Journal list menu

Volume 283, Issue 12 p. 2233-2244
State-of-the-Art Review
Free Access

Immune checkpoint blockade in lymphoid malignancies

Gita Thanarajasingam

Gita Thanarajasingam

Division of Hematology, Mayo Clinic, Rochester, MN, USA

Search for more papers by this author
Uma Thanarajasingam

Uma Thanarajasingam

Division of Rheumatology, Mayo Clinic, Rochester, MN, USA

Search for more papers by this author
Stephen M. Ansell

Corresponding Author

Stephen M. Ansell

Division of Hematology, Mayo Clinic, Rochester, MN, USA

Correspondence

S. M. Ansell, Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA

Fax: +1 507 266 4972

Tel: +1 507 284 2511

E-mail: [email protected]

Search for more papers by this author
First published: 25 January 2016
Citations: 17

Abstract

Malignant cells may subvert and escape endogenous host immune surveillance by up-regulation of immune inhibitory signals known as immune checkpoints. These checkpoints are important therapeutic targets, and antibodies that block checkpoint signaling have shown remarkable efficacy in some solid tumors as well as in some refractory hematologic malignancies. In hematologic cancers, the mechanism of these checkpoints is complex, as the tumor and immune system are one and the same. In this review, we evaluate the biology of checkpoint inhibition, review the current data on its efficacy in lymphoid tumors, and explore uncertainties in the field, including those involving the precise mechanisms of action, the appropriate timing of therapy, and the differences in response rate between lymphoid tumor types.

Abbreviations

  • cHL
  • classical Hodgkin lymphoma
  • CLL
  • chronic lymphocytic leukemia
  • CTLA4
  • cytotoxic T lymphocyte-associated antigen 4
  • DLBCL
  • diffuse large B cell lymphoma
  • FL
  • follicular lymphoma
  • HL
  • Hodgkin lymphoma
  • NHL
  • non-Hodgkin lymphoma
  • NOS
  • not otherwise specified
  • PD-1
  • programmed cell death-1
  • PD-L1
  • programmed cell death ligand 1
  • PD-L2
  • programmed cell death ligand 2
  • Introduction

    Immune therapies are rapidly becoming an important part of cancer treatment for both solid tumors and hematologic malignancies due to advances in our understanding of the intrinsic antitumor activity of the immune system. Immunotherapies affect not only malignant cells but incite the host immune cells to target the tumor. Hematologic neoplasms are distinguished from solid tumors in that the malignancy arises from the immune system itself. The vast majority of adult lymphoid malignancies originate from mature B cells, with a minor proportion arising from mature T cells [1]. The role of immune pathways in these tumors is therefore complex. Immunological ‘checkpoints’ are inhibitory feedback loops that down-regulate the magnitude of immune responses to protect the host from autoimmunity or damage from inflammation. This mechanism of keeping the immune response contained is frequently subverted by malignant cells, which escape immune surveillance by increasing inhibitory immune checkpoint ligands, leading to host T cell exhaustion. Therefore, these immune checkpoints have become important therapeutic targets, with US Food and Drug Administration-approved agents for the treatment of advanced solid tumors and very promising results in studies of patients with hematologic malignancies.

    Immune checkpoint inhibitors enhance the cytotoxic activity of host T cells by blocking inhibitory signals from tumor cells. Rather than targeting the cancer cell directly, these agents stimulate the host immune system to exert an antitumor effect [2]. The most clinically relevant checkpoints to date are cytotoxic T lymphocyte-associated antigen 4 (CTLA4; CD152) and programmed cell death 1 (PD-1; CD279). In lymphoid malignancies, these inhibitory pathways resemble their state in the native immune system, and differences may be mediated in part by the tumor microenvironment. Here we review immune checkpoint inhibition in lymphoid malignancies, including the underlying biology and the efficacy of available therapeutic agents, as well as uncertainties in the field of checkpoint blockade for treatment of lymphoid tumors.

    Biology of immune checkpoints

    An effective immune response in malignancy is similar to that seen in states of infection or inflammation. T lymphocytes become activated by antigen delivery to the T-cell receptor and concurrent engagement of co-stimulatory receptors, including CD28. Immune checkpoints regulate suppressive pathways that diminish immune activation. These counter-regulatory circuits exist to dampen immune responses in order to maintain peripheral self-tolerance as well as to prevent collateral tissue damage that may occur during an immune response [3]. Numerous inhibitory and stimulatory checkpoints have been described, with the best characterized of these being the co-inhibitory pathways exemplified by CTLA4 and PD-1.

    CTLA4

    CTLA4 is the archetypal immune regulatory checkpoint, expressed exclusively on T cells. Although first recognized on cytotoxic CD8+ lymphocytes, its main effect is mediated through profound changes in CD4+ helper and regulatory T cells. The importance of this inhibitory pathway was demonstrated in studies of knockout mice, in which deletion of the CTLA4 gene resulted in systemic immune activation, culminating in expansive lymphoproliferation and death at ~ 3–4 weeks old [4]. The ligands for CTLA4, CD80 (B7.1) and CD86 (B7.2), are expressed primarily on antigen-presenting cells and other immune cells, including T cells themselves. CTLA4 is stored in cytoplasmic vesicles in resting T cells, and moves to the cell surface when T cells are activated by antigen binding to the T-cell receptor. CTLA4 then competes with the immune stimulatory receptor CD28 for binding of CD80 and CD86. The ligands CD80 and CD86 have a higher affinity for CTLA4 than for CD28, and CTLA4 therefore has the ability to dampen CD28-mediated immune activation [5], resulting in impaired co-stimulation. Additionally, CTLA4 binding activates intracellular phosphatases that interfere with downstream signaling cascades that are important for T-cell activation.

    PD-1

    PD-1 and its endogenous ligands form an inhibitory checkpoint at a different stage of the lymphocyte response to infection or inflammation in peripheral tissues. In healthy individuals, the PD-1 receptor is expressed on antigen-presenting cells, activated T cells and other immune cells in peripheral tissues, including natural killer and dendritic cells. PD-1 interactions may down-regulate signaling via the B-cell receptor and natural killer cells, although the majority of its inhibitory function has been established in the setting of T-cell responses [6]. The ligand PD-L1 (PDCD1LG1; CD274; B7-H1) has a broad expression profile, and is seen on the same cells as PD-1 but also on non-hematopoietic cells in non-lymphoid organs, such as heart, lungs and muscle, implying a role in regulation of effector T-cell responses in peripheral tissues. Expression of the ligand PD-L2 (PDCD1LG2; CD273; B7-DC) appears to be restricted to B cells, macrophages and dendritic cells. Initial stimulation of a T cell results in expression of PD-1, which persists when the cell migrates to the tissues. Within the tissues, inflammatory cytokines induce expression of its ligands. PD-1 engagement increases phosphorylation of Src homology 2 domain containing non-transmembrane PTP (SHP-2), a phosphatase that may be important in attenuating T cell receptor sign. In addition, PD-1 ligation inhibits T-cell proliferation by blocking cell-cycle progression without increasing cell death [7]. The end result is reversible suppression of T-cell activity and proliferation, known as T-cell exhaustion, which has also been demonstrated in the setting of chronic infection [8]. T-cell exhaustion typically occurs at a site of infection or inflammation to limit damage to surrounding tissues and prevent autoimmunity [9].

    Subversion of immune checkpoints in lymphoid malignancy

    Cancers may subvert either of these two inhibitory checkpoints to evade host anti-tumor immune responses. Malignant cells escape immune surveillance by up-regulating their expression of checkpoint receptor ligands, as well as by increased expression of these ligands within the tumor microenvironment in areas of dense T-cell infiltration. This leads to inhibition of effector T-cell function. CTLA4 contributes to the suppressive function of regulatory T cells, which play a role in T-cell anergy as well as in suppression of T- and B-cell responses [10, 11]. Regulatory T cells in the tumor microenvironment constitutively express CTLA4 [12]. Tumor engagement of the CTLA4 pathway may therefore promote the suppressive function of regulatory T cells and dampen the immune response in the microenvironment.

    With the exception of chronic lymphocytic leukemia (CLL), PD-1 is rarely expressed on malignant lymphoid cells themselves, but is commonly found on tumor-infiltrating lymphocytes in the microenvironment, particularly in follicular lymphoma (FL) and Hodgkin lymphoma (HL) [13, 14]. Its ligands, particularly PD-L1, are more commonly expressed on lymphoid tumors [15]. Both PD-L1 and PD-L2 appear to be up-regulated in some B-cell lymphomas, including Hodgkin lymphoma and primary mediastinal B-cell lymphoma, due to amplification of the genomic locus encoding these two proteins on chromosome 9p as well as specific gene fusions that lead to PD-L1 and PD-L2 over-expression [16-18]. Classical Hodgkin lymphomas (cHLs) with normal 9p copy number may also express detectable PD-L1. An alternative mechanism of PD-L1 up-regulation in cHLs is Epstein–Barr virus infection (Fig. 1). Epstein–Barr virus infection promotes PD-L1 expression and PD-L1 promotor activity, implying a role for the PD-1 checkpoint in the pathogenesis of a variety of Epstein–Barr virus-related lymphoproliferative disorders [19]. PD-L1 expression may also be induced by interferon γ [7]. By expressing suppressive PD-1 ligands and engaging PD-1-bearing immune cells, tumors induce T-cell exhaustion and evade the host immune response. Immune checkpoints are therefore an attractive and important therapeutic target to harness the endogenous anti-tumor activity of the immune system.

    Details are in the caption following the image
    Immune evasion by PD-1 ligand up-regulation in classical Hodgkin lymphoma: putative mechanisms. PD-L1 and PD-L2 are over-expressed in Reed–Sternberg (RS) tumor cells with the 9p24.1 amplification. In addition, expression of Janus kinase 2 (JAK2) increases, which in turn increases PD-L1 transcription [16] (A). In Reed–Sternberg (RS) tumor cells with diploid 9p24.1 copy numbers, Epstein–Barr virus (EBV) promotes PD-L1 transcription in an AP-1-dependent manner [19] (B). AP-1, activator protein-1.

    Efficacy of immune checkpoint blockade in lymphoid malignancy

    Use of humanized IgG monoclonal antibodies primarily targeting CTLA4–CD80/86 and PD-1–PD-L1/PD-L2 interactions has shown promise in cancer therapy. These include the anti-CTLA4 IgG humanized antibodies, ipilimumab and tremelimumab, the former of which was the first checkpoint inhibitor to demonstrate a survival advantage in melanoma [20]. The US Food and Drug Administration approved ipilimumab for unresectable or metastatic melanoma in 2011. The anti-PD-1 IgG monoclonal antibodies pidilizumab, pembrolizumab and nivolumab followed. Single-agent pembrolizumab is approved for use in metastatic melanoma, as is single-agent nivolumab, which is also approved as second-line therapy for metastatic non-small cell lung cancer. Due to a more manageable immune toxicity profile, these agents have largely supplanted single-agent ipilimumab as the standard therapy in solid tumor oncology. The combination of ipilimumab and nivolumab was recently approved, and represents the first combination of checkpoint inhibitors available as a standard therapy. Large trials of checkpoint inhibitors in solid tumors have therefore paved the way for studies in hematologic malignancies.

    Hematologic malignancies are unique in that the tumor is itself constituted by cells of the immune system. They are attractive targets for immune checkpoint blockade given the demonstrated curative potential of other adoptive immune strategies, such as allogeneic stem cell transplant. Lymphoid malignancies have previously demonstrated significant responsiveness to immunological manipulations. B-cell lymphomas demonstrate inter-play between tumor and the host immune system that appears to directly affect lymphoma growth. This is supported by their ability to undergo spontaneous regression, their reported response to non-specific immune activators such as infectious triggers, and their response to B cell-directed monoclonal antibodies [21-23]. Furthermore, some B-cell malignancies, such as follicular lymphoma, contain many tumor-infiltrating lymphocytes in the tumor microenvironment that are potential candidates for checkpoint blockade to elicit local immune activation against malignant B cells.

    Studies of checkpoint inhibitors in lymphoid malignancies are reviewed below by agent, focusing first on the anti-CTLA4 antibody, ipilimumab, then on the first PD-1 antibody, pidilizumab, and concluding with nivolumab and pembrolizumab. Landmark clinical trials of these agents in lymphoid malignancies are summarized in Table 1.

    Ipilimumab

    Ipilimumab was the first checkpoint inhibitor studied in lymphoid malignancies. A dose-escalation study in 18 patients with relapsed B-cell lymphoma included 14 patients with follicular lymphoma (FL), three with diffuse large B-cell lymphoma (DLBCL) and one with mantle cell lymphoma. The drug was safe and tolerable, although diarrhea was noted in 56% of patients (grade 3 in 28%), probably a reflection of the autoimmune colitis that is now a well-recognized adverse event of the drug. The overall response rate was 11%, with one patient with DLBCL achieving a complete response of 31 months’ duration and another with FL achieving a partial response of 19 months’ duration. As a correlative endpoint, ipilimumab was shown to double T-cell proliferation in five of the 16 patients in whom it was evaluated [24].

    Ipilimumab has also been studied in the context of relapsed malignancy after allogeneic stem cell transplant as a strategy to augment the graft-versus-tumor effect. A phase I study evaluated 29 patients, including 14 with Hodgkin disease, six with myeloma, two with CLL and one with non-Hodgkin lymphoma (NHL). There were no dose-limiting toxicities, and 15 patients were treated with the target dose of 3 mg·kg−1, one of whom developed grade 4 pneumonitis possibly related to the drug. No patients developed grade 3 or 4 graft-versus-host disease after ipilimumab. Of the three patients who showed objective responses after ipilimumab, one had mantle cell lymphoma (partial response) and two had Hodgkin disease (complete responses) [25].

    Pidilizumab

    Pidilizumab is a humanized IgG1 monoclonal anti-PD-1 antibody that was first assessed in a phase I trial of 17 patients with hematologic malignancies, including three with CLL, four with NHL, one with HL and one with myeloma. The maximum tolerated dose was not reached. One patient with FL achieved a complete response, and one with HL and two with CLL had stable disease [26]. A phase II single-arm international study of pidilizumab in relapsed or refractory DLBCL or primary mediastinal B-cell lymphoma after autologous stem cell transplant was performed with the rationale that PD-1 blockade post-transplant would be effective due to low-volume disease and the potential for immune remodeling. The study included 66 patients [27]. In the 35 patients with measurable disease, the overall response rate was 51%, with 34% achieving complete response and 37% achieving stable disease. Correlative studies demonstrated an increase in PD-L1-bearing circulating lymphocytes, apparent 24 h after pidilizumab treatment, and sustained until almost 16 weeks. Subsequently, the drug was evaluated in conjunction with rituximab in a phase II single-arm open label study of 29 eligible patients with relapsed, refractory follicular lymphoma. There were no high-grade treatment-related adverse events, and the overall response rate was 66%, with an impressive 52% complete response rate. Correlative analyses suggested an association between up-regulated T-cell activation determined by gene expression profiling and improved progression-free survival [28].

    Nivolumab and pembrolizumab

    The newer PD-1 receptor-blocking antibodies, nivolumab and pembrolizumab, have received attention in hematologic malignancies due to very encouraging responses observed in early phase studies in cHL. In dose-escalation study of 23 relapsed and refractory patients with cHL, 20 patients (87%) had received at least three prior lines of treatment: 18 (78%) had previously received autologous stem cell transplant, and 18 (78%) had prior exposure to the anti-CD30 immunotoxin brentuximab vedotin. A remarkable 20 (87%) of patients on this study achieved a response, with four patients achieving a complete response. Progression-free survival at 24 weeks was 86% (90% CI = 62–95%). Notably, only 60% of the responses occurred within 8 weeks of initiating therapy, reflecting a delayed effect. Grade 3–4 toxicities were observed in 12 patients (52%), most commonly rash (22%) and thrombocytopenia (17%). Most patients received the full intended dosing schedule. Studies on a sub-group of ten patients with available tumor specimens prior to treatment demonstrated increased copy number of the PDL1 and PDL2 genes, and over-expression of PD-L1 and PD-L2 proteins in Reed–Sternberg cells. A confirmatory phase II open-label study of nivolumab in patients who have failed an autologous stem cell transplant is currently ongoing (NCT02181738) [16].

    Pembrolizumab has demonstrated similarly impressive results in cHL. A recently described phase Ib study enrolled 29 extensively pre-treated cHL patients: 15 patients (52%) had undergone five or more prior lines of therapy, all had received prior brentuximab vedotin, and 20 (69%) had received an autograft. Of 15 evaluable patients at 12 weeks, 3 patients (20%) had CR and five patients (33%) had PR, yielding an overall response rate of 53%. Grade 3 toxicities included pneumonitis, axillary discomfort and joint swelling, and one patient with pneumonitis discontinued treatment [29]. No grade 4 toxicities have been observed to date. The trial is ongoing at the present time (NCT01953692).

    To assess the effect of PD-1 blockade in hematologic malignancies other than cHL, a phase I dose-escalation study of nivolumab in patients with relapsed or refractory lymphoid malignancies has been completed and preliminary results were recently reported [30]. The patients included 27 patients with multiple myeloma, 31 with B-cell NHL (11 patients with DLBCL, 2 patients with PMBCL, 10 with FL, 8 with other B cell NHL), 23 patients with T-cell lymphoma (13 with mycosis fungoides, five with peripheral T-cell lymphoma not otherwise specified (NOS) and give with other T-cell lymphomas), and one patient with chronic myeloid leukemia. Responses were seen in four of eleven DLBCL patients (including one complete response), and four of the ten FL patients (including one complete response). The only other responses seen were a partial response in two of the 13 patients (15%) with mycosis fungoides and two of the five patients (40%) with peripheral T-cell lymphoma NOS. There were no responses in the 27 patients with multiple myeloma, although 18 (67%) of these patients achieved stable disease [30].

    Persistent questions relating to checkpoint inhibition in lymphoid malignancies

    As described above, immune interventions in the treatment of hematologic malignancies have demonstrated encouraging results in some disease sub-types. However, many questions remain regarding their specific mechanism of action, the reason for differing response rates in different tumor types, and whether immune checkpoint inhibitors may effectively be incorporated into frontline therapy.

    Checkpoint inhibition in lymphoid malignancies: how does it work?

    Our understanding of the mechanism of checkpoint inhibitors is incomplete. Despite a rudimentary grasp of what immune checkpoint blockade accomplishes on a cellular and molecular level, the precise mechanisms of action remain unclear. At the most basic level, PD-1 and CTLA4 blocking antibodies function by releasing a brake on the immune system, thereby facilitating T-cell activation and proliferation, and both CD4+ CD8+ T cells appear to play a role. However, the unleashing of activated T cells probably represents a bird's eye view that disregards the intricate network of pathways that regulate the immune system in lymphoid neoplasms [31]. The difficulty in establishing a reliable biomarker reflects the uncertainty in this area. Tumors appear to up-regulate expression of PD-L1. However, substantial responses to nivolumab have been seen even in tumors that appear to be PD-L1-negative. The studies of PD-1 ligand expression have been complicated by different definitions of what constitutes positivity, heterogeneity in the assays used, expression of PD-L1 on tumor cells compared to immune cells, and inclusion of patients at various stages of disease who may have received differing doses of therapy. Overall, the lack of a reliably identifiable biomarker in lymphoid malignancies may reflect our over-simplification of the mechanisms involved.

    It is also unclear to what degree features of tumor-infiltrating lymphocytes in the microenvironment are predictive of response, given the many complexities of the tumor–host relationship. This is particularly the case in hematologic malignancies, where lines are blurred because the tumor itself comprises immune cells. Malignant cells may evade the host immune response by up-regulation of PD-1 ligands and signaling through PD-1 on CD8+ T cells. The endogenous T-cell compartment is then unable to perform its function of recognizing peptide epitopes displayed on major histocompatibility complexes (MHCs) on the surface of malignant cells, a phenomenon known as adaptive immune resistance. Pre-existing CD8+ T cells located on the border of invasive tumor margins appear to express PD-1/PD-L1. In a study of 46 patients with metastatic melanoma, samples were obtained before and during anti-PD-1 therapy with pembrolizumab. Responders demonstrated proliferation of intratumoral CD8+ T cells that correlated directly with tumor size reduction. The pre-treatment specimens obtained from these patients showed more CD8+-, PD-1- and PD-L1 expressing cells at the tumor margin and inside the tumors. The authors contend that therapeutic PD-1 blockade therefore requires pre-existing CD8+ T cells in the tumor microenvironment (particularly at the border of invasion) that are negatively regulated by immune checkpoints [32]. The theory of silenced immune cells on the periphery of tumors becoming awakened by checkpoint inhibitors and then ‘rushing in’ appears to make sense intuitively in solid tumors, but the situation is less clear in hematologic malignancies, where the tumor and the immune system are one and the same, and T cells and malignant cells are typically inter-mixed at sites of disease.

    In addition to the immune cells in the tumor microenvironment, there are many unanswered questions about how much the malignant cell itself contributes to the mechanism of action of checkpoint inhibitors. These queries relate to longstanding uncertainty regarding the nature of the antigens involved in how the immune system distinguishes cancer cells from non-cancer cells. One predominant theory is that certain peptides arise as a consequence of tumor-specific mutations. These ‘neo-antigens’ are entirely absent from the normal human genome, and when the mutant peptide is presented by major histocompatibility complex molecules, they are recognized as tumor by the immune system. Therefore, tumors with more somatic mutations form neo-antigens more frequently that may be recognized by autologous T cells, and are therefore more susceptible to checkpoint inhibition. Solid tumors such as lung cancer and melanoma have the highest prevalence of somatic mutations, and indeed appear to be highly responsive to immune checkpoint blockade [33]. However, hematologic malignancies appear to fall somewhere in the middle in terms of the number of somatic mutations per megabase of coding DNA. Additionally, MHC expression is commonly lost in lymphoid malignancies, and this may interfere with neoantigen presentation. Does this imply that the formation of neo-antigens by lymphoid tumors and their subsequent presentation is less than in solid tumors? If so, theories of neo-antigenic immunogenicity and mutational burden may be less relevant in lymphoid malignancy than in solid tumors. These questions merit further investigation.

    Checkpoint inhibition in various lymphoid histologies

    Related to the many questions about the exact mechanism of action of checkpoint inhibition, there are also uncertainties as to what accounts for the striking differences in the response rate between different histologies of lymphoid tumors. Checkpoint inhibition is undoubtedly a promising strategy in cHL. On the other end of the spectrum, the apparent lack of responses seen in multiple myeloma patients is also striking, particularly because myeloma cells have been shown to express PD-L1 [34]. The biological basis for this difference is not understood, and there is little in the literature exploring this question. Are the differences due to less effective identification of the tumor in the first place, and, if so, is that caused by differences in tumor neo-antigens and mutational burden? Or is it due to substantial variation in the composition in the microenvironment amongst different lymphoid tumors?

    There may be differences in the ways that the host immune system interacts with different lymphoid malignancies. Recent studies have revealed a role for T cells in affecting the immunogenicity of cancers early in their formation by selectively destroying malignant cells that express strong tumor-specific neo-antigens, leaving behind malignant cells that express weaker antigens (which are perhaps still tumor-specific mutant antigens) [35]. Second, the chronic T-cell response to a tumor may silence expression of certain neo-antigens through epigenetic mechanisms [36]. In this manner, differential tumor ‘editing’ by the immune system may lead to antigenic heterogeneity in different tumors, resulting in different responses to checkpoint blockade [33].

    In addition to tumor immunoediting, host immunity may play a variety of roles in different tumor types that further confound the issue of disparate response rates in different lymphoid malignancies. Data suggest that there are two populations of PD-1-expressing T-cell sub-populations in follicular lymphoma, and the prevalence of the two subsets may affect patient outcome [37]. PD-1 is expressed on exhausted intra-tumoral effector T cells, but it is expressed more highly on follicular helper T cells. The native function of follicular helper T cells is rooted in their close interaction with B cells and their promotion of B-cell growth for germinal center formation: it is felt that they may ‘edit’ B cells. It is possible that follicular helper T cells may function in the host immune system to promote somatic hypermutation, affinity maturation and class-switch recombination, resulting in endogenously manufactured anti-tumor antibodies that help host B cells to fight malignant cells [38]. Therefore, PD-1 blockade in these cells may be inappropriate. The complex interplay between host B and T cells, as well as their interactions with tumor cells, is not well understood in the context of hematologic malignancy. Therefore, differences in the immune system itself in different tumor types may account for some of the varied response rates in different histologies.

    Checkpoint inhibitors: timing and combinations

    Aside from the questions related to the precise mechanism of checkpoint inhibition and the different responses amongst lymphoid malignancies, many questions exist regarding when in the disease course to use checkpoint blockade and whether it should be used as part of combination therapy. There have not yet been any studies of checkpoint inhibitors in the frontline setting, and therefore our experience is limited to patients with relapsed or refractory disease. It is unknown whether responses will differ between newly diagnosed and relapsed patients. In cHL in particular, the question arises as to how much improvement on the excellent frontline success rates with cytotoxic chemotherapy may be achieved. As a tumor becomes more chemo-resistant, it acquires more mutations and may display higher neo-antigenic immunogenicity than a recently diagnosed, less mutated tumor. It is also unclear whether the increase in mutations in tumors from chemo-resistant patients may include mutations that result in higher expression of PD-1/PD-L1, something that may be seen less frequently in chemo-naive patients. If it is the case that recurrent tumors are more antigenic and express PD-L1 more highly, the response rates observed in the relapsed or refractory population may be reflective of a population in whom checkpoint blockade works better than it would in the frontline. On the other hand, one may argue that a patient who has been through multiple rounds of cytotoxic chemotherapy has an immune system that is depleted. In this sense, checkpoint blockade may be more effective in a patient who is therapy-naive, i.e. a patient whose more functional immune system is ripe for activation. However, this theory is challenged by the outstanding responses seen in immuno-suppressed post-transplant patients [16, 27].

    Aside from these biological hypotheses, it is necessary to consider whether immune therapy should precede chemotherapy to avoid the increased toxicity of standard chemotherapy, or whether it should follow chemotherapy to address minimal residual disease and avoid relapse. It appears to make intuitive sense to deplete the malignant cell that is subverting the immune system in the first place, and then follow this with immune therapy. Conventional cancer therapies may led to tumor cell death and release antigens to activate T cells, priming an ‘immunogenic’ tumor microenvironment in which subsequent checkpoint blockade is more effective [39]. Clinical trials in development are likely to address these important questions regarding sequential or combined use of checkpoint inhibitors and conventional chemotherapy.

    If not given sequentially, should immune therapy be given in combination, either as CTLA4 or PD-1 blocking agents together or as combinations of checkpoint inhibitors with other agents? In metastatic melanoma, there appears to be benefit in terms of response rate to the combination of ipilimumab and nivolumab, albeit at the cost of increased treatment-related toxicity [40]. It is unclear whether the benefits and risks will be similar with combined checkpoint blockade in lymphoid neoplasms. Combining immune checkpoint inhibitors with immuno-modulators and targeted agents is an area of active investigation, as small molecules directed at signaling pathways involved in malignancy affect immune responses. Lenalidomide is an immune modulator that affects the tumor microenvironment. Preclinical studies in multiple myeloma have suggested various mechanisms by which the drug may stimulate antitumor immunity. A recent report suggests that lenalidomide promotes sustained proteasomal degradation of the substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3) in a cereblon-dependent manner, resulting in down-regulation of c-MYC and interferon regulatory factor 4 and ultimately tumor-cell apoptosis [41, 42]. Degradation of Ikaros and Aiolos also appears to potentiate production of interleukin-2 by T cells [43]. Lenalidomide may also promote T-cell proliferation by decreasing T-cell expression of PD-1 and decreasing the number of regulatory T cells in the microenvironment [44]. Further support for this mechanism comes from a trial of rituximab and lenalidomide in relapsed indolent NHL. Correlative studies of immune cell subsets demonstrated increased CD8+ T cells and increased PD-1-positive cells in patients receiving lenalidomide, implying activation of CD8+ intra-tumoral lymphocytes. There also appeared to be fewer suppressive CD4+ regulatory T cells [45]. Preclinical data also support the combination of checkpoint inhibitor therapy and the Bruton tyrosine kinase inhibitor ibrutinib. This drug inhibits Bruton tyrosine kinase but also blocks an enzyme known as interleukin-2-inducible T cell kinase, which is essential in Th2 cells. The combination of anti-PD-L1 antibody and ibrutinib demonstrated enhanced antitumor T-cell immune responses in mouse models of lymphoma, and led to suppression of tumor growth [46]. These studies open the door for clinical studies of combinations of immunotherapy with immunomodulators such as lenalidomide, or targeted agents, including Bruton tyrosine kinase inhibitors, phosphoinositide 3-kinase inhibitors and others.

    Future directions and conclusions

    The future of immune therapy is promising, and extends beyond inhibition of CTLA4 and PD-1. It has the potential to affect multiple other immunological pathways, in both a positive and negative fashion. Multiple other negative checkpoint regulators exist and have the ability to restrict T-cell activation and antitumor activity. These other inhibitory pathways include the T-cell immunoglobulin and mucin-containing protein 3 [47], the lymphocyte-activated gene 3, the B and T lymphocyte attenuator, and the V-domain Ig suppressor of T-cell activation, among others [47, 48]. These negative checkpoint regulators have similar but not necessarily redundant function to the more familiar CTLA4 and PD1 checkpoints, and blockade of several of these pathways synergistically may be necessary to restoring an efficient antitumor immune response.

    In addition to blocking multiple inhibitory immune checkpoints, another potential target for immune therapies is to use agonistic antibodies to simultaneously activate co-stimulatory pathways. The TNF receptor family co-stimulatory molecule OX40 (CD134) enhances antitumor immunity, promotes T-cell differentiation and decreases the number of regulatory T cells. Preclinical work suggests a synergistic role for an anti-OX40 agonistic antibody with CTLA4 blockade [49]. CD137 (4-1BB) is another co-stimulatory molecule with an agonistic antibody that has therapeutic potential [49, 50]. The inducible co-stimulatory molecule inducible T cell co-stimulator (ICOS) may have a direct role in the antitumor effect observed with CTLA4 blockade. Concomitant CTLA4 blockade and engagement of ICOS with a tumor cell vaccine enhanced antitumor immune responses in mouse models of prostate cancer and melanoma, further supporting a role for combined therapies [51].

    With a promising array of novel targets and combinations to pursue in the laboratory and the clinic, immune checkpoint inhibitors are already part of standard treatment protocols for solid tumors, and they may soon be an important complement to the treatment of hematologic malignancies. However, many unanswered questions remain regarding their mechanism, their sequential or combined use, and their efficacy in various lymphoid malignancy sub-types. Ongoing collaborative translational research to define relevant immune pathways and biomarkers that are predictive of efficacy is crucial. Participation in clinical trials of checkpoint inhibitors should be encouraged as we enter a promising new era of immunotherapy to treat lymphoid cancers.

    Author contributions

    GT and SMA developed the concept and outline for this article. GT and UT reviewed the literature, developed the figure and table, and wrote and revised the manuscript with contributions from SMA. All authors reviewed the final version of the manuscript.