Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy

Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy

Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy A. Rotte1, J. Y. Jin1, V. Lema...

1MB Sizes 0 Downloads 39 Views

Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy A. Rotte1, J. Y. Jin1, V. Lemaire1 1-Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, California, USA.

Corresponding Authors: Dr. Anand Rotte, Department of Clinical Pharmacology Genentech Research and Early Development, South San Francisco, 94080, California, USA. Tel: +1-650-225-1346 Email: [email protected]; [email protected] Dr. Vincent Lemaire Department of Clinical Pharmacology Genentech Research and Early Development, South San Francisco, 94080, California, USA. Tel: +1-650-225-5320 Email: [email protected] & [email protected]

Running title: T-cell activation checkpoints and cancer immunotherapy

© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: [email protected].

Abstract Checkpoint receptor blockers, known to act by blocking the pathways that inhibit immune cell activation and stimulate immune responses against tumor cells, have been immensely successful in the treatment of cancer. Among several checkpoint receptors of immune cells, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), T-cell immunoglobulin and ITIM domain (TIGIT), T-cell immunoglobulin-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3) are the most commonly targeted checkpoints for cancer immunotherapy. Six drugs including one CTLA-4 blocker (ipilimumab), two PD-1 blockers (nivolumab and pembrolizumab), and three PD-L1 blockers (atezolizumab, avelumab and durvalumab) are approved for the treatment of different types of cancers including both solid tumors such as melanoma, lung cancer, head and neck cancer, bladder cancer and Merkel cell cancer as well as hematological tumors such as classic Hogdkins lymphoma. The main problem with checkpoint blockers is that only a fraction of patients respond to the therapy. Insufficient immune activation is considered as one of the main reason for low response rates and combination of checkpoint blockers has been proposed to increase the response rates. The combination of checkpoint blockers was successful in melanoma but had significant adverse events. A combination that is selected based on the mechanistic differences between checkpoints and the differences in expression of checkpoints and their ligands in the tumor microenvironment (TME) could have a synergistic effect in a given cancer subtype and also have a manageable safety profile. This review aims to help in design of optimal checkpoint blocker combinations by discussing the mechanistic details and outlining the subtle differences between major checkpoints targeted for cancer immunotherapy.

Key words: T-cells, activation, exhaustion, checkpoints, PD-1, cancer immunotherapy

2

Introduction Monoclonal antibodies targeting T-cell inhibitory checkpoint receptors have been highly successful in inducing anti-tumor immune responses and have shown enormous potential in the treatment of cancer (Figure 1). To date six checkpoint receptor blockers (commonly known as checkpoint blockers) are approved for treatment of melanoma, lung cancer, head and neck cancer, bladder cancer, Merkel cell cancer as well as classic Hodgkins lymphoma and seventeen more are in advanced stages of clinical testing (Table 1 and Supplementary Table S1) [1-28]. The greatest advantages of checkpoint blockers have been impressive durable response rates and manageable safety profile [29-31]. However, the achievements of cancer immunotherapy are eclipsed by low response rates with only a fraction of patients benefitting from therapy. Although the use of combinational immunotherapy as in the combination of ipilimumab and nivolumab increased response rates in metastatic melanoma patients, a sizeable proportion of patients remained unresponsive [32, 33]. More importantly, the immune related adverse events commonly associated with checkpoint blockers such as diarrhea, colitis, myocarditis and endocrine disorders were found to be more severe in patients treated with the combination and the activity of the combination also may not extrapolate to lung cancer patients based on the available data [32-41]. As listed in Table 2, checkpoints inhibit immune responses through unique mechanisms; checkpoint receptors as well as their ligands are differently expressed on immune cells and the down stream signaling that follows the receptor activation is also varied. While some checkpoints inhibit immune cell activation in lymph nodes and peripheral tissues, others regulate activation only at peripheral tissues (Table 3). Similarly, some checkpoints induce tolerogenic phenotype in antigen presenting cells (APCs) and/or drive the priming of regulatory Tcells (TRegs), whereas others are involved only in induction of T-cell dysfunction. The differences in checkpoint-mediated regulation could be the reason for varied response rates and adverse effects of checkpoint blocker combinations. A review discussing how the different checkpoints converge in inhibition of anti-tumor immune response could provide a platform for the development of effective checkpoint combinations. To our knowledge there have been very few attempts to compare different checkpoints and did not include all the major checkpoint targets in the discussion [42-46]. The present review

3

therefore aims to provide a foundation for rational design of checkpoint combinations by discussing checkpoints that are currently evaluated in clinical trials such as cytotoxic Tlymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), Tcell immunoglobulin and ITIM domain (TIGIT), T-cell immunoglobulin-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3). The basic concepts of T-cell activation in the lymph

node,

regulation

of

T-cell

activity

in

peripheral

tissues

and

the

exhaustive/dysfunctional phenotype of T-cells are discussed in the following sections to aid in the better understanding of checkpoint mechanisms. T-cell activation Activation of T-cells involves presentation of antigens on MHC I/II molecules to T-cell receptor (TCR) on T-cells by APCs such as dendritic cells (DCs), which results in phosphorylation of CD3ζ subunits and activation of zeta-chain associated protein kinase 70 (ZAP-70) that phosphorylates linker for activation of T-cells (LAT), a scaffold protein for other signaling molecules, and activates PI3K-Akt-MTORC1, MAPK and NFkB pathways [47-56]. However, antigen recognition alone is not sufficient to activate T-cells and requires a second activating signal (also known as costimulation), which most commonly originates from the interaction between CD28 receptors on T-cells and B7ligands (CD80/CD86) on APCs; presentation of antigens to T-cells in the absence of costimulation causes T-cell anergy and apoptosis [57]. In addition to CD28, several other receptors also function as costimulatory receptors for T-cell activation; the receptors and their respective ligands are listed in Supplementary Table S2. Uncontrolled T-cell activation is also prevented by specialized receptors on T-cells called ‘checkpoint receptors’, which act by binding to their cognate ligands on APCs (Supplementary Table S2). Thus the fate of the T-cell in lymph nodes depends on the degree of APC-antigen mediated TCR activation, number of costimulatory receptors, presence of costimulatory ligands on APCs as well as on the number of checkpoint receptors (Figure 2). Interestingly, T-cell activity can also be regulated (activation or inhibition) outside the lymph node and T-cells can interact again with a new APC or other immune cells before reaching the target site. Although the second interaction is not a compulsory event, colocalization of immune cells in tissue spaces introduces the possibility of interaction between T-cell:APC, T-cell:T-cell, T-cell:NK cell and so on. The second interaction in

4

the tissues could decide whether the activated T-cell should proceed with effector functions or should be shutdown and prevented from causing damage to the tissues (Figure 2). To explain the significance of second interaction, some immunologists proposed a two-step activation theory of T-cells: the first step of activation takes place in the lymph nodes and the second step of activation takes place in the peripheral tissues. According to this model, T-cells are primed and activated in the first stage but are not fully committed to the effector phenotype; complete differentiation and commitment to effector functions takes place only after the second interaction, also referred to as ‘second touch’ [58]. Regardless of whether there is enough experimental evidence to support the two-step activation model, the possibility of interactions between immune cells and the outcome cannot be ignored. Costimulatory receptors on T-cells further activate the Tcells and amplify the immune response whereas checkpoint receptors inhibit the T-cells and dampen the response. Tumor microenvironment (TME) is enriched with mediators of immunosuppression, which induce the expression of checkpoint receptors on the surface of T-cells and promote a dysfunctional/exhausted phenotype in T-cells, which is briefly described in the following section [59-65]. T-cell exhaustion Long-term exposure to antigens in presence of inflammatory cytokines induces a distinct phenotype in T-cells, characterized by loss of effector functions, sustained expression of inhibitory receptors, poor proliferative capacity and decreased cytotoxic functions [6668]. Progressive loss of T-cell effector functions, termed ‘T-cell exhaustion’ is commonly seen during chronic viral infections and in cancer. The significance of T-cell exhaustion in cancer has rapidly gained importance and several studies have identified the presence of ‘exhausted’ T-cells in TME [59-65]. Negative regulatory signals arising due to activation of immunosuppressive checkpoints are thought to be the main mechanism for effector T-cell dysfunction in TME. Interestingly, exhausted T-cells were found to have residual effector functions and the blockade of checkpoints of T-cell activity has been shown to rescue and reverse the phenotype in at least a subset of the cells [68-71]. Indeed, monoclonal antibodies targeting CTLA-4 and PD-1 checkpoint pathways have shown tremendous potential in the treatment of some of the cancers types with very poor prognosis (Table 1). The mechanisms of checkpoint blockers are debated as to whether

5

their effects are through promotion of increased T-cell activation by APCs or through reversal of the phenotype of a subset of exhausted T-cells or both. Strikingly, PD-1 receptor expression is considered as an important marker for degree of T-cell exhaustion and the cells with high T-bet (T-box transcription factor) and low-to-intermediate PD-1 expression are considered as reversible [67, 68]. The fact that PD-1:PD-L1 pathway targeting antibodies have produced durable responses in some patients provides credence to the argument that reversal of T-cell exhaustion is one important mechanism by which anti-tumor immune response can be stimulated. However, although the response rates to PD-1:PD-L1 targeting antibodies are encouraging, a significant fraction of patients still do not respond to therapy. The focus of ongoing research is to effectively treat these nonresponding patients. Increasing evidence indicates that exhausted T-cells in the TME express multiple checkpoints and that blockade of a single checkpoint may not be sufficient to stimulate an immune response [59, 60]. Using immune profiling of peripheral blood from melanoma patients, Wherry and associates demonstrated that CD8 T-cells expressing multiple checkpoints were the most responsive to anti-PD-1 therapy [69]. Deeper understanding of the regulation of T-cell activity by checkpoints with emphasis on similarities and differences between the mechanisms could therefore help in the development of a checkpoint blocker combination with higher response rates. Checkpoints commonly associated with T-cell exhaustion are discussed in the next section followed by factors relevant to checkpoint blocker combinations. Checkpoints of T-cell activation CTLA-4 CTLA-4, was discovered in 1987 by Brunet et al, through screening of mouse cytolytic T-cell derived cDNA libraries. The protein was found to be expressed mainly in activated lymphocytes and was co-induced with T-cell mediated cytotoxicity [72]. Loss of Ctla-4 in mice was reported to be associated with development of rapidly progressive and fatal disease characterized by massive lymphoproliferation, multiorgan tissue destruction and death by 3-4 weeks of age [73, 74]. Studies on human CD28/CTLA-4:B7-1 crystal structure revealed that during T-cell activation, CTLA-4 binds to B7 ligands on APCs with higher affinity and at a much lower surface density compared to costimulatory receptor CD28 and suppresses the activation by competing with CD28 for binding with 6

B7 ligands [75]. CTLA-4 expression was shown to be minimal in resting murine T-cells and induced at both mRNA and protein level in response to TCR activation [76, 77]. Unlike other effector cells, murine TRegs were shown to constitutively express CTLA-4 owing to their high expression of forkhead transcription factor FoxP3, a known regulator of CTLA-4 expression [42, 78-80]. CTLA-4:B7-ligand interaction in mice was shown to cause sequestration of B7-ligands, transendocytosis and degradation of endocytosed ligands resulting in tolerogenic APCs due to significant depletion of the B7 ligands from the surface [81]. Furthermore, studies in murine T-cells that followed intracellular events after CTLA-4 engagement also illustrated activation of cell-instrinsic signaling cascades and cross-talk with pathways such as MAPK, PI3K, and NFκB that regulate cell survival and proliferation [82-87]. Anti-CTLA-4 antibodies were the first to show promising results in cancer treatment and 3 antibodies, ipilimumab, tremelimumab and MK1308 progressed into clinical trials. Tremelimumab and MK1308 are currently under development (Supplementary Table S1), whereas ipilimumab is approved for treatment of unresectable stage III/IV metastatic melanoma and as adjuvant therapy for surgically treated ‘high-risk’ melanoma patients [1-3]. PD-1 PD-1 was first described as a cell surface receptor expressed commonly on T cells by Honjo and coworkers from studies on pathways of programmed cell death [88]. Unlike CTLA-4, the phenotype of PD-1 deficient mice is relatively mild, has delayed onset and is dependent on the genetic background of the mouse [89-91]. In humans, PD-1 is expressed on activated T-cells and B-cells, TRegs, natural killer T-cells (NKT) cells, natural killer (NK) cells, activated monocytes and on myeloid DCs. PD-L1 is the widely expressed ligand for PD-1, found on T-cells, B-cells, macrophages, DCs as well as nonhematopoietic cell types such as vascular endothelial cells, fibroblastic reticular cells, epithelia, pancreatic islet cells, astrocytes, neurons and also on cells at sites of immune privilege including trophoblasts in the placenta and retinal pigment epithelial cells. PDL2 has a comparatively narrow expression profile and is seen mainly on activated macrophages and DCs [92-95]. The interaction between PD-1 and its ligands PD-L1/PDL2 impairs T-cell survival and blocks the characteristic features of T-cell response such as cell proliferation, cytokine secretion and cytotoxic ability [96, 97]. In addition, PD-1

7

signaling was also shown to enhance FoxP3 expression in murine models and to regulate the differentiation of induced TReg (iTReg) cells [97]. Recently, PD-1 was shown to be expressed on murine and human tumor-associated macrophages and inhibit their phagocytic potency against tumor cells [98]. The PD-1:PD-L1 pathway is a tissue protective pathway; tumor cells utilize this normal physiological mechanism by expressing PD-L1 and thereby develop resistance to anti-tumor immune responses [42]. Additionally, chronic exposure to increased levels of inflammatory cytokines and antigens can also result in increased expression of PD-1 and PD-L1, a characteristic feature of T-cell exhaustion and dysfunction [68]. PD-1-mediated signaling involves recruitment of phosphatases SHP1 and SHP2, dephosphorylation of effector kinases leading to inhibition of glucose consumption, cytokine production, and the proliferation and survival of T-cells [99]. In contrast to CTLA-4, PD-1 blocks the proximal activation of PI3K/Akt; the extent of its T-cell inhibition thus depends on TCR signal strength and was shown to be abrogated in tumor bearing mice, in presence of T-cell costimulation [70]. While PD-1:PD-L1/PD-L2 pathway could be inhibited by blocking PD-1 or PD-L1 or PD-L2, PD-1 blockers are expected to have greater activation of immune response as they inhibit both PD-1: PD-L1 and PD-1:PD-L2 axis. PD-L1 blockade would only inhibit PD-1:PD-L1 axis, but is expected to activate the immune response significantly because PD-L1 is broadly expressed, whereas PD-L2 blockade can only inhibit PD-1:PD-L2 axis and due to the low expression of PD-L2 ligands, PD-L2 blockers are not expected to significantly increase the immune response. PD-1:PD-L1 pathway has been the most promising

target

for

cancer

immunotherapy

and

two

anti-PD-1

antibodies

(pembrolizumab and nivolumab), and three anti-PD-L1 antibodies (atezolizumab, avelumab and durvalumab) are approved for the treatment of various types of cancer [428]. Further, eight monoclonal antibodies against PD-1 and five against PD-L1 are under clinical development (Supplementary Table S1). Strikingly, anti-PD-1 therapy is the first cancer treatment to receive approval for patients with solid tumors that are positive for microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) abnormalities [100-102].

8

TIGIT TIGIT is an inhibitory receptor recently discovered by scientists from Genentech and independently by other researchers through genomic search for T-cell specific genes with protein domain structures representative of potential inhibitory receptors [103-105]. In humans, TIGIT expression is mainly seen on resting TReg cells, Tr1 cells, memory Tcells, activated T-cells, exhausted CD8+ T-cells, TFH cells, NKT cells and NK cells, but it is not detected on naïve CD4+ T-cells [103-108]. TIGIT has greater affinity towards poliovirus receptor (PVR; also known as CD155 & nectin-like protein 5) ligands and like CTLA-4 it acts mainly by competing with the costimulatory receptors CD226 and CD96 expressed on T-cells for PVR and PVRL2 ligands (also known as CD112 and nectin 2) [109-112]. Studies in mice showed that TIGIT-PVR interaction inhibits the activation, proliferation and differentiation of T-cells and at the same time activates the survival pathways thereby ensuring long-term survival of inhibited T cells. The interaction of PVR on murine DCs with TIGIT was shown to induce a tolerogenic phenotype in DCs by skewing the cytokine secretion profile of DCs towards decreased IL-12 production and increased IL-10 production [103]. Activation of TIGIT on human NK cells reportedly resulted in decreased IFN-γ production, cytotoxic granule polarization & NK cell cytotoxicity [105]. Finally, experiments in mice revealed shift in cytokine balance, promotion of Th2 phenotype and suppression of Th1 or Th17-phenotype upon TIGIT engagement on TRegs [113]. Intracellular TIGIT signaling has been mostly demonstrated in NK cells where its intracellular domain was shown to associate with β-arrestin 2, recruit SHIP1 (SH2-containing inositol phosphatase 1) and regulate PI3K, MAPK and NF-κB signaling cascades [114-116]. Genentech is leading the development of antiTIGIT antibodies for the treatment of cancer and its molecule MTIG7192A/RG6058 is currently in clinical trials (Supplementary Table S1). TIM-3 TIM-3, was identified by Kuchroo and associates in 2002, as a cell surface receptor selectively expressed on differentiated CD4+ Th1 cells [117]. Apart from Th1 cells, TIM3 is also expressed on activated CD8+ T-cells, Th17 cells, TRegs and other innate immune cells such as DCs, NK cells and monocytes [118]. The ligands for TIM-3 include galectin-9, a C-type lectin, carcinoembryonic antigen-related cell adhesion molecule 1

9

(Ceacam-1), high mobility group box1 protein (HMGB-1) and phosphatidyl serine [119]. Unlike the ligands for other checkpoints such as TIGIT, the ligands for TIM-3 have a broad expression profile and are expressed on a variety of cell types including cancer cells [118]. TIM-3 has been shown to regulate the proliferation and cytokine release of Th1 cells and Tim-3-/- mice were found to be refractory to induction of antigen-specific tolerance [120, 121]. Studies in mice also implicated TIM-3 in expansion of TRegs and myeloid derived suppressor cells (MDSCs) and inhibition of the phagocytic activity and maintenance of peripheral tolerance by DCs [122, 123]. Additionally, TIM-3 was also found to be involved in T-cell exhaustion; high TIM-3 expression has been observed on CD8+ T-cells from tumor bearing mice as well as from cancer patients [124-126]. Human TIM-3 does not have a conventional signaling motif in its cytoplasmic tail and depends on the conserved tyrosine residues in the cytoplasmic region for initiation of signaling cascade. BAT-3 (HLA-B associated transcript 3) plays an important role in the control of TIM-3 signaling; under basal conditions when TIM-3 is not activated by its ligand, BAT3 is bound to TIM-3 and promotes the TCR signaling by blocking SH2 domain interaction with inhibitory Src kinases. TIM-3 activation leads to phosphorylation of tyrosine residues mediated release of BAT-3 from TIM-3 tail and inhibits TCR signaling [44, 127, 128]. The clinical development of anti-TIM-3 antibodies is currently being pursued by Novartis, Tesaro and by Roche (Supplementary Table S1). LAG-3 LAG-3 (also known as CD223) was discovered more than 25 years ago as a CD4-related molecule found on activated T-cells as well as NK cells [129]. In addition, LAG-3 expression is also seen on both natural and induced TRegs, Tr1 cells, exhausted T-cells, B-cells and DCs. The ligands for LAG-3 receptor include MHC class II molecules expressed on APCs, Galectin-3 and liver sinusoidal endothelial cell lectin (LSECtin), a member of dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) family of molecules, expressed in liver as well as several tumor subtypes [130-134]. Unlike other negative regulators of immune response, the effects of LAG-3-mediated suppression are comparatively mild, as the LAG-3-deficient mice develop autoimmune disorders only when tested under permissive genetic background [135-138]. LAG-3 is thought to function in coordination with other checkpoints such as

10

PD-1 to promote tolerance, T-cell dysfunction and exhaustion [44, 68, 139]. Studies in human peripheral blood mononuclear cells showed that the negative effects of LAG-3 on effector T-cell functions were due to its association with CD3 and the subsequent LAG3:CD3 ligation-mediated inhibition of T-cell proliferation, cytokine production and calcium flux [140]. Interestingly, LAG-3 has differential effects on T-cell subtypes; while it inhibits the activity of effector T-cells, LAG-3 promotes the suppressive activity and the IL-10 secretion capacity of TRegs as seen by studies in mouse as well as human cells [141, 142]. The mechanisms involved in these varying effects and the signaling events that occur after LAG-3 activation are not completely understood. While the detailed mechanisms are yet to be elucidated, the KIEELE motif in the cytoplasmic region was shown to be critical for the inhibition of CD4+ effector T-cells [44, 143, 144]. Anti-LAG3 antibodies are under clinical development and recently, anti-LAG-3 (BMS986016) plus anti-PD-1 (nivolumab) combination was reported to show encouraging efficacy in melanoma patients who progressed after anti-PD-1 therapy (Supplementary Table S3) [145, 146]. Apart from BMS, anti-LAG-3 antibodies developed by Roche, Novartis, and Tesaro are also in clinical trials for treatment of cancer (Supplementary Table S1). Perspective Immunotherapy has transformed cancer treatment in the past decade with dramatic increase in durable response rates. While other strategies like cancer vaccines, indoleamine oxidase (IDO) inhibitors and chimeric antigen receptor engineered T-cells (CAR-T-cells) are also promising, the review only focused on discussing checkpoint blockers because they are the most successful class of immunotherapeutics and are approved for multiple tumor-types both as monotherapy and in combination [94, 147]. By discussing the mechanistic differences between checkpoints, the review aims to support the development of future combinations of checkpoint blockers. As pointed out in the previous sections, checkpoint receptors play a significant role in the initial T-cell activation in lymph nodes, second activation in tissues and also in T-cell exhaustion. A key point to be noted here is that though there is some overlap in inhibitory functions, each checkpoint receptor also has distinct functions (Figure 3). For example, CTLA-4 is primarily involved in regulation of T-cell activation in lymph nodes/tissues and in TRegmediated suppression of DC activity. CTLA-4 is not expressed on NK cells and therefore

11

does not regulate NK cell functions. The fact that CTLA-4 knockout mouse has a lethal autoimmune phenotype indicates its predominant role in priming and tolerance to selfantigens [73, 74]. PD-1 receptors on the other hand are expressed on activated T-cells as well as NK cells, and therefore regulate T-cell activation in lymph nodes/tissues, differentiation into TRegs as well as NK cell activity. PD-1 knockout mouse also develops autoimmune phenotype but the onset is delayed and is less severe compared to phenotype of Ctla-4-/- mouse, suggesting that PD-1 has basal role in induction of tolerance to self-antigens and priming, and that it mainly regulates the immune responses in peripheral tissues [89-91]. TIGIT receptors are expressed on activated T-cells and NK cells; they shift the cytokine profile of DCs from Th1-promoting towards TReg promoting and also regulate T-cell and NK cell activation in the tissues. Milder phenotype of Tigit-/- mouse suggests less critical role of TIGIT in the induction of tolerance to self-antigens and points to its importance in peripheral tissues [103]. TIM-3 receptors have broader expression profile and are seen on activated T-cells, NK cells, DCs and monocytes. They mainly promote the expansion of immune suppressor cells like TRegs, MDSCs and macrophages and inhibit the activity of effector T-cells and NK cells in the peripheral tissues. Finally, LAG-3 is expressed in activated T-cells and NK cells and functions by promoting the expansion of TRegs and inhibiting the activity of effector T-cells and NK cells. Tim-3-/- and Lag-3-/- mice have even milder phenotype that can be manifested only upon induction of autoimmunity in the mice and in mice with genetically permissive background respectively, indicating a late-stage role of TIM-3 and LAG-3 in immune response [120, 121, 135]. Inhibition of effector T-cells and NK cells in non-lymphoid/peripheral tissues due to cell-cell interactions as well as exhaustion of effector T-cells due to continuous antigen release under immunosuppressive conditions appear to be important mechanisms operating in the TME. Blockade of PD-1:PD-L1 pathway, has been the most successful cancer immunotherapy strategy to date possibly because it abrogates both peripheral inhibition and T-cell exhaustion and also affects the priming of TRegs. CTLA-4 pathway blockade acts mainly by increasing the T-cell activation in lymph nodes, which is possibly responsible for severe immune-related side effects but not sufficient for stimulating anti-tumor immune responses in most patients. A combination of CTLA-4

12

and PD-1:PD-L1 pathway blockers was expected to increase immune cell activation in lymph nodes as well as peripheral tissues and also reverse T-cell exhaustion. The combination did show remarkable success in melanoma patients but was associated with severe immune-related adverse events and also was unable to achieve similar response rates in lung cancer patients [32-36]. The reasons for this varied success of the combination could be due to differences in the pathways that lead to inhibition of effector cells in TME. The degree of T-cell exhaustion as well as the characteristic checkpoints in melanoma patients may differ from lung cancer patients. Tumor mutation load, neoantigen generation, rate of antigen release and immunogenicity has been reported to vary between cancer subtypes and between cancer patients; these differences could lead to disparities in priming of naïve T-cells, activation of immune response and in degree of T-cell exhaustion [148-154]. Analysis of tumor samples from non-small cell lung cancer (NSCLC) patients showed that the PD-1 expressing CD8+ T-cells had lowest percentages of other inhibitory receptors such as CTLA-4, whereas analysis of immune cells from melanoma patients showed that PD-1+ CD8+ T-cells also expressed other checkpoints such as CTLA-4 and were in fact the most responsive to anti-PD-1 therapy, pointing towards the significance of differences in checkpoint expression and the mechanisms of T-cell exhaustion operating in the TME [59, 69]. TIGIT expression was reported to correlate significantly with PD-1 expression in tumor samples from lung cancer patients and the combination of anti-TIGIT and anti-PD-1 antibodies showed synergistic effects in mouse tumor models suggesting that targeting TIGIT and PD-1 could be advantageous in lung cancer [106]. Along those lines, TIM-3 expression was shown to be altered in CD8+ T-cells following PD-1 therapy indicating the benefits of combining anti-PD-1 and anti-TIM-3 antibodies [69]. Degree of effector T-cell infiltration into tumor sites is another factor that influences the response to immunotherapy and the combination that can increase tumor infiltration of T-cells could have synergistic benefits. For example, in a relatively small group of metastatic renal cell carcinoma patients, a combination of antivascular endothelial growth factor (anti-VEGF) antibody, known to increase vascular permeability and anti-PD-L1 antibody was shown to significantly increase CD8 T-cells in tumor tissues and induce tumor regression [155, 156].

13

An effective combination could thus be cancer-specific and dependent on multiple factors such as status of immune response, mechanisms operating in the TME and the levels of checkpoint receptor co-expression. Assays such as whole exome sequencing, immunohistochemical analyses and peripheral blood immunophenotyping could be employed to analyze biomarkers that help in understanding patient-specific mechanisms of immunosuppression such as neoantigen expression, presence of antigen-specific Tcells, relative expression of checkpoint receptors on effector T-cells, neutrophil-tolymphocyte ratio (NLR) and serum lactate dehydrogenase (LDH), and the information could be used in rational selection of checkpoint combinations (Supplementary Table S4) [100-102, 153, 154, 157-168]. In cancer types where lack of sufficient priming of naïve T-cells and activation of immune responses are expected, a combination of CTLA-4 plus PD-1:PD-L1 pathway blockers could be useful, whereas when the lack of immune response is predominantly due to checkpoints such as TIGIT, TIM-3 and LAG-3, then combining anti-PD-1 with anti-TIGIT, anti-TIM-3 or anti-LAG-3 could be helpful. Similarly, when the immune-related adverse events cannot be tolerated, combining antiPD-1 therapy with targets that can have relatively milder phenotypes such as TIGIT, TIM-3 or LAG-3 would be favorable. In summary, a combination that considers the subtle differences in checkpoint expression and control as well as the information on subtype-specific immunosuppressive pathways in TME may help address the issue of improving response rates to cancer immunotherapy.

Acknowledgements: Authors gratefully acknowledge Dr Madhuri Bhandaru for assistance in the preparation of manuscript and Dr Bert Lum, Dr Helen Winter and Dr Joe Ware for their valuable feedback on the manuscript.

Funding: None declared

Disclosure: All authors are employees of Genentech, a member of the Roche Group. They have no other conflicts of interest to declare.

14

Key Message: Combinations of immune checkpoint blockers showed promising response rates in melanoma patients but also increased the severity and incidence of adverse events. To extend the success of checkpoint blocker combinations into other cancer types and to overcome the problems of adverse events, the combinations need to be selected based on the mechanistic differences between checkpoint pathways.

15

References 1. Hodi FS, O'Day SJ, McDermott DF et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363: 711-723. 2. Robert C, Thomas L, Bondarenko I et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364: 2517-2526. 3. Eggermont AM, Chiarion-Sileni V, Grob JJ et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 2015; 16: 522-530. 4. Weber JS, D'Angelo SP, Minor 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. 5. Robert C, Long GV, Brady B et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015; 372: 320-330. 6. Ribas A, Puzanov I, Dummer R et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015; 16: 908-918. 7. Robert C, Schachter J, Long GV et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 2015; 372: 2521-2532. 8. Garon EB, Rizvi NA, Hui R et al. Pembrolizumab for the treatment of non-smallcell lung cancer. N Engl J Med 2015; 372: 2018-2028. 9. Reck M, Rodriguez-Abreu D, Robinson AG et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016; 375: 1823-1833. 10. Borghaei H, Paz-Ares L, Horn L et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373: 1627-1639. 11. Brahmer J, Reckamp KL, Baas P et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015; 373: 123-135. 12. Chow LQ, Haddad R, Gupta S et al. Antitumor Activity of Pembrolizumab in Biomarker-Unselected Patients With Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma: Results From the Phase Ib KEYNOTE-012 Expansion Cohort. J Clin Oncol 2016. 13. Ferris RL, Blumenschein G, Jr., Fayette J et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 2016; 375: 1856-1867. 14. Ansell SM, Lesokhin AM, Borrello I et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med 2015; 372: 311-319. 15. Younes A, Santoro A, Shipp M et al. Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol 2016; 17: 1283-1294. 16. Chen R, Zinzani PL, Fanale MA 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. 17. Motzer RJ, Escudier B, McDermott DF et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 2015; 373: 1803-1813.

16

18. Sharma P, Retz M, Siefker-Radtke A 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. 19. Bellmunt J, de Wit R, Vaughn DJ et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017; 376: 1015-1026. 20. Peters S, Gettinger S, Johnson ML et al. Phase II Trial of Atezolizumab As FirstLine or Subsequent Therapy for Patients With Programmed Death-Ligand 1-Selected Advanced Non-Small-Cell Lung Cancer (BIRCH). J Clin Oncol 2017; JCO2016719476. 21. Balar AV, Galsky MD, Rosenberg JE 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. 22. Rosenberg JE, Hoffman-Censits J, Powles T 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. 23. Fehrenbacher L, Spira A, Ballinger M 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. 24. Rittmeyer A, Barlesi F, Waterkamp D 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. 25. Kaufman HL, Russell J, Hamid O et al. Avelumab in patients with chemotherapyrefractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 2016; 17: 1374-1385. 26. Apolo AB, Infante JR, Balmanoukian A et al. Avelumab, an Anti-Programmed Death-Ligand 1 Antibody, In Patients With Refractory Metastatic Urothelial Carcinoma: Results From a Multicenter, Phase Ib Study. J Clin Oncol 2017; 35: 2117-2124. 27. Massard C, Gordon MS, Sharma S et al. Safety and Efficacy of Durvalumab (MEDI4736), an Anti-Programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients With Advanced Urothelial Bladder Cancer. J Clin Oncol 2016; 34: 3119-3125. 28. Powles T, O'Donnell PH, Massard C 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; e172411. 29. Rotte A, Bhandaru M, Zhou Y, McElwee KJ. Immunotherapy of melanoma: present options and future promises. Cancer Metastasis Rev 2015; 34: 115-128. 30. Tarhini A. Immune-mediated adverse events associated with ipilimumab ctla-4 blockade therapy: the underlying mechanisms and clinical management. Scientifica (Cairo) 2013; 2013: 857519. 31. Michot JM, Bigenwald C, Champiat S et al. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer 2016; 54: 139-148. 32. Postow MA, Chesney J, Pavlick AC et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015; 372: 2006-2017. 33. Larkin J, Chiarion-Sileni V, Gonzalez R et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015; 373: 23-34.

17

34. Champiat S, Lambotte O, Barreau E et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol 2016; 27: 559574. 35. Johnson DB, Balko JM, Compton ML et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N Engl J Med 2016; 375: 1749-1755. 36. Hellmann MD, Rizvi NA, Goldman JW et al. Nivolumab plus ipilimumab as firstline treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol 2017; 18: 31-41. 37. Antonia SJ, Lopez-Martin JA, Bendell J et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016; 17: 883-895. 38. Long GV, Atkinson V, Cebon JS et al. Standard-dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial. Lancet Oncol 2017. 39. Ribas A, Martín-Algarra S, Bhatia S et al. Abstract CT073: Immunomodulatory effects of nivolumab and ipilimumab in combination or nivolumab monotherapy in advanced melanoma patients: CheckMate 038. In AACR Annual Meeting 2017. Washington, DC: American Association for Cancer Research 2017. 40. Antonia S, Goldberg SB, Balmanoukian A et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol 2016; 17: 299-308. 41. Hammers HJ, Plimack ER, Infante JR et al. Safety and Efficacy of Nivolumab in Combination With Ipilimumab in Metastatic Renal Cell Carcinoma: The CheckMate 016 Study. J Clin Oncol 2017; JCO2016721985. 42. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252-264. 43. Intlekofer AM, Thompson CB. At the bench: preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. J Leukoc Biol 2013; 94: 25-39. 44. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016; 44: 9891004. 45. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol 2016; 39: 98-106. 46. Granier C, De Guillebon E, Blanc C et al. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO Open 2017; 2: e000213. 47. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12: 265-277. 48. Combadiere B, Reis e Sousa C, Trageser C et al. Differential TCR signaling regulates apoptosis and immunopathology during antigen responses in vivo. Immunity 1998; 9: 305-313. 49. Montoya MC, Sancho D, Bonello G et al. Role of ICAM-3 in the initial interaction of T lymphocytes and APCs. Nat Immunol 2002; 3: 159-168. 50. Wang H, Kadlecek TA, Au-Yeung BB et al. ZAP-70: an essential kinase in T-cell signaling. Cold Spring Harb Perspect Biol 2010; 2: a002279. 51. Guy CS, Vignali KM, Temirov J et al. Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. Nat Immunol 2013; 14: 262-270.

18

52. Thaker YR, Schneider H, Rudd CE. TCR and CD28 activate the transcription factor NF-kappaB in T-cells via distinct adaptor signaling complexes. Immunol Lett 2015; 163: 113-119. 53. Chakraborty AK, Weiss A. Insights into the initiation of TCR signaling. Nat Immunol 2014; 15: 798-807. 54. Navarro MN, Cantrell DA. Serine-threonine kinases in TCR signaling. Nat Immunol 2014; 15: 808-814. 55. Kass I, Buckle AM, Borg NA. Understanding the structural dynamics of TCRpMHC complex interactions. Trends Immunol 2014; 35: 604-612. 56. Shi JH, Sun SC. TCR signaling to NF-kappaB and mTORC1: Expanding roles of the CARMA1 complex. Mol Immunol 2015; 68: 546-557. 57. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol 2009; 27: 591-619. 58. Ley K. The second touch hypothesis: T cell activation, homing and polarization. F1000Res 2014; 3: 37. 59. Thommen DS, Schreiner J, Muller P et al. Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors. Cancer Immunol Res 2015; 3: 1344-1355. 60. Webb JR, Milne K, Nelson BH. PD-1 and CD103 Are Widely Coexpressed on Prognostically Favorable Intraepithelial CD8 T Cells in Human Ovarian Cancer. Cancer Immunol Res 2015; 3: 926-935. 61. Zhang L, Wang J, Wei F et al. Profiling the dynamic expression of checkpoint molecules on cytokine-induced killer cells from non-small-cell lung cancer patients. Oncotarget 2016; 7: 43604-43615. 62. Djenidi F, Adam J, Goubar A et al. CD8+CD103+ tumor-infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients. J Immunol 2015; 194: 3475-3486. 63. Gaillard S, Dumbauld C, Bilewski A et al. Evaluating the repertoire of immune checkpoint markers expressed on peripheral and ascites CD8+ T cells in ovarian cancer. In ASCO annual meeting. Chicago: American Society of Clinical Oncology 2017. 64. Chen J, Chen Z. The effect of immune microenvironment on the progression and prognosis of colorectal cancer. Med Oncol 2014; 31: 82. 65. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P et al. Tumor-infiltrating NY-ESO1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A 2010; 107: 7875-7880. 66. Kahan SM, Wherry EJ, Zajac AJ. T cell exhaustion during persistent viral infections. Virology 2015; 479-480: 180-193. 67. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol 2015; 36: 265-276. 68. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015; 15: 486-499. 69. Huang AC, Postow MA, Orlowski RJ et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017; 545: 60-65. 70. Kamphorst AO, Wieland A, Nasti T et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 2017; 355: 1423-1427.

19

71. Philip M, Fairchild L, Sun L et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 2017. 72. Brunet JF, Denizot F, Luciani MF et al. A new member of the immunoglobulin superfamily--CTLA-4. Nature 1987; 328: 267-270. 73. Tivol EA, Borriello F, Schweitzer AN et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995; 3: 541-547. 74. Chambers CA, Sullivan TJ, Allison JP. Lymphoproliferation in CTLA-4-deficient mice is mediated by costimulation-dependent activation of CD4+ T cells. Immunity 1997; 7: 885-895. 75. Stamper CC, Zhang Y, Tobin JF et al. Crystal structure of the B7-1/CTLA-4 complex that inhibits human immune responses. Nature 2001; 410: 608-611. 76. Perkins D, Wang Z, Donovan C et al. Regulation of CTLA-4 expression during T cell activation. J Immunol 1996; 156: 4154-4159. 77. Alegre ML, Noel PJ, Eisfelder BJ et al. Regulation of surface and intracellular expression of CTLA4 on mouse T cells. J Immunol 1996; 157: 4762-4770. 78. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003; 299: 1057-1061. 79. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003; 4: 330-336. 80. Gavin MA, Rasmussen JP, Fontenot JD et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature 2007; 445: 771-775. 81. Wing K, Onishi Y, Prieto-Martin P et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008; 322: 271-275. 82. Rotte A, Bhandaru M. Ipilimumab. Switzerland: Springer International Publishing,2016. 83. Carreno BM, Bennett F, Chau TA et al. CTLA-4 (CD152) can inhibit T cell activation by two different mechanisms depending on its level of cell surface expression. J Immunol 2000; 165: 1352-1356. 84. Fraser JH, Rincon M, McCoy KD, Le Gros G. CTLA4 ligation attenuates AP-1, NFAT and NF-kappaB activity in activated T cells. Eur J Immunol 1999; 29: 838-844. 85. Chikuma S, Abbas AK, Bluestone JA. B7-independent inhibition of T cells by CTLA-4. J Immunol 2005; 175: 177-181. 86. Schneider H, Valk E, Leung R, Rudd CE. CTLA-4 activation of phosphatidylinositol 3-kinase (PI 3-K) and protein kinase B (PKB/AKT) sustains T-cell anergy without cell death. PLoS One 2008; 3: e3842. 87. Olsson C, Riesbeck K, Dohlsten M, Michaelsson E. CTLA-4 ligation suppresses CD28-induced NF-kappaB and AP-1 activity in mouse T cell blasts. J Biol Chem 1999; 274: 14400-14405. 88. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992; 11: 3887-3895. 89. Nishimura H, Nose M, Hiai H et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999; 11: 141-151.

20

90. Nishimura H, Okazaki T, Tanaka Y et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001; 291: 319-322. 91. Nishimura H, Honjo T. PD-1: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol 2001; 22: 265-268. 92. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008; 26: 677-704. 93. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunological Reviews 2010; 236: 219-242. 94. Rotte A, Bhandaru M. Nivolumab. Switzerland: Springer International Publishing,2016. 95. Cheng X, Veverka V, Radhakrishnan A et al. Structure and interactions of the human programmed cell death 1 receptor. J Biol Chem 2013; 288: 11771-11785. 96. Latchman Y, Wood CR, Chernova T et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2001; 2: 261-268. 97. Francisco LM, Salinas VH, Brown KE et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 2009; 206: 30153029. 98. Gordon SR, Maute RL, Dulken BW et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017. 99. Chemnitz JM, Parry RV, Nichols KE et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 2004; 173: 945-954. 100. FDA. FDA approves first cancer treatment for any solid tumor with a specific genetic feature. In. FDA 2017. 101. Le DT, Durham JN, Smith KN et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017. 102. FDA. FDA grants nivolumab accelerated approval for MSI-H or dMMR colorectal cancer. In. FDA 2017. 103. Yu X, Harden K, Gonzalez LC et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 2009; 10: 48-57. 104. Boles KS, Vermi W, Facchetti F et al. A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC. Eur J Immunol 2009; 39: 695-703. 105. Stanietsky N, Simic H, Arapovic J et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A 2009; 106: 17858-17863. 106. Johnston RJ, Comps-Agrar L, Hackney J et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell 2014; 26: 923-937. 107. Kurtulus S, Sakuishi K, Ngiow SF et al. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest 2015; 125: 4053-4062. 108. Chew GM, Fujita T, Webb GM et al. TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PLoS Pathog 2016; 12: e1005349.

21

109. Fuchs A, Cella M, Giurisato E et al. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J Immunol 2004; 172: 3994-3998. 110. Gilfillan S, Chan CJ, Cella M et al. DNAM-1 promotes activation of cytotoxic lymphocytes by nonprofessional antigen-presenting cells and tumors. J Exp Med 2008; 205: 2965-2973. 111. Chan CJ, Martinet L, Gilfillan S et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol 2014; 15: 431438. 112. Manieri NA, Chiang EY, Grogan JL. TIGIT: A Key Inhibitor of the Cancer Immunity Cycle. Trends Immunol 2017; 38: 20-28. 113. Joller N, Lozano E, Burkett PR et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 2014; 40: 569-581. 114. Liu S, Zhang H, Li M et al. Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ 2013; 20: 456-464. 115. Li M, Xia P, Du Y et al. T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus receptor (PVR) ligand engagement suppresses interferon-gamma production of natural killer cells via beta-arrestin 2-mediated negative signaling. J Biol Chem 2014; 289: 17647-17657. 116. Gur C, Ibrahim Y, Isaacson B et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015; 42: 344-355. 117. Monney L, Sabatos CA, Gaglia JL et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002; 415: 536-541. 118. Gorman JV, Colgan JD. Regulation of T cell responses by the receptor molecule Tim-3. Immunol Res 2014; 59: 56-65. 119. Zhu C, Anderson AC, Schubart A et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 2005; 6: 1245-1252. 120. Sabatos CA, Chakravarti S, Cha E et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol 2003; 4: 1102-1110. 121. Sanchez-Fueyo A, Tian J, Picarella D et al. Tim-3 inhibits T helper type 1mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol 2003; 4: 1093-1101. 122. Sehrawat S, Suryawanshi A, Hirashima M, Rouse BT. Role of Tim-3/galectin-9 inhibitory interaction in viral-induced immunopathology: shifting the balance toward regulators. J Immunol 2009; 182: 3191-3201. 123. Dardalhon V, Anderson AC, Karman J et al. Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J Immunol 2010; 185: 1383-1392. 124. Sakuishi K, Apetoh L, Sullivan JM et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010; 207: 21872194.

22

125. Fourcade J, Sun Z, Benallaoua M et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med 2010; 207: 2175-2186. 126. Baitsch L, Legat A, Barba L et al. Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization. PLoS One 2012; 7: e30852. 127. Ngiow SF, von Scheidt B, Akiba H et al. Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors. Cancer Res 2011; 71: 3540-3551. 128. Anderson AC. Tim-3: an emerging target in the cancer immunotherapy landscape. Cancer Immunol Res 2014; 2: 393-398. 129. Triebel F, Jitsukawa S, Baixeras E et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med 1990; 171: 1393-1405. 130. Liu W, Tang L, Zhang G et al. Characterization of a novel C-type lectin-like gene, LSECtin: demonstration of carbohydrate binding and expression in sinusoidal endothelial cells of liver and lymph node. J Biol Chem 2004; 279: 18748-18758. 131. Tang L, Yang J, Liu W et al. Liver sinusoidal endothelial cell lectin, LSECtin, negatively regulates hepatic T-cell immune response. Gastroenterology 2009; 137: 14981508 e1491-1495. 132. Xu F, Liu J, Liu D et al. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res 2014; 74: 3418-3428. 133. Kouo T, Huang L, Pucsek AB et al. 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. 134. Yang J, Wang H, Wang M et al. Involvement of LSECtin in the hepatic natural killer cell response. Biochem Biophys Res Commun 2016; 476: 49-55. 135. Miyazaki T, Dierich A, Benoist C, Mathis D. LAG-3 is not responsible for selecting T helper cells in CD4-deficient mice. Int Immunol 1996; 8: 725-729. 136. Workman CJ, Cauley LS, Kim IJ et al. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol 2004; 172: 5450-5455. 137. Bettini M, Szymczak-Workman AL, Forbes K et al. Cutting edge: accelerated autoimmune diabetes in the absence of LAG-3. J Immunol 2011; 187: 3493-3498. 138. Jha V, Workman CJ, McGaha TL et al. Lymphocyte Activation Gene-3 (LAG-3) negatively regulates environmentally-induced autoimmunity. PLoS One 2014; 9: e104484. 139. Richter K, Agnellini P, Oxenius A. On the role of the inhibitory receptor LAG-3 in acute and chronic LCMV infection. Int Immunol 2010; 22: 13-23. 140. Hannier S, Tournier M, Bismuth G, Triebel F. CD3/TCR complex-associated lymphocyte activation gene-3 molecules inhibit CD3/TCR signaling. J Immunol 1998; 161: 4058-4065. 141. Huang CT, Workman CJ, Flies D et al. Role of LAG-3 in regulatory T cells. Immunity 2004; 21: 503-513. 142. Gagliani N, Magnani CF, Huber S et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med 2013; 19: 739-746.

23

143. He Y, Rivard CJ, Rozeboom L et al. Lymphocyte-activation gene-3, an important immune checkpoint in cancer. Cancer Sci 2016; 107: 1193-1197. 144. Andrews LP, Marciscano AE, Drake CG, Vignali DA. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev 2017; 276: 80-96. 145. Ascierto PA, Melero I, Bhatia S et al. Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. In ASCO Annual Meeting. Chicago: American Society of Clinical Oncology 2017. 146. Ascierto PA, Bono P, Bhatia S et al. Efficacy of BMS-986016, a Monoclonal Antibody That Targets LAG-3, in Combination With Nivolumab in Pts With Melanoma Who Progressed During Prior Anti–PD-1/PD-L1 Therapy (mel prior IO) in All-Comer and Biomarker-Enriched Populations. In ESMO. Annals of Oncology 2017; V605-649. 147. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 2017; 541: 321-330. 148. Lawrence MS, Stojanov P, Polak P et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013; 499: 214-218. 149. Alexandrov LB, Nik-Zainal S, Wedge DC et al. Deciphering signatures of mutational processes operative in human cancer. Cell Rep 2013; 3: 246-259. 150. Alexandrov LB, Nik-Zainal S, Wedge DC et al. Signatures of mutational processes in human cancer. Nature 2013; 500: 415-421. 151. Alexandrov LB, Stratton MR. Mutational signatures: the patterns of somatic mutations hidden in cancer genomes. Curr Opin Genet Dev 2014; 24: 52-60. 152. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015; 348: 69-74. 153. Rizvi NA, Hellmann MD, Snyder A et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124-128. 154. Snyder A, Nathanson T, Funt SA et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis. PLoS Med 2017; 14: e1002309. 155. Wallin JJ, Bendell JC, Funke R et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun 2016; 7: 12624. 156. Powles T, McDermott DF, Rini B et al. Novel Radiological Endpoints and Updated Data From a Randomized Phase II Trial Investigating Atezolizumab With or Without Bevacizumab vs Sunitinib in Untreated Metastatic Renal Cell Carcinoma. In ESMO. Annals of Oncology 2017; V605-v649. 157. Robbins PF, Lu YC, El-Gamil M et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013; 19: 747-752. 158. van Rooij N, van Buuren MM, Philips D et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013; 31: e439-442. 159. Snyder A, Makarov V, Merghoub T et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014; 371: 2189-2199.

24

160. Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest 2015; 125: 3413-3421. 161. Linnemann C, van Buuren MM, Bies L et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat Med 2015; 21: 81-85. 162. Alexander GS, Palmer JD, Tuluc M et al. Immune biomarkers of treatment failure for a patient on a phase I clinical trial of pembrolizumab plus radiotherapy. J Hematol Oncol 2016; 9: 96. 163. Manson G, Norwood J, Marabelle A et al. Biomarkers associated with checkpoint inhibitors. Ann Oncol 2016; 27: 1199-1206. 164. Martens A, Wistuba-Hamprecht K, Geukes Foppen M et al. Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22: 2908-2918. 165. Koh YW, Choi JH, Ahn MS et al. Baseline neutrophil-lymphocyte ratio is associated with baseline and subsequent presence of brain metastases in advanced nonsmall-cell lung cancer. Sci Rep 2016; 6: 38585. 166. Martens A, Wistuba-Hamprecht K, Yuan J et al. Increases in Absolute Lymphocytes and Circulating CD4+ and CD8+ T Cells Are Associated with Positive Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22: 4848-4858. 167. Ferrucci PF, Ascierto PA, Pigozzo J et al. Baseline neutrophils and derived neutrophil-to-lymphocyte ratio: prognostic relevance in metastatic melanoma patients receiving ipilimumab. Ann Oncol 2017. 168. Cassidy MR, Wolchok RE, Zheng J et al. Neutrophil to Lymphocyte Ratio is Associated With Outcome During Ipilimumab Treatment. EBioMedicine 2017; 18: 5661.

25

Table Legends Table 1. List of FDA approved drugs that target T-cell checkpoints as of September 20, 2017 Table 2. Checkpoints of T-cell activation Table 3. Differences in effects of checkpoint blockade

Figure Legends Figure 1. Milestones in the clinical development of checkpoint blockers. Source: references 1, 3, 5, 7, 8, 10, 11, 21, 22, 23, 24, 25, 28, 32, 33, 100, 101 & 102

Figure 2. Regulation of T-cell activation. The outcome of T-cell priming in lymph node depends on the degree of TCR activation and on the levels of costimulatory/checkpoint receptor expression on T-cells. Optimal TCR activation results in minimal CTLA-4 and PD-1 expression (checkpoints) and maximal CD28 expression (costimulatory receptor), driving the T-cell towards an effector phenotype. Hyperactivation of TCR during priming induces expression of CTLA-4 and/or PD-1 and drives the T-cells into anergy, dysfunction or differentiation into TRegs depending on the checkpoint receptor and the naïve T-cell sub-type. Similarly, peripheral interaction between effector T-cells and other immune cells such as DCs, macrophages, B-cells and Th1 cells can also result in amplification or dampening of the effector responses depending on the TCR-antigen interaction and the subsequent costimulatory or checkpoint receptor activation.

Figure 3. Checkpoints in regulation of immune response. The distinct role checkpoint receptors CTLA-4, PD-1, TIGIT, TIM-3 and LAG-3 in regulation of T-cell responses are shown in the figure. (+) sign indicates positive regulation of the flow of reaction and (-) sign indicates negative regulation of the flow of reaction. Amplification of effector function and induction of dysfunctional phenotype is indicated (arbitrarily) using corresponding darker shade of the color. Th1 cells promote formation of mature dendritic cells (mDCs) whereas CTLA-4 and TIGIT induce formation of tolerogenic DCs (tDCs) in lymph nodes (represented as lymph) and peripheral tissues (peripheral sites) and

26

reduce the number of mature DCs (mDCs). The differentiation of naïve CD4 and CD8 Tcells into Th1 and CTLs, and amplification of Th1/CTL effector functions are promoted by costimulation. PD-1 promotes priming of TRegs, T-effector cell dysfunction and inhibition of NK cell effector functions. CTLA-4 induces anergy during priming of naïve CD4/CD8 T-cells and also favor T-effector cell dysfunction. TIGIT, TIM-3 and LAG-3 promote T-effector cell dysfunction and inhibit NK cell effector functions.

Supplementary Tables Table S1. Checkpoint targeting monoclonal antibodies in development Table S2. Checkpoints and costimulators of T-cell activity Table S3. Efficacy of checkpoint combinations Table S4. Commonly tested biomarkers that aid in the understanding of immune response in TME

27

Figure 1 180x113mm (300 x 300 DPI)

Figure 2 180x180mm (300 x 300 DPI)

Figure 3 180x192mm (300 x 300 DPI)

Table 1: List of FDA approved drugs that target T-cell checkpoints as of September 20, 2017 Drug; Approved indications Recommended dose & Reference Brand name; Route Marketed by CTLA-4 Blockers Ipilimumab; Melanoma Metastatic disease: not more [1-3] Yervoy; than 4 doses of 3 mg/kg IV for Bristol-Myers every 3 weeks Squibb Adjuvant setting: 4 doses of 10 mg/kg IV for every 3 weeks, followed by 10 mg/kg IV for every 12 weeks, for up to 3 years PD-1 Blockers Nivolumab; Metastatic melanoma, For melanoma, NSCLC, RCC, [4], [5], Opdivo; Metastatic Non-small cell metastatic urothelial [10], [11], Bristol-Myers lung cancer (NSCLC), carcinoma, HCC & colorectal [13-15], Squibb Renal cell carcinoma cancer with MSI-H and MMR [17], [18] & (RCC), aberrations: 240 mg IV [102] Classical Hodgkins infusion for every 2 weeks until lymphoma, disease progression or toxicity Head and Neck Squamous cell carcinoma (HNSCC), For Classical Hodgkins Metastatic urothelial lymphoma & HNSCC: 3 mg/kg Carcinoma IV infusion for every 2 weeks Hepatocellular carcinoma until disease progression or (HCC) toxicity Colorectal cancer with MSIH and MMR aberrations Pembrolizumab; Metastatic melanoma, 200 mg IV infusion for every 3 [6-9], [12], Keytruda; Merck Metastatic NSCLC, weeks until disease [16], [19], Classical Hodgkins progression, toxicity or up to [100] & lymphoma, 24 months [101] HNSCC Gastric cancer Solid tumors with MSI-H and MMR aberrations Metastatic urothelial carcinoma PD-L1 Blockers Atezolizumab; Metastatic urothelial 1200 mg IV infusion for every [19-23] Tecentriq; carcinoma, Metastatic 3 weeks until disease Genentech/Roche NSCLC progression or toxicity Avelumab; Merkel cell carcinoma, 10 mg/kg IV infusion for every [26] Bavencio; Metastatic urothelial 2 weeks until disease Pfizer carcinoma progression or toxicity Durvalumab Imfinzi; Astrazeneca

Metastatic urothelial carcinoma

10 mg/kg IV infusion for every 2 weeks until disease progression or toxicity

[27] & [28]

Table 2: Checkpoints of T-cell activation Receptor CTLA-4 PD-1

TIGIT

TIM-3

LAG-3

Synonyms

CD152

WUCAM

HAVCR2

CD223

Gene location

Chromosome 2q33

PDCD1 & CD279 Chromosome 2q37.3

Chromosome 3q13.31

Chromosome 5q33.3

Chromosome 12p13.32

Number of amino acids

223-amino acids

288-amino acids

244-amino acids

301-amino acids

498 aminoacids

Signaling motif

Cytoplasmic tail

ITSM

ITT-ITIM

KIEELE motif in the cytoplasmic tail

Ligands

CD80 & CD86

PD-L1 & PD-L2

PVR/ CD155 & CD112

Cells expressing receptor

Activated Tcells, TRegs, exhausted effector T cells

Activated T-cells, TRegs, NK cells, macrophages & exhausted effector T cells

Cells expressing Ligand

APCs

Reference

[72-80]

APCs, hematopoetic & nonhematopoetic cells & tumor cells [70], [88] & [9298]

Activated Tcells, memory Tcells, TFH cells, TRegs, Tr1 cells, NK cells, NKT cells & exhausted effector T cells APCs, fibroblasts, endothelial cells & tumor cells [103-112]

Tyrosine residues in the cytoplasmic tail Galectin-9, Ceacam-1, HMGB-1 & phosphatidyl serine Activated T cells, TH17 cells, TRegs, DCs, NK cells, monocytes & exhausted effector T cells

APCs, tumor cells

[117-126]

MHC II class, LSECtin, Galectin-3

Activated CD4+ T-cells, TRegs, Tr1 cells, activated + CD8 T-cells, NK cells, DCs, B-cells & exhausted effector Tcells APCs Liver cells and tumor cells [129-134], [143] & [144]

Table 3 Differences in effects of checkpoint blockade Check- AutoimmPhases of Cells effected points une immune by checkpoint Phenotype response blockade of knockeffected by out mice checkpoint blockade CTLA-4

Spontaneo us & lethal; mice die within 3-4 weeks of age

Mainly early stage activation in lymph nodes; also effects exhaustion & activation in tissues Mainly activation in tissues, exhaustion & also activation in lymph nodes

PD-1

Spontaneo us but less severe

TIGIT

Mild; not spontaneo us

Exhaustion & activation in tissues

TIM-3

Mild; not spontaneo us

Exhaustion & activation in tissues

LAG-3

Mild; not spontaneo us

Exhaustion & activation in tissue

Activated Tcells in lymph nodes & periphery, DCs & Exhausted effector T-cells

Activated Tcells in lymph nodes & periphery, TRegs, macrophages, Exhausted effector T-cells, & NK cells DC phenotype, TRegs, Activated Tcells in periphery, Exhausted effector T-cells & NK cells DC phenotype, MDSCs, TRegs, Activated Tcells in periphery, Exhausted effector T-cells & NK cells TRegs, Activated Tcells in periphery, Exhausted effector T-cells & NK cells

Expected stimulation of immune response with checkpoint blockade +++

Expected safety of checkpoint blockade

Reference

+

[7274] & [81]

++

++

[8991], [96] & [97]

+

+++

[103108] & [113]

+

+++

[118], [120] & [121123]

+

+++

[135140]