Open access peer-reviewed chapter

Perspective Chapter: Role of Immunosuppressive and Immunomodulatory Agents in Cancer

Written By

Poppy Diah Palupi, Mohammed Safwan Ali Khan and Nur Dina Amalina

Submitted: 21 October 2022 Reviewed: 21 November 2022 Published: 11 December 2022

DOI: 10.5772/intechopen.109072

From the Edited Volume

Immunosuppression and Immunomodulation

Edited by Rajeev K. Tyagi, Prakriti Sharma and Praveen Sharma

Chapter metrics overview

80 Chapter Downloads

View Full Metrics

Abstract

Immunosuppressants offer some benefits and disadvantages. Like a blade with two edges, immunosuppressants are categorized as drugs but also cause decreased immunity, which eventually cause cancer. Immunosuppressants are widely used in organ transplantation patients and autoimmune illnesses to suppress the immune response and provide a significant risk of cancer. According to epidemiological and cancer research, malignancies are higher among transplant patients. However, the risk varies significantly between studies due to methods and patient selection variations. A more accurate illustration of the effects of mild-to-moderate immunosuppression concerning the risk of cancer can be seen in the rising use of immunosuppressant medications in non-transplant patients. Generally, cancer cells have an approach to avoid immune surveillance and create a complex balance in which many immune subtypes may be responsible for controlling tumor development, metastasis, and resistance. Therefore, the main objective of most cancer immunotherapies is to reestablish effective immune control. Immunomodulators help to maintain immune system function and promote the immune system’s capacity to fight and defeat cancer. One of them is immune checkpoint inhibitors.

Keywords

  • immune system
  • immunosuppressant
  • immunomodulator
  • immune checkpoint inhibitor
  • cancer

1. Introduction

Malignancies are reported to be linked with the immune suppression system. Consequently, approximately, about 8.2 million annual casualties are expected to increase [1]. The concept, innate and adaptive immune cells can regulate tumor growth. However, neoplasm tissue tumors are identifiable as malignant cells and evolve new defense mechanisms that imitate peripheral immunological tolerance to fight against tumoricidal strikes [2]. In addition, due to the cancerous cells having antigens that make them different from normal cells, the immune system can find cells that have become cancerous [3]. The immune system recognizes and eliminates abnormal cells as part of its normal function, most likely preventing or slowing cancer progression [4].

Present chapter discusses the significance of comprehending the immune system’s function in the emergence of cancer, including the often prescribed immunosuppressant medications for autoimmune and organ transplant patients. Also, this manuscript shows the importance of the immunomodulators, including immune checkpoint blockade, in cancer immunotherapy.

Advertisement

2. Cancer and immune suppression

The host immune system is well established to contribute to the evolution and progression of cancer, as significant as the tumor immune system. The complex interactions between the immune system and the tumor commonly occur in either the tumor’s immune deterrence or the termination of cancer [5]. Moreover, significant discoveries in the last few decades have demonstrated that the immune system plays an important role in maintaining the equivalence between immune recognition and cancer development. And it might both promote and inhibit tumor growth [6].

Immune cells that have entered the tumor microenvironment (TME) regulate the growth and dissemination of cancer (TME) [7]. The disruption of the TME induces an inflammatory immune response, as evidenced by the presence of innate and adaptive immune cells in histopathological examinations, and is classified as tumor growth [8]. Interactions between the morphological and molecular elements of the TME through a complex and multistep metastatic cascade enable cancer cells to spread from the initial site to distant regions and become invasive [9]. Immune evasion also frequently occurs due to interactions between the elements of the immune system and the tumor cells in the TME, which promotes the development of tumors [10].

Tumor formation is initiated when immune cells like CD8+ T cells and natural killer (NK) cells attack and destroy most cancer cells. However, specific tumor cells can avoid these immune defenses by suppressing effector cells or stimulating tolerogenic cells during the immunological escape period. External and internal mechanisms that reduce antitumor immune responses and increase immunosuppressive cells, like regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs), promote immune surveillance escape [11]. Regulatory T (Treg) cells expressing Foxp3+ reduce the dysfunctional immune response to self-antigens and the antitumor immune response. Additionally, Treg cell infiltration into tumor tissues is frequently associated with poor clinical outcomes [12]. Autoimmune diseases occur when Treg cells are insufficient due to immune suppression of Foxp3+, CD25+, and CD4+ Treg cells; these cells have been identified as one of the most important mechanisms of immunological self-tolerance [13]. In addition, a study has shown that Treg cell ablation can induce antitumor immunity effectively. However, it can also result in autoimmunity, particularly if Treg cells are eliminated systemically [14].

Treg cells have the ability to regulate T-cells, B-cells, NK cells, dendritic cells (DCs), and macrophages via humoral and cell–cell contact pathways. Several molecules, including TGF (transforming growth factor), GITR (glucocorticoid-induced TNF receptor), CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), LAG3 (lymphocyte-activation gene 3), IL-2, IL-10, IL-35, granzyme B, adenosine, and cAMP, are implicated in Treg-mediated suppression pathways [15]. Foxp3+ regulates the expression of these molecules, and deficiencies of IL-2, CD25, CD122, and CTLA-4 are associated with autoimmune disorders. According to these hypotheses, only a few molecules have their expressions directly or indirectly regulated by Foxp3+, such as IL-2, IL-2 receptor subunits, and CTLA-4. The absence of these molecules results in severe autoimmune disorders and the loss of Treg-suppressive function [16, 17, 18].

In addition, the suppression mechanisms dependent on Tregs are necessary for establishing self-tolerance and have a significant impact on tumor immunity. The expression of CD25 and CTLA-4, dependence on exogenous IL-2, and T-cell receptor (TCR) activation produce Treg functions, particularly suppression mediated by Tregs. Regarding tumor immunity, these molecular mechanisms are also effective targets for regulating the activity and expansion of Tregs [16]. Therefore, recent advances in cancer immunotherapy targeted Treg cells suggest that Treg depletion or functional modification may be facilitated by Treg-specific drugs. These molecules are CD25, GITR, OX-40, and LAG3 [12]. CTLA-4, programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1 (PD-L1) will be discussed further in this chapter.

Advertisement

3. Cancer and immunosuppressive agents

Since their discovery, many immunosuppressive medications have been used in transplantation and autoimmune diseases. Many organ transplant recipients, for instance, take medication to suppress their immune systems and reduce the graft rejection episodes. The immunosuppressive therapy after transplantation aims to prevent acute and chronic rejection while reducing adverse pharmacological effects in transplant recipients. The majority of therapies modify immune response mechanisms but lack immunological specificity [19].

Any immunosuppressive medication carries a potentially serious risk of cancer. Due to the increasing use of immunosuppressive medications among transplant and non-transplant patients, it is possible to define the effects of mild-to-moderate immunosuppression on the risk of neoplasms [20]. The use of immunosuppressive drugs in organ transplant patients is associated with a wide range of adverse side effects. The potential cancer risks—well-documented since the late 1960s—represent a significant cause of morbidity, mortality, and late failure in patients who otherwise have healthy grafts. Since transplantation first became popular, two agents have been utilized: azathioprine and corticosteroids [21]. Tacrolimus, cyclosporine, mycophenolate mofetil, everolimus, and sirolimus are the most common immunosuppressive drugs used in the combination therapy [19].

Cancers in transplant patients have been the subject of the current study, collecting information from single and multicenter studies. The type of malignancies and estimated risks differ significantly from study to study due to a variety of factors, including geographic differences, the use of various immunosuppressive regimens and antiviral therapy prevention, the duration of follow-up, the type of organ transplant and multiple techniques for estimating the occurrence [20]. The malignancy incidence in transplant recipients is higher in young adults, with significant clinical aggressiveness and a relatively short time in initiation after transplantation. Additionally, a key risk factor for immunosuppressive medication use is the dosing frequency and schedule [22].

A high prevalence of cancer among transplant recipients was seen/observed. Still, there is a debate over which factors—such as the type of immunosuppressive regimens, the overall level of immunosuppression, the course of treatment, or the dosage—is most important to assess the risks involved. The early agent used, azathioprine, can potentially cause cancer directly or indirectly. A study in premalignant dysplastic keratotic lesions showed that azathioprine might have a carcinogenic effect rather than simply suppressing the immune system [23]. Although cyclosporine unexpectedly showed the high rates of lymphomas and Kaposi’s sarcomas, there is no substantial evidence that this medication increases the risk of tumors compared to those seen with the other immunosuppressive drugs such as, traditional azathioprine-based regimens [24]. On the other hand, tacrolimus-induced post-transplant malignancies were shown to have a high prevalence and pathological characteristics comparable to other immunosuppressive drugs [25].

In addition to treating non-transplant patients, such as Inflammatory Bowel Disease (IBD), immunomodulators such as thiopurines or methotrexate and TNF- antagonists may also reduce the incidence of inflammation-related cancers. However, although there is little chance of developing cancer when taking azathioprine in non-transplant patients, there is a potential risk that may rise with time and in a dose-dependent manner [26]. Although cyclophosphamide is typically used for cancer patients or bone marrow transplantation regimens, its immunosuppressive effects have also been applied to many chronic inflammatory diseases. For example, in people treated for cancer or non-malignant illnesses, it was shown to have the ability to increase the risk of bladder cancer. However, long-term cyclophosphamide therapy in a non-neoplasm patient is linked to an increased frequency of some malignancies, suggesting an immunosuppressive agent’s potential side effect [27].

Immunomodulators and biological agents affect the immune system and might promote cancer development [28]. Thiopurines and methotrexate contribute to the development of the cancer by activating oncogenes, altering DNA directly, reducing physiologic immunosurveillance of malignant cells, and impairing the immune system’s capacity to regulate oncogenic viruses [27, 29]. Infliximab, a chimeric IgG antibody, is primarily directed against TNF-α to neutralize the cytotoxic effects in a dose-dependent manner and has recently been licensed to treat rheumatoid arthritis and Crohn’s disease [20]. However, TNF-α has many different impacts on the immune system, but the carcinogenic potential is less understood because of unreliable molecular information. TNF-α has been demonstrated to have antitumor activity by inducing the cellular death of malignant cells. Moreover, as a pro-tumor inflammatory cytokine, TNF-α is generated by most tumors to stimulate cellular survival and accelerate cancer growth [30, 31].

Another immunosuppressant agent that is used in the transplant population is Rapamycin. Streptomyces hygroscopicus is the source of the fermentation product rapamycin (RAPA), also referred to as sirolimus [32]. Many studies revealed that RAPA was a potent immunosuppressive drug to prevent allograft rejection in the heart, liver, lung, and kidney transplantation [33, 34, 35]. Sirolimus, and its analogs, including deforolimus, everolimus, and temsirolimus (a rapamycin prodrug), block the mechanistic target of Rapamycin (mTOR). Therefore, rapalogs are used in some clinical applications, such as organ transplant management and cancer therapy. The immunosuppressive properties of Rapalogs justified their use in organ recipients. Despite the fact that rapalogs were predicted to promote tumor growth and increase cancer incidence, they are frequently used in cancer treatment [36].

Commonly, growth factors, nutrient-rich environments, and oxygen levels excessively stimulate cultured cells. The environment stimulates growth-promoting pathways, including the PI3K/mTOR axis and mitogen-activated protein kinases (MAPKs). The activation of the oncosuppressor p53 and the accumulation of cell cycle inhibitors, such as cyclin-dependent kinase inhibitor 1A (CDKN1A) and cyclin-dependent kinase inhibitor 2A, can induce a cell cycle arrest under certain stress conditions (CDKN2A). In the case of malignant cells, growth factors and oncogenic signaling pathways continue to activate cultured cells by promoting mTOR and MAPK signaling even though the cell cycle has finished [37, 38]. Therefore, rapalogs are increasingly recommended for cancer treatment, especially for mTOR-dependent cancer subtypes [39, 40]. Thus, rapalogs could be thought of as anti-inflammatory substances that demonstrated anticancer. In addition, Rapamycin and its analogs also lowered the risk of cancer related to organ transplants and extended the overall and disease-free longevity of patients with certain malignancies.

Advertisement

4. Immune checkpoint inhibitors (ICI) as immunomodulatory agents

In adaptive immunity, two immune cells, B and T. B-cells, recognize circulating antigens in their natural state and produce protective antibodies in response [41]. T-cells are a powerful weapon the immune system uses to fight cancer [42]. T-cells identify peptide antigens from intracellularly degraded proteins filled onto the Cell’s surface of major histocompatibility complex (MHC) molecules, and this process is known as antigen presentation [43]. Immunological checkpoints on the cell surface are activated when their surface proteins recognize and bind to partner proteins on other cells, such as specific tumor cells [42]. Self-tolerance requires immune checkpoints to prevent autoimmunity and protect tissues from destruction [44]. Tregs are drawn to cancer cells, which causes them to express less tumor antigen and release immune-suppressive cytokines that activate inhibitory immunological checkpoints [10] that create an immunosuppressive TME [45]. Immune checkpoint inhibitors work by obstructing particular inhibitory pathways’ actions to combat immunosuppressive conditions [44]. The “brake system” of the immune system that cancers routinely exploit to halt immunological responses and defend themselves are immune checkpoints. Checkpoint inhibitors can generate new immune responses against cancer and strengthen already-existing ones to remove malignant cells.

The CTLA-4, PD-1, and PD-L1 are the common inhibitory checkpoints [46]. These antibodies have biological effects on different body parts during the T cell’s lifecycle [47]. In addition, they functionally complement one another, establishing that T cell responses retain self-tolerance while defending the body from infections and cancer [43]. The immune checkpoint inhibitors approved by the FDA are shown in Table 1.

TargetDrugsMechanism of actionApproval yearTreatment
CTLA-4IpilimumabInhibited CTLA-4, T-cell activation2010CRC, HCC, melanoma, mesothelioma, NSCLC, RCC
PD-1NivolumabInhibited PD-1, T-cell activation2014CRC, esophageal SCC, HCC, HL, HNSCC, melanoma, mesothelioma, NSCLC, RCC, UC
PembrolizumabInhibited PD-1, T-cell activation2014BC, CVC, CRC, CSCC, EnC, EsC, GC, HCC, HL, HNSCC, melanoma, mesothelioma, MCC, NSCLC, LBCL, RCC, SCLC, UC
CemiplimabInhibited PD-1, T-cell activation2018BCC, CSCC, NSCLC
DostarlimabInhibited PD-1, T-cell activation2021dMMR, EnC
PD-L1AtezolizumabInhibited PD-L1, T-cell activation2016BC, HCC, melanoma, NSCLC, SCLC, UC
AvelumabInhibited PD-L1, T-cell activation2017MCC, RCC, UC
DurvalumabInhibited PD-L1, T-cell activation2018NSCLC, SCLC, UC

Table 1.

Immune checkpoint inhibitors approved by FDA.

CRC, colorectal cancer; HCC, hepatocellular carcinoma; NSCLC, non-small cell lung cancer; RCC, renal cell carcinoma; BCC, basal cell carcinoma; CSCC, cutaneous squamous cell carcinoma; EnC, endometrial carcinoma; EsC, esophageal carcinoma; GC, gastric carcinoma; SCC, squamous cell carcinoma; HL, Hodgkin lymphoma; HNSCC, head, and neck squamous cell carcinoma; UC, urothelial carcinoma BC; Breast cancer; CVC, cervical cancer; MCC, Merkel cell carcinoma; LBCL, large B cell lymphoma; SCLC, small cell lung cancer; dMMR, mismatch repair deficient.

4.1 CTLA-4

In humans, CTLA4 is the first immune-checkpoint receptor to be studied; its located on T cells and controls T cell activation in the early stages of infection [48]. CTLA-4 is a novel immunoglobulin superfamily member resembling CD28, structurally and pharmacologically [49]. CTLA-4 and CD28 are expressed exclusively in the hematopoietic compartment and found in the exact location of chromosome 2 (2q33.2). Furthermore, CTLA-4 and CD28 have the most sequence similarity in their extracellular binding domain; they bind to the identical CD80 and CD86 ligands expressed by antigen-presenting cells (APCs) [50]. Further characterization revealed that CD28 and CTLA4 have opposing immunoregulatory functions. CTLA4 inhibits T cell activation in a number of ways, including by directly opposing CD28, competing for co-stimulatory ligands, preventing the production of immunological conjugates, and recruiting inhibitory effectors [51]. Moreover, CTLA4 promotes the internalization of its ligands, which prevents them from binding to CD28 and reduces IL-2 production and T-cell proliferation [52].

4.2 Clinical application of CTLA-4 inhibitor

In 2010, the FDA authorized ipilimumab for advanced melanoma treatment, making it the first medicine with a survival advantage for metastatic melanoma. Long-term studies have shown that antitumor immunity persists following CTLA4 inhibition, confirming the stability of this survival effect [53]. Unfortunately, findings from trials in renal cell carcinoma [54], non-small-cell lung cancer [55], small-cell lung cancer [56], and prostate cancer [57] were less effective than those reported in melanoma patients. The FDA has not yet approved tremelimumab, an IgG2 isotype CTLA4-blocking antibody, because it did not extend survival in patients with advanced melanoma. It is believed that the effectiveness of ipilimumab and tremelimumab differs due to differences in binding kinetics and the ability to mediate cytotoxicity [58].

4.3 PD-1/PD-L1

PD1 was initially considered to be a potential modulator of apoptosis. However, later data revealed a role in regulating hyperactivation of the immune system, like CTLA4 [59]. CTLA4 and CD28 have 20% and 15% amino acid identity, respectively [60]. CTLA4 limits T-cell activation in peripheral tissues, whereas PD1 regulates T-cell activation predominantly within lymphoid organs. Relatively, the PD1 axis performs a particular role in self-tolerance in T cells [47].

4.4 Clinical application of PD-1/PD-L1 inhibitor

Pembrolizumab and nivolumab (both IgG4), humanized and completely human anti-PD1 monoclonal antibodies (mAbs), were the first FDA-approved PD1-targeted therapies for refractory and unresectable melanoma in 2014 [61]. Pembrolizumab exceeded ipilimumab in six-month progression-free survival and overall survival [62]. Pembrolizumab was approved in 2015 for treating non-small-cell lung cancer because it increased progression-free survival by 4.3 months compared to platinum-based chemotherapeutics and was more productive than paclitaxel [63]. However, the different organs have distinct immunosuppressive microenvironments, so it’s difficult to anticipate which patients may benefit. Nivolumab has since been approved for treating renal cell carcinoma, head and neck squamous cell carcinoma, urothelial carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, and colorectal cancer, similar to pembrolizumab [64].

PDL1 also targeted several antibodies that have benefits for cancer treatment. Atezolizumab (an IgG4 antibody), the first PDL1-targeted humanized mAbs, was licensed to treat urothelial cancer in 2016 [65]. However, more trial results have not shown that atezolizumab has clinical efficacy in urothelial carcinoma over the standard of treatment, even though it is less toxic than conventional chemotherapy [66]. In 2017, avelumab and durvalumab, two new anti-PDL1 human mAbs, were introduced to the market [67]. As a result, similar to PD1, blocking PDL1 is successful in difficult-to-treat cancers.

4.5 Clinical challenges during the blockade of immune checkpoint

Potent immune effector mechanisms are activated by blocking a naturally occurring immunological checkpoint [68]. According to a meta-analysis study, immune-related adverse events are expected to occur in 15–90% of patients. However, patients treated with CTLA4 and PD1 inhibitors have more severe episodes that require intervention in 15–30% of cases [69]. In addition, patients receiving anti-CTLA4 medication are at a higher risk of hypothyroidism, hepatotoxicity, and pneumonitis. In contrast, the patient that receives PD1-targeted drugs is more likely to develop hypothyroidism, hepatotoxicity, and pneumonitis [70].

Adverse events are particularly problematic in adjuvant chemotherapy because late-onset, frequently severe toxicities can impact tumor-survivor patients even after surgery only [71]. However, the toxicity of immune checkpoint inhibitors is more tolerable than that of conventional chemotherapeutics [72].

Advertisement

5. Conclusion and future perspectives

A decade after immune checkpoint protein discovery, such as PD-1/PDL-1 and CTLA-4, still offers hope for the cure for cancer patients. Although not all patients may benefit from these medications, some of the drugs demonstrated dose-dependent adverse events of mild to moderate. Unfortunately, not every cancer type responds well to the treatment, and the only options are to discontinue therapy or switch to conventional cancer treatments.

Even though several studies showing the side effects of ICI do not discourage researchers from developing the new finding, these drugs will probably be examined in adjuvant or neoadjuvant approaches, based on clinical responses in various cancer types, to increase the overall survival of many cancer patients. In addition, understanding the systemic effect mechanism caused by immunotherapy may help gain better knowledge for an effective antitumor response. Finally, combination therapy with various immunotherapy, chemotherapy, targeted medicines, radiation, and T-cell-based therapies can potentially improve the outcomes, especially in patients who have not responded well to immunotherapy-based treatments.

References

  1. 1. Ferlay J et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer. 2015;136(5):E359-E386. DOI: 10.1002/IJC.29210
  2. 2. Gonzalez H, Hagerling C, Werb Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes & Development. 2018;32(19-20):1267. DOI: 10.1101/GAD.314617.118
  3. 3. Ponomarev AV, Shubina IZ. Insights into mechanisms of tumor and immune system interaction: Association with wound healing. Frontiers in Oncology. 2019;9(OCT):1115. DOI: 10.3389/FONC.2019.01115/XML/NLM
  4. 4. NIH. Immunotherapy for Cancer. National Cancer Institute; 2019
  5. 5. Becker JC, Andersen MH, Schrama D, Thor Straten P. Immune-suppressive properties of the tumor microenvironment. Cancer Immunology, Immunotherapy. 2013;62(7):1137-1148. DOI: 10.1007/S00262-013-1434-6
  6. 6. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: From immunosurveillance to tumor escape. Nature Immunology. 2002;3(11):991-998. DOI: 10.1038/NI1102-991
  7. 7. Guerrouahen BS, Maccalli C, Cugno C, Rutella S, Akporiaye ET. Reverting immune suppression to enhance cancer immunotherapy. Frontiers in Oncology. 2020;9:1554. DOI: 10.3389/FONC.2019.01554/BIBTEX
  8. 8. Fridman WH, Pagès F, Sauts-Fridman C, Galon J. The immune contexture in human tumours: Impact on clinical outcome. Nature Reviews. Cancer. 2012;12(4):298-306. DOI: 10.1038/NRC3245
  9. 9. Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers. 2021;13(9):2053. DOI: 10.3390/CANCERS13092053
  10. 10. Vinay DS et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Seminars in Cancer Biology. 2015;35(Suppl):S185-S198. DOI: 10.1016/J.SEMCANCER.2015.03.004
  11. 11. Shimizu K, Iyoda T, Okada M, Yamasaki S, Fujii SI. Immune suppression and reversal of the suppressive tumor microenvironment. International Immunology. 2018;30(10):445-455. DOI: 10.1093/INTIMM/DXY042
  12. 12. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Research. 2017;27(1):109-118. DOI: 10.1038/CR.2016.151
  13. 13. Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annual Review of Immunology. 2004;22:531-562. DOI: 10.1146/ANNUREV.IMMUNOL.21.120601.141122
  14. 14. Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: A common basis between tumor immunity and autoimmunity. Journal of Immunology. 1999;163(10):5211-5218
  15. 15. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775-787. DOI: 10.1016/J.CELL.2008.05.009
  16. 16. Yamaguchi T et al. Construction of self-recognizing regulatory T cells from conventional T cells by controlling CTLA-4 and IL-2 expression. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(23):E2116-E2125. DOI: 10.1073/PNAS.1307185110
  17. 17. Negi S, Saini S, Tandel N, Sahu K, Mishra RPN, Tyagi RK. Translating treg therapy for inflammatory bowel disease in humanized mice. Cell. 2021;10(8):1-28. DOI: 10.3390/cells10081847
  18. 18. Tyagi RK et al. HLA-restriction of human Treg cells is not required for therapeutic efficacy of low-dose IL-2 in humanized mice. Frontiers in Immunology. 2021;12(February):1-8. DOI: 10.3389/fimmu.2021.630204
  19. 19. Tönshoff B. Immunosuppressants in organ transplantation. Handbook of Experimental Pharmacology. 2020;261:441-469. DOI: 10.1007/164_2019_331
  20. 20. Vial T, Descotes J. Immunosuppressive drugs and cancer. Toxicology. 2003;185(3):229-240. DOI: 10.1016/S0300-483X(02)00612-1
  21. 21. Taylor AL, Watson CJE, Bradley JA. Immunosuppressive agents in solid organ transplantation: Mechanisms of action and therapeutic efficacy. Critical Reviews in Oncology/Hematology. 2005;56(1):23-46. DOI: 10.1016/J.CRITREVONC.2005.03.012
  22. 22. London NJ, Farmery SM, Lodge JPA, Will EJ, Davidson AM. Risk of neoplasia in renal transplant patients. Lancet. 1995;346(8972):403-406. DOI: 10.1016/S0140-6736(95)92780-8
  23. 23. Weaver JL. Establishing the carcinogenic risk of immunomodulatory drugs. Toxicologic Pathology. 2012;40(2):267-271. DOI: 10.1177/0192623311427711
  24. 24. Durnian JM, Stewart RMK, Tatham R, Batterbury M, Kaye SB. Cyclosporin-a associated malignancy. Clinical Ophthalmology. 2007;1(4):421
  25. 25. Carenco C et al. Tacrolimus and the risk of solid cancers after liver transplant: A dose effect relationship. American Journal of Transplantation. 2015;15(3):678-686. DOI: 10.1111/AJT.13018
  26. 26. Sprangers B, Nair V, Launay-Vacher V, Riella LV, Jhaveri KD. Risk factors associated with post–kidney transplant malignancies: An article from the cancer-kidney international network. Clinical Kidney Journal. 2018;11(3):315-329. DOI: 10.1093/CKJ/SFX122
  27. 27. Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nature Reviews. Immunology. 2006;6(10):715-727. DOI: 10.1038/NRI1936
  28. 28. Axelrad JE, Lichtiger S, Yajnik V. Inflammatory bowel disease and cancer: The role of inflammation, immunosuppression, and cancer treatment. World Journal of Gastroenterology. 2016;22(20):4794. DOI: 10.3748/WJG.V22.I20.4794
  29. 29. Münz C, Moormann A. Immune escape by Epstein-Barr virus associated malignancies. Seminars in Cancer Biology. 2008;18(6):381-387. DOI: 10.1016/J.SEMCANCER.2008.10.002
  30. 30. Danial NN, Korsmeyer SJ. Cell death: Critical control points. Cell. 2004;116(2):205-219. DOI: 10.1016/S0092-8674(04)00046-7
  31. 31. Balkwill F. Tumour necrosis factor and cancer. Nature Reviews. Cancer. 2009;9(5):361-371. DOI: 10.1038/NRC2628
  32. 32. Abraham RT, Wiederrecht GJ. Immunopharmacology of rapamycin. Annual Review of Immunology. 2003;14:483-510. DOI: 10.1146/ANNUREV.IMMUNOL.14.1.483
  33. 33. Keogh A et al. Sirolimus in De novo heart transplant recipients reduces acute rejection and prevents coronary artery disease at 2 years. Circulation. 2004;110(17):2694-2700. DOI: 10.1161/01.CIR.0000136812.90177.94
  34. 34. Trotter JF et al. Liver transplantation using sirolimus and minimal corticosteroids (3-day taper). Liver Transplant. 2001;7(4):343-351. DOI: 10.1053/JLTS.2001.23012
  35. 35. Shitrit D et al. Use of sirolimus and low-dose calcineurin inhibitor in lung transplant recipients with renal impairment: Results of a controlled pilot study. Kidney International. 2005;67(4):1471-1475. DOI: 10.1111/J.1523-1755.2005.00224.X
  36. 36. Waldner M, Fantus D, Solari M, Thomson AW. New perspectives on mTOR inhibitors (rapamycin, rapalogs and TORKinibs) in transplantation. British Journal of Clinical Pharmacology. 2016;82(5):1158. DOI: 10.1111/BCP.12893
  37. 37. Arcaro A, Guerreiro AS. The phosphoinositide 3-kinase pathway in human cancer: Genetic alterations and therapeutic implications. Current Genomics. 2007;8(5):271. DOI: 10.2174/138920207782446160
  38. 38. Blagosklonny MV. Hypoxia, MTOR and autophagy: Converging on senescence or quiescence. Autophagy. 2013;9(2):260-262. DOI: 10.4161/AUTO.22783
  39. 39. Blagosklonny MV. Rapalogs in cancer prevention: Anti-aging or anticancer? Cancer Biology & Therapy. 2012;13(14):1349. DOI: 10.4161/CBT.22859
  40. 40. Grabiner BC et al. A diverse array of cancer-associated mTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discovery. 2014;4(5):554. DOI: 10.1158/2159-8290.CD-13-0929
  41. 41. Lebien TW, Tedder TF. B lymphocytes: How they develop and function. Blood. 2008;112(5):1570-1580. DOI: 10.1182/BLOOD-2008-02-078071
  42. 42. He X, Xu C. Immune checkpoint signaling and cancer immunotherapy. Cell Research. 2020;30(8):660-669. DOI: 10.1038/s41422-020-0343-4
  43. 43. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nature Reviews. Immunology. 2020;20(11):651-668. DOI: 10.1038/s41577-020-0306-5
  44. 44. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews. Cancer. 2012;12(4):264. DOI: 10.1038/NRC3239
  45. 45. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27(45):5904-5912. DOI: 10.1038/ONC.2008.271
  46. 46. Marin-Acevedo JA, Kimbrough EMO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. Journal of Hematology & Oncology. 2021;14(1):45. DOI: 10.1186/S13045-021-01056-8
  47. 47. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunological Reviews. 2008;224(1):166-182. DOI: 10.1111/J.1600-065X.2008.00662.X
  48. 48. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunological Reviews. 2009;229(1):12-26. DOI: 10.1111/J.1600-065X.2009.00770.X
  49. 49. Dariavach P, Mattéi M-G, Golstein P, Lefranc M-P. Human Ig superfamily CTLA-4 gene: Chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains. European Journal of Immunology. 1988;18(12):1901-1905. DOI: 10.1002/EJI.1830181206
  50. 50. Pentcheva-Hoang T, Egen JG, Wojnoonski K, Allison JP. B7-1 and B7-2 selectively recruit CTLA-4 and CD28 to the immunological synapse. Immunity. 2004;21(3):401-413. DOI: 10.1016/J.IMMUNI.2004.06.017
  51. 51. Intlekofer AM, Thompson CB. At the bench: Preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. Journal of Leukocyte Biology. 2013;94(1):25-39. DOI: 10.1189/JLB.1212621
  52. 52. Qureshi OS et al. Trans-endocytosis of CD80 and CD86: A molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332(6029):600-603. DOI: 10.1126/SCIENCE.1202947
  53. 53. Maio M et al. Five-year survival rates for treatment-naive patients with advanced melanoma who received ipilimumab plus dacarbazine in a phase III trial. Journal of Clinical Oncology. 2015;33(10):1191-1196. DOI: 10.1200/JCO.2014.56.6018
  54. 54. Yang JC et al. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. Journal of Immunotherapy. 2007;30(8):825-830. DOI: 10.1097/CJI.0B013E318156E47E
  55. 55. Lynch TJ et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: Results from a randomized, double-blind, multicenter phase II study. Journal of Clinical Oncology. 2012;30(17):2046-2054. DOI: 10.1200/JCO.2011.38.4032
  56. 56. Reck M et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: Results from a randomized, double-blind, multicenter phase 2 trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013;24(1):75-83. DOI: 10.1093/ANNONC/MDS213
  57. 57. Kwon ED et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. The Lancet Oncology. 2014;15(7):700-712. DOI: 10.1016/S1470-2045(14)70189-5
  58. 58. He M et al. Remarkably similar CTLA-4 binding properties of therapeutic ipilimumab and tremelimumab antibodies. Oncotarget. 2017;8(40):67129-67139. DOI: 10.18632/ONCOTARGET.18004
  59. 59. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. The EMBO Journal. 1992;11(11):3887-3895. DOI: 10.1002/J.1460-2075.1992.TB05481.X
  60. 60. Carreno BM, Collins M. The B7 family of ligands and its receptors: New pathways for costimulation and inhibition of immune responses. Annual Review of Immunology. 2002;20:29-53. DOI: 10.1146/ANNUREV.IMMUNOL.20.091101.091806
  61. 61. Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: A comprehensive review of registration trials and future considerations. Journal for Immunotherapy of Cancer. 2018;6(1):8. DOI: 10.1186/S40425-018-0316-Z
  62. 62. Robert C et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. The New England Journal of Medicine. 2015;372(26):2521-2532. DOI: 10.1056/NEJMOA1503093
  63. 63. Herbst RS et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet (London, England). 2016;387(10027):1540-1550. DOI: 10.1016/S0140-6736(15)01281-7
  64. 64. Overman MJ et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. The Lancet Oncology. 2017;18(9):1182-1191. DOI: 10.1016/S1470-2045(17)30422-9
  65. 65. Rosenberg JE et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet. 2016;387(10031):1909-1920. DOI: 10.1016/S0140-6736(16)00561-4
  66. 66. Powles T et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748-757. DOI: 10.1016/S0140-6736(17)33297-X
  67. 67. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. International Immunopharmacology. 2018;62:29-39. DOI: 10.1016/J.INTIMP.2018.06.001
  68. 68. Fritz JM, Lenardo MJ. Development of immune checkpoint therapy for cancer. The Journal of Experimental Medicine. 2019;216(6):1244-1254. DOI: 10.1084/JEM.20182395
  69. 69. Michot JM et al. Immune-related adverse events with immune checkpoint blockade: A comprehensive review. European Journal of Cancer. 2016;54:139-148. DOI: 10.1016/J.EJCA.2015.11.016
  70. 70. Kumar V, Chaudhary N, Garg M, Floudas CS, Soni P, Chandra AB. Current diagnosis and Management of Immune Related Adverse Events (irAEs) induced by immune checkpoint inhibitor therapy. Frontiers in Pharmacology. 2017;8(FEB):49. DOI: 10.3389/FPHAR.2017.00049
  71. 71. de Miguel M, Calvo E. Clinical challenges of immune checkpoint inhibitors. Cancer Cell. 2020;38(3):326-333. DOI: 10.1016/J.CCELL.2020.07.004
  72. 72. Nishijima TF, Shachar SS, Nyrop KA, Muss HB. Safety and tolerability of PD-1/PD-L1 inhibitors compared with chemotherapy in patients with advanced cancer: A meta-analysis. The Oncologist. 2017;22(4):470-479. DOI: 10.1634/THEONCOLOGIST.2016-0419

Written By

Poppy Diah Palupi, Mohammed Safwan Ali Khan and Nur Dina Amalina

Submitted: 21 October 2022 Reviewed: 21 November 2022 Published: 11 December 2022