STAT3 Signaling Pathway in Health and Disease

Md Abdus Samad , Iftikhar Ahmad , Aakifah Hasan , Mohammad Hassan Alhashmi , Arusha Ayub , Fahad A. Al-Abbasi , Ajoy Kumer , Shams Tabrez

MedComm ›› 2025, Vol. 6 ›› Issue (4) : e70152

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (4) : e70152 DOI: 10.1002/mco2.70152
REVIEW

STAT3 Signaling Pathway in Health and Disease

Author information +
History +
PDF

Abstract

Signal transducer and activator of transcription 3 (STAT3) is a critical transcription factor involved in multiple physiological and pathological processes. While STAT3 plays an essential role in homeostasis, its persistent activation has been implicated in the pathogenesis of various diseases, particularly cancer, bone-related diseases, autoimmune disorders, inflammatory diseases, cardiovascular diseases, and neurodegenerative conditions. The interleukin-6/Janus kinase (JAK)/STAT3 signaling axis is central to STAT3 activation, influencing tumor microenvironment remodeling, angiogenesis, immune evasion, and therapy resistance. Despite extensive research, the precise mechanisms underlying dysregulated STAT3 signaling in disease progression remain incompletely understood, and no United States Food and Drug Administration (USFDA)-approved direct STAT3 inhibitors currently exist. This review provides a comprehensive evaluation of STAT3's role in health and disease, emphasizing its involvement in cancer stem cell maintenance, metastasis, inflammation, and drug resistance. We systematically discuss therapeutic strategies, including JAK inhibitors (tofacitinib, ruxolitinib), Src Homology 2 domain inhibitors (S3I-201, STATTIC), antisense oligonucleotides (AZD9150), and nanomedicine-based drug delivery systems, which enhance specificity and bioavailability while reducing toxicity. By integrating molecular mechanisms, disease pathology, and emerging therapeutic interventions, this review fills a critical knowledge gap in STAT3-targeted therapy. Our insights into STAT3 signaling crosstalk, epigenetic regulation, and resistance mechanisms offer a foundation for developing next-generation STAT3 inhibitors with greater clinical efficacy and translational potential.

Keywords

cancer / IL-6 pathway / JAK/STAT signaling / nanomaterial-based drug delivery / STAT3 / targeted therapy

Cite this article

Download citation ▾
Md Abdus Samad, Iftikhar Ahmad, Aakifah Hasan, Mohammad Hassan Alhashmi, Arusha Ayub, Fahad A. Al-Abbasi, Ajoy Kumer, Shams Tabrez. STAT3 Signaling Pathway in Health and Disease. MedComm, 2025, 6(4): e70152 DOI:10.1002/mco2.70152

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

S. Chan, Z. Liu, Y. Chen, et al., “The JAK-STAT Signaling-Related Signature Serves as a Prognostic and Predictive Biomarker for Renal Cell Carcinoma Immunotherapy,” Gene 927 (2024): 148719.

[2]

X. H. Xu, J. X. Zhang, H. X. Liu, Z. Zhao, and J. Y. Jiang, “Intervention of inflammation associated With ankylosing spondylitis by triptolide promotes histone H3 Iys-27 trimethylation,” Immunopharmacology and Immunotoxicology 46, no. 6 (2024): 785-792.

[3]

Q. Hu, Q. Bian, D. Rong, et al., “JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens,” Frontiers in Bioengineering and Biotechnology 11 (2023): 1110765.

[4]

S. Wei, J. Li, M. Tang, et al., “STAT3 and p63 in the Regulation of Cancer Stemness,” Frontiers in Genetics 13 (2022): 909251.

[5]

W. Wang, M. C. Lopez McDonald, C. Kim, et al., “The complementary roles of STAT3 and STAT1 in cancer biology: Insights Into tumor pathogenesis and therapeutic strategies,” Frontiers in Immunology 14 (2023): 1265818.

[6]

M. Hashemi, S. Abbaszadeh, M. Rashidi, et al., “STAT3 as a newly emerging target in colorectal cancer therapy: Tumorigenesis, therapy response, and pharmacological/nanoplatform strategies,” Environmental Research 233 (2023): 116458.

[7]

N. Jill, S. Bhootra, S. Kannanthodi, et al., “Interplay Between signal transducers and activators of transcription (STAT) proteins and cancer: Involvement, therapeutic and prognostic perspective,” Clinical and Experimental Medicine 23, no. 8 (2023): 4323-4339.

[8]

C. D. Mohan, S. Rangappa, H. D. Preetham, et al., “Targeting STAT3 signaling pathway in cancer by agents derived From Mother Nature,” Seminars in Cancer Biology 80 (2022): 157-182.

[9]

M. Tolomeo and A. Cascio, “The Multifaced Role of STAT3 in Cancer and Its Implication for Anticancer Therapy,” International Journal of Molecular Sciences 22, no. 2 (2021): 603.

[10]

P. Faida, M. K. I. Attiogbe, U. Majeed, J. Zhao, L. Qu, and D. Fan, “Lung cancer treatment potential and limits associated With the STAT family of transcription factors,” Cell Signalling 109 (2023): 110797.

[11]

J. Li, Z. Yin, B. Huang, K. Xu, and J. Su, “Stat3 Signaling Pathway: A Future Therapeutic Target for Bone-Related Diseases,” Frontiers in Pharmacology 13 (2022): 897539.

[12]

H. Jiang, J. Yang, T. Li, et al., “JAK/STAT3 signaling in cardiac fibrosis: A promising therapeutic target,” Frontiers in Pharmacology 15 (2024): 1336102.

[13]

H. Li, Q. Bi, H. Cui, C. Lv, and M. Wang, “Suppression of autophagy Through JAK2/STAT3 contributes to the therapeutic action of rhynchophylline on asthma,” BMC Complementary Medicine and Therapies 21, no. 1 (2021): 1-12.

[14]

K. Saito, M. Fujimoto, E. Funajima, et al., “Novel germline STAT3 gain-of-function mutation causes autoimmune diseases and severe growth failure,” Journal of Allergy and Clinical Immunology: Global 3, no. 4 (2024): 100312.

[15]

G. N. Zyuz'kov, V. V. Zhdanov, L. A. Miroshnichenko, et al., “The Role of JAK and STAT3 in Regulation of Secretory Function of Neuroglial Cells of Different Types in Ethanol-Induced Neurodegenerationt,” Bulletin of Experimental Biology and Medicine 172, no. 6 (2022): 686-690.

[16]

M. El-Tanani, A. O. Al Khatib, S. M. Aladwan, A. Abuelhana, P. A. McCarron, and M. M. Tambuwala, “Importance of STAT3 signalling in cancer, metastasis and therapeutic interventions,” Cell Signalling 92 (2022): 110275.

[17]

M. M. Kasembeli, E. Kaparos, U. Bharadwaj, et al., “Aberrant function of pathogenic STAT3 mutant proteins is linked to altered stability of monomers and homodimers,” Blood 141, no. 12 (2023): 1411-1424.

[18]

S. Manoharan and E. Perumal, “A strategic review of STAT3 signaling inhibition by phytochemicals for cancer prevention and treatment: Advances and insights,” Fitoterapia 179 (2024): 106265.

[19]

A. Tesoriere, A. Dinarello, and F. Argenton, “The Roles of Post-Translational Modifications in STAT3 Biological Activities and Functions,” Biomed 9, no. 8 (2021): 956.

[20]

M. Garg, M. K. Shanmugam, V. Bhardwaj, et al., “The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting Through natural products for cancer prevention and therapy,” Medicinal Research Reviews 41, no. 3 (2021): 1291-1336.

[21]

C. E. Heron, L. C. Strowd, and S. R. Feldman, “Janus Kinase (JAK) Inhibitors,” Handbook of Systemic Drug Treatment in Dermatology170-176, published online February 13, 2023, https://doi.org/10.1201/9781003016786-25.

[22]

K. Sardana, S. Bathula, and A. Khurana, “Which is the ideal JAK inhibitor for alopecia areata—Baricitinib, tofacitinib, ritlecitinib or ifidancitinib—Revisiting the immunomechanisms of the JAK pathway,” Indian Dermatology Online Journal 14, no. 4 (2023): 465-474.

[23]

J. Song, J. Wang, S. Tian, and H. Li, “Discovery of STAT3 Inhibitors: Recent Advances and Future Perspectives,” Current Medicinal Chemistry 30, no. 16 (2022): 1824-1847.

[24]

Y. Hua, X. Yuan, Y. H. Shen, et al., “Novel STAT3 Inhibitors Targeting STAT3 Dimerization by Binding to the STAT3 SH2 Domain,” Frontiers in Pharmacology 13 (2022): 836724.

[25]

O. Kovecses, F. E. Mercier, and M. McKeague, “Nucleic acid therapeutics as differentiation agents for myeloid leukemias,” Leukemia 38, no. 7 (2024): 1441-1454.

[26]

Q. Feng and K. Xiao, “Nanoparticle-Mediated Delivery of STAT3 Inhibitors in the Treatment of Lung Cancer,” Pharmaceutics 14, no. 12 (2022): 2787.

[27]

H. Q. Wang, Q. W. Man, F. Y. Huo, et al., “STAT3 pathway in cancers: Past, present, and future,” MedComm 3, no. 2 (2022): e124.

[28]

S. Comità, S. Femmino, C. Thairi, et al., “Regulation of STAT3 and its role in cardioprotection by conditioning: Focus on non-genomic roles targeting mitochondrial function,” Basic Research in Cardiology 116, no. 1 (2021): 1-31.

[29]

K. Wu, Q. Sun, D. Liu, et al., “Alternative Splicing Landscape of Head and Neck Squamous Cell Carcinoma,” Technology in Cancer Research & Treatment 23 (2024).

[30]

M. Kise, S. Masaki, N. Kataoka, and K. Suzuki, “Identification of Entinostat as a Novel Modifier of STAT3 Pre-mRNA Alternative Splicing,” Biological & Pharmaceutical Bulletin 47, no. 9 (2024): 1504-1510.

[31]

S. Edtmayer, A. Witalisz-Siepracka, B. Zdársky, et al., “A novel function of STAT3β in suppressing interferon response improves outcome in acute myeloid leukemia,” Cell Death & Disease 15, no. 5 (2024): 1-12.

[32]

J. T. Yu, S. Fan, X. Y. Li, et al., “Novel insights Into STAT3 in renal diseases,” Biomedicine & Pharmacotherapy 165 (2023): 115166.

[33]

Y. Hu, Z. Dong, and K. Liu, “Unraveling the complexity of STAT3 in cancer: Molecular understanding and drug discovery,” Journal of Experimental & Clinical Cancer Research 43, no. 1 (2024): 1-29.

[34]

X. Hou and F. Tian, “STAT3-mediated osteogenesis and osteoclastogenesis in osteoporosis,” Cell Communication and Signaling 20, no. 1 (2022): 1-17.

[35]

Z. Wu, W. Li, K. Jiang, et al., “Regulation of bone homeostasis: Signaling pathways and therapeutic targets,” MedComm 5, no. 8 (2024): e657.

[36]

M. L. Sobah, C. Liongue, and A. C. Ward, “Contribution of Signal Transducer and Activator of Transcription 3 (STAT3) to Bone Development and Repair,” International Journal of Molecular Sciences 25, no. 1 (2024): 389.

[37]

X. Yin, Q. Wang, Y. Tang, T. Wang, Y. Zhang, and T. Yu, “Research progress on macrophage polarization During osteoarthritis disease progression: A review,” Journal of Orthopaedic Surgery and Research 19, no. 1 (2024): 584.

[38]

J. Li, W. Zhang, X. Liu, et al., “Endothelial Stat3 activation promotes osteoarthritis development,” Cell Proliferation 56, no. 12 (2023): e13518.

[39]

V. Molnar, E. Pavelić, K. Vrdoljak, et al., “Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review,” Genes 13, no. 6 (2022): 949.

[40]

B. Chen, K. Ning, M. L. Sun, and X. A. Zhang, “Regulation and therapy, the role of JAK2/STAT3 signaling pathway in OA: A systematic review,” Cell Communication and Signaling 21, no. 1 (2023): 1-14.

[41]

O. Kuryata, O. Akimov, S. Denisenko, et al., “Chondroitin sulfate in osteoarthritis management Among diabetic patients: Molecular mechanisms and clinical potential,” Romanian Journal of Diabetes Nutrition and Metabolic Diseases 30, no. 4 (2023): 481-493.

[42]

A. I. S. Jrad, M. Trad, W. Bzeih, G. El Hasbani, and I. Uthman, “Role of pro-inflammatory interleukins in osteoarthritis: A narrative review,” Connective Tissue Research 64, no. 3 (2023): 238-247.

[43]

Q. Zhou, Q. Ren, L. Jiao, et al., “The potential roles of JAK/STAT signaling in the progression of osteoarthritis,” Front Endocrinol (Lausanne) 13 (2022): 1069057.

[44]

T. Liang, T. Chen, J. Qiu, et al., “Inhibition of nuclear receptor RORα attenuates cartilage damage in osteoarthritis by modulating IL-6/STAT3 pathway,” Cell Death & Disease 12, no. 10 (2021): 1-13.

[45]

G. Dwivedi, L. Flaman, B. Alaybeyoglu, et al., “Inflammatory cytokines and mechanical injury induce post-traumatic osteoarthritis-Like changes in a human cartilage-bone-synovium microphysiological system,” Arthritis Research & Therapy 24, no. 1 (2022): 1-18.

[46]

Z. Zou, H. Li, K. Yu, et al., “The potential role of synovial cells in the progression and treatment of osteoarthritis,” Exploration 3, no. 5 (2023): 20220132.

[47]

A. Wayupat, P. Kongtawelert, P. Pothacharoen, T. H. Shwe, and T. Phitak, Leptin augments the inflammatory effect of Interleukin 1 Beta on synoviocytes mainly Through NF-kB and STAT3 prompting its possible implementation in OA pathogenesis, published online October 8, 2024, https://doi.org/10.21203/RS.3.RS-4878390/V1.

[48]

J. Zhu, G. Ruan, H. Cen, et al., “Association of serum levels of inflammatory markers and adipokines With joint symptoms and structures in participants With knee osteoarthritis,” Rheumatology 61, no. 3 (2022): 1044-1052.

[49]

J. Xu, L. Yu, F. Liu, L. Wan, and Z. Deng, “The effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis: A review,” Frontiers in Immunology 14 (2023): 1222129.

[50]

E. Umur, S. B. Bulut, P. Yiğit, et al., “Exploring the Role of Hormones and Cytokines in Osteoporosis Development,” Biomed 12, no. 8 (2024): 1830.

[51]

W. Da, L. Tao, and Y. Zhu, “The Role of Osteoclast Energy Metabolism in the Occurrence and Development of Osteoporosis,” Frontiers in Endocrinology (Lausanne) 12 (2021): 675385.

[52]

X. Liu, Z. Zhou, W. N. Zeng, Q. Zeng, and X. Zhang, “The role of toll-Like receptors in orchestrating osteogenic differentiation of mesenchymal stromal cells and osteoimmunology,” Frontiers in Cell and Developmental Biology 11 (2023): 1277686.

[53]

T. Jiang, T. Xia, F. Qiao, N. Wang, Y. Jiang, and H. Xin, “Role and Regulation of Transcription Factors in Osteoclastogenesis,” International Journal of Molecular Sciences 24, no. 22 (2023): 16175.

[54]

C. H. Li, Z. R. , Z. D. Zhao, et al., “Nitazoxanide, an Antiprotozoal Drug, Reduces Bone Loss in Ovariectomized Mice by Inhibition of RANKL-Induced Osteoclastogenesis,” Frontiers in Pharmacology 12 (2021): 781640.

[55]

P. Ethiraj, I. A. Haque, A. K. Alford, et al., “Inhibition of NFAM1 suppresses phospho-SAPK/JNK signaling During osteoclast differentiation and bone resorption,” Journal of Cellular Biochemistry 122, no. 10 (2021): 1534-1543.

[56]

J. Xu, W. Jiao, D. B. Wu, et al., “Yishen Tongbi decoction attenuates inflammation and bone destruction in rheumatoid arthritis by regulating JAK/STAT3/SOCS3 pathway,” Frontiers in Immunology 15 (2024): 1381802.

[57]

G. Kour, R. Choudhary, S. Anjum, A. Bhagat, B. K. Bajaj, and Z. Ahmed, “Phytochemicals targeting JAK/STAT pathway in the treatment of rheumatoid arthritis: Is there a future?,” Biochemical Pharmacology 197 (2022): 114929.

[58]

N. Kondo, T. Kuroda, and D. Kobayashi, “Cytokine Networks in the Pathogenesis of Rheumatoid Arthritis,” International Journal of Molecular Sciences 22, no. 20 (2021): 10922.

[59]

S. Liu, H. Ma, H. Zhang, C. Deng, and P. Xin, “Recent advances on signaling pathways and their inhibitors in rheumatoid arthritis,” Clinical Immunology 230 (2021): 108793.

[60]

D. Ilchovska and D. M. Barrow, “An Overview of the NF-kB mechanism of pathophysiology in rheumatoid arthritis, investigation of the NF-kB ligand RANKL and related nutritional interventions,” Autoimmunity Reviews 20, no. 2 (2021): 102741.

[61]

Q. Niu, J. Gao, L. Wang, J. Liu, and L. Zhang, “Regulation of differentiation and generation of osteoclasts in rheumatoid arthritis,” Frontiers in Immunology 13 (2022): 1034050.

[62]

G. M. Kim, H. Park, and S. Y. Lee, “Roles of osteoclast-associated receptor in rheumatoid arthritis and osteoarthritis,” Joint Bone Spine 89, no. 5 (2022): 105400.

[63]

S. Tabrez, M. R. S. Mohammed, N. R. Jabir, and M. I. Khan, “Identification of novel cardiovascular disease associated metabolites using untargeted metabolomics,” Biological Chemistry 402, no. 6 (2021): 749-757.

[64]

S. Tabrez, N. R. Jabir, T. A. Zughaibi, and M. Suhail, “Association of interleukin-18 promoter polymorphism With comorbid conditions of cardiovascular disease,” Journal of King Saud University 35, no. 1 (2023): 102440.

[65]

X. Zhang, S. Chen, G. Yin, et al., “The Role of JAK/STAT Signaling Pathway and Its Downstream Influencing Factors in the Treatment of Atherosclerosis,” Journal of Cardiovascular Pharmacology and Therapeutics 29 (2024).

[66]

Y. Feng, D. Ye, Z. Wang, et al., “The Role of Interleukin-6 Family Members in Cardiovascular Diseases,” Frontiers in Cardiovascular Medicine 9 (2022): 818890.

[67]

A. M. Markin, Y. V. Markina, A. I. Bogatyreva, et al., “The Role of Cytokines in Cholesterol Accumulation in Cells and Atherosclerosis Progression,” International Journal of Molecular Sciences 24, no. 7 (2023): 6426.

[68]

S. Tabrez, N. R. Jabir, T. A. Zughaibi, and M. Suhail, “Association of IL-6 promoter polymorphism hotspots (− 174G/C and − 572G/C) With cardiovascular disease risk factors,” Molecular Biology Reports 49, no. 3 (2022): 2265-2272.

[69]

S. Singh, D. Anshita, and V. Ravichandiran, “MCP-1: Function, regulation, and involvement in disease,” International Immunopharmacology 101 (2021): 107598.

[70]

T. Xia, M. Zhang, W. Lei, et al., “Advances in the role of STAT3 in macrophage polarization,” Frontiers in Immunology 14 (2023): 1160719.

[71]

X. Liu, J. Liu, Y. Li, and H. Zhang, “The Correlation Between the Inflammatory Effects of Activated Macrophages in Atherosclerosis and Aortic Dissection,” Annals of Vascular Surgery 85 (2022): 341-346.

[72]

X. Sun, J. Gao, X. Meng, X. Lu, L. Zhang, and R. Chen, “Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling,” Frontiers in Immunology 12 (2021): 763334.

[73]

P. Lin, H. H. Ji, Y. J. Li, and S. D. Guo, “Macrophage Plasticity and Atherosclerosis Therapy,” Frontiers in Molecular Biosciences 8 (2021): 679797.

[74]

Y. Wan, L. Mo, H. Huang, et al., “Cadmium contributes to atherosclerosis by affecting macrophage polarization,” Food and Chemical Toxicology 173 (2023): 113603.

[75]

B. Guo, Y. Yu, M. Wang, et al., “Targeting the JAK2/STAT3 signaling pathway With natural plants and phytochemical ingredients: A novel therapeutic method for combatting cardiovascular diseases,” Biomedicine & Pharmacotherapy 172 (2024): 116313.

[76]

W. Li, J. Liu, R. Jiao, et al., “Baricitinib alleviates cardiac fibrosis and inflammation induced by chronic sympathetic activation,” International Immunopharmacology 140 (2024): 112894.

[77]

N. V. Dwivedi, S. Datta, K. El-Kersh, et al., “GPCRs and fibroblast heterogeneity in fibroblast-associated diseases,” Faseb Journal 37, no. 8 (2023): e23101.

[78]

S. Perveen, R. Vanni, M. Lo Iacono, R. Rastaldo, and C. Giachino, “Direct Reprogramming of Resident Non-Myocyte Cells and Its Potential for In Vivo Cardiac Regeneration,” Cells 12, no. 8 (2023): 1166.

[79]

N. J. Patel, D. M. Nassal, D. Gratz, and T. J. Hund, “Emerging therapeutic targets for cardiac arrhythmias: Role of STAT3 in regulating cardiac fibroblast function,” Expert Opinion on Therapeutic Targets 25, no. 1 (2021): 63-73.

[80]

S. Lovisa, “Epithelial-to-Mesenchymal Transition in Fibrosis: Concepts and Targeting Strategies,” Frontiers in Pharmacology 12 (2021): 737570.

[81]

Y. Liu, M. Hu, G. Fan, N. Xing, and R. Zhang, “Effect of Baricitinib on the epithelial-mesenchymal transition of alveolar epithelial cells induced by IL-6,” International Immunopharmacology 110 (2022): 109044.

[82]

M. Aliyu, F. T. Zohora, A. U. Anka, et al., “Interleukin-6 cytokine: An overview of the immune regulation, immune dysregulation, and therapeutic approach,” International Immunopharmacology 111 (2022): 109130.

[83]

J. Zhao, Y. F. Qi, and Y. R. Yu, “STAT3: A key regulator in liver fibrosis,” Annals of Hepatology 21 (2021): 100224.

[84]

A. Shahini and A. Shahini, “Role of interleukin-6-mediated inflammation in the pathogenesis of inflammatory bowel disease: Focus on the available therapeutic approaches and gut microbiome,” Journal of Cell Communication and Signaling 17, no. 1 (2022): 55-74.

[85]

A. A. Nikolskii, I. P. Shilovskiy, E. D. Barvinskaia, A. V. Korneev, M. S. Sundukova, and M. R. Khaitov, “Role of STAT3 Transcription Factor in Pathogenesis of Bronchial Asthma,” Biochemistry 86, no. 11 (2021): 1489-1501.

[86]

Y. Zhou, X. Huang, H. Yu, et al., “TMT-based quantitative proteomics revealed protective efficacy of Icariside II Against airway inflammation and remodeling via inhibiting LAMP2, CTSD and CTSS expression in OVA-induced chronic asthma mice,” Phytomedicine 118 (2023): 154941.

[87]

V. Margelidon-Cozzolino, A. Tsicopoulos, C. Chenivesse, and P. de Nadai, “Role of Th17 Cytokines in Airway Remodeling in Asthma and Therapy Perspectives,” Frontiers in Allergy 3 (2022): 806391.

[88]

A. Jafarzadeh, P. Chauhan, M. Nemati, S. Jafarzadeh, and A. Yoshimura, “Aberrant expression of suppressor of cytokine signaling (SOCS) molecules contributes to the development of allergic diseases,” Clinical and Experimental Allergy 53, no. 11 (2023): 1147-1161.

[89]

A. M. Jetten, J. Y. Beak, A. T. Slominski, and B. Jensen, “Retinoic acid-related orphan receptor (ror) inverse agonists: Potential therapeutic strategies for multiple inflammatory diseases?,” Nucl Recept Art Sci Modul Des Discov 349-377, published online September 28, 2021, https://doi.org/10.1007/978-3-030-78315-0_14/FIGURES/7.

[90]

S. Berry, C. Dossou, A. Kashif, et al., “The role of IL-17 and anti-IL-17 agents in the immunopathogenesis and management of autoimmune and inflammatory diseases,” International Immunopharmacology 102 (2022): 108402.

[91]

L. Cui, X. Qin, T. Fu, et al., “Attenuated airways inflammation and remodeling in IL-37a and IL-37b transgenic mice With an ovalbumin-induced chronic asthma,” Cellular Immunology 391-392 (2023): 104759.

[92]

A. Chetty and H. C. Nielsen, “Targeting airway smooth muscle hypertrophy in asthma: An approach whose time has come,” Journal of Asthma and Allergy 14 (2021): 539-556.

[93]

Q. Lu, M. F. Yang, Y. J. Liang, et al., “Immunology of Inflammatory Bowel Disease: Molecular Mechanisms and Therapeutics,” Journal of Inflammation Research 15 (2022): 1825-1844.

[94]

L. Wang, Y. Hu, B. Song, Y. Xiong, J. Wang, and D. Chen, “Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease,” Inflammation Research 70, no. 7 (2021): 753-764.

[95]

U. Tripathi, Y. Stern, I. Dagan, et al., “Genetic Overlap Between Inflammatory Bowel Disease and Neurological Disorders: Insights From GWAS and Gene Expression Analysis,” Medrxiv, published online September 26, 2024:2024.07.29.24311160, https://doi.org/10.1101/2024.07.29.24311160.

[96]

M. Vebr, R. Pomahačová, J. Sýkora, and J. Schwarz, “A Narrative Review of Cytokine Networks: Pathophysiological and Therapeutic Implications for Inflammatory Bowel Disease Pathogenesis,” Biomed 11, no. 12 (2023): 3229.

[97]

D. Aebisher, D. Bartusik-Aebisher, A. Przygórzewska, P. Oleś, P. Woźnicki, and A. Kawczyk-Krupka, “Key Interleukins in Inflammatory Bowel Disease—A Review of Recent Studies,” International Journal of Molecular Sciences 26, no. 1 (2024): 121.

[98]

W. Zhang, X. Liu, Y. Zhu, et al., “Transcriptional and posttranslational regulation of Th17/Treg balance in health and disease,” European Journal of Immunology 51, no. 9 (2021): 2137-2150.

[99]

J. Mackie, C. S. Ma, S. G. Tangye, and A. Guerin, “The ups and downs of STAT3 function: Too much, too little and human immune dysregulation,” Clinical and Experimental Immunology 212, no. 2 (2023): 107-116.

[100]

A. Kondo, S. Ma, M. Y. Y. Lee, et al., “Highly Multiplexed Image Analysis of Intestinal Tissue Sections in Patients With Inflammatory Bowel Disease,” Gastroenterology 161, no. 6 (2021): 1940-1952.

[101]

P. Robinson, K. Montoya, E. Magness, et al., “Therapeutic Potential of a Small-Molecule STAT3 Inhibitor in a Mouse Model of Colitis,” Cancers 15, no. 11 (2023): 2977.

[102]

F. Cordes, E. Lenker, T. Weinhage, et al., “Impaired IFN-γ-dependent STAT3 Activation Is Associated With Dysregulation of Regulatory and Inflammatory Signaling in Monocytes of Ulcerative Colitis Patients,” Inflammatory Bowel Diseases 27, no. 6 (2021): 887-901.

[103]

Z. Du, A. Chen, L. Huang, et al., “STAT3 couples With 14-3-3σ to regulate BCR signaling, B-cell differentiation, and IgE production,” Journal of Allergy and Clinical Immunology 147, no. 5 (2021): 1907-1923. e6.

[104]

M. Michée-Cospolite, M. Boudigou, A. Grasseau, et al., “Molecular Mechanisms Driving IL-10- Producing B Cells Functions: STAT3 and c-MAF as Underestimated Central Key Regulators?,” Frontiers in Immunology 13 (2022): 818814.

[105]

S. Vlachiotis and H. Abolhassani, “Transcriptional regulation of B cell class-switch recombination: The role in development of noninfectious complications,” Expert Review of Clinical Immunology 18, no. 11 (2022): 1145-1154.

[106]

A. Witalisz-Siepracka, K. Klein, B. Zdársky, and D. Stoiber, “The Multifaceted Role of STAT3 in NK-Cell Tumor Surveillance,” Frontiers in Immunology 13 (2022): 947568.

[107]

C. Zalfa and S. Paust, “Natural Killer Cell Interactions With Myeloid Derived Suppressor Cells in the Tumor Microenvironment and Implications for Cancer Immunotherapy,” Frontiers in Immunology 12 (2021): 633205.

[108]

R. Pandey, M. Bakay, and H. Hakonarson, “SOCS-JAK-STAT inhibitors and SOCS mimetics as treatment options for autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis,” Frontiers in Immunology 14 (2023): 1271102.

[109]

P. Kotyla, O. Gumkowska-Sroka, B. Wnuk, and K. Kotyla, “Jak Inhibitors for Treatment of Autoimmune Diseases: Lessons From Systemic Sclerosis and Systemic Lupus Erythematosus,” Pharmaceutics 15, no. 8 (2022): 936.

[110]

T. P. Vogel, J. W. Leiding, M. A. Cooper, and L. R. Forbes Satter, “STAT3 gain-of-function syndrome,” Frontiers in Pediatrics 10 (2023): 770077.

[111]

L. Nussbaum, Y. L. Chen, and G. S. Ogg, “Role of regulatory T cells in psoriasis pathogenesis and treatment,” British Journal of Dermatology 184, no. 1 (2021): 14-24.

[112]

P. Hu, M. Wang, H. Gao, et al., “The Role of Helper T Cells in Psoriasis,” Frontiers in Immunology 12 (2021): 788940.

[113]

M. Kamata and Y. Tada, “Crosstalk: Keratinocytes and immune cells in psoriasis,” Frontiers in Immunology 14 (2023): 1286344.

[114]

A. Orsmond, L. Bereza-Malcolm, T. Lynch, L. March, and M. Xue, “Skin Barrier Dysregulation in Psoriasis,” International Journal of Molecular Sciences 22, no. 19 (2021): 10841.

[115]

M. Kishimoto, M. Komine, M. Sashikawa-Kimura, et al., “STAT3 Activation in Psoriasis and Cancers,” Diagnostics 11, no. 10 (2021): 1903.

[116]

M. Zhang, N. Li, R. Cai, et al., “Rosmarinic acid protects mice From imiquimod induced psoriasis-Like skin lesions by inhibiting the IL-23/Th17 axis via regulating Jak2/Stat3 signaling pathway,” Phytotherapy Research 35, no. 8 (2021): 4526-4537.

[117]

L. I. Ortiz-Lopez, V. Choudhary, and W. B. Bollag, “Updated Perspectives on Keratinocytes and Psoriasis: Keratinocytes are More Than Innocent Bystanders,” Psoriasis: Targets and Therapy 12 (2022): 73-87.

[118]

S. Parab and G. Doshi, “The Experimental Animal Models in Psoriasis Research: A Comprehensive Review,” International Immunopharmacology 117 (2023): 109897.

[119]

M. Ramaswamy, R. Tummala, K. Streicher, A. Nogueira da Costa, and P. Z. Brohawn, “The Pathogenesis, Molecular Mechanisms, and Therapeutic Potential of the Interferon Pathway in Systemic Lupus Erythematosus and Other Autoimmune Diseases,” International Journal of Molecular Sciences 22, no. 20 (2021): 11286.

[120]

M. A. Ameer, H. Chaudhry, J. Mushtaq, et al., “An Overview of Systemic Lupus Erythematosus (SLE) Pathogenesis, Classification, and Management,” Cureus 14, no. 10 (2022): e30330.

[121]

N. Bolouri, M. Akhtari, E. Farhadi, et al., “Role of the innate and adaptive immune responses in the pathogenesis of systemic lupus erythematosus,” Inflammation Research 71, no. 5 (2022): 537-554.

[122]

E. Moysidou, M. Christodoulou, G. Lioulios, et al., “Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis,” International Journal of Molecular Sciences 25, no. 20 (2024): 10905.

[123]

S. P. Pandey, R. Bhaskar, S. S. Han, and K. B. Narayanan, “Autoimmune Responses and Therapeutic Interventions for Systemic Lupus Erythematosus: A Comprehensive Review,” Endocrine, Metabolic & Immune Disorders - Drug Targets 24, no. 5 (2023): 499-518.

[124]

T. Montoya, M. L. Castejón, R. Muñoz-García, and C. Alarcón-De-La-Lastra, “Epigenetic linkage of systemic lupus erythematosus and nutrition,” Nutrition Research Reviews 36, no. 1 (2023): 39-59.

[125]

C. M. Hedrich, “Epigenetics,” Systemic Lupus Erythematosus: Basic, Applied and Clinical Aspects 277-292, published online January 1, 2021, https://doi.org/10.1016/B978-0-12-814551-7.00032-5.

[126]

J. Huang, X. Li, Q. Zhu, M. Wang, Z. Xie, and T. Zhao, “Imbalance of Th17 cells, Treg cells and associated cytokines in patients With systemic lupus erythematosus: A meta-analysis,” Frontiers in Immunology 15 (2024): 1425847.

[127]

F. C. Paquissi and H. Abensur, “The Th17/IL-17 Axis and Kidney Diseases, With Focus on Lupus Nephritis,” Frontiers of Medicine 8 (2021): 654912.

[128]

J. Zhou, B. Lei, F. Shi, et al., “CAR T-cell therapy for systemic lupus erythematosus: Current status and future perspectives,” Frontiers in Immunology 15 (2024): 1476859.

[129]

T. Zhou, Y. Liu, Z. Yang, et al., “IL-17 signaling induces iNOS+ microglia activation in retinal vascular diseases,” Glia 69, no. 11 (2021): 2644-2657.

[130]

K. F. Azman and R. Zakaria, “Recent Advances on the Role of Brain-Derived Neurotrophic Factor (BDNF) in Neurodegenerative Diseases,” International Journal of Molecular Sciences 23, no. 12 (2022): 6827.

[131]

M. Jain, M. K. Singh, H. Shyam, et al., “Role of JAK/STAT in the Neuroinflammation and its Association With Neurological Disorders,” Annals of Neurosciences 28, no. 3-4 (2021): 191-200.

[132]

C. G. Hart and S. Karimi-Abdolrezaee, “Recent insights on astrocyte mechanisms in CNS homeostasis, pathology, and repair,” Journal of Neuroscience Research 99, no. 10 (2021): 2427-2462.

[133]

M. Bhatt, M. Sharma, and B. D. C. Neurobiology, “The Role of Inflammatory Cascade and Reactive Astrogliosis in Glial Scar Formation Post-spinal Cord Injury,” Springer 44, no. 1 (2024): 78. and M, 2024 undefined.

[134]

S. P. Panda, A. Kesharwani, S. Datta, D. Prasanth, S. K. Panda, and A. Guru, “JAK2/STAT3 as a new potential target to manage neurodegenerative diseases: An interactive review,” European Journal of Pharmacology 970 (2024): 176490.

[135]

M. Hoque, A. Samanta, S. S. M. Alam, T. A. Zughaibi, M. A. Kamal, and S. Tabrez, “Nanomedicine-based immunotherapy for Alzheimer's disease,” Neuroscience and Biobehavioral Reviews 144 (2023): 104973.

[136]

M. S. Khan, Z. Khan, N. R. Jabir, et al., “Synthesis and Neurobehavioral Evaluation of a Potent Multitargeted Inhibitor for the Treatment of Alzheimer's Disease,” Molecular Neurobiology 62, no. 2 (2024): 1558-1576.

[137]

S. Thakur, R. Dhapola, P. Sarma, B. Medhi, and D. H. K. Reddy, “Neuroinflammation in Alzheimer's Disease: Current Progress in Molecular Signaling and Therapeutics,” Inflammation 46, no. 1 (2023): 1-17.

[138]

Z. Fan, W. Zhang, Q. Cao, et al., “JAK2/STAT3 pathway regulates microglia polarization involved in hippocampal inflammatory damage due to acute paraquat exposure,” Ecotoxicology and Environmental Safety 234 (2022): 113372.

[139]

Y. Zhong, B. Yin, Y. Ye, et al., “The bidirectional role of the JAK2/STAT3 signaling pathway and related mechanisms in cerebral ischemia-reperfusion injury,” Experimental Neurology 341 (2021): 113690.

[140]

S. Thangwaritorn, C. Lee, E. Metchikoff, et al., “A Review of Recent Advances in the Management of Alzheimer's Disease,” Cureus 16, no. 4 (2024): e58416.

[141]

W. Wu, Y. Ji, Z. Wang, et al., “The FDA-approved anti-amyloid-β monoclonal antibodies for the treatment of Alzheimer's disease: A systematic review and meta-analysis of randomized controlled trials,” European Journal of Medical Research 28, no. 1 (2023): 1-13.

[142]

C. P. Chang, C. W. Wu, and Y. Chern, “Metabolic dysregulation in Huntington's disease: Neuronal and glial perspectives,” Neurobiology of Disease 201 (2024): 106672.

[143]

X. Li, H. Tong, S. Xu, et al., “Neuroinflammatory Proteins in Huntington's Disease: Insights Into Mechanisms, Diagnosis, and Therapeutic Implications,” International Journal of Molecular Sciences 25, no. 21 (2024): 11787.

[144]

Q. Jia, S. Li, X. J. Li, and P. Yin, “Neuroinflammation in Huntington's disease: From animal models to clinical therapeutics,” Frontiers in immunology 13 (2022): 1088124.

[145]

L. Abjean, H. L. Ben, M. Riquelme-Perez, et al., “Reactive astrocytes promote proteostasis in Huntington's disease Through the JAK2-STAT3 pathway,” Brain 146, no. 1 (2023): 149-166.

[146]

K. Vaibhav, M. Ahluwalia, P. Gaur, and J. Luo, “TGF-β as a Key Modulator of Astrocyte Reactivity: Disease Relevance and Therapeutic Implications,” Biomed 10, no. 5 (2022): 1206.

[147]

S. A. Liddelow, M. L. Olsen, and M. V. Sofroniew, “Reactive Astrocytes and Emerging Roles in Central Nervous System (CNS) Disorders,” Cold Spring Harbor Perspectives in Biology 16, no. 7 (2024): a041356.

[148]

Z. He, B. Song, M. Zhu, and J. Liu, “Comprehensive pan-cancer analysis of STAT3 as a prognostic and immunological biomarker,” Scientific Reports 13, no. 1 (2023): 1-18.

[149]

B. Panda, A. Tripathy, S. Patra, B. Kullu, S. Tabrez, and M. Jena, “Imperative connotation of SODs in cancer: Emerging targets and multifactorial role of action,” Iubmb Life, published online 2024, https://doi.org/10.1002/IUB.2821.

[150]

M. You, Z. Xie, N. Zhang, et al., “Signaling pathways in cancer metabolism: Mechanisms and therapeutic targets,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 1-27.

[151]

Y. Wang, Z. Wang, S. Li, J. Ma, X. Dai, and J. Lu, “Deciphering JAK/STAT signaling pathway: A multifaceted approach to tumorigenesis, progression and therapeutic interventions,” International Immunopharmacology 131 (2024): 111846.

[152]

T. M. Ayele, Z. T. Muche, A. B. Teklemariam, A. B. Kassie, and E. C. Abebe, “Role of JAK2/STAT3 Signaling Pathway in the Tumorigenesis, Chemotherapy Resistance, and Treatment of Solid Tumors: A Systemic Review,” Journal of Inflammation Research 15 (2022): 1349-1364.

[153]

O. Rahbar Farzam, S. Najafi, M. Amini, et al., “Interplay of miRNAs and lncRNAs in STAT3 signaling pathway in colorectal cancer progression,” Cancer Cell International 24, no. 1 (2024): 1-13.

[154]

C. Xue, Q. Yao, X. Gu, et al., “Evolving cognition of the JAK-STAT signaling pathway: Autoimmune disorders and cancer,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 1-24.

[155]

Y. X. Liu, B. W. Xu, X. D. Niu, et al., “Inhibition of Src/STAT3 signaling-mediated angiogenesis is involved in the anti-melanoma effects of dioscin,” Pharmacological Research 175 (2022): 105983.

[156]

W. Zhang, D. Li, B. Li, X. Chu, and B. Kong, “STAT3 as a therapeutic target in the metformin-related treatment,” International Immunopharmacology 116 (2023): 109770.

[157]

M. Hashemi, E. Sabouni, P. Rahmanian, et al., “Deciphering STAT3 signaling potential in hepatocellular carcinoma: Tumorigenesis, treatment resistance, and pharmacological significance,” Cellular & Molecular Biology Letters 28, no. 1 (2023): 1-32.

[158]

S. Guo, V. Ramar, A. A. Guo, et al., “TRPM7 transactivates the FOSL1 gene Through STAT3 and enhances glioma stemness,” Cellular and Molecular Life Sciences 80, no. 9 (2023): 1-18.

[159]

J. E. Lefler, C. B. MarElia-Bennett, K. A. Thies, et al., “STAT3 in tumor fibroblasts promotes an immunosuppressive microenvironment in pancreatic cancer,” Life Science Alliance 5, no. 11 (2022).

[160]

M. Sadrkhanloo, M. D. A. Paskeh, M. Hashemi, et al., “STAT3 signaling in prostate cancer progression and therapy resistance: An oncogenic pathway With diverse functions,” Biomedicine & Pharmacotherapy 158 (2023): 114168.

[161]

L. M. Channon, V. M. Tyma, Z. Xu, et al., “Small extracellular vesicles (exosomes) and their cargo in pancreatic cancer: Key roles in the hallmarks of cancer,” Biochimica et Biophysica Acta—Review Cancer 1877, no. 3 (2022): 188728.

[162]

M. Golmohammadi, M. Y. Zamanian, A. M. Al-Ani, et al., “Targeting STAT3 signaling pathway by curcumin and its analogues for breast cancer: A narrative review,” Animal Models and Experimental Medicine 7, no. 6 (2024): 853-867.

[163]

S. Dimri, R. Malhotra, T. Shet, S. Mokal, S. Gupta, and A. De, “Noncanonical pS727 post translational modification dictates major STAT3 activation and downstream functions in breast cancer,” Experimental Cell Research 396, no. 2 (2020): 112313.

[164]

Y. J. Li, C. Zhang, A. Martincuks, A. Herrmann, and H. Yu, “STAT proteins in cancer: Orchestration of metabolism,” Nature Reviews Cancer 23, no. 3 (2023): 115-134.

[165]

S. G. Manore, D. L. Doheny, G. L. Wong, and H. W. Lo, “IL-6/JAK/STAT3 Signaling in Breast Cancer Metastasis: Biology and Treatment,” Frontiers in Oncology 12 (2022): 866014.

[166]

X. Yang and H. Wu, “RAS signaling in carcinogenesis, cancer therapy and resistance mechanisms,” Journal of Hematology & Oncology 17, no. 1 (2024): 1-31.

[167]

L. Raji, A. Tetteh, and A. Amin, “Role of c-Src in Carcinogenesis and Drug Resistance,” Cancers 16, no. 1 (2023): 32.

[168]

Y. Ma, Y. Zhu, L. Shang, et al., “LncRNA XIST regulates breast cancer stem cells by activating proinflammatory IL-6/STAT3 signaling,” Oncogene 42, no. 18 (2023): 1419-1437.

[169]

R. A. Eladwy, H. T. Vu, R. Shah, C. G. Li, D. Chang, and D. J. Bhuyan, “The Fight Against the Carcinogenic Epstein-Barr Virus: Gut Microbiota, Natural Medicines, and Beyond,” International Journal of Molecular Sciences 24, no. 2 (2023): 1716.

[170]

J. Schwestermann, A. Besse, C. Driessen, and L. Besse, “Contribution of the Tumor Microenvironment to Metabolic Changes Triggering Resistance of Multiple Myeloma to Proteasome Inhibitors,” Frontiers in Oncology 12 (2022): 899272.

[171]

B. Gutic, T. Bozanovic, A. Mandic, et al., “Programmed cell death-1 and its ligands: Current knowledge and possibilities in immunotherapy,” Clinics 78 (2023): 100177.

[172]

O. Aksoy, J. Lind, V. Sunder-Plaßmann, S. Vallet, and K. Podar, “Bone marrow microenvironment- induced regulation of Bcl-2 family members in multiple myeloma (MM): Therapeutic implications,” Cytokine 161 (2023): 156062.

[173]

L. Cai, Y. Wang, H. Chen, et al., “Platinum(IV) Complexes as Inhibitors of STAT3 and Regulators of the Tumor Microenvironment To Control Breast Cancer,” Journal of Medicinal Chemistry 66, no. 16 (2023): 11351-11364.

[174]

K. A. F. Pennel, P. Hatthakarnkul, C. S. Wood, et al., “JAK/STAT3 represents a therapeutic target for colorectal cancer patients With stromal-rich tumors,” Journal of Experimental & Clinical Cancer Research 43, no. 1 (2024): 1-20.

[175]

H. S. Tuli, K. Sak, A. Iqubal, et al., “STAT signaling as a target for intervention: From cancer inflammation and angiogenesis to non-coding RNAs modulation,” Molecular Biology Reports 49, no. 9 (2022): 8987-8999. 2022 499.

[176]

Y. Zhong, L. Deng, S. Shi, et al., “The novel STAT3 inhibitor WZ-2-033 causes regression of human triple-negative breast cancer and gastric cancer xenografts,” Acta Pharmacologica Sinica no. 4 (2021): 1013-1023. 2021 434.

[177]

M. Chen, T. Wang, D. Tian, C. Hai, and Z. Qiu, “Induction, growth, drug resistance, and metastasis: A comprehensive summary of the relationship Between STAT3 and gastric cancer,” Heliyon 10, no. 18 (2024).

[178]

D. Standing, E. Feess, S. Kodiyalam, et al., “The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy,” Cancers 2023, no. 9: 2485.

[179]

Y. Wang, J. Wang, C. Yang, et al., “A study of the correlation Between M2 macrophages and lymph node metastasis of colorectal carcinoma,” World Journal of Surgical Oncology 19, no. 1 (2021): 1-8.

[180]

A. N. Gargalionis, K. A. Papavassiliou, and A. G. Papavassiliou, “Targeting STAT3 Signaling Pathway in Colorectal Cancer,” Biomed 9, no. 8 (2021): 1016.

[181]

S. Shi, H. Y. Ma, and Z. G. Zhang, “Clinicopathological and prognostic value of STAT3/p-STAT3 in cervical cancer: A meta and bioinformatics analysis,” Pathology - Research and Practice 227 (2021): 153624.

[182]

Y. Zhang, W. Lu, Y. Chen, et al., “The miR-19b-3p-MAP2K3-STAT3 feedback loop regulates cell proliferation and invasion in esophageal squamous cell carcinoma,” Molecular Oncology 15, no. 5 (2021): 1566-1583.

[183]

Z. Qureshy, H. Li, Y. Zeng, et al., “STAT3 Activation as a Predictive Biomarker for Ruxolitinib Response in Head and Neck Cancer,” Clinical Cancer Research 28, no. 21 (2022): 4737-4746.

[184]

H. Y. Lim, P. S. Ong, L. Wang, et al., “Celastrol in cancer therapy: Recent developments, challenges and prospects,” Cancer Letters 521 (2021): 252-267.

[185]

Y. Kang, H. Li, Y. Liu, and Z. Li, “Regulation of VEGF-A expression and VEGF-A-targeted therapy in malignant tumors,” Journal of Cancer Research and Clinical Oncology 150, no. 5 (2024): 1-10.

[186]

M. Ashrafizadeh, C. D. Mohan, S. Rangappa, et al., “Noncoding RNAs as regulators of STAT3 pathway in gastrointestinal cancers: Roles in cancer progression and therapeutic response,” Medicinal Research Reviews 43, no. 5 (2023): 1263-1321.

[187]

B. Kumar, A. S. Khatpe, J. Guanglong, et al., “Stromal heterogeneity may explain increased incidence of metaplastic breast cancer in women of African descent,” Nature Communications 14, no. 1 (2023): 1-22.

[188]

L. E. Pascal, Y. Wang, M. Zhong, et al., “EAF2 and p53 Co-Regulate STAT3 Activation in Prostate Cancer,” Neoplasia 20, no. 4 (2018): 351-363.

[189]

W. Peng, H. Zhang, M. Yin, et al., “Combined Inhibition of PI3K and STAT3 signaling effectively inhibits bladder cancer growth,” Oncogenesis 13, no. 1 (2024): 1-12.

[190]

S. Singh, H. J. Gomez, S. Thakkar, S. P. Singh, and A. S. Parihar, “Overcoming Acquired Drug Resistance to Cancer Therapies Through Targeted STAT3 Inhibition,” International Journal of Molecular Sciences 24, no. 5 (2023): 4722.

[191]

F. Khan, P. Pandey, M. Verma, and T. K. Upadhyay, “Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies,” Biomol 14, no. 2 (2024): 200.

[192]

M. Sadrkhanloo, M. Entezari, S. Orouei, et al., “STAT3-EMT axis in tumors: Modulation of cancer metastasis, stemness and therapy response,” Pharmacological Research 182 (2022): 106311.

[193]

Z. Ma, F. Zhou, H. Jin, and X. Wu, “Crosstalk Between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway,” Cells 13, no. 12 (2024): 1027.

[194]

R. Y. Jiang, J. Y. Zhu, H. P. Zhang, et al., “STAT3: Key targets of growth-promoting receptor positive breast cancer,” Cancer Cell International 24, no. 1 (2024): 1-41.

[195]

Z. Kang, S. Li, Y. Li, J. Song, Y. Peng, and Y. Chen, “Small molecular inhibitors and degraders targeting STAT3 for cancer therapy: An updated review (From 2022 to 2024),” Chinese Chemical Letters 110447, published online September 11, 2024, https://doi.org/10.1016/J.CCLET.2024.110447.

[196]

T. Adesoye, D. Tripathy, K. K. Hunt, and K. Keyomarsi, “Exploring Novel Frontiers: Leveraging STAT3 Signaling for Advanced Cancer Therapeutics,” Cancers 16, no. 3 (2024): 492.

[197]

S. S. Yu, R. C. Tang, A. Zhang, et al., “Deacetylase Sirtuin 1 mitigates type I IFN- and type II IFN-induced signaling and antiviral immunity,” Journal of Virology 98, no. 3 (2024).

[198]

S. Hashimoto, A. Hashimoto, R. Muromoto, Y. Kitai, K. Oritani, and T. Matsuda, “Central Roles of STAT3-Mediated Signals in Onset and Development of Cancers: Tumorigenesis and Immunosurveillance,” Cells 11, no. 16 (2022): 2618.

[199]

X. Hu, J. li, M. Fu, X. Zhao, and W. Wang, “The JAK/STAT signaling pathway: From bench to clinic,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 1-33.

[200]

J. Xu, H. Lin, G. Wu, M. Zhu, and M. Li, “IL-6/STAT3 Is a Promising Therapeutic Target for Hepatocellular Carcinoma,” Frontiers in Oncology 11 (2021): 760971.

[201]

S. Rose-John, B. J. Jenkins, C. Garbers, J. M. Moll, and J. Scheller, “Targeting IL-6 trans-signalling: Past, present and future prospects,” Nature Reviews Immunology 23, no. 10 (2023): 666-681.

[202]

S. Natani, V. M. Dhople, A. Parveen, et al., “AMPK/SIRT1 signaling Through p38MAPK mediates Interleukin-6 induced neuroendocrine differentiation of LNCaP prostate cancer cells,” Biochimica et Biophysica Acta—Molecular Cell Research 1868, no. 10 (2021): 119085.

[203]

L. Puigdevall, C. Michiels, C. Stewardson, and L. Dumoutier, “JAK/STAT: Why choose a classical or an alternative pathway when you can have both?,” Journal of Cellular and Molecular Medicine 26, no. 7 (2022): 1865-1875.

[204]

S. Sudhesh Dev, S. A. Zainal Abidin, R. Farghadani, I. Othman, and R. Naidu, “Receptor Tyrosine Kinases and Their Signaling Pathways as Therapeutic Targets of Curcumin in Cancer,” Frontiers in Pharmacology 12 (2021): 772510.

[205]

S. Luo, S. Du, M. Tao, J. Cao, and P. Cheng, “Insights on hematopoietic cell kinase: An oncogenic player in human cancer,” Biomedicine & Pharmacotherapy 160 (2023): 114339.

[206]

M. Cao, Y. Wang, G. Lu, et al., “Classical Angiogenic Signaling Pathways and Novel Anti-Angiogenic Strategies for Colorectal Cancer,” Current Issues in Molecular Biology 44, no. 10 (2022): 4447-4471.

[207]

Z. Yang, X. Zhang, X. Bai, X. Xi, W. Liu, and W. Zhong, “Anti-angiogenesis in colorectal cancer therapy,” Cancer Science 115, no. 3 (2024): 734-751.

[208]

Q. B. Liu, R. H. Zhou, and C. M. Liu, “TLR9/FCRL3 regulates B cell viability, apoptosis, and antibody and IL-10 production Through ERK1/2, p38, and STAT3 signaling pathways,” In Vitro Cellular & Developmental Biology—Animal 58, no. 8 (2022): 702-711.

[209]

A. H. Rahmani, A. Almatroudi, K. S. Allemailem, et al., “Myricetin: A Significant Emphasis on Its Anticancer Potential via the Modulation of Inflammation and Signal Transduction Pathways,” International Journal of Molecular Sciences 24, no. 11 (2023): 9665.

[210]

M. Ashrafizadeh, M. H. Gholami, S. Mirzaei, et al., “Dual relationship Between long non-coding RNAs and STAT3 signaling in different cancers: New insight to proliferation and metastasis,” Life Sciences 270 (2021): 119006.

[211]

M. Sajjadi-Dokht, T. A. Merza Mohamad, H. Sulaiman Rahman, et al., “MicroRNAs and JAK/STAT3 signaling: A new promising therapeutic axis in blood cancers,” Genes & Diseases 9, no. 4 (2022): 849-867.

[212]

Y. L. Chen, C. C. Hsieh, P. M. Chu, J. Y. Chen, Y. C. Huang, and C. Y. Chen, “Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review),” Oncology Reports 49, no. 3 (2023).

[213]

Y. Lin, Z. Xiaohan, K. Yang, et al., “Protein tyrosine phosphatase receptor type D gene promotes radiosensitivity via STAT3 dephosphorylation in nasopharyngeal carcinoma,” Oncogene 40 (2021): 3101-3117.

[214]

K. A. Young, K. Wojdyla, T. Lai, et al., “The receptor protein tyrosine phosphatase PTPRK promotes intestinal repair and catalysis-independent tumor suppression,” Journal of Cell Science 137, no. 14 (2024).

[215]

X. Tang, X. Sui, and Y. Liu, “Immune checkpoint PTPN2 predicts prognosis and immunotherapy response in human cancers,” Heliyon 9, no. 1 (2023).

[216]

J. Song, J. Lan, J. Tang, and N. Luo, “PTPN2 in the Immunity and Tumor Immunotherapy: A Concise Review,” International Journal of Molecular Sciences 23, no. 17 (2022): 10025.

[217]

L. Dai, Z. Li, W. Liang, et al., “SOCS proteins and their roles in the development of glioblastoma,” Oncology Letters 23, no. 1 (2022).

[218]

H. Yoshikawa, K. Matsubara, G. S. Qian, et al., “SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity,” Nature Genetics 28 (2001): 29-35, Accessed May 4, 2024, http://genetics.nature.com.

[219]

M. Morelli, S. Madonna, and C. Albanesi, “SOCS1 and SOCS3 as key checkpoint molecules in the immune responses associated to skin inflammation and malignant transformation,” Frontiers in Immunology 15 (2024): 1393799.

[220]

W. Blaszczak, B. White, S. Monterisi, and P. Swietach, “Dynamic IL-6R/STAT3 signaling leads to heterogeneity of metabolic phenotype in pancreatic ductal adenocarcinoma cells,” Cell Reports 43, no. 1 (2024).

[221]

C. Yu, Y. Fan, Y. Zhang, L. Liu, and G. Guo, “LINC00893 inhibits the progression of prostate cancer Through miR-3173-5p/SOCS3/JAK2/STAT3 pathway,” Cancer Cell International 22, no. 1 (2022): 1-17.

[222]

L. Dai, Y. Han, Z. Yang, et al., “Identification and validation of SOCS1/2/3/4 as potential prognostic biomarkers and correlate With immune infiltration in glioblastoma,” Journal of Cellular and Molecular Medicine 27, no. 15 (2023): 2194-2214.

[223]

X. Li, A. Rasul, F. Sharif, and M. Hassan, “PIAS family in cancer: From basic mechanisms to clinical applications,” Frontiers in Oncology 14 (2024): 1376633.

[224]

Q. Ju, Q. Shi, C. Liu, G. Fu, and H. Shi, “Bufalin suppresses esophageal squamous cell carcinoma progression by activating the PIAS3/STAT3 signaling pathway,” Journal of Thoracic Disease 15, no. 4 (2023): 2141-2160.

[225]

M. Jiang, W. Zhang, R. Zhang, et al., “Cancer exosome-derived miR-9 and miR-181a promote the development of early-stage MDSCs via interfering With SOCS3 and PIAS3 respectively in breast cancer,” Oncogene 39 (2020): 4681-4694.

[226]

W. Bao, J. Wang, K. Fan, Y. Gao, and J. Chen, “PIAS3 promotes ferroptosis by regulating TXNIP via TGF-β signaling pathway in hepatocellular carcinoma,” Pharmacological Research 196 (2023): 106915.

[227]

Y. Fei, X. Zhang, X. Wang, et al., “Upregulation of tumor suppressor PIAS3 by Honokiol promotes tumor cell apoptosis via selective inhibition of STAT3 tyrosine 705 phosphorylation,” Journal of Natural Medicines 78, no. 2 (2024): 285-295.

[228]

H. Chen, W. Zhou, A. Bian, et al., “Selectively Targeting STAT3 Using a Small Molecule Inhibitor is a Potential Therapeutic Strategy for Pancreatic Cancer,” Clinical Cancer Research 29, no. 4 (2023): 815-830.

[229]

M. A. Kamal, Y. M. Mandour, M. K. Abd El-Aziz, U. Stein, and H. M. El Tayebi, “Small Molecule Inhibitors for Hepatocellular Carcinoma: Advances and Challenges,” Mol 27, no. 17 (2022): 5537.

[230]

S. Manoharan, A. Balakrishnan, V. Hemamalini, and E. Perumal, “Screening of potent STAT3-SH2 domain inhibitors From JAK/STAT compound library Through molecular dynamics simulation,” Molecular Diversity 27, no. 3 (2023): 1297-1308.

[231]

C. C. Zhang, T. Wu, L. Guan, et al., “Effects of STAT3 Inhibitor BP-1-102 on The Proliferation, Invasiveness, Apoptosis and Neurosphere Formation of Glioma Cells in Vitro,” Cell Biochemistry and Biophysics 80, no. 4 (2022): 723-735.

[232]

H. Guo, Y. Xiao, Z. Yuan, et al., “Inhibition of STAT3Y705 phosphorylation by Stattic suppresses proliferation and induces mitochondrial-dependent apoptosis in pancreatic cancer cells,” Cell Death Discovery 8, no. 1 (2022): 1-12.

[233]

S. Imbaby and Y. Hattori, “Stattic ameliorates the cecal ligation and puncture-induced cardiac injury in septic mice via IL-6-gp130-STAT3 signaling pathway,” Life Sciences 330 (2023): 122008.

[234]

L. Pan, X. Chen, F. V. Rassool, C. Li, and J. Lin, “LLL12B, a Novel Small-Molecule STAT3 Inhibitor, Induces Apoptosis and Suppresses Cell Migration and Tumor Growth in Triple-Negative Breast Cancer Cells,” Biomed 10, no. 8 (2022): 2003.

[235]

S. M. Zarezadeh, A. M. Sharafi, G. Erabi, et al., “Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review,” Recent Patents on Anti-Cancer Drug Discovery 19, no. 4 (2023): 403-502.

[236]

X. Zhang, T. Pang, H. Zhang, et al., “The natural compound periplogenin suppresses the growth of prostate carcinoma cells by directly targeting ATP1A1,” Science Reports 14, no. 1 (2024): 1-8.

[237]

P. He, Y. Miao, Y. Sun, et al., “Discovery of a Novel Potent STAT3 Inhibitor HP590 With Dual p-Tyr705/Ser727Inhibitory Activity for Gastric Cancer Treatment,” Journal of Medicinal Chemistry 65, no. 19 (2022): 12650-12674.

[238]

B. Biersack and M. Höpfner, “Emerging role of MYB transcription factors in cancer drug resistance,” Cancer Drug Resistance 7 (2024): 15.

[239]

X. Gu, T. Zhang, T. Liu, H. Cong, and W. Wu, “Cirsilineol inhibits the proliferation and migration of endometriotic cells,” Tropical Journal of Pharmaceutical Research 22, no. 12 (2023): 2427-2432.

[240]

V. R. Silva, L. S. de Santos, R. B. Dias, C. A. Quadros, and D. P. Bezerra, “Emerging agents that target signaling pathways to eradicate colorectal cancer stem cells,” Cancer Communications 41, no. 12 (2021): 1275-1313.

[241]

Q. Feng and K. Xiao, “Nanoparticle-Mediated Delivery of STAT3 Inhibitors in the Treatment of Lung Cancer,” Pharmaceutics 14, no. 12 (2022): 2787.

[242]

C. Hervieu, N. Christou, S. Battu, and M. Mathonnet, “The Role of Cancer Stem Cells in Colorectal Cancer: From the Basics to Novel Clinical Trials,” Cancers 13, no. 5 (2021): 1092.

[243]

M. Zhu, S. Li, X. Cao, K. Rashid, and T. Liu, “The STAT family: Key transcription factors mediating crosstalk Between cancer stem cells and tumor immune microenvironment,” Seminars in Cancer Biology 88 (2023): 18-31.

[244]

H. Flebbe, M. Spitzner, P. E. Marquet, et al., “Targeting STAT3 Signaling Facilitates Responsiveness of Pancreatic Cancer Cells to Chemoradiotherapy,” Cancers (Basel) 14, no. 5 (2022): 1301.

[245]

D. Pádua, P. Figueira, I. Ribeiro, R. Almeida, and P. Mesquita, “The Relevance of Transcription Factors in Gastric and Colorectal Cancer Stem Cells Identification and Eradication,” Frontiers in Cell and Developmental Biology 8 (2020): 544473.

[246]

S. Y. Ye, J. Y. Li, T. H. Li, et al., “The Efficacy and Safety of Celecoxib in Addition to Standard Cancer Therapy: A Systematic Review and Meta-Analysis of Randomized Controlled Trials,” Current Oncology Reports 29, no. 9 (2022): 6137-6153.

[247]

T. Hu, C. J. Liu, X. Yin, et al., “Selective COX-2 inhibitors do not increase gastrointestinal reactions After colorectal cancer surgery: A systematic review and meta-analysis,” BMC Gastroenterology [Electronic Resource] 23, no. 1 (2023): 1-9.

[248]

L. Chi, L. Huan, C. Zhang, H. Wang, and J. Lu, “Adenosine receptor A2b confers ovarian cancer survival and PARP inhibitor resistance Through IL-6-STAT3 signalling,” Journal of Cellular and Molecular Medicine 27, no. 15 (2023): 2150-2164.

[249]

M. Santoni, F. Miccini, A. Cimadamore, et al., “An update on investigational therapies that target STAT3 for the treatment of cancer,” Expert Opinion on Investigational Drugs 30, no. 3 (2021): 245-251.

[250]

I. Tošić and D. A. Frank, “STAT3 as a mediator of oncogenic cellular metabolism: Pathogenic and therapeutic implications,” Neoplasia 23, no. 12 (2021): 1167-1178.

[251]

Z. Hamel, S. Sanchez, D. Standing, and S. Anant, “Role of STAT3 in pancreatic cancer,” Exploration of Targeted Anti-tumor Therapy 5, no. 5 (2024): 20.

[252]

Z. X. Niu, Y. T. Wang, J. F. Sun, P. Nie, and P. Herdewijn, “Recent advance of clinically approved small-molecule drugs for the treatment of myeloid leukemia,” European Journal of Medicinal Chemistry 261 (2023): 115827.

[253]

J. Greene, A. Segaran, and S. Lord, “Targeting OXPHOS and the electron transport chain in cancer; Molecular and therapeutic implications,” Seminars in Cancer Biology 86 (2022): 851-859.

[254]

D. Liang, Q. Wang, W. Zhang, et al., “JAK/STAT in leukemia: A clinical update,” Molecular Cancer 23, no. 1 (2024): 1-12.

[255]

A. Virzì, A. A. R. Suarez, T. F. Baumert, and J. Lupberger, “Rewiring Host Signaling: Hepatitis C Virus in Liver Pathogenesis,” Cold Spring Harbor Perspectives in Medicine 10, no. 1 (2020): a037366.

[256]

H. Li, S. Ouyang, Y. Zhang, et al., “Structural optimization of Imidazo[1, 2-a]pyridine derivatives for the treatment of gastric cancer via STAT3 signaling pathway,” European Journal of Medicinal Chemistry 244 (2022): 114858.

[257]

Y. Li and Y. Dong, “TTI-101 targets STAT3/c-Myc signaling pathway to suppress cervical cancer progression: An integrated experimental and computational analysis,” Cancer Cell International 24, no. 1 (2024): 1-14.

[258]

D. J. Feith, J. Ung, O. Elghawy, P. Yue, J. Turkson, and T. P. Loughran, “Strategies for Targeting the JAK—STAT Pathway in Lymphoid Malignancies,” Precis Cancer Ther 1 (2023): 381-401.

[259]

S. Ramchandani, C. D. Mohan, J. R. Mistry, et al., “The multifaceted antineoplastic role of pyrimethamine Against human malignancies,” Iubmb Life 74, no. 3 (2022): 198-212.

[260]

S. Molenda, A. Sikorska, A. Florczak, and P. Lorenc, “Dams-Kozlowska H. Oligonucleotide-Based Therapeutics for STAT3 Targeting in Cancer—Drug Carriers Matter,” Cancers 15, no. 23 (2023): 5647.

[261]

S. Song, H. Tang, T. Ran, et al., “Application of deep generative model for design of Pyrrolo[2,3-d] pyrimidine derivatives as new selective TANK binding kinase 1 (TBK1) inhibitors,” European Journal of Medicinal Chemistry 247 (2023): 115034.

[262]

J. Liu; F.; Wang, F. Luo, J. Liu, F. Wang, and F. Luo, “The Role of JAK/STAT Pathway in Fibrotic Diseases: Molecular and Cellular Mechanisms,” Biomol 13, no. 1 (2023): 119.

[263]

Z. E. Walton, M. J. Frigault, and M. V. Maus, “Current and emerging pharmacotherapies for cytokine release syndrome, neurotoxicity, and hemophagocytic lymphohistiocytosis-Like syndrome due to CAR T cell therapy,” Expert Opinion on Pharmacotherapy 25, no. 3 (2024): 263-279.

[264]

C. Keenan, S. Albeituni, K. E. Nichols, and M. Hines, “JAK Inhibitors in Cytokine Storm Syndromes,” Advances in Experimental Medicine and Biology 1448 (2024): 583-600.

[265]

R. Roskoski, “Janus kinase (JAK) inhibitors in the treatment of neoplastic and inflammatory disorders,” Pharmacological Research 183 (2022): 106362.

[266]

H. Zhang, F. He, G. Gao, et al., “Approved Small-Molecule ATP-Competitive Kinases Drugs Containing Indole/Azaindole/Oxindole Scaffolds: R&D and Binding Patterns Profiling,” Mol 28, no. 3 (2023): 943.

[267]

P. Shen, Y. Wang, X. Jia, et al., “Dual-target Janus kinase (JAK) inhibitors: Comprehensive review on the JAK-based strategies for treating solid or hematological malignancies and immune-related diseases,” European Journal of Medicinal Chemistry 239 (2022): 114551.

[268]

X. H. Wei and Y. Y. Liu, “Potential applications of JAK inhibitors, clinically approved drugs Against autoimmune diseases, in cancer therapy,” Frontiers in Pharmacology 14 (2023): 1326281.

[269]

K. Gerlach, K. Lechner, V. Popp, et al., “The JAK1/3 Inhibitor to Tofacitinib Suppresses T Cell Homing and Activation in Chronic Intestinal Inflammation,” J Crohn's Colitis 15, no. 2 (2021): 244-257.

[270]

M. Lensing and A. Jabbari, “An overview of JAK/STAT pathways and JAK inhibition in alopecia areata,” Frontiers in Immunology 13 (2022): 955035.

[271]

P. Beinhoff, L. Sabharwal, V. Udhane, et al., “Second-generation jak2 inhibitors for advanced prostate cancer: Are we ready for clinical development?,” Cancers (Basel) 13, no. 20 (2021): 5204.

[272]

Y. Zhao, X. Zhang, X. Ding, et al., “Efficacy and safety of FLT3 inhibitors in monotherapy of hematological and solid malignancies: A systemic analysis of clinical trials,” Frontiers in Pharmacology 15 (2024): 1294668.

[273]

A. B. Griso, L. Acero-Riaguas, and B. Castelo, “Cebrián-Carretero JL, Sastre-Perona A. Mechanisms of Cisplatin Resistance in HPV Negative Head and Neck Squamous Cell Carcinomas,” Cells 11, no. 3 (2022): 561.

[274]

K. Okuyama, T. Naruse, and S. Yanamoto, “Tumor microenvironmental modification by the current target therapy for head and neck squamous cell carcinoma,” Journal of Experimental & Clinical Cancer Research 42, no. 1 (2023): 1-14.

[275]

S. K. Padda, K. L. Reckamp, M. Koczywas, et al., “A phase 1b study of erlotinib and momelotinib for the treatment of EGFR-mutated, tyrosine kinase inhibitor-naive metastatic non-small cell lung cancer,” Cancer Chemotheraphy and Pharmacology 89, no. 1 (2022): 105-115.

[276]

N. Mahadik, D. Bhattacharya, A. Padmanabhan, K. Sakhare, K. P. Narayan, and R. Banerjee, “Targeting steroid hormone receptors for anti-cancer therapy—A review on small molecules and nanotherapeutic approaches,” Wiley Interdiscip Rev Nanomedicine Nanobiotechnology 14, no. 2 (2022): e1755.

[277]

V. АZolottsev, АS. Latysheva, V. S. Pokrovsky, I. I. Khan, and A. Y. Misharin, “Promising applications of steroid сonjugates for cancer research and treatment,” European Journal of Medicinal Chemistry 210 (2021): 113089.

[278]

Y. Yue, Y. Lou, X. Liu, and X. Peng, “Vasculogenic mimicry in head and neck tumors: A narrative review,” Translational Cancer Research 10, no. 6 (2021): 3044.

[279]

Y. W. Guo, L. Zhu, Y. T. Duan, et al., “Ruxolitinib induces apoptosis and pyroptosis of anaplastic thyroid cancer via the transcriptional inhibition of DRP1-mediated mitochondrial fission,” Cell Death & Disease 15, no. 2 (2024): 1-18.

[280]

K. H. T. Dao, J. Gotlib, M. M. N. Deininger, et al., “Efficacy of Ruxolitinib in Patients With Chronic Neutrophilic Leukemia and Atypical Chronic Myeloid Leukemia,” Journal of Clinical Oncology 38, no. 10 (2020): 1006.

[281]

J. Mascarenhas, “Pacritinib for the treatment of patients With myelofibrosis and thrombocytopenia,” Expert Review of Hematology 15, no. 8 (2022): 671-684.

[282]

Y. Yang, Y. Mou, L. X. Wan, et al., “Rethinking therapeutic strategies of dual-target drugs: An update on pharmacological small-molecule compounds in cancer,” Medicinal Research Reviews 44, no. 6 (2024): 2600-2623.

[283]

P. Wu, J. Zhou, Y. Wu, and L. Zhao, “The emerging role of Interleukin 37 in bone homeostasis and inflammatory bone diseases,” International Immunopharmacology 98 (2021): 107803.

[284]

J. E. Lopes, J. L. Fisher, H. L. Flick, et al., “ALKS 4230: A novel engineered IL-2 fusion protein With an improved cellular selectivity profile for cancer immunotherapy,” Journal for ImmunoTherapy of Cancer 8, no. 1 (2020): e000673.

[285]

M. L. T. Nguyen, K. C. Bui, T. Scholta, et al., “Targeting interleukin 6 signaling by monoclonal antibody siltuximab on cholangiocarcinoma,” Journal of Gastroenterology and Hepatology 36, no. 5 (2021): 1334-1345.

[286]

L. Kruk, M. Mamtimin, A. Braun, et al., “Inflammatory Networks in Renal Cell Carcinoma,” Cancers 15, no. 8 (2023): 2212.

[287]

F. van Rhee, A. Rosenthal, K. Kanhai, et al., “Siltuximab is associated With improved progression-free survival in idiopathic multicentric Castleman disease,” Blood Advances 6, no. 16 (2022): 4773-4781.

[288]

R. Tamura, K. Yoshihara, and T. Enomoto, “Therapeutic Strategies Focused on Cancer-Associated Hypercoagulation for Ovarian Clear Cell Carcinoma,” Cancers 14, no. 9 (2022): 2125.

[289]

H. M. Lee, H. J. Lee, and J. E. Chang, “Inflammatory Cytokine: An Attractive Target for Cancer Treatment,” Biomed 10, no. 9 (2022): 2116.

[290]

S. Parakh, M. Ernst, and A. R. Poh, “Multicellular Effects of STAT3 in Non-small Cell Lung Cancer: Mechanistic Insights and Therapeutic Opportunities,” Cancers 13, no. 24 (2021): 6228.

[291]

C. Ebersbach, A. M. K. Beier, C. Thomas, and H. H. H. Erb, “Impact of STAT Proteins in Tumor Progress and Therapy Resistance in Advanced and Metastasized Prostate Cancer,” Cancers 13, no. 19 (2021): 4854.

[292]

Y. Wang and Y. Zhang, “Prognostic role of interleukin-6 in renal cell carcinoma: A meta-analysis,” Clinical & Translational Oncology 22, no. 6 (2020): 835-843.

[293]

J. Kaur, P. Singh, T. Enzler, and V. Sahai, “Emerging antibody therapies for pancreatic adenocarcinoma: A review of recent phase 2 trials,” Expert Opinion on Emerging Drugs 26, no. 2 (2021): 103-129.

[294]

M. Narazaki and T. Kishimoto, “Current status and prospects of IL-6-targeting therapy,” Expert Review of Clinical Pharmacology 15, no. 5 (2022): 575-592.

[295]

A. B. Avci, E. Feist, and G. R. Burmester, “Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What Have We Learned?,” Biodrugs 38, no. 1 (2024): 61.

[296]

K. Panuciak, M. Margas, K. Makowska, and M. Lejman, “Insights Into Modern Therapeutic Approaches in Pediatric Acute Leukemias,” Cells 11, no. 1 (2022): 139.

[297]

I. M. Chen, M. Donia, C. A. Chamberlain, et al., “Phase 2 study of ipilimumab, nivolumab, and tocilizumab combined With stereotactic body radiotherapy in patients With refractory pancreatic cancer (TRIPLE-R),” European Journal of Cancer 180 (2023): 125-133.

[298]

M. Li, S. Li, R. Zhao, et al., “CD318 is a target of chimeric antigen receptor T cells for the treatment of colorectal cancer,” Clinical and Experimental Medicine 23, no. 6 (2023): 2409-2419.

[299]

M. Choi, J. Shin, C. E. Lee, et al., “Immunogenic cell death in cancer immunotherapy,” BMB Reports 56, no. 5 (2023): 275-286.

[300]

C. Liu, J. Zhou, S. Zhang, et al., “Mesenchymal stem cells-derived IL-6 promotes invasion and metastasis of oral squamous cell carcinoma via JAK-STAT3 signalling,” Oral Diseases 30, no. 4 (2024): 2097-2109.

[301]

M. Markouli, F. Ullah, N. Omar, et al., “Recent Advances in Adult Post-Transplant Lymphoproliferative Disorder,” Cancers 14, no. 23 (2022): 5949.

[302]

P. Ercilla-Rodríguez, M. Sánchez-Díez, N. Alegría-Aravena, et al., “CAR-T lymphocyte-based cell therapies; mechanistic substantiation, applications and biosafety enhancement With suicide genes: New opportunities to melt side effects,” Frontiers in Immunology 15 (2024): 1333150.

[303]

A. Das, K. J. Lavanya, Nandini, K. Kaur, and V. Jaitak, “Effectiveness of Selective Estrogen Receptor Modulators in Breast Cancer Therapy: An Update,” Current Medicinal Chemistry 30, no. 29 (2022): 3287-3314.

[304]

E. Zafar, M. F. Maqbool, A. Iqbal, et al., “A comprehensive review on anticancer mechanism of bazedoxifene,” Biotechnology and Applied Biochemistry 69, no. 2 (2022): 767-782.

[305]

C. Shi, T. Bopp, H. W. Lo, K. Tkaczuk, and J. Lin, “Bazedoxifene as a Potential Cancer Therapeutic Agent Targeting IL-6/GP130 Signaling,” Current Oncology 31, no. 10 (2024): 5737-5751.

[306]

Q. Huang, Y. Zhong, H. Dong, et al., “Revisiting signal transducer and activator of transcription 3 (STAT3) as an anticancer target and its inhibitor discovery: Where are we and where should we go?,” European Journal of Medicinal Chemistry 187 (2020): 111922.

[307]

K. Taniguchi, M. Tsugane, and A. Asai, “A Brief Update on STAT3 Signaling: Current Challenges and Future Directions in Cancer Treatment,” Journal of Cell Communication and Signaling 2, no. 3 (2021): 181-194.

[308]

K. N. Wang, K. Zhou, N. N. Zhong, et al., “Enhancing cancer therapy: The role of drug delivery systems in STAT3 inhibitor efficacy and safety,” Life Sciences 346 (2024): 122635.

[309]

R. Mikyskova, O. Sapega, M. Psotka, et al., “STAT3 inhibitor Stattic and its analogues inhibit STAT3 phosphorylation and modulate cytokine secretion in senescent tumour cells,” Molecular Medicine Reports 27, no. 4 (2023).

[310]

G. D. Qian, J. Xu, X. X. Shen, et al., “BP-1-102 and silencing of Fascin-1 by RNA interference inhibits the proliferation of mouse pituitary adenoma AtT20 cells via the signal transducer and activator of transcription 3/fascin-1 pathway,” International Journal of Neuroscience 131, no. 8 (2021): 810-827.

[311]

W. Liu, Z. Chu, C. Yang, et al., “Discovery of potent STAT3 inhibitors using structure-based virtual screening, molecular dynamic simulation, and biological evaluation,” Frontiers in Oncology 13 (2023): 1287797.

[312]

X. Tan, X. Ma, Y. Dai, et al., “A large-scale transcriptional analysis reveals herb-derived ginsenoside F2 suppressing hepatocellular carcinoma via inhibiting STAT3,” Phytomedicine 120 (2023): 155031.

[313]

A. Ahamed, M. Hasan, A. Samanta, et al., “Prospective pharmacological potential of cryptotanshinone in cancer therapy,” Pharmacological Research - Modern Chinese Medicine 9 (2023): 100308.

[314]

K. Zhao, Q. Zhao, X. Dai, et al., “Alantolactone enhances the sensitivity of melanoma to MAPK pathway inhibitors by targeting inhibition of STAT3 activation and Down-regulating stem cell markers,” Cancer Cell International 24, no. 1 (2024): 1-14.

[315]

L. Long, X. Fei, L. Chen, L. Yao, and X. Lei, “Potential therapeutic targets of the JAK2/STAT3 signaling pathway in triple-negative breast cancer,” Frontiers in Oncology 14 (2024): 1381251.

[316]

S. Verdura, E. Cuyàs, V. Ruiz-Torres, et al., “Lung cancer management With silibinin: A historical and translational perspective,” Pharmaceuticals 14, no. 6 (2021): 559.

[317]

V. Damerell, M. S. Pepper, and S. Prince, “Molecular mechanisms underpinning sarcomas and implications for current and future therapy,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 1-19.

[318]

P. S. Thilakasiri, R. S. Dmello, T. L. Nero, M. W. Parker, M. Ernst, and A. L. Chand, “Repurposing of drugs as STAT3 inhibitors for cancer therapy,” Seminars in Cancer Biology 68 (2021): 31-46.

[319]

Y. Chen, N. Zhai, Y. Zhu, et al., “Azetidine ring, salicylic acid, and salicylic acid bioisosteres as determinants of the binding characteristics of novel potent compounds to Stat3,” Bioorganic & Medicinal Chemistry Letters 97 (2024): 129565.

[320]

A. Caruso, A. Barbarossa, A. Carocci, G. Salzano, M. S. Sinicropi, and C. Saturnino, “Carbazole Derivatives as STAT Inhibitors: An Overview,” Applied Science Letters 11, no. 13 (2021): 6192.

[321]

N. Deravi and N. Rezaei. Signal Transducer and Activator of Transcription as a Potential Therapeutic Target in Breast Cancer, published online 2023: 1-26, https://doi.org/10.1007/16833_2022_107.

[322]

F. Shao, X. Pang, and G. H. Baeg, “Targeting the JAK/STAT Signaling Pathway for Breast Cancer,” Current Medicinal Chemistry 28, no. 25 (2020): 5137-5151.

[323]

P. Yue, Y. Zhu, C. Brotherton-Pleiss, et al., “Novel potent azetidine-based compounds irreversibly inhibit Stat3 activation and induce antitumor response Against human breast tumor growth in vivo,” Cancer Letters 534 (2022): 215613.

[324]

M. A. Thalappil, Targeting STAT3 signaling with essential oils: a potential strategy for adjuvant cancer therapy, published online 2023, accessed November 18, 2024. https://iris.univr.it/handle/11562/1099367.

[325]

E. Khatoon, M. Hegde, A. Kumar, et al., “The multifaceted role of STAT3 pathway and its implication as a potential therapeutic target in oral cancer,” Archives of Pharmacal Research 45, no. 8 (2022): 507-534.

[326]

E. L. Morgan and A. Macdonald, “Manipulation of JAK/STAT Signalling by High-Risk HPVs: Potential Therapeutic Targets for HPV-Associated Malignancies,” Viruses. 12, no. 9 (2020): 977.

[327]

J. Xu, J. Zhang, Q. F. Mao, J. Wu, and Y. Wang, “The Interaction Between Autophagy and JAK/STAT3 Signaling Pathway in Tumors,” Frontiers in Genetics 13 (2022).

[328]

D. P. McLornan, J. E. Pope, J. Gotlib, and C. N. Harrison, “Current and future status of JAK inhibitors,” Lancet 398, no. 10302 (2021): 803-816.

[329]

B. Rah, R. A. Rather, G. R. Bhat, et al., “JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies,” Frontiers in Pharmacology 13 (2022): 821344.

[330]

N. R. Jabir, C. K. Firoz, M. A. Kamal, et al., “Assessment of genetic diversity in IL-6 and RANTES promoters and their level in Saudi coronary artery disease patients,” Journal of Clinical Laboratory Analysis 31, no. 5 (2017): e22092.

[331]

D. Aletaha, A. Kerschbaumer, K. Kastrati, et al., “Consensus statement on blocking interleukin-6 receptor and interleukin-6 in inflammatory conditions: An update,” Annals of the Rheumatic Diseases 82, no. 6 (2023): 773-787.

[332]

P. L. Yang, L. X. Liu, E. M. Li, and L. Y. Xu, “STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy,” Cancers 12, no. 9 (2020): 2459.

[333]

Y. Jiang, L. Liu, Y. Geng, et al., “Feasibility of the inhibitor development for cancer: A systematic approach for drug design,” PLoS ONE 19, no. 8 (2024): e0306632.

[334]

X. Yang, L. Xu, L. Yang, and S. Xu, “Research progress of STAT3-based dual inhibitors for cancer therapy,” Bioorganic & Medicinal Chemistry 91 (2023): 117382.

[335]

T. W. Kim, Y. Kim, H. Keum, W. Jung, M. Kang, and S. Jon, “Combination of a STAT3 inhibitor With anti-PD-1 immunotherapy is an effective treatment regimen for a vemurafenib-resistant melanoma,” Molecular Therapy Oncolytics 26 (2022): 1-14.

[336]

Y. Zhu, M. Chen, D. Xu, et al., “The combination of PD-1 blockade With interferon-α has a synergistic effect on hepatocellular carcinoma,” Cellular & Molecular Immunology 19, no. 6 (2022): 726-737.

[337]

M. Markouli, D. Strepkos, and C. Piperi, “RETRACTED: Impact of Histone Modifications and Their Therapeutic Targeting in Hematological Malignancies,” International Journal of Molecular Sciences 23, no. 21 (2022): 13657.

[338]

X. Chen, L. Pan, J. Wei, et al., “LLL12B, a small molecule STAT3 inhibitor, induces growth arrest, apoptosis, and enhances cisplatin-mediated cytotoxicity in medulloblastoma cells,” Science Reports 11, no. 1 (2021): 1-14.

[339]

S. H. Lee, C. X. Ng, S. R. Wong, and P. P. Chong, “MiRNAs Overexpression and Their Role in Breast Cancer: Implications for Cancer Therapeutics,” Current Drug Targets 24, no. 6 (2023): 484-508.

[340]

A. J. Oweida, L. Darragh, A. Phan, et al., “STAT3 Modulation of Regulatory T Cells in Response to Radiation Therapy in Head and Neck Cancer,” Journal of the National Cancer Institute 111, no. 12 (2019): 1339-1349.

[341]

Y. Ni, J. T. Low, J. Silke, and L. A. O'Reilly, “Digesting the Role of JAK-STAT and Cytokine Signaling in Oral and Gastric Cancers,” Frontiers in Immunology 13 (2022): 835997.

[342]

W. Wang, M. C. Lopez McDonald, R. Hariprasad, T. Hamilton, and D. A. Frank, “Oncogenic STAT Transcription Factors as Targets for Cancer Therapy: Innovative Strategies and Clinical Translation,” Cancers 16, no. 7 (2024): 1387.

[343]

T. Hu, R. Shi, Y. Gu, et al., “Cancer-derived non-coding RNAs endow tumor microenvironment With immunosuppressive properties,” Wiley Interdisciplinary Reviews RNA 15, no. 1 (2024): e1822.

[344]

C. J. Chen, H. C. Wang, Y. Y. Wu, C. C. Shieh, and Y. S. Shan, A New Gene Therapy for Pancreatic Cancer: STAT6-CYBB Decoy Oligodeoxynucleotides Focusing on Inhibiting M2 Macrophages, published online May 31, 2022, https://doi.org/10.21203/RS.3.RS-1690410/V1.

[345]

M. Jiang and B. Li, “STAT3 and Its Targeting Inhibitors in Oral Squamous Cell Carcinoma,” Cells 11, no. 19 (2022): 3131.

[346]

Y. Pan, J. Cheng, Y. Zhu, J. Zhang, W. Fan, and X. Chen, “Immunological nanomaterials to combat cancer metastasis,” Chemical Society Reviews 53, no. 12 (2024): 6399-6444.

[347]

M. Corte-Real, F. Veiga, A. C. Paiva-Santos, and P. C. Pires, “Improving Skin Cancer Treatment by Dual Drug Co-Encapsulation Into Liposomal Systems—An Integrated Approach towards Anticancer Synergism and Targeted Delivery,” Pharmaceutics 16, no. 9 (2024): 1200.

[348]

A. S. Widjaya, Y. Liu, Y. Yang, W. Yin, J. Liang, and Y. Jiang, “Tumor-permeable smart liposomes by modulating the tumor microenvironment to improve the chemotherapy,” Journal of Controlled Release 344 (2022): 62-79.

[349]

G. Basirinia, M. Ali, A. Comelli, et al., “Theranostic Approaches for Gastric Cancer: An Overview of In Vitro and In Vivo Investigations,” Cancers 16, no. 19 (2024): 3323.

[350]

S. Kumari, P. K. Choudhary, R. Shukla, A. Sahebkar, and P. Kesharwani, “Recent advances in nanotechnology based combination drug therapy for skin cancer,” Journal of Biomaterials Science, Polymer Edition 33, no. 11 (2022): 1435-1468.

[351]

Y. Jiang, C. Yan, M. Li, et al., “Delivery of natural products via polysaccharide-based nanocarriers for cancer therapy: A review on recent advances and future challenges,” International Journal of Biological Macromolecules 278 (2024): 135072.

[352]

S. Abdullah, F. Goher, and A. N. Awan, Nanoparticles: A Treatment Modality for Lung Cancer, published online 2024: 139-159, https://doi.org/10.1007/16833_2024_306.

[353]

Y. Dong, J. Chen, Y. Chen, and S. Liu, “Targeting the STAT3 oncogenic pathway: Cancer immunotherapy and drug repurposing,” Biomedicine & Pharmacotherapy 167 (2023): 115513.

[354]

R. Bellavita, S. Braccia, A. Falanga, and S. Galdiero, “An Overview of Supramolecular Platforms Boosting Drug Delivery,” Bioinorganic Chemistry and Applications 2023, no. 1 (2023): 8608428.

[355]

M. R. Tavares, K. Hrabánková, R. Konefał, et al., “HPMA-Based Copolymers Carrying STAT3 Inhibitor Cucurbitacin-D as Stimulus-Sensitive Nanomedicines for Oncotherapy,” Pharmaceutics 13, no. 2 (2021): 179.

[356]

T. Hu, H. Gong, J. Xu, Y. Huang, F. Wu, and Z. He, “Nanomedicines for Overcoming Cancer Drug Resistance,” Pharmaceutics 14, no. 8 (2022): 1606.

[357]

Y. Sun, M. Li, M. Zheng, Y. Zou, and B. Shi, “Blood-brain barrier penetrating nanosystems enable synergistic therapy of glioblastoma,” Nano Today 56 (2024): 102310.

[358]

N. Bie, T. Yong, Z. Wei, L. Gan, and X. Yang, “Extracellular vesicles for improved tumor accumulation and penetration,” Advanced Drug Delivery Reviews 188 (2022): 114450.

[359]

W. Ngamcherdtrakul, M. Reda, M. A. Nelson, et al., “In Situ Tumor Vaccination With Nanoparticle Co-Delivering CpG and STAT3 siRNA to Effectively Induce Whole-Body Antitumor Immune Response,” Advanced Materials 33, no. 31 (2021): 2100628.

[360]

L. Yang, Q. Hu, and T. Huang, “Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert “Cold” Tumors to “Hot” Tumors,” International Journal of Molecular Sciences 25, no. 13 (2024): 7208.

[361]

F. Della Sala, A. Fabozzi, M. di Gennaro, et al., “Advances in Hyaluronic-Acid-Based (Nano)Devices for Cancer Therapy,” Macromolecular Bioscience 22, no. 1 (2022): 2100304.

[362]

S. Karthik, S. Mohan, I. Magesh, et al., “Chitosan nanocarriers for non-coding RNA therapeutics: A review,” International Journal of Biological Macromolecules 263 (2024): 130361.

[363]

A. Ahmad, S. Rashid, A. A. Chaudhary, et al., “Nanomedicine as potential cancer therapy via targeting dysregulated transcription factors,” Seminars in Cancer Biology 89 (2023): 38-60.

[364]

K. Thapa Magar, G. F. Boafo, X. Li, Z. Chen, and W. He, “Liposome-based delivery of biological drugs,” Chinese Chemical Letters 33, no. 2 (2022): 587-596.

[365]

J. Zhao, J. Yang, J. Jiao, X. Wang, Y. Zhao, and L. Zhang, “Biomedical applications of artificial exosomes for intranasal drug delivery,” Frontiers in Bioengineering and Biotechnology 11 (2023): 1271489.

[366]

B. You, C. Jin, J. Zhang, et al., “MSC-Derived Extracellular Vesicle-Delivered L-PGDS Inhibit Gastric Cancer Progression by Suppressing Cancer Cell Stemness and STAT3 Phosphorylation,” Stem Cells International 2022 (2022).

[367]

S. F. Liang, F. F. Zuo, B. C. Yin, and B. C. Ye, “Delivery of siRNA based on engineered exosomes for glioblastoma therapy by targeting STAT3,” Biomaterials Science 10, no. 6 (2022): 1582-1590.

[368]

T. Chen, B. Ma, S. Lu, et al., “Cucumber-Derived Nanovesicles Containing Cucurbitacin B for Non-Small Cell Lung Cancer Therapy,” International Journal of Nanomedicine 17 (2022): 3583-3599.

[369]

I. Ahmad, S. Ahmad, A. Ahmad, T. A. Zughaibi, M. Alhosin, and S. Tabrez, “Curcumin, its derivatives, and their nanoformulations: Revolutionizing cancer treatment,” Cell Biochemistry and Function 42, no. 1 (2024).

[370]

D. Guimarães, A. Cavaco-Paulo, and E. Nogueira, “Design of liposomes as drug delivery system for therapeutic applications,” International Journal of Pharmaceutics 601 (2021): 120571.

[371]

P. Liu, G. Chen, and J. Zhang, “A Review of Liposomes as a Drug Delivery System: Current Status of Approved Products, Regulatory Environments, and Future Perspectives,” Mol 27, no. 4 (2022): 1372.

[372]

K. Shi, J. Xue, Y. Fang, et al., “Inorganic Kernel-Reconstituted Lipoprotein Biomimetic Nanovehicles Enable Efficient Targeting “Trojan Horse” Delivery of STAT3-Decoy Oligonucleotide for Overcoming TRAIL Resistance,” Theranostics 7, no. 18 (2017): 4480-4497.

[373]

K. Veselá, Z. Kejík, M. Masařík, et al., “Curcumin: A Potential Weapon in the Prevention and Treatment of Head and Neck Cancer,” ACS Pharmacology & Translational Science 7, no. 11 (2024): 3394-3418.

[374]

F. Zahedipour, M. Bolourinezhad, Y. Teng, and A. Sahebkar, “The Multifaceted Therapeutic Mechanisms of Curcumin in Osteosarcoma: State-of-the-Art,” Journal of Oncology 2021, no. 1 (2021): 3006853.

[375]

T. Ikeda, M. Kawabori, Y. Zheng, et al., “Intranasal Administration of Mesenchymal Stem Cell-Derived Exosome Alleviates Hypoxic-Ischemic Brain Injury,” Pharmaceutics 16, no. 4 (2024): 446.

[376]

H. Zheng, Z. Chen, A. Cai, et al., “Nanoparticle mediated codelivery of nifuratel and doxorubicin for synergistic anticancer therapy Through STAT3 inhibition,” Colloids Surfaces B Biointerfaces 193 (2020): 111109.

[377]

Z. Li, G. Chen, L. Ding, et al., “Increased Survival by Pulmonary Treatment of Established Lung Metastases With Dual STAT3/CXCR4 Inhibition by siRNA Nanoemulsions,” Molecular Therapy 27, no. 12 (2019): 2100-2110.

[378]

H. Tabasi, S. Mollazadeh, E. Fazeli, et al., “Transitional Insight Into the RNA-Based Oligonucleotides in Cancer Treatment,” Applied Biochemistry and Biotechnology 196, no. 3 (2023): 1685-1711.

[379]

N. O. Alafaleq, T. A. Zughaibi, N. R. Jabir, A. U. Khan, M. S. Khan, and S. Tabrez, “Biogenic Synthesis of Cu-Mn Bimetallic Nanoparticles Using Pumpkin Seeds Extract and Their Characterization and Anticancer Efficacy,” Nanomater (Basel, Switzerland) 13, no. 7 (2023).

[380]

T. A. Zughaibi, N. R. Jabir, A. U. Khan, M. S. Khan, and S. Tabrez, “Screening of Cu4O3 NPs efficacy and its anticancer potential Against cervical cancer,” Cell Biochemistry and Function 41, no. 8 (2023): 1174-1187.

[381]

M. Rashid Khan, N. Omar Alafaleq, A. Kumar Ramu, et al., “Evaluation of biogenically synthesized MgO NPs anticancer activity Against breast cancer cells,” Saudi Journal of Biological Sciences 31, no. 1 (2024).

[382]

L. Sun, Z. Li, J. Lan, Y. Wu, T. Zhang, and Y. Ding, “Better together: Nanoscale co-delivery systems of therapeutic agents for high-performance cancer therapy,” Frontiers in Pharmacology 15 (2024): 1389922.

[383]

N. Pore, S. Wu, N. Standifer, et al., “Resistance to durvalumab and durvalumab plus tremelimumab is associated With functional STK11 mutations in patients With non-small cell lung cancer and is reversed by STAT3 knockdown,” Cancer Discovery 11, no. 11 (2021): 2828-2845.

[384]

D. Mukherjee and S. Raikwar, “Recent Update on Nanocarrier(s) as the Targeted Therapy for Breast Cancer,” Aaps Pharmscitech [Electronic Resource] 25, no. 6 (2024): 1-25.

[385]

M. I. Khan, M. I. Hossain, M. K. Hossain, et al., “Recent Progress in Nanostructured Smart Drug Delivery Systems for Cancer Therapy: A Review,” ACS Applied Bio Materials 5, no. 3 (2022): 971-1012.

[386]

Y. Zhang, Y. Wu, H. Du, et al., “Nano-Drug Delivery Systems in Oral Cancer Therapy: Recent Developments and Prospective,” Pharmaceutics 16, no. 1 (2023): 7.

[387]

K. K. Chenab, H. Malektaj, A. A. R. Nadinlooie, S. Mohammadi, and M. R. Zamani-Meymian, “Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials,” Microchimica Acta 191, no. 9 (2024): 1-51.

[388]

B. Yang, F. Meng, J. Zhang, et al., “Engineered drug delivery nanosystems for tumor microenvironment normalization therapy,” Nano Today 49 (2023): 101766.

[389]

C. Pacheco, A. Baião, T. Ding, W. Cui, and B. Sarmento, “Recent advances in long-acting drug delivery systems for anticancer drug,” Advanced Drug Delivery Reviews 194 (2023): 114724.

[390]

H. Xie, Z. Chen, N. Zhang, et al., “A review of recent advances in the stability, efficacy, and biosafety of black phosphorus-based drug delivery,” Journal of Materials Science 59, no. 27 (2024): 12129-12153.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

9

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/