Transforming Growth Factor-Beta Signaling in Cancer: Therapeutic Implications, Challenges, and Pathways to Progress

Faizah A. Alabi , Izuchukwu F. Okpalanwaka , Adenike Oyegbesan , Ebenezar Okoyeocha , Mariam Oladejo , Sikiru O. Imodoye

MEDCOMM - Oncology ›› 2025, Vol. 4 ›› Issue (3) : e70027

PDF
MEDCOMM - Oncology ›› 2025, Vol. 4 ›› Issue (3) : e70027 DOI: 10.1002/mog2.70027
REVIEW ARTICLE

Transforming Growth Factor-Beta Signaling in Cancer: Therapeutic Implications, Challenges, and Pathways to Progress

Author information +
History +
PDF

Abstract

The transforming growth factor-β (TGF-β) family consists of evolutionarily conserved cytokines that regulate various physiological processes across nearly all tissue and cell types. While TGF-β signaling plays a critical role in immune homeostasis and tissue repair, its dysregulation is implicated in multiple diseases, particularly cancer. Given its dual role in tumor suppression and promotion, TGF-β has emerged as a promising yet challenging therapeutic target. Preclinical studies have demonstrated significant tumor inhibition through TGF-β signaling blockade using diverse classes of inhibitors. However, despite extensive research and clinical trials spanning over two decades, no TGF-β inhibitors have been approved for cancer therapy, underscoring a significant disconnect between preclinical promise and clinical efficacy. This review systematically examines the tumorigenic mechanisms driven by TGF-β and evaluates the therapeutic landscape of anti-TGF-β inhibitors, including receptor kinase inhibitors, neutralizing antibodies, bifunctional ligand traps, integrin-mediated TGF-β therapy, antisense oligonucleotides, TGF-β-targeted vaccines, and various combination strategies. By comparing preclinical and clinical findings, we highlight key challenges and propose novel approaches to improve the translational success of TGF-β-targeted therapies. These insights provide a foundation for optimizing future research and advancing the clinical application of TGF-β inhibitors in oncology.

Keywords

cancer therapy / immunotherapy / targeted therapy / transforming growth factor-beta (TGF-β) / tumor microenvironment

Cite this article

Download citation ▾
Faizah A. Alabi, Izuchukwu F. Okpalanwaka, Adenike Oyegbesan, Ebenezar Okoyeocha, Mariam Oladejo, Sikiru O. Imodoye. Transforming Growth Factor-Beta Signaling in Cancer: Therapeutic Implications, Challenges, and Pathways to Progress. MEDCOMM - Oncology, 2025, 4(3): e70027 DOI:10.1002/mog2.70027

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

H. L. Moses, A. B. Roberts, and R. Derynck, “The Discovery and Early Days of TGF-β: A Historical Perspective,” Cold Spring Harbor Perspectives in Biology 8, no. 7 (2016): a021865.

[2]

A. F. Teixeira, P. ten Dijke, and H. J. Zhu, “On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges?,” Frontiers in Cell and Developmental Biology 8 (2020): 605.

[3]

Z. Deng, T. Fan, C. Xiao, et al., “TGF-β Signaling in Health, Disease, and Therapeutics,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 61.

[4]

D. Danielpour, “Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective,” Pharmaceuticals 17, no. 4 (2024): 533.

[5]

H. Peng, J. Shen, X. Long, et al., “Local Release of TGF-β Inhibitor Modulates Tumor-Associated Neutrophils and Enhances Pancreatic Cancer Response to Combined Irreversible Electroporation and Immunotherapy,” Advanced Science 9, no. 10 (2022): 2105240.

[6]

S. O. Imodoye, K. A. Adedokun, A. O. Muhammed, et al., “Understanding the Complex Milieu of Epithelial-Mesenchymal Transition in Cancer Metastasis: New Insight Into the Roles of Transcription Factors,” Frontiers in Oncology 11 (2021): 762817.

[7]

Y. Yu, C. H. Xiao, L. D. Tan, Q. S. Wang, X. Q. Li, and Y. M. Feng, “Cancer-Associated Fibroblasts Induce Epithelial-Mesenchymal Transition of Breast Cancer Cells Through Paracrine TGF-β Signalling,” British Journal of Cancer 110, no. 3 (2014): 724-732.

[8]

C. Anderberg, S. I. Cunha, Z. Zhai, et al., “Deficiency for Endoglin in Tumor Vasculature Weakens the Endothelial Barrier to Metastatic Dissemination,” Journal of Experimental Medicine 210, no. 3 (2013): 563-579.

[9]

Y. Tie, F. Tang, D. Peng, Y. Zhang, and H. Shi, “TGF-Beta Signal Transduction: Biology, Function and Therapy for Diseases,” Molecular Biomedicine 3, no. 1 (2022): 45.

[10]

C. H. Heldin and A. Moustakas, “Role of Smads in TGFβ Signaling,” Cell and Tissue Research 347, no. 1 (2012): 21-36.

[11]

C. H. Heldin and A. Moustakas, “Signaling Receptors for TGF-β Family Members,” Cold Spring Harbor Perspectives in Biology 8, no. 8 (2016): a022053.

[12]

C. L. Pervan, “Smad-Independent TGF-β2 Signaling Pathways in Human Trabecular Meshwork Cells,” Experimental Eye Research 158 (2017): 137-145.

[13]

A. Hata and Y. G. Chen, “TGF-β Signaling From Receptors to Smads,” Cold Spring Harbor Perspectives in Biology 8, no. 9 (2016): a022061.

[14]

K. E. Schmidt, A. L. Höving, S. Kiani Zahrani, et al., “Serum-Induced Proliferation of Human Cardiac Stem Cells Is Modulated via TGFβRI/II and SMAD2/3,” International Journal of Molecular Sciences 25, no. 2 (2024): 959.

[15]

Y. Zhang, P. B. Alexander, and X. F. Wang, “TGF-β Family Signaling in the Control of Cell Proliferation and Survival,” Cold Spring Harbor Perspectives in Biology 9, no. 4 (2017): a022145.

[16]

G. Zhong, Q. Zhao, Z. Chen, and T. Yao, “TGF-β Signaling Promotes Cervical Cancer Metastasis via CDR1as,” Molecular Cancer 22, no. 1 (2023): 66.

[17]

K. Li, J. Guo, Y. Ming, et al., “A Circular RNA Activated by TGFβ Promotes Tumor Metastasis Through Enhancing IGF2BP3-Mediated PDPN mRNA Stability,” Nature Communications 14, no. 1 (2023): 6876.

[18]

E. M. O'Leary, Y. Tian, R. Nigdelioglu, et al., “TGF-β Promotes Metabolic Reprogramming in Lung Fibroblasts via mTORC1-dependent ATF4 Activation,” American Journal of Respiratory Cell and Molecular Biology 63, no. 5 (2020): 601-612.

[19]

X. Xu, L. Zheng, Q. Yuan, et al., “Transforming Growth Factor-β in Stem Cells and Tissue Homeostasis,” Bone Research 6, no. 1 (2018): 2.

[20]

A. M. Lee, C. Shimizu, T. Oharaseki, et al., “Role of TGF-β Signaling in Remodeling of Noncoronary Artery Aneurysms in Kawasaki Disease,” Pediatric and Developmental Pathology 18, no. 4 (2015): 310-317.

[21]

R. Halwani, S. Al-Muhsen, H. Al-Jahdali, and Q. Hamid, “Role of Transforming Growth Factor-β in Airway Remodeling in Asthma,” American Journal of Respiratory Cell and Molecular Biology 44, no. 2 (2011): 127-133.

[22]

E. Batlle and J. Massagué, “Transforming Growth Factor-β Signaling in Immunity and Cancer,” Immunity 50, no. 4 (2019): 924-940.

[23]

Z. Liu, C. Li, N. Kang, H. Malhi, V. H. Shah, and J. L. Maiers, “Transforming Growth Factor β (TGFβ) Cross-Talk With the Unfolded Protein Response Is Critical for Hepatic Stellate Cell Activation,” Journal of Biological Chemistry 294, no. 9 (2019): 3137-3151.

[24]

C. Leduc, L. Sobilo, H. Toumi, et al., “TGF-Beta-Induced Early Gene-1 Overexpression Promotes Oxidative Stress Protection and Actin Cytoskeleton Rearrangement in Human Skin Fibroblasts,” Biochimica et Biophysica Acta (BBA) - General Subjects 1860, no. 6 (2016): 1071-1078.

[25]

H. Zhang, Y. Caudle, C. Wheeler, et al., “TGF-β1/Smad2/3/Foxp3 Signaling Is Required for Chronic Stress-Induced Immune Suppression,” Journal of Neuroimmunology 314 (2018): 30-41.

[26]

Y. E. Zhang, “Non-Smad Signaling Pathways of the TGF-β Family,” Cold Spring Harbor Perspectives in Biology 9, no. 2 (2017): a022129.

[27]

C. Hough, M. Radu, and J. J. E. Doré, “TGF-Beta Induced Erk Phosphorylation of Smad Linker Region Regulates Smad Signaling,” PLoS One 7, no. 8 (2012): e42513.

[28]

R. Batlle, E. Andrés, L. Gonzalez, et al., “Regulation of Tumor Angiogenesis and Mesenchymal-Endothelial Transition by p38α Through TGF-β and JNK Signaling,” Nature Communications 10, no. 1 (2019): 3071.

[29]

B. P. Muthusamy, E. H. Budi, Y. Katsuno, et al., “ShcA Protects Against Epithelial-Mesenchymal Transition Through Compartmentalized Inhibition of TGF-β-Induced Smad Activation,” PLoS Biology 13, no. 12 (2015): e1002325.

[30]

S. Jin, J. Gao, Y. Qi, et al., “TGF-β1 Fucosylation Enhances the Autophagy and Mitophagy via PI3K/Akt and Ras-Raf-MEK-ERK in Ovarian Carcinoma,” Biochemical and Biophysical Research Communications 524, no. 4 (2020): 970-976.

[31]

M. Sisto, L. Lorusso, G. Ingravallo, D. Ribatti, and S. Lisi, “TGFβ1-Smad Canonical and -Erk Noncanonical Pathways Participate in interleukin-17-Induced Epithelial-Mesenchymal Transition in Sjögren's Syndrome,” Laboratory Investigation 100, no. 6 (2020): 824-836.

[32]

A. C. M. Sousa-Squiavinato, M. R. Rocha, P. Barcellos-de-Souza, W. F. de Souza, and J. A. Morgado-Diaz, “Cofilin-1 Signaling Mediates Epithelial-Mesenchymal Transition by Promoting Actin Cytoskeleton Reorganization and Cell-Cell Adhesion Regulation in Colorectal Cancer Cells,” Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1866, no. 3 (2019): 418-429.

[33]

A. Korol, A. Taiyab, and J. A. West-Mays, “RhoA/ROCK Signaling Regulates TGFβ-Induced Epithelial-Mesenchymal Transition of Lens Epithelial Cells Through MRTF-A,” Molecular Medicine 22, no. 1 (2016): 713-723.

[34]

M. E. Menezes, X. N. Shen, S. K. Das, L. Emdad, D. Sarkar, and P. B. Fisher, “MDA-9/Syntenin (SDCBP) Modulates Small GTPases Rhoa and Cdc42 via Transforming Growth Factor β1 to Enhance Epithelial-Mesenchymal Transition in Breast Cancer,” Oncotarget 7, no. 49 (2016): 80175-80189.

[35]

Y. He, M. M. Sun, G. G. Zhang, et al., “Targeting PI3K/Akt Signal Transduction for Cancer Therapy,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 425.

[36]

L. Y. Tang, M. Heller, Z. Meng, et al., “Transforming Growth Factor-β (TGF-β) Directly Activates the JAK1-STAT3 Axis to Induce Hepatic Fibrosis in Coordination With the SMAD Pathway*,” Journal of Biological Chemistry 292, no. 10 (2017): 4302-4312.

[37]

R. L. Philips, Y. Wang, H. Cheon, et al., “The JAK-STAT Pathway at 30: Much Learned, Much More to Do,” Cell 185, no. 21 (2022): 3857-3876.

[38]

F. Huang and Y. G. Chen, “Regulation of TGF-β Receptor Activity,” Cell & Bioscience 2, no. 1 (2012): 9.

[39]

K. Luo, “Positive and Negative Regulation of Type II TGF-Beta Receptor Signal Transduction by Autophosphorylation on Multiple Serine Residues,” EMBO Journal 16, no. 8 (1997): 1970-1981.

[40]

J. Massagué and D. Sheppard, “TGF-β Signaling in Health and Disease,” Cell 186, no. 19 (2023): 4007-4037.

[41]

M. Wu, S. Wu, W. Chen, and Y. P. Li, “The Roles and Regulatory Mechanisms of TGF-β and BMP Signaling in Bone and Cartilage Development, Homeostasis and Disease,” Cell Research 34, no. 2 (2024): 101-123.

[42]

R. Li, “Disrupted TGF-β Signaling: A Link Between Bronchopulmonary Dysplasia and Alveolar Type 1 Cells,” Journal of Clinical Investigation 134, no. 6 (2024): e178562.

[43]

S. K. Ahlfeld, J. Wang, Y. Gao, P. Snider, and S. J. Conway, “Initial Suppression of Transforming Growth Factor-β Signaling and Loss of TGFBI Causes Early Alveolar Structural Defects Resulting in Bronchopulmonary Dysplasia,” American Journal of Pathology 186, no. 4 (2016): 777-793.

[44]

E. A. Meyers and J. A. Kessler, “TGF-β Family Signaling in Neural and Neuronal Differentiation, Development, and Function,” Cold Spring Harbor Perspectives in Biology 9, no. 8 (2017): a022244.

[45]

X. Deng, Y. Chen, Q. Duan, et al., “Genetic and Molecular Mechanisms of Hydrocephalus,” Frontiers in Molecular Neuroscience 17 (2025): 1512455.

[46]

C. Zhan, G. Xiao, X. Zhang, X. Chen, Z. Zhang, and J. Liu, “Decreased MiR-30a Promotes TGF-β1-Mediated Arachnoid Fibrosis in Post-Hemorrhagic Hydrocephalus,” Translational Neuroscience 11, no. 1 (2020): 60-74.

[47]

S. Liarte, Á. Bernabé-García, and F. J. Nicolás, “Role of TGF-β in Skin Chronic Wounds: A Keratinocyte Perspective,” Cells 9, no. 2 (2020): 306.

[48]

Y. M. Kim, E. Kang, J. H. Choi, G. H. Kim, H. W. Yoo, and B. H. Lee, “Clinical Characteristics and Treatment Outcomes in Camurati-Engelmann Disease,” Medicine 97, no. 14 (2018): e0309.

[49]

Q. Chen, Y. Yao, K. Chen, et al., “Aberrant Activation of TGF-β1 Induces High Bone Turnover via Rho GTPases-Mediated Cytoskeletal Remodeling in Camurati-Engelmann Disease,” Frontiers in Endocrinology 13 (2022): 913979.

[50]

I. van der Pluijm, N. van Vliet, J. H. von der Thusen, et al., “Defective Connective Tissue Remodeling in Smad3 Mice Leads to Accelerated Aneurysmal Growth Through Disturbed Downstream TGF-β Signaling,” EBioMedicine 12 (2016): 280-294.

[51]

M. E. Lindsay, D. Schepers, N. A. Bolar, et al., “Loss-of-Function Mutations in TGFB2 Cause a Syndromic Presentation of Thoracic Aortic Aneurysm,” Nature Genetics 44, no. 8 (2012): 922-927.

[52]

A. M. Bertoli-Avella, E. Gillis, H. Morisaki, et al., “Mutations in a TGF-β Ligand, TGFB3, Cause Syndromic Aortic Aneurysms and Dissections,” Journal of the American College of Cardiology 65, no. 13 (2015): 1324-1336.

[53]

R. Jan and G. S. Chaudhry, “Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics,” Advanced Pharmaceutical Bulletin 9, no. 2 (2019): 205-218.

[54]

S. Qian, Z. Wei, W. Yang, J. Huang, Y. Yang, and J. Wang, “The Role of BCL-2 Family Proteins in Regulating Apoptosis and Cancer Therapy,” Frontiers in Oncology 12 (2022): 985363.

[55]

K. A. Adedokun, S. O. Imodoye, I. O. Bello, A. A. Lanihun, and I. O. Bello, “Therapeutic potentials of medicinal plants and significance of computational tools in anti-cancer drug discovery,” in Phytochemistry, Computational Tools and Databases in Drug Discovery, ed. C, Egbuna, M, Rudrapal, H, Tijjani, 1st ed. (Elsevier, 2023), 393-455.

[56]

A. B. Baba, B. Rah, G. R. Bhat, et al., “Transforming Growth Factor-Beta (TGF-β) Signaling in Cancer-A Betrayal Within,” Frontiers in Pharmacology 13 (2022): 791272.

[57]

J. J. Huang and G. C. Blobe, “Dichotomous Roles of TGF-β in Human Cancer,” Biochemical Society Transactions 44, no. 5 (2016): 1441-1454.

[58]

Q. Hao, J. Chen, J. Liao, et al., “p53 induces ARTS to Promote Mitochondrial Apoptosis,” Cell Death & Disease 12, no. 2 (2021): 204.

[59]

C. J. David, Y. H. Huang, M. Chen, et al., “TGF-β Tumor Suppression Through a Lethal EMT,” Cell 164, no. 5 (2016): 1015-1030.

[60]

J. Wang, L. Yang, J. Yang, et al., “Transforming Growth Factor β Induces Apoptosis Through Repressing the Phosphoinositide 3-Kinase/AKT/Survivin Pathway in Colon Cancer Cells,” Cancer Research 68, no. 9 (2008): 3152-3160.

[61]

K. C. Kawabata, S. Ehata, A. Komuro, K. Takeuchi, and K. Miyazono, “TGF-β-Induced Apoptosis of B-Cell Lymphoma Ramos Cells Through Reduction of MS4A1/CD20,” Oncogene 32, no. 16 (2013): 2096-2106.

[62]

J. Korah, N. Falah, A. Lacerte, and J. J. Lebrun, “A Transcriptionally Active pRb-E2F1-P/CAF Signaling Pathway Is Central to TGFβ-Mediated Apoptosis,” Cell Death & Disease 3, no. 10 (2012): e407.

[63]

M. J. Lim, T. Lin, and S. B. Jakowlew, “Signaling Mechanism of Transforming Growth Factor (TGF-β) in Cancer; TGF-β Induces Apoptosis in Lung Cells by a Smad-Dependent Mechanism,” in Tumor Suppressor Genes (InTech, 2012), 145-180.

[64]

M. Rudrapal, A. K. Mishra, L. Rani, et al., “Nanodelivery of Dietary Polyphenols for Therapeutic Applications,” Molecules 27, no. 24 (2022): 8706.

[65]

A. K. Adedokun, S. O. Imodoye, Z. S. Yahaya, et al., “ Nanodelivery of Polyphenols as Nutraceuticals in Anticancer Interventions.” in Polyphenols: Food, Nutraceutical, and Nanotherapeutic Applications, ed. M. Rudrapal (Wiley, 2024), 1st ed, 188-224.

[66]

L. R. Pack, L. H. Daigh, and T. Meyer, “Putting the Brakes on the Cell Cycle: Mechanisms of Cellular Growth Arrest,” Current Opinion in Cell Biology 60 (2019): 106-113.

[67]

J. Seoane and R. R. Gomis, “TGF-β Family Signaling in Tumor Suppression and Cancer Progression,” Cold Spring Harbor Perspectives in Biology 9, no. 12 (2017): a022277.

[68]

S. Haque and J. C. Morris, “Transforming Growth Factor-β: A Therapeutic Target for Cancer,” Human Vaccines & Immunotherapeutics 13, no. 8 (2017): 1741-1750.

[69]

L. Ding, J. Cao, W. Lin, et al., “The Roles of Cyclin-Dependent Kinases in Cell-Cycle Progression and Therapeutic Strategies in Human Breast Cancer,” International Journal of Molecular Sciences 21, no. 6 (2020): 1960.

[70]

M. Zhang, L. Zhang, R. Hei, et al., “CDk Inhibitors in Cancer Therapy, an Overview of Recent Development,” American Journal of Cancer Research 11, no. 5 (2021): 1913-1935.

[71]

L. Kubiczkova, L. Sedlarikova, R. Hajek, and S. Sevcikova, “TGF-β - An Excellent Servant but a Bad Master,” Journal of Translational Medicine 10, no. 1 (2012): 183.

[72]

D. R. Principe, J. A. Doll, J. Bauer, et al., “TGF-β: Duality of Function Between Tumor Prevention and Carcinogenesis,” JNCI Journal of the National Cancer Institute 106, no. 2 (2014): djt369.

[73]

Y. Ke and X. J. Wang, “TGFβ Signaling in Photoaging and UV-Induced Skin Cancer,” Journal of Investigative Dermatology 141, no. 4 (2021): 1104-1110.

[74]

R. Derynck, S. J. Turley, and R. J. Akhurst, “TGFβ Biology in Cancer Progression and Immunotherapy,” Nature Reviews Clinical Oncology 18, no. 1 (2021): 9-34.

[75]

T. S. Anderson, A. L. Wooster, S. L. Piersall, I. F. Okpalanwaka, and D. B. Lowe, “Disrupting Cancer Angiogenesis and Immune Checkpoint Networks for Improved Tumor Immunity,” Seminars in Cancer Biology 86, no. Pt 3 (2022): 981-996.

[76]

D. Song, J. Lan, Y. Chen, et al., “NSD2 Promotes Tumor Angiogenesis Through Methylating and Activating STAT3 Protein,” Oncogene 40, no. 16 (2021): 2952-2967.

[77]

S. Liu, S. Chen, and J. Zeng, “TGF-β Signaling: A Complex Role in Tumorigenesis,” Molecular Medicine Reports 17, no. 1 (2018): 699-704.

[78]

L. A. van Meeteren and P. ten Dijke, “Regulation of Endothelial Cell Plasticity by TGF-β,” Cell and Tissue Research 347, no. 1 (2012): 177-186.

[79]

S. Liu, J. Ren, and P. ten Dijke, “Targeting TGFβ Signal Transduction for Cancer Therapy,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 8.

[80]

G. A. Cabral-Pacheco, I. Garza-Veloz, C. Castruita-De la rosa, et al., “The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases,” International Journal of Molecular Sciences 21, no. 24 (2020): 9739.

[81]

S. Quintero-Fabián, R. Arreola, E. Becerril-Villanueva, et al., “Role of Matrix Metalloproteinases in Angiogenesis and Cancer,” Frontiers in Oncology 9 (2019): 1370.

[82]

A. H. Webb, B. T. Gao, Z. K. Goldsmith, et al., “Inhibition of MMP-2 and MMP-9 Decreases Cellular Migration, and Angiogenesis in In Vitro Models of Retinoblastoma,” BMC Cancer 17, no. 1 (2017): 434.

[83]

T. Chen, Y. You, H. Jiang, and Z. Z. Wang, “Epithelial-Mesenchymal Transition (EMT): A Biological Process in the Development, Stem Cell Differentiation, and Tumorigenesis,” Journal of Cellular Physiology 232, no. 12 (2017): 3261-3272.

[84]

E. Sahai, I. Astsaturov, E. Cukierman, et al., “A Framework for Advancing Our Understanding of Cancer-Associated Fibroblasts,” Nature Reviews Cancer 20, no. 3 (2020): 174-186.

[85]

S. O. Imodoye and K. A. Adedokun, “EMT-Induced Immune Evasion: Connecting the Dots From Mechanisms to Therapy,” Clinical and Experimental Medicine 23, no. 8 (2023): 4265-4287.

[86]

D. Yang, J. Liu, H. Qian, and Q. Zhuang, “Cancer-Associated Fibroblasts: From Basic Science to Anticancer Therapy,” Experimental & Molecular Medicine 55, no. 7 (2023): 1322-1332.

[87]

X. Mao, J. Xu, W. Wang, et al., “Crosstalk Between Cancer-Associated Fibroblasts and Immune Cells in the Tumor Microenvironment: New Findings and Future Perspectives,” Molecular Cancer 20, no. 1 (2021): 131.

[88]

N. Cui, M. Hu, and R. A. Khalil, “Chapter One Biochemical and Biological Attributes of Matrix Metalloproteinases,” Progress in Molecular Biology and Translational Science 147 (2017): 1-73.

[89]

L. Han, B. Sheng, Q. Zeng, W. Yao, and Q. Jiang, “Correlation Between MMP2 Expression in Lung Cancer Tissues and Clinical Parameters: A Retrospective Clinical Analysis,” BMC Pulmonary Medicine 20, no. 1 (2020): 283.

[90]

G. Y. Lian, Q. M. Wang, T. S. K. Mak, X. R. Huang, X. Q. Yu, and H. Y. Lan, “Inhibition of Tumor Invasion and Metastasis by Targeting TGF-β-Smad-MMP2 Pathway With Asiatic Acid and Naringenin,” Molecular Therapy - Oncolytics 20 (2021): 277-289.

[91]

U. Blank and S. Karlsson, “The Role of Smad Signaling in Hematopoiesis and Translational Hematology,” Leukemia 25, no. 9 (2011): 1379-1388.

[92]

F. Xie, L. Ling, H. van Dam, F. Zhou, and L. Zhang, “TGF-β Signaling in Cancer Metastasis,” Acta Biochimica et Biophysica Sinica 50, no. 1 (2017): 121-132.

[93]

Y. Yoshimatsu, I. Wakabayashi, S. Kimuro, et al., “TNF-α Enhances TGF-β-Induced Endothelial-to-Mesenchymal Transition via TGF-β Signal Augmentation,” Cancer Science 111, no. 7 (2020): 2385-2399.

[94]

X. Jiang, Z. Zhang, C. Song, et al., “Glaucocalyxin A Reverses EMT and TGF-β1-Induced EMT by Inhibiting TGF-β1/Smad2/3 Signaling Pathway in Osteosarcoma,” Chemico-Biological Interactions 307 (2019): 158-166.

[95]

J. Ma, G. van der Zon, M. A. F. V. Gonçalves, et al., “TGF-β-Induced Endothelial to Mesenchymal Transition Is Determined by a Balance Between SNAIL and ID Factors,” Frontiers in Cell and Developmental Biology 9 (2021): 616610.

[96]

X. Gong, Z. Hou, M. P. Endsley, et al., “Interaction of Tumor Cells and Astrocytes Promotes Breast Cancer Brain Metastases Through TGF-β2/ANGPTL4 Axes,” npj Precision Oncology 3, no. 1 (2019): 24.

[97]

H. Yang, L. Wang, J. Zhao, et al., “TGF-β-Activated SMAD3/4 Complex Transcriptionally Upregulates N-Cadherin Expression in Non-Small Cell Lung Cancer,” Lung Cancer 87, no. 3 (2015): 249-257.

[98]

Y. Katsuno and R. Derynck, “Epithelial Plasticity, Epithelial-Mesenchymal Transition, and the TGF-β Family,” Developmental Cell 56, no. 6 (2021): 726-746.

[99]

J. Nong, S. Shen, F. Hong, et al., “Verteporfin Inhibits TGF-β Signaling by Disrupting the Smad2/3-Smad4 Interaction,” Molecular Biology of the Cell 35, no. 7 (2024): ar95.

[100]

H. Ungefroren, S. Groth, S. Sebens, H. Lehnert, F. Gieseler, and F. Fändrich, “Differential Roles of Smad2 and Smad3 in the Regulation of TGF-β1-Mediated Growth Inhibition and Cell Migration in Pancreatic Ductal Adenocarcinoma Cells: Control by Rac1,” Molecular Cancer 10, no. 1 (2011): 67.

[101]

Y. Hao, D. Baker, and P. ten Dijke, “TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis,” International Journal of Molecular Sciences 20, no. 11 (2019): 2767.

[102]

G. Han and X. J. Wang, “Roles of TGFβ Signaling Smads in Squamous Cell Carcinoma,” Cell & Bioscience 1, no. 1 (2011): 41.

[103]

T. J. Bell, D. J. Nagel, C. F. Woeller, and R. M. Kottmann, “Ogerin Mediated Inhibition of TGF-β(1) Induced Myofibroblast Differentiation Is Potentiated by Acidic pH,” PLoS One 17, no. 7 (2022): e0271608.

[104]

Z. Zi, “Molecular Engineering of the TGF-β Signaling Pathway,” Journal of Molecular Biology 431, no. 15 (2019): 2644-2654.

[105]

D. Peng, M. Fu, M. Wang, Y. Wei, and X. Wei, “Targeting TGF-β Signal Transduction for Fibrosis and Cancer Therapy,” Molecular Cancer 21, no. 1 (2022): 104.

[106]

P. Sritananuwat, N. Sueangoen, P. Thummarati, K. Islam, and T. Suthiphongchai, “Blocking ERK1/2 Signaling Impairs TGF-β1 Tumor Promoting Function but Enhances Its Tumor Suppressing Role in Intrahepatic Cholangiocarcinoma Cells,” Cancer Cell International 17, no. 1 (2017): 85.

[107]

D. R. Principe, A. M. Diaz, C. Torres, et al., “TGFβ Engages MEK/ERK to Differentially Regulate Benign and Malignant Pancreas Cell Function,” Oncogene 36, no. 30 (2017): 4336-4348.

[108]

M. Zhao, L. Mishra, and C. X. Deng, “The Role of TGF-β/SMAD4 Signaling in Cancer,” International Journal of Biological Sciences 14, no. 2 (2018): 111-123.

[109]

M. Deng, X. Cai, L. Long, et al., “CD36 Promotes the Epithelial-Mesenchymal Transition and Metastasis in Cervical Cancer by Interacting With TGF-Β,” Journal of Translational Medicine 17, no. 1 (2019): 352.

[110]

N. Zhang and M. J. Bevan, “TGF-β Signaling to T Cells Inhibits Autoimmunity During Lymphopenia-Driven Proliferation,” Nature Immunology 13, no. 7 (2012): 667-673.

[111]

S. Sanjabi, S. A. Oh, and M. O. Li, “Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection,” Cold Spring Harbor Perspectives in Biology 9, no. 6 (2017): a022236.

[112]

S. Regis, A. Dondero, F. Caliendo, C. Bottino, and R. Castriconi, “NK Cell Function Regulation by TGF-β-Induced Epigenetic Mechanisms,” Frontiers in Immunology 11 (2020): 311.

[113]

S. A. Oh and M. O. Li, “TGF-β: Guardian of T Cell Function,” Journal of Immunology 191, no. 8 (2013): 3973-3979.

[114]

B. V. Park, Z. T. Freeman, A. Ghasemzadeh, et al., “TGFβ1-Mediated SMAD3 Enhances PD-1 Expression on Antigen-Specific T Cells in Cancer,” Cancer Discovery 6, no. 12 (2016): 1366-1381.

[115]

Z. Z. Yang, D. M. Grote, B. Xiu, et al., “TGF-β Upregulates CD70 Expression and Induces Exhaustion of Effector Memory T Cells in B-Cell Non-Hodgkin's Lymphoma,” Leukemia 28, no. 9 (2014): 1872-1884.

[116]

Z. Z. Yang, D. M. Grote, S. C. Ziesmer, et al., “Soluble and Membrane-Bound TGF-β-Mediated Regulation of Intratumoral T Cell Differentiation and Function in B-Cell Non-Hodgkin Lymphoma,” PLoS One 8, no. 3 (2013): e59456.

[117]

Y. Tsuchida, S. Sumitomo, K. Ishigaki, et al., “TGF-β3 Inhibits Antibody Production by Human B Cells,” PLoS One 12, no. 1 (2017): e0169646.

[118]

F. Zhang, H. Wang, X. Wang, et al., “TGF-β Induces M2-Like Macrophage Polarization via Snail-Mediated Suppression of a Pro-Inflammatory Phenotype,” Oncotarget 7, no. 32 (2016): 52294-52306.

[119]

D. Gong, W. Shi, S. Yi, H. Chen, J. Groffen, and N. Heisterkamp, “TGFβ Signaling Plays a Critical Role in Promoting Alternative Macrophage Activation,” BMC Immunology 13, no. 1 (2012): 31.

[120]

Z. G. Fridlender, J. Sun, S. Kim, et al., “Polarization of Tumor-Associated Neutrophil Phenotype by TGF-β: ‘N1’ Versus ‘N2’ TAN,” Cancer Cell 16, no. 3 (2009): 183-194.

[121]

F. Qin, X. Liu, J. Chen, et al., “Anti-TGF-β Attenuates Tumor Growth via Polarization of Tumor Associated Neutrophils Towards an Anti-Tumor Phenotype in Colorectal Cancer,” Journal of Cancer 11, no. 9 (2020): 2580-2592.

[122]

J. P. Stevenson, H. L. Kindler, E. Papasavvas, et al., “Immunological Effects of the TGFβ-blocking Antibody GC1008 in Malignant Pleural Mesothelioma Patients,” Oncoimmunology 2, no. 8 (2013): e26218.

[123]

F. Wu, J. Yang, J. Liu, et al., “Signaling Pathways in Cancer-Associated Fibroblasts and Targeted Therapy for Cancer,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 218.

[124]

A. Chakravarthy, L. Khan, N. P. Bensler, P. Bose, and D. D. De Carvalho, “TGF-β-Associated Extracellular Matrix Genes Link Cancer-Associated Fibroblasts to Immune Evasion and Immunotherapy Failure,” Nature Communications 9, no. 1 (2018): 4692.

[125]

E. Henke, R. Nandigama, and S. Ergün, “Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy,” Frontiers in Molecular Biosciences 6 (2020): 160.

[126]

H. Salmon and E. Donnadieu, “Within Tumors, Interactions Between T Cells and Tumor Cells Are Impeded by the Extracellular Matrix,” Oncoimmunology 1, no. 6 (2012): 992-994.

[127]

P. Papageorgis and T. Stylianopoulos, “Role of TGFβ in Regulation of the Tumor Microenvironment and Drug Delivery (Review),” International Journal of Oncology 46, no. 3 (2015): 933-943.

[128]

M. Papaspyridonos, I. Matei, Y. Huang, et al., “Id1 Suppresses Anti-Tumour Immune Responses and Promotes Tumour Progression by Impairing Myeloid Cell Maturation,” Nature Communications 6, no. 1 (2015): 6840.

[129]

B. A. Hanks, A. Holtzhausen, K. S. Evans, et al., “Type III TGF-β Receptor Downregulation Generates an Immunotolerant Tumor Microenvironment,” Journal of Clinical Investigation 123, no. 9 (2013): 3925-3940.

[130]

O. Shinde and P. Li, “The Molecular Mechanism of dsDNA Sensing Through the cGAS-STING Pathway,” Advances in Immunology 162 (2024): 1-21.

[131]

J. Zhang, S. Yu, Q. Peng, P. Wang, and L. Fang, “Emerging Mechanisms and Implications of cGAS-STING Signaling in Cancer Immunotherapy Strategies,” Cancer Biology & Medicine 21, no. 1 (2024): 1-20.

[132]

J. Zhou, Z. Zhuang, J. Li, and Z. Feng, “Significance of the cGAS-STING Pathway in Health and Disease,” International Journal of Molecular Sciences 24, no. 17 (2023): 13316.

[133]

D. Zhang, D. Zhan, R. Zhang, et al., “Treg-Derived TGF-β1 Dampens cGAS-STING Signaling to Downregulate the Expression of Class I MHC Complex in Multiple Myeloma,” Scientific Reports 14, no. 1 (2024): 11593.

[134]

J. Luo, S. Wang, Q. Yang, et al., “γδ T Cell-Mediated Tumor Immunity Is Tightly Regulated by STING and TGF-β Signaling Pathways,” Advancement of Science 12 (2024): e2404432.

[135]

R. Fujii, C. Jochems, S. R. Tritsch, H. C. Wong, J. Schlom, and J. W. Hodge, “An IL-15 Superagonist/IL-15Rα Fusion Complex Protects and Rescues NK Cell-Cytotoxic Function From TGF-β1-Mediated Immunosuppression,” Cancer Immunology, Immunotherapy 67, no. 4 (2018): 675-689.

[136]

P. M. K. Tang, S. Zhou, X. M. Meng, et al., “Smad3 Promotes Cancer Progression by Inhibiting E4BP4-Mediated NK Cell Development,” Nature Communications 8, no. 1 (2017): 14677.

[137]

A. G. Bozward, F. Warricker, Y. H. Oo, and S. I. Khakoo, “Natural Killer Cells and Regulatory T Cells Cross Talk in Hepatocellular Carcinoma: Exploring Therapeutic Options for the Next Decade,” Frontiers in Immunology 12 (2021): 643310.

[138]

V. S. Cortez, T. K. Ulland, L. Cervantes-Barragan, et al., “SMAD4 Impedes the Conversion of NK Cells Into ILC1-like Cells by Curtailing Non-Canonical TGF-β Signaling,” Nature Immunology 18, no. 9 (2017): 995-1003.

[139]

Y. Gao, F. Souza-Fonseca-Guimaraes, T. Bald, et al., “Tumor Immunoevasion by the Conversion of Effector NK Cells Into Type 1 Innate Lymphoid Cells,” Nature Immunology 18, no. 9 (2017): 1004-1015.

[140]

K. Tzavlaki and A. Moustakas, “TGF-β Signaling,” Biomolecules 10, no. 3 (2020): 487.

[141]

H. C. Tran, Z. Wan, M. A. Sheard, et al., “TGFβR1 Blockade With Galunisertib (LY2157299) Enhances Anti-Neuroblastoma Activity of the Anti-GD2 Antibody Dinutuximab (ch14.18) With Natural Killer Cells,” Clinical Cancer Research 23, no. 3 (2017): 804-813.

[142]

S. Herbertz, J. S. Sawyer, A. J. Stauber, et al., “Clinical Development of Galunisertib (LY2157299 Monohydrate), a Small Molecule Inhibitor of Transforming Growth Factor-Beta Signaling Pathway,” Drug Design, Development and Therapy 9 (2015): 4479-4499.

[143]

R. B. Holmgaard, D. A. Schaer, Y. Li, et al., “Targeting the TGFβ Pathway With Galunisertib, a TGFβRI Small Molecule Inhibitor, Promotes Anti-Tumor Immunity Leading to Durable, Complete Responses, as Monotherapy and in Combination With Checkpoint Blockade,” Journal for Immunotherapy of Cancer 6, no. 1 (2018): 47.

[144]

D. Melisi, D. Y. Oh, A. Hollebecque, et al., “Safety and Activity of the TGFβ Receptor I Kinase Inhibitor Galunisertib Plus the anti-PD-L1 Antibody Durvalumab in Metastatic Pancreatic Cancer,” Journal for Immunotherapy of Cancer 9, no. 3 (2021): e002068.

[145]

V. Santini, D. Valcárcel, U. Platzbecker, et al., “Phase II Study of the ALK5 Inhibitor Galunisertib in Very Low-, Low-, and Intermediate-Risk Myelodysplastic Syndromes,” Clinical Cancer Research 25, no. 23 (2019): 6976-6985.

[146]

S. Faivre, A. Santoro, R. K. Kelley, et al., “Novel Transforming Growth Factor Beta Receptor I Kinase Inhibitor Galunisertib (LY2157299) in Advanced Hepatocellular Carcinoma,” Liver International 39, no. 8 (2019): 1468-1477.

[147]

A. A. Brandes, A. F. Carpentier, S. Kesari, et al., “A Phase II Randomized Study of Galunisertib Monotherapy or Galunisertib Plus Lomustine Compared With Lomustine Monotherapy in Patients With Recurrent Glioblastoma,” Neuro-Oncology 18, no. 8 (2016): 1146-1156.

[148]

Definition of vactosertib - “NCI Drug Dictionary - NCI,” Accessed, September 9, 2024, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/vactosertib.

[149]

E. Hong, W. Barczak, S. Park, et al., “Combination Treatment of T1-44, a PRMT5 Inhibitor With Vactosertib, An Inhibitor of TGF-β Signaling, Inhibits Invasion and Prolongs Survival in a Mouse Model of Pancreatic Tumors,” Cell Death & Disease 14, no. 2 (2023): 93.

[150]

S. T. Kim, J. Y. Hong, Y. S. Park, S. J. Kim, and J. O. Park, “Phase 1b Study of Vactosertib in Combination With Oxaliplatin With 5FU/LV (FOLFOX) in Patients With Metastatic Pancreatic Cancer Who Have Failed First-Line Gemcitabine/Nab-Paclitaxel,” Journal of Clinical Oncology 40, no. 16_suppl (2022): e16299.

[151]

C. H. Jin, M. Krishnaiah, D. Sreenu, et al., “Discovery of N-((4-([1,2,4]Triazolo[1,5-a]Pyridin-6-yl)-5-(6-Methylpyridin-2-yl)-1H-Imidazol-2-yl)Methyl)-2-Fluoroaniline (EW-7197): A Highly Potent, Selective, and Orally Bioavailable Inhibitor of TGF-β Type I Receptor Kinase as Cancer Immunotherapeutic/Antifibrotic Agent,” Journal of Medicinal Chemistry 57, no. 10 (2014): 4213-4238.

[152]

J. Y. Son, S. Y. Park, S. J. Kim, et al., “EW-7197, a Novel ALK-5 Kinase Inhibitor, Potently Inhibits Breast to Lung Metastasis,” Molecular Cancer Therapeutics 13, no. 7 (2014): 1704-1716.

[153]

S. H. Choi, J. T. Myers, S. L. Tomchuck, et al., “Oral Transforming Growth Factor-Beta Receptor 1 Inhibitor Vactosertib Promotes Osteosarcoma Regression by Targeting Tumor Proliferation and Enhancing Anti-Tumor Immunity,” Cancer Communications 44, no. 8 (2024): 884-888.

[154]

E. Malek, P. S. Rana, M. Swamydas, et al., “Vactosertib, a Novel TGFb Type I Receptor Kinase Inhibitor, Improves T-Cell Fitness: A Single-Arm, Phase 1b Trial in Relapsed/Refractory Multiple Myeloma,” Blood 142, no. Suppl 1 (2023): 4749.

[155]

S. Y. Jung, S. Hwang, J. M. Clarke, et al., “Pharmacokinetic Characteristics of Vactosertib, a New Activin Receptor-Like Kinase 5 Inhibitor, in Patients With Advanced Solid Tumors in a First-in-Human Phase 1 Study,” Investigational New Drugs 38, no. 3 (2020): 812-820.

[156]

T. W. Kim, K. W. Lee, J. B. Ahn, et al., “Efficacy and Safety of Vactosertib and Pembrolizumab Combination in Patients With Previously Treated Microsatellite Stable Metastatic Colorectal Cancer,” Journal of Clinical Oncology 39, no. 15_suppl (2021): 3573.

[157]

J. van den Bulk, N. F. C. C. de Miranda, and P. ten Dijke, “Therapeutic Targeting of TGF-β in Cancer: Hacking a Master Switch of Immune Suppression,” Clinical Science 135, no. 1 (2021): 35-52.

[158]

A. Gupta, S. Budhu, K. Fitzgerald, et al., “Isoform Specific Anti-TGFβ Therapy Enhances Antitumor Efficacy in Mouse Models of Cancer,” Communications Biology 4, no. 1 (2021): 1296.

[159]

S. Busch, A. Acar, Y. Magnusson, P. Gregersson, L. Rydén, and G. Landberg, “TGF-Beta Receptor Type-2 Expression in Cancer-Associated Fibroblasts Regulates Breast Cancer Cell Growth and Survival and Is a Prognostic Marker in Pre-Menopausal Breast Cancer,” Oncogene 34, no. 1 (2015): 27-38.

[160]

J. C. Morris, A. R. Tan, T. E. Olencki, et al., “Phase I Study of GC1008 (Fresolimumab): A Human Anti-Transforming Growth Factor-Beta (TGFβ) Monoclonal Antibody in Patients With Advanced Malignant Melanoma or Renal Cell Carcinoma,” PLoS One 9, no. 3 (2014): e90353.

[161]

A. W. Tolcher, J. D. Berlin, J. Cosaert, et al., “A Phase 1 Study of Anti-TGFβ Receptor Type-II Monoclonal Antibody LY3022859 in Patients With Advanced Solid Tumors,” Cancer Chemotherapy and Pharmacology 79, no. 4 (2017): 673-680.

[162]

C. L. Arteaga, S. D. Hurd, A. R. Winnier, M. D. Johnson, B. M. Fendly, and J. T. Forbes, “Anti-Transforming Growth Factor (TGF)-Beta Antibodies Inhibit Breast Cancer Cell Tumorigenicity and Increase Mouse Spleen Natural Killer Cell Activity. Implications for a Possible Role of Tumor Cell/Host TGF-Beta Interactions in Human Breast Cancer Progression,” Journal of Clinical Investigation 92, no. 6 (1993): 2569-2576.

[163]

G. J. Prud'homme, “Pathobiology of Transforming Growth Factor β in Cancer, Fibrosis and Immunologic Disease, and Therapeutic Considerations,” Laboratory Investigation 87, no. 11 (2007): 1077-1091.

[164]

R. Kumar, A. V. Grinberg, H. Li, et al., “Functionally Diverse Heteromeric Traps for Ligands of the Transforming Growth Factor-β Superfamily,” Scientific Reports 11, no. 1 (2021): 18341.

[165]

Y. Yang, O. Dukhanina, B. Tang, et al., “Lifetime Exposure to a Soluble TGF-β Antagonist Protects Mice Against Metastasis Without Adverse Side Effects,” Journal of Clinical Investigation 109, no. 12 (2002): 1607-1615.

[166]

T. Qin, L. Barron, L. Xia, et al., “A Novel Highly Potent Trivalent TGF-β Receptor Trap Inhibits Early-Stage Tumorigenesis and Tumor Cell Invasion in Murine Pten-Deficient Prostate Glands,” Oncotarget 7, no. 52 (2016): 86087-86102.

[167]

T. A. Yap, N. J. Lakhani, D. V. Araujo, et al., “AVID200, First-in-Class TGF-Beta 1 and 3 Selective and Potent Inhibitor: Safety and Biomarker Results of a Phase I Monotherapy Dose-Escalation Study in Patients With Advanced Solid Tumors,” Journal of Clinical Oncology 38, no. 15_suppl (2020): 3587.

[168]

A. Rodríguez, C. Yang, E. Furutani, et al., “Inhibition of TGFβ1 and TGFβ3 Promotes Hematopoiesis in Fanconi Anemia,” Experimental Hematology 93 (2021): 70-84.e4.

[169]

P. Dobosz, M. Stępień, A. Golke, and T. Dzieciątkowski, “Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment,” International Journal of Molecular Sciences 23, no. 5 (2022): 2847.

[170]

S. Mariathasan, S. J. Turley, D. Nickles, et al., “TGFβ Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells,” Nature 554, no. 7693 (2018): 544-548.

[171]

D. V. F. Tauriello, S. Palomo-Ponce, D. Stork, et al., “TGFβ Drives Immune Evasion in Genetically Reconstituted Colon Cancer Metastasis,” Nature 554, no. 7693 (2018): 538-543.

[172]

L. G. Feun, Y. Y. Li, C. Wu, et al., “Phase 2 Study of Pembrolizumab and Circulating Biomarkers to Predict Anticancer Response in Advanced, Unresectable Hepatocellular Carcinoma,” Cancer 125, no. 20 (2019): 3603-3614.

[173]

C. Vanpouille-Box and S. C. Formenti, “Dual Transforming Growth Factor-β and Programmed Death-1 Blockade: A Strategy for Immune-Excluded Tumors?,” Trends in Immunology 39, no. 6 (2018): 435-437.

[174]

M. Terabe, F. C. Robertson, K. Clark, et al., “Blockade of Only TGF-β 1 and 2 Is Sufficient to Enhance the Efficacy of Vaccine and PD-1 Checkpoint Blockade Immunotherapy,” Oncoimmunology 6, no. 5 (2017): e1308616.

[175]

X. Chen, L. Wang, P. Li, et al., “Dual TGF-β and PD-1 Blockade Synergistically Enhances MAGE-A3-specific CD8+ T Cell Response in Esophageal Squamous Cell Carcinoma,” International Journal of Cancer 143, no. 10 (2018): 2561-2574.

[176]

J. M. David, C. Dominguez, K. K. McCampbell, J. L. Gulley, J. Schlom, and C. Palena, “A Novel Bifunctional anti-Pd-L1/TGF-β Trap Fusion Protein (M7824) Efficiently Reverts Mesenchymalization of Human Lung Cancer Cells,” Oncoimmunology 6, no. 10 (2017): e1349589.

[177]

R. Ravi, K. A. Noonan, V. Pham, et al., “Bifunctional Immune Checkpoint-Targeted Antibody-Ligand Traps That Simultaneously Disable TGFβ Enhance the Efficacy of Cancer Immunotherapy,” Nature Communications 9, no. 1 (2018): 741.

[178]

Y. Lan, D. Zhang, C. Xu, et al., “Enhanced Preclinical Antitumor Activity of M7824, a Bifunctional Fusion Protein Simultaneously Targeting PD-L1 and TGf-Β,” Science Translational Medicine 10, no. 424 (2018): eaan5488.

[179]

B. Tan, A. Khattak, E. Felip, et al., “Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Patients With Esophageal Adenocarcinoma: Results From a Phase 1 Cohort,” Targeted Oncology 16, no. 4 (2021): 435-446.

[180]

C. C. Lin, T. Doi, K. Muro, et al., “Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGFβ and PD-L1, in Patients With Esophageal Squamous Cell Carcinoma: Results From a Phase 1 Cohort in Asia,” Targeted Oncology 16, no. 4 (2021): 447-459.

[181]

J. Strauss, M. E. Gatti-Mays, B. C. Cho, et al., “Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Patients With Human Papillomavirus-Associated Malignancies,” Journal for Immunotherapy of Cancer 8, no. 2 (2020): e001395.

[182]

M. Khasraw, M. Weller, D. Lorente, et al., “Bintrafusp Alfa (M7824) a Bifunctional Fusion Protein Targeting TGF-β and PD-L1: Results From a Phase 1 Expansion Cohort in Patients With Recurrent Glioblastoma,” Neuro-Oncology Advances 3, no. 1 (2021): vdab058.

[183]

F. Barlesi, N. Isambert, E. Felip, et al., “Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Patients With Non-Small Cell Lung Cancer Resistant or Refractory to Immune Checkpoint Inhibitors,” Oncologist 28, no. 3 (2022): 258-267.

[184]

V. K. Morris, M. J. Overman, M. Lam, et al., “Bintrafusp Alfa, An anti-PD-L1:TGF-β Trap Fusion Protein, in Patients With ctDNA-Positive, Liver-Limited Metastatic Colorectal Cancer,” Cancer Research Communications 2, no. 9 (2022): 979-986.

[185]

A. Spira, M. S. Wertheim, E. J. Kim, et al., “Bintrafusp Alfa: A Bifunctional Fusion Protein Targeting PD-L1 and TGF-β, in Patients With Pretreated Colorectal Cancer: Results From a Phase I Trial,” Oncologist 28, no. 2 (2022): e124-e127.

[186]

Y. K. Kang, Y. J. Bang, S. Kondo, et al., “Safety and Tolerability of Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGFβ and PD-L1, in Asian Patients With Pretreated Recurrent or Refractory Gastric Cancer,” Clinical Cancer Research 26, no. 13 (2020): 3202-3210.

[187]

C. Yoo, D. Y. Oh, H. J. Choi, et al., “Phase I Study of Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Patients With Pretreated Biliary Tract Cancer,” Journal for Immunotherapy of Cancer 8, no. 1 (2020): e000564.

[188]

C. Yoo, M. M. Javle, H. Verdaguer Mata, et al., “Phase 2 Trial of Bintrafusp Alfa as Second-Line Therapy for Patients With Locally Advanced/Metastatic Biliary Tract Cancers,” Hepatology 78, no. 3 (2023): 758-770.

[189]

A. A. Strait and X. J. Wang, “Setting Up Clinical Trials for Success: Applying Preclinical Advances in Combined TGFβ/Pd-L1 Inhibition to Ongoing Clinical Studies,” Molecular Carcinogenesis 61, no. 2 (2022): 239-242.

[190]

P. Fenaux, U. Platzbecker, G. J. Mufti, et al., “Luspatercept in Patients With Lower-Risk Myelodysplastic Syndromes,” New England Journal of Medicine 382, no. 2 (2020): 140-151.

[191]

M. D. Cappellini and A. T. Taher, “The Use of Luspatercept for Thalassemia in Adults,” Blood Advances 5, no. 1 (2021): 326-333.

[192]

M. D. Cappellini, V. Viprakasit, A. T. Taher, et al., “A Phase 3 Trial of Luspatercept in Patients With Transfusion-Dependent Β-Thalassemia,” New England Journal of Medicine 382, no. 13 (2020): 1219-1231.

[193]

A. T. Taher, M. D. Cappellini, A. Kattamis, et al., “Luspatercept for the Treatment of Anaemia in Non-Transfusion-Dependent β-Thalassaemia (BEYOND): A Phase 2, Randomised, Double-Blind, Multicentre, Placebo-Controlled Trial,” Lancet Haematology 9, no. 10 (2022): e733-e744.

[194]

A. Bandyopadhyay, F. López-Casillas, S. N. Malik, et al., “Antitumor Activity of a Recombinant Soluble Betaglycan in Human Breast Cancer Xenograft,” Cancer Research 62, no. 16 (2002): 4690-4695.

[195]

A. P. Conley, C. L. Roland, A. Bessudo, et al., “Beta Prime: A First-in-Man Phase 1 Study of AdAPT-001, An Armed Oncolytic Adenovirus for Solid Tumors,” Cancer Gene Therapy 31, no. 4 (2024): 517-526.

[196]

C. Su, J. Li, L. Zhang, et al., “The Biological Functions and Clinical Applications of Integrins in Cancers,” Frontiers in Pharmacology 11 (2020): 579068.

[197]

J. D. Hood and D. A. Cheresh, “Role of Integrins in Cell Invasion and Migration,” Nature Reviews Cancer 2, no. 2 (2002): 91-100.

[198]

D. Valdembri and G. Serini, “The Roles of Integrins in Cancer,” Faculty Reviews 10 (2021): 45.

[199]

H. Hamidi and J. Ivaska, “Every Step of the Way: Integrins in Cancer Progression and Metastasis,” Nature Reviews Cancer 18, no. 9 (2018): 533-548.

[200]

C. Bergonzini, K. Kroese, A. J. M. Zweemer, and E. H. J. Danen, “Targeting Integrins for Cancer Therapy - Disappointments and Opportunities,” Frontiers in Cell and Developmental Biology 10 (2022): 863850.

[201]

I. Malenica, J. Adam, S. Corgnac, et al., “Integrin-αV-Mediated Activation of TGF-β Regulates Anti-Tumour CD8 T Cell Immunity and Response to PD-1 Blockade,” Nature Communications 12, no. 1 (2021): 5209.

[202]

E. Dodagatta-Marri, H. Y. Ma, B. Liang, et al., “Integrin αvβ8 on T Cells Suppresses Anti-Tumor Immunity in Multiple Models and Is a Promising Target for Tumor Immunotherapy,” Cell Reports 36, no. 1 (2021): 109309.

[203]

J. Cooper and F. G. Giancotti, “Integrin Signaling in Cancer: Mechanotransduction, Stemness, Epithelial Plasticity, and Therapeutic Resistance,” Cancer Cell 35, no. 3 (2019): 347-367.

[204]

S. L. Nishimura, “Integrin-Mediated Transforming Growth Factor-β Activation, a Potential Therapeutic Target in Fibrogenic Disorders,” American Journal of Pathology 175, no. 4 (2009): 1362-1370.

[205]

N. F. Brown and J. F. Marshall, “Integrin-Mediated TGFβ Activation Modulates the Tumour Microenvironment,” Cancers 11, no. 9 (2019): 1221.

[206]

V. Sarrazy, A. Koehler, M. L. Chow, et al., “Integrins αvβ5 and αvβ3 Promote Latent TGF-β1 Activation by Human Cardiac Fibroblast Contraction,” Cardiovascular Research 102, no. 3 (2014): 407-417.

[207]

A. Bagati, S. Kumar, P. Jiang, et al., “Integrin αvβ6-TGFβ-SOX4 Pathway Drives Immune Evasion in Triple-Negative Breast Cancer,” Cancer Cell 39, no. 1 (2021): 54-67.e9.

[208]

M. Zhou, J. Niu, J. Wang, et al., “Integrin αvβ8 Serves as a Novel Marker of Poor Prognosis in Colon Carcinoma and Regulates Cell Invasiveness Through the Activation of TGF-β1,” Journal of Cancer 11, no. 13 (2020): 3803-3815.

[209]

L. A. Koopman Van Aarsen, D. R. Leone, S. Ho, et al., “Antibody-Mediated Blockade of Integrin αvβ6 Inhibits Tumor Progression In Vivo by a Transforming Growth Factor-β-Regulated Mechanism,” Cancer Research 68, no. 2 (2008): 561-570.

[210]

J. Stockis, S. Liénart, D. Colau, et al., “Blocking Immunosuppression by Human Tregs In Vivo With Antibodies Targeting Integrin Αvβ8,” Proceedings of the National Academy of Sciences 114, no. 47 (2017): E10161-E10168.

[211]

N. Takasaka, R. I. Seed, A. Cormier, et al., “Integrin αvβ8-Expressing Tumor Cells Evade Host Immunity by Regulating TGF-β Activation in Immune Cells,” JCI Insight 3, no. 20 (2018): e122591.

[212]

D. Wang, V. Shinde, M. Singh, R. Brake, and A. Kolodziej, “Abstract 706: CRB-601, An avβ8 Blocking Antibody, Prevents Activation of TGFb and Exhibits Anti-Tumor Activity Associated With Immune Cell Remodeling of the Tumor Microenvironment,” Cancer Research 83, no. 7_Suppl (2023): 706.

[213]

C. S. Reader, S. Vallath, C. W. Steele, et al., “The Integrin αvβ6 Drives Pancreatic Cancer Through Diverse Mechanisms and Represents an Effective Target for Therapy,” Journal of Pathology 249, no. 3 (2019): 332-342.

[214]

T. Yap, M. Barve, J. Gainor, et al., 532 First-in-Human Phase 1 Trial of SRK-181: A Latent TGFβ1 Inhibitor, Alone or in Combination With Anti-PD-(L)1 Treatment in Patients With Advanced Solid Tumors (DRAGON trial). Regular and Young Investigator Award Abstracts. Published online. 2021: A563-A563.

[215]

C. J. Martin, A. Datta, C. Littlefield, et al., “Selective Inhibition of TGFβ1 Activation Overcomes Primary Resistance to Checkpoint Blockade Therapy by Altering Tumor Immune Landscape,” Science Translational Medicine 12, no. 536 (2020): eaay8456.

[216]

R. J. Kelly, J. L. Gulley, and G. Giaccone, “Targeting the Immune System in Non-Small-Cell Lung Cancer: Bridging the Gap Between Promising Concept and Therapeutic Reality,” Clinical Lung Cancer 11, no. 4 (2010): 228-237.

[217]

J. Nemunaitis, R. O. Dillman, P. O. Schwarzenberger, et al., “Phase II Study of Belagenpumatucel-L, a Transforming Growth Factor Beta-2 Antisense Gene-Modified Allogeneic Tumor Cell Vaccine in Non-Small-Cell Lung Cancer,” Journal of Clinical Oncology 24, no. 29 (2006): 4721-4730.

[218]

G. Giaccone, L. A. Bazhenova, J. Nemunaitis, et al., “A Phase III Study of Belagenpumatucel-L, An Allogeneic Tumour Cell Vaccine, as Maintenance Therapy for Non-Small Cell Lung Cancer,” European Journal of Cancer 51, no. 16 (2015): 2321-2329.

[219]

E. J. Lipson, W. H. Sharfman, S. Chen, et al., “Safety and Immunologic Correlates of Melanoma GVAX, a GM-CSF Secreting Allogeneic Melanoma Cell Vaccine Administered in the Adjuvant Setting,” Journal of Translational Medicine 13, no. 1 (2015): 214.

[220]

J. Nemunaitis, M. Nemunaitis, N. Senzer, et al., “Phase II Trial of Belagenpumatucel-L, a TGF-β2 Antisense Gene Modified Allogeneic Tumor Vaccine in Advanced Non Small Cell Lung Cancer (NSCLC) Patients,” Cancer Gene Therapy 16, no. 8 (2009): 620-624.

[221]

J. E. Ward and D. G. McNeel, “GVAX: An Allogeneic, Whole-Cell, GM-CSF-Secreting Cellular Immunotherapy for the Treatment of Prostate Cancer,” Expert Opinion on Biological Therapy 7, no. 12 (2007): 1893-1902.

[222]

I. Rastogi, A. Muralidhar, and D. G. McNeel, “Vaccines as Treatments for Prostate Cancer,” Nature Reviews Urology 20, no. 9 (2023): 544-559.

[223]

M. Yarchoan, C. Y. Huang, Q. Zhu, et al., “A Phase 2 Study of GVAX Colon Vaccine With Cyclophosphamide and Pembrolizumab in Patients With Mismatch Repair Proficient Advanced Colorectal Cancer,” Cancer Medicine 9, no. 4 (2020): 1485-1494.

[224]

A. A. Wu, K. M. Bever, W. J. Ho, et al., “A Phase II Study of Allogeneic GM-CSF-Transfected Pancreatic Tumor Vaccine (GVAX) With Ipilimumab as Maintenance Treatment for Metastatic Pancreatic Cancer,” Clinical Cancer Research 26, no. 19 (2020): 5129-5139.

[225]

R. P. Rocconi, E. A. Grosen, S. A. Ghamande, et al., “Gemogenovatucel-T (Vigil) Immunotherapy as Maintenance in Frontline Stage III/IV Ovarian Cancer (Vital): A Randomised, Double-Blind, Placebo-Controlled, Phase 2b Trial,” Lancet Oncology 21, no. 12 (2020): 1661-1672.

[226]

Y. Zhang, L. Zhang, Y. Zhao, S. Wang, and L. Feng, “Efficacy and Safety of Gemogenovatucel-T (Vigil) Immunotherapy for Advanced Ovarian Carcinoma: A Systematic Review and Meta-Analysis of Randomized Controlled Trials,” Frontiers in Oncology 12 (2022): 945867.

[227]

M. Gagliardi and A. T. Ashizawa, “The Challenges and Strategies of Antisense Oligonucleotide Drug Delivery,” Biomedicines 9, no. 4 (2021): 433.

[228]

P. S. Rana, D. C. Soler, J. Kort, and J. J. Driscoll, “Targeting TGF-β Signaling in the Multiple Myeloma Microenvironment: Steering CARs and T Cells in the Right Direction,” Frontiers in Cell and Developmental Biology 10 (2022): 1059715.

[229]

E. Tamara, H. Michael, J. Piotr, et al., “Preclinical Efficacy Data on TGF-Beta1 Suppression in Colorectal and Non-Small Cell Lung Cancer by the Antisense Oligonucleotide AP 11014,” Cancer Research 64, no. 7_Suppl (2004): 1019.

[230]

F. Jaschinski, T. Rothhammer, P. Jachimczak, C. Seitz, A. Schneider, and K. H. Schlingensiepen, “The Antisense Oligonucleotide Trabedersen (AP 12009) for the Targeted Inhibition of TGF-β2,” Current Pharmaceutical Biotechnology 12, no. 12 (2011): 2203-2213.

[231]

H. Oettle, A. Hilbig, T. Seufferlein, et al., “Phase I/II Study With Trabedersen (AP 12009) Monotherapy for the Treatment of Patients With Advanced Pancreatic Cancer, Malignant Melanoma, and Colorectal Carcinoma,” Journal of Clinical Oncology 29, no. 15_suppl (2011): 2513.

[232]

M. de Miguel and E. Calvo, “Clinical Challenges of Immune Checkpoint Inhibitors,” Cancer Cell 38, no. 3 (2020): 326-333.

[233]

Y. Jiang and H. Zhan, “Communication Between EMT and PD-L1 Signaling: New Insights Into Tumor Immune Evasion,” Cancer Letters 468 (2020): 72-81.

[234]

M. Z. Noman, B. Janji, A. Abdou, et al., “The Immune Checkpoint Ligand PD-L1 Is Upregulated in EMT-Activated Human Breast Cancer Cells by a Mechanism Involving ZEB-1 and miR-200,” Oncoimmunology 6, no. 1 (2017): e1263412.

[235]

A. Dongre and R. A. Weinberg, “New Insights Into the Mechanisms of Epithelial-Mesenchymal Transition and Implications for Cancer,” Nature Reviews Molecular Cell Biology 20, no. 2 (2019): 69-84.

[236]

S. Bao, X. Jiang, S. Jin, P. Tu, and J. Lu, “TGF-β1 Induces Immune Escape by Enhancing PD-1 and CTLA-4 Expression on T Lymphocytes in Hepatocellular Carcinoma,” Frontiers in Oncology 11 (2021): 694145.

[237]

S. Funaki, Y. Shintani, T. Kawamura, R. Kanzaki, M. Minami, and M. Okumura, “Chemotherapy Enhances Programmed Cell Death 1/Ligand 1 Expression via TGF-β Induced Epithelial Mesenchymal Transition in Non-Small Cell Lung Cancer,” Oncology Reports 38, no. 4 (2017): 2277-2284.

[238]

A. J. Hou, Z. L. Chang, M. H. Lorenzini, E. Zah, and Y. Y. Chen, “TGF-β-responsive CAR-T Cells Promote Anti-Tumor Immune Function,” Bioengineering & Translational Medicine 3, no. 2 (2018): 75-86.

[239]

M. Liu, F. Kuo, K. J. Capistrano, et al., “TGF-β Suppresses Type 2 Immunity to Cancer,” Nature 587, no. 7832 (2020): 115-120.

[240]

M. D. O'connor-Mccourt, A. E. Lenferink, J. Zwaagstra, et al., “Abstract 4688: AVID200: A Novel Computationally-Designed TGF Beta Trap Promoting Anti-Tumor T Cell Activity,” Cancer Research 77, no. 13_Suppl (2017): 4688.

[241]

S. Li, M. Liu, M. H. Do, et al., “Cancer Immunotherapy via Targeted TGF-β Signalling Blockade in TH Cells,” Nature 587, no. 7832 (2020): 121-125.

[242]

Y. Wang, Z. Gao, X. Du, et al., “Co-Inhibition of the TGF-β Pathway and the PD-L1 Checkpoint by pH-Responsive Clustered Nanoparticles for Pancreatic Cancer Microenvironment Regulation and Anti-Tumor Immunotherapy,” Biomaterials Science 8, no. 18 (2020): 5121-5132.

[243]

A. Wallace, V. Kapoor, J. Sun, et al., “Transforming Growth Factor-β Receptor Blockade Augments the Effectiveness of Adoptive T-Cell Therapy of Established Solid Cancers,” Clinical Cancer Research 14, no. 12 (2008): 3966-3974.

[244]

M. Shou, H. Zhou, and L. Ma, “New Advances in Cancer Therapy Targeting TGF-β Signaling Pathways,” Molecular Therapy - Oncolytics 31 (2023): 100755.

[245]

M. Hong, J. D. Clubb, and Y. Y. Chen, “Engineering CAR-T Cells for Next-Generation Cancer Therapy,” Cancer Cell 38, no. 4 (2020): 473-488.

[246]

M. B. Khawar, F. Ge, A. Afzal, and H. Sun, “From Barriers to Novel Strategies: Smarter CAR T Therapy Hits Hard to Tumors,” Frontiers in Immunology 14 (2023): 1203230.

[247]

Y. Qin and G. Xu, “Enhancing Car T-Cell Therapies Against Solid Tumors: Mechanisms and Reversion of Resistance,” Frontiers in Immunology 13 (2022): 1053120.

[248]

C. C. Kloss, J. Lee, A. Zhang, et al., “Dominant-Negative TGF-β Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation and Augments Prostate Cancer Eradication,” Molecular Therapy 26, no. 7 (2018): 1855-1866.

[249]

J. Sun, G. Dotti, L. E. Huye, et al., “T Cells Expressing Constitutively Active Akt Resist Multiple Tumor-Associated Inhibitory Mechanisms,” Molecular Therapy 18, no. 11 (2010): 2006-2017.

[250]

T. L. Roth, P. J. Li, F. Blaeschke, et al., “Pooled Knockin Targeting for Genome Engineering of Cellular Immunotherapies,” Cell 181, no. 3 (2020): 728-744.e21.

[251]

A. B. Roberts and L. M. Wakefield, “The Two Faces of Transforming Growth Factor β in Carcinogenesis,” Proceedings of the National Academy of Sciences 100, no. 15 (2003): 8621-8623.

[252]

K. J. Gordon and G. C. Blobe, “Role of Transforming Growth Factor-β Superfamily Signaling Pathways in Human Disease,” Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1782, no. 4 (2008): 197-228.

[253]

S. Colak and P. ten Dijke, “Targeting TGF-β Signaling in Cancer,” Trends in Cancer 3, no. 1 (2017): 56-71.

[254]

J. Rodon, M. A. Carducci, J. M. Sepulveda-Sánchez, et al., “First-in-Human Dose Study of the Novel Transforming Growth Factor-β Receptor I Kinase Inhibitor LY2157299 Monohydrate in Patients With Advanced Cancer and Glioma,” Clinical Cancer Research 21, no. 3 (2015): 553-560.

[255]

J. G. Quatromoni, E. Suzuki, O. Okusanya, et al., “The Timing of TGF-β Inhibition Affects the Generation of Antigen-Specific CD8+ T Cells,” BMC Immunology 14, no. 1 (2013): 30.

[256]

C. Egbuna, K. C. Patrick-Iwuanyanwu, E. N. Onyeike, et al., “Phytochemicals and Bioactive Compounds Effective Against Acute Myeloid Leukemia: A Systematic Review,” Food Science & Nutrition 11, no. 7 (2023): 4191-4210.

[257]

C. Egbuna, K. C. Patrick-Iwuanyanwu, E. N. Onyeike, et al., “Wnt/β-Catenin Signaling Pathway Inhibitors, Glycyrrhizic Acid, Solanine, Polyphyllin I, Crocin, Hypericin, Tubeimoside-1, Diosmin, and Rutin in Medicinal Plants Have Better Binding Affinities and Anticancer Properties: Molecular Docking and Admet Study,” Food Science & Nutrition 11, no. 7 (2023): 4155-4169.

[258]

X. Liu, Y. Zhang, G. J. Zhou, et al., “Natural Alkaloid 8-Oxo-Epiberberine Inhibited TGF-β1-Triggred Epithelial-Mesenchymal Transition by Interfering Smad3,” Toxicology and Applied Pharmacology 404 (2020): 115179.

[259]

A. Maier, A. L. Peille, V. Vuaroqueaux, and M. Lahn, “Anti-Tumor Activity of the TGF-β Receptor Kinase Inhibitor Galunisertib (LY2157299 Monohydrate) in Patient-Derived Tumor Xenografts,” Cellular Oncology 38, no. 2 (2015): 131-144.

[260]

U. Ben-David, G. Ha, Y. Y. Tseng, et al., “Patient-Derived Xenografts Undergo Mouse-Specific Tumor Evolution,” Nature Genetics 49, no. 11 (2017): 1567-1575.

[261]

J. S. Munger and D. Sheppard, “Cross Talk Among TGF-β Signaling Pathways, Integrins, and the Extracellular Matrix,” Cold Spring Harbor Perspectives in Biology 3, no. 11 (2011): a005017.

[262]

G. D. Orvis, S. P. Jamin, K. M. Kwan, et al., “Functional Redundancy of TGF-Beta Family Type I Receptors and Receptor-Smads in Mediating Anti-Müllerian Hormone-Induced Müllerian Duct Regression in the Mouse1,” Biology of Reproduction 78, no. 6 (2008): 994-1001.

[263]

M. Zhang, Y. Y. Zhang, Y. Chen, J. Wang, Q. Wang, and H. Lu, “TGF-β Signaling and Resistance to Cancer Therapy,” Frontiers in Cell and Developmental Biology 9 (2021): 786728.

[264]

M. Y. Wang, W. J. Liu, L. Y. Wu, G. Wang, C. L. Zhang, and J. Liu, “The Research Progress in Transforming Growth Factor-β2,” Cells 12, no. 23 (2023): 2739.

[265]

Z. J. Chia, Y. Cao, P. J. Little, and D. Kamato, “Transforming Growth Factor-β Receptors: Versatile Mechanisms of Ligand Activation,” Acta Pharmacologica Sinica 45, no. 7 (2024): 1337-1348.

[266]

C. Montemagno and G. Pagès, “Resistance to Anti-Angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs,” Frontiers in Cell and Developmental Biology 8 (2020): 584.

[267]

T. Mashima, T. Wakatsuki, N. Kawata, et al., “Neutralization of the Induced VEGF-A Potentiates the Therapeutic Effect of an anti-VEGFR2 Antibody on Gastric Cancer In Vivo,” Scientific Reports 11, no. 1 (2021): 15125.

[268]

B. J. Raphael, R. H. Hruban, A. J. Aguirre, et al., “Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma,” Cancer Cell 32, no. 2 (2017): 185-203.e13.

[269]

A. Lechner, H. Schlößer, S. I. Rothschild, et al., “Characterization of Tumor-Associated T-Lymphocyte Subsets and Immune Checkpoint Molecules in Head and Neck Squamous Cell Carcinoma,” Oncotarget 8, no. 27 (2017): 44418-44433.

[270]

E. Fessler, J. Drost, S. R. van Hooff, et al., “TGFβ Signaling Directs Serrated Adenomas to the Mesenchymal Colorectal Cancer Subtype,” EMBO Molecular Medicine 8, no. 7 (2016): 745-760.

[271]

D. Yang, Y. Sun, L. Hu, et al., “Integrated Analyses Identify a Master MicroRNA Regulatory Network for the Mesenchymal Subtype in Serous Ovarian Cancer,” Cancer Cell 23, no. 2 (2013): 186-199.

[272]

B. D. Lehmann, J. A. Bauer, X. Chen, et al., “Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies,” Journal of Clinical Investigation 121, no. 7 (2011): 2750-2767.

[273]

K. Araki, T. Shimura, H. Suzuki, et al., “E/N-Cadherin Switch Mediates Cancer Progression via TGF-β-Induced Epithelial-To-Mesenchymal Transition in Extrahepatic Cholangiocarcinoma,” British Journal of Cancer 105, no. 12 (2011): 1885-1893.

[274]

M. Kowanetz, U. Valcourt, R. Bergström, C. H. Heldin, and A. Moustakas, “Id2 and Id3 Define the Potency of Cell Proliferation and Differentiation Responses to Transforming Growth Factor β and Bone Morphogenetic Protein,” Molecular and Cellular Biology 24, no. 10 (2004): 4241-4254.

[275]

L. Yu, “TGF-Beta Receptor-Activated p38 Map Kinase Mediates Smad-Independent TGF-Beta Responses,” EMBO Journal 21, no. 14 (2002): 3749-3759.

[276]

Y. Yang, H. H. Yang, B. Tang, et al., “The Outcome of TGFβ Antagonism in Metastatic Breast Cancer Models In Vivo Reflects a Complex Balance Between Tumor-Suppressive and Proprogression Activities of TGFβ,” Clinical Cancer Research 26, no. 3 (2020): 643-656.

[277]

J. Boudreault, N. Wang, M. Ghozlan, and J. J. Lebrun, “Transforming Growth Factor-β/Smad Signaling Inhibits Melanoma Cancer Stem Cell Self-Renewal, Tumor Formation and Metastasis,” Cancers 16, no. 1 (2024): 224.

[278]

T. Biswas, X. Gu, J. Yang, L. G. Ellies, and L. Z. Sun, “Attenuation of TGF-β Signaling Supports Tumor Progression of a Mesenchymal-Like Mammary Tumor Cell Line in a Syngeneic Murine Model,” Cancer Letters 346, no. 1 (2014): 129-138.

[279]

S. J. Modi, A. Tiwari, and V. M. Kulkarni, “Reversal of TGF-β-Induced Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma by Sorafenib, a VEGFR-2 and Raf Kinase Inhibitor,” Current Research in Pharmacology and Drug Discovery 2 (2021): 100014.

[280]

J. H. Jeong, H. J. Jang, S. Kwak, et al., “Novel TGF-β1 Inhibitor Antagonizes TGF-β1-Induced Epithelial-Mesenchymal Transition in Human A549 Lung Cancer Cells,” Journal of Cellular Biochemistry 120, no. 1 (2019): 977-987.

[281]

S. M. Zhu, Y. R. Park, S. Y. Seo, I. H. Kim, S. T. Lee, and S. W. Kim, “Parthenolide Inhibits Transforming Growth Factor β1-Induced Epithelial-Mesenchymal Transition in Colorectal Cancer Cells,” Intestinal Research 17, no. 4 (2019): 527-536.

[282]

M. H. Barcellos-Hoff and J. L. Gulley, “Molecular Pathways and Mechanisms of TGFβ in Cancer Therapy,” Clinical Cancer Research 29, no. 11 (2023): 2025-2033.

[283]

A. Castiglioni, Y. Yang, K. Williams, et al., “Combined PD-L1/TGFβ Blockade Allows Expansion and Differentiation of Stem Cell-Like CD8 T Cells in Immune Excluded Tumors,” Nature Communications 14, no. 1 (2023): 4703.

[284]

T. Li, X. Wang, M. Niu, et al., “Bispecific Antibody Targeting TGF-β and PD-L1 for Synergistic Cancer Immunotherapy,” Frontiers in Immunology 14 (2023): 1196970.

[285]

J. L. Gulley, J. Schlom, M. H. Barcellos-Hoff, et al., “Dual Inhibition of TGF-β and PD-L1: A Novel Approach to Cancer Treatment,” Molecular Oncology 16, no. 11 (2022): 2117-2134.

[286]

J. Kment, D. Newsted, S. Young, et al., “Blockade of TGF-β and PD-L1 by Bintrafusp Alfa Promotes Survival in Preclinical Ovarian Cancer Models by Promoting T Effector and NK Cell Responses,” British Journal of Cancer 130, no. 12 (2024): 2003-2015.

[287]

A. E. Metropulos, H. G. Munshi, and D. R. Principe, “The Difficulty in Translating the Preclinical Success of Combined TGFβ and Immune Checkpoint Inhibition to Clinical Trial,” EBioMedicine 86 (2022): 104380.

[288]

M. Yi, J. Zhang, A. Li, et al., “The Construction, Expression, and Enhanced Anti-Tumor Activity of YM101: A Bispecific Antibody Simultaneously Targeting TGF-β and PD-L1,” Journal of Hematology & Oncology 14, no. 1 (2021): 27.

[289]

M. Yi, Y. Wu, M. Niu, et al., “Anti-TGF-β/PD-L1 Bispecific Antibody Promotes T Cell Infiltration and Exhibits Enhanced Antitumor Activity in Triple-Negative Breast Cancer,” Journal for Immunotherapy of Cancer 10, no. 12 (2022): e005543.

[290]

E. M. O'Reilly, T. Golan, M. Ikeda, et al., “Phase III Study (daNIS-2) of the anti-TGF-β Monoclonal Antibody (mAb) NIS793 With Nab-Paclitaxel/Gemcitabine (NG) Versus NG Alone in Patients (pts) With First-Line Metastatic Pancreatic Ductal Adenocarcinoma (mPDAC),” Journal of Clinical Oncology 40, no. 16_suppl (2022): TPS4193.

[291]

E. Hong, S. Park, A. Ooshima, et al., “Inhibition of TGF-β Signalling in Combination With Nal-Iri Plus 5-Fluorouracil/Leucovorin Suppresses Invasion and Prolongs Survival in Pancreatic Tumour Mouse Models,” Scientific Reports 10, no. 1 (2020): 2935.

[292]

S. O. Imodoye, K. A. Adedokun, and I. O. Bello, “From Complexity to Clarity: Unravelling Tumor Heterogeneity Through the Lens of Tumor Microenvironment for Innovative Cancer Therapy,” Histochemistry and Cell Biology 161, no. 4 (2024): 299-323.

[293]

H. Huang, Y. Zhang, V. Gallegos, et al., “Targeting TGF βR2-Mutant Tumors Exposes Vulnerabilities to Stromal TGF β Blockade in Pancreatic Cancer,” EMBO Molecular Medicine 11, no. 11 (2019): e10515.

[294]

Z. Zi, D. A. Chapnick, and X. Liu, “Dynamics of TGF-β/Smad Signaling,” FEBS Letters 586, no. 14 (2012): 1921-1928.

RIGHTS & PERMISSIONS

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

AI Summary AI Mindmap
PDF

42

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/