoroughly discussed. We present this article in accordance with the Narrative Review reporting checklist (available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-1015/rc).


Methods

We searched PubMed for references with the terms “lung cancer”, “cell therapy”, “CAR-T”, “stem cells”, “cytokine-induced killer (CIK)”, and “tumor-infiltrating lymphocyte (TIL)” or their combination in the title or abstract. We also identified relevant articles from the reference lists of the selected articles. After inclusion and exclusion criteria were applied, with all authors implementing the same selection process, 81 papers were ultimately included. The search strategy is summarized in Table 1.

Table 1

Summary of the literature search strategy

Item Specification
Date of search March 28, 2023 and June 10, 2023
Databases and other sources searched PubMed
Search terms used Search terms: “lung cancer” OR “cell therapy” OR “lung cancer” AND “CAR-T” OR “lung cancer” AND “stem cells” OR “lung cancer” AND “CIK” OR “lung cancer” AND “TIL”
Time frame 1995–2023
Inclusion and exclusion criteria Inclusion: all articles related to lung cancer and studies involving cell therapy
Exclusion: articles not written in English
Selection process L.L. conducted the literature search and analysis. All authors reviewed the final list of studies included in the review
Any additional considerations Some papers were not obtained from PubMed but were rather referenced in papers from the original PubMed search

CAR-T, chimeric antigen receptor T cell; CIK, cytokine-induced killer cell; TIL, tumor-infiltrating lymphocyte.


Cell therapy and lung cancer

Cell therapy is a type of ACT, which is a therapeutic method that involves isolating immune cells with antitumor activity from patients, expanding them in vitro, and then transplanting them back into patients to improve host immunity against diseases (Figure 1). A meta-analysis by Zhao et al. (11) showed that ACT could significantly improve the overall survival (OS) and progression-free survival (PFS) of patients with non-small cell lung cancer (NSCLC). Most cell therapies are still in the early experimental stage of development. Recent studies have demonstrated that these immunoregulatory cells are highly differentiated. If these cells are used in the clinical research of cell therapy, the deficiency in the treatment effect of traditional methods, especially in cancer therapy, can be remedied to a large extent (12). These cells, including T cells, stem cells, killer cells, dendritic cells (DCs), and TILs, among others, all exert immune function and can recognize and eliminate tumor-associated antigens (TAAs) (13).

Figure 1 The principle of adoptive cell therapy involves isolating immune cells with antitumor activity from patients, expanding them in vitro, and reinfusing the cells into patients.

CAR-Ts

CAR-Ts refers to the chimeric antigen receptor (CAR) expressed in T cells through genetic engineering technology, which can specifically recognize the antigen molecules of tumor cells so as to obtain T cells capable of specifically killing the antigen of tumor cells (Figure 2). The key structural element of CAR-Ts is the CAR, which is composed of a TAA binding domain (a single-chain fragment variable usually derived from the antigen-binding region of a monoclonal antibody), an extracellular hinge domain, a transmembrane domain, and an intracellular signaling domain (14). CAR-Ts specifically recognize tumor cell-surface antigens via major histocompatibility complex (MHC)-independent pathways to kill tumor cells, thereby avoiding the immune escape caused by the loss or downregulation of MHC. Moreover, CAR-Ts can increase costimulatory molecular signals, enhance the tumor destruction of T cells, and achieve an antitumor effect.

Figure 2 The tumor-associated antigen binding domain (single-chain fragment variable) on the surface of chimeric antigen receptor T cells specifically recognizes the surface antigen of tumor cells. CAR-T, chimeric antigen receptor T cell; scFv, single-chain fragment variable.

CAR-Ts were first applied in hematological diseases, exhibiting significant effectiveness against several hematological cancers (15). The safety and efficacy of CAR-T therapy in solid tumors are currently being examined in depth in a number of studies. Among these, the majority of feasibility clinical trials are related to CAR-T therapy for lung cancer. The optimal targets for CAR-T therapy are those that are specifically expressed or universally overexpressed in tumor cells and that are expressed at very low or limited levels in normal peripheral cells or tissues. Mesothelin (MSLN), mucin 1 (MUC1), glypican 3 (GPC3), prostate stem cell antigen (PSCA), epidermal growth factor receptor (EGFR), carcinoembryonic antigen (CEA), human epidermal growth factor receptor 2 (HER2), programmed death-ligand 1 (PD-L1), receptor tyrosine kinase-like orphan receptor 1 (ROR1), and other promising targets have been the focus of clinical trials of CAR-T for the treatment of NSCLC and small cell lung cancer (SCLC) (16). Zhang et al. (17) used EGFR subtype III-specific third-generation CAR-Ts in a mouse model of lung cancer metastasis. Their findings indicated that there was a considerable decrease in mouse mortality, decreased metastasis of lung cancer, and no overt signs of adverse reactions. Feng et al. (18) used CAR-Ts in the treatment of patients with advanced relapsed or refractory NSCLC expressing EGFR. Among 11 patients, two patients achieved partial response (PR), and five patients were stable for 2 to 8 months. In a phase I clinical trial conducted by Zhang et al. (19), EGFR-specific CAR-Ts generated by the piggyBac transposon system were used in patients with advanced relapsed or refractory NSCLC. The results showed that after 2 months of CAR-T cell infusion in nine patients, EGFR-CAR T cells were detected in the peripheral blood of eight patients, with one patient experiencing PR that lasted for more than 13 months, six patients with stable disease (SD), and two patients with progressive disease, which confirmed the safety and feasibility of CAR-T therapy for lung cancer. Common adverse effects after CAR-T infusion were mild skin toxicity, nausea, vomiting, dyspnea, and hypotension. MSLN is a tumor differentiation antigen that is underexpressed in normal mesothelial cells and overexpressed in a variety of solid cancers. including lung cancer, mesothelioma, and pancreatic cancer. In one study, MSLN-targeted CAR-T in vitro showed significant inhibitory effects on the proliferation and invasion of cancer cells (20).

T-cell receptor-engineered T cell (TCR-T) therapy is a more targeted cellular immunotherapy for cancer treatment. The modified T cells with new genes can express the T-cell receptor (TCR). The TCR-Ts can specifically recognize TAA and kill tumor cells. A recent study reported that TCR-T therapy could achieve anticancer activity in the treatment of NSCLC (21). PD-L1 is a significant immune checkpoint, is upregulated in multiple tumors, and can lead to the immune escape of tumor cells by binding to programmed cell death protein 1 (PD-1) on T cells. Agents targeting PD-1/PD-L1 signaling are flagship ICIs and constitute a breakthrough in the treatment of multiple types of advanced solid tumors. Studies have shown that PD-1 blockade could enhance the efficacy of CAR-T therapy in solid tumors (22,23) and represents an effective treatment strategy for NSCLC (24,25).

However, CAR-Ts have sufficient antitumor activity in solid tumors (26) because solid tumors rarely express specific tumor-associated antigens, resulting in off-target effects (27). CAR-Ts have limited persistence in vivo and the function is impaired (28). Prevention of CAR-T exhaustion may improve clinical outcomes for patients (29,30). A lack of highly immunosuppressive and metabolically challenging tumor microenvironments is an urgent problem that needs to be solved (31). In addition, CAR-T therapy induces a cascade of toxicity, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS). Multiple organ damage and functional failure, along with other issues such as antitumor immune evasion response, physical barrier, and CAR-T depletion, can result from antigen-targeting and nontumor toxicity (16,32).


Stem cells

Stem cells are clonal cell populations derived from single cells that have the ability to indefinitely self-renew and differentiate into various cell types (33). Research into stem cells has revealed that the therapy of acute respiratory distress syndrome, myocardial infarction, and liver injury may all benefit from the use of stem cells (34). With their ability to invade cancer cells, release bioactive substances, and suppress the immune system, stem cells have substantial potential in treating cancer (35).

Mesenchymal stem cells (MSCs) are adult stem cells with the ability to differentiate into a variety of cell types and can be isolated from bone marrow, umbilical cord blood, peripheral blood, fallopian tube tissues, fetal liver tissue, and lung tissue. MSCs have the ability to specifically localize tumors and their metastases and have been employed to treat degenerative illnesses in preclinical and clinical trials (36). Among the types of MSCs, bone marrow MSCs can be used to deliver various anticancer drugs directly into tumors (Figure 3) (37). According to Loebinger et al.’s study (38), MSCs expressing tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) migrate to tumors, which can cause apoptosis and the death of lung cancer cells, hinder the formation of side population (SP) cells, reduce the growth and metastasis of primary tumor, and exert a synergistic effect with chemotherapy to induce apoptosis, which plays a role in preventing cancer recurrence. Sage et al. (39) are conducting phase I–II clinical trials of TRAIL-transduced MSCs in lung cancer treatment and remain in the experimental stage. TRAIL activates exogenous apoptotic pathways by binding to cell-surface death receptors and selectively induces apoptosis in cancer cells without affecting healthy cells. Additionally, MSCs’ low immunogenicity, lack of MHC II, and expression of their costimulatory molecules (CD80, CD86, and CD40) may prevent allogeneic cells from inducing an immune response in patients, negating the necessity for human leukocyte antigen (HLA) matching and facilitating brief treatment with good safety (40). Chen et al. (41) prolonged the lifespan in a Lewis lung carcinoma model by using MSCs to express pigment epithelium-derived factor (PEDF). PEDF directly inhibited tumor effects via anti-angiogenesis, tumor differentiation, and apoptosis, indicating that MSCs can be used as an effective vector for tumor gene therapy. Yan et al. (42) applied human umbilical cord MSCs (HUC-MSCs) to express secretable soluble TRAIL (sTRAIL). These cells migrated via the monocyte chemoattractant protein-1/CC chemokine receptor 2 (MCP-1/CCR2) axis to mouse lung cancer tissues that had been injected with A549 human lung cancer cells and exhibited antitumor effects in vivo without significant side effects. As the precursors of most stromal cells, MSCs can affect the disease progression of various tumors, including lung cancer (40,43), for which they may exert an inhibitory effect. Moreover, studies have shown that MSCs can effectively inhibit the proliferation and induce apoptosis of A549 cells (44,45).

Figure 3 Mesenchymal stem cells carry anticancer drugs target to tumor cells and then release the drugs. MSC, mesenchymal stem cell.

MSCs are a critical factor in the growth, metastasis, and drug resistance of lung cancer cells and may be used as an effective method for the treatment of lung cancer. However, some studies have shown that MSCs can inhibit the apoptosis of lung cancer cells, promote tumor growth, and facilitate the occurrence and development of lung cancer (46). Liu et al. found that MSCs can spontaneously hybridize with NSCLC cells, which can enable these tumor cells to acquire abilities of epithelial-mesenchymal transition (EMT) and stem cell-like self-renewal. Indeed, the interaction between MSCs and lung cancer remains ambiguous, and further study in this direction is needed (43). In addition, the proliferation and differentiation potential of MSCs are greatly affected by environmental factors, and it has been shown the proliferation and differentiation abilities of MSCs gradually decrease after they are passaged in vitro (36).


CIKs

CIKs, a newly discovered antitumor effector cell, are a subtype of T lymphocytes. CIKs have the phenotype of natural killer (NK) T cells and express both CD3 and CD56 markers. In many studies, the CD3+CD56+ subsets show the highest levels of antitumor activity against a variety of tumor target cells with non-MHC-restricted cytotoxicity. Activated T cells induced by cytokines such as interferon-γ (IFN-γ) and interleukin-2 (IL-2) in vitro can directly kill tumor cells and generally enhance the immune function of patients with cancer (Figure 4) (47). Schmeel et al. (47) examined 2,729 patients for clinical trials of 22 kinds of solid tumors. The results showed that the average effective rate of CIKs against solid tumors was 39%, the OS was significantly increased, the quality of life was significantly improved, and the side effects were slight. CIKs may have considerable potential in improving tumor prognosis. Wang et al. (48) reported that compared with conventional treatment—including chemotherapy—in patients with lung cancer, adoptive immunotherapy with adjuvant CIK cells yielded a significantly higher objective response rate (ORR) and OS. The percentage of CD3+, CD4+, CD4+CD8+, CD3+CD56+, and NK cells increased after CIK combined treatment, and CEA was more likely to decrease to normal levels. In Han et al.’s (49) study, patients with advanced NSCLC treated with CIK cells combined with PD-1 inhibitor had an ORR of 42.9%. In these patients, the T-cell level was significantly increased but not the incidence of adverse events. CIKs can be effectively expanded by peripheral blood mononuclear cell culture in vitro and are easy to obtain. CIK therapy is associated with a low incidence of graft versus host disease (GVHD), can strengthen the immune function of patients with lung cancer to help prevent cancer recurrence and metastasis, and can prolong the OS and PFS of these patients (50,51).

Figure 4 Activated T cells induced by IFN-γ and IL-2 in vitro can express both CD3 and CD56 markers, which can bind tumor cells with non-MHC-restricted cytotoxicity. IL-2, interleukin-2; IFN-γ, interferon-γ; MHC, major histocompatibility complex.

Some studies have shown that CIKs in combination with PD-1 blockade can enhance the efficacy of CIK therapy for patients with NSCLC, providing an experimental basis for clinical practice (52-54). Zhao et al. investigated the efficacy of PD-1 blockade combined with CIKs in patients with advanced NSCLC who failed at least two treatment regimens. They reported a disease control rate (DCR) of 85.7% and found this treatment to be safe and effective. Among the seven patients, two patients achieved PR and four patients experienced SD (55). Hui et al. reported clinical evidence that PD-1 blockade plus autologous CIK therapy was effective in treating patients with advanced squamous NSCLC and severe thrombocytopenia (56).

Notably, compared with the administration of DCs alone, the cocultivation of CIKs and DCs can significantly enhance the antitumor immunity and cytotoxic activity. Xiao et al. (57) examined 28 randomized controlled trials of DC-CIK therapy combined with chemotherapy in the treatment of NSCLC. The results showed that DC-CIK treatment could significantly increase the proportion of peripheral blood T-lymphocyte subsets. The antitumor effect was enhanced, and the ORR and DCR of the patients were effectively improved. DCs are by far the most powerful antigen-presenting cells, which are crucial to the host’s immune response. CIKs exert stronger tumor-killing activity than do lymphokine-activated killer cells (LAKs). Therefore, DC-CIK therapy, which involves coculturing DCs and CIKs, has enormous potential for the targeted treatment of malignant tumors (58). One study demonstrated that PD-1 blockade–activated combined DC-CIK treatment in patients with advanced solid tumors was safe and effective (59). Despite these promising findings, patients treated with CIKs should be aware of issues such as recurrence, ineffective treatment, and unclear toxic effects (60).


TILs

TILs are a cell population that can exert antitumor activity. They can be obtained by stimulating and activating infiltrating lymphocytes isolated from tumor tissues with IL-2 in vitro and further expansion after culture (Figure 5) (61). TILs were initially used to treat metastatic malignant melanoma and found to be capable of providing a good therapeutic effect (62). According to the relevant research, the remission rate of patients with malignant melanoma treated with TILs can reach 36% (63). Creelan et al. (64) conducted a phase I clinical trial in 20 patients with early progression of advanced NSCLC who were treated with nivolumab in PD1 inhibitor monotherapy. Of these patients, 68.75% experienced tumor shrinkage after TIL treatment, and CR was achieved in two patients. Adverse effects of TIL include hypoproteinemia, nausea, hyponatremia, and diarrhea, but they can be effectively controlled in most patients. Iovance Biotherapeutics, a biotechnology company, opened a phase II, multicenter study (IOV-COM-202; NCT03645928) to evaluate autologous TILs in patients with solid tumors. After one cycle of LN-145 therapy, the overall response rate of 28 patients with metastatic NSCLC was 21.4%, one patient experienced CR, five patients experienced PR, and two patients had a PD-L1-negative response. Disease control was achieved in 64.3% of patients. Treatment-related adverse effects were fever, nausea, and cytopenia (65), and no unexpected adverse effects have been identified or encountered thus far. Additionally, Iovance has initiated another phase II trial (IOV-LUN-202; NCT04614103) to examine LN-145 in the second-line treatment of patients with metastatic NSCLC who progressed on previous ICI therapy and chemotherapy.

Figure 5 The tumor tissues isolated from patients are activated with IL-2 in vitro, and after tumor-specific recognition and selection, the cells with anti-tumor activity are expanded and then reinfused into patients. IL-2, interleukin-2.

In recent years, the combination of TIL therapy with tumor-specific antigen (TSA) has provided a new direction in the development of TIL therapy. TSA, also known as neoantigen, is an abnormal protein produced by the somatic mutation of tumor cells (66). Achilles Therapeutics have begun the phase I–IIa CHIRON clinical trial to evaluate the safety, tolerability, and clinical activity of clonal neoantigen-reactive T cell (cNeT) therapy as a single dose in adult patients with advanced metastatic NSCLC. Among the eight enrolled patients, one experienced PR (56% tumor reduction maintained at week 36) and six experienced SD, with an overall durable clinical benefit at 12 weeks in 71% of evaluable patients (5/7) with advanced NSCLC. The safety and tolerability of cNeT therapy was observed to be better than those of standard TIL therapy, with the most common adverse events being lymphopenia and neutropenia (67).


Conclusions

Most of the research related to cell therapy for lung cancer thus far remains in the stages of preclinical or clinical trials. The United States National Library of Medicine clinical trials registered website lists almost 100 cell treatments for clinical trials in lung cancer (http://www.clinicaltrials.gov; accessed in April 2023). A substantial amount of clinical research data is required to support cell therapy for lung cancer before it can be widely applied in clinical practice. Despite the progress made in this area, there are critical challenges that need to be overcome, including the complexity of preparation technology, high cost, tolerability to immunotherapy, insufficient antitumor activity in solid tumors, the high heterogeneity of tumors, unclear side effects, and adverse reactions.

Nonetheless, cell therapy represents a novel option for patients with lung cancer and has favorable potential in the treatment of lung cancer. Moreover, cell therapy, while not a substitute for conventional means of treatment, can complement these therapies (details in Table 2). Cell therapy can be administered to postoperative patients with a small tumor burden to remove small residual lesions, prevent tumor recurrence and metastasis, and thus improve the cure rate (17,46,68). Many studies have shown that for patients undergoing chemotherapy, combination with cell therapy can enhance the immune function of patients, prolong OS and PFS, improve the quality of life of patients, reduce the incidence of adverse reactions, and inhibit tumor progression (38,69-76,79,80). Ratto et al. (77) reported that compared with chemoradiotherapy alone, TIL therapy combined with chemoradiotherapy could significantly improve the survival rate of patients with stage III NSCLC. Targeted therapy combined with cell therapy is considered to be a safe and effective treatment method, which can exert strong antitumor activity and improve the control rate of the disease (55,78,81). Thus far, cell therapy has shown good antitumor activity in lung cancer, especially in NSCLC. Due to the lack of understanding in SCLC regarding the molecular mechanism underlying rapid progression, tumor genetic heterogeneity, and treatment resistance, good efficacy has not been achieved for this disease type.

Table 2

Studies on conventional treatments combined with cell therapy for lung cancer

Conventional treatment Cell therapy Diseases Number of patients Results Year References
Surgery TIL Stage III NSCLC 24 The median survival was 14 months, and the 2-year survival was 40% 1995 Ratto et al. (68)
Chemotherapy DC-CIK Advanced NSCLC 60 The median survival time of was 13.8 months. More than two immunotherapies improved TTP and OS 2014 Zhong et al. (69)
DC-CIK Stage IB NSCLC 66 Patients in the DC-CIK therapy group had significantly longer OS and PFS 2015 Li et al. (70)
CIK Extensive stage NSCLC 44 The total response rate and PFS in the combined treatment group were longer than those of the control group 2017 Huang et al. (71)
DC-CIK Advanced NSCLC 135 The PFS and OS for DC-CIK plus chemotherapy were higher. The abundance CD3+ T cells increased after immunotherapy 2019 Zhao et al. (72)
CIK Different progression stages NSCLC and SCLC 68 The median OS and the 3-year OS of patients in the CIK group were longer than those in the control group 2018 Chen et al. (73)
DC-CIK Locally advanced NSCLC 142 The OS and PFS in the DC-CIK group were superior to those in the chemoradiotherapy group 2020 Tian et al. (74)
DC-CIK Intermediate to advanced NSCLC 60 The overall cancer control rate of treatment in the DC-CIK immunotherapy plus chemotherapy group was higher 2021 Wang et al. (75)
DC-CIK Advanced NSCLC 507 Chemotherapy reduced the incidence of DTH in patients receiving DC-CIK therapy 2016 Zhang et al. (76)
Chemoradiotherapy TIL NSCLC 131 The 3-year survival was better for patients who underwent TIL therapy, especially for patients with stage IIIb NSCLC 1996 Ratto et al. (77)
Targeted therapy R-CIK Advanced NSCLC 7 Partial remission was achieved in two patients and stable disease in four patients, one patient experiencing progressive disease. The median TTP was 4.8 months 2018 Zhao et al. (55)
CIK NSCLC 1 The patient achieved a near-complete response. No adverse events occurred during treatment 2018 Wang et al. (78)

TIL, tumor-infiltrating lymphocyte; NSCLC, non-small cell lung cancer; DC, dendritic cell; CIK, cytokine-induced killer cell; TTP, time to progression; OS, overall survival; PFS, progression-free survival; SCLC, small cell lung cancer; R-CIK, RetroNectin-activated CIK; DTH, delayed-type hypersensitivity.

Cell therapy has been applied in the treatment of lung cancer and has been used in combination with conventional methods to provide new solutions in clinical practice. Many relevant issues, including finding stably expressed sites, reducing off-target toxicity, mitigating cytokine storm, understanding the mechanism of cell therapy, and developing a clinical application strategy, still need to be researched further. In the future, studies into cell therapy can examine their combination with traditional methods in the treatment of lung cancer to improve the efficacy of drugs and reduce the occurrence of adverse reactions, thus providing a more effective treatment of lung cancer.


Acknowledgments

Funding: This work was supported by the Key Research and Development Project of the Science and Technology Department of Zhejiang Province (No. 2019C03041).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-1015/rc

Peer Review File: Available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-1015/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-1015/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. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Wei W, Zeng H, Zheng R, et al. Cancer registration in China and its role in cancer prevention and control. Lancet Oncol 2020;21:e342-9. [Crossref] [PubMed]
  3. Zheng R, Zhang S, Zeng H, et al. Cancer incidence and mortality in China, 2016. J Natl Cancer Cent 2022;2:1-9.
  4. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung Cancer Staging Project: Proposals for Revision of the TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM Classification for Lung Cancer. J Thorac Oncol 2016;11:39-51. [Crossref] [PubMed]
  5. Kim ES. Chemotherapy Resistance in Lung Cancer. Adv Exp Med Biol 2016;893:189-209. [Crossref] [PubMed]
  6. Hanania AN, Mainwaring W, Ghebre YT, et al. Radiation-Induced Lung Injury: Assessment and Management. Chest 2019;156:150-62. [Crossref] [PubMed]
  7. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin 2020;70:86-104. [Crossref] [PubMed]
  8. Allemani C, Matsuda T, Di Carlo V, et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 2018;391:1023-75. [Crossref] [PubMed]
  9. Curtis E, Martin JR, Gabel B, et al. A First-in-Human, Phase I Study of Neural Stem Cell Transplantation for Chronic Spinal Cord Injury. Cell Stem Cell 2018;22:941-950.e6. [Crossref] [PubMed]
  10. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371:1507-17. [Crossref] [PubMed]
  11. Zhao B, Zhang W, Yu D, et al. Adoptive immunotherapy shows encouraging benefit on non-small cell lung cancer: a systematic review and meta-analysis. Oncotarget 2017;8:113105-19. [Crossref] [PubMed]
  12. Mount NM, Ward SJ, Kefalas P, et al. Cell-based therapy technology classifications and translational challenges. Philos Trans R Soc Lond B Biol Sci 2015;370:20150017. [Crossref] [PubMed]
  13. Hung LVM, Ngo HT, Van Pham P. Clinical Trials with Cytokine-Induced Killer Cells and CAR-T Cell Transplantation for Non-small Cell Lung Cancer Treatment. Adv Exp Med Biol 2020;1292:113-30. [Crossref] [PubMed]
  14. Ma S, Li X, Wang X, et al. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int J Biol Sci 2019;15:2548-60. [Crossref] [PubMed]
  15. Park JH, Rivière I, Gonen M, et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N Engl J Med 2018;378:449-59. [Crossref] [PubMed]
  16. Xiao BF, Zhang JT, Zhu YG, et al. Chimeric Antigen Receptor T-Cell Therapy in Lung Cancer: Potential and Challenges. Front Immunol 2021;12:782775. [Crossref] [PubMed]
  17. Zhang Z, Jiang J, Wu X, et al. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy. Front Med 2019;13:57-68. [Crossref] [PubMed]
  18. Feng K, Guo Y, Dai H, et al. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci 2016;59:468-79. [Crossref] [PubMed]
  19. Zhang Y, Zhang Z, Ding Y, et al. Phase I clinical trial of EGFR-specific CAR-T cells generated by the piggyBac transposon system in advanced relapsed/refractory non-small cell lung cancer patients. J Cancer Res Clin Oncol 2021;147:3725-34. [Crossref] [PubMed]
  20. Ye L, Lou Y, Lu L, et al. Mesothelin-targeted second generation CAR-T cells inhibit growth of mesothelin-expressing tumors in vivo. Exp Ther Med 2019;17:739-47. [Crossref] [PubMed]
  21. Hong DS, Van Tine BA, Biswas S, et al. Autologous T cell therapy for MAGE-A4(+) solid cancers in HLA-A*02(+) patients: a phase 1 trial. Nat Med 2023;29:104-14. [Crossref] [PubMed]
  22. Davies JS, Karimipour F, Zhang L, et al. Non-synergy of PD-1 blockade with T-cell therapy in solid tumors. J Immunother Cancer 2022;10:e004906. [Crossref] [PubMed]
  23. John LB, Devaud C, Duong CP, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res 2013;19:5636-46. [Crossref] [PubMed]
  24. Liu M, Wang X, Li W, et al. Targeting PD-L1 in non-small cell lung cancer using CAR T cells. Oncogenesis 2020;9:72. [Crossref] [PubMed]
  25. Wang J, Wang Y, Pan H, et al. Chemokine Receptors CCR6 and PD1 Blocking scFv E27 Enhances Anti-EGFR CAR-T Therapeutic Efficacy in a Preclinical Model of Human Non-Small Cell Lung Carcinoma. Int J Mol Sci 2023;24:5424. [Crossref] [PubMed]
  26. Kosti P, Maher J, Arnold JN. Perspectives on Chimeric Antigen Receptor T-Cell Immunotherapy for Solid Tumors. Front Immunol 2018;9:1104. [Crossref] [PubMed]
  27. Cortés-Selva D, Dasgupta B, Singh S, et al. Innate and Innate-Like Cells: The Future of Chimeric Antigen Receptor (CAR) Cell Therapy. Trends Pharmacol Sci 2021;42:45-59. [Crossref] [PubMed]
  28. Majzner RG, Mackall CL. Clinical lessons learned from the first leg of the CAR T cell journey. Nat Med 2019;25:1341-55. [Crossref] [PubMed]
  29. Delgoffe GM, Xu C, Mackall CL, et al. The role of exhaustion in CAR T cell therapy. Cancer Cell 2021;39:885-8. [Crossref] [PubMed]
  30. Chen J, Qiu S, Li W, et al. Tuning charge density of chimeric antigen receptor optimizes tonic signaling and CAR-T cell fitness. Cell Res 2023;33:341-54. [Crossref] [PubMed]
  31. Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through the solid-tumour microenvironment. Nat Rev Drug Discov 2021;20:531-50. [Crossref] [PubMed]
  32. Wing A, Fajardo CA, Posey AD Jr, et al. Improving CART-Cell Therapy of Solid Tumors with Oncolytic Virus-Driven Production of a Bispecific T-cell Engager. Cancer Immunol Res 2018;6:605-16. [Crossref] [PubMed]
  33. Tran C, Damaser MS. Stem cells as drug delivery methods: application of stem cell secretome for regeneration. Adv Drug Deliv Rev 2015;82-83:1-11. [Crossref] [PubMed]
  34. Xu T, Zhang Y, Chang P, et al. Mesenchymal stem cell-based therapy for radiation-induced lung injury. Stem Cell Res Ther 2018;9:18. [Crossref] [PubMed]
  35. Zhang CL, Huang T, Wu BL, et al. Stem cells in cancer therapy: opportunities and challenges. Oncotarget 2017;8:75756-66. [Crossref] [PubMed]
  36. Brown C, McKee C, Bakshi S, et al. Mesenchymal stem cells: Cell therapy and regeneration potential. J Tissue Eng Regen Med 2019;13:1738-55. [Crossref] [PubMed]
  37. Kimbrel EA, Lanza R. Next-generation stem cells - ushering in a new era of cell-based therapies. Nat Rev Drug Discov 2020;19:463-79. [Crossref] [PubMed]
  38. Loebinger MR, Sage EK, Davies D, et al. TRAIL-expressing mesenchymal stem cells kill the putative cancer stem cell population. Br J Cancer 2010;103:1692-7. [Crossref] [PubMed]
  39. Sage EK, Thakrar RM, Janes SM. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy 2016;18:1435-45. [Crossref] [PubMed]
  40. Kolluri KK, Laurent GJ, Janes SM. Mesenchymal stem cells as vectors for lung cancer therapy. Respiration 2013;85:443-51. [Crossref] [PubMed]
  41. Chen Q, Cheng P, Yin T, et al. Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma. Int J Mol Med 2012;30:527-34. [Crossref] [PubMed]
  42. Yan C, Song X, Yu W, et al. Human umbilical cord mesenchymal stem cells delivering sTRAIL home to lung cancer mediated by MCP-1/CCR2 axis and exhibit antitumor effects. Tumour Biol 2016;37:8425-35. [Crossref] [PubMed]
  43. Liu R, Wei S, Chen J, et al. Mesenchymal stem cells in lung cancer tumor microenvironment: their biological properties, influence on tumor growth and therapeutic implications. Cancer Lett 2014;353:145-52. [Crossref] [PubMed]
  44. Tian LL, Yue W, Zhu F, et al. Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo. J Cell Physiol 2011;226:1860-7. [Crossref] [PubMed]
  45. Li L, Tian H, Chen Z, et al. Inhibition of lung cancer cell proliferation mediated by human mesenchymal stem cells. Acta Biochim Biophys Sin (Shanghai) 2011;43:143-8. [Crossref] [PubMed]
  46. Cortes-Dericks L, Galetta D. The therapeutic potential of mesenchymal stem cells in lung cancer: benefits, risks and challenges. Cell Oncol (Dordr) 2019;42:727-38. [Crossref] [PubMed]
  47. Schmeel LC, Schmeel FC, Coch C, et al. Cytokine-induced killer (CIK) cells in cancer immunotherapy: report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol 2015;141:839-49. [Crossref] [PubMed]
  48. Wang M, Cao JX, Pan JH, et al. Adoptive immunotherapy of cytokine-induced killer cell therapy in the treatment of non-small cell lung cancer. PLoS One 2014;9:e112662. [Crossref] [PubMed]
  49. Han Y, Mu D, Liu T, et al. Autologous cytokine-induced killer (CIK) cells enhance the clinical response to PD-1 blocking antibodies in patients with advanced non-small cell lung cancer: A preliminary study. Thorac Cancer 2021;12:145-52. [Crossref] [PubMed]
  50. Jin C, Li J, Wang Y, et al. Impact of cellular immune function on prognosis of lung cancer patients after cytokine-induced killer cell therapy. Asian Pac J Cancer Prev 2014;15:6009-14. [Crossref] [PubMed]
  51. Introna M, Correnti F. Innovative Clinical Perspectives for CIK Cells in Cancer Patients. Int J Mol Sci 2018;19:358. [Crossref] [PubMed]
  52. Li Y, Sharma A, Wu X, et al. A Combination of Cytokine-Induced Killer Cells With PD-1 Blockade and ALK Inhibitor Showed Substantial Intrinsic Variability Across Non-Small Cell Lung Cancer Cell Lines. Front Oncol 2022;12:713476. [Crossref] [PubMed]
  53. Liu S, Meng Y, Liu L, et al. Rational pemetrexed combined with CIK therapy plus anti-PD-1 mAbs administration sequence will effectively promote the efficacy of CIK therapy in non-small cell lung cancer. Cancer Gene Ther 2023;30:277-87. [Crossref] [PubMed]
  54. Chen J, Chen Y, Feng F, et al. Programmed cell death protein-1/programmed death-ligand 1 blockade enhances the antitumor efficacy of adoptive cell therapy against non-small cell lung cancer. J Thorac Dis 2018;10:6711-21. [Crossref] [PubMed]
  55. Zhao L, Han L, Zhang Y, et al. Combination of PD-1 blockade and RetroNectin(®)-activated cytokine-induced killer in preheavily treated non-small-cell lung cancer: a retrospective study. Immunotherapy 2018;10:1315-23. [Crossref] [PubMed]
  56. Hui Z, Zhang X, Ren B, et al. Rapid Response of Advanced Squamous Non-Small Cell Lung Cancer with Thrombocytopenia after First-Line Treatment with Pembrolizumab Plus Autologous Cytokine-Induced Killer Cells. Front Immunol 2015;6:633. [Crossref] [PubMed]
  57. Xiao Z, Wang CQ, Zhou MH, et al. The Antitumor Immunity and Tumor Responses of Chemotherapy with or without DC-CIK for Non-Small-Cell Lung Cancer in China: A Meta-Analysis of 28 Randomized Controlled Trials. J Immunol Res 2018;2018:9081938. [Crossref] [PubMed]
  58. Wang S, Wang X, Zhou X, et al. DC-CIK as a widely applicable cancer immunotherapy. Expert Opin Biol Ther 2020;20:601-7. [Crossref] [PubMed]
  59. Chen CL, Pan QZ, Weng DS, et al. Safety and activity of PD-1 blockade-activated DC-CIK cells in patients with advanced solid tumors. Oncoimmunology 2018;7:e1417721. [Crossref] [PubMed]
  60. Sharma A, Schmidt-Wolf IGH. 30 years of CIK cell therapy: recapitulating the key breakthroughs and future perspective. J Exp Clin Cancer Res 2021;40:388. [Crossref] [PubMed]
  61. Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol 2014;192:5451-8. [Crossref] [PubMed]
  62. van den Berg JH, Heemskerk B, van Rooij N, et al. Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J Immunother Cancer 2020;8:e000848. [Crossref] [PubMed]
  63. Sarnaik A, Khushalani NI, Chesney JA, et al. Long-term follow up of lifileucel (LN-144) cryopreserved autologous tumor infiltrating lymphocyte therapy in patients with advanced melanoma progressed on multiple prior therapies. J Clin Oncol 2020;38:10006.
  64. Creelan BC, Wang C, Teer JK, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med 2021;27:1410-8. [Crossref] [PubMed]
  65. Meaningful Response to TILs in NSCLC. Cancer Discov 2021;11:2117-8. [Crossref] [PubMed]
  66. van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013;31:e439-42. [Crossref] [PubMed]
  67. Achilles Therapeutics Presents Encouraging Phase I/IIa Update on Clonal Neoantigen Reactive T Cells in Advanced NSCLC and Melanoma at ESMO IO Congress 2022. [retrieved Oct 7th, 2022]. Available online: https://ir.achillestx.com/news-releases/news-release-details/achilles-therapeutics-presents-encouraging-phase-iiia-update
  68. Ratto GB, Melioli G, Zino P, et al. Immunotherapy with the use of tumor-infiltrating lymphocytes and interleukin-2 as adjuvant treatment in stage III non-small-cell lung cancer. A pilot study. J Thorac Cardiovasc Surg 1995;109:1212-7. [Crossref] [PubMed]
  69. Zhong R, Han B, Zhong H. A prospective study of the efficacy of a combination of autologous dendritic cells, cytokine-induced killer cells, and chemotherapy in advanced non-small cell lung cancer patients. Tumour Biol 2014;35:987-94. [Crossref] [PubMed]
  70. Li DP, Li W, Feng J, et al. Adjuvant chemotherapy with sequential cytokine-induced killer (CIK) cells in stage IB non-small cell lung cancer. Oncol Res 2015;22:67-74. [Crossref] [PubMed]
  71. Huang J, Kan Q. Chemotherapy in combination with cytokine-induced killer cell transfusion: An effective therapeutic option for patients with extensive stage small cell lung cancer. Int Immunopharmacol 2017;46:170-7. [Crossref] [PubMed]
  72. Zhao Y, Qiao G, Wang X, et al. Combination of DC/CIK adoptive T cell immunotherapy with chemotherapy in advanced non-small-cell lung cancer (NSCLC) patients: a prospective patients' preference-based study (PPPS). Clin Transl Oncol 2019;21:721-8. [Crossref] [PubMed]
  73. Chen D, Sha H, Hu T, et al. Cytokine-induced killer cells as a feasible adoptive immunotherapy for the treatment of lung cancer. Cell Death Dis 2018;9:366. [Crossref] [PubMed]
  74. Tian L, Wang W, Yu B, et al. Efficacy of dendritic cell-cytokine induced killer cells combined with concurrent chemoradiotherapy on locally advanced non-small cell lung cancer. J BUON 2020;25:2364-70.
  75. Wang L, Dai Y, Zhu F, et al. Efficacy of DC-CIK-based immunotherapy combined with chemotherapy in the treatment of intermediate to advanced non-small cell lung cancer. Am J Transl Res 2021;13:13076-83.
  76. Zhang L, Yang X, Sun Z, et al. Dendritic cell vaccine and cytokine-induced killer cell therapy for the treatment of advanced non-small cell lung cancer. Oncol Lett 2016;11:2605-10. [Crossref] [PubMed]
  77. Ratto GB, Zino P, Mirabelli S, et al. A randomized trial of adoptive immunotherapy with tumor-infiltrating lymphocytes and interleukin-2 versus standard therapy in the postoperative treatment of resected nonsmall cell lung carcinoma. Cancer 1996;78:244-51. [Crossref] [PubMed]
  78. Wang Z, Liu X, Till B, et al. Combination of Cytokine-Induced Killer Cells and Programmed Cell Death-1 Blockade Works Synergistically to Enhance Therapeutic Efficacy in Metastatic Renal Cell Carcinoma and Non-Small Cell Lung Cancer. Front Immunol 2018;9:1513. [Crossref] [PubMed]
  79. Xiao Z, Wang CQ, Feng JH, et al. Effectiveness and safety of chemotherapy with cytokine-induced killer cells in non-small cell lung cancer: A systematic review and meta-analysis of 32 randomized controlled trials. Cytotherapy 2019;21:125-47. [Crossref] [PubMed]
  80. Xiao Z, Wang CQ, Zhou MH, et al. Clinical efficacy and safety of CIK plus radiotherapy for lung cancer: A meta-analysis of 16 randomized controlled trials. Int Immunopharmacol 2018;61:363-75. [Crossref] [PubMed]
  81. Gargett T, Yu W, Dotti G, et al. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade. Mol Ther 2016;24:1135-49. [Crossref] [PubMed]
Cite this article as: Lv L, Chen W, Chen N, Cui E. Advances of cell therapy in lung cancer: a narrative review. J Thorac Dis 2023;15(12):7050-7062. doi: 10.21037/jtd-23-1015

Download Citation