Role of Immunity in Spontaneous Cure of Cancer

Review Article


Abstract views: 60 / PDF downloads: 31

Authors

  • Nasira Tajamal

DOI:

https://doi.org/10.58372/2835-6276.1090

Abstract

This review article provides a brief overview of recent developments in a particular topic. In general, this article summarizes the current knowledge on the subject. This provides the reader with an understanding of the subject by discussing the results of a recent research paper.

Interestingly, some cancers have a particular tendency to regress spontaneously. There are several examples of such referrals. Among them, metastatic melanoma (MM), leukemia, lung cancer, and Merkel cell carcinoma deserve special mention. MM is fascinating because it can regress in the final stage of the disease. [42] The significant contribution of host immunity to early tumor genesis has recently been recognized due to our better understanding of the molecular pathways that regulate tumor cell biology and tumor-lymphocyte interactions. [3]

Targeted therapies designed to target oncogenic pathways in tumor cells may also up-regulate the endogenous immune response and tumor microenvironment. [3]Identifying T-cell inhibitory signals has led to the development of immune checkpoint inhibitors, which specifically impede the inhibition of immune effectors, amplifying and potentially expanding the pre-existing immune response against cancer. [3] This immunity against cancer can be enhanced through immunotherapies, mainly in vaccines, which stimulate natural T lymphocyte clones that specifically recognize tumor antigens. [3] Consequently, a promising anticancer therapy will aim to activate patients' natural anticancer immunity, either to eliminate residual tumor cells or to extend the dormancy period in metastatic tumor cells. Such endogenous cancer immunity plays an essential role in controlling the balance between dormant tumor cells and tumor leakage and in limiting metastasis. [3] 

Malignant melanoma (MM) is the most aggressive and difficult-to-treat form of skin cancer. It is estimated that up to 90% of deaths from skin tumors are due to this malignancy. [8] Regression is more common in melanoma than in other types of tumors; it would be six times higher than in other malignant tumors. Up to 50% of primary MM undergo spontaneous regression. However, spontaneous regression of the metastatic form of the tumor is a rare phenomenon observed in only 0.23% of cases. [8]The most frequently mentioned factors leading to spontaneous regression of MM are surgical trauma, infection, vaccination (BCG and rabies vaccines), and immunological factors. Other well-documented conditions associated with the regression of metastatic MM are blood transfusion and various endocrine factors. [8] 

Among the New Age methods, auto fiction, alternative gene therapy, combined bacteriolytic therapy, and enzyme-prodrug therapy targeting bacteria are some of the potential cancer treatment modalities that use microorganisms. There is an interconnection between microorganisms, the response of the immune system, and possible mechanisms involved in spontaneous tumor regression. [33] The role of NK cells evolves with tumor progression. It offers new opportunities to manipulate NK cell function to improve response rates to cancer immunotherapy in a broader range of cancers. [23] 

Self-recognition of missing target cells. Signaling from activating and inhibitory receptors regulates the activation of natural killer (NK) cells. Stressed cells, such as tumor cells, lose their MHC class I molecules, a ligand for inhibitory receptors on NK cells. At the same time, they acquire stress-associated molecules that act as ligands for receptor activation. Thus, the absence of inhibitory signaling combined with the induction of activating signaling shifts the balance towards the activation of NK cells, leading to the secretion of cytokines and the destruction of tumor cells.

References

Abdelrazeq, A. S. (2007). Spontaneous regression of colorectal cancer: a review of cases from 1900 to 2005. International journal of colorectal disease, 22(7), 727-736.

Balkwill, F. (2006). TNF-α in promotion and progression of cancer. Cancer and Metastasis Reviews, 25(3), 409-416.

Baxevanis, C. N., & Perez, S. A. (2015). Cancer dormancy: a regulatory role for endogenous immunity in establishing and maintaining the tumor dormant state. Vaccines, 3(3), 597-619.

Bilich T, Nelde A, Bichmann L, Roerden M, Salih HR, Kowalewski DJ, Schuster H, Tsou CC, Marcu A, Neidert MC, Lübke M, Rieth J, Schemionek M, Brümmendorf TH, Vucinic V, Niederwieser D, Bauer J, Märklin M, Peper JK, Klein R, Kohlbacher O, Kanz L, Rammensee HG, Stevanović S, Walz JS. The HLA ligandome landscape of chronic myeloid leukemia delineates novel T-cell epitopes for immunotherapy. Blood. 2019 Feb 7;133(6):550-565. doi: 10.1182/blood-2018-07-866830. Epub 2018 Dec 10. PMID: 30530751.

Cann, S. H., Van Netten, J. P., Van Netten, C., & Glover, D. W. (2002). Spontaneous regression: a hidden treasure buried in time. Medical hypotheses, 58(2), 115-119.

Cavallo F, Di Carlo E, Butera M, Verrua R, Colombo MP, Musiani P, Forni G. Immune events associated with the cure of established tumors and spontaneous metastases by local and systemic interleukin 12. Cancer Res. 1999 Jan 15; 59(2):414-21. PMID: 9927056.

Cavallo, F., Di Carlo, E., Butera, M., Verrua, R., Colombo, M. P., Musiani, P., & Forni, G. (1999). Immune events associated with the cure of established tumors and spontaneous metastases by local and systemic interleukin 12. Cancer Research, 59(2), 414-421.

Du N, Guo F, Wang Y, Cui J. NK Cell Therapy: A Rising Star in Cancer Treatment. Cancers (Basel). 2021 Aug 17;13(16):4129. doi: 10.3390/cancers13164129. PMID: 34439285; PMCID: PMC8394762.

Cervinkova, M., Kucerova, P., & Cizkova, J. (2017). Spontaneous regression of malignant melanoma is based on the interplay between the host immune system and melanoma antigens. Anti-cancer drugs, 28(8), 819-830.

Cole, W. H. (1981). Efforts to explain spontaneous regression of cancer. Journal of Surgical Oncology, 17 (3), 201-209.

Cornberg M, Wong VW, Locarnini S, Brunetto M, Janssen HLA, Chan HL. The role of quantitative hepatitis B surface antigen revisited. J Hepatol. 2017 Feb;66(2):398-411. doi: 10.1016/j.jhep.2016.08.009. Epub 2016 Aug 27. PMID: 27575311.

Curiel, T. J. (2007). Tregs and rethinking cancer immunotherapy. The Journal of Clinical Investigation, 117(5), 1167-1174.

De Giovanni C, Nicoletti G, Landuzzi L, Rossi I, Astolfi A, Ricci C, Di Carlo E, Musiani P, Forni G, Fradelizi D, Nanni P, Lollini PL. Therapy of lung metastases through combined vaccination with

carcinoma cells engineered to release IL-13 and IFN-gamma. Gene Ther. 2001 Nov;8(22):1698-704. doi: 10.1038/sj.gt.3301584. PMID: 11892837.

De Visser, K. E., Eichten, A., & Coussens, L. M. (2006). Paradoxical roles of the immune system during cancer development. Nature Reviews Cancer, 6(1), 24-37.

Dobosz, P., & Dzieciątkowski, T. (2019). The intriguing history of cancer immunotherapy. Frontiers in immunology, 10, 2965.

Doyle, H. A., Gee, R. J., Masters, T. D., Gee, C. R., Booth, C. J., Peterson-Roth, E., ... & Mamula, M. J. (2021). Vaccine-induced ErbB (EGFR/HER2)-specific immunity in spontaneous canine cancer. Translational oncology, 14(11), 101205.

Dunn, G. P., Koebel, C. M., & Schreiber, R. D. (2006). Interferons, immunity, and cancer immunoediting. Nature Reviews Immunology, 6(11), 836-848.

Dunn, G. P., Old, L. J., & Schreiber, R. D. (2004). The immunobiology of cancer immunosurveillance and immunoediting. Immunity, 21(2), 137-148.

Dunn, G. P., Old, L. J., & Schreiber, R. D. (2004). The three Es of cancer immunoediting. Annu. Rev. Immunol., 22, 329-360.

Foon KA, Smalley RV, Riggs CW, Gale RP. The role of immunotherapy in acute myelogenous leukemia. Arch Intern Med. 1983 Sep;143(9):1726-31. PMID: 6577818.

Grivennikov, S. I., Greten, F. R., & Karin, M. (2010). Immunity, inflammation, and cancer. Cell, 140(6), 883-899.

Gu T, Kilinc MO, Egilmez NK. Transient activation of tumor-associated T-effector/memory cells promotes tumor eradication via NK-cell recruitment: a minimal role for long-term T-cell immunity in the cure of metastatic disease. Cancer Immunol Immunother. 2008 Jul;57(7):997-1005. doi: 10.1007/s00262-007-0430-0. PMID: 18049819.

Hellstrom, K. E., & Hellstrom, I. N. G. E. G. E. R. D. (1972). Immunity to neuroblastomas and melanomas. Annual review of medicine, 23(1), 19-38.

Huntington, N. D., Cursons, J., & Rautela, J. (2020). The cancer–natural killer cell immunity cycle. Nature Reviews Cancer, 1-18.

Ichim, C. V. (2005). Revisiting immunosurveillance and immunostimulation: Implications for cancer immunotherapy. Journal of translational medicine, 3(1), 1-13.

Ikeda, H., Chamoto, K., Tsuji, T., Suzuki, Y., Wakita, D., Takeshima, T., & Nishimura, T. (2004). The critical role of type‐1 innate and acquired immunity in tumor immunotherapy. Cancer Science, 95(9), 697-703.

Janssen, L. M., Ramsay, E. E., Logsdon, C. D., & Overwijk, W. W. (2017). The immune system in cancer metastasis: friend or foe? Journal for immunotherapy of cancer, 5(1), 1-14.

Jessy T. Immunity over inability: The spontaneous regression of cancer. J Nat Sci Biol Med. 2011 Jan;2(1):43-9. doi: 10.4103/0976-9668.82318. PMID: 22470233; PMCID: PMC3312698.

Jessy, T. (2011). Immunity over inability: The spontaneous regression of cancer. Journal of natural science, biology, and medicine, 2(1), 43.

Kappauf, H., Gallmeier, W. M., Wünsch, P. H., Mittelmeier, H. O., Birkmann, J., Büschel, G., ... & Kraus, J. (1997). Complete spontaneous remission in a patient with metastatic non-small-cell lung cancer: case report, review of literature, and discussion of possible biological pathways involved. Annals of Oncology, 8(10), 1031-1039.

Khong, H. T., & Restifo, N. P. (2002). Natural selection of tumor variants in the generation of “tumor escape” phenotypes. Nature Immunology, 3(11), 999-1005.

Kim, R. (2007). Cancer immunoediting: from immune surveillance to immune escape. Cancer Immunotherapy, 9-27.

Koebel, C. M., Vermi, W., Swann, J. B., Zerafa, N., Rodig, S. J., Old, L. J., ... & Schreiber, R. D. (2007). Adaptive immunity maintains occult cancer in an equilibrium state. Nature, 450(7171), 903-907.

Kucerova, P., & Cervinkova, M. (2016). Spontaneous regression of tumor and the role of microbial infection–possibilities for cancer treatment. Anti-Cancer Drugs, 27(4), 269.

Lollini, P. L., & Forni, G. (1999). Specific and nonspecific immunity in the prevention of spontaneous tumors. Immunology Today, 20(8), 347-350.

Lubet RA, Carlson DE. Therapy of the murine plasmacytoma MOPC 104E: role of the immune response. J Natl Cancer Inst. 1978 Sep;61(3):897-903. PMID: 278867.

Mantovani, A., Romero, P., Palucka, A. K., & Marincola, F. M. (2008). Tumor immunity: effector response to tumor and role of the microenvironment. The Lancet, 371(9614), 771-783.

Matsuzawa A, Kaneko T, Takeda Y, Ozawa H. Characterization of a strongly immunogenic mouse leukemia line (DL811) undergoing spontaneous cure with tumor dormancy. Cancer Lett. 1991 Jun 14;58(1-2):69-74. doi: 10.1016/0304-3835(91)90025-d. PMID: 2049784.

Mylvaganam G, Yanez AG, Maus M, Walker BD. Toward T Cell-Mediated Control or Elimination of HIV Reservoirs: Lessons From Cancer Immunology. Front Immunol. 2019 Sep 10;10:2109. doi: 10.3389/fimmu.2019.02109. PMID: 31552045; PMCID: PMC6746828.

Nair RE, Kilinc MO, Jones SA, Egilmez NK. Chronic immune therapy induces a progressive increase in intratumoral T suppressor activity and a concurrent loss of tumor-specific CD8+ T effectors in her-2/neu transgenic mice bearing advanced spontaneous tumors. J Immunol. 2006 Jun 15;176(12):7325-34. doi: 10.4049/jimmunol.176.12.7325. PMID: 16751376.

Ni X, Wang Q, Gu J, Lu L. Clinical and Basic Research Progress on Treg-Induced Immune Tolerance in Liver Transplantation. Front Immunol. 2021 May 20;12:535012. doi: 10.3389/fimmu.2021.535012. PMID: 34093514; PMCID: PMC8173171.

Penn, I. (1981). Depressed immunity and the development of cancer. Clinical and experimental immunology, 46(3), 459.

Radha, G., & Lopus, M. (2021). The spontaneous remission of cancer: Current insights and therapeutic significance. Translational Oncology, 14(9), 101166.

Rogers, L. M., Olivier, A. K., Meyerholz, D. K., & Dupuy, A. J. (2013). Adaptive immunity does not strongly suppress spontaneous tumors in a Sleeping Beauty model of cancer. The Journal of Immunology, 190(8), 4393-4399.

Saegusa Y, Eriguchi M, Kidani Y, Matsuzawa A, Takeda Y. Antitumor effects of a new lipophilic platinum compound, trans-bis(n-valerate)(1R, 2R-cyclohexane diamine)(oxalate)platinum(IV), in mice. Anticancer Res. 2001 Jan-Feb; 21(1A):245-52. PMID: 11299742.

Satgé, D. (2018). A tumor profile in primary immune deficiencies challenges the cancer immune surveillance concept. Frontiers in immunology, 9, 1149.

Schreiber, R. D., Old, L. J., & Smyth, M. J. (2011). Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science, 331(6024), 1565-1570.

Smyth, M. J., Dunn, G. P., & Schreiber, R. D. (2006). Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Advances in immunology, 90, 1-50.

Smyth, Mark J., Erika Cretney, Michael H. Kershaw, and Yoshihiro Hayakawa. "Cytokines in cancer immunity and immunotherapy." Immunological Reviews 202, no. 1 (2004): 275-293.

Swann, J. B., & Smyth, M. J. (2007). Immune surveillance of tumors. The Journal of Clinical Investigation, 117(5), 1137-1146.

Thomas, J. A., & Badini, M. (2011). The role of innate immunity in spontaneous regression of cancer. Indian journal of cancer, 48(2), 246.

Tu SM, Pisters LL. Curing Cancer: Lessons from a Prototype. Cancers (Basel). 2021 Feb 7;13(4):660. doi: 10.3390/cancers13040660. PMID: 33562202; PMCID: PMC7915721.

Vernon, L. F. (2018). William Bradley Coley, MD, and the phenomenon of spontaneous regression. Immune Targets and therapy, 7, 29.

Vesely, M. D., Kershaw, M. H., Schreiber, R. D., & Smyth, M. J. (2011). Natural innate and adaptive immunity to cancer. Annual review of immunology, 29, 235-271.

Waldman, A.D., Fritz, J.M. & Lenardo, M.J. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 20, 651–668 (2020). https://doi.org/10.1038/s41577-020-0306-5

Zitvogel, L., Tesniere, A., & Kroemer, G. (2006). Cancer despite immunosurveillance: immunoselection and immunosubversion. Nature Reviews Immunology, 6(10), 715-727. Du, N., Guo, F., Wang, Y., & Cui, J. (2021). NK.

Downloads

Published

2023-11-02

How to Cite

Nasira Tajamal. (2023). Role of Immunity in Spontaneous Cure of Cancer: Review Article. American Journal of Medical and Clinical Research & Reviews, 2(11), 1–17. https://doi.org/10.58372/2835-6276.1090

Issue

Section

Articles