Research article

STAT3 down-regulation for IL-2 inducing TIL-A clinical clue for spatial quantitative pathway analysis

  • † These two authors contributed equally.
  • Received: 30 June 2024 Revised: 18 November 2024 Accepted: 20 November 2024 Published: 02 December 2024
  • Tumor-infiltrating lymphocytes (TILs) can be used in two clinical areas: Adoptive cell therapy (ACT) and studying TIL immune properties ex vivo. After we studied ACT for more than thirty years, we are now going to study T-cells' immune properties for personalized immunotherapy. A spatial-quantitative network model can be applied to analyze the immune properties of T-cells and then develop new strategies under a machine learning algorithm. This study utilized STAT3 complex down-regulation for IL-2 inducing cytotoxic T-lymphocyte (CTL) to study T-cell characteristics. After studying spatial-order regulation from the SMAD-STAT3 axis of multiple protein complexes, we focused on a second protein, or STAT3, knockdown for IL-2-inducing CTL. Because machine learning algorithms require a good pathway configuration to mine targeting genes and targeting drugs, the spatial-time quantification network can address the spatial competition of complex proteins binding to downstream proteins/DNA in network analysis. The research demonstrated that STAT3 downregulation can reactivate CTL under a spatial-order quantification network combined with IL2 inducing. After studying the algorithm of a spatial quantitative network under STAT3 downregulation, we can use the spatial-complex configurations to analyze complex regulation in artificial intelligence for future personalized therapy.

    Citation: Yun Yang, Shuyi Chen, Wenqian Li, Jason Chengwu Duan, Yuan Yuan, Victor S. Xie, Biaoru Li. STAT3 down-regulation for IL-2 inducing TIL-A clinical clue for spatial quantitative pathway analysis[J]. AIMS Allergy and Immunology, 2024, 8(4): 283-295. doi: 10.3934/Allergy.2024017

    Related Papers:

    [1] Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda . IL-17 signaling is regulated through intrinsic stability control of mRNA during inflammation. AIMS Allergy and Immunology, 2022, 6(3): 188-199. doi: 10.3934/Allergy.2022014
    [2] Boel De Paepe . A recipe for myositis: nuclear factor κB and nuclear factor of activated T-cells transcription factor pathways spiced up by cytokines. AIMS Allergy and Immunology, 2017, 1(1): 31-42. doi: 10.3934/Allergy.2017.1.31
    [3] Manal Alkan, Fadel Sayes, Abdulraouf Ramadan, Francois Machavoine, Michel Dy, Elke Schneider, Nathalie Thieblemont . Basophil activation through TLR2 and TLR4 signaling pathways. AIMS Allergy and Immunology, 2018, 2(3): 126-140. doi: 10.3934/Allergy.2018.3.126
    [4] Sabrine Louhaichi, Mariem Salhi, Anissa Berraïes, Besma Hamdi, Jamel Ammar, Kamel Hamzaoui, Agnès Hamzaoui . Co-inhibitory receptors in female asthmatic patients: Correlation with IL-17 and IL-26. AIMS Allergy and Immunology, 2018, 2(1): 10-23. doi: 10.3934/Allergy.2018.1.10
    [5] Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda . Tyk2-mediated homeostatic control by regulating the PGE2-PKA-IL-10 axis. AIMS Allergy and Immunology, 2021, 5(3): 175-183. doi: 10.3934/Allergy.2021013
    [6] Daniil Shevyrev, Valeriy Tereshchenko, Olesya Manova, Vladimir kozlov . Homeostatic proliferation as a physiological process and a risk factor for autoimmune pathology. AIMS Allergy and Immunology, 2021, 5(1): 18-32. doi: 10.3934/Allergy.2021002
    [7] Kochar Khasro Saleh, Semih Dalkiliç, Lütfiye Kadioğlu Dalkiliç, Bahra Radhaa Hamarashid, Sevda Kirbağ . Targeting cancer cells: from historic methods to modern chimeric antigen receptor (CAR) T-Cell strategies. AIMS Allergy and Immunology, 2020, 4(2): 32-49. doi: 10.3934/Allergy.2020004
    [8] Caterina Marangio, Rosa Molfetta, Erisa Putro, Alessia Carnevale, Rossella Paolini . Exploring the dynamic of NKG2D/NKG2DL axis: A central regulator of NK cell functions. AIMS Allergy and Immunology, 2025, 9(2): 70-88. doi: 10.3934/Allergy.2025005
    [9] Mansur Aliyu, Sayed-Hamidreza Mozhgani, Omid Kohandel Gargari, Mustapha Ahmed Yusuf, Ali Akbar Saboor-Yaraghi . The host immune responses to SARS-CoV-2 and therapeutic strategies in the treatment of COVID-19 cytokine storm. AIMS Allergy and Immunology, 2021, 5(4): 240-258. doi: 10.3934/Allergy.2021018
    [10] Tuba Karakurt, Nurhan Kasap, Kübra Aslan, Hayrunnisa Bekis Bozkurt, Fatma Bal Cetinkaya, Gizem Uslu, Zafer Bicakci, Filiz Özen, Özlem Cavkaytar, Ahmet Eken, Mustafa Arga . Suspected ALPS with clinical and laboratory findings: Three patients—three different diagnoses. AIMS Allergy and Immunology, 2023, 7(4): 304-312. doi: 10.3934/Allergy.2023020
  • Tumor-infiltrating lymphocytes (TILs) can be used in two clinical areas: Adoptive cell therapy (ACT) and studying TIL immune properties ex vivo. After we studied ACT for more than thirty years, we are now going to study T-cells' immune properties for personalized immunotherapy. A spatial-quantitative network model can be applied to analyze the immune properties of T-cells and then develop new strategies under a machine learning algorithm. This study utilized STAT3 complex down-regulation for IL-2 inducing cytotoxic T-lymphocyte (CTL) to study T-cell characteristics. After studying spatial-order regulation from the SMAD-STAT3 axis of multiple protein complexes, we focused on a second protein, or STAT3, knockdown for IL-2-inducing CTL. Because machine learning algorithms require a good pathway configuration to mine targeting genes and targeting drugs, the spatial-time quantification network can address the spatial competition of complex proteins binding to downstream proteins/DNA in network analysis. The research demonstrated that STAT3 downregulation can reactivate CTL under a spatial-order quantification network combined with IL2 inducing. After studying the algorithm of a spatial quantitative network under STAT3 downregulation, we can use the spatial-complex configurations to analyze complex regulation in artificial intelligence for future personalized therapy.



    Tumor-infiltrating lymphocytes (TILs) are heterogeneous immune cells that kills tumor cells in clinical applications [1][7], but the cytotoxic T-lymphocyte (CTL) can be blocked by the immunosuppressive tumor microenvironment (TME) within tumors from myeloid-derived suppressor cells (MDSCs), regulatory T-cells (Tregs), and Programmed Death-Ligan 1 (PD-L1) [8][12]. Signal transducer and activator of transcription-3 (STAT3) is produced from MDSC and Tregs to participate in the TGF-β pathway, resulting in immune suppressive action in the TME [13]. STAT3 upregulation improves the propagation of tumor cells and crosstalk among vascular endothelial growth factor (VEGF), interleukin 6 (IL-6), interleukin 10 (IL-10), and interleukin 23 (IL-23) to promote tumor growth, angiogenesis, and invasion [14][17]. STAT3 can also inhibit the expression of immune-stimulatory factors, resulting in a TME decreasing immune responses to tumors [18],[19]. Because we have reported that a spatial-timely quantification network can address the spatial competition of complex proteins binding to downstream proteins/DNA in network analysis which have been reported, we apply for the algorithm to study STAT3 downregulation. After we set up Suppressor of Mothers Against Decapentaplegic (SMAD)-STAT3 complex spatial-order regulation to compete for downstream binding sites, we use siRNA knockdown STAT3 to study the hypothesis due to SMAD-STAT3 complex inhibiting CTL functions. We used SMAD-STAT3 complex configuration under STAT3 downregulation to study IL-2-inducing CTL. The research goal is to confirm that spatial-order quantification network is more powerful than traditional pathway and study that STAT3 down-regulation will increase TIL activity. After studying SMAD-STAT3 complex downregulation in the spatial order quantitative network, we can use the spatial-complex configurations to analyze complex regulation in machine learning algorithms for future personalized therapy.

    According to TIL CD8 immune networks which we have studied, at least three pathways are majorly involved in T-cell immune response to tumor cells: CD8+ T-cell receptor (TCR) signal from TCR antigen associated with MHC class I; T-cells reactivated with IL-2 to produce Tumor necrosis factor alpha (TNF-α), Interferon-gamma (IFN-γ), Granzyme-B (GZMB), and perforin-1 (PRF1) to kill tumor cells; TGF-β pathway with SMAD-STAT3 combination to maintain quiescent TIL to block the antitumor functions in tumor tissues [20][22]. To study spatial-timely pathway and SMAD-STAT3 complex inhibiting CTL, we put promoters of IL-2, TNF-α, IFN-γ, GZMB, and PRF1 into TRANSFAC to screen binding protein (http://bioinfo.wilmer.jhu.edu/PDI/index.html) by using TRANSFAC Matrices (Cutoff < 0.001).

    After the discovery of STAT3 protein-predicted binding site in the promoters of IL-2, TNF-α, IFN-γ, GZMB, and PRF1, suggesting the STAT3 regulates CTL functions, two of the genes (TNF-α and IFN-γ) were studied by chromatin immunoprecipitation (ChIP). In detail, we used the cloned and cultured TIL from liver tumor tissues without IL-2 on day-7 and with IL-2 induction on day-10 for IL-2 induction 3 days as per our previous report and design [23][27]. The ChIP assay for the two groups was performed as routine protocol [28]. Briefly, both groups of cells were incubated with 1 mL lysis buffer (20 mM HEPES [pH 7.9], 350 mM NaCl, 0.1% NP-40, 1 mM DTT, 0.2 mM PMSF) on ice for 30 min, DNA fragmentation by sonication into 400–800 bp, Protein A (Thermo Scientific™ 88802) beads with 1 µg STAT3 antibodies were incubated for two hours under at 4 °C to pull down the bound DNA fragments. The fragments with the appropriate size of DNA were purified and ran PCR. AP1 also binds the promoters of TNF-α and IFN-γ, so AP1 was used as a positive control and IgG was used as a negative control.

    After discovering the SMAD-STAT3 DNA binding site within promoters of TNF-α, IFN-γ, GZMB, and PRF1, we used siRNA STAT3 to downregulate STAT3 and study inhibited STAT3 regarding enhancing CTL genes' expression. The culture cloned TIL from liver tumor tissues was cultured for 21 days as the previous report [23][27],[29],[30]. In detail, after culturing from day-0 to day-12, the growth medium was replaced with Opti-MEM Invitrogen, Carlsbad CA). For each transfection, 100 nm and 200 nm targeting STAT3 siRNA (CAT#: SR321907, Origene) with scrambled control using DharmaFECT (Dharmacon, Lafayette, CO) according to the manufacturer's protocol. Each experimental condition was performed in triplicate. After cells were transfected for 72 hours, the cells were harvested and assayed for mRNA and protein for CTL gene expression, as in our previous reports [31][33]. Briefly, mRNA expression was measured for STAT3 downregulation with two of the CTL genes, TNF-α and INF-γ, by Q-rtPCR. Western blot is used as per our previous manuals [34][36] to confirm the protein level of TNF-α and INF-γ with STAT3 down-expression (STAT3 antibodies are ab31370).

    To study CTL from siRNA STAT3 down-regulation, CTL function was defined by MTT as previous manual reports [37][40]. Briefly, CTL was assayed for the HepG2 cell line as our report [41],[42]. The cells were seeded in 96-well tissue culture plates at a density of 5 × 103 cells/well. TIL was measured for effector-to-target ratio by 1:25 and 1:50 at culture from day-15. On the day-15, the medium was removed and 100 µL of fresh medium containing 0.5 mg/mL MTT (Roche; Manheim, Germany) was added to each well. The cells were incubated at 37 °C in a humidified 5% CO2 atmosphere for 4 hours, followed by the addition of 150 µL of solubilization solution (0.01 mol/L HCl in 100 g/L sodium dodecyl [SDS]) to each well and the incubation of cells for a further 10 minutes at 37 °C with gentle shaking. The optical density of the plates was measured using spectrophotometric absorbance at 570 nm in the Microplate Reader Model 550 (Bio-Rad; CA).

    According to the research purpose, the final mimic included IL2 activating pathway with CTL under TNF-α, INF-γ, GZMB, and PRF1 participation and blocked STAT3 within the network of TCR signal and IL2 activating pathway. To study the spatial-timely quantitative network from the SMAD-STAT3 complex regulation, TCR signal/and IL2 activating pathway/quiescent TIL-cell pathway hubs derive from our previous published genomic profile [26],[43],[44] and targeting SMAD-STAT3 within TCR signal and IL2 activating pathway from published STKE database. The complex regulation within spatial-timely quantitative network modeling is SMAD-STAT3 complex (Smad-2, Smad-3, and Smad-4) with combination ratio (0%, 25%, 50%, 75%, and 100%) and downstream transcriptional factor complex binding sites with combination ratio (0%, 25%, 50%, 75%, and 100%) under our Python script plugin into Cytoscape (http://www.cytoscape.org/) which we have written and reported [45],[46]. We are finally combining targeting SMAD-STAT3 with IL2 activating pathway for CTL under blocked STAT3 status.

    According to TIL CD8 immune networks, T-cells reactivated with IL-2 to produce TNF-α, IFN-γ, GZMB, and PRF1 to kill tumor cells. In order to discover STAT3 inhibiting CTL functions and to study spatial-timely pathways, we put promoters of IL2, TNF-α, IFN-γ, GZMB, and PRF1 into a platform of transcriptional factors binding screening (https://biogrid-lasagna.engr.uconn.edu/lasagna_search/). As Figure 1 shows, there are two matrix logos as STAT3 protein binding promoters: M00225 (Figure 1A) and M00497 (Figure 1B). As Table 1 shows, STAT3 protein in silico binding sites are demonstrated within the promoter of TNF-α, IFN-γ, GZMB, PRF1, and IL2, in which STAT3 has five binding sequences in PRF1 promoter, four binding sites within that of IL2, three binding sites within that of TNF-α and two binding sites within that of IFN-γ and GZMB. As Table 1 shows, the P-values are satisfactory within both strands.

    Because the promoters of CTL genes were discovered for binding by STAT3 proteins, two of the genes (TNF-α and IFN-γ) are applied for chromatin immunoprecipitation (ChIP) assay. After we harvested the cultured cloned TIL from liver tumor tissues without IL-2 groups on day-7 and on day-10 after IL-2 induction, we performed a chromatin immunoprecipitation (ChIP) assay. We observed low TNF-α and IFN-γ binding on day-7 and high TNF-α and IFN-γ binding about 2.5-times significantly increased on day-10 (P = 0.044 and P = 0.0072, respectively). The results showed that STAT3 regulates TNF-α and IFN-γ expression after IL2 induction TILs. Because AP1 located with TNF-α and IFN-γ promoter was predicted in silico, AP1 was used as a positive control, as shown in Figure 1C, while IgG was used as a negative control.

    Figure 1.  STAT3 promoter binding Logo and ChIP assay. A) Shown M00225 logo; B) for M0049 logo; C) Observed low TNF-α and IFN-γ binding at day-7 and compared to about 2.5-times significantly increase by day-10, respectively. Because AP1 also located with TNF-α and IFN-γ promoter was predicted by in silico, AP1 was used as a positive control as shown, while IgG was used as a negative control as shown.
    Table 1.  CTL gene promoter binding motif.
    Name of promoters Binding protein name Promoters sequence Position Strand Score P-value E-value
    TNF-alpha STAT3 (M00225) CCGGGAATTCACA 608 12.52 0.0002 0.192
    TNF-alpha STAT3 (M00497) AATTCCC 612 + 7.3 0.00065 0.63
    TNF-alpha STAT3 (M00497) GAATTCCC 611 + 8.33 7.50E-05 0.073
    INF-gamma STAT3 (M00225) CGGAAACGATG 466 + 11.26 0.0004 0.38
    INF-gamma STAT3 (M00497) GAGTTCCC 868 7.58 0.0003 0.292
    GZMB STAT3 (M00225) CGGAAATGCCA 642 12.81 0.000125 0.146
    GZMB STAT3 (M00497) GAGTTCCC 809 7.58 0.0003 0.3
    PRF1 STAT3 (M00225) CGGGAAAAGAGA 132 10.47 0.00075 0.93
    PRF1 STAT3 (M00225) GGAAGTGGAT 794 + 11.54 0.0004 0.5
    PRF1 STAT3 (M00497) AATTCCA 1 + 7.31 0.00045 0.56
    PRF1 STAT3 (M00497) GATTCCG 318 + 7.79 0.000175 0.219
    PRF1 STAT3 (M00497) AGATTCCG 317 + 8.29 0.000125 0.157
    IL2 STAT3 (M00497) GAATTCCA 0 + 8.35 5.00E-05 0.063
    IL2 STAT3 (M00497) AGATTCCG 317 + 8.29 0.000125 0.157
    IL2 STAT3 (M00497) GATTCCG 318 + 7.79 0.000175 0.219
    IL2 STAT3 (M00497) AATTCCA 1 + 7.31 0.00045 0.56

     | Show Table
    DownLoad: CSV

    Two of the three pathways are majorly involved in T-cell immune CTL to kill tumor cells: CD8+ TCR signal from TCR antigen associated with MHC class I and T-cells reactivated with IL-2 to induce TNF-α, INF-γ, GZMB, and PRF1.

    Figure 2.  STAT3 downregulation for CTL gene expression and function change. A) Day-15 RT-qPCR quantification of targeting STAT3 mRNA. B) Confirmation of STAT3 downregulation by Western blot at day-15. C) Confirmation of STAT3 downregulation by CTL increase at day-15.

    After STAT3 was mined by transcriptional binding in promoters of TNF-α, INF-γ, GZMB, and PRF1 for CTL, according to the research goal, two of the CTL genes (TNF-α and IFN-γ) were also studied for the gene expression after STAT3 knockdown [23],[24],[47],[48]. The results showed that STAT3 down-regulating will increase TNF-α and IFN-γ expression as Figure 2A indicates that down-regulation STAT3 is expressing a level of 50% in the 100 nm group and 37% in the 200 nm group. TNF-α mRNA is 2.54 and 5.54 times higher transfected TILs than those in the scramble control group to compare 100 nm and 200 nm scramble TIL group (significantly increased in 200 nm group, P = 0.04172), respectively. The IFN-γ level is about 1.99 and 7.18 times higher in the transfected TILs group than those in the 100 nm and 200 nm scramble group (significantly increased in the 200 nm group, P = 0.049). As Figure 2B, Western blot further confirmed TNF-α and IFN-γ higher expression after STAT3 down-regulation.

    After STAT3 was downregulated to confirm the increase of TNF-α and INF-γ to expression, the culture cloned TIL was further studied CTL function assay under TIL for effector: target ratio by 1:25 and 1:50 at culture from day-12 to day-15 as our previous report [25],[42],[49],[50]. The results of CTL killing activity showed about 31% (1:25) and 59% (1:50) at 200 nm siRNA STAT3 in the transfected TILs group to compare the scrambled group (about 25%) at 200 nm shown in Figure 2C. Obviously, cytotoxic T lymphocyte of knockdown STAT3 TIL was (significantly increased on 1:50, P = 0.0023) much higher than scrambled groups.

    To study spatial-timely quantitative network from the complex proteins and STAT3 downregulation for reactivating IL-2 induced TILs, in silico experiment is SMAD-STAT3 complex regulation from TGF-β/SMAD signaling for TCR signaling and IL-2 reactivated to kill tumor cells. After the spatial-timely quantitative network set up with SMAD combination-regulation ratio (0%, 25%, 50%, 75%, and 100%), the results demonstrated SMAD-STAT3 complex regulation ratio (0%, 25%, 50%, 75%, and 100%). Eventually, we can harvest maximal spatial-timely combination-regulation with the activity of SMAD-STAT3 complex including 15,625 combination regulation as Table S1, including maximal reactivity values (spatial-timely order) for SMAD-STAT3 complex, so that results in silico can discover maximal regulation from a TGF-β/SMAD-STAT3 to regulating CTL functions. IL-2-inducing CTL genes are shown in Figure 3A. Additionally, we can achieve combination regulation activity for SMAD-STAT3 complex regulation, as shown in Figure 3B. The results demonstrated that IL-2 is completely blocked under STAT3 higher expression as Figure 3C2. Further, during in silico experiments, as demonstrated in Figure 3D, STAT3 is blocked to 70% CTL function to increase relating with IL2 inducing will show liner increase under the maximal spatial-timely combination-regulation Figure 3D2.

    Figure 3.  In silico support Smad-STAT3 complex to CTL. A) Shown is a CTL pathway; B) STAT3 in CTL pathways; C) Smad-STAT3 complex regulation, as Figure 3C2; D) STAT3 is blocked, CTL function to positively related with IL2 inducing IL2 inducing will show liner increase under the maximal spatial-timely combination-regulation. Figure 3D2.

    Tumor-infiltrating lymphocytes are a group of heterogeneous T-cells that have relevance in administrating TIL adoptive cell therapy (ACT) [51] and studying immune characteristics regarding T-cell, B-cell, NK cells, macrophage, and neutrophil within the tumor tissues [5],[7],[52],[53]. In order to study efficacy in treating solid tumor diseases, we began to study TIL genomic profiles from single-cell mRNA differential display from TIL published in 2007 [31] to single-cell RNA-seq from TIL published in 2013 [54], establishing a TIL quantitative network published in 2015 [55] and studying machine learning algorithms by artificial intelligence (AI) from TIL published in 2022 [56].

    Because the SMAD-STAT3 complex is focused on immune tolerance and T-cell quiescence through multiple mechanisms, as an oncogene, STAT3 was first described by Takeda et al. in 1999 as a negative regulator of Th1 immune responses [57], and other studies further support the role of STAT3 in immune evasion of human cancers [58]. The evidence demonstrated that multiple oncogenic signaling pathways regulate STAT3 expression in cancer cells, one of those from JAK/STAT3 [59] while the SMAD-STAT3 axis controls immune effects in the TGF-β pathway [60]. The TGF-β pathway has two effects same “double-edged sword” as “immune stimulation and inhibition”. Here, we report that STAT3 signaling attenuates immune responses through direct SMAD3-STAT3 interaction. Activated STAT3 suppresses the immune response with SMAD3, causing STAT3 and SMAD3 to block some immune responses. The evidence demonstrated that STAT3 activation suppresses the expression of CTL functions, such as IFN-γ, which are required for innate and adaptive antitumor immunity [61]. These findings suggest that STAT3 blockade could be used in cancer immunotherapy with increasing CTL functions.

    According to these investigations, the spatial-timely quantitative network can mimicking complexes binding to complexes regulation because three pathways are majorly involved in T-cell immune CTL to tumor cells: CD8+ TCR signal from TCR antigen associated with MHC class I; T-cells reactivated with IL-2 to induce TNF-α, INF-γ, GZMB, and PRF1 and TGF-β pathway for quiescent TIL-cell in tumor tissues. After we found that STAT3 has a binding site in promoters in CTL genes such as TNF-α, INF-γ, GZMB, and PRF1 while SMAD-STAT3 complex also involves the four promoters related to TGF-β/SMAD signaling-dependent activation of the TGF-β pathway, STAT3 down-regulation can be used to study CTL function and spatial-timely pathway. STAT3 knockdown results demonstrated TNF-α and IFN-γ higher expression than our previous results from traditional pathway configuration [62][65]. Moreover, Western blot not only supported mRNA results but also CTL increase after knockdown STAT3. The experiment data and in silico data support the spatial-timely quantitative pathway related to the TIL function. Now we can use the algorism to activate and inhibit the genes by targeting in vivo for combination therapy or ex vivo for analysis of other immune cells [66].

    CTL can be blocked by the immunosuppressive TME within tumors. STAT3 is key to the TGF-β pathway, resulting in immune suppressive action. In order to study STAT3 regulation, we study the SMAD-STAT3 complex within a spatial-order regulation pathway. Therefore, we use siRNA knockdown STAT3 to study the SMAD-STAT3 complex regulating CTL functions. We discovered the complex configuration of SMAD-STAT3 complex downregulation that induces CTL functions. The research evidence supported spatial-order quantification networks with more power than traditional pathways and confirmed that STAT3 down-regulation increases TIL activity. After studying the SMAD-STAT3 complex downregulation in the spatial order quantitative network, we can use the spatial-complex regulation algorithm to perform a machine learning algorithm for “big-data” analysis for personalized therapy.

    The authors declare they have not used Artificial Intelligence (AI) tools in the creation of this article.


    Acknowledgments



    Under the support of Dr. H. D. Preisler, we have set up a method to analyze genomic profiles of CD3, CD4, and CD8 from TIL. This work was supported by the National Cancer Institute IRG-91-022-09, USA (to BL). Mentions of trade names or commercial products in this article are solely for the purpose of providing specific information and do not imply recommendation.

    Conflict of interest



    Biaoru Li is an editorial board member for AIMS Allergy and Immunology and was not involved in the editorial review or the decision to publish this article. All authors declare that there are no competing interests.

    Author contributions



    YY (1) all papers with references check, VX for ChIP with statistics support and SYC rtPCR and CTL with statistics support. WQL and JD perform data, bioinformatics and biostatistics, YY (5) teaches YY (1) rtPCR and Western blot and VX performs rtPCR and Western blot under BL. BL conceived and designed the experiments.

    [1] Li BR, Tong SQ, Zhang XH, et al. (1994) A new experimental and clinical approach of combining usage of highly active tumor-infiltrating lymphocytes and highly sensitive antitumor drugs for the advanced malignant tumor. Chin Med J (Engl) 107: 803-807.
    [2] Li B, Tong S, Zhang XH, et al. (2020) Chapter-9: Development of adoptive T-cell immunotherapy-Future of personalized immunotherapy. Personalized immunotherapy for tumor diseases and beyond . UAE: Bentham Science Publishers 137-154. https://doi.org/10.2174/9789811482755120010012
    [3] Hu BC, Li GW, Cheng W, et al. (1997) Clinical application of infiltrating lymphocytes in malignant brain tumors. J Immunol (Chin) 2: 1-2.
    [4] Hua ZD, Lu J, Li HF, et al. (1996) Clinical study of tumor infiltrating lymphocytes in ovarian cancer. Chin J Obstet Gynecol 31: 55-57.
    [5] Lu J, Hu LLW, Hua ZD, et al. (1996) Analysis of the therapeutic effects of different therapeutic approaches for TIL. Chin J Cancer Biother 3: 127-129.
    [6] Cai XM, Lu J, Hua ZD, et al. (1996) Clinical application of TIL from different sources. J Immunol (Chin) 12: 251-254.
    [7] Li B, Ding J, Larson A, et al. (1999) Tumor tissue recycling-a new combination treatment for solid tumors: Experimental and preliminarily clinical research. In Vivo 13: 433-438.
    [8] Chen HY, Huang N, Tian HW, et al. (2021) Splenectomy provides protective effects against CLP-induced sepsis by reducing TRegs and PD-1/PD-L1 expression. Int J Biochem Cell Biolo 136: 105970. https://doi.org/10.1016/j.biocel.2021.105970
    [9] Wu L, Mao L, Liu JF, et al. (2019) Blockade of TIGIT/CD155 signaling reverses T-cell exhaustion and enhances antitumor capability in head and neck squamous cell carcinoma. Cancer Immunol Res 7: 1700-1713. https://doi.org/10.1158/2326-6066.CIR-18-0725
    [10] Labiano S, Palazon A, Melero I (2015) Immune response regulation in the tumor microenvironment by hypoxia. Semin Oncol 42: 378-386. https://doi.org/10.1053/j.seminoncol.2015.02.009
    [11] Chevolet I, Speeckaert R, Schreuer M, et al. (2015) Clinical significance of plasmacytoid dendritic cells and myeloid-derived suppressor cells in melanoma. J Transl Med 13: 1-9. https://doi.org/10.1186/s12967-014-0376-x
    [12] Minnema-Luiting J, Vroman H, Aerts J, et al. (2018) Heterogeneity in immune cell content in malignant pleural mesothelioma. Int J Mol Sci 19: 1041. https://doi.org/10.3390/ijms19041041
    [13] Oweida AJ, Darragh L, Phan A, et al. (2019) STAT3 modulation of regulatory T cells in response to radiation therapy in head and neck cancer. J Natl Cancer Inst 111: 1339-1349. https://doi.org/10.1093/jnci/djz036
    [14] Dong S, Guo XG, Han F, et al. (2022) Emerging role of natural products in cancer immunotherapy. Acta Pharm Sin B 12: 1163-1185. https://doi.org/10.1016/j.apsb.2021.08.020
    [15] Liao F, Liu L, Luo E, et al. (2018) Curcumin enhances anti-tumor immune response in tongue squamous cell carcinoma. Arch Oral Biol 92: 32-37. https://doi.org/10.1016/j.archoralbio.2018.04.015
    [16] Zhai NC, Li HJ, Song HX, et al. (2017) Hepatitis C virus induces MDSCs-like monocytes through TLR2/PI3K/AKT/STAT3 signaling. PLoS One 12: e0170516. https://doi.org/10.1371/journal.pone.0170516
    [17] Huang LL, Zhao Y, Shan MY, et al. (2023) Targeting crosstalk of STAT3 between tumor-associated M2 macrophages and Tregs in colorectal cancer. Cancer Biol Ther 24: 2226418. https://doi.org/10.1080/15384047.2023.2226418
    [18] Xiong WJ, Chen YH, Zhang CT, et al. (2023) Pharmacologic inhibition of IL11/STAT3 signaling increases MHC-I expression and T cell infiltration. J Transl Med 21: 416. https://doi.org/10.1186/s12967-023-04079-6
    [19] Yu L, Ji Kim H, Kim B, et al. (2023) Ethacrynic acid suppresses B7-H4 expression involved in epithelial-mesenchymal transition of lung adenocarcinoma cells via inhibiting STAT3 pathway. Biochem Pharmacol 212: 115537. https://doi.org/10.1016/j.bcp.2023.115537
    [20] Alahdal M, Xing Y, Tang TT, et al. (2018) 1-Methyl-D-tryptophan reduces tumor CD133+ cells, Wnt/β-catenin and NF-κβp65 while enhances lymphocytes NF-κβ2, STAT3, and STAT4 pathways in murine pancreatic adenocarcinoma. Sci Rep 8: 9869. https://doi.org/10.1038/s41598-018-28238-8
    [21] Han QJ, Wang YQ, Pang M, et al. (2017) STAT3-blocked whole-cell hepatoma vaccine induces cellular and humoral immune response against HCC. J Exp Clin Cancer Res 36: 1-11. https://doi.org/10.1186/s13046-017-0623-0
    [22] Li M, Shang DS, Zhao WD, et al. (2009) Amyloid β interaction with receptor for advanced glycation end products up-regulates brain endothelial CCR5 expression and promotes T cells crossing the blood-brain barrier. J Immunol 182: 5778-5788. https://doi.org/10.4049/jimmunol.0803013
    [23] Li B (2008) A strategy to identify genomic expression profiles at single-T-cell level and a small number of cells (review paper). J Biotechnol 7: 71-82. http://dx.doi.org/10.2225/vol8-issue1-fulltext-3
    [24] Li B, Perabekam S, Liu G, et al. (2002) Experimental and bioinformatics comparison of gene expression between T cells from TIL of liver cancer and T cells from UniGene. J Gastroenterol 37: 275-282. https://doi.org/10.1007/s005350200035
    [25] Li B, Chang TF, Larson A, et al. (2000) Identification of mRNAs expressed in tumor-infiltrating lymphocytes by a strategy for rapid and high throughput screening. Gene 255: 273-279. https://doi.org/10.1016/S0378-1119(00)00330-9
    [26] Li B (2015) Breakthrough of 2015-Personalized immunotherapy based on individual GWAS and biomarkers. Biomark J 1: 1-2.
    [27] Zhang W, Yan Q, Lin MH, et al. (2020) Immune cells signaling-personalized immunotherapy for tumor diseases and beyond. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 20-42. https://doi.org/10.2174/9789811482755120010005
    [28] Li B, Zhu XG, Ward CM, et al. (2019) MIR-144-mediated NRF2 gene silencing inhibits fetal hemoglobin expression in sickle cell disease. Exp Hematol 70: 85-96.e5. https://doi.org/10.1016/j.exphem.2018.11.002
    [29] Ren YX, Yang J, Sun RM, et al. (2016) Viral IL-10 down-regulates the “MHC-I antigen processing operon” through the NF-κB signaling pathway in nasopharyngeal carcinoma cells. Cytotechnology 68: 2625-2636. https://doi.org/10.1007/s10616-016-9987-9
    [30] Li Z, Abraham BJ, Berezovskaya A, et al. (2017) APOBEC signature mutation generates an oncogenic enhancer that drives LMO1 expression in T-ALL. Leukemia 31: 2057-2064. https://doi.org/10.1038/leu.2017.75
    [31] Zhang W, Ding JQ, Qu Y, et al. (2009) Genomic expression analysis of quiescent CD8 T-cells from tumor-infiltrating lymphocytes of in vivo liver tumor by single-cell mRNA differential display. Immunology 127: 83-90. https://doi.org/10.1111/j.1365-2567.2008.02926.x
    [32] Li B, Hu HL, Ding J, et al. (2015) Functional cell-proliferation and differentiation by system modeling for cell therapy. Int J Latest Res Sci Technol 4: 180-187.
    [33] Li B, Liu G, Hu HL, et al. (2015) Biomarkers analysis for heterogeneous immune responses of quiescent CD8+ cells-a clue for personalized immunotherapy. Biomark J 1: 1-12.
    [34] Min Tao, Li B, Nayini J, et al. (2000) SCF, IL-1 beta, IL-1ra and GM-CSF in the bone marrow and serum of normal individuals and of AML CML patients. Cytokine 12: 699-707. https://doi.org/10.1006/cyto.2000.0666
    [35] Preisler HD, Li B, Yang BL, et al. (2000) Suppression of telomerase activity and cytokine messenger RNA levels in acute myelogenous leukemia cells in vivo in patients by amifostine and interleukin 4. Clin Cancer Res 6: 807-812.
    [36] Mundle SD, Mativi BY, Cartlidge JD, et al. (2000) Signal antonymy unique to myelodysplastic marrows correlates with altered expression of E2F1. Br J Haematol 109: 376-381. https://doi.org/10.1046/j.1365-2141.2000.01937.x
    [37] Yang J, Li BR, Nayini J, et al. (1999) Tyrosine phosphorylation of Shc proteins in normal CD34+ progenitor cells and leukemia cells. Blood 94: 373-374. https://doi.org/10.1182/blood.V94.1.373.413a48b_373_374
    [38] Preisler HD, Gao XZ, Ming T, et al. (1999) Marrow cytokine transcripts and the secondary hematologic disorders. Leuk Lymphoma 35: 297-302. https://doi.org/10.3109/10428199909145733
    [39] Perambakam S, Li B, Preisler H (2001) Quantitation of interferon regulatory factor transcripts in patients with AML. Cancer Invest 19: 346-351.
    [40] Li B (2008) A strategy to identify genomic expression profiles at single-T-cell level and a small number of cells. Electron J Biotechnol 8. http://dx.doi.org/10.2225/vol8-issue1-fulltext-3
    [41] Li B, Zhu YM, Hu BY, et al. (1994) Establishment of a method for separation of tumor infiltrating lymphocytes with high vitality. J Immunol (Chin) 10: 44-47.
    [42] Li S, Perabekam S, Devemy E, et al. (2020) Genetically modified T-cells affinity to tumor cells-development of adoptive T-cell immunotherapy. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 174-149. https://doi.org/10.2174/9789811482755120010014
    [43] Li B, Xu YB (2022) Single-cell whole RNA sequencing from tumor-infiltrating lymphocyte. New Horizons in Medicine and Medical Research : 167-173. https://doi.org/10.9734/bpi/nhmmr/v10/6129F
    [44] Liu G, Zheng J, Li B (2020) Bioinformatics of T-cell and primary tumor cells. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 118-136. https://doi.org/10.2174/9789811482755120010011
    [45] Wang ZH, Hu HL, Zheng J, et al. (2011) Gene expression and pathway analysis of quiescent CD8+ T cells from liver cancer, liver sinusoid and peripheral blood-study on toxicogenomic and prevention targeting. “2011 IEEE 11th International Conference on Bioinformatics and Bioengineering” : 72-77. https://doi.org/10.1109/BIBE.2011.18
    [46] Li B, Liu G, Zheng J (2020) System modeling of T-cell function development of adoptive T-cell immunotherapy. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 197-223. https://doi.org/10.2174/9789811482755120010015
    [47] Tan SZ, Zhu X (2020) Molecular targeting checkpoint in cancer. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 70-89. https://doi.org/10.2174/9789811482755120010008
    [48] Hu HL Zhang QH, Li S, et al. (2015) A therapeutic targeting identification from microarray data and quantitative network analysis. Open Access J Sci Technol 3. https://doi.org/10.11131/2015/101114
    [49] Xiao HS, Gao Y, Li S, et al. (2016) A case report of personalized chemotherapy for metastatic cardiac sarcoma. J Clin Epigenet 1: 1-6.
    [50] Lu J, Cheng SJ, Li B (2007) Pathway-based approaches for analysis of genome-wide association studies-a case report for metastatic small cell lung cancer. Int J Hematol Ther 10: 2381-1404.
    [51] Li B, Larson A, Li S (2020) Personalized Immunotherapy for Tumor Diseases and Beyond. UAE: Bentham Science Publishers. https://doi.org/10.2174/97898114827551200101
    [52] Ding JQ, Qian GX, Li B, et al. (1995) A preliminary study of tumor necrosis factor gene transduction of tumor infiltrating lymphocytes Application. Chin J Cancer Biother 1: 11-15.
    [53] Jing LH, Li B (2020) Immunoassay of personalized immunotherapy. Personalized Immunotherapy for Tumor Diseases and Beyond . UAE: Bentham Science Publishers 43-56. https://doi.org/10.2174/9789811482755120010006
    [54] Xu YB, Hu HL, Zheng J, et al. (2013) Feasibility of whole RNA sequencing from single-cell mRNA amplification. Genet Res Int . https://doi.org/10.1155/2013/724124
    [55] Li B, Hu HL, Ding JQ, et al. (2015) Functional cell-proliferation and differentiation by system modelling for cell therapy. Int J Latest Res Sci Technol 4: 180-187. Available from: https://www.mnkjournals.com/journal/ijlrst/index.php
    [56] Ying XN, Li B (2022) Machine-learning modeling for personalized immunotherapy-an evaluation module. Biomed J Scie Tech Res 47: 38211. https://doi.org/10.26717/BJSTR.2022.47.007462
    [57] Takeda K, Clausen BE, Kaisho T, et al. (1999) Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity 10: 39-49. https://doi.org/10.1016/S1074-7613(00)80005-9
    [58] Goswami K, Barik S, Sarkar M, et al. (2014) Targeting STAT3 phosphorylation by neem leaf glycoprotein prevents immune evasion exerted by supraglottic laryngeal tumor induced M2 macrophages. Mol Immunol 59: 119-127. https://doi.org/10.1016/j.molimm.2014.01.015
    [59] Üremiş MM, Üremis N, Türköz Y (2023) Cucurbitacin E shows synergistic effect with sorafenib by inducing apoptosis in hepatocellular carcinoma cells and regulates Jak/Stat3, ERK/MAPK, PI3K/Akt/mTOR signaling pathways. Steroids 198: 109261. https://doi.org/10.1016/j.steroids.2023.109261
    [60] Wang G, Yu Y, Sun C, et al. (2016) STAT3 selectively interacts with Smad3 to antagonize TGF-β signaling. Oncogene 35: 4388-4398. https://doi.org/10.1038/onc.2015.446
    [61] Németh Á, Mózes MM, Calvier L, et al. (2019) The PPARγ agonist pioglitazone prevents TGF-β induced renal fibrosis by repressing EGR-1 and STAT3. BMC Nephrol 20. https://doi.org/10.1186/s12882-019-1431-x
    [62] Sun ZJ, Lin DD, Shen Y, et al. (2023) Critical role of MXRA7 in differentiation blockade in human acute promyelocytic leukemia cells. Exp Hematol 125: 45-54. https://doi.org/10.1016/j.exphem.2023.07.001
    [63] Zhu YZ, Wang XH, He ZW, et al. (2023) Enterovirus 71 enters human brain microvascular endothelial cells through an ARF6-mediated endocytic pathway. J Med Virol 95: e28915. https://doi.org/10.1002/jmv.28915
    [64] Kongkaew T, Thaiwong R, Tudsamran S, et al. (2022) TIL expansion with high dose IL-2 or low dose IL-2 with anti-CD3/anti-CD28 stimulation provides different quality of TIL-expanded T cell clones. J Immunol Methods 503: 113229. https://doi.org/10.1016/j.jim.2022.113229
    [65] Knochelmann HM, Rivera-Reyes AM, Wyatt MM, et al. (2021) Modeling ex vivo tumor-infiltrating lymphocyte expansion from established solid malignancies. Oncoimmunology 10: 1959101. https://doi.org/10.1080/2162402X.2021.1959101
    [66] Li B (2022) Why will TIL produce different efficacy to treat solid tumor. Front Immunol 2: 1-13. https://doi.org/10.3389/fimmu.2022.973881
  • allergy-08-04-017-s001.xlsx
  • Reader Comments
  • © 2024 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(856) PDF downloads(28) Cited by(0)

Figures and Tables

Figures(3)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog