Review

Combinatorial application of artificial intelligence and CRISPR/Cas9 on the next-generation CAR-T immunotherapy

  • Published: 05 September 2025
  • Chimeric Antigen Receptor T-cell (CAR-T) therapy has emerged as one effective treatment against complex diseases, including malignancies, infectious disorders, and autoimmune conditions. However, its clinical applications remain hindered by severe side effects, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Recent innovations in genome editing and computational biology, specifically CRISPR/Cas9 and artificial intelligence (AI), offer promising avenues to improve CAR-T safety and efficacy. In this article, we reviewed recent advances in CAR-T therapy, highlighting the key role CRISPR/Cas9 plays in various aspects of CAR-T therapy through gene editing. We then studied and analyzed how AI leverages large datasets and its powerful learning capabilities to advance CAR-T therapies and CRISPR/Cas9. Finally, focusing on CAR-T therapy, we discussed how CRISPR/Cas9 and AI work synergistically to advance the development of CAR-T therapy, including reducing toxic side effects, improving therapeutic efficacy, sustaining long-term outcomes, discovering new therapeutic targets, and ensuring safety monitoring. By combining the predictive power of AI with the precision of CRISPR/Cas9, researchers can develop next-generation immunotherapies that are safe, effective, and tailored to the patients' requirements. This synergy can address previously untreatable diseases and reshape the landscape of precision medicine.

    Citation: Jiesheng Cui, Dini Zhang, Guanyu Wang. Combinatorial application of artificial intelligence and CRISPR/Cas9 on the next-generation CAR-T immunotherapy[J]. AIMS Molecular Science, 2025, 12(3): 292-317. doi: 10.3934/molsci.2025018

    Related Papers:

  • Chimeric Antigen Receptor T-cell (CAR-T) therapy has emerged as one effective treatment against complex diseases, including malignancies, infectious disorders, and autoimmune conditions. However, its clinical applications remain hindered by severe side effects, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Recent innovations in genome editing and computational biology, specifically CRISPR/Cas9 and artificial intelligence (AI), offer promising avenues to improve CAR-T safety and efficacy. In this article, we reviewed recent advances in CAR-T therapy, highlighting the key role CRISPR/Cas9 plays in various aspects of CAR-T therapy through gene editing. We then studied and analyzed how AI leverages large datasets and its powerful learning capabilities to advance CAR-T therapies and CRISPR/Cas9. Finally, focusing on CAR-T therapy, we discussed how CRISPR/Cas9 and AI work synergistically to advance the development of CAR-T therapy, including reducing toxic side effects, improving therapeutic efficacy, sustaining long-term outcomes, discovering new therapeutic targets, and ensuring safety monitoring. By combining the predictive power of AI with the precision of CRISPR/Cas9, researchers can develop next-generation immunotherapies that are safe, effective, and tailored to the patients' requirements. This synergy can address previously untreatable diseases and reshape the landscape of precision medicine.



    加载中

    Acknowledgments



    This work was partly supported by Natural Science Foundation of Shenzhen (JCYJ20240813113606009), National Natural Science Foundation of China (32070681), National Key R&D Program of China (2019YFA0906002), the Shenzhen-Hong Kong Cooperation Zone for Technology and Innovation (HZQB-KCZYB-2020056).

    Conflict of interest



    The authors declare no conflict of interest in this paper.

    [1] Zheng PP, Kros JM, Wang G (2019) Elusive neurotoxicity in T cell-boosting anticancer therapies. Trends Immunol 40: 274-278. https://doi.org/10.1016/j.it.2019.02.005
    [2] Zheng PP, Kros JM, Li J (2018) Approved CAR T cell therapies: Ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today 23: 1175-1182. https://doi.org/10.1016/j.drudis.2018.02.012
    [3] Awasthi R, Maier HJ, Zhang J, et al. (2023) Kymriah®(tisagenlecleucel)–An overview of the clinical development journey of the first approved CAR-T therapy. Hum Vaccines 19: 2210046. https://doi.org/10.1080/21645515.2023.2210046
    [4] Westin JR, Oluwole OO, Kersten MJ, et al. (2023) Survival with axicabtagene ciloleucel in large B-cell lymphoma. N Engl J Med 389: 148-157. https://doi.org/10.1056/NEJMoa2301665
    [5] Belin C, Devic P, Ayrignac X, et al. (2020) Description of neurotoxicity in a series of patients treated with CAR T-cell therapy. Sci Rep 10: 18997. https://doi.org/10.1038/s41598-020-76055-9
    [6] Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. (2018) Cytokine release syndrome. J Immunother Cancer 6: 56. https://doi.org/10.1186/s40425-018-0343-9
    [7] Morris EC, Neelapu SS, Giavridis T, et al. (2022) Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol 22: 85-96. https://doi.org/10.1038/s41577-021-00547-6
    [8] Lee DW, Gardner R, Porter DL, et al. (2014) Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124: 188-195. https://doi.org/10.1182/blood-2014-05-552729
    [9] Kroschinsky F, Stölzel F, Von Bonin S, et al. (2017) New drugs, new toxicities: Severe side effects of modern targeted and immunotherapy of cancer and their management. Crit Care 21: 89. https://doi.org/10.1186/s13054-017-1678-1
    [10] Boretti A (2024) The transformative potential of AI-driven CRISPR-Cas9 genome editing to enhance CAR T-cell therapy. Comput Biol Med 182: 109137. https://doi.org/10.1016/j.compbiomed.2024.109137
    [11] Hort S, Herbst L, Bäckel N, et al. (2022) Toward rapid, widely available autologous CAR-T cell therapy–Artificial intelligence and automation enabling the smart manufacturing hospital. Front Med 9: 913287. https://doi.org/10.3389/fmed.2022.913287
    [12] Joy R, Phair K, O'Hara R, et al. (2024) Recent advances and current challenges in CAR-T cell therapy. Biotechnol Lett 46: 115-126. https://doi.org/10.1007/s10529-023-03461-0
    [13] Krueger GRF, Brandt ME, Wang G (2002) A computational analysis of Canale-Smith syndrome: Chronic lymphadenopathy simulating malignant lymphoma. Anticancer Res 22: 2365-2371.
    [14] Wang G, Krueger GRF (2010) Computational analysis of mTOR signaling pathway: Bifurcation, carcinogenesis, and drug discovery. Anticancer Res 30: 2683-2688.
    [15] Wang G (2010) Singularity analysis of the AKT signaling pathway reveals connections between cancer and metabolic diseases. Phys Biol 7: 046015. https://doi.org/10.1088/1478-3975/7/4/046015
    [16] Li T, Wang G (2014) Computer-aided targeting of the PI3K/Akt/mTOR pathway: Toxicity reduction and therapeutic opportunities. Int J Mol Sci 15: 18856-18891. https://doi.org/10.3390/ijms151018856
    [17] Sun L, Li X, Pan J, et al. (2019) Seeking mTORC1 inhibitors through molecular dynamics simulation of arginine analogs inhibiting CASTOR1. Cancer Genom Proteom 16: 465-479. https://doi.org/10.21873/cgp.20150
    [18] Xing F, Liu YC, Huang S, et al. (2021) Accelerating precision anti-cancer therapy by time-lapse and label-free 3D tumor slice culture platform. Theranostics 11: 9415-9430. https://doi.org/10.7150/thno.59533
    [19] June CH, Sadelain M (2018) Chimeric antigen receptor therapy. N Engl J Med 379: 64-73. https://doi.org/10.1056/NEJMra1706169
    [20] Yu T, Jiao JH, Wu MF (2025) CAR-T cells in the treatment of multiple myeloma: An encouraging cell therapy. Front Immunol 16: 1499590. https://doi.org/10.3389/fimmu.2025.1499590
    [21] Wang L, Guo M, Hou S (2025) Advances in primary large B-cell lymphoma of immune-privileged sites. Front Immunol 16: 1533444. https://doi.org/10.3389/fimmu.2025.1533444
    [22] Csizmar CM, Litzow MR, Saliba AN (2025) Antibody-based and other novel agents in adult B-cell acute lymphoblastic leukemia. Cancers 17: 779. https://doi.org/10.3390/cancers17050779
    [23] Brentjens RJ, Davila ML, Riviere I, et al. (2013) CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5: 177ra38. https://doi.org/10.1126/scitranslmed.3005930
    [24] Grupp SA, Kalos M, Barrett D, et al. (2013) Chimeric antigen receptor–modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509-1518. https://doi.org/10.1056/NEJMoa1215134
    [25] Scheuermann RH, Racila E (1995) CD19 antigen in leukemia and lymphoma diagnosis and Immunotherapy. Leukemia Lymphoma 18: 385-397. https://doi.org/10.3109/10428199509059636
    [26] Porter DL, Levine BL, Kalos M, et al. (2011) Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. N Engl J Med 365: 725-733. https://doi.org/10.1056/NEJMoa1103849
    [27] Sadelain M (2017) CD19 CAR T cells. Cell 171: 1471. https://doi.org/10.1016/j.cell.2017.12.002
    [28] Chung JB, Brudno JN, Borie D, et al. (2024) Chimeric antigen receptor T cell therapy for autoimmune disease. Nat Rev Immunol 24: 830-845. https://doi.org/10.1038/s41577-024-01035-3
    [29] Lyu X, Gupta L, Tholouli E, et al. (2024) Chimeric antigen receptor T cell therapy: A new emerging landscape in autoimmune rheumatic diseases. Rheumatology 63: 1206-1216. https://doi.org/10.1093/rheumatology/kead616
    [30] Chen Y, Liu C, Fang Y, et al. (2024) Developing CAR-immune cell therapy against SARS-CoV-2: Current status, challenges and prospects. Biochem Pharmacol 222: 116066. https://doi.org/10.1016/j.bcp.2024.116066
    [31] Guo X, Kazanova A, Thurmond S, et al. (2021) Effective chimeric antigen receptor T cells against SARS-CoV-2. iScience 24: 103295. https://doi.org/10.1016/j.isci.2021.103295
    [32] Singh MS, Arun PPS, Ansari MA (2024) Unveiling common markers in COVID-19: ADAMTS2, PCSK9, and OLAH emerged as key differential gene expression profiles in PBMCs across diverse disease conditions. AIMS Mol Sci 11: 189-205. https://doi.org/10.3934/molsci.2024011
    [33] Shukor MSA, Shukor MYA (2023) Molecular docking and dynamics studies show: Phytochemicals from Papaya leaves extracts as potential inhibitors of SARS–CoV–2 proteins targets and TNF–alpha and alpha thrombin human targets for combating COVID-19. AIMS Mol Sci 10: 213-262. https://doi.org/10.3934/molsci.2023015
    [34] Navale AM, Devangan V, Goswami A, et al. (2023) Effects of pre-existing metformin therapy on platelet count, serum creatinine, and hospitalization in COVID-19 patients with diabetes mellitus. AIMS Mol Sci 10: 311-321. https://doi.org/10.3934/molsci.2023018
    [35] Sharma B, Angel B, Naik VU, et al. (2023) Repurposed drug molecules targeting NSP12 protein of SARS-CoV-2: An in-silico study. AIMS Mol Sci 10: 322-342. https://doi.org/10.3934/molsci.2023019
    [36] Thirumalaisamy R, Vasuki S, Sindhu SM, et al. (2025) FDA-approved Cchimeric Antigen Receptor (CAR)-T cell therapy for different cancers-A recent perspective. Mol Biotechnol 67: 469-483. https://doi.org/10.1007/s12033-024-01090-0
    [37] Von Kalle C, Deichmann A, Schmidt M (2014) Vector integration and tumorigenesis. Human Gene Ther 25: 475-481. https://doi.org/10.1089/hum.2014.2525
    [38] Jensen TI, Axelgaard E, Bak RO (2019) Therapeutic gene editing in haematological disorders with CRISPR/Cas9. Brit J Haematol 185: 821-835. https://doi.org/10.1111/bjh.15851
    [39] Chandran SS, Klebanoff CA (2019) T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance. Immunol Rev 290: 127-147. https://doi.org/10.1111/imr.12772
    [40] Pulé M, Mekkaoui L, Amrolia P, et al. (2019) Chimeric antigen receptor (CAR) comprising a CD19-binding domain. Google Patents .
    [41] Hartmann J, Schüßler-Lenz M, Bondanza A, et al. (2017) Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med 9: 1183-1197. https://doi.org/10.15252/emmm.201607485
    [42] Dagar G, Gupta A, Masoodi T, et al. (2023) Harnessing the potential of CAR-T cell therapy: Progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med 21: 449. https://doi.org/10.1186/s12967-023-04292-3
    [43] Romeo C, Seed B (1991) Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides. Cell 64: 1037-1046. https://doi.org/10.1016/0092-8674(91)90327-U
    [44] Irving BA, Weiss A (1991) The cytoplasmic domain of the T cell receptor ζ chain is sufficient to couple to receptor-associated signal transduction pathways. Cell 64: 891-901. https://doi.org/10.1016/0092-8674(91)90314-O
    [45] Harding FA, McArthur JG, Gross JA, et al. (1992) CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature 356: 607-609. https://doi.org/10.1038/356607a0
    [46] Imai C, Mihara K, Andreansky M, et al. (2004) Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18: 676-684. https://doi.org/10.1038/sj.leu.2403302
    [47] Ramos CA, Rouce R, Robertson CS, et al. (2018) In vivo fate and activity of second- versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkin's lymphomas. Mol Ther 26: 2727-2737. https://doi.org/10.1016/j.ymthe.2018.09.009
    [48] Chmielewski M, Abken H (2015) TRUCKs: The fourth generation of CARs. Expert Opin Biol Ther 15: 1145-1154. https://doi.org/10.1517/14712598.2015.1046430
    [49] Chmielewski M, Hombach AA, Abken H (2014) Of CARs and TRUCKs: Chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol Rev 257: 83-90. https://doi.org/10.1111/imr.12125
    [50] Kagoya Y, Tanaka S, Guo T, et al. (2018) A novel chimeric antigen receptor containing a JAK–STAT signaling domain mediates superior antitumor effects. Nat Med 24: 352-359. https://doi.org/10.1038/nm.4478
    [51] Schuster SJ, Bishop MR, Tam CS, et al. (2019) Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 380: 45-56. https://doi.org/10.1056/NEJMoa1804980
    [52] Neelapu SS, Tummala S, Kebriaei P, et al. (2018) Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat Rev Clin Oncol 15: 47-62. https://doi.org/10.1038/nrclinonc.2017.148
    [53] Santomasso BD, Park JH, Salloum D, et al. (2018) Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov 8: 958-971. https://doi.org/10.1158/2159-8290.CD-17-1319
    [54] Jinek M, Chylinski K, Fonfara I, et al. (2012) A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science 337: 816-821. https://doi.org/10.1126/science.1225829
    [55] Cong L, Ran FA, Cox D, et al. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823. https://doi.org/10.1126/science.1231143
    [56] Mali P, Yang L, Esvelt KM, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823-826. https://doi.org/10.1126/science.1232033
    [57] Gasiunas G, Barrangou R, Horvath P, et al. (2012) Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci USA 109: E2579-E2586. https://doi.org/10.1073/pnas.1208507109
    [58] Doudna JA, Charpentier E (2014) The new frontier of genome engineering with CRISPR-Cas9. Science 346: 1258096. https://doi.org/10.1126/science.1258096
    [59] Akhtar J, Imran M, Wang G (2023) CRISPR/Cas9-mediated CtBP1 gene editing enhances chemosensitivity and inhibits metastatic potential in esophageal squamous cell carcinoma cells. Int J Mol Sci 24: 14030. https://doi.org/10.3390/ijms241814030
    [60] Sterner RM, Sakemura R, Cox MJ, et al. (2019) GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood 133: 697-709. https://doi.org/10.1182/blood-2018-10-881722
    [61] Nukala U, Rodriguez Messan M, Yogurtcu ON, et al. (2021) A systematic review of the efforts and hindrances of modeling and simulation of CAR T-cell therapy. AAPS J 23: 52. https://doi.org/10.1208/s12248-021-00579-9
    [62] Akhtar J, Imran M, Wang G (2023) CRISPR/Cas9-mediated CtBP1 gene editing enhances chemosensitivity and inhibits metastatic potential in esophageal squamous cell carcinoma cells. Int J Mol Sci 24: 14030. https://doi.org/10.3390/ijms241814030
    [63] Razeghian E, Nasution MKM, Rahman HS, et al. (2021) A deep insight into CRISPR/Cas9 application in CAR-T cell-based tumor immunotherapies. Stem Cell Res Ther 12: 428. https://doi.org/10.1186/s13287-021-02510-7
    [64] Terrett J, Mcewan B, Hostetter D, et al. (2021) 133 CRISPR/Cas9 gene-edited allogeneic CAR-T cells targeting CD33 show high preclinical efficacy against AML without long-term hematopoietic toxicity. J ImmunoTher Cancer . https://doi.org/10.1136/jitc-2021-SITC2021.133
    [65] Eyquem J, Mansilla-Soto J, Giavridis T, et al. (2017) Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543: 113-117. https://doi.org/10.1038/nature21405
    [66] Meng H, Nan M, Li Y, et al. (2023) Application of CRISPR-Cas9 gene editing technology in basic research, diagnosis and treatment of colon cancer. Front Endocrinol 14: 1148412. https://doi.org/10.3389/fendo.2023.1148412
    [67] Ren J, Liu X, Fang C, et al. (2017) Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res 23: 2255-2266. https://doi.org/10.1158/1078-0432.CCR-16-1300
    [68] Huan M, Wang G (2024) Harnessing naturally occurring bistable switches for their application in synthetic biology. SynBio 2: 363-377. https://doi.org/10.3390/synbio2040023
    [69] Young E, Alper H (2010) Synthetic biology: Tools to design, build, and optimize cellular processes. Biomed Res J 1–12. https://doi.org/10.1155/2010/130781
    [70] Dimitri A, Herbst F, Fraietta JA (2022) Engineering the next-generation of CAR T-cells with CRISPR-Cas9 gene editing. Mol Cancer 21: 78. https://doi.org/10.1186/s12943-022-01559-z
    [71] Cho JH, Collins JJ, Wong WW (2018) Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173: 1426-1438.e11. https://doi.org/10.1016/j.cell.2018.03.038
    [72] Shalem O, Sanjana NE, Hartenian E, et al. (2014) Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343: 84-87. https://doi.org/10.1126/science.1247005
    [73] Liu D, Zhao X, Tang A, et al. (2020) CRISPR screen in mechanism and target discovery for cancer immunotherapy. BBA-Rev Cancer 1874: 188378. https://doi.org/10.1016/j.bbcan.2020.188378
    [74] Carnevale J, Shifrut E, Kale N, et al. (2022) RASA2 ablation in T cells boosts antigen sensitivity and long-term function. Nature 609: 174-182. https://doi.org/10.1038/s41586-022-05126-w
    [75] Freitas KA, Belk JA, Sotillo E, et al. (2022) Enhanced T cell effector activity by targeting the Mediator kinase module. Science 378: eabn5647. https://doi.org/10.1126/science.abn5647
    [76] Belk JA, Yao W, Ly N, et al. (2022) Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell 40: 768-786.E7. https://doi.org/10.1016/j.ccell.2022.06.001
    [77] Trefny MP, Kirchhammer N, Auf Der Maur P, et al. (2023) Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy. Nat Commun 14: 86. https://doi.org/10.1038/s41467-022-35583-w
    [78] Zhang X, Zhang C, Qiao M, et al. (2022) Depletion of BATF in CAR-T cells enhances antitumor activity by inducing resistance against exhaustion and formation of central memory cells. Cancer Cell 40: 1407-1422.E7. https://doi.org/10.1016/j.ccell.2022.09.013
    [79] Wang D, Prager BC, Gimple RC, et al. (2021) CRISPR screening of CAR T cells and cancer stem cells reveals critical dependencies for cell-based therapies. Cancer Discov 11: 1192-1211. https://doi.org/10.1158/2159-8290.CD-20-1243
    [80] Gurusamy D, Henning AN, Yamamoto TN, et al. (2020) Multi-phenotype CRISPR-Cas9 screen identifies p38 kinase as a target for adoptive immunotherapies. Cancer Cell 37: 818-833.E9. https://doi.org/10.1016/j.ccell.2020.05.004
    [81] Geiger R, Rieckmann JC, Wolf T, et al. (2016) L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167: 829-842.E13. https://doi.org/10.1016/j.cell.2016.09.031
    [82] Ye L, Park JJ, Peng L, et al. (2022) A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies proline metabolism as a means to enhance CAR-T therapy. Cell Metab 34: 595-614.E14. https://doi.org/10.1016/j.cmet.2022.02.009
    [83] Wu Y, Wang G (2018) Machine learning based toxicity prediction: From chemical structural description to transcriptome analysis. Int J Mol Sci 19: 2358. https://doi.org/10.3390/ijms19082358
    [84] Mao J, Akhtar J, Zhang X, et al. (2021) Comprehensive strategies of machine-learning-based quantitative structure-activity relationship models. iScience 24: 103052. https://doi.org/10.1016/j.isci.2021.103052
    [85] Guan S, Wang G (2024) Drug discovery and development in the era of artificial intelligence: From machine learning to large language models. Artif Intell Chem 2: 100070. https://doi.org/10.1016/j.aichem.2024.100070
    [86] Han Y, Akhtar J, Liu G, et al. (2023) Early warning and diagnosis of liver cancer based on dynamic network biomarker and deep learning. Compu Struct Biotech 21: 3478-3489. https://doi.org/10.1016/j.csbj.2023.07.002
    [87] Wu Y, Guan S, Wang G (2023) Drug effect deep learner based on graphical convolutional network. Machine learning and deep learning in computational toxicology . Cham: Springer 83-140. https://doi.org/10.1007/978-3-031-20730-3_4
    [88] Wang G (2016) Chemoinformatics in the new era: From molecular dynamics to systems dynamics. Molecules 21: 71. https://doi.org/10.3390/molecules21030071
    [89] Naghizadeh A, Tsao W, Hyun Cho J, et al. (2022) In vitro machine learning-based CAR T immunological synapse quality measurements correlate with patient clinical outcomes. PLoS Comput Biol 18: e1009883. https://doi.org/10.1371/journal.pcbi.1009883
    [90] Zhao Z, Wu J, Li T, et al. (2021) Challenges and opportunities of AI-enabled monitoring, diagnosis & prognosis: A review. Chin J Mech Eng 34: 56. https://doi.org/10.1186/s10033-021-00570-7
    [91] Daniels KG, Wang S, Simic MS, et al. (2022) Decoding CAR T cell phenotype using combinatorial signaling motif libraries and machine learning. Science 378: 1194-1200. https://doi.org/10.1126/science.abq0225
    [92] Zhang R, Han X, Lei Z, et al. (2022) RCMNet: A deep learning model assists CAR-T therapy for leukemia. Comput Biol Med 150: 106084. https://doi.org/10.1016/j.compbiomed.2022.106084
    [93] Qiu S, Chen J, Wu T, et al. (2024) CAR-Toner: An AI-driven approach for CAR tonic signaling prediction and optimization. Cell Res 34: 386-388. https://doi.org/10.1038/s41422-024-00936-1
    [94] Hartmann FJ, Babdor J, Gherardini PF, et al. (2019) Comprehensive immune monitoring of clinical trials to advance human immunotherapy. Cell Rep 28: 819-831.E4. https://doi.org/10.1016/j.celrep.2019.06.049
    [95] Rodriques SG, Stickels RR, Goeva A, et al. (2019) Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution. Science 363: 1463-1467. https://doi.org/10.1126/science.aaw1219
    [96] Lu T, Zhang Z, Zhu J, et al. (2021) Deep learning-based prediction of the T cell receptor–antigen binding specificity. Nat Mach Intell 3: 864-875. https://doi.org/10.1038/s42256-021-00383-2
    [97] Hayden PJ, Roddie C, Bader P, et al. (2022) Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 33: 259-275. https://doi.org/10.1016/j.annonc.2021.12.003
    [98] Dixit S, Kumar A, Srinivasan K, et al. (2024) Advancing genome editing with artificial intelligence: opportunities, challenges, and future directions. Front Bioeng Biotechnol 11: 1335901. https://doi.org/10.3389/fbioe.2023.1335901
    [99] Wang J, Zhang X, Cheng L, et al. (2020) An overview and metanalysis of machine and deep learning-based CRISPR gRNA design tools. RNA Biol 17: 13-22. https://doi.org/10.1080/15476286.2019.1669406
    [100] Kim GB, Kim WJ, Kim HU, et al. (2020) Machine learning applications in systems metabolic engineering. Curr Opin Biotechnol 64: 1-9. https://doi.org/10.1016/j.copbio.2019.08.010
    [101] Lin J, Wong KC (2018) Off-target predictions in CRISPR-Cas9 gene editing using deep learning. Bioinformatics 34: i656-i663. https://doi.org/10.1093/bioinformatics/bty554
    [102] Dimauro G, Colagrande P, Carlucci R, et al. (2019) CRISPRLearner: A deep learning-based system to predict CRISPR/Cas9 sgRNA on-target cleavage efficiency. Electronics 8: 1478. https://doi.org/10.3390/electronics8121478
    [103] Oh J, Hessel M, Czarnecki WM, et al. (2020) Discovering reinforcement learning algorithms. Adv Neural Inform Process Syst 33: 1060-1070.
    [104] Baidya S, Choudhury S, De RK (2023) A novel CRISPR-MultiTargeter Multi-agent Reinforcement learning (CMT-MARL) algorithm to identify editable target regions using a Hybrid scoring from multiple similar sequences. Appl Intell 53: 9562-9579. https://doi.org/10.1007/s10489-022-03871-z
    [105] Ferdous S, Shihab IF, Chowdhury R, et al. (2024) Reinforcement learning-guided control strategies for CAR T-cell activation and expansion. Biotechnol Bioeng 121: 2868-2880. https://doi.org/10.1002/bit.28753
    [106] Chuai G, Wang QL, Liu Q (2017) In silico meets in vivo: Towards computational CRISPR-based sgRNA design. Trends Biotechnol 35: 12-21. https://doi.org/10.1016/j.tibtech.2016.06.008
    [107] Henry VJ, Bandrowski AE, Pepin AS, et al. (2014) OMICtools: An informative directory for multi-omic data analysis. Database 2014: bau069. https://doi.org/10.1093/database/bau069
    [108] Tsai SQ, Zheng Z, Nguyen NT, et al. (2015) GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol 33: 187-197. https://doi.org/10.1038/nbt.3117
    [109] Frock RL, Hu J, Meyers RM, et al. (2015) Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat Biotechnol 33: 179-186. https://doi.org/10.1038/nbt.3101
    [110] Ran FA, Cong L, Yan WX, et al. (2015) In vivo genome editing using Staphylococcus aureus Cas9. Nature 520: 186-91. https://doi.org/10.1038/nature14299
    [111] Wang X, Wang Y, Wu X, et al. (2015) Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors. Nat Biotechnol 33: 175-178. https://doi.org/10.1038/nbt.3127
    [112] Chuai G, Ma H, Yan J, et al. (2018) DeepCRISPR: Optimized CRISPR guide RNA design by deep learning. Genome Biol 19: 80. https://doi.org/10.1186/s13059-018-1459-4
    [113] Bengio Y, Courville A, Vincent P (2013) Representation learning: A review and new perspectives. IEEE Trans Pattern Anal Mach Intell 35: 1798-1828. https://doi.org/10.1109/TPAMI.2013.50
    [114] Seyyed-Kalantari L, Zhang H, McDermott MBA, et al. (2021) Underdiagnosis bias of artificial intelligence algorithms applied to chest radiographs in under-served patient populations. Nat Med 27: 2176-2182. https://doi.org/10.1038/s41591-021-01595-0
    [115] Longo L, Brcic M, Cabitza F, et al. (2024) Explainable Artificial Intelligence (XAI) 2.0: A manifesto of open challenges and interdisciplinary research directions. Inform Fusion 106: 102301. https://doi.org/10.1016/j.inffus.2024.102301
    [116] Raza MM, Venkatesh KP, Kvedar JC (2024) Generative AI and large language models in health care: pathways to implementation. Npj Digit Med 7: 62. https://doi.org/10.1038/s41746-023-00988-4
  • Reader Comments
  • © 2025 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(867) PDF downloads(52) Cited by(1)

Article outline

Figures and Tables

Figures(4)  /  Tables(3)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog