Open access peer-reviewed chapter

Immune Checkpoint Inhibitors Programmed Cell Death-1/Programmed Cell Death-Ligand1 (PD-1/PD-L1) for Cancer Therapy

Written By

Shaimaa M.M. Bebars

Submitted: 06 August 2022 Reviewed: 29 September 2022 Published: 30 January 2023

DOI: 10.5772/intechopen.108366

From the Edited Volume

Immune Checkpoint Inhibitors - New Insights and Recent Progress

Edited by Afsheen Raza

Chapter metrics overview

103 Chapter Downloads

View Full Metrics

Abstract

Monoclonal antibodies that inhibit “immune checkpoint” through programmed cell death-1 and its ligand (PD-1/PD-L1) blockage have proven remarkable therapeutic action toward a range of cancer types. Hence, immunotherapy, binding the immune system to act against malignant tumors, has generated encouraging outcomes in clinical practice. Nevertheless, the robust advantage is not observed in a large number of patients. Recognizing patients that will probably respond and using therapies covering a larger number of patients necessitate an enhanced understanding of the biological action of PD-1 and cytotoxic T lymphocyte antigen (CTLA) at the cell level and reviewing the performed clinical studies and their outcomes to recognize the accumulating proof of its clinical significance. In this chapter, we will discuss and review the clinical and preclinical data regarding Immune Checkpoint Inhibitors PD-1/PD-L1 to recognize the advances and challenges of their implication in clinical practice.

Keywords

  • immune checkpoint
  • PD-1
  • PD-L1
  • target therapy
  • immunotherapy
  • resistance

1. Introduction

Twenty-two years ago, Freeman et al. were the first to introduce programmed cell death-1 and its ligand (PD-1/PD-L1) as an immune checkpoint that later on was the immune inhibitor target in cancer therapy. Generally, checkpoints play a role as a brake to slow down the immune function, and it was proposed that inhibition of these checkpoints may stimulate T cells and eradicate malignant cells more effectively [1, 2].

In immune regulation, CD4+ regulatory T cell’s role is well recognized; they suppress autoimmunity and allergic reactions, on the other side, they enable cancer growth by inhibiting immunity against neoplasms [3, 4].

Immune evasion by cancer cells is mediated through acquiring mechanisms of resistance and escape including creating an environment of immunosuppression using immunosuppressive components such as molecules, cytokines of immune suppression type, and cells including CD4+ regulatory T cells to constrain active immunity against the tumor, thus endorsing tumor growth [4]. There is a suggestion of more malleability of the CD4+ regulatory T cell compartment expression with various aspects such as suppression function deactivation, inflammatory cytokines expression, and transcription reprogramming. Even though it is uncertain which element is responsible for CD4+ regulatory T cell malleability, there is probability that precise microenvironments have the inscription on CD4+ regulatory T cell destiny [5].

Numerous checkpoints of immune reaction have been established to bound the hyper-activation of immune cells involved in self-tolerance. One of the significant checkpoints is PD-1 and its ligands, PD-L1 and PD-L2 expressed mainly on the surface and in the body of lymphocytes, in antigen-presenting cells (APCs), and cancer cells and regulate responses toward antigens. Managing PD-1/PD-L actions facilitates the regulation of numerous immune-related disorders including infection, autoimmune disorders, and malignancies [6]. PD-L1 expression is related to microenvironment of T cells, cytokines produced by T cells of helper type, chemical mediators, interferons, and precise characters of gene expression. Continuous stimulation with an elevated level of PD-1 is frequently established in cancer inflammatory infiltrates mainly lymphocytes, as an expression of PD-L1 is utilized by neoplastic cells to escape immune damage. Blocking of these immune checkpoints is exploited to release the prospective of antagonistic therapy against cancer immune response as cancer treatment strategy [7].

Although there is a significant benefit in the outcome of patients attained with PD-L1/PD-1 blocking therapies, resistances are also commonly observed [8].

Advertisement

2. PD-1/PD-L1 immune checkpoint

Programmed cell death-1(PD-1/CD279) has two identified ligands, PD-L1 and 2 (CD274 and CD273 respectively), each with distinct expression patterns and regulations, of which PD-L1 is expressed in numerous cancers [9]. PD-1 is a transmembrane glycoprotein type 1 of the immunoglobulin superfamily, out of its total 288 amino acids, a 20% amino acid shows distinctiveness to cytotoxic T lymphocyte antigen 4 (CTLA4) and it is encoded by the PDCD1 gene on chromosome 2. PD-1 is formed of an extracellular domain of IgV-like and a transmembrane section. Its tyrosine-based switch motif (ITSM) and inhibitory motif tyrosine base are forming the tail which is located intracellularly. It was considered a CD28 receptor family member of T cells accessory molecules. Although PD-1 has similarities to the CD28 family, it shows distinctive properties distinguishing it from members of such family [10, 11].

PD-1 expression on effector T cells is mediated due to stimulation of the T cell receptor (TCR) and it plays a role as a receptor of immune inhibition. It binds the PD-L1 B7 homologs (B7-H1) and PD-L2 (B7-DC), existing mainly on APCs, and can be prompted in other tissues by cytokines of inflammation [1, 6]. PD-1 has a tendency concerning moderate local activation of T cells in tissues of the periphery. PD-1 may have delayed actions in the T cell activation and decay. Generally, PD-1/PD-L plays a significant role in sustaining T cell self-tolerance with the prevention of autoimmunity, and it reduces some anti-apoptotic molecules expression such as B-cell lymphoma-extra-large (BCL-XL) in addition to pro-inflammatory cytokines secretion [12, 13]. The inhibitory function of PD-1/PD-L1 is mediated primarily on effector T cells but also it acts on regulatory T cells by affecting phosphatidyl-inositol 3 kinase (PI3K) to control T cell autoimmunity and tolerance. In general, tolerance is the failure of T cells that may be manifested as ignorance (failure of activation), or “anergic” status (responding cells are persistent in a refractory status), or deletion (apoptosis of T cell) [3, 14].

PDCDL1 gene on chromosome 9 is responsible for PD-L1 coding in humans, PD-L1 a transmembrane protein type 1 was documented as a member of the family B7 protein. The length of PD-L1 is 290 amino acids of 40 kDa protein. PD-L1 consists of extracellular domains (IgV-like & IgC-like), a transmembrane domain (hydrophobic), and a 30 amino acids cytoplasmic tail. The PD-L1 constitutive expression can be detected at low levels, on inactive lymphocytes, APCs, and in syncytiotrophoblasts, corneal, endothelial, keratinocytes, and Langerhans’ islet cells of the pancreas as it plays a role in inflammatory response tissue homeostasis and giving a state of “immune privileged”, as the introduction of external antigens is tolerated with no immune or inflammatory response. PD-L1 is prompted as an inhibitory signal in inflammation acting upon immune, epithelial, and endothelial cells [15, 16].

Toll-like receptors (TLRs) in APCs affect PD-L1 expression through MEK/ERK (extracellular signal-regulated kinase) kinases activation, in addition to receptors of Interferon-gamma (IFN-γ) 1 & 2 via Jak (Janus kinase)/STAT (Signal transducer and activator of transcription)-mediated activation that can also influence MEK/ERK and PI3K/AKT pathway [17]. PD-1/PDL-1 ligation leads to SHP-1/SHP-2 recruitment to the ITSM with dephosphorylation of kinases e.g. CD3ζ, PKCθ, and ZAP70 leading to a general inhibition of T cell spreading out which results from PI3K-Akt and Ras-MEK-ERK cascade inhibition mostly through direct inactivation effect of PD-1of Ras &dephosphorylation of phospholipase Cγ [15, 18]. Dephosphorylation of Casein kinase 2 (CK-2), which is an SHP-2 target, causes uncontrolled activation of PTEN (PI3K-Akt signaling antagonist) [16, 17]. It was also suggested that CD28 receptor co-stimulation, maybe the main dephosphorylation target by SHP2 phosphatase [19]. PD-1/PD-L1 engagement modifies variable T cell activities including T cell proliferation deactivation, cytokine induction, survival, and other functions [20], the reaction between PI3K signaling and BCL-XL is a significant point of control where PD-1-inhibition of P13K decreases BCL-XL and endorses apoptosis [21] (Figure 1).

Figure 1.

Activation pathways of PD-1/PD-L1 expression.

The PD-1/PD-L1 interaction is critical for immune tolerance development, whether central or peripheral in primary or secondary lymphoid tissue respectively [22]. PD-1/PD-L1 knock-out in animal experiments causes autoimmunity with glomerulonephritis lupus-like arthritis and diabetes. While in humans, using antibodies against PD-1/PD-L1 leads to immune-related disorders such as endocrinopathy, colitis, and dermatoses [23, 24, 25]. A principal feature of T cell exhaustion, which is a marked weakening of effector T cell function, embraces the generation of several co-inhibitory pathways such as PD-1/PD-L1. Such impairment could be detected through apoptosis or inhibition of T cell development or production of regulatory T cells [26, 27]. The role of PD-1/PD-L1 is manifested in cases of T cell exhaustion, not only in chronic infection but also in cancer state [28, 29].

The greatest documented evidence of this inhibitory role in human immunity is derived from the usage of mediators to block the PD-1/PD-L1 pathway, an important target for immunotherapy in malignancy. Nevertheless, PD-1–PD-L1 interaction inhibition in patients suffering from malignancy causes anticancer immunity activation and autoimmune symptoms known as immune-associated opposing incidents [30].

Advertisement

3. PD-1/PD-L1 and cancer

3.1 PD-1/PD-L1 role in cancer

As PD-1/PD-L1 cardinal role is known in avoiding autoimmunity and preservation of normal peripheral tolerance, it is utilized by cancer cells in immune evasion eventually facilitating tumor growth, proliferation, and metastasis [6, 24].

Signaling of PD-1 in the tumor microenvironment, generated through interaction between cancer cells and non-transformed cells is an important player in the development and persistence of cancer through evasion of immune surveillance. PD-1 is markedly expressed in lymphocytes infiltrating a large number of cancers, also myeloid cells. While, PD-L1 is mainly conveyed on variable types of neoplastic cells such as melanoma, lung, renal and ovarian types. Moreover, PD-L1 expression can be up-regulated in different types of malignancy through carcinogenic signaling via aberrant PI3K-AKT activation or chromosomal amplifications and alterations, unrelated to inflammatory signaling in the microenvironment of malignancy [31, 32, 33].

Sometimes, PD-L1 expression is induced as a reaction to inflammation as an antitumor immune response via variable cytokines with IFN-γ being the most potent. IFN-γ causes PD-L1 activation and progression of ovarian neoplastic cells, while inhibition of IFN-γ receptor 1 can decrease PD-L1 expression in acute myeloid leukemia via the MEK/ERK and MYD88/TRAF6 pathways. Furthermore, IFN-γ prompts PKD2 (protein kinase D isoform 2), one of the PD-L1 regulator proteins. PD-L1 is also frequently expressed in cells of the microenvironment of tumors such as macrophages, dendritic cells, endothelial cells, and fibroblasts. PD-1/PD-L1 in tumor microenvironment endorses dysfunction and exhaustion of T cell, apoptosis, neutralization, and IL-10 secretion in a neoplastic mass generating resistance status against cancer cell destruction by a cytotoxic T cell (CD8+). Further infiltration by CD4+ regulatory T cells helps more suppression of effector immune response in tumors [31, 32].

The signaling of PD-L1 occurs via stimulation of PD-1which lead to pro-survival neoplastic signals induction helping anti-apoptotic effect via the cytoplasmic domain. In addition, PD-L1 protects neoplastic cells from the IFNs (I & II) cytotoxicity and CTL cytolysis. Furthermore, targeting PD-L1 decreases the activity of mTOR and neoplastic cell glycolytic metabolism without T cells [31, 34, 35].

PD-L1 positivity or negativity in tumor cells can be achieved via variable biological processes. T cells infiltrating the malignant tumor may produce surface expression of PDL-1 which can be lost in absence of T cells. Also, genetic proceedings inside the neoplastic cells could prevent PD-L1 expression. Thus, the neoplastic cell surface expression or absence of PD-L1 may implicate diverse functional significances and treatment allegations according to the causal expression mechanism [36].

Immunohistochemical expression of PD-L1 in neoplastic tissues shows that PD-L1 positive immune reaction may appear membranous or cytoplasmic. Trans membranous structure of PD-L1 suggests that the positive immune reaction may be related to the binding of PD-L1 antibody to a specific domain. While the cytoplasmic reaction may be related to the translocation of receptors onto the surface as a part of the immune response [37, 38, 39, 40].

Immunohistochemical expression of PD-L1 is still considered the merely broadly accessible, applicable, and cost-effective method for reviewing PD-L1 expression in cancer. Moreover, this method aids in recognizing patients who probably benefit from immunotherapy targeting PD-1/PD-L1. The FDA approval of such targeting therapy used trials depending on variable immunohistochemical platforms with different antibodies to evaluate PD-L1 expression on neoplastic cells, microenvironment immune cells, or both using specific scoring methods. These trials utilized particular PD-L1 inhibitors with particular assays and specific antibody reagents, thresholds, and protocols which should be standardized and validated. PD-L1 immunohistochemical procedure should be reproducible, both the staining technique and interpretation by pathologists which should be quality controlled starting from the tissue fixation and processing steps. Evolving applicable and reproducible scoring systems for PD-L1 is clinically important to identify patients who will probably benefit from targeted therapy. PD-L1 expression on both neoplastic and microenvironment immune cells has a greater association with clinical consequences in some neoplasms [41].

A recent systemic review and data analysis revealed the prognostic value of PD-L1. It shows high expression in solid malignancies which represents a bad prognostic feature regarding overall survival and progression-free survival [42]. The cytoplasmic expression and circulating tumor cells of PD-L1 were linked to better survival in thyroid carcinoma [43].

Blocking of PD-1 leads to suppression of transplanted myeloma cells growth in mice model while overexpressing PD-L1 in transplanted cells of mice model leads to the neoplastic establishment, load, and invasiveness which were reversed by using antibodies against PD-1/PD-L1. Thus, down-regulation of PD-1 and/or PD-L1 enriches T-cell activation against malignant cells which is the base for immunotherapy [33, 44].

3.2 PD-1/PD-L1 as a targeted therapy

Enhanced cancer suppression can be perceived when engaging different methodologies of PD-1/PD-L1 signaling disturbance, such as blocking using an antibody against PD-L1, DNA vaccination of PD-1 extracellular region, and injection of neoplasm-specific clones of T cells. The use of several immunotherapy approaches in combination may increase the therapeutic outcome [43, 44, 45].

The regulation of neoplastic cells’ expression of PD-L1 includes signaling pathways (MAPK, PI3K/Akt), transcription factors (e.g., Hypoxia-Inducible Factor 1, STAT3, Nuclear Factor kappa B, Transforming Growth Factor beta, GATA-3, and T-bet), and epigenetic and micro RNAs regulation [11].

In clinical practice, blocking of PD-1/PD-L1 causes inhibition of immune checkpoints which provides long-term responses, and as such the blocking antibodies are approved to treat solid and hematologic neoplasms. As well, the cytoplasmic PD-L1 knockdown using particular RNAs could benefit tumor immunotherapy [46].

The PD-L1/PD-1 blocking antibodies are the mainstay of immunotherapy due to improved survival and their clinical effectiveness in various malignancies such as non-small cell lung carcinoma (NSCLC) [47, 48]. Neoplasms showing an increased capacity for mutation and antigenicity, e.g., high microsatellite instability (MSI) and mismatch repair deficiencies (dMMR) are good targets for PD-1 blocking therapies. Numerous elements play significant roles as a determinant of clinical response to blocking PD-L1/PD-1 pathway such as neoplasm mutation load densities of immune cells and tumor microenvironment types of cells, PD-1/PD-L1 level of expression, and cytokines [49, 50].

Immunotherapy is considered a safe treatment compared with other strategies such as chemotherapy, irradiation, and surgery, as the mechanism involves augmentation of self-immunity against malignancy. Moreover, as a checkpoint inhibitor, it is extremely precise to a targeted cell with fewer side effects and keeps antigenic memory of neoplasm. However, it has been observed that associated toxicities with PD-1 blocking antibodies is lower than the associated toxicity with other immunotherapies, e.g., CTLA-4 blocking agents [45, 47, 51, 52, 53].

PD-1/PD-L1-induced opposing events related to immunity are one of the disadvantages of this type of therapy. It can induce side effects related to immunity in different organs such as the endocrine, pancreas, skin, gastrointestinal tract, liver, and renal system. Furthermore, such antibodies induced lethal xenogeneic hypersensitivity reactions in an experimental model of breast cancer after repeated administration. It is worth mentioning that PD-1 blocking-related pneumonitis is a significant side event mostly observed in NSCLC patients. Other systemic side effects, e.g., cardiac arrhythmia and even heart failure due to myocarditis have also been reported [30, 54, 55]. Clinically, subcutaneous or intravenous route of administration in this type of therapy is considered as one of the drawbacks, especially with humble penetration of neoplastic tissue [56].

PD-1 blocking mediators are associated with increased rates of recurrence and progression of the disease, nevertheless, local therapy can produce long-term survival without progression in some of these patients [57]. PD-L1 expression is used as an authenticated and main biomarker for the prediction of therapy; still, this biomarker alone, due to tumor heterogeneity is considered insufficient for defining patients who can benefit from PD-1/PD-L1 blocking therapy. Then again, PDL-1 single nucleotide polymorphisms (genetic copy number gains) have been suggested to help predict treatment responders, [52, 58, 59], especially in lymphoma [60, 61]. This is because de-glycosylation of the natural heavily glycosylated surface PD-1 molecules by enzymes during immunohistochemistry increases antibody binding ability of anti-PD-L1, thus increasing the intensity of signals leading to better outcome prediction [62].

The prediction of patient response is an important issue in PD-1/PD-L1 blocking therapy as only one to two-thirds of patients show resistance to treatment due to heterogeneity [52, 63]. The absence of neoplastic antigens, neoantigens or gene mutations, dysfunction of T cell, expression of PD-L1 and tumor microenvironment, noncoding RNA, and gut microbiome are also underlying factors serving as mechanisms of resistance to PD-1/PD-L1 blocking treatment [64].

Furthermore, PD-1 blocking therapy is largely costing more than other immunotherapy treatments and original lines of treatment, especially in old patients with low income and there is a debate regarding the cost-benefit relationship in combined therapy [65]. Using such an immunotherapy line of treatment in patients with immune disease whether hyperactive or autoimmune or hypoactive or even deficient immunity is considered a challenge, particularly with the toxicity effect of such therapy [53, 66].

3.3 Improving the PD-1/PD-L1 blocking efficacy

The regulatory mechanisms and pathways of PD-L1 expression have been widely investigated to understand side effects and deficient responses in some patients. The combination approaches were introduced to concurrently enhance several cancer-immunity processes, eliminate brakes of immunosuppression, and coordinate an immune-enhanced neoplastic microenvironment [53, 67].

Combination lines of treatment, such as combining anti-PD-1/PD-L1 with radiotherapy, chemotherapy, other immune checkpoint inhibitors or targeted therapy, interferon genes agonists stimulator, transplantation/manipulation of microbiome, epigenetic or metabolic modulators may produce better treatment response. Furthermore, agents containing both PD-1 and PD-L1 targeting antibodies may also provoke a potent effect. Regulation of PD-L1 expression in the tumor microenvironment through medical treatment or regulation of genes expands the -PD-1/PD-L1 therapy effect [68, 69, 70].

Possible small-molecule mediators were suggested to be used for targeting PD-L1 which may help overcome the restrictions of monoclonal antibodies used in blocking PD-1/PD-L1. Preclinical investigations suggest that the combination of these small-molecule mediators with other immune checkpoints targeting agents may cause augmented antineoplastic action or use such small molecules to up-regulate PD-L1 and elevate the blocking efficacy [55, 71]. Moreover, prodrug nanoparticles conjugated with anti-PD-L1 peptide were suggested to be used to help inhibit neoplastic growth with minimum side events [72].

Other effective antineoplastic agents, e.g., antiangiogenic mediators, and immunogenic cell death inducers combined with immune checkpoint inhibitors may help as a preventive therapeutic method in improving blocking agents’ efficacy [73, 74].

Advertisement

4. Conclusion

Taking into account the various potential strategies for successful PD-1/PD-L1 checkpoint inhibition such as blocking using antibodies against PD-L1, DNA vaccination of PD-1 extracellular region, injection of neoplasm-specific clones of T cell, with understanding mechanisms of action is important in clinical practice. A deep consideration of the mechanisms of resistance, whether cellular or molecular will benefit patients and improve therapeutic approaches. The combination approaches of immunotherapy or with other lines and strategies of therapy were introduced, also, small-molecule mediators and prodrug nanoparticle conjugation were suggested to be helpful in cases of anticipated resistance. The future perspective of combination therapy and investigation of predictive biomarkers will provide an important pathway for cancer patient care in cases treated with PD-1/PD-L1 checkpoint inhibition.

References

  1. 1. 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. The Journal of Experimental Medicine. 2000;192(7):1027-1034. DOI: 10.1084/jem.192.7.1027
  2. 2. Ljunggren HG, Jonsson R, Hoglund P. Seminal immunologic discoveries with direct clinical implications: The 2018 nobel prize in physiology or medicine honours discoveries in cancer immunotherapy. Scandinavian Journal of Immunology. 2018;88:e12731. DOI: 10.1111/sji.12731
  3. 3. Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nature Immunology. 2010;11(1):7-13. DOI: 10.1038/ni.1818
  4. 4. Takeuchi Y, Nishikawa H. Roles of regulatory T cells in cancer immunity. International Immunology. 2016;28(8):401-409. DOI: 10.1093%2Fintimm%2Fdxw025
  5. 5. Hatzioannou A, Boumpas A, Papadopoulou M, Papafragkos I, Varveri A, Alissafi T, et al. Regulatory T cells in autoimmunity and cancer: A duplicitous lifestyle. Frontiers in Immunology. 2021;12:731947. DOI: 10.3389/fimmu.2021.731947
  6. 6. Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Current Opinion in Immunology. 2014;27:1-7. DOI: 10.1016/j.coi.2013.12.005
  7. 7. Chamoto K, Al-Habsi M, Honjo T. Role of PD-1 in immunity and diseases. In: Akihiko Y, editor. Emerging Concepts Targeting Immune Checkpoints in Cancer and Autoimmunity. 2017. pp. 75-97. DOI: 10.1007/978-3-319-68929-6
  8. 8. Pardoll D. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews. Cancer. 2012;12:252-264. DOI: 10.1038/nrc3239
  9. 9. Hudson K, Cross N, Jordan-Mahy N, Leyland R. The extrinsic and intrinsic roles of PD-L1 and its receptor PD-1: Implications for immunotherapy treatment. Frontiers in Immunology. 2020;2362:568931. DOI: 10.3389/fimmu.2020.568931
  10. 10. 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. Nature Medicine. 2002;8:793-800. DOI: 10.1038/nm730
  11. 11. Carreno BM, Collins M. The B7 family of ligands and its receptors: New pathways of costimulation and inhibition of immune responses. Annual Review of Immunology. 2002;20:29. DOI: 10.1146/annurev.immunol.20.091101.091806
  12. 12. Dermani FK, Samadi P, Rahmani G, Kohlan AK, Najafi R. PD-1/PD-L1 immune checkpoint: Potential target for cancer therapy. Journal of Cellular Physiology. 2019;234(2):1313-1325. DOI: 10.1002/jcp.27172
  13. 13. 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
  14. 14. Tan CL, Kuchroo JR, Sage PT, Liang D, Francisco LM, Buck J, et al. PD-1 restraint of regulatory T cell suppressive activity is critical for immune tolerance. Journal of Experimental Medicine. 2021;218(1):e20182232. DOI: 10.1084/jem.20182232
  15. 15. 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
  16. 16. Kythreotou A, Siddique A, Mauri FA, Bower M, Pinato DJ. PD-L1. Journal of Clinical Pathology. 2018;71(3):189-194. DOI: 10.1136/jclinpath-2017-204853
  17. 17. Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. New England Journal of Medicine. 2016;375(18):1767-1778. DOI: 10.1056/nejmra1514296
  18. 18. Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Molecular Cancer. 2019;18(1):1-7. DOI: 10.1186/s12943-018-0928-4
  19. 19. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. The Journal of Experimental Medicine. 2012;209:1201-1217. DOI: 10.1084/jem.20112741
  20. 20. Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355:1428-1433
  21. 21. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111-122
  22. 22. 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%2Fjlb.1212621
  23. 23. Xing Y, Hogquist KA. T-cell tolerance: Central and peripheral. Cold Spring Harbor Perspectives in Biology. 2012;4(6):a006957
  24. 24. Nishimura H, Nose M, Hiai H, et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141-151
  25. 25. Keir ME, Liang SC, Guleria I, et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. The Journal of Experimental Medicine. 2006;203:883-895
  26. 26. Kumar V, Chaudhary N, Garg M, et al. Current diagnosis and management of immune related adverse events (irAEs) induced by immune checkpoint inhibitor therapy. Frontiers in Pharmacology. 2017;8:49
  27. 27. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nature Reviews. Immunology. 2015;15:486-499
  28. 28. Wherry EJ. T cell exhaustion. Nature Immunology. 2011;131:492-499
  29. 29. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682-687
  30. 30. Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S, et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. European Journal of Cancer. 2016;60:190-209. DOI: 10.1016/j.ejca.2016.02.025
  31. 31. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Research. 2005;65:1089-1096
  32. 32. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. Journal of Hematology & Oncology. 2019;12(1):1-3. DOI: 10.1186/s13045-019-0779-5
  33. 33. Ai L, Xu A, Xu J. Roles of PD-1/PD-L1 pathway: Signaling, cancer, and beyond. Regulation of Cancer Immune Checkpoints. 2020;1248:33-59. DOI: 10.1007/978-981-15-3266-5_3
  34. 34. Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen L. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood. 2008;111:3635-3643
  35. 35. Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229-1241
  36. 36. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? The Journal of Experimental Medicine. 2016;213(13):2835-2840. DOI: 10.1084/jem.20161462
  37. 37. Gato-Cañas M, Zuazo M, Arasanz H, Ibañez-Vea M, Lorenzo L, Fernandez-Hinojal G, et al. PDL1 signals through conserved sequence motifs to overcome interferon-mediated cytotoxicity. Cell Reports. 2017;20:1818-1829
  38. 38. Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: A novel role of pro-survival signalling in cancer. Annals of Oncology. 2016;27(3):409-416. DOI: 10.1093/annonc/mdv615
  39. 39. Mahoney KM, Sun H, Liao X, Hua P, Callea M, Greenfield EA, et al. PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells immunohistochemical detection of membrane PD-L1. Cancer Immunology Research. 2015;3(12):1308-1315. DOI: 10.1158/2326-6066.CIR-15-0116
  40. 40. Akhtar M, Rashid S, Al-Bozom IA. PD− L1 immunostaining: What pathologists need to know. Diagnostic Pathology. 2021;16(1):1-2
  41. 41. Chowdhury S, Veyhl J, Jessa F, Polyakova O, Alenzi A, MacMillan C, et al. Programmed death-ligand 1 overexpression is a prognostic marker for aggressive papillary thyroid cancer and its variants. Oncotarget. 2016;7(22):32318
  42. 42. D’Andréa G, Lassalle S, Guevara N, Mograbi B, Hofman P. From biomarkers to therapeutic targets: The promise of PD-L1 in thyroid autoimmunity and cancer. Theranostics. 2021;11(3):1310. DOI: 10.7150/thno.50333
  43. 43. Wang Q , Liu F, Liu L. Prognostic significance of PD-L1 in solid tumor: An updated meta-analysis. Medicine. 2017;96(18):e6369 DOI: 10.1097/md.0000000000006369
  44. 44. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proceedings of the National Academy of Sciences. 2002;99(19):12293-12297. DOI: 10.1073/pnas.192461099
  45. 45. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity. Nature Medicine. 2003;9(5):562-567. DOI: 10.1038/nm863
  46. 46. He YF, Zhang GM, Wang XH, Zhang H, Yuan Y, Li D, et al. Blocking programmed death-1 ligand-PD-1 interactions by local gene therapy results in enhancement of antitumor effect of secondary lymphoid tissue chemokine. The Journal of Immunology. 2004;173(8):4919-4928. DOI: 10.4049/jimmunol.173.8.4919
  47. 47. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Research. 2004;64(3):1140-1145. DOI: 10.1158/0008-5472.can-03-3259
  48. 48. Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Computational and Structural Biotechnology Journal. 2019;17:661-674. DOI: 10.1016%2Fj.csbj.2019.03.006
  49. 49. Cui G. The mechanisms leading to distinct responses to PD-1/PD-L1 blockades in colorectal cancers with different MSI statuses. Frontiers in Oncology. 2021;11:573547. DOI: 10.3389/fonc.2021.573547
  50. 50. Robert C, Ribas A, Schachter J, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): Post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. The Lancet Oncology. 2019;20(9):1239-1251. DOI: 10.1016/S1470-2045(19)30388-2
  51. 51. Khan M, Lin J, Liao G, Tian Y, Liang Y, Li R, et al. Comparative analysis of immune checkpoint inhibitors and chemotherapy in the treatment of advanced non-small cell lung cancer: A meta-analysis of randomized controlled trials. Medicine. 2018;97(33):e11936 DOI: 10.1097%2FMD.0000000000011936
  52. 52. Kopetz S, Lonardi S, McDermott RS, Aglietta M, Hendlisz A, Morse M, et al. Concordance of DNA mismatch repair deficient (dMMR)/microsatellite instability (MSI) assessment by local and central testing in patients with metastatic CRC (mCRC) receiving nivolumab (nivo) in CheckMate 142 study. DOI: 10.1200/JCO.2017.35.15_suppl.3548
  53. 53. Wu K, Yi M, Qin S, Chu Q , Zheng X, Wu K. The efficacy and safety of the combination of PD-1 and CTLA-4 inhibitors: A meta-analysis. Experimental Hematology & Oncology. 2019;8(1):1-2. DOI: 10.1186/s40164-019-0150-0
  54. 54. Constantinidou A, Alifieris C, Trafalis DT. Targeting programmed cell death-1 (PD-1) and ligand (PD-L1): A new era in cancer active immunotherapy. Pharmacology & Therapeutics. 2019;194:84-106. DOI: 10.1016/j.pharmthera.2018.09.008
  55. 55. Makuku R, Khalili N, Razi S, Keshavarz-Fathi M, Rezaei N. Current and future perspectives of PD-1/PDL-1 blockade in cancer immunotherapy. Journal of Immunology Research. 2021;2021:6661406. DOI: 10.1155/2021/6661406
  56. 56. Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. Journal of Clinical Oncology. 2017;35(7):709. DOI: 10.1200/JCO.2016.68.2005
  57. 57. Läubli H, Balmelli C, Bossard M, Pfister O, Glatz K, Zippelius A. Acute heart failure due to autoimmune myocarditis under pembrolizumab treatment for metastatic melanoma. Journal for Immunotherapy of Cancer. 2015;3(1):1-6. Available from: https://link.gale.com/apps/doc/A541676778/AONE?u=anon~af3806cf&sid=googleScholar&xid=e07d2490
  58. 58. Yamaguchi H, Hsu JM, Yang WH, Hung MC. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nature Reviews Clinical Oncology. 2022;19(5):287-305. DOI: 10.1038/s41571-022-00601-9
  59. 59. Klemen ND, Wang M, Feingold PL, Cooper K, Pavri SN, Han D, et al. Patterns of failure after immunotherapy with checkpoint inhibitors predict durable progression-free survival after local therapy for metastatic melanoma. Journal for Immunotherapy of Cancer. 2019;7(1):1-9. DOI: 10.1186/s40425-019-0672-3
  60. 60. Chen Q , Li T, Yue W. Drug response to PD-1/PD-L1 blockade: Based on biomarkers. OncoTargets and Therapy. 2018;11:4673. DOI: 10.2147/OTT.S168313
  61. 61. Udall M, Rizzo M, Kenny J, Doherty J, Dahm S, Robbins P, et al. PD-L1 diagnostic tests: A systematic literature review of scoring algorithms and test-validation metrics. Diagnostic Pathology. 2018;13(1):1-1. DOI: 10.1186/s13000-018-0689-9
  62. 62. Budczies J, Denkert C, Győrffy B, Schirmacher P, Stenzinger A. Chromosome 9p copy number gains involving PD-L1 are associated with a specific proliferation and immune-modulating gene expression program active across major cancer types. BMC Medical Genomics. 2017;10(1):1-8. DOI: 10.1186/s12920-017-0308-8
  63. 63. Nayak L, Iwamoto FM, LaCasce A, Mukundan S, Roemer MG, Chapuy B, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood, The Journal of the American Society of Hematology. 2017;129(23):3071-3073. DOI: 10.1182/blood-2017-01-764209
  64. 64. Xu J, Yang X, Mao Y, Mei J, Wang H, Ding J, et al. Removal of N-linked glycosylation enhances PD-L1 detection in colon cancer: Validation research based on immunohistochemistry analysis. Technology in Cancer Research & Treatment. 2021;20:15330338211019442
  65. 65. LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 pathway in tumor therapy. The Journal of Immunology. 2018;200(2):375-383. DOI: 10.4049/jimmunol.1701044
  66. 66. Sun JY, Zhang D, Wu S, Xu M, Zhou X, Lu XJ, Ji J. Resistance to PD-1/PD-L1 blockade cancer immunotherapy: Mechanisms, predictive factors, and future perspectives. Biomarker Research 2020;8(1):1-0. DOI: 10.1186/s40364-020-00212-5
  67. 67. Pike E, Hamidi V, Saeterdal I, Odgaard-Jensen J, Klemp M. Multiple treatment comparison of seven new drugs for patients with advanced malignant melanoma: A systematic review and health economic decision model in a Norwegian setting. BMJ Open. 2017;7(8):e014880. DOI: 10.1136/bmjopen-2016-014880
  68. 68. Alexander S, Swami U, Kaur A, Gao Y, Fatima M, Ginn MM, et al. Safety of immune checkpoint inhibitors in patients with cancer and pre-existing autoimmune disease. Annals of Translational Medicine. 2021;9(12):1033. DOI: 10.21037/atm-20-8124
  69. 69. 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
  70. 70. Yi M, Zheng X, Niu M, Zhu S, Ge H, Wu K. Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Molecular Cancer. 2022;21(1):1-27. DOI: 10.1186/s12943-021-01489-2
  71. 71. Dai X, Bu X, Gao Y, Guo J, Hu J, Jiang C, et al. Energy status dictates PD-L1 protein abundance and anti-tumor immunity to enable checkpoint blockade. Molecular Cell. 2021;81(11):2317-2331. DOI: 10.1016/j.molcel.2021.03.037
  72. 72. Moon Y, Shim MK, Choi J, Yang S, Kim J, Yun WS, et al. Anti-PD-L1 peptide-conjugated prodrug nanoparticles for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death. Theranostics. 2022;12(5):1999. DOI: 10.7150/thno.69119
  73. 73. Song Y, Fu Y, Xie Q , Zhu B, Wang J, Zhang B. Anti-angiogenic agents in combination with immune checkpoint inhibitors: A promising strategy for cancer treatment. Frontiers in Immunology. 2020;11:1956. DOI: 10.3389/fimmu.2020.01956
  74. 74. Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF, Oves M. Revolutionization in cancer therapeutics via targeting major immune checkpoints PD-1, PD-L1 and CTLA-4. Pharmaceuticals. 2022;15(3):335. DOI: 10.3390/ph15030335

Written By

Shaimaa M.M. Bebars

Submitted: 06 August 2022 Reviewed: 29 September 2022 Published: 30 January 2023