Open access

Introductory Chapter: Are We There Yet? The Long and Winding Road to Cancer Immunotherapy

Written By

Hilal Arnouk and Sana Moqeet

Submitted: 26 July 2019 Published: 13 November 2019

DOI: 10.5772/intechopen.89553

From the Edited Volume

Cancer Immunotherapy and Biological Cancer Treatments

Edited by Hilal Arnouk

Chapter metrics overview

970 Chapter Downloads

View Full Metrics

1. History of cancer immunotherapy

Cancer immunotherapy has become an innovative approach that both pushes the medical and scientific community to better understand our own immune system and charts a new frontier in the fight against one of the leading killers in the world, cancer. The advent of this line of work dates back to the discovery of vaccine. Luminary scientists in the nineteenth century such as Joseph Lister, Louis Pasteur, Robert Koch, and most notably William Coley have allowed for a better understanding of the immune responses and the establishment of vaccines, including vaccines directed against malignant tumors [1, 2, 3, 4].

While the basic concept of using immunotherapy to combat cancer was practiced in the scientific community, specifically by Coley, scientists Thomas and Burney were the first to propose the theory of cancer immunosurveillance in 1957. The premise behind their theory was that lymphocytes, an integral part of the immune system, have the capability to eliminate the mutated cancerous cells throughout the human body [5, 6, 7, 8]. However, due to a lack of scientific proof-of-concept and the inability to culture the lymphocytes ex vivo for extended periods of time, the next development did not occur until a few decades later. From the 1970s onward, the scientific community has discovered the utility of several different immune therapies to treat cancer including interferon alpha (IFN-α) [9], the T cell growth factor interleukin 2 (IL-2), monoclonal antibodies targeting tumor-associated antigens, and the first FDA-approved cell-based cancer vaccine developed for patients with advanced prostate cancer in 2010 (Figure 1).

Figure 1.

Key events in the history of cancer immunotherapy.

This chapter will give a brief overview of few of these therapies that scientists and physicians are currently utilizing in the fight against cancer.

Advertisement

2. Tumor antigens

Tumor cells are distinguished from normal cells in a tissue by the presence of unique proteins known as tumor antigens. They can be further divided into two broad categories, tumor-associated antigens and tumor-specific antigens.

Tumor-associated antigens can arise from oncofetal genes that become aberrantly expressed in malignant cells, such as alpha-fetoprotein (AFP) in liver cancer and the melanoma-specific antigens of the MAGE family [10], or from cancer-testis antigens that are expressed normally in the germ cells and become activated in cancers, such as the New York esophageal squamous cell carcinoma 1 (NY-ESO-1) antigen [11]. Tumor-associated antigens also include tissue differentiation antigens, such as tyrosinase in melanoma [12], and viral antigens in a number of malignancies where viral oncogenesis is implicated, such as HPV E6 and E7 oncoproteins in HPV-associated cervical cancers [13]. On the other hand, tumor-specific antigens are usually the products of mutated oncogenes or tumor suppressor genes, such as ras and p53.

Advertisement

3. Cancer vaccines

While it is well established that vaccines are a way to stimulate the immune system against infectious agents, the concept of harnessing the power of a vaccine to eliminate cancers remains revolutionary. Cancer vaccines work through inducing a specific antitumor T cell response.

Cancer vaccines are divided into prophylactic and therapeutic vaccines. For instance, a prophylactic vaccine against human papillomavirus (HPV) can prevent several cancer types caused by virus, such as cervical cancer and some cancers of the oropharynx [14, 15], and hepatitis B virus (HBV) vaccine provides protection against liver cancers initiated by the hepatitis B virus [16]. Alternatively, therapeutic cancer vaccines trigger an immune response against an existing tumor by inducing T cell response within the tumor microenvironment. Various formats have been developed for therapeutic cancer vaccines, including peptide fragments and full-length tumor antigens, vectors for genetically encoded tumor antigens, whole tumor cell contents, and autologous dendritic cell (DC) vaccines. Most notably, Sipuleucel-T (Provenge) was the first FDA-approved cell-based cancer vaccine developed for patients with hormone-refractory prostate cancer and has shown to prolong the life of affected patients by several months [17, 18].

Since most tumor-associated antigens are not highly immunogenic on their own, an immune adjuvant is typically added to vaccine formulas. Examples of vaccine adjuvants include recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) and heat shock proteins [19, 20, 21].

Advertisement

4. Adoptive cellular immunotherapy

Adoptive cellular immunotherapy (ACT) is the process of transferring effector immune cells, such as T lymphocytes, to cancer patients, including modifying and expanding these immune cells ex vivo to target specific cancer cells [22, 23]. The transferred immune cells can have autologous or allogeneic origin. Two major approaches have been utilized for adoptive cellular immunotherapy, tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor-modified T cell (CAR-T) therapy.

4.1 Tumor-infiltrating lymphocytes (TILs)

A tumor-infiltrating lymphocyte is an immune cell that has moved into a tumor in the attempt to destroy the cancer. In this therapy, TILs are extracted from tumor tissue biopsies and cultured in vitro with IL-2 to expand the tumor-reactive clones. Once activated, lymphocytes are infused back into the patient [24].

4.2 Chimeric antigen receptor-modified T cell (CAR-T) therapy

Chimeric antigen receptor-modified T cell (CAR-T) therapy has emerged as a successful method in the fight against cancer, especially B cell hematologic malignancies [25]. This success lies within the chimeric antigen receptors (CAR) composed of single-chain variable fragment (scFv) and the costimulatory signaling molecules that can stimulate T cells and allow for direct antigen binding and activation, bypassing the requirement for antigen presentation by antigen-presenting cells (APCs).

CAR-T cell therapy has been especially effective against certain types of lymphoma and leukemia, such as refractory B cell lymphoma and acute lymphoblastic leukemia (ALL) [26, 27]. CAR-T cell therapy is a multistep process. First, patients are evaluated to determine if CAR-T cell therapy is an appropriate treatment. Second, T cells are isolated from blood. The patient’s T cells are then taken into the laboratory to be genetically modified to express the chimeric antigen receptors (CARs) on their surfaces. As discussed above, these essential receptors allow for the modified T cells to recognize tumor antigens. Finally, the CAR-T cells are infused back into the patient, and a recovery period of 2–3 months is expected [28].

Overall, CAR-T cell therapy has become a leading area of continued growth and research in the field of cancer immunotherapy. It has had much success in both preclinical and clinical applications, and the next steps involve harnessing the power of modified T cells to aid in eliminating solid tumors including aggressive cancers, such as sarcomas.

4.3 NK cell therapy

Natural killer (NK) cells are large granular lymphocytes that can destroy cells organically, without the requirement for priming first. One of the major hypotheses related to NK cells is the “missing self” hypothesis, which states that NK cells have the ability to destroy cells that do not display major histocompatibility complex (MHC) class I molecules [29]. Given the extraordinary functions of NK cells in recognizing and destroying altered cells, NK cell-based therapies have become a wide area of research for diseases like cancer. One such targeted therapy is the use of NK alloreactivity, specifically in acute leukemia and other types of hematologic cancers. MHC class I inhibitory receptors, specifically killer immunoglobulin-like receptors (KIRs), have been utilized to exert NK cell alloreactivities as a way to combat leukemic cells [30].

Advertisement

5. Antibody-based treatments

Antibodies are arguably one of the most important aspects of the human immune system. They serve to bind to antigens and allow for flagging of cells with specific antigens for eradication. An antitumor monoclonal antibody is a genetically engineered immunoglobulin that is produced to recognize a specific tumor antigen on the surface of a cancer cell [31].

Monoclonal antibodies work in a multitude of ways including flagging cancer cells, blocking growth of cells, blocking immune inhibitors, directly attacking cancer cells, and bridging cancer and immune cells [32]. Monoclonal antibodies come in two different varieties, naked and conjugated. Naked monoclonal antibodies work individually and can attach themselves to antigens on cancer cells. For instance, trastuzumab (Herceptin) can bind to HER2 oncogene/tumor antigen that is overexpressed in a subset of breast cancers, which blocks the growth and proliferation of the malignant cells [33]. Conjugated monoclonal antibodies work by carrying a radioactive or cytotoxic drug into proximity to the tumor cells, thus killing these malignant cells [34, 35]. As with other cancer immunotherapies, antibody-based treatments carry the risk of adverse side effects that include fever, weakness, nausea, and rashes.

Advertisement

6. Immune checkpoint inhibitors

Immune checkpoint inhibitors are inhibitory molecules on immune cells that prevent the immune system from attacking the organism’s own tissues. Two of the identified immune checkpoints are programmed cell death-1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) [36].

Interaction of PD-1 and its ligand PDL-1 leads to T lymphocytes’ dysfunction and exhaustion. PDL-1 is shown to be highly expressed on cancer cells [37]. Consequently, the blockage of PD-1 and PDL-1 interactions using immune checkpoint inhibitors allows tumor-specific T lymphocytes to exert their function by recognizing and destroying cancer cells. Nivolumab and pembrolizumab are monoclonal antibodies that function as PD-1 blockers and are being currently being used to treat patients with advanced melanoma and non-small cell lung cancer [38, 39]. Similarly, CTLA-4 inhibits T cell activation. Thus, CTLA-4 blockage with inhibitors, such as ipilimumab, enhances the immune responses against malignant tumors such as melanoma [40].

It is important to note, however, that the use of immune checkpoint inhibitors in cancer immunotherapy is frequently followed by inflammatory and autoimmune side effects that include endocrine effects, rash, and hepatitis [41, 42].

In conclusion, cancer immunotherapy remains a field that has tremendously changed our understanding of how to best treat cancers. While advancements have been made, there are many more discoveries to be made as the field of tumor immunology is growing exponentially.

References

  1. 1. Gradmann C. A matter of methods: The historicity of Koch’s postulates 1840-2000. Medizinhistorisches Journal. 2008;43:121-148
  2. 2. Herr HW. Ignorance is bliss: The Listerian revolution and education of American surgeons. The Journal of Urology. 2007;177:457-460
  3. 3. Ullmann A. Pasteur-Koch: Distinctive ways of thinking about infectious diseases. Microbe. 2007;2:383-387
  4. 4. Coley WB. II. Contribution to the knowledge of sarcoma. Annals of Surgery. 1891;14(3):199-220
  5. 5. Thomas L. Discussion of cellular and humoral aspects of hypersensitive states. In: Lawrence HS, editor. New York: Hoeber-Harper; 1959
  6. 6. Burnet FM. Cancer A biological approach. 1. The process of control. British Medical Journal. 1957;1:779-782
  7. 7. Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ, et al. Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:7556-7561
  8. 8. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410:1107-1111
  9. 9. Talpaz M, McCredie KB, Mavligit GM, Gutterman JU. Leukocyte interferon-induced myeloid cytoreduction in chronic myelogenous leukemia. Blood. 1983;62:689-692
  10. 10. Van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643-1647
  11. 11. Karbach J, Neumann A, Atmaca A, et al. Efficient in vivo priming by vaccination with recombinant NY-ESO-1 protein and CpG in antigen naive prostate cancer patients. Clinical Cancer Research. 2011;17(4):861-870
  12. 12. Parkhurst MR, Fitzgerald EB, Southwood S, Sette A, Rosenberg SA, Kawakami Y. Identification of a shared HLA-A*0201-restricted T-cell epitope from the melanoma antigen tyrosinase-related protein 2 (TRP2). Cancer Research. 1998;58(21):4895-4901
  13. 13. DeFilippis RA, Goodwin EC, Wu L, DiMaio D. Endogenous human papillomavirus E6 and E7 proteins differentially regulate proliferation, senescence, and apoptosis in HeLa cervical carcinoma cells. Journal of Virology. 2003;77(2):1551-1563
  14. 14. Kash N, Lee MA, Kollipara R, Downing C, Guidry J, Tyring SK. Safety and efficacy data on vaccines and immunization to human papillomavirus. Journal of Clinical Medicine. 2015;4(4):614-633
  15. 15. Roden R, Wu TC. How will HPV vaccines affect cervical cancer? Nature Reviews. Cancer. 2006;6:753-763
  16. 16. Chang MH, Chen CJ, Lai MS, Hsu HM, Wu TC, Kong MS, et al. Universal hepatitis B vaccination in Taiwan and the incidence of hepatocellular carcinoma in children. Taiwan childhood Hepatoma study group. The New England Journal of Medicine. 1997;336(26):1855-1859
  17. 17. Higano CS, Schellhammmer PF, Small EJ, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670-3679
  18. 18. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. The New England Journal of Medicine. 2010;363(5):411-422
  19. 19. Obeid J, Hu Y, Slingluff CL Jr. Vaccines, adjuvants, and dendritic cell activators-current status and future challenges. Seminars in Oncology. 2015;42(4):549-561
  20. 20. Mach N, Gillessen S, Wilson SB, Sheehan C, Mihm M, Dranoff G. Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte macrophage colony-stimulating factor or Flt3-ligand. Cancer Research. 2000;60(12):3239-3246
  21. 21. Manjili MH, Wang XY, MacDonald IJ, Arnouk H, Yang GY, Pritchard MT, et al. Cancer immunotherapy and heat-shock proteins: Promises and challenges. Expert Opinion on Biological Therapy;4(3):363-373
  22. 22. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62-68
  23. 23. Rohaan MW, Wilgenhof S, Haanen JBAG. Adoptive cellular therapies: The current landscape. Virchows Archiv. 2019;474(4):449-461
  24. 24. Verdegaal EM. Adoptive cell therapy: A highly successful individualized therapy for melanoma with great potential for other malignancies. Current Opinion in Immunology. 2016;39:90-95
  25. 25. Brudno JN, Somerville RP, Shi V, et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that Progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. Journal of Clinical Oncology. 2016;34(10):1112-1121
  26. 26. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. Journal of Clinical Oncology. 2015;33(6):540-549
  27. 27. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England Journal of Medicine. 2014;371(16):1507-1517
  28. 28. Pang Y, Hou X, Yang C, Liu Y, Jiang G. Advances on chimeric antigen receptor-modified T-cell therapy for oncotherapy. Molecular Cancer. 2018;17(1):91
  29. 29. Ljunggren HG, Karre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunology Today. 1990;11:237-244
  30. 30. Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood. 1999;94(1):333-339
  31. 31. Coulson A, Levy A, Gossell-Williams M. Monoclonal antibodies in cancer therapy: Mechanisms, successes and limitations. The West Indian Medical Journal. 2014;63(6):650-654
  32. 32. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nature Reviews. Cancer. 2012;12(4):237-251
  33. 33. Pinto AC, Ades F, de Azambuja E, Piccart-Gebhart M. Trastuzumab for patients with HER2 positive breast cancer: Delivery, duration and combination therapies. Breast. 2013;22(Suppl 2):S152-S155
  34. 34. Steiner M, Neri D. Antibody-radionuclide conjugates for cancer therapy: Historical considerations and new trends. Clinical Cancer Research. 2011;17(20):6406-6416
  35. 35. Flygare JA, Pillow TH, Aristoff P. Antibody-drug conjugates for the treatment of cancer. Chemical Biology and Drug Design. 2013;81(1):113-121
  36. 36. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell. 2015;161(2):205-214
  37. 37. Tsai HF, Hsu PN. Cancer immunotherapy by targeting immune checkpoints: Mechanism of T cell dysfunction in cancer immunity and new therapeutic targets. Journal of Biomedical Science. 2017;24(1):35
  38. 38. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. The New England Journal of Medicine. 2015;372(21):2006-2017
  39. 39. Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous nonsmall-cell lung cancer: A randomised, phase 2 cohort of the open-label KEYNOTE-021 study. The Lancet Oncology. 2016;17(11):1497-1508
  40. 40. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England Journal of Medicine. 2010;363(8):711-723
  41. 41. Corsello SM, Barnabei A, Marchetti P, Vecchis LD, Salvatori R, Torino F. Endocrine side effects induced by immune checkpoint inhibitors. The Journal of Clinical Endocrinology and Metabolism. 2013;98(4):1361-1375
  42. 42. Di Giacomo AM, Biagioli M, Maio M. The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. Seminars in Oncology. 2010;37:499-507

Written By

Hilal Arnouk and Sana Moqeet

Submitted: 26 July 2019 Published: 13 November 2019