Next Article in Journal
Role of Micronutrients in the Response to SARS-CoV-2 Infection in Pediatric Patients
Previous Article in Journal
Synergistic Effect of Co-Administered SARS-CoV-2 Vaccines Improves Immune Responses in BALB/c Mice: A Preliminary Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions

1
Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
2
Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
*
Author to whom correspondence should be addressed.
Immuno 2024, 4(3), 186-210; https://doi.org/10.3390/immuno4030013
Submission received: 24 April 2024 / Revised: 21 June 2024 / Accepted: 28 June 2024 / Published: 5 July 2024

Abstract

:
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.

1. Introduction

Over the past few decades, many types of cancer immunotherapies have been studied such as monoclonal antibodies (mAb), cancer vaccines and adoptive T-cell therapies [1,2,3]. They each target different components of the immune system and have shown varying levels of promise as well as limitations [4]. However, no immunotherapy has shown as much promise in recent years as immune checkpoint inhibitors (ICI) [5]. Cancer cells can evade immunosurveillance through activation of immune checkpoint pathways, inhibiting T-cell activation and diminishing anti-tumour immune responses [6]. Immune checkpoint pathways are activated when co-inhibitory molecules on the surface of cancer cells or antigen-presenting cells (APCs) bind to T-cell surface molecules known as immune checkpoints [7]. ICIs act to block immunosuppressive signalling by inhibiting this binding, reviving anti-tumour activity and preventing cancer progression [5]. In 2011, the Food and Drug Administration (FDA) approved ipilimumab, the first ICI for clinical use in the treatment of metastatic melanoma. Ipilumumab is an inhibitory mAb targeting CD152, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [8]. This marked the beginning of a new era of cancer immunotherapy, and since then, a number of other ICIs have also been approved including: pembrolizumab targeting the protein CD279, programmed cell death protein (PD-1); atezolizumab targeting its ligand CD274, programmed cell death-ligand 1 (PD-L1) and most recently, relatlimab targeting the protein CD223, Lymphocyte-activation gene 3 (LAG-3) [9,10]. Clinical trials have shown promising therapeutic outcomes of ICIs in several different cancers, including melanoma, lung cancer, urothelial cancer, renal cancer, hepatocellular cancer and many more, for which mainstream use has been approved [11,12,13,14]. Additionally, there are hundreds of active clinical trials underway to evaluate the efficacy and safety of multiple ICIs across many different cancers [3]. However, despite the success of ICIs in studied patient populations, only a fraction of cancer patients benefit from ICIs in practice [5]. The efficacy of ICI relies on the basis that there is an underlying anti-tumour immune response present, and the ICI is simply removing the brakes to release this function. However, the degree of anti-tumour immunity is variable and regulated by complex factors in the tumour microenvironment (TME), the immunogenicity of the tumour and its mutational burden [15,16,17]. ICIs are also associated with a unique spectrum of immune-related adverse effects (irAEs), such as dermatological, gastrointestinal and endocrine inflammatory reactions [18]. These are some limitations that must be addressed if ICIs are to be effective in wider patient populations.

2. Immune Checkpoint Pathways

Immune checkpoints are important regulators of the immune system. They act as T-cell receptor (TCR) co-signalling partners, delivering either activating or inhibitory signals to T cells upon stimulation. These signalling pathways are vital for maintaining self-tolerance as well as ensuring an adequate immune response [19]. An example of a stimulatory checkpoint protein is the CD28 molecule, a protein highly expressed on CD4+ and CD8+ T cells, which initiates co-stimulatory signals when it binds to its corresponding ligands, CD80 and CD86, found on APCs [20]. To date, a total of seven other stimulatory checkpoints have been identified, all of which act in a similar way to activate and stimulate T-cell activity [19]. However, inhibitory immune checkpoints have been the main subject of attention in recent years. Their co-inhibitory pathways have been shown to promote tumour formation and progression, through the dampening of cancer immunosurveillance [21]. In fact, cancer cells have been shown to utilise these pathways by expression of their ligands, activating immunosuppressive signalling and allowing for immune evasion [22]. The therapeutic potential surrounding these pathways is evident, and since the success of the first ICI, ipilimumab, targeting CTLA-4, many more inhibitory immune checkpoints have been identified. As of now, the immune checkpoints which have been successfully targeted with approved ICIs are CTLA-4, PD-1 (CD279), its ligand PD-L1 (CD274) and LAG-3 (CD223) [5,10]. Other co-inhibitory immune checkpoints include T-cell immunoglobulin and mucin-domain-containing-3 (TIM-3, CD366), T-cell immunoglobulin and ITIM domain (TIGIT), V-domain Ig suppressor of T-cell activation (VISTA) and many more (Figure 1) [23]. These are all emerging ICI targets, currently under clinical trial or development, with many showing promising outcomes [23]. However, before we focus on what the future holds, it is important to understand the current established ICI targets, as ultimately it is their success and current limitations that are guiding research into these new targets. The next section will look in more depth at the physiological roles of these immune checkpoints, the mechanism of their blockade by ICIs and analyse the success of this immunotherapy in treatment of advanced cancers.

3. CTLA-4 Physiological Role

CTLA-4 is a transmembrane protein belonging to the immunoglobulin superfamily, and its expression and function are intrinsically linked to T-cell activation [8,19]. In fact, CTLA-4 is not detectable on naïve T cells but is immediately induced and upregulated in response to TCR activation [21]. Its expression is regulated by the strength of TCR signalling, whereby stronger TCR signalling induces greater expression of CTLA-4 [24]. CTLA-4 is a homologue of the co-stimulatory checkpoint CD28 and, as such, shares its ligands, CD80 (B7-1) and CD86 (B7-2), also known as the B7 ligands [21]. Following its expression, it begins to compete with CD28 for the B7 ligands; however, as it boasts a higher affinity and avidity for these ligands, it can effectively outcompete CD28, blocking co-stimulatory signals and attenuating T-cell activation [24]. Thus, it regulates the availability of B7 ligands to CD28, forming a homeostatic feedback loop in response to TCR stimulation. The net outcome following TCR stimulation is dependent on the relative binding of CD28 versus CTLA-4 to the B7 ligands and can result in either T-cell activation or anergy [25]. Other than its primary cell-intrinsic role in regulating T-cell activity, it also influences immunological tolerance through cell-extrinsic mechanisms, mainly through the action of T-regulatory cells (T-reg) [21]. Unlike effector T cells, CTLA-4 is constitutively expressed on T-reg cells, a cell with a key role in maintaining immune tolerance [26]. CTLA-4 expressed on T-reg cells acts in a similar way, limiting the availability of B7 ligands, in a cell-extrinsic manner, and blocking CD28 costimulatory activation of nearby effector T cells through competitive inhibition [21]. Additionally, beyond just the binding of B7 ligands, CTLA-4 has also been reported to limit the overall availability of B7 ligands through trans-endocytosis of these ligands from APCs [27]. It is quite evident that CTLA-4’s function is inherently linked to the interaction of its homologue CD28 and its ligands, as its primary mechanism is to impede this signalling. However, some evidence suggests that CTLA-4 may also have a role independent of CD28, in which it directly antagonises TCR signalling following B7-1 engagement, through its own cell-intrinsic signalling [28]. A proven mechanism has yet to be established, and further research is needed to understand the intracellular signalling of CTLA-4. Ultimately, CTLA-4 has a fundamental role in maintaining self-tolerance, and as such, it primarily regulates T-cell activation at sites of T-cell priming, such as lymph nodes [21]. Its critical role in immune tolerance has been proven by observations seen in CTLA-4 knockout mice, which developed uncontrolled lymphoproliferative disease and fatal autoimmunity, due to unrestrained CD28 co-stimulation [29].
Considering it has such an important role in self-tolerance, it makes sense that on the opposite end of the scale, excessive CTLA-4 activity can produce an immunosuppressive environment conducive with tumorigenesis [24]. Such a concept was given more weight by pre-clinical studies conducted in the late 1990s, which showed decreased tumour growth and improved survival in mouse models following CTLA-4 pathway blockade [30]. Of course, the next step was to develop an approved anti-CTLA-4 therapy that could replicate such results in human cancer patients. Initially, three anti-CTLA4 antibodies entered clinical trials: tremelimumab, its parental antibody known as CP-642,570 and ipilimumab [8]. CP-642,570 did not make it past the first human trial, and tremelimumab showed initial promise in phase I and II trials, but ultimately failed to meet its endpoints in phase III trials [8,31,32]. This left ipilimumab, a fully humanized IgG1 mAb, that went on to achieve FDA and European Medicines Agency (EMA) approval for the treatment of unresectable stage III and metastatic melanoma, following many successful clinical trials [8]. In the last few years, it has also been approved as part of combination therapy with nivolumab, an anti-PD-1 mAb, for unresectable or metastatic melanoma, advanced renal cell carcinoma (RCC), some forms of advanced colorectal carcinoma and many more (Table 1) [33].

3.1. Mechanism of CTLA-4 Blockade-Induced Tumour Rejection

The exact mechanism of CTLA-4 blockade-induced tumour rejection by ipilimumab is still not completely clear, with many distinct mechanisms thought to play a role. The primary mechanism is centred around the direct blockade of CTLA-4 competition for B7 ligands, allowing unrestricted CD28 co-stimulation and increasing activation of effector T cells in response to tumour-associated antigens (TAAs) (Figure 2) [21]. As B7 ligands are not found on cancer cells, this mainly occurs in the tumour-draining lymph nodes, where APCs cross-present TAAs to tumour-reactive effector cells [21]. The result is increased activation and activity of effector CD8+ cytotoxic T-lymphocytes (CTLs) and CD4+ type 1 helper (Th1) cells, the primary mediators of the anti-tumour immune response [21]. Additionally, within the TME, CTLA-4 blockade acts to reactivate and expand exhausted CD8+ T-cell and Th1 cells through the blockade of CTLA-4 [40]. Exhaustion occurs due to chronic TAA-induced TCR stimulation, resulting in persistently high CTLA-4 expression and greatly reduced activity of primed immune cells [41]. Therefore, CTLA-4 blockade removes this inhibitory control, reviving effector T-cell function [40].
Depletion of T-reg cells within the TME is the other main mechanism that is thought to contribute to CTLA-4 blockade-induced tumour rejection [42]. CTLA-4 signalling plays an important role in the immunosuppressive function of T-regs, and as such, blockade of such signalling removes this immunosuppressive influence that otherwise promotes tumour growth and progression [42]. This is achieved through the selective depletion of intra-tumoural T-reg cells, not peripheral populations [43]. This selectivity is likely due to increased expression of CTLA-4 T-regs in the TME or, additionally, differences in activity and availability of receptors for crystallizable fragment of immunoglobulin (FcR), which is thought to mediate the mechanism of T-reg depletion. Laboratory investigations showed that binding of the Fc portion of anti-CTLA4 antibodies to FcR found on APCs were necessary for the depletion of intra-tumoural T-regs and subsequent anti-tumour effect [44].
Interestingly, a recent study has gone as far as to say that the efficacy of ipilimumab is completely independent of B7 ligands, instead solely driven by antibody-mediated T-reg depletion [45]. Such a conclusion of course contradicts years of prior evidence, and although recent studies do suggest a greater role of T-reg depletion, a large body of research backs the concept of B7 ligand regulation as a critical mechanism of ipilimumab [21]. Also, there is not yet any definitive evidence of T-reg depletion in humans treated with ipilimumab. In reality, both mechanisms could likely contribute to the efficacy of ipilimumab to some degree, as both effector T cells and T-regs express CTLA-4 and have roles in checkpoint signalling pathways [46]. However, the relative contribution of cell intrinsic expansion of effector T-cell function versus T-reg depletion remains unclear, and as such, further research is needed to better understand the weight of each mechanism. To be able to delineate the primary and more effective mechanism behind CTLA-4 blockade-induced tumour rejection could potentially guide development of new anti-CTLA-4 therapies, more focused on targeting these mechanisms. For example, ipilimumab was initially developed based on the knowledge that CTLA-4 blockade would enhance T-cell activity and, as such, was not developed to be a depleting antibody and target T-regs in any way [47]. If T-reg depletion did in fact play a more central role in tumour rejection in humans, then this would change the direction of future anti-CTLA4 therapies.

3.2. Efficacy of Ipilimumab in Treatment of Advanced Melanoma

For decades prior to the introduction to ipilimumab, there was little improvement in the treatment available for patients with advanced or metastatic melanoma, with no significant improvement seen in overall survival (OS) of patients in this time [48]. The chemotherapeutic drug dacarbazine remained the reference single agent of choice, despite it showing no impact on survival [49]. Ipilumumab was the first immunotherapy to show improved OS in this patient population with high unmet needs, completely changing the therapeutic outlook for this cancer following its FDA approval [8]. Pre-clinical data of CTLA-4 blockade in mice showed that it could not only enhance the endogenous anti-tumour immunity against cancers but also be used synergistically in combination with other interventions such as chemotherapy or vaccines to improve response against less immunogenic cancer [50]. Findings from phase I trials extended these promising results, showing anti-tumour immunity in various advanced tumours, especially melanoma, and demonstrated a favourable safety and toxicity profile both alone and in combination [49].
As ipilimumab’s clinical development progressed into phase II trials, it continued to show promising outcomes. In a Phase II trial (MDX010-08), 72 previously untreated advanced melanoma patients were randomised to receive either ipilimumab 3 mg/kg monotherapy or in combination with dacarbazine. In both cases, ipilimumab showed strong objective clinical outcomes and promising OS in patients [51]. When comparing the long-term survival data in this trial against another trial, MDX010-15, a phase 1/2 dose-ranging study in which 23 patients were treated with ipilimumab at 10 mg/kg dose every 3 weeks, a trend towards better survival with a higher dose was demonstrated. A 2-year survival rate of 36% was seen in the 10 mg/kg dose vs. 22% for the 3 mg/kg dose [52]. A similar trend was seen in a phase 1/2 study (CA184-022), in which 217 previously treated advanced melanoma patients underwent induction therapy with one of three doses of ipilimumab, followed by maintenance therapy. Disease control rate (DCR) was 13.7%, 26.4%, and 29.2% in the 0.3 mg/kg, 3 mg/kg, and 10 mg/kg groups, respectively. There was a similar safety profile between the two higher doses, with an irAE rate of 70% with 10 mg/kg and 65% with 3 mg/kg [53]. Overall, a series of phase II trials evaluated the efficacy and safety of ipilimumab in over 500 patients [54]. The results were of course very promising, but there were two key lessons derived from the findings of these trials. Firstly, irAEs were the most common toxicity associated with ipilimumab; however, they were in most cases reversible [54]. Secondly, these trials revealed the unique response pattern of ipilimumab, in which some patients showed response following apparent disease progression. In fact, objective responses were sometimes observed 6–12 months after treatment initiation [54]. Therefore, it was recognised that overall survival (OS) was the superior endpoint to reflect the clinical activity of ipilimumab, and as such, it was chosen as the primary endpoint for future phase III trials [54].
Phase III trials were the last step in the journey towards FDA approval for ipilimumab. A key phase III trial, MDX010-20, evaluated ipilimumab treatment at 3 mg/kg in previously treated patients, comparing it against the cancer vaccine gp100 alone, and in combination [13]. A significantly improved OS was observed in patients treated with ipilimumab, with or without gp100, compared to patients receiving gp100 alone. There was almost double the patients in the 1- and 2-year survival rates with ipilimumab compared to gp100. As well as this, clinical outcomes were independent of important prognostic factors of melanoma, such as stage at presentation, baseline lactate dehydrogenase levels and age. Once again, there was a high frequency of irAEs, of approximately 60%, but they were low grade and manageable in most cases [13]. The results of this study were the basis for approval of ipilimumab at 3 mg/kg dose for previously treated metastatic melanoma and, in some countries, for treatment-naïve metastatic melanoma [49]. A recent meta-analysis of 1861 patients across 12 clinical trials of ipilimumab in melanoma showed a favourable median OS of 11.4 months and 3-year survival of 22% across all patients [55]. It also showed that ipilimumab was more effective in improving OS and other clinical outcomes in combination with PD-1 inhibitors, such as nivolumab. This synergistic relationship between these two ICI has therefore been utilised and approved for treatment of melanoma and many other cancers (Table 1) [55]. In summary, ipilimumab was the first instance of success for ICI therapy as a whole, paving the way for further research, development and clinical application of immune checkpoint blockade.

4. PD-1/PD-L1 Physiological Role

PD-1 is a transmembrane protein which, much like CTLA-4, has an important role in co-inhibitory regulation of TCR activation [56]. Although both CTLA-4 and PD-1 have similar inhibitory effects on T-cell activity, the responsible signalling mechanisms, timing of downregulation and anatomic location of immune inhibition differ significantly [24]. Unlike CTLA-4 expression, which is confined to T cells, PD-1 is expressed more broadly in activated T cells, B cells and myeloid cells. Furthermore, while CTLA-4 primarily functions during the priming phase of T-cell activation within secondary lymphoid organs, PD-1 functions during the effector phase and therefore within peripheral tissue [57]. PD-1 has two ligands to which it binds to mediate its effects, PD-L1 and PD-L2, the expression of which also differs in comparison to CTLA-4 ligands [21]. Whereas B7 ligands are expressed on APCs, the PD-1 ligands are expressed widely on leukocytes, non-hematopoietic cells and non-lymphoid tissues [57].
When PD-1 binds to its ligands in the presence of TCR or B-cell receptor activation, it transduces a direct inhibitory signal to the activating receptor (Figure 3). This co-inhibitory signal prevents phosphorylation of key signalling intermediates, inhibiting activation of T cells or B cells [58]. Signalling through PD-1 also heavily influences cytokine production by T cells, inhibiting the release of pro-inflammatory cytokines such as Interferon-gamma (INF-γ), tumour necrosis factor-alpha (TNF-α) and interleukin-2 (IL-2), which ordinarily act to promote T-cell activity and proliferation [59]. Additionally, PD-1 signalling also inhibits the upregulation of Bcl-xl, an anti-apoptotic protein, and limits the expression of transcriptional factors which are important for T-cell effector function [59]. Like CTLA-4, the net effect of PD-1 co-inhibitory signalling is dependent on the extent of the opposing co-stimulatory signalling of CD28 and other stimulatory signals. These immune checkpoints all act together to form a homeostatic feedback loop, which closely regulates the degree of T-cell activation and activity [24]. Considering the widespread distribution of PD-1 and its ligands, alongside its role in inhibitory regulation, it follows that the inherent role of PD-1 is to maintain peripheral tolerance of effector lymphocytes [21]. Experimental evidence supporting this concept has been seen in mice studies, in which genetic deletion of the Pdcd1 gene (encoding for PD-1) led to various autoimmune pathologies [59,60].

4.1. Mechanism of PD-1/PD-L1 Blockade-Induced Tumour Rejection

PD-L1 expression has been detected in many different cancer cell types. They are able to utilise the PD-1 signalling pathway to evade the immune system, taking advantage of its immunosuppressive function to grow and progress [21,61]. As a result, several different ICIs have been developed targeting the PD-1 signalling pathways. Currently, pembrolizumab, nivolumab and cemiplimab are the approved anti-PD-1 ICIs, and atezolizumab, avelumab and durvalumab are approved anti-PD-L1 ICIs. They are collectively able to target 18 different cancers (Table 2), which highlights the significance of PD-1/PD-L1 signalling in cancer progression and the great deal of therapeutic benefit that can be derived from the blockade of these pathways [62]. PD-1 blockade acts to induce tumour rejection through reinvigoration of exhausted T cells and overall enhancement of their effector function through blockade of the many inhibitory mechanisms of PD-1 mentioned before [59]. PD-L1 blockade is thought to induce tumour rejection in a largely similar manner to anti-PD1 antibodies, due to the dominance in the expression of PD-L1 compared to PD-L2 [24]. Also, considering that PD-L1 is induced predominantly by Th1 cytokines whereas PD-L2 is induced by Th2 cytokines, PD-L1 blockade would produce a Th-1 skewed response that would favour anti-tumour immunity [63]. Furthermore, selective inhibition of PD-L1 blocks PD-1/PD-L1 interactions while preserving PD-1/PD-L2 interactions. This, in theory, could produce a more targeted response with less unwanted toxicity, as there will be partial preservation of PD-1 self-tolerance mechanisms, via PD-L2 binding. However, as of yet, there are limited clinical data to prove this concept [24]. Also, PD-L1 is known to bind to B7-1, as well as PD-1, to induce inhibitory signals to T cells. Therefore PD-L1 blockade could also stimulate T-cell activation in a very similar manner to CTLA-4 blockade, increasing availability of B7-1 ligand for CD28 co-stimulation [64]. Ultimately, more research is needed to better understand the potential multifaceted mechanisms of both anti-PD-1 and anti-PD-L1 therapies, in order to optimize their use in the many different targeted cancers.

4.2. Efficacy of Anti-PD-1/PD-L1 Agents

4.2.1. Pembrolizumab

Of the ICIs that target the PD-1/PD-L1 signalling pathway, the PD-1 ICI pembrolizumab and the PD-L1 ICI atezolizumab are the most established, as such they are the best ICIs to assess in order to understand the efficacy of PD-1/PD-L1 therapies as a whole [62]. Pembrolizumab is a fully humanised mAb that received accelerated FDA approval in 2014 for treatment of unresectable or metastatic melanoma, and shortly after for non-small-cell lung cancer (NSCLC) in 2015 [91]. KEYNOTE-001, a phase I dose-finding trial of melanoma patients previously treated with ipilimumab, assessed pembrolizumab given intravenously at multiple doses ranging from 1 to 10 mg/kg, either every 2 or 3 weeks [92]. The overall response rate (ORR) was highest in the dose-dense and dose-intense groups at the expense of higher irAE incidence [92]. Consequent expansion cohorts of this study explored the safety and efficacy of many pembrolizumab regimes in patients with advanced melanoma and NSCLC [91]. The KEYNOTE-001 data showed significant treatment activity in patients with advanced melanoma, regardless of previous ipilimumab treatment. However, notably higher response rates were seen in ipilimumab-naïve patients [91]. A recent 5-year follow-up of this patient cohort in KEYNOTE-001 showed favourable 5-year OS rates, further proving the durable anti-tumour activity and tolerability of pembrolizumab in advanced melanoma [65]. A phase 3 trial, KEYNOTE-006, compared the efficacy of pembrolizumab against ipilimumab and 5-year follow-up results also recently became available. After a median follow-up of 57.7 months, the median OS was 32.7 months in the pembrolizumab group compared to 15.9 months in the ipilimumab group. There was also a lower incidence of high-grade treatment-related adverse effects seen with pembrolizumab. These results extended the finding in the original trial, showing the superiority of pembrolizumab over ipilimumab in patients with advanced melanoma [93].
Pembrolizumab also showed impressive outcomes in 5-year follow-up of the NSCLC expansion cohort of the KEYNOTE-001 trial. Based on these results estimated OS rates was 23.2% for treatment-naïve patients and 15.5% for previously treated patients, which are very favourable when compared to pre-immunotherapy OS rates of around 5%. There was also a very low incidence of treatment-related AEs [94]. A phase 2/3 trial, KEYNOTE-010, also showed improved OS with ipilimumab when compared to docetaxel in previously treated patients with advanced NSCLC, with long-term follow-up data from this same trial extending this positive trend [66]. Overall, through many trials, pembrolizumab has demonstrated durable responses and prolonged OS in NSCLC, especially in variants with high expression of PD-L1. Thus, this has been recognized as an important predictive biomarker of the efficacy of pembrolizumab in this patient population [91]. Beyond melanoma and NSLC, pembrolizumab has been approved and shown success in the treatment of many cancers including head and neck cancer, gastric cancer, Hodgkin lymphoma, urothelial cancer and many more. Table 2 highlights the wide-reaching therapeutic potential of pembrolizumab and PD-1 ICIs as a whole [91].

4.2.2. Atezolizumab

Atezolizumab was the first anti-PD-L1 ICI of its kind, receiving FDA approval in 2016 for treatment of locally advanced or metastatic urothelial carcinoma (mUC) resistant to platinum-based chemotherapy. The following year, it also received accelerated approval as an initial treatment in patients who were ineligible for cisplatin chemotherapy [95]. In both cases, approval was primarily based on data from the IMvigor210 study, a phase II trial which evaluated the safety and efficacy of atezolizumab in patients with locally advanced or mUC, regardless of PD-L1 expression. Patients were split into 2 cohorts, cohort 1 comprised of treatment-naïve patients and cohort 2 comprised of previously treated patients. The median follow-up for cohort 1 was 29 months, and 33 months for cohort 2. In cohort 1, the ORR was 24%, and in cohort 2 the ORR 16%, which were both very favourable when compared to the historical control ORR of 10%. Additionally, in both cohorts, a higher PDL-1 expression of tumour-infiltrating lymphocytes (>5%) was associated with improved ORR. The incidence of irAEs in cohort 1 was 14% and in cohort 2 10%, with the incidence of high-grade irAEs being 7% and 6%, respectively [96,97].
In 2016, atezolizumab also received accelerated approval for treatment of cisplatin-resistant metastatic NSCLC, specifically associated with EGFR or ALK gene abnormalities [98]. This approval is based on results from the Phase III OAK and Phase II POPLAR studies [99,100]. The largest study, OAK, evaluated the efficacy and safety of atezolizumab compared with docetaxel in 1225 patients. The results showed that those treated with atezolizumab had a median OS of 13.8 months, 4.2 months longer than those treated with docetaxel chemotherapy [100]. Since then, it has also been approved as an initial treatment in combination with chemotherapy for metastatic NSCLC and extensive-stage small-cell lung cancer [101].
Additionally, in 2019, atezolizumab received accelerated approval for the treatment of PD-L1-positive, metastatic triple-negative breast cancer (TNBC), in combination with chemotherapy [102]. TNBC is an aggressive variant of breast cancer with few treatment options, in which cancer cells lack expression of hormone receptors and do not overexpress the HER2 protein [103]. The accelerated approval was based on data from the Phase III IMpassion130 study, which evaluated the efficacy and safety of atezolizumab plus nab-paclitaxel compared with placebo plus nab-paclitaxel in unresectable locally advanced or metastatic TNBC patients [103]. The study demonstrated that the combination therapy significantly reduced the risk of cancer progression or death by 40% compared with nab-paclitaxel alone specifically in patients who had PD-L1-positive disease and had not received any prior chemotherapy. The combination therapy was also shown to be safe and tolerable, consistent with the known individual safety profiles of each medication and with no new safety concerns identified [103]. Overall, much like PD-1 ICIs, atezolizumab and other PD-L1 inhibitors have proven to be a versatile and effective therapy for many end-stage cancers, as outlined in Table 3 [104].

5. LAG-3 Physiological Role

LAG-3 is a co-inhibitory transmembrane protein expressed on a range of effector leukocytes, including CD4+ and CD8+ T cells, natural killer (NK) cells, dendritic cells and T-reg cells [118]. Structurally similar to CD4, LAG-3 shares its primary ligand, major histocompatibility complex class II (MHC-II), with a binding affinity approximately 100 times that of CD4 [119]. LAG-3 also interacts with other ligands to a lesser extent, such as fibrinogen-like protein 1 (FGL-1), a soluble protein primarily secreted by hepatocytes, and galectin-3, an inflammatory lectin secreted by activated macrophages [120,121]. The binding of LAG-3 to its ligands induces co-inhibitory signalling, inhibiting TCR activation, signalling, proliferation and inflammatory cytokine production. As a result, LAG-3 contributes to immune regulation and tolerance mechanisms, much like CTLA-4 and PD-1 [122]. Studies have shown that LAG-3 signalling is crucial for CD4+ T-reg suppression of autoimmune responses, emphasizing its role in maintaining immune homeostasis [123]. Furthermore, LAG-3 has been linked to functional exhaustion in T cells, particularly in chronic infections like HIV, where upregulation of LAG-3 correlates with disease progression and impaired T-cell function [124]. In the context of the TME, chronic exposure to TAAs can lead to T-cell exhaustion, characterized by the upregulation of multiple co-inhibitory receptors, including LAG-3 [125].
The exact mechanism of LAG-3 inhibitory signalling is still not completely well understood. Some studies have suggested that its inhibitory function is through competitive inhibition of CD4 due to its structural homology and much stronger binding affinity [126,127]. However, little evidence supports this as the major mechanism of LAG-3 co-inhibitory effects, in fact, there is strong evidence to suggest the contrary [128]. Many studies have shown that LAG-3 inhibitory function relies mainly on the intracellular domain, implying the role of intracellular signalling rather than receptor-competitive inhibition [126,128,129]. The presence of its other ligands also suggests LAG-3’s functional independence from CD4, as these alternative ligands do not bind to CD4 [120,121]. Once it binds to its ligand, the exact mechanism by which LAG-3 communicates an inhibitory signal to suppress TCR activation remains largely unknown [128]. As such, the intricacies of the LAG-3 signalling pathway still require further research, a better understanding could guide immunotherapies targeting this immune checkpoint.
A key point of research interest has been the synergistic relationship between LAG-3 and PD-1/PD-L1 signalling. A study by Huaeng et al. suggested that they may share intracellular localisation and trafficking pathways, with LAG-3 potentially facilitating the translocation of PD-1 to the immunological synapse [130]. Both signalling pathways also involve the recruitment of Src homology region 2 domain-containing phosphatase-1 (SHP-1) and SHP-2, which dephosphorylate key signalling molecules downstream of the TCR, inhibiting T-cell activation [130]. Preclinical studies have also shown compensatory upregulation of LAG-3 and other immune checkpoints in response to PD-1/PD-L1 blockade [131,132]. Co-expression of LAG-3 and PD-1 has been observed in tumour-infiltrating lymphocytes, with this co-expression having been associated with worse outcomes in various cancer types such as colorectal, breast, renal cell carcinoma, NSCLC and many more [133,134,135,136,137]. This was a key initial driver for research into combination therapies targeting these two immune checkpoints.

5.1. Mechanism of LAG-3 Blockade-Induced Tumour Rejection

Cancer cells can exploit LAG-3 signalling pathways to dampen the effector T-cell-mediated anti-tumour immunity and promote an immunosuppressive TME. It had established itself as a key player in cancer immune evasion, with its expression being upregulated and associated with worse outcomes in many tumour types [137]. Pre-clinical data showed its close synergy with PD-1 signalling, with evidence of these two immune checkpoints acting via distinct, non-redundant pathways to regulate T-cell function and immune tolerance mechanisms [138]. The next step was to explore if the concomitant blockade of these checkpoints could further impede cancer immune evasion and add to the arsenal of therapies against advanced cancers. In March 2022, the FDA approved the first anti-LAG-3 agent, relatlimab, in combination with nivolumab, for the treatment of unresectable or metastatic melanoma, representing another significant advancement in cancer immunotherapy [10]. Relatlimab is a human IgG4 mAb which acts to block LAG-3 binding to its ligands, primarily LAG-3:MHC-II interaction (Figure 4) [139]. Blockade of LAG-3 pathways acts to remove its co-inhibitory influence and restore unopposed TCR activation, proliferation and inflammatory cytokine production [139]. This reverses T-cell exhaustion and restores the functional capacity of T cells to respond to TAAs and exert cytotoxic effects on cancer cells [125]. Additionally, the LAG-3-blockade-induced release of inflammatory cytokines (e.g., IL-2, IFN-γ and TNF-α) by effector T cells modulates the TME, which is often rich in immunosuppressive cytokines, resulting in an immune-inflamed TME conducive with anti-tumour immunity [140]. Finally, much like CTLA-4, LAG-3 is often highly expressed on T-regs, enhancing their suppressive functions within the TME [141]. As such, LAG-3 inhibition reduces T-reg-mediated suppression of effector T cells. When combined with PD-1 inhibitors like nivolumab, this combination therapy provides a dual blockade of inhibitory pathways, leading to a more substantial enhancement of effector T-cell activity and anti-tumour immunity [142].

5.2. Efficacy of Relatimab Plus Nivolumab in the Treatment of Advanced Melanoma

RELATIVITY-020 was one of the key initial clinical trials that demonstrated the efficacy of relatlimab and nivolumab combination therapy [143]. It was a phase I/IIA, open-label trial that investigated the combination of relatlimab and nivolumab in 518 patients with advanced melanoma who had progressed after prior anti-PD-1/PD-L1 therapy. The trial included two cohorts: D1 cohort was comprised of 354 patients who had been treated with one prior anti-PD-1/PD-L1 regimen, and D2 cohort with 164 patients with multiple such regimens. The primary endpoint was the safety of the combination therapy. ORR and progression-free survival (PFS) were also evaluated by blinded independent central review (BICR). The ORR was 12.0% for the D1 cohort and 9.2% for the D2 cohort. Median PFS was 2.1 months in D1 and 3.2 months in D2. The 6-month PFS rates were 29.1% and 27.7%, respectively, and 12-month PFS rates were 21.4% and 16.0%. The combination therapy showed a manageable safety profile with grade 3–4 treatment-related adverse events occurring in 15.0% of the D1 cohort and 12.8% of the D2 cohort. These results were very promising, showing that this combination could provide clinical benefits in heavily pretreated advanced melanoma patients, demonstrating durable responses and a tolerable safety profile [143].
The trial that directly led to FDA approval of relatlimab was the RELATIVITY-047 trial, a phase II/III, double-blind, randomized control trial including 714 patients with previously untreated metastatic or unresectable melanoma [144]. Patients were randomised to receive either relatlimab with nivolumab (n = 355) or nivolumab monotherapy (n = 359), with efficacy and safety profile comparison of the treatment regimes. The trial met its primary endpoints with the combination group demonstrating a far superior median PFS of 10.1 months compared to a median PFS of 4.6 in the nivolumab monotherapy group. Hazard ratio (HR) was 0.75, indicating a 25% reduction in the risk of disease progression or death with the combination therapy compared to nivolumab alone. Additionally, ORR in the combination group was 43.1% versus 32.6% in the monotherapy group. The incidence of irAEs of grade 3 or 4 was higher in the combination therapy group compared to the monotherapy group (18.9% vs. 9.7%); however, the combination was generally well tolerated. These significant findings and manageable safety profile were enough to seal FDA approval of relatlimab for the treatment of adult and paediatric patients (12 years of age or older) with unresectable or metastatic melanoma [10].

6. Adverse Effects of Immune Checkpoint Inhibitors

Immune checkpoints have an important role in maintaining immune homeostasis and preventing autoimmunity. Therefore, although the blockade of its immunosuppressive pathways by ICIs may revive anti-tumour immunity, it can lead to potentially severe and even lethal autoimmune pathologies known as immune-related adverse events (irAEs) [145]. IrAEs are clinically diverse with most organs being potentially affected such as the skin, liver, lung, pituitary, thyroid, gastrointestinal tract and even the central nervous system (Figure 5) [145]. These toxicities are often unpredictable and highly variable; however, most cases are mild and easily managed [146]. Dermatological reactions are the most common irAEs associated with ICIs, such as rash, pruritis and vitiligo, occurring in more than 40% of patients with ipilimumab and in around 20% of those with PD-1/PD-L1 inhibitors [146]. Gastrointestinal irAEs are also frequently seen, commonly presenting as diarrhoea and colitis, once again incidence of which is reported to be higher in anti-CTLA4 therapies [146]. In general, the incidence of irAEs is higher in anti-CTLA4 therapies compared to anti-PD1/PDL1 therapies, including high-grade cases [24]. For example, in a phase III trial of melanoma patients, the overall rate of grade ≥3 irAEs was higher in ipilimumab compared with pembrolizumab (20% vs. 13%) [147]. In general, the management of mild-to-moderate cases is with supportive care and conservative measures; however, in severe cases, corticosteroid, biological therapy or even treatment cessation may be indicated [146].
Interestingly, although irAEs are unwanted side effects and can have potentially serious implications, in some cases their presence, and in particular their onset, can be a positive prognostic marker of ICI efficacy [148]. As a predictive marker of treatment response, irAE onset has been more strongly associated with anti-PD-1/PD-L1 therapies compared to anti-CTLA4 therapies, a link demonstrated by a growing body of research [148]. Other characteristics of irAEs such as site, severity and timing of onset have also been considered as potential clinical biomarkers; however, current research is limited, and further studies are needed to understand the true implications of these parameters on ICI efficacy [148]. Overall, there is a close link between autoimmunity and the anti-tumour effect elicited by ICIs; therefore, it is an important area of research interest to determine whether these two aspects of ICI therapy can be uncoupled to maximize benefit while minimizing toxicities for patients.

7. Predictive Biomarkers

Although ICIs have transformed the treatment landscape for many patients with advanced cancers, response rates range from 15 to 60%, leaving a significant proportion of patients who do not derive any benefit [148]. Therefore, identifying predictive biomarkers is critical to delineate responders from non-responders; prevent unnecessary treatment and adverse effects of therapy; and improve the cost-effectiveness of this often expensive therapy by only applying it to those most likely to benefit [5,149]. Key biomarkers that have been identified include PDL-1 overexpression, immune cell constitution of TME, neoantigens, genetic and epigenetic signatures of a tumour, and the gut microbiome [150].
PD-L1 over-expression has proven to be a very accurate predictive biomarker of anti-PD-1/PD-L1 ICI efficacy, so much so that it is a vital criterion for treatment of cancers such as urothelial cancer and NSCLC with such therapies [151]. The efficacy of ICI therapy relies on the underlying presence of anti-tumour immunity; thus, the immune constitution within the TME is another important prognostic biomarker. For example, in responders, the TME is associated with high levels of tumour-infiltrating lymphocytes, a high T-effector/T-reg ratio and increased secretion of inflammatory cytokines within the TME [5]. The TME is classified into three major types based on the degree of immune cell infiltration: immune desert, immune-excluded and immune-inflamed. Immune-inflamed phenotype is characterised by infiltration of multiple immune cell subtypes and is associated with the best prognosis. Immune-excluded phenotype exhibits the presence of many inflammatory cells and mediators; however, there is a lack of TME infiltration of accumulated T cells. Immune desert phenotype is characterised by the absence of T cells in the TME and unsurprisingly does not respond well to ICI therapy [15]. The degree of anti-tumour immune response is also determined by the immunogenicity of a cancer, so biomarkers such as tumour mutational burden (TMB) and the presence of neoantigens are also important predictors of response [152]. Furthermore, certain genetic signatures of tumours can also confer a better prognosis with ICI therapy. For example, microsatellite instability (MSI)-positive colorectal cancers show a better prognosis following ICI as they are associated with greater degree of CD8+ T-cell infiltration as well as high TMB and neoantigen expression [153,154]. Another example of an epigenetic biomarker is DNA methylation of certain genes, for example, a study showed methylation status of a single gene, FOXP3, which regulates T-reg cell function, was also found to be predictive of ICI response in NSCLC patients [155]. Finally, research has also found the gut microbiome to be a potential predictive biomarker for ICI efficacy. Research has established that a diverse and healthy microbiome has profound positive immunomodulatory effects. As such, it makes sense that this would extend to anti-tumour immunity [156,157]. For example, Gopalakrishnan et al. analysed the gut microbiomes of melanoma patients undergoing anti-PD-1 therapy and found that responders had a higher diversity of gut bacteria and distinct microbial compositions compared to non-responders [158]. Specifically, the presence of certain bacterial species such as Faecalibacterium and Clostridiales was associated with a favourable response to treatment [158].
There are many more biomarkers that have been identified but as of yet there is no single predictive model for ICI efficacy [159]. There is a great degree of heterogeneity seen in each clinical case as immune responses are not uniform across all malignancies and individuals [160]. Therefore, in reality, a case-by-case approach is needed in which multiple potential biomarkers are utilised to best predict the efficacy of each ICI therapy [159]. Ultimately, continued research into this area will help identify and guide clinical application of predictive biomarkers, in hope of extending the reach of ICI therapy and maximising patient benefit.

8. Future Directions

ICIs have proven to be a major breakthrough in the field of cancer immunotherapy, revealing a promising avenue for treatment of a high-risk and diverse patient population. Despite the ICIs to date showing compelling clinical effectiveness in certain tumour types, the overall efficiency of ICI therapy remains unsatisfactory [23]. Therefore, going forward, research goals are to optimise the current ICI therapies as well as explore additional immune checkpoints in hope of creating a more effective, safer and wider-reaching therapy.
Beyond CTLA-4, PD-1 and LAG-3, other novel immune checkpoints have been discovered, and many ICIs are currently under development or clinical trial [23]. An example of a promising ICI target is TIM-3, a co-inhibitory receptor expressed on a variety of immune cells including Th1 cells, T-reg cells, and certain dendritic cells [161]. Upon binding to its ligands, such as galectin-9, it can lead to the inhibition of Th1-driven immune responses and promote immune tolerance [162]. Currently, there are over 15 drugs targeting TIM-3 in various stages of clinical trials, ranging from phase I to phase III [161]. Among the most advanced is sabatolimab (MBG453), an anti-TIM-3 humanised mAb, which is a promising treatment option for advanced haematological malignancies [163]. In a phase Ib study, it was shown to have favourable outcomes in combination with a hypomethylating agent in patients with high or very high-risk myelodysplastic syndromes (MDS) and newly diagnosed acute myeloid leukaemia (AML) [164]. This therapy has received fast-track designation by the FDA due to preliminary data, with the potential for an expedited review process due to the urgent need for new treatments in this area [165]. STIMULUS-MDS2, a phase III trial, is currently underway evaluating sabatolimab in combination with the chemotherapeutic agent azacitidine in patients with very high/high/intermediate-risk MDS [166].
TIGIT is another novel immune checkpoint, which inhibits TCR activation and NK cell cytotoxicity by interacting with its ligands, such as CD155 present on APCs and tumour cells [167]. A promising anti-TIGIT therapy is domvanalimab, an Fc-silent humanized IgG1 mAb, which has shown promising clinical activity when used in combination with the novel anti-PD-1 mAb zimberelimab in upper gastrointestinal (GI) cancers and NSCLC [168]. The EDGE-Gastric trial, a multi-arm, global, phase 2 trial, showed excellent outcomes of this ICI combination in patients with advanced metastatic upper GI cancer, with an ORR of 59% across all patients and notably higher ORR of 80% in patients with high PD-1 expression (<5%) [169]. The combination treatment was well tolerated, with a safety profile similar to that of anti-PD-1 therapy and chemotherapy used in this setting. This combination has also shown efficacy against NSCLC, as shown by the Phase 2 ARC-7 clinical trial targeting metastatic, PD-L1 highly expressed NSCLC [170]. Based on these positive results, the Phase 3 ARC-10 clinical trial is currently underway, comparing domvanalimab plus zimberelimab to chemotherapy as first-line treatment for PD-L1 highly expressed, locally advanced or metastatic NSCLC [171]. There is also more to understand when it comes to the biology and signalling mechanisms of many immune checkpoints, for example, the identification of ligands for the immune checkpoints VISTA and B7-H3, which would be the key to fully understanding their therapeutic potential [23].
While monotherapies have shown very promising results, more attempts have been made to design ICI-based combination therapies that target multiple non-redundant pathways to achieve synergistic anti-tumour effects [172]. The combination of ipilimumab and nivolumab was the first such combination therapy, utilising together the centrally acting CTLA-4 and peripherally acting PD-1 pathways [173]. This has shown greater efficacy in cancers such as melanoma, but also a higher incidence of adverse effects. Therefore, it seems that synergistic effects of combined immune checkpoint blockade also come at the cost of greater loss of intrinsic immune-tolerant mechanisms [173]. Additionally, many novel ICIs are also under trial in combination with PD-1 inhibitors as there is convincing rationale that other immune checkpoints are upregulated following PD-1/PD-L1 blockade [174]. There is also potential for combining ICIs with other forms of immunotherapy such as adoptive T-cell therapies or cellular vaccines, which could also act via complementary mechanisms to produce synergistic effects [172]. Finally, ICIs already have a basis for combination with conventional treatments such as chemotherapeutic drugs, but now combination with radiotherapy is also being considered [175]. While radiotherapy can be immunosuppressive, it can also enhance antigenicity by triggering the release of TAAs [172]. Preclinical data have also supported this theory with positive results seen in mouse models treated with radiotherapy and anti-CTLA4 therapy, and as such, there are several clinical trials exploring this potential combination [176].
Beyond the identification and development of new ICI-based regimes, in order to optimise its current and future clinical use, accurate biomarkers must be identified to predict both efficacy and adverse effects [159]. Subsequently, such adverse effects should be diagnosed and managed promptly and effectively [146]. Research into these areas will help formulate, design and improve upon the current management guidelines in place. Overall, there is so much left to learn and understand regarding immune checkpoints, and thus, continued preclinical and clinical research into all avenues is needed to guide further advancements.

9. Conclusions

ICI therapies have led to important clinical advancements and provided a new weapon against advanced cancers, giving hope to a patient population with a previously dismal prognosis. This therapy has elicited durable clinical responses with significant improvements in patient survival, showing its potential as a curative option. However, the therapy is still held back by limitations such as the small proportion of responders, toxicity caused by irAEs and significant resource implications. There are many avenues of research being undertaken in relation to ICIs with the aim of addressing such limitations and ultimately guiding the development of new and more effective agents.

Author Contributions

Writing—original draft preparation, A.Y.; supervision, writing—review and editing of manuscript, J.G. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mellman, I.; Coukos, G.; Dranoff, G. Cancer Immunotherapy Comes of Age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
  2. Borghaei, H.; Smith, M.R.; Campbell, K.S. Immunotherapy of Cancer. Eur. J. Pharmacol. 2009, 625, 41–54. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, H.; Chen, J. Current Status and Future Directions of Cancer Immunotherapy. J. Cancer 2018, 9, 1773–1781. [Google Scholar] [CrossRef] [PubMed]
  4. Voena, C.; Chiarle, R. Advances in Cancer Immunology and Cancer Immunotherapy. Discov. Med. 2016, 21, 125–133. [Google Scholar] [PubMed]
  5. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune Checkpoint Inhibitors: Recent Progress and Potential Biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed]
  6. Pardoll, D.M. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
  7. Kates, M.; Sopko, N.A.; Matsui, H.; Drake, C.G.; Hahn, N.M.; Bivalacqua, T.J. Immune Checkpoint Inhibitors: A New Frontier in Bladder Cancer. World J. Urol. 2016, 34, 49–55. [Google Scholar] [CrossRef] [PubMed]
  8. Camacho, L.H. CTLA-4 Blockade with Ipilimumab: Biology, Safety, Efficacy, and Future Considerations. Cancer Med. 2015, 4, 661–672. [Google Scholar] [CrossRef] [PubMed]
  9. Barbee, M.S.; Ogunniyi, A.; Horvat, T.Z.; Dang, T.O. Current Status and Future Directions of the Immune Checkpoint Inhibitors Ipilimumab, Pembrolizumab, and Nivolumab in Oncology. Ann. Pharmacother. 2015, 49, 907–937. [Google Scholar] [CrossRef]
  10. Paik, J. Nivolumab Plus Relatlimab: First Approval. Drugs 2022, 82, 925–931. [Google Scholar] [CrossRef]
  11. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; Van Der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; 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, 1909–1920. [Google Scholar] [CrossRef]
  12. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef] [PubMed]
  13. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  14. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, D.S.; Mellman, I. Elements of Cancer Immunity and the Cancer-Immune Set Point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  16. Blankenstein, T.; Coulie, P.G.; Gilboa, E.; Jaffee, E.M. The Determinants of Tumour Immunogenicity. Nat. Rev. Cancer 2012, 12, 307–313. [Google Scholar] [CrossRef]
  17. Fancello, L.; Gandini, S.; Pelicci, P.G.; Mazzarella, L. Tumor Mutational Burden Quantification from Targeted Gene Panels: Major Advancements and Challenges. J. Immunother. Cancer 2019, 7, 183. [Google Scholar] [CrossRef] [PubMed]
  18. Kourie, H.R.; Klastersky, J. Immune Checkpoint Inhibitors Side Effects and Management. Immunotherapy 2016, 8, 799–807. [Google Scholar] [CrossRef] [PubMed]
  19. O’Neill, R.E.; Cao, X. Co-Stimulatory and Co-Inhibitory Pathways in Cancer Immunotherapy. Adv. Cancer Res. 2019, 143, 145–194. [Google Scholar] [CrossRef]
  20. Nirschl, C.J.; Drake, C.G. Molecular Pathways: Coexpression of Immune Checkpoint Molecules: Signaling Pathways and Implications for Cancer Immunotherapy. Clin. Cancer Res. 2013, 19, 4917–4924. [Google Scholar] [CrossRef]
  21. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [PubMed]
  22. Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Nicholas Haining, W.; Freeman, G.J.; Sharpe, A.H. PD-L1 on Tumor Cells Is Sufficient for Immune Evasion in Immunogenic Tumors and Inhibits CD8 T Cell Cytotoxicity. J. Exp. Med. 2017, 214, 895–904. [Google Scholar] [CrossRef]
  23. Qin, S.; Xu, L.; Yi, M.; Yu, S.; Wu, K.; Luo, S. Novel Immune Checkpoint Targets: Moving beyond PD-1 and CTLA-4. Mol. Cancer 2019, 18, 1–14. [Google Scholar] [CrossRef]
  24. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. Cancer Clin. Trials 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed]
  25. Sansom, D.M. CD28, CTLA-4 and Their Ligands: Who Does What and to Whom? Immunology 2000, 101, 169–177. [Google Scholar] [CrossRef] [PubMed]
  26. Takahashi, T.; Tagami, T.; Yamazaki, S.; Uede, T.; Shimizu, J.; Sakaguchi, N.; Mak, T.W.; Sakaguchi, S. Immunologic Self-Tolerance Maintained by Cd25+Cd4+Regulatory T Cells Constitutively Expressing Cytotoxic T Lymphocyte–Associated Antigen 4. J. Exp. Med. 2000, 192, 303–310. [Google Scholar] [CrossRef] [PubMed]
  27. Qureshi, O.S.; Zheng, Y.; Nakamura, K.; Attridge, K.; Manzotti, C.; Schmidt, E.M.; Baker, J.; Jeffery, L.E.; Kaur, S.; Briggs, Z.; et al. Trans-Endocytosis of CD80 and CD86: A Molecular Basis for the Cell-Extrinsic Function of CTLA-4. Science 2011, 332, 600–603. [Google Scholar] [CrossRef] [PubMed]
  28. Fallarino, F.; Fields, P.E.; Gajewski, T.F. B7-1 Engagement of Cytotoxic T Lymphocyte Antigen 4 Inhibits T Cell Activation in the Absence of CD28. J. Exp. Med. 1998, 188, 205–210. [Google Scholar] [CrossRef] [PubMed]
  29. Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative Disorders with Early Lethality in Mice Deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
  30. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of Antitumor Immunity by CTLA-4 Blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef]
  31. Camacho, L.H.; Antonia, S.; Sosman, J.; Kirkwood, J.M.; Gajewski, T.F.; Redman, B.; Pavlov, D.; Bulanhagui, C.; Bozon, V.A.; Gomez-Navarro, J.; et al. Phase I/II Trial of Tremelimumab in Patients with Metastatic Melanoma. J. Clin. Oncol. 2009, 27, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
  32. Ribas, A.; Kefford, R.; Marshall, M.A.; Punt, C.J.A.; Haanen, J.B.; Marmol, M.; Garbe, C.; Gogas, H.; Schachter, J.; Linette, G.; et al. Phase III Randomized Clinical Trial Comparing Tremelimumab with Standard-of-Care Chemotherapy in Patients with Advanced Melanoma. J. Clin. Oncol. 2013, 31, 616–622. [Google Scholar] [CrossRef] [PubMed]
  33. Rotte, A. Combination of CTLA-4 and PD-1 Blockers for Treatment of Cancer. J. Exp. Clin. Cancer Res. 2019, 38, 255. [Google Scholar] [CrossRef]
  34. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  35. Overman, M.J.; McDermott, R.; Leach, J.L.; Lonardi, S.; Lenz, H.-J.; Morse, M.A.; Desai, J.; Hill, A.; Axelson, M.; Moss, R.A.; 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. Lancet Oncol. 2017, 18, 1182–1191. [Google Scholar] [CrossRef] [PubMed]
  36. Yau, T.; Kang, Y.-K.; Kim, T.-Y.; El-Khoueiry, A.B.; Santoro, A.; Sangro, B.; Melero, I.; Kudo, M.; Hou, M.-M.; Matilla, A.; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol. 2020, 6, e204564. [Google Scholar] [CrossRef]
  37. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef]
  38. Baas, P.; Scherpereel, A.; Nowak, A.K.; Fujimoto, N.; Peters, S.; Tsao, A.S.; Mansfield, A.S.; Popat, S.; Jahan, T.; Antonia, S.; et al. First-Line Nivolumab plus Ipilimumab in Unresectable Malignant Pleural Mesothelioma (CheckMate 743): A Multicentre, Randomised, Open-Label, Phase 3 Trial. Lancet 2021, 397, 375–386. [Google Scholar] [CrossRef]
  39. Chau, I.; Doki, Y.; Ajani, J.A.; Xu, J.; Wyrwicz, L.; Motoyama, S.; Ogata, T.; Kawakami, H.; Hsu, C.-H.; Adenis, A.; et al. Nivolumab (NIVO) plus Ipilimumab (IPI) or NIVO plus Chemotherapy (Chemo) versus Chemo as First-Line (1L) Treatment for Advanced Esophageal Squamous Cell Carcinoma (ESCC): First Results of the CheckMate 648 Study. J. Clin. Oncol. 2021, 39, LBA4001. [Google Scholar] [CrossRef]
  40. Wei, S.C.; Levine, J.H.; Cogdill, A.P.; Zhao, Y.; Anang, N.A.A.S.; Andrews, M.C.; Sharma, P.; Wang, J.; Wargo, J.A.; Pe’er, D.; et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 2017, 170, 1120–1133.e17. [Google Scholar] [CrossRef]
  41. Wherry, E.J. T Cell Exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  42. Simpson, T.R.; Li, F.; Montalvo-Ortiz, W.; Sepulveda, M.A.; Bergerhoff, K.; Arce, F.; Roddie, C.; Henry, J.Y.; Yagita, H.; Wolchok, J.D.; et al. Fc-Dependent Depletion of Tumor-Infiltrating Regulatory t Cells Co-Defines the Efficacy of Anti-CTLA-4 Therapy against Melanoma. J. Exp. Med. 2013, 210, 1695–1710. [Google Scholar] [CrossRef] [PubMed]
  43. Selby, M.J.; Engelhardt, J.J.; Quigley, M.; Henning, K.A.; Chen, T.; Srinivasan, M.; Korman, A.J. Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells. Cancer Immunol. Res. 2013, 1, 32–42. [Google Scholar] [CrossRef] [PubMed]
  44. Bulliard, Y.; Jolicoeur, R.; Windman, M.; Rue, S.M.; Ettenberg, S.; Knee, D.A.; Wilson, N.S.; Dranoff, G.; Brogdon, J.L. Activating Fc γ Receptors Contribute to the Antitumor Activities of Immunoregulatory Receptor-Targeting Antibodies. J. Exp. Med. 2013, 210, 1685–1693. [Google Scholar] [CrossRef] [PubMed]
  45. Du, X.; Tang, F.; Liu, M.; Su, J.; Zhang, Y.; Wu, W.; Devenport, M.; Lazarski, C.A.; Zhang, P.; Wang, X.; et al. A Reappraisal of CTLA-4 Checkpoint Blockade in Cancer Immunotherapy. Cell Res. 2018, 28, 416–432. [Google Scholar] [CrossRef] [PubMed]
  46. Peggs, K.S.; Quezada, S.A.; Chambers, C.A.; Korman, A.J.; Allison, J.P. Blockade of CTLA-4 on Both Effector and Regulatory T Cell Compartments Contributes to the Antitumor Activity of Anti-CTLA-4 Antibodies. J. Exp. Med. 2009, 206, 1717–1725. [Google Scholar] [CrossRef] [PubMed]
  47. Ramagopal, U.A.; Liu, W.; Garrett-Thomson, S.C.; Bonanno, J.B.; Yan, Q.; Srinivasan, M.; Wong, S.C.; Bell, A.; Mankikar, S.; Rangan, V.S.; et al. Structural Basis for Cancer Immunotherapy by the First-in-Class Checkpoint Inhibitor Ipilimumab. Proc. Natl. Acad. Sci. USA 2017, 114, E4223–E4232. [Google Scholar] [CrossRef] [PubMed]
  48. Altekruse, S.F.; Kosary, C.L.; Krapcho, M.; Neyman, N.; Aminou, R.; Waldron, W.; Ruhl, J.; Howlader, N.; Tatalovich, Z.; Cho, H.; et al. (Eds.) SEER Cancer Statistics Review, 1975–2007; Based on November 2009 SEER Data Submission, Posted to the SEER Web Site; National Cancer Institute: Bethesda, MD, USA, 2010. [Google Scholar]
  49. Tarhini, A.; Lo, E.; Minor, D.R. Releasing the Brake on the Immune System: Ipilimumab in Melanoma and Other Tumors. Cancer Biother. Radiopharm 2010, 25, 601–613. [Google Scholar] [CrossRef]
  50. Peggs, K.S.; Quezada, S.A.; Korman, A.J.; Allison, J.P. Principles and Use of Anti-CTLA4 Antibody in Human Cancer Immunotherapy. Curr. Opin. Immunol. 2006, 18, 206–213. [Google Scholar] [CrossRef]
  51. Hersh, E.M.; O’Day, S.J.; Powderly, J.; Khan, K.D.; Pavlick, A.C.; Cranmer, L.D.; Samlowski, W.E.; Nichol, G.M.; Yellin, M.J.; Weber, J.S. A Phase II Multicenter Study of Ipilimumab with or without Dacarbazine in Chemotherapy-Naïve Patients with Advanced Melanoma. Investig. New Drugs 2011, 29, 489–498. [Google Scholar] [CrossRef]
  52. Hersh, E.; Weber, J.; John, P., II; Pavlik, A.; Nichol, G.; Yellin, M.; Cranmer, L.; Urba, W.; O’Day, S. Long-term survival of patients (pts) with advanced melanoma treated with ipilimumab with or without dacarbazine. J. Clin. Oncol. 2009, 27 (Suppl. S15), 9038. [Google Scholar] [CrossRef]
  53. Wolchok, J.D.; Neyns, B.; Linette, G.; Negrier, S.; Lutzky, J.; Thomas, L.; Waterfield, W.; Schadendorf, D.; Smylie, M.; Guthrie, T.; et al. Ipilimumab Monotherapy in Patients with Pretreated Advanced Melanoma: A Randomised, Double-Blind, Multicentre, Phase 2, Dose-Ranging Study. Lancet Oncol. 2010, 11, 155–164. [Google Scholar] [CrossRef] [PubMed]
  54. Wolchok, J.D.; Hodi, F.S.; Weber, J.S.; Allison, J.P.; Urba, W.J.; Robert, C.; O’Day, S.J.; Hoos, A.; Humphrey, R.; Berman, D.M.; et al. Development of Ipilimumab: A Novel Immunotherapeutic Approach for the Treatment of Advanced Melanoma. Ann. N. Y. Acad. Sci. 2013, 1291, 1–13. [Google Scholar] [CrossRef]
  55. Schadendorf, D.; Hodi, F.S.; Robert, C.; Weber, J.S.; Margolin, K.; Hamid, O.; Patt, D.; Chen, T.-T.; Berman, D.M.; Wolchok, J.D. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J. Clin. Oncol. 2015, 33, 1889–1894. [Google Scholar] [CrossRef]
  56. Dong, Y.; Sun, Q.; Zhang, X. PD-1 and Its Ligands Are Important Immune Checkpoints in Cancer. Oncotarget 2017, 8, 2171–2186. [Google Scholar] [CrossRef]
  57. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef]
  58. Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed Cell Death 1 Forms Negative Costimulatory Microclusters That Directly Inhibit T Cell Receptor Signaling by Recruiting Phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217. [Google Scholar] [CrossRef]
  59. Riella, L.V.; Paterson, A.M.; Sharpe, A.H.; Chandraker, A. Role of the PD-1 Pathway in the Immune Response. Am. J. Transplant. 2012, 12, 2575–2587. [Google Scholar] [CrossRef] [PubMed]
  60. 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, 141–151. [Google Scholar] [CrossRef]
  61. Xiang, X.; Yu, P.C.; Long, D.; Liao, X.L.; Zhang, S.; You, X.M.; Zhong, J.H.; Li, L.Q. Prognostic Value of PD -L1 Expression in Patients with Primary Solid Tumors. Oncotarget 2018, 9, 5058–5072. [Google Scholar] [CrossRef]
  62. PD-1/PD-L1 Landscape Analysis—Cancer Research Institute (CRI). Available online: https://www.cancerresearch.org/scientists/immuno-oncology-landscape/pd-1-pd-l1-landscape (accessed on 23 March 2020).
  63. Loke, P.; Allison, J.P. PD-L1 and PD-L2 Are Differentially Regulated by Th1 and Th2 Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 5336–5341. [Google Scholar] [CrossRef]
  64. Butte, M.J.; Keir, M.E.; Phamduy, T.B.; Sharpe, A.H.; Freeman, G.J.; Sharpe, H. PD-L1 Interacts Specifically with B7-1 to Inhibit T Cell Proliferation. Immunity 2009, 27, 111–122. [Google Scholar] [CrossRef]
  65. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-Year Survival Outcomes for Patients with Advanced Melanoma Treated with Pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef]
  66. Herbst, R.S.; Garon, E.B.; Kim, D.-W.; Cho, B.C.; Perez-Gracia, J.L.; Han, J.-Y.; Arvis, C.D.; Majem, M.; Forster, M.D.; Monnet, I.; et al. Long-Term Outcomes and Retreatment Among Patients With Previously Treated, Programmed Death-Ligand 1–Positive, Advanced Non–Small-Cell Lung Cancer in the KEYNOTE-010 Study. J. Clin. Oncol. 2020, 38, 1580–1590. [Google Scholar] [CrossRef] [PubMed]
  67. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and Clinical Activity of Pembrolizumab for Treatment of Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-012): An Open-Label, Multicentre, Phase 1b Trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
  68. Chen, R.; Zinzani, P.L.; Fanale, M.A.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 2125–2132. [Google Scholar] [CrossRef] [PubMed]
  69. Balar, A.V.; Castellano, D.; O’Donnell, P.H.; Grivas, P.; Vuky, J.; Powles, T.; Plimack, E.R.; Hahn, N.M.; de Wit, R.; Pang, L.; et al. First-Line Pembrolizumab in Cisplatin-Ineligible Patients with Locally Advanced and Unresectable or Metastatic Urothelial Cancer (KEYNOTE-052): A Multicentre, Single-Arm, Phase 2 Study. Lancet Oncol. 2017, 18, 1483–1492. [Google Scholar] [CrossRef] [PubMed]
  70. Diaz, L.; Marabelle, A.; Kim, T.W.; Geva, R.; Cutsem, E.V.; André, T.; Ascierto, P.A.; Maio, M.; Delord, J.-P.; Gottfried, M.; et al. Efficacy of Pembrolizumab in Phase 2 KEYNOTE-164 and KEYNOTE-158 Studies of Microsatellite Instability High Cancers. Ann. Oncol. 2017, 28, v128–v129. [Google Scholar] [CrossRef]
  71. Bang, Y.-J.; Kang, Y.-K.; Catenacci, D.V.; Muro, K.; Fuchs, C.S.; Geva, R.; Hara, H.; Golan, T.; Garrido, M.; Jalal, S.I.; et al. Pembrolizumab Alone or in Combination with Chemotherapy as First-Line Therapy for Patients with Advanced Gastric or Gastroesophageal Junction Adenocarcinoma: Results from the Phase II Nonrandomized KEYNOTE-059 Study. Gastric Cancer 2019, 22, 828–837. [Google Scholar] [CrossRef]
  72. Chung, H.C.; Ros, W.; Delord, J.-P.; Perets, R.; Italiano, A.; Shapira-Frommer, R.; Manzuk, L.; Piha-Paul, S.A.; Xu, L.; Zeigenfuss, S.; et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Cervical Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2019, 37, 1470–1478. [Google Scholar] [CrossRef]
  73. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in Patients with Advanced Hepatocellular Carcinoma Previously Treated with Sorafenib (KEYNOTE-224): A Non-Randomised, Open-Label Phase 2 Trial. Lancet Oncol. 2018, 19, 940–952. [Google Scholar] [CrossRef] [PubMed]
  74. Nghiem, P.; Bhatia, S.; Lipson, E.J.; Sharfman, W.H.; Kudchadkar, R.R.; Brohl, A.S.; Friedlander, P.A.; Daud, A.; Kluger, H.M.; Reddy, S.A.; et al. Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J. Clin. Oncol. 2019, 37, 693–702. [Google Scholar] [CrossRef] [PubMed]
  75. Rini, B.I.; Plimack, E.R.; Stus, V.; Gafanov, R.; Hawkins, R.; Nosov, D.; Pouliot, F.; Alekseev, B.; Soulières, D.; Melichar, B.; et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1116–1127. [Google Scholar] [CrossRef] [PubMed]
  76. Kojima, T.; Shah, M.A.; Muro, K.; Francois, E.; Adenis, A.; Hsu, C.-H.; Doi, T.; Moriwaki, T.; Kim, S.-B.; Lee, S.-H.; et al. Randomized Phase III KEYNOTE-181 Study of Pembrolizumab Versus Chemotherapy in Advanced Esophageal Cancer. J. Clin. Oncol. 2020, 38, 4138–4148. [Google Scholar] [CrossRef] [PubMed]
  77. Cortes, J.; Cescon, D.W.; Rugo, H.S.; Nowecki, Z.; Im, S.-A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Holgado, E.; et al. KEYNOTE-355: Randomized, Double-Blind, Phase III Study of Pembrolizumab + Chemotherapy versus Placebo + Chemotherapy for Previously Untreated Locally Recurrent Inoperable or Metastatic Triple-Negative Breast Cancer. J. Clin. Oncol. 2020, 38, 1000. [Google Scholar] [CrossRef]
  78. Grob, J.-J.; Gonzalez, R.; Basset-Seguin, N.; Vornicova, O.; Schachter, J.; Joshi, A.; Meyer, N.; Grange, F.; Piulats, J.M.; Bauman, J.R.; et al. Pembrolizumab Monotherapy for Recurrent or Metastatic Cutaneous Squamous Cell Carcinoma: A Single-Arm Phase II Trial (KEYNOTE-629). J. Clin. Oncol. 2020, 38, 2916–2925. [Google Scholar] [CrossRef]
  79. O’Malley, D.M.; Bariani, G.M.; Cassier, P.A.; Marabelle, A.; Hansen, A.R.; De Jesus Acosta, A.; Miller, W.H.; Safra, T.; Italiano, A.; Mileshkin, L.; et al. Pembrolizumab in Patients With Microsatellite Instability–High Advanced Endometrial Cancer: Results From the KEYNOTE-158 Study. J. Clin. Oncol. 2022, 40, 752–761. [Google Scholar] [CrossRef] [PubMed]
  80. Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D.; et al. Nivolumab versus Chemotherapy in Patients with Advanced Melanoma Who Progressed after Anti-CTLA-4 Treatment (CheckMate 037): A Randomised, Controlled, Open-Label, Phase 3 Trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef] [PubMed]
  81. Weber, J.; Mandala, M.; Del Vecchio, M.; Gogas, H.J.; Arance, A.M.; Cowey, C.L.; Dalle, S.; Schenker, M.; Chiarion-Sileni, V.; Marquez-Rodas, I.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N. Engl. J. Med. 2017, 377, 1824–1835. [Google Scholar] [CrossRef]
  82. Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Procopio, G.; Plimack, E.R.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1803–1813. [Google Scholar] [CrossRef]
  83. Younes, A.; Santoro, A.; Zinzani, P.L.; Timmerman, J.; Ansell, S.M.; Armand, P.; Fanale, M.A.; Ratanatharathorn, V.; Kuruvilla, J.; Cohen, J.B.; et al. Checkmate 205: Nivolumab (Nivo) in Classical Hodgkin Lymphoma (cHL) after Autologous Stem Cell Transplant (ASCT) and Brentuximab Vedotin (BV)—A Phase 2 Study. J. Clin. Oncol. 2016, 34, 7535. [Google Scholar] [CrossRef]
  84. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  85. Sharma, P.; Retz, M.; Siefker-Radtke, A.; Baron, A.; Necchi, A.; Bedke, J.; Plimack, E.R.; Vaena, D.; Grimm, M.-O.; Bracarda, S.; et al. Nivolumab in Metastatic Urothelial Carcinoma after Platinum Therapy (CheckMate 275): A Multicentre, Single-Arm, Phase 2 Trial. Lancet Oncol. 2017, 18, 312–322. [Google Scholar] [CrossRef] [PubMed]
  86. Ready, N.; Farago, A.F.; De Braud, F.; Atmaca, A.; Hellmann, M.D.; Schneider, J.G.; Spigel, D.R.; Moreno, V.; Chau, I.; Hann, C.L.; et al. Third-Line Nivolumab Monotherapy in Recurrent SCLC: CheckMate 032. J. Thorac. Oncol. 2019, 14, 237–244. [Google Scholar] [CrossRef]
  87. Cho, B.C.; Kato, K.; Takahashi, M.; Okada, M.; Lin, C.-Y.; Chin, K.; Kadowaki, S.; Ahn, M.-J.; Hamamoto, Y.; Doki, Y.; et al. Nivolumab versus Chemotherapy in Advanced Esophageal Squamous Cell Carcinoma (ESCC): The Phase III ATTRACTION-3 Study. Ann. Oncol. 2019, 30, v873–v874. [Google Scholar] [CrossRef]
  88. Migden, M.R.; Rischin, D.; Schmults, C.D.; Guminski, A.; Hauschild, A.; Lewis, K.D.; Chung, C.H.; Hernandez-Aya, L.; Lim, A.M.; Chang, A.L.S.; et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2018, 379, 341–351. [Google Scholar] [CrossRef] [PubMed]
  89. Stratigos, A.J.; Sekulic, A.; Peris, K.; Bechter, O.; Prey, S.; Kaatz, M.; Lewis, K.D.; Basset-Seguin, N.; Chang, A.L.S.; Dalle, S.; et al. Cemiplimab in Locally Advanced Basal Cell Carcinoma after Hedgehog Inhibitor Therapy: An Open-Label, Multi-Centre, Single-Arm, Phase 2 Trial. Lancet Oncol. 2021, 22, 848–857. [Google Scholar] [CrossRef]
  90. Sezer, A.; Kilickap, S.; Gümüş, M.; Bondarenko, I.; Özgüroğlu, M.; Gogishvili, M.; Turk, H.M.; Cicin, I.; Bentsion, D.; Gladkov, O.; et al. Cemiplimab Monotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer with PD-L1 of at Least 50%: A Multicentre, Open-Label, Global, Phase 3, Randomised, Controlled Trial. Lancet 2021, 397, 592–604. [Google Scholar] [CrossRef]
  91. Khoja, L.; Butler, M.O.; Kang, S.P.; Ebbinghaus, S.; Joshua, A.M. Pembrolizumab. J. Immunother. Cancer 2015, 3, 36. [Google Scholar] [CrossRef]
  92. Robert, C.; Ribas, A.; Wolchok, J.D.; Hodi, F.S.; Hamid, O.; Kefford, R.; Weber, J.S.; Joshua, A.M.; Hwu, W.J.; Gangadhar, T.C.; et al. Anti-Programmed-Death-Receptor-1 Treatment with Pembrolizumab in Ipilimumab-Refractory Advanced Melanoma: A Randomised Dose-Comparison Cohort of a Phase 1 Trial. Lancet 2014, 384, 1109–1117. [Google Scholar] [CrossRef]
  93. Robert, C.; Ribas, A.; Schachter, J.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.M.; Lotem, M.; 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. Lancet Oncol. 2019, 20, 1239–1251. [Google Scholar] [CrossRef] [PubMed]
  94. Garon, E.B.; Hellmann, M.D.; Rizvi, N.A.; Carcereny, E.; Leighl, N.B.; Ahn, M.J.; Eder, J.P.; Balmanoukian, A.S.; Aggarwal, C.; Horn, L.; et al. Five-Year Overall Survival for Patients with Advanced Non-Small-Cell Lung Cancer Treated with Pembrolizumab: Results from the Phase i KEYNOTE-001 Study. J. Clin. Oncol. 2019, 37, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  95. Atezolizumab for Urothelial Carcinoma|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/atezolizumab-urothelial-carcinoma (accessed on 9 April 2020).
  96. Hoffman-Censits, J.H.; Grivas, P.; Van Der Heijden, M.S.; Dreicer, R.; Loriot, Y.; Retz, M.; Vogelzang, N.J.; Perez-Gracia, J.L.; Rezazadeh, A.; Bracarda, S.; et al. IMvigor 210, a Phase II Trial of Atezolizumab (MPDL3280A) in Platinum-Treated Locally Advanced or Metastatic Urothelial Carcinoma (mUC). J. Clin. Oncol. 2016, 34, 355. [Google Scholar] [CrossRef]
  97. Balar, A.V.; Galsky, M.D.; Rosenberg, J.E.; Powles, T.; Petrylak, D.P.; Bellmunt, J.; Loriot, Y.; Necchi, A.; Hoffman-Censits, J.; Perez-Gracia, J.L.; et al. Atezolizumab as First-Line Treatment in Cisplatin-Ineligible Patients with Locally Advanced and Metastatic Urothelial Carcinoma: A Single-Arm, Multicentre, Phase 2 Trial. Lancet 2017, 389, 67–76. [Google Scholar] [CrossRef]
  98. Atezolizumab (TECENTRIQ)|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/atezolizumab-tecentriq (accessed on 9 April 2020).
  99. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus Docetaxel for Patients with Previously Treated Non-Small-Cell Lung Cancer (POPLAR): A Multicentre, Open-Label, Phase 2 Randomised Controlled Trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef] [PubMed]
  100. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus Docetaxel in Patients with Previously Treated Non-Small-Cell Lung Cancer (OAK): A Phase 3, Open-Label, Multicentre Randomised Controlled Trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
  101. Hematology/Oncology (Cancer) Approvals & Safety Notifications|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/hematologyoncology-cancer-approvals-safety-notifications (accessed on 9 April 2020).
  102. FDA Approves Atezolizumab for PD-L1 Positive Unresectable Locally Advanced or Metastatic Triple-Negative Breast Cancer|FDA. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-pd-l1-positive-unresectable-locally-advanced-or-metastatic-triple-negative (accessed on 9 April 2020).
  103. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.-A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef] [PubMed]
  104. Clinical Trials Using Atezolizumab—National Cancer Institute. Available online: https://www.cancer.gov/about-cancer/treatment/clinical-trials/intervention/atezolizumab (accessed on 9 April 2020).
  105. Horn, L.; Mansfield, A.S.; Szczęsna, A.; Havel, L.; Krzakowski, M.; Hochmair, M.J.; Huemer, F.; Losonczy, G.; Johnson, M.L.; Nishio, M.; et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2220–2229. [Google Scholar] [CrossRef]
  106. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  107. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, Vemurafenib, and Cobimetinib as First-Line Treatment for Unresectable Advanced BRAFV600 Mutation-Positive Melanoma (IMspire150): Primary Analysis of the Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
  108. Chen, A.P.; Sharon, E.; O’sullivan-Coyne, G.; Moore, N.; Foster, J.C.; Hu, J.S.; Van Tine, B.A.; Conley, A.P.; Read, W.L.; Riedel, R.F.; et al. Atezolizumab for Advanced Alveolar Soft Part Sarcoma. N. Engl. J. Med. 2023, 389, 911–921. [Google Scholar] [CrossRef] [PubMed]
  109. Kaufman, H.L.; Russell, J.S.; Hamid, O.; Bhatia, S.; Terheyden, P.; D’Angelo, S.P.; Shih, K.C.; Lebbé, C.; Milella, M.; Brownell, I.; et al. Updated Efficacy of Avelumab in Patients with Previously Treated Metastatic Merkel Cell Carcinoma after ≥1 Year of Follow-up: JAVELIN Merkel 200, a Phase 2 Clinical Trial. J. Immunother. Cancer 2018, 6, 7. [Google Scholar] [CrossRef] [PubMed]
  110. Patel, M.R.; Ellerton, J.; Infante, J.R.; Agrawal, M.; Gordon, M.; Aljumaily, R.; Britten, C.D.; Dirix, L.; Lee, K.-W.; Taylor, M.; et al. Avelumab in Metastatic Urothelial Carcinoma after Platinum Failure (JAVELIN Solid Tumor): Pooled Results from Two Expansion Cohorts of an Open-Label, Phase 1 Trial. Lancet Oncol. 2018, 19, 51–64. [Google Scholar] [CrossRef] [PubMed]
  111. Powles, T.; Park, S.H.; Voog, E.; Caserta, C.; Valderrama, B.P.; Gurney, H.; Kalofonos, H.; Radulović, S.; Demey, W.; Ullén, A.; et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2020, 383, 1218–1230. [Google Scholar] [CrossRef] [PubMed]
  112. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115. [Google Scholar] [CrossRef] [PubMed]
  113. Powles, T.; O’Donnell, P.H.; Massard, C.; Arkenau, H.-T.; Friedlander, T.W.; Hoimes, C.J.; Lee, J.L.; Ong, M.; Sridhar, S.S.; Vogelzang, N.J.; et al. Efficacy and Safety of Durvalumab in Locally Advanced or Metastatic Urothelial Carcinoma: Updated Results From a Phase 1/2 Open-Label Study. JAMA Oncol. 2017, 3, e172411. [Google Scholar] [CrossRef] [PubMed]
  114. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; De Wit, M.; et al. Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef] [PubMed]
  115. Paz-Ares, L.; Dvorkin, M.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; et al. Durvalumab plus Platinum–Etoposide versus Platinum–Etoposide in First-Line Treatment of Extensive-Stage Small-Cell Lung Cancer (CASPIAN): A Randomised, Controlled, Open-Label, Phase 3 Trial. Lancet 2019, 394, 1929–1939. [Google Scholar] [CrossRef] [PubMed]
  116. Oh, D.-Y.; Ruth He, A.; Qin, S.; Chen, L.-T.; Okusaka, T.; Vogel, A.; Kim, J.W.; Suksombooncharoen, T.; Ah Lee, M.; Kitano, M.; et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced Biliary Tract Cancer. NEJM Evid. 2022, 1, EVIDoa2200015. [Google Scholar] [CrossRef]
  117. Abou-Alfa, G.K.; Chan, S.L.; Kudo, M.; Lau, G.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Dao, T.V.; De Toni, E.N.; et al. Phase 3 Randomized, Open-Label, Multicenter Study of Tremelimumab (T) and Durvalumab (D) as First-Line Therapy in Patients (Pts) with Unresectable Hepatocellular Carcinoma (uHCC): HIMALAYA. J. Clin. Oncol. 2022, 40, 379. [Google Scholar] [CrossRef]
  118. MacLachlan, B.J.; Mason, G.H.; Greenshields-Watson, A.; Triebel, F.; Gallimore, A.; Cole, D.K.; Godkin, A. Molecular Characterization of HLA Class II Binding to the LAG-3 T Cell Co-Inhibitory Receptor. Eur. J. Immunol. 2021, 51, 331–341. [Google Scholar] [CrossRef] [PubMed]
  119. Huard, B.; Mastrangeli, R.; Prigent, P.; Bruniquel, D.; Donini, S.; El-Tayar, N.; Maigret, B.; Dréano, M.; Triebel, F. Characterization of the Major Histocompatibility Complex Class II Binding Site on LAG-3 Protein. Proc. Natl. Acad. Sci. USA 1997, 94, 5744–5749. [Google Scholar] [CrossRef] [PubMed]
  120. Wang, J.; Sanmamed, M.F.; Datar, I.; Su, T.T.; Ji, L.; Sun, J.; Chen, L.; Chen, Y.; Zhu, G.; Yin, W.; et al. Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell 2019, 176, 334–347.e12. [Google Scholar] [CrossRef] [PubMed]
  121. Kouo, T.; Huang, L.; Pucsek, A.B.; Cao, M.; Solt, S.; Armstrong, T.; Jaffee, E. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol. Res. 2015, 3, 412–423. [Google Scholar] [CrossRef] [PubMed]
  122. Abdel-Rahman, S.A.; Rehman, A.U.; Gabr, M.T. Discovery of First-in-Class Small Molecule Inhibitors of Lymphocyte Activation Gene 3 (LAG-3). ACS Med. Chem. Lett. 2023, 14, 629–635. [Google Scholar] [CrossRef]
  123. Grosso, J.F.; Kelleher, C.C.; Harris, T.J.; Maris, C.H.; Hipkiss, E.L.; Marzo, A.D.; Anders, R.; Netto, G.; Getnet, D.; Bruno, T.C.; et al. LAG-3 Regulates CD8+ T Cell Accumulation and Effector Function in Murine Self- and Tumor-Tolerance Systems. J. Clin. Investig. 2007, 117, 3383–3392. [Google Scholar] [CrossRef] [PubMed]
  124. Tian, X.; Zhang, A.; Qiu, C.; Wang, W.; Yang, Y.; Qiu, C.; Liu, A.; Zhu, L.; Yuan, S.; Hu, H.; et al. The Upregulation of LAG-3 on T Cells Defines a Subpopulation with Functional Exhaustion and Correlates with Disease Progression in HIV-Infected Subjects. J. Immunol. 2015, 194, 3873–3882. [Google Scholar] [CrossRef] [PubMed]
  125. Wherry, E.J.; Kurachi, M. Molecular and Cellular Insights into T Cell Exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  126. Maruhashi, T.; Okazaki, I.-M.; Sugiura, D.; Takahashi, S.; Maeda, T.K.; Shimizu, K.; Okazaki, T. LAG-3 Inhibits the Activation of CD4+ T Cells That Recognize Stable pMHCII through Its Conformation-Dependent Recognition of pMHCII. Nat. Immunol. 2018, 19, 1415–1426. [Google Scholar] [CrossRef]
  127. Huard, B.; Prigent, P.; Tournier, M.; Bruniquel, D.; Triebel, F. CD4/Major Histocompatibility Complex Class II Interaction Analyzed with CD4- and Lymphocyte Activation Gene-3 (LAG-3)-Ig Fusion Proteins. Eur. J. Immunol. 1995, 25, 2718–2721. [Google Scholar] [CrossRef]
  128. Graydon, C.G.; Mohideen, S.; Fowke, K.R. LAG3’s Enigmatic Mechanism of Action. Front. Immunol. 2021, 11, 615317. [Google Scholar] [CrossRef] [PubMed]
  129. Workman, C.J.; Dugger, K.J.; Vignali, D.A.A. Cutting Edge: Molecular Analysis of the Negative Regulatory Function of Lymphocyte Activation Gene-3. J. Immunol. 2002, 169, 5392–5395. [Google Scholar] [CrossRef]
  130. Huang, R.-Y.; Eppolito, C.; Lele, S.; Shrikant, P.; Matsuzaki, J.; Odunsi, K. LAG3 and PD1 Co-Inhibitory Molecules Collaborate to Limit CD8+ T Cell Signaling and Dampen Antitumor Immunity in a Murine Ovarian Cancer Model. Oncotarget 2015, 6, 27359–27377. [Google Scholar] [CrossRef] [PubMed]
  131. Huang, R.Y.; Francois, A.; McGray, A.R.; Miliotto, A.; Odunsi, K. Compensatory Upregulation of PD-1, LAG-3, and CTLA-4 Limits the Efficacy of Single-Agent Checkpoint Blockade in Metastatic Ovarian Cancer. OncoImmunology 2017, 6, e1249561. [Google Scholar] [CrossRef]
  132. Yap, T.A.; LoRusso, P.M.; Wong, D.J.; Hu-Lieskovan, S.; Papadopoulos, K.P.; Holz, J.-B.; Grabowska, U.; Gradinaru, C.; Leung, K.-M.; Marshall, S.; et al. A Phase 1 First-in-Human Study of FS118, a Tetravalent Bispecific Antibody Targeting LAG-3 and PD-L1 in Patients with Advanced Cancer and PD-L1 Resistance. Clin. Cancer Res. 2023, 29, 888–898. [Google Scholar] [CrossRef]
  133. Chen, J.; Chen, Z. The Effect of Immune Microenvironment on the Progression and Prognosis of Colorectal Cancer. Med. Oncol. 2014, 31, 82. [Google Scholar] [CrossRef] [PubMed]
  134. Giraldo, N.A.; Becht, E.; Pagès, F.; Skliris, G.; Verkarre, V.; Vano, Y.; Mejean, A.; Saint-Aubert, N.; Lacroix, L.; Natario, I.; et al. Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer. Clin. Cancer Res. 2015, 21, 3031–3040. [Google Scholar] [CrossRef]
  135. He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 Protein Expression in Non–Small Cell Lung Cancer and Its Relationship with PD-1/PD-L1 and Tumor-Infiltrating Lymphocytes. J. Thorac. Oncol. 2017, 12, 814–823. [Google Scholar] [CrossRef]
  136. Burugu, S.; Gao, D.; Leung, S.; Chia, S.K.; Nielsen, T.O. LAG-3+ Tumor Infiltrating Lymphocytes in Breast Cancer: Clinical Correlates and Association with PD-1/PD-L1+ Tumors. Ann. Oncol. 2017, 28, 2977–2984. [Google Scholar] [CrossRef]
  137. Maruhashi, T.; Sugiura, D.; Okazaki, I.; Okazaki, T. LAG-3: From Molecular Functions to Clinical Applications. J. Immunother. Cancer 2020, 8, e001014. [Google Scholar] [CrossRef]
  138. Grosso, J.F.; Goldberg, M.V.; Getnet, D.; Bruno, T.C.; Yen, H.-R.; Pyle, K.J.; Hipkiss, E.; Vignali, D.A.A.; Pardoll, D.M.; Drake, C.G. Functionally Distinct LAG-3 and PD-1 Subsets on Activated and Chronically Stimulated CD8 T Cells. J. Immunol. 2009, 182, 6659–6669. [Google Scholar] [CrossRef] [PubMed]
  139. Su, J.; Fu, Y.; Cui, Z.; Abidin, Z.; Yuan, J.; Zhang, X.; Li, R.; Zhao, C. Relatlimab: A Novel Drug Targeting Immune Checkpoint LAG-3 in Melanoma Therapy. Front. Pharmacol. 2024, 14, 1349081. [Google Scholar] [CrossRef] [PubMed]
  140. Long, L.; Zhang, X.; Chen, F.; Pan, Q.; Phiphatwatchara, P.; Zeng, Y.; Chen, H. The Promising Immune Checkpoint LAG-3: From Tumor Microenvironment to Cancer Immunotherapy. Genes Cancer 2018, 9, 176–189. [Google Scholar] [CrossRef]
  141. Huang, C.-T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in Regulatory T Cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef] [PubMed]
  142. Nguyen, L.T.; Ohashi, P.S. Clinical Blockade of PD1 and LAG3—Potential Mechanisms of Action | Nature Reviews Immunology. Nat. Rev. Immunol. 2015, 15, 45–56. [Google Scholar] [CrossRef] [PubMed]
  143. Ascierto, P.A.; Lipson, E.J.; Dummer, R.; Larkin, J.; Long, G.V.; Sanborn, R.E.; Chiarion-Sileni, V.; Dréno, B.; Dalle, S.; Schadendorf, D.; et al. Nivolumab and Relatlimab in Patients With Advanced Melanoma That Had Progressed on Anti–Programmed Death-1/Programmed Death Ligand 1 Therapy: Results From the Phase I/IIa RELATIVITY-020 Trial. J. Clin. Oncol. 2023, 41, 2724–2735. [Google Scholar] [CrossRef] [PubMed]
  144. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  145. Weinmann, S.C.; Pisetsky, D.S. Mechanisms of Immune-Related Adverse Events during the Treatment of Cancer with Immune Checkpoint Inhibitors. Rheumatol. Oxf. Engl. 2019, 58, vii59–vii67. [Google Scholar] [CrossRef] [PubMed]
  146. Liu, Y.H.; Zang, X.Y.; Wang, J.C.; Huang, S.S.; Xu, J.; Zhang, P. Diagnosis and Management of Immune Related Adverse Events (irAEs) in Cancer Immunotherapy. Biomed. Pharmacother. 2019, 120, 109437. [Google Scholar] [CrossRef]
  147. Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef]
  148. Das, S.; Johnson, D.B. Immune-Related Adverse Events and Anti-Tumor Efficacy of Immune Checkpoint Inhibitors. J. Immunother. Cancer 2019, 7, 306. [Google Scholar] [CrossRef] [PubMed]
  149. Andrews, A. Treating with Checkpoint Inhibitors-Figure $1 Million per Patient. Am. Health Drug Benefits 2015, 8, 9. [Google Scholar] [PubMed]
  150. Arora, S.; Velichinskii, R.; Lesh, R.W.; Ali, U.; Kubiak, M.; Bansal, P.; Borghaei, H.; Edelman, M.J.; Boumber, Y. Existing and Emerging Biomarkers for Immune Checkpoint Immunotherapy in Solid Tumors. Adv. Ther. 2019, 36, 2638–2678. [Google Scholar] [CrossRef] [PubMed]
  151. Davis, A.A.; Patel, V.G. The Role of PD-L1 Expression as a Predictive Biomarker: An Analysis of All US Food and Drug Administration (FDA) Approvals of Immune Checkpoint Inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef]
  152. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed]
  153. Schlötterer, C.; Harr, B.; Schlo, C. Microsatellite Instability. In Encyclopedia of Life Sciences; Nature Publishing Group: London, UK, 2017; pp. 1–4. [Google Scholar] [CrossRef]
  154. Drescher, K.M.; Sharma, P.; Watson, P.; Gatalica, Z.; Thibodeau, S.N.; Lynch, H.T. Lymphocyte Recruitment into the Tumor Site Is Altered in Patients with MSI-H Colon Cancer. Fam. Cancer 2009, 8, 231–239. [Google Scholar] [CrossRef] [PubMed]
  155. Duruisseaux, M.; Martínez-Cardús, A.; Calleja-Cervantes, M.E.; Moran, S.; Castro de Moura, M.; Davalos, V.; Piñeyro, D.; Sanchez-Cespedes, M.; Girard, N.; Brevet, M.; et al. Epigenetic Prediction of Response to Anti-PD-1 Treatment in Non-Small-Cell Lung Cancer: A Multicentre, Retrospective Analysis. Lancet Respir. Med. 2018, 6, 771–781. [Google Scholar] [CrossRef] [PubMed]
  156. Wu, H.-J.; Wu, E. The Role of Gut Microbiota in Immune Homeostasis and Autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef]
  157. Zitvogel, L.; Ayyoub, M.; Routy, B.; Kroemer, G. Microbiome and Anticancer Immunosurveillance. Cell 2016, 165, 276–287. [Google Scholar] [CrossRef]
  158. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut Microbiome Modulates Response to Anti–PD-1 Immunotherapy in Melanoma Patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  159. Havel, J.J.; Chowell, D.; Chan, T.A. The Evolving Landscape of Biomarkers for Checkpoint Inhibitor Immunotherapy. Nat. Rev. Cancer 2019, 19, 133–150. [Google Scholar] [CrossRef] [PubMed]
  160. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [PubMed]
  161. Sauer, N.; Janicka, N.; Szlasa, W.; Skinderowicz, B.; Kołodzińska, K.; Dwernicka, W.; Oślizło, M.; Kulbacka, J.; Novickij, V.; Karłowicz-Bodalska, K. TIM-3 as a Promising Target for Cancer Immunotherapy in a Wide Range of Tumors. Cancer Immunol. Immunother. 2023, 72, 3405–3425. [Google Scholar] [CrossRef] [PubMed]
  162. Zhu, C.; Anderson, A.C.; Schubart, A.; Xiong, H.; Imitola, J.; Khoury, S.J.; Zheng, X.X.; Strom, T.B.; Kuchroo, V.K. The Tim-3 Ligand Galectin-9 Negatively Regulates T Helper Type 1 Immunity. Nat. Immunol. 2005, 6, 1245–1252. [Google Scholar] [CrossRef]
  163. Xu, S.; Zhang, N.; Rinne, M.L.; Sun, H.; Stein, A.M. Sabatolimab (MBG453) Model-Informed Drug Development for Dose Selection in Patients with Myelodysplastic Syndrome/Acute Myeloid Leukemia and Solid Tumors. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 1653–1665. [Google Scholar] [CrossRef] [PubMed]
  164. Brunner, A.M.; Esteve, J.; Porkka, K.; Knapper, S.; Traer, E.; Scholl, S.; Garcia-Manero, G.; Vey, N.; Wermke, M.; Janssen, J.; et al. Efficacy and Safety of Sabatolimab (MBG453) in Combination with Hypomethylating Agents (HMAs) in Patients (Pts) with Very High/High-Risk Myelodysplastic Syndrome (vHR/HR-MDS) and Acute Myeloid Leukemia (AML): Final Analysis from a Phase Ib Study. Blood 2021, 138, 244. [Google Scholar] [CrossRef]
  165. Crescioli, S.; Kaplon, H.; Chenoweth, A.; Wang, L.; Visweswaraiah, J.; Reichert, J.M. Antibodies to Watch in 2024. mAbs 2024, 16, 2297450. [Google Scholar] [CrossRef]
  166. Zeidan, A.M.; Giagounidis, A.; Sekeres, M.A.; Xiao, Z.; Sanz, G.F.; Hoef, M.V.; Ma, F.; Hertle, S.; Santini, V. STIMULUS-MDS2 Design and Rationale: A Phase III Trial with the Anti-TIM-3 Sabatolimab (MBG453) + Azacitidine in Higher Risk MDS and CMML-2. Future Oncol. 2023, 19, 631–642. [Google Scholar] [CrossRef]
  167. Zhao, J.; Li, L.; Yin, H.; Feng, X.; Lu, Q. TIGIT: An Emerging Immune Checkpoint Target for Immunotherapy in Autoimmune Disease and Cancer. Int. Immunopharmacol. 2023, 120, 110358. [Google Scholar] [CrossRef]
  168. Chu, X.; Tian, W.; Wang, Z.; Zhang, J.; Zhou, R. Co-Inhibition of TIGIT and PD-1/PD-L1 in Cancer Immunotherapy: Mechanisms and Clinical Trials. Mol. Cancer 2023, 22, 93. [Google Scholar] [CrossRef]
  169. Janjigian, Y.Y.; Oh, D.-Y.; Pelster, M.; Wainberg, Z.A.; Sison, E.A.R.; Scott, J.R.; Ronayne, J.; Wishengrad, D.; Rhee, J.; Nuyten, D.S.A.; et al. EDGE-Gastric Arm A1: Phase 2 Study of Domvanalimab, Zimberelimab, and FOLFOX in First-Line (1L) Advanced Gastroesophageal Cancer. J. Clin. Oncol. 2023, 41, 433248. [Google Scholar] [CrossRef]
  170. Johnson, M.L.; Fox, W.; Lee, Y.-G.; Lee, K.H.; Ahn, H.K.; Kim, Y.-C.; Lee, K.-Y.; Lee, J.-S.; He, X.; Park, C.; et al. ARC-7: Randomized Phase 2 Study of Domvanalimab + Zimberelimab ± Etrumadenant versus Zimberelimab in First-Line, Metastatic, PD-L1-High Non-Small Cell Lung Cancer (NSCLC). J. Clin. Oncol. 2022, 40, 397600. [Google Scholar] [CrossRef]
  171. Chouaid, C.; Ho, G.F.; Runglodvatana, Y.; He, X.; Ahlers, C.M.; Pomponio, D.; Todd, T.; Dang, T.; Naidoo, J. ARC-10: A Phase 3 Study to Evaluate Zimberelimab + Domvanalimab versus Pembrolizumab in Front-Line, PD-L1-High, Locally Advanced or Metastatic Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2023, 41, TPS9148. [Google Scholar] [CrossRef]
  172. Seliger, B. Combinatorial Approaches with Checkpoint Inhibitors to Enhance Anti-Tumor Immunity. Front. Immunol. 2019, 10, 999. [Google Scholar] [CrossRef] [PubMed]
  173. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
  174. Saleh, R.; Toor, S.M.; Khalaf, S.; Elkord, E. Breast Cancer Cells and PD-1/PD-L1 Blockade Upregulate the Expression of PD-1, CTLA-4, TIM-3 and LAG-3 Immune Checkpoints in CD4+ T Cells. Vaccines 2019, 7, 149. [Google Scholar] [CrossRef] [PubMed]
  175. Vanpouille-Box, C.; Formenti, S.C.; Demaria, S. Toward Precision Radiotherapy for Use with Immune Checkpoint Blockers. Clin. Cancer Res. 2018, 24, 259–265. [Google Scholar] [CrossRef]
  176. Rudqvist, N.P.; Pilones, K.A.; Lhuillier, C.; Wennerberg, E.; Sidhom, J.W.; Emerson, R.O.; Robins, H.S.; Schneck, J.; Formenti, S.C.; Demaria, S. Radiotherapy and CTLA-4 Blockade Shape the Tcr Repertoire of Tumor-Infiltrating t Cells. Cancer Immunol. Res. 2018, 6, 139–150. [Google Scholar] [CrossRef]
Figure 1. Current and emerging immune checkpoint receptors and their respective ligands. They transmit co-inhibitory or co-stimulatory signals to TCR upon binding to their ligands. Figure generated using BioRender.com.
Figure 1. Current and emerging immune checkpoint receptors and their respective ligands. They transmit co-inhibitory or co-stimulatory signals to TCR upon binding to their ligands. Figure generated using BioRender.com.
Immuno 04 00013 g001
Figure 2. CTLA4:CD80/86 binding inhibits TCR activation via competitive inhibition of CD28 co-stimulatory signalling. Anti-CTLA4 antibodies such as ipilimumab block CTLA4:CD80/86 binding, allowing unrestrained CD28 co-stimulation and increased TCR activation. Figure generated using BioRender.com.
Figure 2. CTLA4:CD80/86 binding inhibits TCR activation via competitive inhibition of CD28 co-stimulatory signalling. Anti-CTLA4 antibodies such as ipilimumab block CTLA4:CD80/86 binding, allowing unrestrained CD28 co-stimulation and increased TCR activation. Figure generated using BioRender.com.
Immuno 04 00013 g002
Figure 3. Tumour cells can express PD-L1, which can directly bind to inhibitory immune checkpoint PD-1 on the surface of T cells. PD-1:PD-L1 binding induces co-inhibitory signalling that inhibits TCR activation. Anti-PD-1 antibodies, such as pembrolizumab, and anti-PD-L1 antibodies, such as atezolizumab, inhibit PD:PD-L1 signalling, allowing unrestrained TCR activation. Figure generated using BioRender.com.
Figure 3. Tumour cells can express PD-L1, which can directly bind to inhibitory immune checkpoint PD-1 on the surface of T cells. PD-1:PD-L1 binding induces co-inhibitory signalling that inhibits TCR activation. Anti-PD-1 antibodies, such as pembrolizumab, and anti-PD-L1 antibodies, such as atezolizumab, inhibit PD:PD-L1 signalling, allowing unrestrained TCR activation. Figure generated using BioRender.com.
Immuno 04 00013 g003
Figure 4. LAG-3 binding to its ligands inhibits TCR activation, primarily through co-inhibitory downstream intracellular signalling. Anti-LAG-3 antibodies such as relatlimab block LAG-3 ligand binding, allowing for unopposed CD4 co-stimulation and increased TCR activation. Figure generated using BioRender.com.
Figure 4. LAG-3 binding to its ligands inhibits TCR activation, primarily through co-inhibitory downstream intracellular signalling. Anti-LAG-3 antibodies such as relatlimab block LAG-3 ligand binding, allowing for unopposed CD4 co-stimulation and increased TCR activation. Figure generated using BioRender.com.
Immuno 04 00013 g004
Figure 5. Immune-related adverse effects can affect almost every organ system. Examples of inflammatory pathology in each organ are shown above. Figure generated using BioRender.com.
Figure 5. Immune-related adverse effects can affect almost every organ system. Examples of inflammatory pathology in each organ are shown above. Figure generated using BioRender.com.
Immuno 04 00013 g005
Table 1. List of cancers for which ipilimumab is an FDA-approved therapy, including key trials that led to approval in each case.
Table 1. List of cancers for which ipilimumab is an FDA-approved therapy, including key trials that led to approval in each case.
Ipilimumab
Cancer TypeFDA Approval YearKey Clinical Trial (Phase)Monotherapy/Combination Therapy
Melanoma2011MDX010-20 (Phase 3) [13]Monotherapy
2015CheckMate-067 (Phase 3) [34]Combination with nivolumab
Renal Cell Carcinoma2018CheckMate-214 (Phase 3) [12]Combination with nivolumab
Colorectal Cancer—Microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR)2018CheckMate 142 (Phase 2) [35]Combination with nivolumab
Hepatocellular Carcinoma2020CheckMate 040 (Phase 1/2) [36]Combination with nivolumab
Non-small-cell lung carcinoma2020CheckMate 227 (Phase 3) [37]Combination with nivolumab
Pleural Mesothelioma2020CheckMate 743 (Phase 3) [38]Combination with nivolumab
Oesophageal Squamous Cell Carcinoma2021CheckMate 648 (Phase 3) [39]Combination with nivolumab
Table 2. List of FDA-approved Anti-PD-1 ICIs, including the specific cancers they are approved for and the key clinical trials that led to approval in each case.
Table 2. List of FDA-approved Anti-PD-1 ICIs, including the specific cancers they are approved for and the key clinical trials that led to approval in each case.
ANTI-PD-1 Immune Checkpoint Inhibitors
DrugCancer TypeFDA Approval YearKey Trial (Phase)Monotherapy/Combination Therapy
PembrolizumabMelanoma2014KEYNOTE-006 (Phase 3) [65]Monotherapy
Non-Small-Cell Lung Carcinoma2015KEYNOTE-010 (Phase 2/3) [66]Monotherapy
Squamous Cell Carcinoma ofHead and Neck2016KEYNOTE-012 (Phase 1b) [67]Monotherapy
Hodgkin’s Lymphoma2017KEYNOTE-087 (Phase 2) [68]Monotherapy
Urothelial Carcinoma2017KEYNOTE-052 (Phase 2) [69]Monotherapy
Colorectal cancer (MSI-H/dMMR) 2017KEYNOTE-164 (Phase 2) [70]Monotherapy
Gastric/Gastroesophageal junction (GEJ) Carcinoma2017KEYNOTE-059 (Phase 2) [71]Monotherapy or Combination with chemotherapy
Cervical Carcinoma2018KEYNOTE-158 (Phase 2) [72]Monotherapy
Hepatocellular Carcinoma2018KEYNOTE-224 (Phase 2) [73]Monotherapy
Merkel Cell Carcinoma2018KEYNOTE-017 (Phase 2) [74]Monotherapy
Renal Cell Carcinoma2019KEYNOTE-426 (Phase 3) [75]Combination with axitinib
Oesophageal Carcinoma2019KEYNOTE-181 (Phase 3) [76]Monotherapy
Triple-Negative Breast Carcinoma2020KEYNOTE-355 (Phase 3) [77]Combination with chemotherapy
Cutaneous Squamous Cell Carcinoma2020KEYNOTE-629 (Phase 2) [78]Monotherapy
Endometrial Carcinoma (MSI-H/dMMR)2022KEYNOTE-158 (Phase 2) [79]Monotherapy
Biliary Tract Carcinoma2023KEYNOTE-966 (Phase 3) [78]Combination with chemotherapy
NivolumabMelanoma 2014CheckMate-037 (Phase 3) [80]Monotherapy
2015CheckMate-067 (Phase 3) [34]Combination with ipilimumab
2017CheckMate-238 (Phase 3) [81]Adjuvant treatment
Non-Small-Cell Lung Carcinoma2015CheckMate-057 (Phase 3) [14]Monotherapy
2020CheckMate-227 (Phase 3) [37]Combination with ipilimumab
Renal Cell Carcinoma2015CheckMate-025 (Phase 3) [82]Monotherapy
2018CheckMate-214 (Phase 3) [12]Combination with ipilimumab
Classical Hodgkin Lymphoma2016CheckMate-205 (Phase 2) [83]Monotherapy
Squamous Cell Carcinoma of Head and Neck Carcinoma2016CheckMate-141 (Phase 3) [84]Monotherapy
Urothelial Carcinoma2017CheckMate-275 (Phase 2) [85]Monotherapy
Colorectal Carcinoma (MSI-H/dMMR)2017CheckMate-142 (Phase 2) [34]Combination with ipilimumab
Hepatocellular carcinoma2020CheckMate-040 (Phase 1/2) [36]Combination with ipilimumab
Small-Cell Lung Carcinoma2018CheckMate-032 (Phase 1/2) [86]Monotherapy
Oesophageal Squamous Cell Carcinoma2020ATTRACTION-3 (Phase 3) [87]Monotherapy
2020CheckMate-648 (Phase 3) [39]Combination with Ipilimumab or Chemotherapy
Pleural Mesothelioma2020CheckMate-743 (Phase 3) [38]Combination with Ipilimumab
Gastric/GEJ Carcinoma2021CheckMate-649 (Phase 3)Combination with Chemotherapy
CemiplimabCutaneous Squamous Cell Carcinoma2018EMPOWER-CSCC-1 (Phase 2) [88]Monotherapy
Basal Cell Carcinoma2021Study-1620 (Phase 2) [89]Monotherapy
Non-Small-Cell Lung Cancer2021EMPOWER-Lung 1 (Phase 3) [90]Monotherapy
Table 3. List of FDA-approved anti-PD-L1 ICIs, including the specific cancers they are approved for and the key clinical trial that led to approval in each case.
Table 3. List of FDA-approved anti-PD-L1 ICIs, including the specific cancers they are approved for and the key clinical trial that led to approval in each case.
ANTI-PD-L1 Immune Checkpoint Inhibitors
DrugCancer TypeFDA Approval YearKey TrialsMonotherapy/Combination
AtezolizumabUrothelial Carcinoma2016IMvigor210 (Phase 2) [97]Monotherapy
Non-Small-Cell Lung Carcinoma2016OAK (Phase 3) [100]Monotherapy
Small-Cell Lung Carcinoma2019IMpower133 (Phase 3) [105]Combination with chemotherapy
Triple-Negative Breast Cancer2019IMpassion130 (Phase 3) [103]Combination with chemotherapy
Hepatocellular Carcinoma2020IMbrave150 (Phase 3) [106]Combination with bevacizumab
Melanoma (BRAF V600 mutation-positive)2020IMspire150 (Phase 3) [107]Combination with cobimetinib and vemurafenib
Alveolar Soft Part Sarcoma2022Study ML39345 (Phase 2) [108]Monotherapy
AvelumabMerkel Cell Carcinoma2017JAVELIN Merkel 200 (Phase 2) [109]Monotherapy
Urothelial Carcinoma2017JAVELIN Solid Tumor (Phase 1) [110]Monotherapy
2020JAVELIN Bladder 100 (Phase 3) [111]1st line maintenance: Combination with best supportive care
Renal Cell Carcinoma2019JAVELIN Renal 101 (Phase 3) [112]Combination with Axitinib
DurvalumabUrothelial Carcinoma2017Study 1108 (Phase 1/2) [113]Monotherapy
Non-Small-Cell Lung Carcinoma2018PACIFIC (Phase 3) [114]Monotherapy
Small-Cell Lung Carcinoma2020CASPIAN (Phase 3) [115]Combination with chemotherapy
Biliary Tract Cancer2022TOPAZ-1 (Phase 3) [116]Combination with chemotherapy
Hepatocellular Carcinoma2020HIMALAYA (Phase 3) [117]Combination with tremelimumab
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Younis, A.; Gribben, J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. Immuno 2024, 4, 186-210. https://doi.org/10.3390/immuno4030013

AMA Style

Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. Immuno. 2024; 4(3):186-210. https://doi.org/10.3390/immuno4030013

Chicago/Turabian Style

Younis, Abdullah, and John Gribben. 2024. "Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions" Immuno 4, no. 3: 186-210. https://doi.org/10.3390/immuno4030013

Article Metrics

Back to TopTop