Aslı Babayakalı1, Oytun Erbaş2

1Institute of Experimental Medicine, Gebze-Kocaeli, Turkey
2Department of Physiology, Medical Faculty of Demiroğlu Bilim University, Istanbul, Turkey

Keywords: Cancer immunotherapy, immune checkpoint, monoclonal antibody, PD-1 inhibition, PD-L1 inhibition

Abstract

In response to a foreign organism or material, our bodies react with a variety of immunological agents. This response mechanism is called immunogenicity. While some of the proteins that regulate the immune response function to activate this response, some of them are responsible for inhibiting this response. The main "brake" proteins involved in this negative regulation are cytotoxic T-lymphocyte-associated protein-4, programmed cell death protein-1 (PD-1), T-cell immunoglobulin and mucin domain-containing protein-3, and lymphocyte-activation gene-3. The cell can differentiate endogenous and exogenous substances through these brakes and other signal pathways. In the presence of brake proteins, the tumor cells are not perceived as a threat by the immune system, so a corresponding response does not occur. To generate this response, drugs containing monoclonal antibodies are produced for use in cancer treatments. Monoclonal antibodies are designed to block braking processes while also eliciting an immunological response. In this review, the PD-1 and programmed cell death-ligand 1 pathway and cancer immunotherapy are mentioned.

IMMUNE CHECKPOINTS

The proliferating tumor cells encounter various cell types involved in the immune system, and as a result of this encounter, the immune system creates a response. Receptors that activate and inhibit T cells regulate the balance between immune response and immune tolerance. T lymphocytes are divided into CD4+ (helper) and CD8+ (cytotoxic) T lymphocytes. For the immature CD8+ T (naive) cell to become active, a peptide presented by the T-cell receptor complex (TCR) and another by the major histocompatibility complex (MHC) must be combined, followed by secondary stimulation.[1] Self and foreign antigens in the body are presented to T cells by antigen-presenting cells (APCs). The antigen presented to T cells by the MHC is recognized by the TCR and the first signal is formed. Afterward, a secondary signal is required for T cells to complete the activation process. The most important secondary stimulus of CD8+ T cell precursors is the interaction of CD28 on the T lymphocyte surface with CD80/CD86 (B7 family molecules) on the APC.[1,2] Thus, the T cell passes from the naive form to the active form. CD80/CD86 is also the target site for cytotoxic T-lymphocyte-associated protein-4 (CTLA-4). If CTLA-4 binds with CD80/CD86, it cannot complete T cell activation, and subsequently, immune response is inhibited. In order to be active for the other immune checkpoint, programmed cell death protein-1 (PD-1) must combine with its ligands programmed cell deathligand 1 (PD-L1) and programmed cell deathligand-2 (PD-L2).[1,3-5] While PD-1 binding occurs only in tumor cells, CTLA-4 binding also occurs between T cells and other normal cells in the body. Unlike CTLA-4, the direct interaction of PD-1 with cancer cells is associated with the potency of PD-1 inhibitors to be more potent and less toxic. In addition, the different immune control points act through the inhibition of two or more control points in combination as a new strategy in the treatment of various diseases, especially cancer.[6] Allison[7] found that the CTLA-4 molecule acts as a brake on T cells in the 1990s and designed an antibody to eliminate this brake. In this study, tumor transplanted mice were treated with monoclonal antibodies against CTLA-4. This treatment designed for the immune system has been found to be curative in mice with tumors. Consequently, a number of studies were conducted, including studies on prostate cancer, breast cancer, skin cancer (malignant melanoma) samples, and animal tumor models.[8-10] Treatment with ipilimumab was successful in a clinical study conducted on malignant melanoma patients in 2010. Thus, ipilimumab, an anti-CTLA-4, immunoglobulin (Ig) G1 monoclonal antibody, was approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2011.[11] Ishida et al.[12] defined the PD-1 molecule and its functions in 1992 and showed that PD-1 also acts as a brake like CTLA-4 but works through different mechanisms. It has been shown in clinical studies that drugs providing PD-1 inhibition have positive results in the treatment of cancer, thus providing long-term treatment options in cancer patients with metastases, who previously could not be treated. Therefore, nivolumab, an antibody-based treatment used today, has been developed.[13] In 2018, Allison and Honjo[14] were awarded the Nobel Prize in Physiology or Medicine due to their work in the field of “discoveries in cancer treatment through suppression of the negative regulation of immunity.”[6]

DEFINING CTLA-4 AS A NEGATIVE REGULATOR

Cytotoxic T-lymphocyte-associated protein-4 is a molecule that binds to CD80/CD86 with a higher affinity than CD28.[15] It is known that CD28-associated CTLA-4 protein is found inside non-activated T cells but is rapidly transferred to the membrane after activation.[16]

In a study on CTLA-4 gene inactivation in mice, it was confirmed that CTLA-4 is a negative regulator, since these mice develop very severe autoimmune disease.[17] In 1999, an anti-CTLA-4 IgG1 monoclonal antibody named MDX-010 was developed and later named ipilimumab.[18,19]

Defining LAG-3 as a negative regulator

Lymphocyte-activation gene-3 (LAG-3 or CD223) is a cell surface molecule expressed on the surface of activated T cells, natural killer (NK) cells, B cells, and plasmacytoid dendritic cells.[20-24] The interaction between LAG-3 and its major ligand, MHC class II molecule, plays an important role in regulating dendritic cell function.[24,25] In addition, LAG-3 has been shown to have various roles on CD8+ T cell exhaustion.[26] Inhibition of the interaction between LAG-3 and MHC class II with the LAG-3 Ig fusion protein continues to be applied as a therapy in many cancer patients.

Defining TIM-3 as negative regulator

T-cell immunoglobulin and mucin domain-containing protein (TIM)-3 is a member of the TIM gene family, which includes TIM-1, TIM-3, and TIM-4 in humans, and it is expressed particularly on the surface of cells belonging to the myeloid family of T helper (Th) 1, Th17, and CD8+ T cells and in mice.[27-29] T-cell immunoglobulin and mucin domain-containing protein-3 and its ligands have been found to suppress Th1 and Th17 responses.[30] It has four different ligands shown to interact with the immunoglobulin variable (IgV) domain.[31] These are galectin-9 (Gal-9), high mobility group protein B1 (HMGB1), carcinoembryonic antigen cell adhesion molecule-1 (Ceacam-1), and phosphatidylserine (PtdSer).[32] Antibodies inhibiting TIM-3 have been shown to exacerbate experimental autoimmune encephalomyelitis (EAE) in a mouse model for human multiple sclerosis (MS).[29] This shows that TIM-3 has a negative regulatory role.

Discovery of the PD-1 receptor

Programmed cell death protein-1 is a 55 kDa transmembrane protein containing an immunoreceptor tyrosine-based inhibition motif (ITIM), first described by Ishida et al.[12] in 1992.[33] Ishida et al.[12] identified two ligands of PD-1 to characterize the function of PD-1. In a later study, they analyzed signal transduction and examined the pathophysiology of autoimmune diseases in PD-1 knockout mice.[33]

The most prominent feature of autoimmune diseases observed in PD-1 knockout mice is the variation in symptoms between strains. Mice in the C57BL strain developed lupus-like arthritis and glomerulonephritis.[2,33] In contrast, mice in the BALB strain produced autoantibodies against cardiac troponin I and developed dilated cardiomyopathy.[2,33-35] In addition, mice in the non-obese diabetic (NOD) strain developed type 1 diabetes as a result of extensive destruction of the islets of Langerhans.[36] From these observations, it was revealed that PD-1 provides negative costimulation against lymphocytes critical for establishing or maintaining self-tolerance.

The occurrence of autoimmune diseases in PD-1 knockout mice suggests that blocking the PD-1/PD-L1 pathway will facilitate the recognition of cancer cells as foreign. One study showed that tumor cells in mice express PD-L1 to escape the immune system by inhibiting T cell activity.[36] Similarly, it has been observed that cancer patients who express high levels of PD-L1 in cancer cells have a worse prognosis.[37]

PD-1 LIGANDS

Programmed cell death-ligand 1 and PD-L2 are type 1 transmembrane proteins with regions similar to IgV and immunoglobulin constant (IgC) in their extracellular regions.[2] Programmed cell death-ligand 1 and PD-L2 were discovered in 2000 and 2001 as ligands of PD-1.[4,5] Programmed cell death-ligand 1 is expressed in both lymphoid and non-lymphoid tissues. In particular, it shows up-regulation in the activation of APCs such as dendritic cells, macrophages, and B cells.[2,38,39] Programmed cell death protein-1 is also expressed in activated T cells. It is upregulated by interferon (IFN)-γ and other inflammatory cytokines secreted by activated T cells.[36,40,41] In addition, PD-L1 is expressed in a variety of tumor cells and virusinfected cells. Programmed cell death-ligand 1 expression in target cells enables PD-1 to directly inhibit the T cell response against the target cell.[42] Unlike PD-L1, PD-L2 is expressed only in APCs such as dendritic cells and macrophages.[39]

PD-1 INHIBITION MECHANISM

Programmed cell death protein-1 is mainly expressed on activated CD4+ and CD8+ T cells and B cells.[2,12,43] Inhibition occurs by binding with the PD-1 ligand PD-L1. The expression of PD-1 induced by activation regulates the late-phase immune response (memory response, chronic infection, etc.) in peripheral tissues while regulating early stimulation in lymphoid organs. The extracellular domain of PD-1 consists of a single IgV region-like fragment, 20-amino acid stem, transmembrane fragment, and cytoplasmic tail, and the cytoplasmic tail consists of an ITIM and an immunoreceptor tyrosine-based switch motif (ITSM).[2] Phosphorylated tyrosines on ITIM and ITSM bind to Src homology protein (SHP), a phosphatase. The main purpose of SHPs is to suppress T cell receptor signals by blocking intermediate molecules during T cell activation.[1] While ITIM binds to SHP-2 only, ITSM binds to both SHP-1 and SHP-2. PD-1/ PD-L1 pathway inhibition loses its function in the presence of ITSM mutation.[30,31,44,45] When PD-1 binds to its ligands, PD-L1 and PD-L2, it suppresses T cell activation by recombining with SHP-2, which dephosphorylates and inactivates ZAP-70, an important integrator of TCR-mediated signaling.[45-47] As a result, PD-1 prevents responses such as T cell proliferation, IFN production, and cytotoxic activity. The main target of the PD-1 pathway is the PI3K/Akt/mTOR pathway, which is involved in cell survival, development, and proliferation.[1]

Under normal conditions, protein tyrosine phosphatase and tensin homolog (PTEN) is inactivated when phosphorylated, while dephosphorylated it is activated and inhibits the PI3K/Akt/mTOR pathway.[1] Another important pathway targeted by PD-1 is the Ras/MEK/ERK pathway. It inhibits this pathway, which affects cell proliferation, cell division, and differentiation, and enables the release of interleukin (IL)-2, an inflammatory cytokine.[48]

Two transcription factor binding sites in the promoter region of the programmed cell death 1 (PDCD1) gene are important in regulating PD-1 expression. T-cell receptor complex-mediated calcium influx in undeveloped T cells initiates transcription of PDCD1 by activation of NFATc1, which binds to the 5' promoter region of the PDCD1 gene.[49] On the other hand, IFN-α and interferon regulatory factor 9 (IRF9) in chronically activated T cells cause long-term transcription of PDCD1 by binding to the PDCD1 promoter.[50] In addition, the PDCD1 promoter region becomes demethylated during chronic infection, causing high PD-1 expression in fatigued CD8+ T cells.[51] Fatigued CD8s+ T cells express high amounts of Eomesodermin (EOMES), a protein regulated by the FoxO1 transcription factor. The FoxO1 also binds to the PDCD1 promoter and enhances PD-1 expression.[52,53]

TREATMENT METHODS USED IN CANCER IMMUNOTHERAPY

Two different approaches have been developed to activate the immune system against cancer. The first is antigen-specific immunotherapy or therapeutic cancer vaccines, and the second is non-antigen-specific immune system modulation or immune checkpoint inhibition.[54] Immune checkpoint inhibitors are currently used in the treatment of many solid tumors such as malignant melanoma, renal cell cancer, and urothelial cancer.

Among other immunotherapeutic methods, monoclonal antibodies have the most clinical studies and are the most validated ones.[55,56] The working principle of monoclonal antibodies is that they activate the immunity by connecting with receptors on the cell surface. This activity varies: While the anti-CD20 (B lymphocyte antigen) monoclonal antibody induces apoptosis, the epidermal growth factor receptor (EGFR) binding antibody blocks the receptors by preventing the binding of naturally occurring ligands.[56]

In the adaptive immunotherapy method, immunologically active cells are given to patients, and T cells are used in this treatment method. These cells are tumor-infiltrating lymphocytes, T cells engineered for cancer-specific TCR expression, and T cells engineered for chimeric antigen receptor expression.[56]

In the formation of antibodies against the antigen in the tumor vaccines, it is aimed to activate CD4+ helper and CD8+ cytotoxic T cells.[54,57] An ideal tumor antigen should be expressed in an excessive amount and only on tumor cells, and the expression of the antigen should continue during the progression of the disease. Vaccines are developed on antigenic targets such as a recombinant protein, peptide, ganglioside, or tumor cell and in combination with an adjuvant that enhances the immune response.[54]

IMMUNOMODULATORY THERAPIES IN NON-SMALL CELL LUNG CANCER

Non-small-cell lung carcinoma (NSCLC) is one of the leading causes of cancer-related death in the world. Five-year survival rates of NSCLC cases are less than 20% despite chemotherapy and targeted treatments.[54,58] Nivolumab and pembrolizumab, which are PD-1 antibodies, and atezolizumab, durvalumab, and avelumab, which are among the PD-L1 antibodies, are agents that have been shown to be effective in different stages of NSCLC. Currently, three immunotherapy agents, approved by the FDA for use in patients with advanced-stage NSCLC, are nivolumab, pembrolizumab, and atezolizumab. All three agents have shown an overall survival (OS) advantage over docetaxel, a chemotherapy drug.[54,59-62]

Nivolumab is an IgG4 type anti-PD1 monoclonal antibody of human origin. Nivolumab’s efficacy was investigated in phase III studies after 17.6% response rates, and 42% OS rates for one year were reported in phase I studies with nivolumab in intensively treated non-squamous NSCLC cases. As a result, nivolumab was shown to be more effective than docetaxel.[54,63]

Pembrolizumab is a human IgG4 type monoclonal antibody developed against PD-1. In a study; in a group that had previously received platinum-based chemotherapy and tumor PD-L1 expression greater than 50%, regard to their OS results, it was approved by the FDA.[54,64] In another subsequent study, an advantage over docetaxel on OS was demonstrated in NSCLC cases who had previously received platinumbased chemotherapy and had PD-L1 expression of more than 1%.[65] The survival advantage is particularly pronounced in the group with PD-L1 expression greater than 50%, and the risk of death was reduced by approximately 50% with pembrolizumab.[54]

Atezolizumab is an IgG1 type anti-PD-L1 monoclonal antibody of human origin. The approval of atezolizumab for use in NSCLC cases is based on the results of an international phase III study.[62] The use of atezolizumab compared with docetaxel has been shown to provide a significant survival advantage in previously treated NSCLC patients.

METASTATIC RENAL CELL CARCINOMA TREATMENT WITH THE NIVOLUMAB AND IPILIMUMAB COMBINATION

Renal cell carcinoma (RCC) accounts for 2.4% of cancer cases worldwide, with 338,000 new diagnoses each year.[65,66] At the time of diagnosis, 25 to 30% of patients show metastatic disease associated with high mortality.[65,67,68] The combined use of immune checkpoint inhibitors results in an increased anti-tumor activity in tumor types such as melanoma compared to monotherapy.[69]

In the reference study, different doses of anti-PD-1 monoclonal antibody nivolumab and anti-CTLA-4 monoclonal antibody ipilimumab were used. A study was designed to find out both combination efficacy and which pathway inhibition is more effective. Nivolumab and ipilimumab doses were equal in the first group of the study, the dose of nivolumab was higher in the second group, and the dose of ipilimumab was higher in the third group.[65] Less adverse events (AEs) were encountered in the arm where nivolumab was used more than ipilimumab, and fewer patients required immune-modulating medication to cope with the side effects of AEs (mainly in the skin, endocrine and gastrointestinal system).[65]

PROGRESSIVE MULTIFOCAL LEUKOENCEPHALOPATHY TREATMENT WITH PEMBROLIZUMAB

Progressive multifocal leukoencephalopathy (PML) is a brain infection caused by the human polyomavirus-2 (JC virus) and is fatal unless the immune system is repaired.[70] The virus is estimated to be latent in the kidneys of more than 50% of healthy adults. It can transform into a neurotropic virus by undergoing genetic rearrangements in non-coding regions, thus infecting glia cells and causing PML.[70,71]

Programmed cell death protein-1 expression is upregulated in CD4+ and CD8+ T cells of PML patients.[70,72] An autopsy revealed increased PD-1 and PD-L1 expression in PML lesions. Pembrolizumab negatively regulates PD-1 expression in peripheral blood and cerebrospinal fluid. Programmed cell death protein-1 inhibition reactivates antitumor immune activity against various types of cancer.[70,73]

Findings suggest that pembrolizumab decreases JC viral load and increases CD4+ and CD8+ activity against the JC virus in some PML patients. In addition, magnetic resonance imaging showed that the size of PML lesions decreased.[70]

In conclusion, while some of the proteins activate the immune response, others are responsible for inhibiting this response. The main brake proteins involved in this negative regulation are CTLA-4, PD-1, TIM-3, and LAG-3. The cell can differentiate endogenous and exogenous substances through these brake proteins. Monoclonal antibodies are used to inhibit negative regulation in the treatment of various diseases. The main monoclonal antibodies that inhibit the PD-1/PD-L1 pathway are nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab. Recently, it has been proven that cancer immunotherapy based on PD-1 and PD-L1 is effective in generating an antitumor immune response with less toxicity in many tumor types. As a result of studies on melanoma and other diseases, it has been found that treatment with a combination of monoclonal antibodies is more successful than monotherapy.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Baş Aksu Ö, Şengül Ş. Immune checkpoints and inhibitors. J Ankara Univ Fac Med 2019;72:262-7.
  2. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci 2017;24:26.
  3. Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: The unique properties of PD-1 and their advantages for clinical application. Nat Immunol 2013;14:1212-8.
  4. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027- 34.
  5. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2001;2:261-8.
  6. Guo ZS. The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer). BMC Cancer 2018;18:1086.
  7. Allison AC. Adjuvants and immune enhancement. Int J Technol Assess Health Care 1994;10:107-20.
  8. Kwon ED, Hurwitz AA, Foster BA, Madias C, Feldhaus AL, Greenberg NM, et al. Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc Natl Acad Sci U S A 1997;94:8099-103.
  9. Gazdar AF, Kurvari V, Virmani A, Gollahon L, Sakaguchi M, Westerfield M, et al. Characterization of paired tumor and non-tumor cell lines established from patients with breast cancer. Int J Cancer 1998;78:766-74.
  10. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med 1999;190:355-66.
  11. Cameron F, Whiteside G, Perry C. Ipilimumab: First global approval. Drugs 2011;71:1093-104.
  12. 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. EMBO J 1992;11:3887-95.
  13. Raedler LA. Opdivo (Nivolumab): Second PD-1 inhibitor receives FDA approval for unresectable or metastatic melanoma. am health drug benefits 2015;8(Spec Feature):180-3.
  14. Allison JP, Honjo T. Press release: The Nobel Prize in physiology or medicine 2018. Available at: https:// www.nobelprize.org/prizes/medicine/2018/pressrelease/
  15. Sansom DM. CD28, CTLA-4 and their ligands: who does what and to whom? Immunology 2000;101:169-77.
  16. Xu Z, Juan V, Ivanov A, Ma Z, Polakoff D, Powers DB, et al. Affinity and cross-reactivity engineering of CTLA4-Ig to modulate T cell costimulation. J Immunol 2012;189:4470-7.
  17. Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD, et al. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med 2015;212:1603-21.
  18. Movva S, Verschraegen C. The monoclonal antibody to cytotoxic T lymphocyte antigen 4, ipilimumab (MDX-010), a novel treatment strategy in cancer management. Expert Opin Biol Ther 2009;9:231-41.
  19. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122-33.
  20. Huard B, Gaulard P, Faure F, Hercend T, Triebel F. Cellular expression and tissue distribution of the human LAG-3-encoded protein, an MHC class II ligand. Immunogenetics 1994;39:213-7.
  21. Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med 1990;171:1393-405.
  22. Kisielow M, Kisielow J, Capoferri-Sollami G, Karjalainen K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur J Immunol 2005;35:2081-8.
  23. Workman CJ, Wang Y, El Kasmi KC, Pardoll DM, Murray PJ, Drake CG, et al. LAG-3 regulates plasmacytoid dendritic cell homeostasis. J Immunol 2009;182:1885-91.
  24. Goldberg MV, Drake CG. LAG-3 in cancer immunotherapy. Curr Top Microbiol Immunol 2011;344:269-78.
  25. Andreae S, Piras F, Burdin N, Triebel F. Maturation and activation of dendritic cells induced by lymphocyte activation gene-3 (CD223). J Immunol 2002;168:3874-80.
  26. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 2009;10:29-37.
  27. Anderson AC, Anderson DE, Bregoli L, Hastings WD, Kassam N, Lei C, et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science 2007;318:1141-3.
  28. Hastings WD, Anderson DE, Kassam N, Koguchi K, Greenfield EA, Kent SC, et al. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur J Immunol 2009;39:2492-501.
  29. Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002;415:536-41.
  30. Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 2005;6:1245-52.
  31. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016;44:989-1004.
  32. Joller N, Kuchroo VK. Tim-3, Lag-3, and TIGIT. Curr Top Microbiol Immunol 2017;410:127-56.
  33. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999;11:141-51.
  34. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001;291:319-22.
  35. Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med 2003;9:1477-83.
  36. Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A 2005;102:11823-8.
  37. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther 2016;9:5023-39.
  38. Ishida M, Iwai Y, Tanaka Y, Okazaki T, Freeman GJ, Minato N, et al. Differential expression of PD-L1 and PD-L2, ligands for an inhibitory receptor PD-1, in the cells of lymphohematopoietic tissues. Immunol Lett 2002;84:57-62.
  39. Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, et al. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol 2002;169:5538-45.
  40. Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med 2003;198:39-50.
  41. Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 2002;9:133-45.
  42. Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 2002;9:133-45.
  43. Iwai Y, Okazaki T, Nishimura H, Kawasaki A, Yagita H, Honjo T. Microanatomical localization of PD-1 in human tonsils. Immunol Lett 2002;83:215-20.
  44. Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med 2016;375:1767-78.
  45. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat Med 2002;8:793-800.
  46. Nayak L, Iwamoto FM, LaCasce A, Mukundan S, Roemer MGM, Chapuy B, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017;129:3071-3.
  47. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015-26.
  48. Henson SM, Macaulay R, Riddell NE, Nunn CJ, Akbar AN. Blockade of PD-1 or p38 MAP kinase signaling enhances senescent human CD8(+) T-cell proliferation by distinct pathways. Eur J Immunol 2015;45:1441-51.
  49. Oestreich KJ, Yoon H, Ahmed R, Boss JM. NFATc1 regulates PD-1 expression upon T cell activation. J Immunol 2008;181:4832-9.
  50. Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, et al. IFN-a directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol 2011;186:2772-9.
  51. Youngblood B, Oestreich KJ, Ha SJ, Duraiswamy J, Akondy RS, West EE, et al. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8(+) T cells. Immunity 2011;35:400-12.
  52. Staron MM, Gray SM, Marshall HD, Parish IA, Chen JH, Perry CJ, et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8(+) T cells during chronic infection. Immunity 2014;41:802-14.
  53. Eno J. Immunotherapy through the years. J Adv Pract Oncol 2017;8:747-53.
  54. Özer L. Akci¤er kanserinde immünmodulatuvar tedaviler. Güncel Gö¤üs Hastalıkları Serisi 2018;6:85-98.
  55. Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003;9:269-77.
  56. Barbaros M, Dikmen M. Cancer immunotherapy. Erciyes Univ J of Sci and Tech 2015;31:177-82.
  57. Cuppens K, Vansteenkiste J. Vaccination therapy for non-small-cell lung cancer. Curr Opin Oncol 2014;26:165-70.
  58. Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL, et al. SEER Cancer Statistics Review, 1975-2014, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2014/, based on November 2016 SEER data submission, posted to the SEER web site, April 2017
  59. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123-35.
  60. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;373:1627-39.
  61. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced nonsmall-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540-50.
  62. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated nonsmall-cell lung cancer (OAK): A phase 3, openlabel, multicentre randomised controlled trial. Lancet 2017;389:255-65.
  63. Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced nonsmall-cell lung cancer. J Clin Oncol 2015;33:2004- 12.
  64. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018-28.
  65. Hammers HJ, Plimack ER, Infante JR, Rini BI, McDermott DF, Lewis LD, et al. Safety and Efficacy of Nivolumab in Combination With Ipilimumab in Metastatic Renal Cell Carcinoma: The CheckMate 016 Study. J Clin Oncol 2017;35:3851-8.
  66. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359-86.
  67. Cairns P. Renal cell carcinoma. Cancer Biomark 2010;9:461-73.
  68. Gupta K, Miller JD, Li JZ, Russell MW, Charbonneau C. Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): A literature review. Cancer Treat Rev 2008;34:193-205.
  69. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377:1345-56.
  70. Cortese I, Muranski P, Enose-Akahata Y, Ha SK, Smith B, Monaco M, et al. Pembrolizumab treatment for progressive multifocal leukoencephalopathy. N Engl J Med 2019;380:1597-605.
  71. Major EO, Yousry TA, Clifford DB. Pathogenesis of progressive multifocal leukoencephalopathy and risks associated with treatments for multiple sclerosis: a decade of lessons learned. Lancet Neurol 2018;17:467-80.
  72. Tan CS, Bord E, Broge TA Jr, Glotzbecker B, Mills H, Gheuens S, et al. Increased program cell death-1 expression on T lymphocytes of patients with progressive multifocal leukoencephalopathy. J Acquir Immune Defic Syndr 2012;60:244-8.
  73. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med 2018;50:1-11.