The blockade of immune checkpoints in cancer immunotherapy

Drew M Pardoll, Drew M Pardoll

Abstract

Among the most promising approaches to activating therapeutic antitumour immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumours co-opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumour antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibodies were the first of this class of immunotherapeutics to achieve US Food and Drug Administration (FDA) approval. Preliminary clinical findings with blockers of additional immune-checkpoint proteins, such as programmed cell death protein 1 (PD1), indicate broad and diverse opportunities to enhance antitumour immunity with the potential to produce durable clinical responses.

Figures

Figure 1. Multiple co-stimulatory and inhibitory interactions…
Figure 1. Multiple co-stimulatory and inhibitory interactions regulate T cell responses
Depicted are various ligand–receptor interactions between T cells and antigen-presenting cells (APCs) that regulate the T cell response to antigen (which is mediated by peptide–major histocompatibility complex (MHC) molecule complexes that are recognized by the T cell receptor (TCR)). These responses can occur at the initiation of T cell responses in lymph nodes (where the major APCs are dendritic cells) or in peripheral tissues or tumours (where effector responses are regulated). In general, T cells do not respond to these ligand–receptor interactions unless they first recognize their cognate antigen through the TCR. Many of the ligands bind to multiple receptors, some of which deliver co-stimulatory signals and others deliver inhibitory signals. In general, pairs of co-stimulatory–inhibitory receptors that bind the same ligand or ligands — such as CD28 and cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) — display distinct kinetics of expression with the co-stimulatory receptor expressed on naive and resting T cells, but the inhibitory receptor is commonly upregulated after T cell activation. One important family of membrane-bound ligands that bind both co-stimulatory and inhibitory receptors is the B7 family. All of the B7 family members and their known ligands belong to the immunoglobulin superfamily. Many of the receptors for more recently identified B7 family members have not yet been identified. Tumour necrosis factor (TNF) family members that bind to cognate TNF receptor family molecules represent a second family of regulatory ligand–receptor pairs. These receptors predominantly deliver co-stimulatory signals when engaged by their cognate ligands. Another major category of signals that regulate the activation of T cells comes from soluble cytokines in the microenvironment. Communication between T cells and APCs is bidirectional. In some cases, this occurs when ligands themselves signal to the APC. In other cases, activated T cells upregulate ligands, such as CD40L, that engage cognate receptors on APCs. A2aR, adenosine A2a receptor; B7RP1, B7-related protein 1; BTLA, B and T lymphocyte attenuator; GAL9, galectin 9; HVEM, herpesvirus entry mediator; ICOS, inducible T cell co-stimulator; IL, interleukin; KIR, killer cell immunoglobulin-like receptor; LAG3, lymphocyte activation gene 3; PD1, programmed cell death protein 1; PDL, PD1 ligand; TGFβ, transforming growth factor-β; TIM3, T cell membrane protein 3.
Figure 2. Clinical responses and immune-mediated toxicities…
Figure 2. Clinical responses and immune-mediated toxicities on antibody blockade of the CTLA4-mediated immune checkpoint
Depicted on the left of the figure are examples of regressions of lung (top two panels) and brain (lower panel) metastases in a patient with melanoma who was treated with the cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibody, ipilimumab. 25–30% of patients treated with extended doses of anti-CTLA4 therapy can develop immune-related ‘on-target’ toxicities. However, the frequency of severe adverse toxicities was lower (10–15%) with the short course that was used in the Phase III trial that led to the approval of ipilimumab. This short-course regimen (4 doses at a cost of US$30,000 per dose) was recommended by the US Food and Drug Administration (FDA). As shown on the right of the figure, common tissues affected by immune-related toxicities from treatment with anti-CTLA4 therapy include the skin (dermatitis) and the colon (colitis). Tissues that do not undergo such rapid regeneration as the skin and colon, such as lung and liver and the pituitary and thyroid glands, are less frequently affected. Immune toxicities from anti-CTLA4 therapy are usually successfully mitigated by treatment with systemic steroids and tumour necrosis factor (TNF) blockers when systemic steroids are not effective. Ongoing tumour responses typically continue even after a course of steroids. Figure is reproduced, with permission, from REF. 39 © (2003) National Academy of Sciences, USA.
Figure 3. Immune checkpoints regulate different components…
Figure 3. Immune checkpoints regulate different components in the evolution of an immune response
a | The cytotoxic T-lymphocyte-associated antigen 4 (CTLA4)-mediated immune checkpoint is induced in T cells at the time of their initial response to antigen. The level of CTLA4 induction depends on the amplitude of the initial T cell receptor (TCR)-mediated signalling. High-affinity ligands induce higher levels of CTLA4, which dampens the amplitude of the initial response. The key to the regulation of T cell activation levels by the CD28–CTLA4 system is the timing of surface expression. Naive and memory T cells express high levels of cell surface CD28 but do not express CTLA4 on their surface. Instead, CTLA4 is sequestered in intracellular vesicles. After the TCR is triggered by antigen encounter, CTLA4 is transported to the cell surface. The stronger the stimulation through the TCR (and CD28), the greater the amount of CTLA4 that is deposited on the T cell surface. Therefore, CTLA4 functions as a signal dampener to maintain a consistent level of T cell activation in the face of widely varying concentrations and affinities of ligand for the TCR. b | By contrast, the major role of the programmed cell death protein 1 (PD1) pathway is not at the initial T cell activation stage but rather to regulate inflammatory responses in tissues by effector T cells recognizing antigen in peripheral tissues. Activated T cells upregulate PD1 and continue to express it in tissues. Inflammatory signals in the tissues induce the expression of PD1 ligands, which downregulate the activity of T cells and thus limit collateral tissue damage in response to a microorganism infection in that tissue. The best characterized signal for PD1 ligand 1 (PDL1; also known as B7-H1) induction is interferon-γ (IFNγ), which is predominantly produced by T helper 1 (TH1) cells, although many of the signals have not yet been defined completely. Excessive induction of PD1 on T cells in the setting of chronic antigen exposure can induce an exhausted or anergic state in T cells. MHC, major histocompatibility complex.
Figure 4. Two general mechanisms of expression…
Figure 4. Two general mechanisms of expression of immune-checkpoint ligands on tumour cells
The examples in this figure use the programmed cell death protein 1 (PD1) ligand, PDL1 (also known as B7-H1), for illustrative purposes, although the concept probably applies to multiple immune-checkpoint ligands, including PDL2 (also known as B7-DC). a | Innate immune resistance. In some tumours, constitutive oncogenic signalling can upregulate PDL1 expression on all tumour cells, independently of inflammatory signals in the tumour microenvironment. Activation of the AKT and signal transducer and activator of transcription 3 (STAT3) pathways has been reported to drive PDL1 expression. b | Adaptive immune resistance. In some tumours, PDL1 is not constitutively expressed, but rather it is induced in response to inflammatory signals that are produced by an active antitumour immune response. The non-uniform expression of PDL1, which is commonly restricted to regions of the tumour that have tumour-infiltrating lymphocytes, suggests that PDL1 is adaptively induced as a consequence of immune responses within the tumour microenvironment. Adaptive induction may be a common mechanism for the expression of multiple immune-checkpoint molecules in tumours. IFNγ, interferon-γ; MHC, major histocompatibility complex; TCR, T cell receptor.
Figure 5. Implications of the adaptive immune…
Figure 5. Implications of the adaptive immune resistance mechanism for combinatorial immunotherapy of cancer
The adaptive immune resistance mechanism implies that the blockade of an induced immune-checkpoint protein, such as programmed cell death protein 1 (PD1), as a single intervention will only induce tumour regressions when there is a pre-existing antitumour immune response to be ‘unleashed’ when the pathway is blocked. Multiple interventions, such as vaccines, that activate a de novo antitumour immune response may not induce tumour regressions because tumours respond by upregulating immune-checkpoint ligands. Therefore, combining the two approaches may induce tumour regressions in patients that would not have responded to either treatment alone. PDL1, PD1 ligand 1; TAM, tumour-associated macrophage.

Source: PubMed

3
Tilaa