Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 1.
Published in final edited form as: Expert Rev Anticancer Ther. 2019 Mar 19;19(5):393–404. doi: 10.1080/14737140.2019.1589374

Immune checkpoint-based therapy in myeloid malignancies: a promise yet to be fulfilled

Jan Philipp Bewersdorf 1, Maximilian Stahl 2, Amer M Zeidan 1,*
PMCID: PMC6527485  NIHMSID: NIHMS1523321  PMID: 30887841

Abstract

Introduction:

Immune system evasion is essential for tumor cell survival and is mediated by the immunosuppressive tumor microenvironment and the activation of inhibitory immune checkpoints. While immune checkpoint-based therapy yielded impressive results in several advanced solid malignancies such as melanoma and non-small cell lung cancer, its role in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) is still evolving.

Areas covered:

Here we review the immunology in the tumor microenvironment in the bone marrow and discuss the current preclinical and clinical data for immune checkpoint-based therapy in myeloid neoplasms.

Expert opinion:

Clinical trials of immune checkpoint inhibitors (ICI) in AML and MDS are still in early stages and reported results so far have been modest especially for monotherapy use in the refractory settings. However, there are preliminary data for synergistic effects for combination of multiple ICI with hypomethylating agents and conventional chemotherapy. ICI might also be effective in eradicating minimal residual disease and to prevent relapse following induction chemotherapy or hematopoietic stem cell transplant. Additional trials to provide insight into the efficacy and safety profile of immune checkpoint-based therapy, its optimal timing and potential combination with other types of therapy as well as identification of predictive biomarkers are needed.

Keywords: acute myeloid leukemia, immune checkpoint blockade, PD-1, CTLA-4, combination therapy, tumor immunology, T-cells

1.). Introduction

Creating an immunosuppressive microenvironment is essential for malignant cells to avoid elimination by the immune system 13. Elimination of malignantly transformed cells is dependent on T-cell mediated immunity 2. T-cells are activated by a two-step process consisting of (I) the presentation of major histocompatibility complex (MHC)-bound tumor antigens on antigen-presenting cells (APC) to the T-cell receptor (TCR) and (II) a co-stimulatory signal triggered by binding of B7–1 (CD80) or B7–2 (CD86) on APCs to CD28 on T-cells 2,4. This two-step process is essential to maintain self-tolerance and protect tissues from autoimmune-mediated destruction and is further supplemented by various immune checkpoints that regulate T-cell activation 5. These include co-stimulatory receptors that are either expressed on T-cells (CD28, 4–1BB, CD27, inducible T-cell co-stimulator (ICOS)) or APCs (CD80 and CD86), and co-inhibitory receptors (cytotoxic T-lymphocyte-associated-protein 4 (CTLA-4) and programed cell-death protein 1 (PD-1), T-cell immunoglobulin mucin-3 (TIM-3) and lymphocyte activation gene-3 (LAG-3))5.

One mechanism of immune system evasion is the upregulation of immune checkpoints in the tumor microenvironment 6. Stimulating an anti-tumor immune response by monoclonal antibodies directed mainly against CTLA-4, PD-1 and PD-1 ligand (PD-L1) has been clinically successful leading to several FDA-approvals in various advanced solid tumors 79. In hematologic malignancies, immune checkpoint inhibitors (ICI) appear promising in Hodgkin’s lymphoma (HL) 10 and some forms of non-Hodgkin’s lymphoma (NHL) 11. The proof of principle for the utility of immunotherapy in acute myeloid leukemia (AML) is allogeneic hematopoietic stem cell transplant (HSCT) which remains the most effective therapy to prevent relapse in AML patients in remission following chemotherapy and relies on the graft-versus-leukemia effect to eliminate residual AML blasts 12. Additionally, the anti-CTLA4 inhibitor ipilimumab and anti-PD1 antibodies nivolumab and pembrolizumab have shown some success in treating extramedullary relapse in AML following HSCT 13,14. However, the results from trials using ICI as monotherapy in AML and myelodysplastic syndrome (MDS) have shown only modest results thus far 15,16. Besides CTLA-4 and PD-1/PD-L1-directed approaches, other T-cell immune checkpoints, such as TIM-3 and LAG-3 as well as the macrophage checkpoint CD47 are also under investigation as therapeutic targets for AML and MDS (Figure 1).

Figure 1:

Figure 1:

Overview of potential targets for immune checkpoint-mediated therapy in myeloid malignancies Antigens, including tumor antigens, are presented bound to major histocompatibility complex (MHC)-I/II molecules by antigen-presenting cells (APC) and activate T-cells by binding to the T-cell receptor (TCR). However, a co-stimulatory signal such as activation of CD28 on T-cells by CD80 or CD86 is required for T-cell activation. The activity of T-cells is further regulated by various co-inhibitory (PD-1, CTLA-4, TIM-3, LAG-3) and co-stimulatory receptors (ICOS, OX-40) which can serve as potential targets for monoclonal antibodies that mainly block the activity of co-inhibitory receptors. However, as in the case of PF-04518600, activating co-stimulatory receptor function can also be employed to increase the anti-tumor immune response. Myeloid blasts can also express various receptors on their surfaces that create an immunosuppressive microenvironment and contribute to immune system evasion. While T-cells have been the main focus of immune checkpoint-mediated therapy thus far, current research has also identified the interaction of CD47 on APCs and myeloid blasts and SIRPα on macrophages as a potentially targetable mechanism of checkpoint therapy.

In this review, we discuss the recent clinical and pre-clinical data for ICI in AML and MDS and look forward into the future of ICB-based research in myeloid malignancies.

2.). Immune evasion in the tumor microenvironment of myeloid malignancies

The presence of T-cells in the tumor microenvironment has been shown to be a prognostic marker and to predict the response to ICI in various solid and hematologic malignancies including AML 1720. While several studies showed a greater effect of immune checkpoint inhibition with a higher expression of CTLA-4 and PD-1 on tumor-infiltrating T-cells and PD-L1 on tumor cells, respectively, clinical trials in non-small-cell lung cancer and melanoma showed clinical responses even in patients with low levels of baseline PD-L1 expression 2123. Furthermore, given the shared cell differentiation lineage with APCs, expression of the co-stimulatory receptors CD80 and CD86 is also seen in AML and MDS with controversial data regarding its impact on prognosis 2426. Additionally, several studies have demonstrated that an increased expression of PD-L1 and PD-L2 is associated with an adverse prognosis in AML patients and a lower likelihood of response to hypomethylating agents (HMA) 27,28. Another study also showed an increase in PD-1+T-cells in the bone marrow of patients with relapsed AML 29. Furthermore, an increase in regulatory T-cells in the peripheral blood has been shown in patients with AML and their persistence following induction chemotherapy has been correlated with a higher risk of relapse 3032.

There is a scarcity of data on the composition of the tumor microenvironment within the bone marrow in AML regarding the expression of immune checkpoint receptors, T-cell functionality, and distribution of various T-cell subsets. One requirement for the use of immune checkpoint blockade (ICB) is the presence of T-cells within the tumor microenvironment. Several studies could show that the absolute percentage of T-cells in the bone marrow from AML patients were either similar or increased compared to healthy volunteers 4,33,34. While peripheral T-cells appear to be functionally intact and able to respond to co-stimulatory signals, bone marrow T-cells showed an overexpression of the inhibitory immune checkpoint receptor PD-1 which could be overcome by concomitant application of a PD-1 inhibitor 4,29,35,36. Therefore, ideally bone marrow samples should be used to assess the expression of immune markers for clinical trials especially as there is significant heterogeneity between AML patients and T-cell function can change during the disease course and in response to treatment.

In addition to the number and distribution of T-cells in AML, their differentiation and activation status are of great importance. While CD8+T-cells can directly induce target cell apoptosis by secretion of granzyme B and perforin, CD4+T-cells can differentiate into various effector cell types including regulatory T-cells depending on the cytokines present in the tumor microenvironment 18. Currently, there are conflicting results regarding the ratio of CD8+/CD4+T-cells with one study showing an increased CD8+/CD4+-ratio compared to healthy controls that normalizes after chemotherapy, while other studies could not reproduce this finding 33,37. One potential explanation for immune evasion in AML is T-cell exhaustion, which is characterized by increased expression of various inhibitory receptors such as TIM-3, LAG-3, PD-1, and T-cell Immunoglobulin and Immunoreceptor Tyrosine-Based Inhibitory Motif Domain (TIGIT), and leads to impaired T-cell proliferation and cytokine production 18. A recent study in a mouse model and human cells which showed that tumor evasion in monocytic leukemias is mediated by LILRB4, a T-cell activity suppressing immunoreceptor tyrosine-based inhibitory motifs receptor, supports this hypothesis 38. This immune exhaustion seems to be related to a persistent antigen stimulation and multiple lines of prior chemotherapies and is most prevalent in bone marrow samples from patients with relapsed AML 36,39,40.

Lastly, immune regulation within the bone marrow is a complex interplay between T-cells, mesenchymal stromal cells, myeloid-derived suppressor cells, and various soluble factors. These interactions are highly complex and have been reviewed in detail elsewhere 18,41,42.

3.). Immune checkpoint inhibitors as monotherapy

3.1.). Anti-CTLA-4

CTLA-4 (CD152) is expressed on T-cells and inhibits maturation and differentiation of T-cells by competing with the costimulatory receptor CD28 for CD80 (B7.1) and CD86 (B7.2) on APCs 43. Additionally, CTLA-4 is constitutively expressed on and activates regulatory T-cells 44. In AML, the increased expression of CD80 and CD86 on AML blasts may contribute to immune evasion and has been associated with poor outcomes and higher rates of relapse 24,25,45,46. Remarkably, CTLA-4 was also found to be constitutively expressed on leukemic blasts in bone marrow samples from both untreated and chemo-resistant AML patients 47. Given the high expression of CTLA-4 on leukemia cells, blocking CTLA-4 seems to be a promising target to stimulate an anti-leukemia immune response that has been successfully tested in various preclinical models 4850.

Ipilimumab is an anti-CTLA-4 monoclonal antibody that enhances T-cell-mediated cytotoxicity by abrogating the inhibitory effect of CTLA-4 51. Several trials tested ipilimumab monotherapy in high-risk MDS and AML as well as in the post-HSCT relapse setting. Zeidan et al reported the results of a phase Ib study of 29 MDS patients with IPSS intermediate-1 risk and higher who had progressed while on HMAs that showed a marrow CR in 1 patient (3.4%), a prolonged stable disease >46 weeks in 24% of patients and 17% of patients were able to undergo HSCT without excessive toxicity 16. Of note, patients with a response to ipilimumab had a significantly higher expression of ICOS on CD8+ and CD4+ T-cells, which has been shown to be important in enhancing T-cell activation and response, and could be potentially used as a predictive response biomarker 13,16,52. Of note, 20.7% of patients in this study developed immune-related adverse events (IRAE) ≥ grade 2 that required removal from the study and was more frequent at higher doses of ipilimumab. Notably, all of these resolved after discontinuation of ipilimumab and administration of corticosteroids 16. Recently presented early results of ongoing trials showed more promising results using anti-CTLA4 and other ICB monotherapy in myeloid malignancies (Table 1). Should these early results hold, it would be important to understand reasons for differences in clinical activity through detailed correlative work.

Table 1:

Selected completed trials of immune checkpoint blockade monotherapy in AML/MDS

Drug Target / mechanism Phase [ref] N Patient characteristics Outcomes
Pidilizumab Anti-PD-1 antibody I57 17 (8 AML, 1 MDS) Advanced hematologic malignancies, age <65 1 AML patient with reduction in peripheral blasts
Nivolumab Anti-PD-1 antibody II77 15 MDS frontline and HMA failure 0% CR/CRp; 25% 1-year survival
II83 14 HR-AML in CR ineligible for HSCT 71% 1-year CR rate
Pembrolizumab Anti-PD-1 antibody I58 28 MDS after HMA failure ORR: 4% (1 PR); 49% 24-week survival
Ipilimumab Anti-CTLA-4 antibody I 13 28 (12 pts w/ AML) Hematologic malignancies with relapse after HSCT No response with 3mg/kg
Response with 10mg/kg
CR 23%, PR 9%
23% decrease in tumor burden
CR in 1 AML patient secondary to MDS
I 16 29 MDS patients who failed HMAs CR 3.4%, PSD for >46 weeks 27%, >54 weeks 10%
Median OS 294 days (censoring at allogeneic HSCT),
II77 20 MDS frontline and HMA failure 15% CR/CRp; 45% 1-year survival

3.2.). Anti-PD-1

Under physiologic conditions, interaction of PD-1 on activated T-cells and regulatory T-cells with PD-L1/2, which is expressed on various immune cells, serves to limit T-cell activation after TCR/MHC engagement and CD28 activation and can be upregulated in the presence of interferon-γ 5355. An increased expression of PD-L1 on AML blasts and PD-1 on T-cells has been found in a subset of AML and MDS patients, especially at relapse, and has been associated with a poor prognosis 28,35.

After mouse models showed that an increased PD-1/PD-L1 expression led to a state of T-cell exhaustion which could be overcome by ICB 56, several monoclonal antibodies targeting PD-1/PD-L1 signaling have been developed and tested in AML patients. Pidilizumab (CT-011) was one of the first monoclonal anti-PD-1 antibodies to be tested in hematologic malignancies. In a phase I study of 17 patients with advanced hematologic neoplasms (8 AML, one MDS) treated with a single dose of pidilizumab, only one AML patient achieved a reduction in bone marrow blast count but progressed subsequently 57. Other anti-PD-1 antibodies that are currently being studied as monotherapy for AML or MDS at various disease stages include nivolumab and pembrolizumab (Table 1). In the KEYNOTE-013 trial (NCT01953692), 28 MDS patients who progressed on therapy with HMAs and were treated with pembrolizumab achieved an overall response rate of only 4% (1 partial remission) with hematologic improvement in 11% of patients 58.

Given the only modest efficacy for anti-PD-1 and anti-CTLA-4 monotherapy, subsequent trials have tested combinations of either CTLA-4 and PD-1 blockade, checkpoint blockade in combination with HMAs, conventional chemotherapy or other forms of immunotherapy.

4.). Immune checkpoint inhibitors in combination with other treatment modalities

4.1). Immune checkpoint inhibitors in combination with hypomethylating agents

Gene expression is regulated by DNA methylation and DNA hydroxymethylation and mutations in genes affecting these key epigenetic pathways (e.g. DNMT3A, TET2) have been linked to malignant transformation by inactivating tumor suppressor genes and are seen in 22% and 23% of AML patients, respectively 5961. HMAs, such as 5-azacytidine (5-AZA) and its analogue 5-aza-2’deoxycytidine (decitabine) inhibit DNA methyltransferase and work by non-selectively reversing DNA methylation and restoring gene expression 6264. In addition to increased expression of tumor suppressor genes, this leads to activation of the immune system by increased expression of MHC-I and other co-stimulatory receptors (CD80, CD86, ICAM1) 6567. One of the potential mechanisms of resistance to 5-AZA is a dose dependent upregulation of the PD-1/PD-L1 axis, which might lead to synergistic effects of combination of HMAs with PD-1/PD-L1 inhibitors 27,56. Both preclinical experiments combining 5-AZA with ipilimumab as well as HMAs in combination with PD-1/PD-L1 blockade in patients with NSCLC supported this presumed synergistic effect 6870.

Another potential mechanism underlying the synergy between azacitidine and ICB might be the upregulation of antiviral defense mechanisms such as interferon-γ signaling. It has previously been shown that a component of the antitumor activity of HMA is the activation of viral defense pathways by induction of HLA class I antigens and interferon-γ signaling 71. Previous studies in melanoma patients treated with anti-CTLA4 antibodies showed increased expression of similar anti-viral defense genes and mouse models showed synergistic effects of anti-CTLA4 inhibitors and azacitidine, which lends additional support to combining HMA with ICB 71,72.

These findings provided the rationale for various clinical trials in AML and MDS of HMAs in combination with several PD-1 inhibitors (nivolumab ((NCT02397720), pembrolizumab (NCT02845297)), PD-L1 inhibitors (durvalumab (NCT02775903), atezolizumab (NCT02508870)), or the CTLA-4 inhibitor ipilimumab (NCT02890329, NCT02397720) for various disease stages (Tables 2 and 3).

Table 2:

Selected completed clinical trials of immune checkpoint blockade as part of combination therapy in AML/MDS

Drug Target / mechanism Phase [ref] N Patient characteristics Intervention Outcomes
Nivolumab Anti-PD-1 antibody II77 20 MDS frontline and HMA failure Nivolumab + 5-AZA 75% CR/CRp; 50% 1-year survival
II73 70 RR-AML Nivolumab + 5-AZA ORR: 33% (22% CR/CRi); median OS 6.3 months
II81 41 Frontline AML or high-risk MDS; age<65 Idarubicin + cytarabine +/− nivolumab 77% CR/CRi; median OS 18.54 (nivolumab group) vs 13.2 months (I+A alone), p =0.2
Pembrolizumab Anti-PD-1 antibody I75 17 RR-AML Pembrolizumab + decitabine 1 MRD-negative CR; median OS 7 months
Ipilimumab Anti-CTLA-4 antibody II77 21 MDS frontline and HMA failure Ipilimumab + 5-AZA 71% CR/CRp; 68% 1-year survival

Table 3:

Selected ongoing trials of immune checkpoint therapy

Drug Target/mechanism Phase NCT Patient characteristics Intervention
Nivolumab Anti-PD-1 antibody I NCT02846376 AML/MDS after SCT at high risk for relapse Nivolumab + ipilimumab
II NCT02275533 Eradication of MRD in AML in CR1 Nivolumab
II NCT02397720 RR-AML, ND-AML >65 years Nivolumab + 5-AZA +/− ipilimumab
II NCT03417154 RR-AML, HR-MDS Nivolumab + Cyclophosphamide
Pembrolizumab Anti-PD-1 antibody I NCT02981914 Relapsed MDS, AML, B-cell lymphoma following allogeneic HSCT Pembrolizumab
I NCT03286114 Relapsed MDS, AML, ALL following allogeneic HSCT Pembrolizumab
II NCT02768792 RR-AML HiDAC + pembrolizumab
II NCT02845297 RR MDS/AML and newly diagnosed AML patients >65 5-AZA + pembrolizumab
II NCT02708641 AML in CR ineligible for HSCT Pembrolizumab
II NCT02771197 HR-AML ineligible for allogeneic HSCT Pembrolizumab + Fludarabine/melphalan conditioning + autologous HSCT
Durvalumab Anti-PD-1 antibody I NCT02117219 125 MDS after HMA failure Durvalumab +/− 5-AZA +/− tremelimumab
II NCT02775903126 High risk MDS, elderly AML patients Durvalumab + 5-AZA
Ipilimumab Anti-CTLA-4-Antibody I NCT02890329 RR MDS/AML Ipilimumab + decitabine
Hu5F9-G4 Anti-CD47 antibody I NCT02678338 RR-AML, HR-MDS Hu5F9-G4
I NCT03248479 RR MDS/AML or unfit ND-AML/MDS Hu5F9-G4 + 5-AZA
TTI-621 (SIRPαFc) Anti-CD47 antibody I NCT02663518 Hematologic and solid tumors TTI-621 +/− rituximab or nivolumab
MBG453 Anti-TIM-3 antibody I NCT03066648 AML, HR-MDS MBG453 + Decitabine or PDR001 (anti-PD-1 antibody)
DC AML vaccine Tumor cell vaccine I NCT01096602 ND-AML or RR-AML PD-1 blockade + DC AML vaccine

A recent phase II study of 70 patients with relapsed/refractory AML (RR-AML) treated with nivolumab and 5-AZA showed a sustained ORR of 33% (22% complete remission (CR) or complete remission with insufficient recovery of counts (CRi)) with a median overall survival (OS) of 6.3 months which is superior to historical controls (ORR: 20%, median OS: 4.6 months) of HMA-based therapy including a combination of 5-AZA and venetoclax 73. IRAEs grade ≥3 occurred in 11% of patients with 2 deaths due to a corticosteroid- and infliximab-refractory IRAEs (NCT02397720). Interestingly, response rates were higher in HMA-naïve patients, patients with CD3+, CD4+ T-effector cells, and CD8+ T-cells in the pretherapy bone marrow aspirates, and in patients with an ASXL1 mutation, which has been previously linked to a higher immunogenicity 73. Of note, non-responders had an increased expression of CTLA-4 on T-cells which suggests that there might be a different efficacy of PD-1 vs. CTLA-4 inhibition. Studies investigating the combination of different ICI with or without HMAs are an interesting area of future investigation. Several of these trials are currently ongoing (nivolumab + ipilimumab + 5-AZA [NCT02397720], nivolumab + ipilimumab for AML after HSCT [NCT02846376]) 74. Similar preliminary results for the combination of pembrolizumab and decitabine in RR-AML were also presented at the 2018 ASH meeting. In a phase I trial of 10 patients (NCT02996474), 1 patient achieved a minimal residual disease (MRD)-negative CR for 337 days and the median OS in the entire study population was 7 months 75. Preliminary data from a phase II study (NCT03094637) of azacitidine and pembrolizumab in 18 high-risk MDS patients presented at the 2018 ASH meeting showed 2 CRs and 3 hematologic improvements in 12 patients evaluable for response of whom 7 had progressed on HMA (1 CR and 1 HI) 76. Treatment was well-tolerated and the clinical efficacy will need to be further evaluated.

A multi-arm phase II clinical trial tested nivolumab and ipilimumab as monotherapy or in combination with 5-AZA in both the frontline setting (41 patients) or after HMA failure (35 patients) in intermediate/high risk MDS (NCT02530463). Preliminary data available in abstract form showed overall response rates of 75% (15/20; CR/CRp 50%), 71% (15/21; CR/CRp 38%), 13% (2/15; CR/CRp 0%), and 35% (7/20; CR/CRp 15%) for 5-AZA + nivolumab, 5-AZA + ipilimumab, nivolumab monotherapy, and ipilimumab monotherapy, respectively. Furthermore, the combination of 5-AZA with either nivolumab or ipilimumab was efficacious both in the frontline and in the HMA-refractory setting with a median OS of 17 months and 8 months, respectively 77.

Safety and especially IRAEs remain a major concern for checkpoint inhibitor therapy. While most IRAEs respond promptly to corticosteroids and even a re-challenge with these agents has been shown to be feasible in selected patients, fatal courses of IRAEs have been reported and a clinical trial of 5-AZA with atezolizumab (NCT02508870) had to be discontinued due to safety concerns 78. Future studies to address the safety profile of checkpoint inhibitors are therefore warranted prior to their broader clinical application especially when combining PD-1/PD-L1 and CTLA-4 blockade which has been shown to have a substantial increase in IRAEs in solid malignancies 7.

4.2). Combination of checkpoint blockade with conventional chemotherapy

DNA damage either by cytotoxic chemotherapy or gamma-irradiation has been shown to stimulate anti-leukemia immune responses in a murine model of AML by inducing expression of the co-stimulatory receptors CD80 and CD86 and decreasing PD-L1 expression 79,80. An increased CD80 and CD86 expression after exposure to cytarabine could also be shown in human AML cells 80. Release of tumor antigens following cytotoxic chemotherapy might also stimulate an anti-leukemia immune response. Several trials investigating anti-PD-1 antibodies are currently active, but no results have been published yet. These include nivolumab + “7+3” induction chemotherapy (NCT02464657), nivolumab + cyclophosphamide (NCT03417154) and pembrolizumab + high-dose cytarabine (NCT02768792). Preliminary data from a phase II trial of nivolumab in combination with idarubicin and cytarabine in newly-diagnosed AML reported a 77% CR/CRi (28 CR, 6 CRi; 18/34 (53%) MRD-negative by flow-cytometry) rate and a non-significant trend towards an improved median OS (18.5 months vs. 13.2 months) with the addition of nivolumab 81.

4.3). Checkpoint inhibitors for minimal residual disease eradication and post HSCT

Minimal residual disease (MRD) as measured by next-generation sequencing has been shown to have a significant prognostic effect on relapse and survival in patients in CR following induction chemotherapy 82. Since preclinical data suggest that immune checkpoint pathways contribute to immune system evasion of dormant leukemia cells and that these cells are resistant to T-cell-mediated cytolysis, ICI therapy might provide a promising option to eradicate MRD in patients in CR 46. Preliminary results of a phase II study (NCT02532231) of maintenance nivolumab in AML patients in CR following induction and consolidation chemotherapy but at high risk of relapse showed promising results with a 1-year OS rate of 86% and grade 3/4 IRAEs in 5 out of 14 patients 83.

For patients with relapse after HSCT the prognosis is poor and treatment modalities to prevent or treat relapse are of great clinical importance 84,85. A potential mechanism of relapse following HSCT appears to be the downregulation of patient specific HLA haplotypes leading to immune evasion of donor T-cells 86,87. Inspired by the graft-versus-leukemia effect observed in HSCT and donor lymphocyte infusion and the hypothesis that this beneficial effect can be further enhanced by ICB, several clinical trials testing PD-1/PD-L1 and CTLA-4-targeted antibodies in the post-HSCT setting have been conducted. In a phase I trial of ipilimumab in relapsed hematologic malignancies following HSCT (NCT01822509), 5 out of 28 patients (4 out of 12 AML patients) had a CR with a median 1-year OS rate of 49% 13. Additionally, ipilimumab appeared to be well-tolerated with GVHD of the liver and gastrointestinal tract in 4 patients as well as one case of IRAE being the only dose-limiting toxicities 13. Conversely, in another trial (NCT00060372) of 29 patients who relapsed after HSCT or donor lymphocyte infusion (2 AML patients), only 3 patients with lymphoid malignancies and none of the AML patients achieved an objective response 88. Furthermore, a recent case series of 3 patients with relapsed AML post-HSCT treated with nivolumab showed a durable CR in 1 patient and disease stabilization in another patient 14. Additional trials of ipilimumab, nivolumab or combination therapy to either treat (NCT01822509) or prevent relapse (NCT02846376) are currently active.

While stimulating the graft-versus-leukemia-effect by ICB might prove to be beneficial, this also poses the risk of increasing incidence and severity of GVHD. In fact, cases of fatal GVHD associated with PD-1 blockade post-HSCT have been reported repeatedly and seemed to respond poorly to corticosteroids 89,90. Therefore, additional studies to evaluate both safety and efficacy of checkpoint inhibitors in the post-HSCT setting in myeloid malignancies are warranted.

5.). Future directions and other potential targets for immune checkpoint-based therapies

5.1.). TIM-3 and LAG-3

In addition to PD-1 and CTLA-4 there are several other inhibitory immune checkpoints that can be therapeutically targeted to enhance the host’s immune response to malignant cells. T-cell immunoglobulin mucin-3 (TIM-3) is a glycoprotein that is expressed on the surface of regulatory T-cells, INF-γ producing T-cells, and innate immune cells and has been shown to be expressed at increased levels on AML and leukemic stem cells 9193. The effect of TIM-3 activation is context-dependent and includes both inhibition of T-cell-mediated immune responses and pro-inflammatory effects by activation of macrophages 94,95. Galactin-9 is a major activator of TIM-3 and high levels of galectin-9-TIM-3 coexpression have been found on leukemic stem cells in AML, but not on normal hematopoietic stem cells and were linked to leukemic transformation and progression by inhibition of Th1-cell immune responses 96,97. Additionally, coexpression of PD-1 and TIM-3 has been associated with immune exhaustion and relapse in AML patients after HSCT 40. Currently, a phase I study of the anti-TIM-3 monoclonal antibody MBG453 alone or in combination with the anti-PD-1 antibody PDR001 or decitabine is being tested in patients with AML or MDS (NCT03066648). Given that TIM-3 is expressed to a higher level on leukemic stem cells than non-neoplastic hematopoietic stem cells and is often expressed in conjunction with other surface antigens such as CD33, CD123, and CLL, targeting TIM-3 might limit on-target-off-leukemia side effects both as a single agent and in combination with another target antigen 92.

Lymphocyte-activation gene 3 (LAG-3) is an inhibitory receptor that has been shown to be upregulated on Th-cells in bone marrow aspirates from AML and MDS patients and has been associated with immune exhaustion especially if coexpressed with PD-1 and CTLA-4 98. Therefore, inhibition with anti-LAG-3 antibodies alone or in combination with other checkpoint inhibitors might provide an additional therapeutic target to overcome T-cell exhaustion in AML.

LILRB4 is an immunoreceptor tyrosine-based inhibition motif-containing receptor that inhibits T-cell activity and has been shown to be associated with immune evasion of monocytic leukemia cells in both mouse models and human cells 38. Importantly, antibody-mediated inhibition has been shown to inhibit AML development and makes LILRB4 another potential therapeutic target especially in monocytic AML 38,99.

5.2.). OX-40

Boosting the immune response by activating co-stimulatory receptors such as 4–1BB (CD137) or OX-40 (CD134) is another promising treatment strategy. OX-40 has been shown to promote activation and proliferation of effector T-cells while suppressing the differentiation of regulatory T-cells 100,101. In preclinical models OX-40 agonists have been demonstrated to lead to tumor regression by stimulating cytotoxic T-cell and NK-cell activity 102. OX40 is also expressed on AML cells and its expression pattern varies between patients 36. Application of tumor-necrosis factor in in vitro models could further increase OX40 expression 101. Antibodies targeting OX40 have been successfully tested in various hematologic and solid malignancies 100,102. A multi-arm phase I trial of the OX40 agonist PF-04518600 as monotherapy or in combination with 5-AZA, PD-1 inhibitors, or 4–1BB agonists in AML patients is currently ongoing (NCT03390296). However, OX40-ligand is highly expressed on other cells within the tumor microenvironment and the effect of this OX40-OX40L interaction needs to be further elucidated 101. Additionally, stimulating the immune response by activating OX40 may lead to increased side effects especially when combined with other checkpoint inhibitors.

5.3). Anti-CD47

While T-cells are essential in the anti-leukemia immune response, the innate immune system and especially macrophages are playing a role in immune evasion as well. CD47 is expressed by normal cells to inhibit phagocytosis by binding to SIRPα on macrophages and has been shown to be overexpressed on various tumor cells including AML 103,104. An increased rate of CD47 expression has also been linked to a higher rate of transformation to AML in MDS patients 105. There is increasing preclinical evidence that blocking the interaction between CD47 and SIRPα with anti-CD47 antibodies can promote the phagocytosis of AML cells 106. Of note, CD47 is expressed on normal hematopoietic stem cells and progenitor cells as well 107,108. In addition to acting as an antigen sink and potentially limiting the anti-CD47 antibody’s ability to reach tumor cells, in vitro studies of anti-CD47 antibodies have been associated with platelet aggregation and hemolytic anemia 104,106,109.

Hu5F9-G4, a monoclonal anti-CD47 antibody, is currently being tested in clinical trials as monotherapy and in combination with azacitidine for RR-AML patients (NCT02678338, NCT03248479). In a phase Ib/II clinical trial (NCT02953509) in 22 patients with relapsed/refractory NHL, Hu5F9-G4 in combination with the anti-CD20 antibody rituximab showed an ORR of 50% (32% CR) with only 3 grade 3 adverse events 110. TTI-621 (SIRPαFc), a fusion protein of the N-terminal portion of SIRPα and the IgG1 Fc region, binds to both CD47 and the Fcγ receptor on macrophages to enhance phagocytosis and has been successfully tested in NHL as well 104,111. However, another phase I study of CC-90002, an anti-CD47 antibody, in RR-AML and high-risk MDS has recently been terminated as “data did not offer a sufficiently encouraging profile for further development” according to a statement on www.clinicaltrials.gov (NCT02641002).

5.4). Combination of tumor vaccines and checkpoint inhibitors

As outlined above, the immunosuppressive tumor microenvironment is of great importance. Dendritic cells (DC) are potent antigen-presenting cells that function as a mediator between the innate and adaptive immune system by expressing co-stimulatory signals and presenting antigens that are required to mount an immune response 112,113. Recently, dendritic cells have been shown to contribute to tumor cell immune evasion by expressing inhibitory immune checkpoints such as PD-1 leading to a reduction in proinflammatory cytokine production and proliferation of CD8+ T-cells 114,115. Combination of dendritic cell/AML vaccination and PD-1 blockade might therefore have synergistic effects and is currently being studied in a phase II clinical trial of AML patients in remission following chemotherapy (NCT01096602).

5.5. Combination of chimeric antigen receptor (CAR) T-cell therapy and ICB

CAR T-cells, a novel therapeutic concept in which autologous T-cells are first transduced with a retro- or lentiviral vector carrying the CAR gene, are expanded in vitro and are infused back into the patient, has shown remarkable results in B-cell-derived hematologic malignancies such as B-ALL and non-Hodgkin’s lymphoma 15,116118. Co-administration of ICB may increase the potency of CAR T-cells as samples from patients with B-cell malignancies treated with anti-CD19 CAR T-cells have shown that CAR T-cells can acquire an exhausted phenotype characterized by increased expression of PD-1, LAG-3, and TIM-3 119,120. Preclinical and clinical data have also shown a synergistic effect of ICB and CAR T-cells therapy 121. Notably, a case report of a patient with DLBCL who had been treated with anti-CD19 CAR T-cells but showed progressive disease on follow-up responded well to addition of pembrolizumab 122. Treatment of patients with CD19+ lymphomas who failed to respond to anti-CD19 CAR T-cells with pembrolizumab is currently studied in a phase I/II trial (NCT02650999). However, there are currently no clinical trials combining CAR T-cells and ICB in AML.

5.6). Balancing therapeutic efficacy and IRAE of ICB in AML

Unleashing the immune system can lead to a variety of adverse events ranging from colitis to pneumonitis, endocrine dysfunction, cardio- and neurotoxicity and patients treated with combinations of PD-1 and CTLA-4 inhibition tended to be more likely to be develop IRAEs 78,123. Fatal toxicity has been reported in 0.6% of patients with any malignancy treated with ICB 78. While most of the safety data are from patients with melanoma and NSCLC, available data from patients with hematologic malignancies appear to be comparable and most of the IRAEs observed responded well to corticosteroids 16,124. A special consideration is the concern for a higher incidence of GVHD in patients undergoing HSCT after ICB therapy. Data on the incidence and severity of GVHD are controversial and cases of fatal GVHD associated with PD-1 blockade post-HSCT have been reported which seemed to respond poorly to corticosteroids 13,89,90. Therefore, additional studies are warranted to elucidate the safety profile of ICB in AML treatment especially in combination therapy and in the post-HSCT setting.

7.). Conclusion

Similar to some solid malignancies, enhancing the host’s anti-tumor immune response in myeloid malignancies might be a promising therapeutic approach. While monotherapy with ICI has so far been reported to have modest effects, combination of ICI with HMAs and chemotherapy appear to offer synergistic effects in early clinical trial results. The optimal combinations and timing of ICI in the disease course need to be further elucidated by clinical trials. As treatment of AML and MDS is becoming increasingly individualized, biomarkers that predict response to immune checkpoint therapy are needed to guide patient selection. Early identification and aggressive management of immune related adverse events is necessary to minimize complications and optimize clinical results.

8.). Expert opinion

While ICI has been extremely successful even if used as monotherapy in some advanced solid malignancies, early clinical trial results in myeloid neoplasms have been underwhelming, though longer term results are still pending. Recent preclinical studies have demonstrated the presence of T-cells in an immunologically exhausted state as evidenced by the upregulation of inhibitory immune checkpoints in AML bone marrow samples which suggests that ICI might be a feasible strategy in myeloid neoplasms as well. However, results from monotherapy with ICI have been only modest thus far and predictive biomarkers are missing. Given the genetic heterogeneity of patients with AML and MDS, responses to ICI are expected to be heterogenous and additional studies to identify predictive biomarkers and to improve our understanding of the immunology in the bone marrow microenvironment are warranted to optimize patient selection.

Therefore, combination therapy of ICI and other treatment modalities such as HMAs and conventional chemotherapy seems a logical way forward. For example, upregulation of CTLA-4 and PD-1/PD-L1/2 has been found in MDS patients who failed treatment with HMAs and HMAs have been successfully combined with various ICI agents already. Similar to studies in solid malignancies that suggested tumors with a higher mutational burden had a higher likelihood to respond to ICI, the DNA damage inflicted by cytotoxic chemotherapy has been shown to stimulate anti-leukemia immune responses in a murine model of AML by inducing expression of the co-stimulatory receptors CD80 and CD86 and decreasing PD-L1 expression. Combination of ICI and conventional chemotherapy could therefore have synergistic effects as it releases the brakes from the immune system and provides additional stimulation simultaneously. Along the same lines are efforts to develop antibodies to co-stimulatory immune checkpoints like OX-40 that could be combined with ICI as well to increase the anti-leukemia immune response. However, several caveats remain especially with regard to excess toxicity associated with combination of various immune checkpoint-targeting therapies. IRAEs have been documented already in monotherapy studies and while they appear manageable with corticosteroids in most cases, fatal toxicities have been reported. Early recognition and aggressive management of IRAEs are of paramount importance in the conduction of these trials, and in myeloid malignancies impose special challenges. For example, the frequency of bacterial and fungal pneumonias in advanced myeloid malignancies can make physicians more concerned about prompt use of steroids for suspected pneumonitis. Similarly, severe and refractory thrombocytopenia, which is common among patients with advanced MDS and AML, make obtaining lung biopsies to differentiate infectious from inflammatory pneumonitis riskier in such cases.

Finally, there has been some concern for inducing or worsening graft-versus-host-disease (GVHD) in patients treated with ICI and proceeding to, or who have relapsed after HSCT. However, early data regarding the few patients who did proceed to HSCT from different clinical trials of ICI in myeloid malignancies do not suggest an increase incidence of severe GVHD. Nonetheless, more data, longer follow-up and careful monitoring are still needed to exclude this concern.

ICI in myeloid malignancies remains in early stages and carefully conducted clinical trials are needed to assess the safety and efficacy of immune checkpoint-based therapy, its optimal timing and potential combination with other treatment modalities. Over the next 5 years we expect our understanding of the immunology of the bone marrow microenvironment to improve further and to maybe enable a correlation of various expression patterns of immune checkpoint molecules on both leukemia cells and immune cells with different disease stages. Analysis of immune checkpoint expression profiles and careful correlative studies should be performed in all such clinical trials to help identify predictive biomarkers and to inform rational combination therapies. Several large clinical trials of various combination therapies are currently ongoing and will provide additional information on the safety, efficacy and long-term outcome of ICI in AML and MDS. Additional trials to investigate the efficacy of ICI to eradicate MRD in patients post-HSCT or induction chemotherapy will also be needed. It will be interesting to see if the responses to ICI in AML are as durable as with some patients with solid malignancies. We remain optimistic that ICI especially if included in well-designed combination therapy have the potential to provide significant benefit to the treatment of patients with myeloid malignancies and various stages of the disease course.

Article highlights.

  • Immune system evasion is essential for tumor cell survival in both solid malignancies as well as in myeloid neoplasms and is mediated by the activation of inhibitory immune checkpoints such as cytotoxic T-lymphocyte-associated-protein 4 (CTLA-4) and programed cell-death protein 1 (PD-1), T-cell immunoglobulin mucin-3 (TIM-3) and lymphocyte activation gene-3 (LAG-3).

  • Immune checkpoint therapy has the potential to reverse immune system evasion of myeloid malignancies. While monotherapy with immune checkpoint inhibitors has only yielded modest results, combination with hypomethylating agents, intensive chemotherapy and multiple immune checkpoint inhibitors as shown promising results.

  • Further research to identify predictive biomarkers, optimize timing, combination partners and treatment settings is warranted

Acknowledgments

Funding

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health (P30CA016359). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Declaration of interest

AM Zeidan reports receiving research funding from Celgene, Acceleron, Abbvie, Otsuka, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Trovagene, Incyte, Takeda, and ADC Therapeutics. They also report acting in a consultant capacity and receiving honoraria from Abbvie, Otsuka, Pfizer, Celgene, Ariad, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, and Takeda. AM Zeidan has also received honoraria for speaker roles from Takeda. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Footnotes

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

References

Articles or patents of special interest have been highlighted as either of interest (*) or of considerable interest (**) to readers.

  • 1.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646–674. [DOI] [PubMed] [Google Scholar]
  • 2.Goodman A, Patel SP, Kurzrock R. PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nat Rev Clin Oncol 2017;14(4):203–220. [DOI] [PubMed] [Google Scholar]
  • 3.Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 2012;21(3):309–322. [DOI] [PubMed] [Google Scholar]
  • 4.Boddu P, Kantarjian H, Garcia-Manero G, Allison J, Sharma P, Daver N. The emerging role of immune checkpoint based approaches in AML and MDS. Leuk Lymphoma 2018;59(4):790–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013;13(4):227–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002;8(8):793–800. [DOI] [PubMed] [Google Scholar]
  • 7.Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 2017;377(14):1345–1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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(2):123–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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(10031):1909–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.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(4):311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol 2016;34(23):2698–2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Antin JH. Graft-versus-leukemia: no longer an epiphenomenon. Blood 1993;82(8):2273–2277. [PubMed] [Google Scholar]
  • *13.Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N Engl J Med 2016;375(2):143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is a landmark clinical trial showing the efficacy and safety of ipilimumab in the post-HSCT setting
  • 14.Albring JC, Inselmann S, Sauer T, et al. PD-1 checkpoint blockade in patients with relapsed AML after allogeneic stem cell transplantation. Bone Marrow Transplant 2017;52(2):317–320. [DOI] [PubMed] [Google Scholar]
  • **15.Liu Y, Bewersdorf JP, Stahl M, Zeidan AM. Immunotherapy in acute myeloid leukemia and myelodysplastic syndromes: The dawn of a new era? Blood Rev 2019;34:67–83. [DOI] [PubMed] [Google Scholar]; comprehensive review on various modalities of immune therapy in myeloid malignancies
  • 16.Zeidan AM, Knaus HA, Robinson TM, et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin Cancer Res 2018;24(15):3519–3527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003;348(3):203–213. [DOI] [PubMed] [Google Scholar]
  • 18.Lamble AJ, Lind EF. Targeting the Immune Microenvironment in Acute Myeloid Leukemia: A Focus on T Cell Immunity. Front Oncol 2018;8:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Schumacher K, Haensch W, Roefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res 2001;61(10):3932–3936. [PubMed] [Google Scholar]
  • 20.Ismail MM, Abdulateef NAB. Bone marrow T-cell percentage: A novel prognostic indicator in acute myeloid leukemia. Int J Hematol 2017;105(4):453–464. [DOI] [PubMed] [Google Scholar]
  • 21.Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 2014;20(19):5064–5074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.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(17):1627–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372(4):320–330. [DOI] [PubMed] [Google Scholar]
  • 24.Alatrash G, Daver N, Mittendorf EA. Targeting Immune Checkpoints in Hematologic Malignancies. Pharmacological Reviews 2016;68(4):1014–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Graf M, Reif S, Hecht K, et al. High expression of costimulatory molecules correlates with low relapse-free survival probability in acute myeloid leukemia (AML). Ann Hematol 2005;84(5):287–297. [DOI] [PubMed] [Google Scholar]
  • 26.Whiteway A, Corbett T, Anderson R, Macdonald I, Prentice HG. Expression of co-stimulatory molecules on acute myeloid leukaemia blasts may effect duration of first remission. Br J Haematol 2003;120(3):442–451. [DOI] [PubMed] [Google Scholar]
  • 27.Yang H, Bueso-Ramos C, DiNardo C, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2014;28(6):1280–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen X, Liu S, Wang L, Zhang W, Ji Y, Ma X. Clinical significance of B7-H1 (PD-L1) expression in human acute leukemia. Cancer Biol Ther 2008;7(5):622–627. [DOI] [PubMed] [Google Scholar]
  • 29.Daver N, Basu S, Garcia-Manero G, et al. Abstract 3205: Defining the immune checkpoint landscape of acute myeloid leukemia (AML). Cancer Research 2016;76(14 Supplement):3205–3205. [Google Scholar]
  • 30.Szczepanski MJ, Szajnik M, Czystowska M, et al. Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin Cancer Res 2009;15(10):3325–3332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Shenghui Z, Yixiang H, Jianbo W, et al. Elevated frequencies of CD4(+) CD25(+) CD127lo regulatory T cells is associated to poor prognosis in patients with acute myeloid leukemia. Int J Cancer 2011;129(6):1373–1381. [DOI] [PubMed] [Google Scholar]
  • 32.Kanakry CG, Hess AD, Gocke CD, et al. Early lymphocyte recovery after intensive timed sequential chemotherapy for acute myelogenous leukemia: peripheral oligoclonal expansion of regulatory T cells. Blood 2011;117(2):608–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Le Dieu R, Taussig DC, Ramsay AG, et al. Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood 2009;114(18):3909–3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vidriales MB, Orfao A, Lopez-Berges MC, et al. Lymphoid subsets in acute myeloid leukemias: increased number of cells with NK phenotype and normal T-cell distribution. Ann Hematol 1993;67(5):217–222. [DOI] [PubMed] [Google Scholar]
  • 35.Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol 2015;8:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **36.Williams P, Basu S, Garcia-Manero G, et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the largest study investigating the phenotypic profile of T-cells in the bone marrow of patients at various stages of the disease course. The authors identified distinct subpopulations that could be potential targets for combination therapies.
  • 37.Panoskaltsis N, Reid CD, Knight SC. Quantification and cytokine production of circulating lymphoid and myeloid cells in acute myelogenous leukaemia. Leukemia 2003;17(4):716–730. [DOI] [PubMed] [Google Scholar]
  • 38.Deng M, Gui X, Kim J, et al. LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature 2018;562(7728):605–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhou Q, Munger ME, Veenstra RG, et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 2011;117(17):4501–4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kong Y, Zhang J, Claxton DF, et al. PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J 2015;5:e330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chokr N, Patel R, Wattamwar K, Chokr S. The Rising Era of Immune Checkpoint Inhibitors in Myelodysplastic Syndromes. Advances in Hematology 2018;2018:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Behrmann L, Wellbrock J, Fiedler W. Acute Myeloid Leukemia and the Bone Marrow Niche-Take a Closer Look. Front Oncol 2018;8:444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sehgal A, Whiteside TL, Boyiadzis M. PD-1 Checkpoint Blockade in Acute Myeloid Leukemia. Expert opinion on biological therapy 2015;15(8):1191–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 2000;192(2):303–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Costello RT, Mallet F, Sainty D, Maraninchi D, Gastaut JA, Olive D. Regulation of CD80/B7–1 and CD86/B7–2 molecule expression in human primary acute myeloid leukemia and their role in allogenic immune recognition. Eur J Immunol 1998;28(1):90–103. [DOI] [PubMed] [Google Scholar]
  • 46.Saudemont A, Quesnel B. In a model of tumor dormancy, long-term persistent leukemic cells have increased B7-H1 and B7.1 expression and resist CTL-mediated lysis. Blood 2004;104(7):2124–2133. [DOI] [PubMed] [Google Scholar]
  • 47.Laurent S, Palmisano GL, Martelli AM, et al. CTLA-4 expressed by chemoresistant, as well as untreated, myeloid leukaemia cells can be targeted with ligands to induce apoptosis. Br J Haematol 2007;136(4):597–608. [DOI] [PubMed] [Google Scholar]
  • 48.Fevery S, Billiau AD, Sprangers B, et al. CTLA-4 blockade in murine bone marrow chimeras induces a host-derived antileukemic effect without graft-versus-host disease. Leukemia 2007;21(7):1451–1459. [DOI] [PubMed] [Google Scholar]
  • 49.Bai XF, Liu J, May KF Jr., Guo Y, Zheng P, Liu Y. B7-CTLA4 interaction promotes cognate destruction of tumor cells by cytotoxic T lymphocytes in vivo. Blood 2002;99(8):2880–2889. [DOI] [PubMed] [Google Scholar]
  • 50.LaBelle JL, Hanke CA, Blazar BR, Truitt RL. Negative effect of CTLA-4 on induction of T-cell immunity in vivo to B7–1+, but not B7–2+, murine myelogenous leukemia. Blood 2002;99(6):2146–2153. [DOI] [PubMed] [Google Scholar]
  • 51.Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363(8):711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hutloff A, Dittrich AM, Beier KC, et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 1999;397(6716):263–266. [DOI] [PubMed] [Google Scholar]
  • 53.Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192(7):1027–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Garcia-Diaz A, Shin DS, Moreno BH, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep 2017;19(6):1189–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Berthon C, Driss V, Liu J, et al. In acute myeloid leukemia, B7-H1 (PD-L1) protection of blasts from cytotoxic T cells is induced by TLR ligands and interferon-gamma and can be reversed using MEK inhibitors. Cancer Immunol Immunother 2010;59(12):1839–1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood 2009;114(8):1545–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Berger R, Rotem-Yehudar R, Slama G, et al. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res 2008;14(10):3044–3051. [DOI] [PubMed] [Google Scholar]
  • 58.Garcia-Manero G, Tallman MS, Martinelli G, et al. Pembrolizumab, a PD-1 Inhibitor, in Patients with Myelodysplastic Syndrome (MDS) after Failure of Hypomethylating Agent Treatment. Blood 2016;128(22):345–345. [Google Scholar]
  • 59.Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell 2012;150(1):12–27. [DOI] [PubMed] [Google Scholar]
  • 60.Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol 2012;30(7):742–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Metzeler KH, Maharry K, Radmacher MD, et al. TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol 2011;29(10):1373–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stahl M, DeVeaux M, Montesinos P, et al. Hypomethylating agents in relapsed and refractory AML: outcomes and their predictors in a large international patient cohort. Blood Adv 2018;2(8):923–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bewersdorf JP, Shallis R, Stahl M, Zeidan AM. Epigenetic therapy combinations in acute myeloid leukemia: what are the options? Ther Adv Hematol 2019;10:2040620718816698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sun Y, Chen BR, Deshpande A. Epigenetic Regulators in the Development, Maintenance, and Therapeutic Targeting of Acute Myeloid Leukemia. Front Oncol 2018;8:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Srivastava P, Paluch BE, Matsuzaki J, et al. Immunomodulatory action of the DNA methyltransferase inhibitor SGI-110 in epithelial ovarian cancer cells and xenografts. Epigenetics 2015;10(3):237–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang LX, Mei ZY, Zhou JH, et al. Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses. PLoS One 2013;8(5):e62924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gbolahan OB, Zeidan AM, Stahl M, et al. Immunotherapeutic Concepts to Target Acute Myeloid Leukemia: Focusing on the Role of Monoclonal Antibodies, Hypomethylating Agents and the Leukemic Microenvironment. Int J Mol Sci 2017;18(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Katoh M Combination immuno-oncology therapy with immune checkpoint blockers targeting PD-L1, PD-1 or CTLA4 and epigenetic drugs targeting MYC and immune evasion for precision medicine. J Thorac Dis 2018;10(3):1294–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wrangle J, Wang W, Koch A, et al. Alterations of immune response of Non-Small Cell Lung Cancer with Azacytidine. Oncotarget 2013;4(11):2067–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell 2017;169(2):361. [DOI] [PubMed] [Google Scholar]
  • 71.Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell 2015;162(5):974–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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(23):2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Daver N, Garcia-Manero G, Basu S, et al. Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Non-randomized, Open-label, Phase 2 Study. Cancer Discov 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **74.Daver N, Boddu P, Garcia-Manero G, et al. Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Leukemia 2018;32(5):1094–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]; This very important trial is the first to show synergistic effects of HMAs and ICB in myeloid malignancies with excellent response rates especially in the HMA-naïve setting
  • 75.Lindblad KE, Thompson J, Gui G, et al. Pembrolizumab and Decitabine for Refractory or Relapsed Acute Myeloid Leukemia. Blood 2018;132(Suppl 1):1437–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chien KS, Cortes JE, Borthakur G, et al. Preliminary Results from a Phase II Study of the Combination of Azacitidine and Pembrolizumab in Patients with Higher-Risk Myelodysplastic Syndrome. Blood 2018;132(Suppl 1):464–464.30072413 [Google Scholar]
  • 77.Garcia-Manero G, Sasaki K, Montalban-Bravo G, et al. A Phase II Study of Nivolumab or Ipilimumab with or without Azacitidine for Patients with Myelodysplastic Syndrome (MDS). Blood 2018;132(Suppl 1):465–465.30072414 [Google Scholar]
  • 78.Wang DY, Salem JE, Cohen JV, et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Vereecque R, Buffenoir G, Gonzalez R, et al. gamma-ray irradiation induces B7.1 expression in myeloid leukaemic cells. Br J Haematol 2000;108(4):825–831. [DOI] [PubMed] [Google Scholar]
  • 80.Vereecque R, Saudemont A, Quesnel B. Cytosine arabinoside induces costimulatory molecule expression in acute myeloid leukemia cells. Leukemia 2004;18(7):1223–1230. [DOI] [PubMed] [Google Scholar]
  • 81.Assi R, Kantarjian HM, Daver NG, et al. Results of a Phase 2, Open-Label Study of Idarubicin (I), Cytarabine (A) and Nivolumab (Nivo) in Patients with Newly Diagnosed Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS). Blood 2018;132(Suppl 1):905–905. [Google Scholar]
  • 82.Jongen-Lavrencic M, Grob T, Hanekamp D, et al. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N Engl J Med 2018;378(13):1189–1199. [DOI] [PubMed] [Google Scholar]
  • 83.Kadia TM, Cortes JE, Ghorab A, et al. Nivolumab (Nivo) maintenance (maint) in high-risk (HR) acute myeloid leukemia (AML) patients. Journal of Clinical Oncology 2018;36(15_suppl):7014–7014. [Google Scholar]
  • 84.Soiffer RJ, Davids MS, Chen YB. Tyrosine kinase inhibitors and immune checkpoint blockade in allogeneic hematopoietic cell transplantation. Blood 2018;131(10):1073–1080. [DOI] [PubMed] [Google Scholar]
  • 85.Soiffer RJ, Chen YB. Pharmacologic agents to prevent and treat relapse after allogeneic hematopoietic cell transplantation. Blood Adv 2017;1(25):2473–2482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Vago L, Perna SK, Zanussi M, et al. Loss of Mismatched HLA in Leukemia after Stem-Cell Transplantation. New England Journal of Medicine 2009;361(5):478–488. [DOI] [PubMed] [Google Scholar]
  • 87.Toffalori C, Cavattoni I, Deola S, et al. Genomic loss of patient-specific HLA in acute myeloid leukemia relapse after well-matched unrelated donor HSCT. Blood 2012;119(20):4813–4815. [DOI] [PubMed] [Google Scholar]
  • 88.Bashey A, Medina B, Corringham S, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood 2009;113(7):1581–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood 2017;129(18):2471–2478. [DOI] [PubMed] [Google Scholar]
  • 90.Haverkos BM, Abbott D, Hamadani M, et al. PD-1 blockade for relapsed lymphoma post–allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. Blood 2017;130(2):221–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunological reviews 2017;276(1):97–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Haubner S, Perna F, Kohnke T, et al. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Roth CG, Garner K, Eyck ST, Boyiadzis M, Kane LP, Craig FE. TIM3 expression by leukemic and non-leukemic myeloblasts. Cytometry B Clin Cytom 2013;84(3):167–172. [DOI] [PubMed] [Google Scholar]
  • 94.Ruvolo PP. Galectins as regulators of cell survival in the leukemia niche. Adv Biol Regul 2018. [DOI] [PubMed] [Google Scholar]
  • 95.Anderson AC, Anderson DE, Bregoli L, et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science 2007;318(5853):1141–1143. [DOI] [PubMed] [Google Scholar]
  • 96.Kikushige Y, Shima T, Takayanagi S-i, et al. TIM-3 Is a Promising Target to Selectively Kill Acute Myeloid Leukemia Stem Cells. Cell Stem Cell 2010;7(6):708–717. [DOI] [PubMed] [Google Scholar]
  • 97.Sakai K, Kawata E, Ashihara E, et al. Galectin-9 ameliorates acute GVH disease through the induction of T-cell apoptosis. Eur J Immunol 2011;41(1):67–75. [DOI] [PubMed] [Google Scholar]
  • 98.Ozkazanc D, Yoyen-Ermis D, Tavukcuoglu E, Buyukasik Y, Esendagli G. Functional exhaustion of CD4(+) T cells induced by co-stimulatory signals from myeloid leukaemia cells. Immunology 2016;149(4):460–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.John S, Chen H, Deng M, et al. A Novel Anti-LILRB4 CAR-T Cell for the Treatment of Monocytic AML. Mol Ther 2018;26(10):2487–2495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Linch SN, McNamara MJ, Redmond WL. OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal. Front Oncol 2015;5:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nuebling T, Schumacher CE, Hofmann M, et al. The Immune Checkpoint Modulator OX40 and Its Ligand OX40L in NK-Cell Immunosurveillance and Acute Myeloid Leukemia. Cancer Immunol Res 2018;6(2):209–221. [DOI] [PubMed] [Google Scholar]
  • 102.Curti BD, Kovacsovics-Bankowski M, Morris N, et al. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res 2013;73(24):7189–7198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ponce LP, Fenn NC, Moritz N, et al. SIRPalpha-antibody fusion proteins stimulate phagocytosis and promote elimination of acute myeloid leukemia cells. Oncotarget 2017;8(7):11284–11301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Petrova PS, Viller NN, Wong M, et al. TTI-621 (SIRPalphaFc): A CD47-Blocking Innate Immune Checkpoint Inhibitor with Broad Antitumor Activity and Minimal Erythrocyte Binding. Clin Cancer Res 2017;23(4):1068–1079. [DOI] [PubMed] [Google Scholar]
  • 105.Pang WW, Pluvinage JV, Price EA, et al. Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes. Proc Natl Acad Sci U S A 2013;110(8):3011–3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Pietsch EC, Dong J, Cardoso R, et al. Anti-leukemic activity and tolerability of anti-human CD47 monoclonal antibodies. Blood Cancer Journal 2017;7:e536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jaiswal S, Jamieson CH, Pang WW, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009;138(2):271–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 2009;138(2):286–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dorahy DJ, Thorne RF, Fecondo JV, Burns GF. Stimulation of platelet activation and aggregation by a carboxyl-terminal peptide from thrombospondin binding to the integrin-associated protein receptor. J Biol Chem 1997;272(2):1323–1330. [DOI] [PubMed] [Google Scholar]
  • 110.Ranjana H Advani IF, Popplewell Leslie, Forero-Torres Andres, Bartlett Nancy L., Ghosh Nilanjan, Paul Kline Justin, Tran Thu, Lynn Judith, Chen James Y, Agoram Balaji, Huang Jenny, Takimoto Chris H.M., Chao Mark, Smith Sonali M.;. Activity and tolerabilty of the first-in-class anti-CD47 antibody Hu5F9-G4 with rituximab tolerated in relapsed/refractory non-Hodgkin lymphoma: Initial phase 1b/2 results. J Clin Oncol 2018;36. [Google Scholar]
  • 111.Ansell SM, Chen RW, Flinn I, et al. A phase 1 study of TTI-621, a novel immune checkpoint inhibitor targeting CD47, in subjects with relapsed or refractory hematologic malignancies. Journal of Clinical Oncology 2016;34(15_suppl):TPS7585–TPS7585. [Google Scholar]
  • 112.Pyzer AR, Avigan DE, Rosenblatt J. Clinical trials of dendritic cell-based cancer vaccines in hematologic malignancies. Hum Vaccin Immunother 2014;10(11):3125–3131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Weinstock M, Rosenblatt J, Avigan D. Dendritic Cell Therapies for Hematologic Malignancies. Mol Ther Methods Clin Dev 2017;5:66–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lim TS, Chew V, Sieow JL, et al. PD-1 expression on dendritic cells suppresses CD8(+) T cell function and antitumor immunity. Oncoimmunology 2016;5(3):e1085146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Krempski J, Karyampudi L, Behrens MD, et al. Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer. J Immunol 2011;186(12):6905–6913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Park JH, Riviere I, Gonen M, et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N Engl J Med 2018;378(5):449–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013;5(177):177ra138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Locke FL, Neelapu SS, Bartlett NL, et al. Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma. Mol Ther 2017;25(1):285–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 2015;33(6):540–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Brudno JN, Somerville RP, Shi V, et al. Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress After Allogeneic Hematopoietic Stem-Cell Transplantation Without Causing Graft-Versus-Host Disease. J Clin Oncol 2016;34(10):1112–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yoon DH, Osborn MJ, Tolar J, Kim CJ. Incorporation of Immune Checkpoint Blockade into Chimeric Antigen Receptor T Cells (CAR-Ts): Combination or Built-In CAR-T. International journal of molecular sciences 2018;19(2):340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chong EA, Melenhorst JJ, Lacey SF, et al. PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: refueling the CAR. Blood 2017;129(8):1039–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncol 2018;4(2):173–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Garcia-Manero G, Daver NG, Montalban-Bravo G, et al. A Phase II Study Evaluating the Combination of Nivolumab (Nivo) or Ipilimumab (Ipi) with Azacitidine in Pts with Previously Treated or Untreated Myelodysplastic Syndromes (MDS). Blood 2016;128(22):344–344. [Google Scholar]
  • 125.Garcia-Manero G, Schiffer CA, Godley LA, et al. Phase 1 study to evaluate the safety and tolerability of MEDI4736, an anti-programmed cell death ligand-1 (PD-L1) antibody, in myelodysplastic syndrome (MDS) after treatment with hypomethylating agents. Journal of Clinical Oncology 2015;33(15_suppl):TPS7103–TPS7103. [Google Scholar]
  • 126.Silverman LR, Dohner H, Dombret H, et al. A randomized, open-label, phase II study of azacitidine (AZA) in combination with durvalumab in patients (pts) with previously untreated higher-risk myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) ineligible for hematopoietic stem cell transplantation (HSCT). Journal of Clinical Oncology 2017;35(15_suppl):TPS7074–TPS7074. [Google Scholar]

RESOURCES