Targeted Therapy and Its Types: Advances, Challenges, and the Role of Bacterial Toxins in Cancer Treatment

Main Article Content

Mohammad Maroufi
Tayyebeh Khoshbakht
Maryam Rastgoo
Matin Vafaei

Abstract

Cancer continues to be a major global health challenge, ranking among the leading causes of mortality despite advancements in conventional treatments such as surgery, chemotherapy, and radiotherapy. Targeted therapy has revolutionized cancer treatment by selectively targeting molecular abnormalities in cancer cells, offering enhanced efficacy and reduced toxicity compared to traditional approaches. This review provides a comprehensive analysis of the principal types of targeted therapy, including monoclonal antibodies, small molecule inhibitors, and emerging modalities such as antibody-drug conjugates and immune checkpoint inhibitors. It also examines the challenges posed by resistance mechanisms, including genetic mutations, tumor microenvironment adaptations, and efflux pump overexpression, which limit therapeutic success. Furthermore, the review explores the innovative application of bacterial toxins, such as diphtheria toxin and Pseudomonas exotoxin, as novel targeted therapeutic agents due to their potent cytotoxic properties and ability to be engineered for specificity. The integration of bioinformatics and artificial intelligence in identifying actionable molecular targets, predicting resistance patterns, and designing personalized treatment strategies is also discussed.

Article Details

Section
Reveiw Article

References

1. Kordkatouli, M., Sateei, A., & Dulskas, A. (2024). Potential roles and mechanisms of Avena sativa in cancer prevention. Multidisciplinary Cancer Investigation 8(2);, 0-0.

2. Ramos, P. and M. Bentires-Alj, Mechanism-based cancer therapy: resistance to therapy, therapy for resistance. Oncogene, 2015. 34(28): p. 3617-3626.

3. Berger, M.F. and E.R. Mardis, The emerging clinical relevance of genomics in cancer medicine. Nature reviews Clinical oncology, 2018. 15(6): p. 353-365.

4. Kordkatouli, M., Janlou, M. A. M., Sateei, A., Mousavi, M. M. H., & Dulskas, A. Recent Progress in Nanoparticle-Driven Drug Delivery Strategies for Cancer Therapy: Focus on Colorectal Cancer. Zahedan Journal of Research in Medical Sciences, 27(1).

5. Köhler, G. and C. Milstein, Continuous cultures of fused cells secreting antibody of predefined specificity. nature, 1975. 256(5517): p. 495-497.

6. Zahavi, D., and Louis Weiner. 2020. "Monoclonal Antibodies in Cancer Therapy" Antibodies 9, no. 3: 34. https://doi.org/10.3390/antib9030034, Monoclonal Antibodies in Cancer Therapy. 2020. 9(3).

7. Murphy, K., et al., Immunobiology. 2012: Garland Science New York, NY.

8. Weiner, L.M., R. Surana, and S. Wang, Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nature Reviews Immunology, 2010. 10(5): p. 317-327.

9. Li, S., et al., Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer cell, 2005. 7(4): p. 301-311.

10. Patel, D., et al., Anti-epidermal growth factor receptor monoclonal antibody cetuximab inhibits EGFR/HER-2 heterodimerization and activation. International journal of oncology, 2009. 34(1): p. 25-32.

11. Chari, R.V., Targeted cancer therapy: conferring specificity to cytotoxic drugs. Accounts of chemical research, 2008. 41(1): p. 98-107.

12. Shi, D.D., et al., Therapeutic avenues for cancer neuroscience: translational frontiers and clinical opportunities. The Lancet Oncology, 2022. 23(2): p. e62-e74.

13. Gharwan, H. and H. Groninger, Kinase inhibitors and monoclonal antibodies in oncology: clinical implications. Nature reviews Clinical oncology, 2016. 13(4): p. 209-227.

14. Cohen, P., D. Cross, and P.A. Jänne, Kinase drug discovery 20 years after imatinib: progress and future directions. Nature reviews drug discovery, 2021. 20(7): p. 551-569.

15. Bedard, P.L., et al., Small molecules, big impact: 20 years of targeted therapy in oncology. The Lancet, 2020. 395(10229): p. 1078-1088.

16. Liu, G.H., et al., Small molecule inhibitors targeting the cancers. MedComm, 2022. 3(4): p. e181.

17. Heersche, N., et al., Clinical implications of germline variations for treatment outcome and drug resistance for small molecule kinase inhibitors in patients with non-small cell lung cancer. Drug Resistance Updates, 2022. 62: p. 100832.

18. Restifo, N.P., M.J. Smyth, and A. Snyder, Acquired resistance to immunotherapy and future challenges. Nature Reviews Cancer, 2016. 16(2): p. 121-126.

19. Ladoire, S., C. Rébé, and F. Ghiringhelli, Associating immunotherapy and targeted therapies: facts and hopes. Clinical Cancer Research, 2023. 29(7): p. 1183-1193.

20. Choi, H.Y. and J.-E. Chang, Targeted therapy for cancers: from ongoing clinical trials to FDA-approved drugs. International Journal of Molecular Sciences, 2023. 24(17): p. 13618.

21. King's College London. "AI tackles huge problem of antimicrobial resistance in intensive care." ScienceDaily. www.sciencedaily.com/releases/2024/11/241101144411.htm (accessed January 1, AI tackles huge problem of antimicrobial resistance in intensive care. 2024.

22. Minchinton, A.I. and I.F. Tannock, Drug penetration in solid tumours. Nature Reviews Cancer, 2006. 6(8): p. 583-592.

23. Forbes, N.S., Engineering the perfect (bacterial) cancer therapy. Nature Reviews Cancer, 2010. 10(11): p. 785-794.

24. MalmgrenBoland and C.C. Flanigan, Localization of the vegetative form of Clostridium tetani in mouse tumors following intravenous spore administration. Cancer research, 1955. 15(7): p. 473-478.

25. Carswell, E., et al., An endotoxin-induced serum factor that causes necrosis of tumors. Proceedings of the National Academy of Sciences, 1975. 72(9): p. 3666-3670.

26. Nougayrède, J.-P., et al., Cyclomodulins: bacterial effectors that modulate the eukaryotic cell cycle. Trends in microbiology, 2005. 13(3): p. 103-110.

27. Mohseni, Z., et al., Potent in vitro antitumor activity of B-subunit of Shiga toxin conjugated to the diphtheria toxin against breast cancer. European Journal of Pharmacology, 2021. 899: p. 174057.

28. Boland, E.L., et al., Structural complementation of the catalytic domain of pseudomonas exotoxin A. Biophysical Journal, 2014. 106(2): p. 614a.

29. Pang, Z., M.-D. Gu, and T. Tang, Pseudomonas aeruginosa in cancer therapy: current knowledge, challenges and future perspectives. Frontiers in Oncology, 2022. 12: p. 891187.

30. Li, X., et al., Tissue distribution and safety evaluation of a claudin-targeting molecule, the C-terminal fragment of Clostridium perfringens enterotoxin. European Journal of Pharmaceutical Sciences, 2014. 52: p. 132-137.

31. Ansiaux, R.g., et al., Botulinum toxin potentiates cancer radiotherapy and chemotherapy. Clinical cancer research: an official journal of the American Association for Cancer Research, 2006. 12(4): p. 1276-1283.

32. Hall, E.H., et al., Inhibition of human breast cancer Matrigel invasion by Streptolysin O activation of the EGF receptor ErbB1. Cellular signalling, 2011. 23(12): p. 1972-1977.

33. Greenfield, L., V.G. Johnson, and R.J. Youle, Mutations in diphtheria toxin separate binding from entry and amplify immunotoxin selectivity. Science, 1987. 238(4826): p. 536-539.

34. Puri, R., Development of a recombinant interleukin-4-Pseudomonas exotoxin for therapy of glioblastoma. Toxicologic pathology, 1999. 27(1): p. 53-57.

35. Callegan, M.C., et al., Role of hemolysin BL in the pathogenesis of extraintestinal Bacillus cereus infection assessed in an endophthalmitis model. Infection and immunity, 1999. 67(7): p. 3357-3366.

36. Honda, T., et al., Enzyme-linked immunosorbent assays for detection of thermostable direct hemolysin of Vibrio parahaemolyticus. Journal of clinical microbiology, 1985. 22(3): p. 383-386.

37. Nauts, H.C., The Beneficial Effects of Bacterial Infections on Host Resistance to Cancer: End Results in 449 Cases: a Study and Abstracts of Reports in the World Medical Literature (1775-1980) and Personal Communications. 1980: Cancer research institute.

38. Camussi, G., et al., The molecular action of tumor necrosis factor‐α. European Journal of Biochemistry, 1991. 202(1): p. 3-14.

39. Shafiee, F., M.G. Aucoin, and A. Jahanian-Najafabadi, Targeted diphtheria toxin-based therapy: a review article. Frontiers in microbiology, 2019. 10: p. 2340.

40. Kojima, T., et al., Claudin-binder C-CPE mutants enhance permeability of insulin across human nasal epithelial cells. Drug Delivery, 2016. 23(8): p. 2703-2710.

41. Jahn, R., A neuronal receptor for botulinum toxin. Science, 2006. 312(5773): p. 540-541.

42. Fischer, A., et al., Pseudomonas exotoxin A based toxins targeting epidermal growth factor receptor for the treatment of prostate cancer. Toxins, 2020. 12(12): p. 753.

43. Melton-Celsa, A.R., Shiga toxin (Stx) classification, structure, and function. Microbiology spectrum, 2014. 2(4): p. 10.1128/microbiolspec. ehec-0024-2013.