PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review)

  • Authors:
    • Shuangcui Wang
    • Changyu Liu
    • Chenxin Yang
    • Yutong Jin
    • Qian Cui
    • Dong Wang
    • Ting Ge
    • Guixin He
    • Wentao Li
    • Guan Zhang
    • Aqing Liu
    • Ying Xia
    • Yunhe Liu
    • Jianchun Yu
  • View Affiliations

  • Published online on: September 26, 2024     https://doi.org/10.3892/ol.2024.14700
  • Article Number: 567
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

T cells play an important role in cancer, and energy metabolism can determine both the proliferation and differentiation of T cells. The inhibition of immune checkpoint molecules programmed cell death protein 1 (PD‑1) and cytotoxic T‑lymphocyte associated protein 4 (CTLA‑4) are a promising cancer treatment. In recent years, research on CD28 has increased. Although numerous reports involve CD28 and its downstream PI3K/AKT/mTOR signaling mechanisms in T cell metabolism, they have not yet been elucidated. A literature search strategy was used for the databases PubMed, Scopus, Web of Science and Cochrane Library to ensure broad coverage of medical and scientific literature, using a combination of keywords including, but not limited to, ‘lung cancer’ and ‘immunotherapy’. Therefore, the present study reviewed the interaction and clinical application of the PD‑1/CTLA‑4/CD28 and PI3K/AKT/mTOR pathways in T cells, aiming to provide a theoretical basis for immunotherapy in clinical cancer patients.

Introduction

Cancer, as a serious health problem, represents one of the main causes of morbidity and mortality worldwide (1). Immunotherapy is a treatment method that utilizes the body's own immune system to combat diseases and has shown potential in cancer treatment. However, despite significant success, immunotherapy still faces multiple challenges (2). Genetic heterogeneity within tumors may lead to inconsistent responses of tumor cells in different regions to immunotherapy, while antigen loss makes it difficult for the immune system to recognize and target all tumor cells (3). Tumor cells may develop resistance to immunotherapy through various mechanisms, such as upregulation or downregulation of programmed death-ligand 1 (PD-L1), changes in the IFNγ signaling pathway and alterations in metabolic pathways (4). For instance, Choi et al (4) found that the consumption of developmentally regulated GTP binding protein 2 in melanoma cells not only increases the expression of PD-L1 in tumor cells, but also increases the proportion of IFN-expressing CD8 T cells in tumor-infiltrating immune cells. Immunotherapy may cause immune-related adverse events (irAEs), such as inflammatory reactions in the endocrine system, skin, digestive tract and lungs (5). Therefore, it is important to overcome these adverse factors by developing new therapies (6).

In previous years, immunotherapy has been developed to design effective treatment methods to enhance the specificity and intensity of the immune system towards cancer (7). T cells are cellular effectors and coordinators in cancer, which serve as adaptive immune cells required for immune tolerance, host defense, immune memory and homeostasis (8). A large amount of data indicates that T cell activation, clone amplification and effector differentiation are closely related to cell energy metabolism (9). Based on studies of immune checkpoint programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), the inhibition of immune checkpoints can more effectively activate T cells and eliminate cancer cells (7) and the immune checkpoint inhibitors may have important therapeutic value (6).

The PI3K/AKT/mTOR pathway represents a primary signaling pathway that regulates processes, including cell metabolism, apoptosis and proliferation (10,11). The activation of PI3K/AKT can strengthen the nutrient intake and energy production of CD8+T cells, whereas mTOR is responsible for participating in regulating both innate and adaptive immune systems and the biological effects of immune cell stimulation (8,12). PI3K/AKT activation can promote PD-L1 expression and co-stimulate CD28, which can enhance T cell activation and metabolism. Therefore, the present article mainly explored the interaction and clinical application of the PD-1/CTLA-4/CD28 and PI3K/AKT/mTOR pathways in T cells.

A literature search strategy was adopted using the PubMed (https://pubmed.ncbi.nlm.nih.gov) database to ensure broad coverage of medical and scientific literature. The present study used a combination of keywords including ‘lung cancer’, ‘PD-1’, ‘CTLA-4’, ‘CD28’, ‘PI3K’, ‘AKT’, ‘mTOR’, ‘T cell’ and ‘immunotherapy’.

Proliferation and differentiation process of Tn/Tm in cancer and changes in energy metabolism

By recognizing and killing tumor cells, T lymphocytes protect the body from cancer (13). According to the functions and phenotypes, T lymphocytes can be mainly classified into immature T cells (Tn) and memory T cells (Tm) (14). To be specific, Tn represents a dormant mature T cell. Through the circulation between the blood and secondary lymphoid organs, Tn exhibits immune surveillance functions (15,16). In addition, Tm is included in maintaining the rapid and long-term immune responses, also known as the memory immune responses (17,18). Situated between Tn and Tcm, Tscm is a significantly different subset (19). Tn and T memory stem cells (Tscm) can self-renew and differentiate into each subset of memory and effector T cells, which are central memory T cells (Tcm), Terminal effector T cells (Tte) and effector memory T cells (Tem) (14). Usually, Tcm cells exist in lymphoid organs and show no direct lytic function (20). Tscm and Tcm go through memory immune responses and quickly clone and proliferate to generate Tem and Tte which particularly kill tumor cells (21).

The energy requirements for T cell proliferation and differentiation are met through the prominent programming of cellular metabolism, and the different phenotypes of T cells can determine their different metabolic modes (2224). Initial T cells maintain the minimum ATP levels by uptaking basic nutrients via oxidative phosphorylation (OXPHOS) while maintaining basic metabolic needs by relying on fatty acid oxidation (FAO) and glutamine metabolism (8). However, under aerobic conditions, rapidly dividing T cells transition their metabolism from oxidative phosphorylation to aerobic glycolysis and glutamine breakdown (25). Despite the existence of oxygen, glucose can still ferment into lactic acid, which can enter the tricarboxylic acid cycle (the TCA cycle), where the main carbon flux is converted from glucose to glutamine (12,25). After encountering antigens, immature T cells rapidly transform into effector T cells, which exhibit increased nutrient absorption and glycolysis rates and a metabolic activation state, mainly relying on OXPHOS and aerobic glycolysis to maintain T cell adaptability and function (26,27).

Given the 10 enzymatic steps of glycolysis (which converts glucose to pyruvate), some intermediates are generated for the various biosynthetic pathways. That can involve de novo fatty acid synthesis, the pentose phosphate pathway, hexosamine biosynthesis and serine biosynthesis, where the pentose phosphate pathway plays an important role in cell growth and offers primary precursors for nucleotide synthesis (28,29). Therefore, glycolysis is not only an energy production pathway in T cells, but also the metabolic foundation for proliferating synthetic organisms (30). Glucose transporter 1 (GLUT1) is a key signaling molecule for T lymphocyte activation and metabolism (31), capturing glucose to convert it into lactic acid, which can be used for oxidative phosphorylation even with sufficient oxygen (32). Glucose is first converted to glucose-6-phosphate (G-6-P), then to fructose-6-phosphate (F-6-P) and further to F-1,6-BP by the key regulatory factor PFK1 in glycolysis (33). Next F-1,6-BP enters the second part of glycolysis, which ultimately produces ATP and pyruvate (34). Therefore, glycolysis contributes much to T cell proliferation and differentiation.

Structure and function of the PI3K/AKT/mTOR pathway

According to differences in structure and function, PI3K is classified into categories I, II and III (35). To generate 3,4,5-triphosphate phosphatidylinositol (PIP3), PI3K phosphorylates 4,5-diphosphate phosphatidylinositol. PIP3 co-acts with target proteins [including Akt and phosphoinositol dependent protein kinase (PDK1)] that involve pleckstrin homologous domains on the inner lobe of the plasma membrane (36). Also known as protein kinase B (PKB), there are three subtypes in Akt, which are Akt1/PKBα, Akt2/PKBβ and Akt3/PKBγ (37). To reach complete activation, Akt should be respectively phosphorylated by PDK1 and mTOR complex 2 (mTORC2) (38). MTOR contributes much as the key element of the two multi-subunit proteins that have various functions. The complex is called mTORC1 and mTORC2 (39). In turn, MTORC1 activation can control protein synthesis, metabolism and cell growth. MTOR is fundamentally part of PI3K related kinases (PIKK), and most PIKK members possess conserved domains (40).

The PI3K/Akt/mTOR signaling pathway exists in each of the mammalian cells and exerts a significant impact on different processes, including cell proliferation, metabolism, differentiation and migration. In T cells, PI3Kδ and PI3Kγ subtypes play an important role in development (41). In the process of thymogenesis, absent or inactivated two isomers hinder the CD4 CD8 double negative stage of T cell development. By contrast, PI3Kδ is a subtype that can contribute significantly to PI3K signal transduction in mature T cells (42). For CD8+T cells, IA type PI3K can be mainly activated by tyrosine kinase-related receptors, including T cell receptors (TCRs), cytokine receptors and co-stimulatory receptors (43). The PI3K/Akt signaling pathway in CD8+T cells is stimulated by the signaling pathways triggered by cytokines IL-12, IL-2, IL-7, IL-15 and IL-21 (44). IL-2 can produce sustained high levels of PIP3, whereas IL-15 can relatively stimulate PI3K weakly, which can cause lower levels of PIP3. Conversely, chemokine receptors and other G protein coupled receptors can activate IB-type PI3K. The mTOR pathway represents an important regulatory factor for cell growth and proliferation and is becoming an attractive target for cancer treatment (45). Apart from the cancer cells, mTOR contributes much to mediating T cell activation and differentiation (46).

Interactions between the PD-1/CTLA-4/CD28 and PI3K/AKT/mTOR pathways affect T cell metabolism

PD-1/CTLA-4/CD28 regulates T cell metabolic balance

T cells are activated via antigen recognition through TCRs and co-stimulatory signals including CD28 (47). CD28 is considered a biosensor for T cell metabolism (48). CD28 co-stimulation plays an important role in strengthening T cell activation and metabolism and is antagonized via the inhibitory and checkpoint immunotherapy receptors CTLA-4 and PD-1 (49).

PD-1 is an inhibitory receptor that can inhibit the T-cell immune responses (6). On the contrary, PD-L1 can promote the stimulation of CD28 without triggering the inhibitory signal of PD-1, thereby facilitating the activation of T cells (50). This reflects the intricate regulatory mechanisms among multiple key factors in the immune system.

In addition to revolutionizing cancer treatment, cancer immunotherapy targeting the inhibitory co-receptors PD-1 and CTLA-4 has also shown long-standing clinical benefits for different tumor types (51,52). CTLA-4 inhibits T cell activation while interacting with CD86 or CD80 through transduction of inhibitory signals and/or the inhibition that involves co-stimulatory protein CD28 (53,54). CTLA-4 can also downregulate T cell glycolysis 1, and related studies have shown that CTLA-4 blockade can affect the metabolic adaptability of T cells in tumors related to tumor glycolysis ability (48).

Interaction between PD-1/CTLA-4/CD28 and PI3K/AKT/ mTOR pathway affects T cell metabolism

CD28 co-stimulation increases T cell synthesis and metabolism, whereas CD28 family members PD-1 and CTLA-4 can inhibit T cell metabolic reprogramming (55). PD-1 can inhibit glycolysis, promote the FAO of endogenous lipids, and change nucleoside synthesis. In addition, PD-1 negatively regulates T cells through changing mitochondrial cristae formation, which hinders oxidative phosphorylation (56). Besides, CTLA-4 can inhibit Akt/mTORC1 on the CD28 signaling and PI3K/Akt/mTORC1 signaling pathways, which can cause reduced glycolysis and mitochondrial oxidative capacity (57,58). AKT can regulate important enzymes in T cell metabolism, including GLUT1, hexokinase 2 and pyruvate kinase isozymes M1/M2, and mTORC1 can promote the expression of GLUT1, which promotes the process of T cell glycolysis (Fig. 1).

Clinical treatment

Blocking treatment of CTLA-4

CTLA-4 is a key negative regulatory molecule recruited to the cell membrane during T-cell activation and can bind to helper molecules of the B7 family expressed by antigen-presenting cells (59,60). Since the binding of CTLA-4 effectively inhibits further activation and proliferation of T cells, the progression of immune response can be controlled and the occurrence of chronic autoimmune inflammation can also be reduced (61). In cancer treatment, the blocking of CTLA-4 can not only relieve the inhibition of anti-cancer T cells but also trigger a new immune response. Related studies have shown that ipilimumab is a therapeutic drug with primary clinical significance, especially for patients in advanced stages (62,63). Different from conventional treatment that directly kills cancer cells and rapidly causes tumor volume reduction, it may take several months for ipilimumab to stimulate T cell responses (64). In addition, other applications of ipizumab have also been pursued, especially in combination with other immunotherapies, such as effective immune checkpoint inhibitors, vaccine administration and small molecule tyrosine kinase inhibitors (65).

Blocking therapy of PD-1/PD-L1

The metabolism can affect the functions of the PD-1 pathway. In the cytoplasmic domain in PD-1, there are two tyrosine motifs. Upon binding to ligands, the phosphorylation of tyrosine residues of PD-1 occurs, which can cause the binding to The Src homology 2 and other protein tyrosine phosphatases (PTP) (66). Then, PTP can phosphorylate kinases and antagonize positive signals that occur via TCR and CD28, which affects the downstream signaling pathways (67), such as Extracellular signal-regulated kinase 1/2 (ERK) (68), PI3K-AKT (69), finally reducing T cell survival, activation and proliferation and altering cytokine production and metabolism.

Given the effectiveness of the PD-1 pathway blockade monotherapy, improved response rates are associated with the infiltration of CD8+T cells at the tumor margin and high levels of PD-L1 expression (70). However, PD-L1 expression is linked to treatment outcomes all the time, as some PD-L1 tumors exhibit poor response to PD-1 pathway blockade while others exhibit a good response. Therefore, multiple biomarkers may play a more effective role in predicting responses of anti-PD-1 monotherapy than PD-L1 expression levels alone (71). In addition, in Renal Cell Carcinoma, certain metabolic signals are linked to treatment failure, which may be caused by the increased metabolic adaptability of tumor cells. Conversely, the immunological markers that involve BACH2 encode transcription factors regulating differentiation and function of effector T cells and memory T cells, as well as CCL3, which can encode chemokines in leukocyte migration, and has a connection to successful RCC treatment of PD-L1 (72,73). In the recent metastatic melanoma data, there is a relationship between mesenchymal and ‘inhibitory inflammatory’ transcriptional phenotypes and the efficacy of PD-1 inhibitors (74).

mTOR inhibitors

The mTOR pathway is a crucial regulatory factor for innate (including the dendritic cells and macrophages) and adaptive effector (including the T and B lymphocytes) immune cell metabolism, proliferation and anti-inflammatory response (75). Since the mTOR pathway is usually dysregulated in various solid tumors and hematological malignancy types, mTOR inhibitors (mTORi) indicate the immunosuppressive method that can prevent transplant rejection in transplant patients (76) and the anti-tumor therapy, which can be combined with immunotherapy and carefully adjusted for the immunosuppressive dose (77). Using mTORi and reducing the dosage of other immunosuppressive drugs is related to improved overall survival in patients with cancer. research indicates that T cell anergy can be maintained by using mTORi and accompanying immune checkpoint inhibitor therapy. Besides, mTORi stimulates the differentiation of immature T cells into Tregs (78).

Hence, mTORi therapy decreases cancer progression in various malignant tumors and exerts antitumor effects, which is seen to be controversial (7981). Currently, only a few mTOR inhibitors are employed in clinical practice. All approved mTOR inhibitors belong to the first-generation mTOR inhibitors. Rapamycin is the first mTOR inhibitor approved by the FDA and is currently suitable for preventing organ rejection after organ transplantation. Also, it can be used alone or in combination with calcineurin inhibitors or corticosteroids (82,83) (Table I).

Table I.

Clinical use of related inhibitors.

Table I.

Clinical use of related inhibitors.

Target pointsDrug nameTumor type
PD-1ToripalimabMelanoma, nasopharyngeal carcinoma, epithelial carcinoma of the urinary tract, esophageal cancer, non-squamous non-small cell lung cancer
SintilimabHodgkin's lymphoma, non-squamous non-small cell lung cancer, lung squamous cell carcinoma, liver cancer, esophageal cancer, gastric or gastroesophageal junction cancer
CamrelizumabHodgkin's lymphoma, liver cancer, EGFR/ALK negative non-small cell lung cancer, esophageal squamous cell carcinoma, nasopharyngeal carcinoma, squamous non-small cell lung cancer
TislelizumabHodgkin's lymphoma, epithelial carcinoma of the urinary tract, squamous non-small cell lung cancer, EGFR/ALK negative non-small cell lung cancer, liver cancer, non-small cell carcinoma, MSI-H/dMMR advanced solid tumors, nasopharyngeal carcinoma, gastroesophageal junction adenocarcinoma (G/GEJ adenocarcinoma), esophageal squamous cell carcinoma
ZimberelimabHodgkin's lymphoma
PenpulimabHodgkin's lymphoma, squamous non-small cell lung cancer
SerplulimabMSI-H advanced solid tumor, non-small cell lung cancer, small cell lung cancer
pucotenlimabMSI-H advanced solid tumor
PembrolizumabMelanoma, non-small cell lung cancer, esophageal cancer, squamous cell carcinoma of the head and neck, MSI-H/dMMR advanced colorectal cancer, liver cancer, esophageal cancer/gastroesophageal junction cancer, triple negative breast cancer
NivolumabNon-small cell lung cancer, squamous cell carcinoma of the head and neck, gastric or gastroesophageal junction cancer, gastric cancer, esophageal adeno-carcinoma, epithelial carcinoma of the urinary tract, pleural mesothelioma, esophageal squamous cell carcinoma
PD-L1SugemalimabIII Nnon-small cell lung cancer, IV non-small cell lung cancer
EnvafolimabMSI-H/dMMR advanced solid tumors
AdebrelimabExtensive stage small cell lung cancer
AtezolizumabSmall cell lung cancer, hepatocellular carcinoma, non-small cell lung cancer, non-squamous non-small cell lung cancer
DurvalumabExtensive stage small cell lung cancer, unremovable III non-small cell lung cancer
CTLA-4IpilimumabMalignant pleural mesothelioma
PD-1/CTLA-4CadonilimabCervical carcinoma
mTORTemsirolimusGlioblastoma multiforme, neuroendocrine tumors, soft tissue sarcoma, prostatic cancer, cervical carcinoma
SirolimusCholangiocarcinoma, liver cancer
EverolimusPancreatic cancer, small cell lung cancer, transitional cell carcinoma, bone and soft tissue sarcoma, epithelial carcinoma of the urinary tract, colorectal cancer
RidaforolimusBone and soft tissue sarcoma, endometrial cancer

[i] PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; CTLA-4, cytotoxic T lymphocyte associated protein 4.

Summary and outlook

Cancer immunotherapy is a major breakthrough in the field of oncology (84). Despite significant clinical success, such as the use of immune checkpoint inhibitors to treat melanoma and other solid tumors, there are still several limitations and potential biases that require further research to overcome (85). Not all patients can benefit from immunotherapy, and irAEs, such as endocrine disorders, enteritis and pneumonia may occur, which can have serious effects on patients and sometimes even be fatal (86,87). The predictive value of biomarkers such as tumor mutational burden and PD-L1 expression levels is not consistent and is influenced by tumor heterogeneity (88,89). The use of precision medicine strategies to customize immunotherapy plans based on each patient's specific situation, develop safer immunotherapy strategies and reduce the incidence and severity of irAEs is currently an urgent problem that needs to be solved (90,91). In summary, although cancer immunotherapy has changed the face of oncology, there is still much work to be done to overcome its limitations, reduce bias and ensure its benefits for as many patients as possible.

Despite significant progress in the field of immunotherapy for lung cancer, there are still a number of challenges to be faced. The present article reviewed the components of the PI3K/AKT/mTOR signaling pathway and the interactions between the PD-1/CTLA-4/CD28 and the PI3K/AKT/mTOR pathways in T cells and their impact on T cell metabolism and proliferation ability. CD28 can promote T cell glycolysis through activating the PI3K/Akt/mTOR pathway, while CD28 is inhibited by PD-1 and CTLA4. Therefore, it is necessary to further consider PI3K/AKT/mTOR pathway inhibitors combined with PD-1/PD-L1 inhibition to regulate T cell metabolism and proliferation, which can prevent and treat cancer.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

SCW designed the review, prepared the figure, wrote the manuscript and made substantial contributions to conception and design, acquisition of data, and analysis and interpretation of data. CYL, CXY, YTJ and QC participated in the conception and design of the study. DW, TG and GXH analysed and interpretated the data. WTL, GZ, AQL, YX and YHL were involved in the conception and design of the study and revised the manuscript. JCY revised the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Shi H, Zhang W, Zhang L, Zheng Y and Dong T: Comparison of different predictive biomarker testing assays for PD-1/PD-L1 checkpoint inhibitors response: A systematic review and network meta-analysis. Front Immunol. 14:12652022023. View Article : Google Scholar : PubMed/NCBI

3 

Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z and Xie Z: Immunomodulatory precision: A narrative review exploring the critical role of immune checkpoint inhibitors in cancer treatment. Int J Mol Sci. 25:54902024. View Article : Google Scholar : PubMed/NCBI

4 

Choi SH, Mani M, Kim J, Cho WJ, Martin TFJ, Kim JH, Chu HS, Jeong WJ, Won YW, Lee BJ, et al: DRG2 is required for surface localization of PD-L1 and the efficacy of anti-PD-1 therapy. Cell Death Discov. 10:2602024. View Article : Google Scholar : PubMed/NCBI

5 

Shen X, Yang J, Qian G, Sheng M, Wang Y, Li G and Yan J: Treatment-related adverse events of immune checkpoint inhibitors in clinical trials: A systematic review and meta-analysis. Front Oncol. 14:13917242024. View Article : Google Scholar : PubMed/NCBI

6 

Han Y, Liu D and Li L: PD-1/PD-L1 pathway: Current researches in cancer. Am J Cancer Res. 10:727–742. 2020.PubMed/NCBI

7 

Chen C, Wang Z and Qin Y: CRISPR/Cas9 system: Recent applications in immuno-oncology and cancer immunotherapy. Exp Hematol Oncol. 12:952023. View Article : Google Scholar : PubMed/NCBI

8 

Guo C, Chen S, Liu W, Ma Y, Li J, Fisher PB, Fang X and Wang XY: Immunometabolism: A new target for improving cancer immunotherapy. Adv Cancer Res. 143:195–253. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Lim SA, Su W, Chapman NM and Chi H: Lipid metabolism in T cell signaling and function. Nat Chem Biol. 18:470–481. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Mafi S, Mansoori B, Taeb S, Sadeghi H, Abbasi R, Cho WC and Rostamzadeh D: mTOR-Mediated regulation of immune responses in cancer and tumor microenvironment. Front Immunol. 12:7741032021. View Article : Google Scholar : PubMed/NCBI

11 

He J, Zeng F, Jin XI, Liang L, Gao M, Li W, Li G and Zhou Y: YWHAH activates the HMGA1/PI3K/AKT/mTOR signaling pathway by positively regulating Fra-1 to affect the proliferation of gastric cancer cells. Oncol Res. 31:615–630. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Liang Y, Wang X, Wang H, Yang W, Yi P, Soong L, Cong Y, Cai J, Fan X and Sun J: IL-33 activates mTORC1 and modulates glycolytic metabolism in CD8+ T cells. Immunology. 165:61–73. 2022. View Article : Google Scholar : PubMed/NCBI

13 

Kruse B, Buzzai AC, Shridhar N, Braun AD, Gellert S, Knauth K, Pozniak J, Peters J, Dittmann P, Mengoni M, et al: CD4+ T cell-induced inflammatory cell death controls immune-evasive tumours. Nature. 618:1033–1040. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Saxena A, Dagur PK and Biancotto A: Multiparametric flow cytometry analysis of naïve, memory, and effector T cells. Methods Mol Biol. 2032:129–140. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Notarbartolo S and Abrignani S: Human T lymphocytes at tumor sites. Semin Immunopathol. 44:883–901. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Goronzy JJ and Weyand CM: Mechanisms underlying T cell ageing. Nat Rev Immunol. 19:573–583. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Zander R, Schauder D, Xin G, Nguyen C, Wu X, Zajac A and Cui W: CD4+ T cell help is required for the formation of a Cytolytic CD8+ T cell subset that protects against chronic infection and cancer. Immunity. 51:1028–1042.e4. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Omilusik KD and Goldrath AW: Remembering to remember: T cell memory maintenance and plasticity. Curr Opin Immunol. 58:89–97. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Fazeli P, Kalani M and Hosseini M: T memory stem cell characteristics in autoimmune diseases and their promising therapeutic values. Front Immunol. 14:12042312023. View Article : Google Scholar : PubMed/NCBI

20 

Liu Q, Sun Z and Chen L: Memory T cells: Strategies for optimizing tumor immunotherapy. Protein Cell. 11:549–564. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Zhang G, Liu A, Yang Y, Xia Y, Li W, Liu Y, Zhang J, Cui Q, Wang D, Liu X, et al: Clinical predictive value of naïve and memory T cells in advanced NSCLC. Front Immunol. 13:9963482022. View Article : Google Scholar : PubMed/NCBI

22 

Reina-Campos M, Scharping NE and Goldrath AW: CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol. 21:718–738. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Wißfeld J, Werner A, Yan X, Ten Bosch N and Cui G: Metabolic regulation of immune responses to cancer. Cancer Biol Med. 19:1528–1542. 2022.PubMed/NCBI

24 

Ma S, Ming Y, Wu J and Cui G: Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol. 21:419–435. 2024. View Article : Google Scholar : PubMed/NCBI

25 

Araujo L, Khim P, Mkhikian H, Mortales CL and Demetriou M: Glycolysis and glutaminolysis cooperatively control T cell function by limiting metabolite supply to N-glycosylation. Elife. 6:e213302017. View Article : Google Scholar : PubMed/NCBI

26 

Kumar A and Chamoto K: Immune metabolism in PD-1 blockade-based cancer immunotherapy. Int Immunol. 33:17–26. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Li F, Liu H, Zhang D, Ma Y and Zhu B: Metabolic plasticity and regulation of T cell exhaustion. Immunology. 167:482–494. 2022. View Article : Google Scholar : PubMed/NCBI

28 

Upadhyay S, Khan S and Hassan MI: Exploring the diverse role of pyruvate kinase M2 in cancer: Navigating beyond glycolysis and the Warburg effect. Biochim Biophys Acta Rev Cancer. 1879:1890892024. View Article : Google Scholar : PubMed/NCBI

29 

Barba I, Carrillo-Bosch L and Seoane J: Targeting the Warburg effect in cancer: Where do we stand? Int J Mol Sci. 25:31422024. View Article : Google Scholar : PubMed/NCBI

30 

von Meyenn L, Bertschi NL and Schlapbach C: Targeting T cell metabolism in inflammatory skin disease. Front Immunol. 10:22852019. View Article : Google Scholar : PubMed/NCBI

31 

Zhang Z and Li X, Yang F, Chen C, Liu P, Ren Y, Sun P, Wang Z, You Y, Zeng YX and Li X: DHHC9-mediated GLUT1 S-palmitoylation promotes glioblastoma glycolysis and tumorigenesis. Nat Commun. 12:58722021. View Article : Google Scholar : PubMed/NCBI

32 

Gerriets VA, Kishton RJ, Nichols AG, Macintyre AN, Inoue M, Ilkayeva O, Winter PS, Liu X, Priyadharshini B, Slawinska ME, et al: Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest. 125:194–207. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Adeva-Andany MM, Pérez-Felpete N, Fernández-Fernández C, Donapetry-García C and Pazos-García C: Liver glucose metabolism in humans. Biosci Rep. 36:e004162016. View Article : Google Scholar : PubMed/NCBI

34 

Icard P, Alifano M, Donnadieu E and Simula L: Fructose-1,6-bisphosphate promotes PI3K and glycolysis in T cells? Trends Endocrinol Metab. 32:540–543. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Bilanges B, Posor Y and Vanhaesebroeck B: PI3K isoforms in cell signalling and vesicle trafficking. Nat Rev Mol Cell Biol. 20:515–534. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Shen Q, Han Y, Wu K, He Y, Jiang X, Liu P, Xia C, Xiong Q, Liu R, Chen Q, et al: MrgprF acts as a tumor suppressor in cutaneous melanoma by restraining PI3K/Akt signaling. Signal Transduct Target Ther. 7:1472022. View Article : Google Scholar : PubMed/NCBI

37 

Akhtar N and Jabeen I: Pharmacoinformatic approaches to design novel inhibitors of protein kinase B pathways in cancer. Curr Cancer Drug Targets. 18:830–846. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Sun K, Luo J, Guo J, Yao X, Jing X and Guo F: The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthritis Cartilage. 28:400–409. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Jhanwar-Uniyal M, Wainwright JV, Mohan AL, Tobias ME, Murali R, Gandhi CD and Schmidt MH: Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regul. 72:51–62. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Chen Y and Zhou X: Research progress of mTOR inhibitors. Eur J Med Chem. 208:1128202020. View Article : Google Scholar : PubMed/NCBI

41 

Dwyer CJ, Arhontoulis DC, Rangel Rivera GO, Knochelmann HM, Smith AS, Wyatt MM, Rubinstein MP, Atkinson C, Thaxton JE, Neskey DM and Paulos CM: Ex vivo blockade of PI3K gamma or delta signaling enhances the antitumor potency of adoptively transferred CD8+ T cells. Eur J Immunol. 50:1386–1399. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Preite S, Gomez-Rodriguez J, Cannons JL and Schwartzberg PL: T and B-cell signaling in activated PI3K delta syndrome: From immunodeficiency to autoimmunity. Immunol Rev. 291:154–173. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Chapman NM, Yoder AN, Barbón KM, Bilal MY, Connolly SF and Houtman JC: Proline-rich tyrosine kinase 2 controls PI3-kinase activation downstream of the T cell antigen receptor in human T cells. J Leukoc Biol. 97:285–296. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Coppola C, Hopkins B, Huhn S, Du Z, Huang Z and Kelly WJ: Investigation of the Impact from IL-2, IL-7, and IL-15 on the growth and signaling of activated CD4+ T Cells. Int J Mol Sci. 21:78142020. View Article : Google Scholar : PubMed/NCBI

45 

Mossmann D, Park S and Hall MN: mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer. 18:744–757. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Werlen G, Jain R and Jacinto E: MTOR signaling and metabolism in Early T cell development. Genes (Basel). 12:7282021. View Article : Google Scholar : PubMed/NCBI

47 

Wen L: The tumor immunotherapy effect of dendritic cells carrying anti CD40 single chain antibodies and the expression and role of CD40 on CD4+T cell surface D. Suzhou University; 2012

48 

Zappasodi R, Serganova I, Cohen IJ, Maeda M, Shindo M, Senbabaoglu Y, Watson MJ, Leftin A, Maniyar R, Verma S, et al: CTLA-4 blockade drives loss of Treg stability in glycolysis-low tumours. Nature. 591:652–658. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Beckermann KE, Hongo R, Ye X, Young K, Carbonell K, Healey DCC, Siska PJ, Barone S, Roe CE, Smith CC, et al: CD28 costimulation drives tumor-infiltrating T cell glycolysis to promote inflammation. JCI Insight. 5:e1387292020. View Article : Google Scholar : PubMed/NCBI

50 

Sugiura D, Okazaki IM, Maeda TK, Maruhashi T, Shimizu K, Arakaki R, Takemoto T, Ishimaru N and Okazaki T: PD-1 agonism by anti-CD80 inhibits T cell activation and alleviates autoimmunity. Nat Immunol. 23:399–410. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Sun C, Mezzadra R and Schumacher TN: Regulation and function of the PD-L1 checkpoint. Immunity. 48:434–452. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Ribas A and Wolchok JD: Cancer immunotherapy using checkpoint blockade. Science. 359:1350–1355. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Lin F, Chen Y, Huang B, Ruan S, Lin J, Chen Z, Huang C and Zhao B: Application of immune checkpoint inhibitors for resectable gastric/gastroesophageal cancer. Front Pharmacol. 15:13915622024. View Article : Google Scholar : PubMed/NCBI

54 

Oyewole-Said D, Konduri V, Vazquez-Perez J, Weldon SA, Levitt JM and Decker WK: Beyond T-cells: Functional characterization of CTLA-4 expression in immune and non-immune cell types. Front Immunol. 11:6080242020. View Article : Google Scholar : PubMed/NCBI

55 

Xia S, Chen Q and Niu B: CD28: A new drug target for immune disease. Curr Drug Targets. 21:589–598. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Kumagai S, Koyama S, Itahashi K, Tanegashima T, Lin YT, Togashi Y, Kamada T, Irie T, Okumura G, Kono H, et al: Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 40:201–218.e9. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Palaskas NJ, Garcia JD, Shirazi R, Shin DS, Puig-Saus C, Braas D, Ribas A and Graeber TG: Global alteration of T-lymphocyte metabolism by PD-L1 checkpoint involves a block of de novo nucleoside phosphate synthesis. Cell Discov. 5:622019. View Article : Google Scholar : PubMed/NCBI

58 

Dong S, Guo X, Han F, He Z and Wang Y: Emerging role of natural products in cancer immunotherapy. Acta Pharm Sin B. 12:1163–1185. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Pulanco MC, Madsen AT, Tanwar A, Corrigan DT and Zang X: Recent advancements in the B7/CD28 immune checkpoint families: New biology and clinical therapeutic strategies. Cell Mol Immunol. 20:694–713. 2023. View Article : Google Scholar : PubMed/NCBI

60 

Sharma P, Goswami S, Raychaudhuri D, Siddiqui BA, Singh P, Nagarajan A, Liu J, Subudhi SK, Poon C, Gant KL, et al: Immune checkpoint therapy-current perspectives and future directions. Cell. 186:1652–1669. 2023. View Article : Google Scholar : PubMed/NCBI

61 

Zhang J, Wei X, Zhang Q, Jiao X, Li K, Geng M, Cao Y, Wang D, Cheng J and Yang J: Fish uses CTLA-4 immune checkpoint to suppress mTORC1-Controlled T-cell glycolysis and immunity. J Immunol. 212:1113–1128. 2024. View Article : Google Scholar : PubMed/NCBI

62 

Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, Park K, Alexandru A, Lupinacci L, de la Mora Jimenez E, et al: Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 381:2020–2031. 2019. View Article : Google Scholar : PubMed/NCBI

63 

VanderWalde A, Bellasea SL, Kendra KL, Khushalani NI, Campbell KM, Scumpia PO, Kuklinski LF, Collichio F, Sosman JA, Ikeguchi A, et al: Ipilimumab with or without nivolumab in PD-1 or PD-L1 blockade refractory metastatic melanoma: A randomized phase 2 trial. Nat Med. 29:2278–2285. 2023. View Article : Google Scholar : PubMed/NCBI

64 

Cascone T, Leung CH, Weissferdt A, Pataer A, Carter BW, Godoy MCB, Feldman H, William WN Jr, Xi Y, Basu S, et al: Neoadjuvant chemotherapy plus nivolumab with or without ipilimumab in operable non-small cell lung cancer: The phase 2 platform NEOSTAR Trial. Nat Med. 29:593–604. 2023. View Article : Google Scholar : PubMed/NCBI

65 

Grimm MO, Esteban E, Barthélémy P, Schmidinger M, Busch J, Valderrama BP, Charnley N, Schmitz M, Schumacher U, Leucht K, et al: Tailored immunotherapy approach with nivolumab with or without nivolumab plus ipilimumab as immunotherapeutic boost in patients with metastatic renal cell carcinoma (TITAN-RCC): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 24:1252–1265. 2023. View Article : Google Scholar : PubMed/NCBI

66 

Liu L, Cheng Y, Zhang Z, Li J, Geng Y, Li Q, Luo D, Liang L, Liu W, Hu J and Ouyang W: Study on the allosteric activation mechanism of SHP2 via elastic network models and neural relational inference molecular dynamics simulation. Phys Chem Chem Phys. 25:23588–23601. 2023. View Article : Google Scholar : PubMed/NCBI

67 

Ai L, Xu A and Xu J: Roles of PD-1/PD-L1 pathway: Signaling, cancer, and beyond. Adv Exp Med Biol. 1248:33–59. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y and Hu LL: ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 19:1997–2007. 2020.PubMed/NCBI

69 

Quan Z, Yang Y, Zheng H, Zhan Y, Luo J, Ning Y and Fan S: Clinical implications of the interaction between PD-1/PD-L1 and PI3K/AKT/mTOR pathway in progression and treatment of non-small cell lung cancer. J Cancer. 13:3434–3443. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Huang Q, Wu X, Wang Z, Chen X, Wang L, Lu Y, Xiong D, Liu Q, Tian Y, Lin H, et al: The primordial differentiation of tumor-specific memory CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell. 185:4049–4066.e25. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Patel SP and Kurzrock R: PD-L1 Expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 14:847–856. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Karim S, Mirza Z, Chaudhary AG, Abuzenadah AM, Gari M and Al-Qahtani MH: Assessment of radiation induced therapeutic effect and cytotoxicity in cancer patients based on transcriptomic profiling. Int J Mol Sci. 17:2502016. View Article : Google Scholar : PubMed/NCBI

73 

Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ, Xu H, Pritchard TS, Fan J, Cheadle C, et al: The intratumoral balance between metabolic and immunologic gene expression is associated with Anti-PD-1 response in patients with renal cell carcinoma. Cancer Immunol Res. 4:726–733. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Van Damme H, Dombrecht B, Kiss M, Roose H, Allen E, Van Overmeire E, Kancheva D, Martens L, Murgaski A, Bardet PMR, et al: Therapeutic depletion of CCR8+ tumor-infiltrating regulatory T cells elicits antitumor immunity and synergizes with anti-PD-1 therapy. J Immunother Cancer. 9:e0017492021. View Article : Google Scholar : PubMed/NCBI

75 

Zou Z, Tao T, Li H and Zhu X: mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 10:312020. View Article : Google Scholar : PubMed/NCBI

76 

Lim LM, Kung LF, Kuo MC, Huang AM and Kuo HT: Timing of mTORI usage and outcomes in kidney transplant recipients. Int J Med Sci. 18:1179–1184. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Kumari A, Gesumaria L, Liu YJ, Hughitt VK, Zhang X, Ceribelli M, Wilson KM, Klumpp-Thomas C, Chen L, McKnight C, et al: mTOR inhibition overcomes RSK3-mediated resistance to BET inhibitors in small cell lung cancer. JCI Insight. 8:e1566572023. View Article : Google Scholar : PubMed/NCBI

78 

Daenthanasanmak A, Lin Y, Zhang M, Bryant BR, Petrus MN, Bamford RN, Thomas CJ, Miljkovic MD, Conlon KC and Waldmann TA: Enhanced efficacy of JAK1 inhibitor with mTORC1/C2 targeting in smoldering/chronic adult T cell leukemia. Transl Oncol. 14:1009132021. View Article : Google Scholar : PubMed/NCBI

79 

Bai S, Taylor SE, Jamalruddin MA, McGonigal S, Grimley E, Yang D, Bernstein KA and Buckanovich RJ: Targeting therapeutic resistance and multinucleate giant cells in CCNE1-Amplified HR-Proficient ovarian cancer. Mol Cancer Ther. 21:1473–1484. 2022. View Article : Google Scholar : PubMed/NCBI

80 

Sawaki M, Muramatsu Y, Togo K and Iwata H: Real-world treatment patterns of subsequent therapy after palbociclib in patients with advanced breast cancer in Japan Breast. 70:1–7. 2023.PubMed/NCBI

81 

Qiu HY, Wang PF and Zhang M: A patent review of mTOR inhibitors for cancer therapy (2011–2020). Expert Opin Ther Pat. 31:965–975. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Vidigal AC, de Lucena DD, Beyerstedt S and Rangel ÉB: A comprehensive update of the metabolic and toxicological considerations for immunosuppressive drugs used during pancreas transplantation. Expert Opin Drug Metab Toxicol. 19:405–427. 2023. View Article : Google Scholar : PubMed/NCBI

83 

Franzin R, Netti GS, Spadaccino F, Porta C, Gesualdo L, Stallone G, Castellano G and Ranieri E: The use of immune checkpoint inhibitors in oncology and the occurrence of AKI: Where do we stand? Front Immunol. 11:5742712020. View Article : Google Scholar : PubMed/NCBI

84 

Zhang Y and Zhang Z: The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 17:807–821. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Catalano M, Iannone LF, Nesi G, Nobili S, Mini E and Roviello G: Immunotherapy-related biomarkers: Confirmations and uncertainties. Crit Rev Oncol Hematol. 192:1041352023. View Article : Google Scholar : PubMed/NCBI

86 

Al-Hussainy A, Adams J, Simmons J and Kennedy J: Immune checkpoint inhibitor associated diarrhoea. BMJ Case Rep. 17:e2590572024. View Article : Google Scholar : PubMed/NCBI

87 

Blum SM, Rouhani SJ and Sullivan RJ: Effects of immune-related adverse events (irAEs) and their treatment on antitumor immune responses. Immunol Rev. 318:167–178. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Ma S, Lei J and Lai X: Modeling tumour heterogeneity of PD-L1 expression in tumour progression and adaptive therapy. J Math Biol. 86:382023. View Article : Google Scholar : PubMed/NCBI

89 

Yi M, Zheng X, Niu M, Zhu S, Ge H and Wu K: Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol Cancer. 21:282022. View Article : Google Scholar : PubMed/NCBI

90 

Chen Z, Yue Z, Wang R, Yang K and Li S: Nanomaterials: A powerful tool for tumor immunotherapy. Front Immunol. 13:9794692022. View Article : Google Scholar : PubMed/NCBI

91 

Li J, Zeng H, Li L, Yang Q, He L and Dong M: Advanced generation therapeutics: Biomimetic nanodelivery system for tumor immunotherapy. ACS Nano. 17:24593–24618. 2023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang S, Liu C, Yang C, Jin Y, Cui Q, Wang D, Ge T, He G, Li W, Zhang G, Zhang G, et al: PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review). Oncol Lett 28: 567, 2024.
APA
Wang, S., Liu, C., Yang, C., Jin, Y., Cui, Q., Wang, D. ... Yu, J. (2024). PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review). Oncology Letters, 28, 567. https://doi.org/10.3892/ol.2024.14700
MLA
Wang, S., Liu, C., Yang, C., Jin, Y., Cui, Q., Wang, D., Ge, T., He, G., Li, W., Zhang, G., Liu, A., Xia, Y., Liu, Y., Yu, J."PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review)". Oncology Letters 28.6 (2024): 567.
Chicago
Wang, S., Liu, C., Yang, C., Jin, Y., Cui, Q., Wang, D., Ge, T., He, G., Li, W., Zhang, G., Liu, A., Xia, Y., Liu, Y., Yu, J."PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review)". Oncology Letters 28, no. 6 (2024): 567. https://doi.org/10.3892/ol.2024.14700