Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jan 1.
Published in final edited form as: Cancer J. 2018 Jan-Feb;24(1):31–35. doi: 10.1097/PPO.0000000000000297

Clinical Development of PD-1 Blockade in Hematologic Malignancies

Matthew J Pianko 1, Aaron D Goldberg 1, Alexander M Lesokhin 1
PMCID: PMC5785103  NIHMSID: NIHMS919162  PMID: 29360725

Abstract

Clinical development of Immune checkpoint inhibitors targeting the programmed cell death 1 (PD-1) pathway has led to clinical benefits for patients with multiple solid tumor and hematologic malignancies and has revolutionized modern oncology. High response rates to PD-1 blockade in patients with classical Hodgkin lymphoma and certain subtypes of non-Hodgkin lymphoma highlight an intrinsic biologic sensitivity to this strategy of treatment. Despite early success of checkpoint inhibitor and immunomodulatory drug combinations in phase II studies in multiple myeloma, safety concerns in patients treated with the combination of immunomodulatory drugs and checkpoint inhibitors in myeloma have stalled drug development in this space. Novel combination approaches exploring PD-1 inhibitors with epigenetic modifiers in leukemia are underway.

Keywords: immune checkpoint inhibitors, hematologic malignancies, leukemia, lymphoma, myeloma

Introduction

As the key mediators of host immunosurveillance, effector T cells are tasked with the maintenance of host immune homeostasis through elimination of intracellular pathogens and neoplasia while also minimizing autoimmune responses and collateral damage to host tissues. At the level of the tissue microenvironment, T cell activation or inhibition requires co-stimulatory or co-inhibitory signaling following T cell receptor (TCR) recognition of its cognate peptide presented in the context of major histocompatibility complex (MHC) I or II receptor on the cell surface. A host of co-stimulatory and co-inhibitory ligand-receptor interactions known as immune checkpoints then ultimately determine T cell fate within the spectrum of activation or inhibition by modulating a balance between expansion, exertion of cellular cytotoxicity and memory formation versus anergy, exhaustion or senescence1.

Clinical development of monoclonal antibodies that block immune checkpoints cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death 1 receptor (PD-1), and programmed death-ligand 1 (PD-L1) has resulted in FDA approvals for use of these agents in multiple solid and hematologic tumors. Herein, we focus on the current status of clinical development of PD-1 pathway inhibitors for the treatment of hematologic malignancies.

The PD-1 Pathway

Biology of PD-1 and its ligands PD-L1 (B7-H1) and PD-L2 (B7-DC)

Programmed cell death 1 receptor (PD-1, CD279) is a type I transmembrane immunoreceptor expressed by activated T cells, natural killer (NK) cells, and B cells cells2,3,4. To prevent immune-related damage to self after binding of TCR to MHC-peptide, T cell activation is inhibited through binding of PD-1 on T cells to its ligands PD-L1 (B7-H1, CD274) and/or PD-L2 (B7-DC, CD273) that are normally expressed on peripheral tissues. PD-1 ligands PD-L1 and PD-L2 are known to be expressed in the tumor microenvironment by both tumor cells{Dong, 2002 #2406;Zou, 2008 #2630} and non-tumor immune and stromal cells{Curiel, 2003 #3251;Zou, 2008 #2630;Wu, 2009 #3249;Nazareth, 2007 #3254;Flies, 2011 #998;Dong, 2002 #2406}.

Binding of PD-1 to its ligands act to inhibit cytotoxic T cell activity via an intracellular signaling cascade involving several kinases downstream of PD-15. Continued PD-1 signaling is important in development of T cell exhaustion, a state in which T cells lose their capacity for cytotoxicity6. Antitumor activity following blockade of the PD-1 axis suggests PD-1 signaling is important for immune escape and tumor persistence, an idea supported by clinical efficacy of PD-1 pathway blockade in multiple tumor types and several hematologic cancers.

PDL1 is encoded on the long arm of chromosome 9p under the control of an AP1 sensitive promoter region downstream of JAK-2-STAT signaling cascade. Expression of PD-L1 is upregulated by many cytokines, most potently by interferon-γ (IFN-γ)8. Interferon-mediated upregulation of PD-L1 and PD-L2 is an important feature of several hematologic cancers that reside in a pro-inflammatory tumor microenvironment, including acute myeloid leukemia11 and multiple myeloma12,13. In addition to cytokine-mediated mechanisms of inducible PD-L1 overexpression, oncogenic viruses relevant to lymphomagenesis such as EBV are directly capable of driving PD-L1 expression via downstream effects of latent membrane protein-1 (LMP-1) on nuclear factor kappa B (NF-κB) signaling and the AP-1 sensitive promoter of PD-L114,15. PD-L1 can also be overexpressed by STAT3 upregulation via upstream activation of ALK16. Correlative analysis of ongoing trials in hematologic malignancies is needed to explore the contribution of these and other mechanisms to response and resistance to PD-1 pathway inhibition.

Classical Hodgkin lymphoma (CHL)

Early phase clinical trials of PD-1 pathway blockade highlighted that certain lymphomas, especially classical Hodgkin lymphoma (cHL), harbor an intrinsic susceptibility to PD-1 pathway blockade. Hodgkin lymphoma is defined by the presence of the Hodgkin Reed Sternberg (HRS) cell among a robust inflammatory infiltrate and is cured with chemotherapy or combined modality therapy in most cases. However, a need exists for cHL patients who relapse or are refractory to existing therapies. In several pre-clinical studies in cHL, near universal overexpression of PD-L1 on the HRS cell likely driven by 9p24.1 amplification were noted, suggesting a biologic importance to the PD-1 pathway in cHL17.

Supporting the importance of PD-1 signaling to the pathogenesis of cHL, a significant efficacy signal was apparent in the phase I clinical trials of nivolumab (Opdivo, Bristol-Myers Squibb), a humanized IgG1 kappa monoclonal antibody, and pembrolizumab (Keytruda, Merck), a humanized IgG4 kappa monoclonal antibody, both of which block the interaction of PD-1 with its ligands PD-L1 and PD-L2. Responses to PD-1 blockade were directly related to the degree of amplification of 9p24.118. In the CheckMate039 phase I clinical trial (nivolumab), relapsed/refractory (R/R) cHL patients had an overall response rate (ORR) of 87% with 17% reaching a complete response (CR), and 70% with a partial response (PR)19,20. Among 31 patients with cHL with relapse after brentuximab vedotin (BV) and autologous stem cell transplant (ASCT) or prior ineligibility for treatment with BV, the ORR was 58%, CR rate 19%, and PR rate 12% with median follow-up of 24.9 months21. The phase II study of pembrolizumab, KEYNOTE-087, evaluated 210 patients among three cohorts, in whom the overall response rate was 69%, and the overall CR rate was 22.4%22. The ORR was 73.9% in R/R cHL patients with progressive lymphoma after ASCT and BV, 64.2% in patients with chemo-resistant disease ineligible for ASCT, and 70.0% in BV-naïve patients with relapsed disease after ASCT. These important studies led the Food and Drug Administration (FDA) to grant accelerated approval to nivolumab and pembrolizumab for the treatment of Hodgkin lymphoma.

Ongoing studies are evaluating use of PD-1 blockade for cHL in the frontline setting and in a variety of patient populations including those who are transplant ineligible, early stage unfavorable disease, and in patients older than 60. Interim results from a phase I/II study of nivolumab plus BV as first salvage after upfront chemotherapy before ASCT showed a CR rate of 63% among 59 patients, a rate much higher than expected with either agent as a monotherapy23. In sum, PD-1 pathway blockade is very likely to transform the treatment landscape for cHL patients in the coming years.

Non-Hodgkin lymphoma (NHL)

Substantial heterogeneity exists among non-Hodgkin lymphomas. Within this large group of malignancies there have been some responses to PD-1 pathway blockade. PD-L1 upregulation via genetic aberrations involving 9p24.1 are found commonly in primary mediastinal B cell lymphoma (PMBL)24, primary testicular lymphoma25, plasmablastic lymphoma, HHV-8 associated primary effusion lymphoma, and primary central nervous system lymphomas25,26, and several of these tumors have been sensitive to PD-1 blockade. Promising activity of pembrolizumab for PMBL was observed in a phase Ib study with 41% responding to treatment among 17 patients24. Chronic viral infection with EBV also plays a role in PD-L1 upregulation, and high response rates to PD-1 blockade in extranodal NK/T cell lymphoma27, EBV(+) and EBV(−) lymphoproliferative disorders have been reported.

Diffuse large B cell lymphoma (DLBCL) represents a heterogeneous group of aggressive malignancies derived from mature B lymphocytes. A retrospective series of 1253 samples from patients with DLBCL showed only 11% were PD-L1 positive and those patients with PD-L1 had a poor prognosis28. In addition, DLBCL patients with a high International Prognostic Index (IPI) score and elevated soluble PD-L1 (sPD-L1) have an inferior prognosis, with a 3 year overall survival of 40.9% in patients with high sPD-L1 compared with 82.1% in those without elevated sPD-L129. In a signal-seeking phase Ib study, 36% of unselected patients with DLBCL responded to treatment with nivolumab30.

PD-1 can also be upregulated on peripheral T cell lymphomas derived from T follicular helper cells (TFH) including angioimmunoblastic T cell lymphoma (AITL), follicular T cell lymphoma, and nodal peripheral T cell lymphoma with TFH phenotype31. A phase II study in mycosis fungoides/Sézary syndrome (MF/SS) demonstrated a response rate of 38% in 24 patients32.

Evaluation for biomarkers predictive of response to PD-1 pathway blockade in lymphoma may aid in optimal patient selection for prospective clinical trials. Markers of immune exhaustion LAG-3 (lymphocyte activation gene 3) and TIM-3 (T-cell immunoglobulin and mucin-domain containing-3) have been found to be co-expressed in T cell infiltrates in NHL, and these may represent viable targets alone or in combination with PD-1 blockade33,34. Biomarkers of poor response to therapy also warrant attention; beta-2 microglobulin (B2M) mutations and loss of MHC-I expression are relevant in melanoma resistant to PD-1 pathway blockade35. Correlation of these biomarkers with outcome after checkpoint blockade in cHL or NHL have not yet been reported, although it is known B2M mutations and loss of MHC-II are frequent findings in cHL.

Multiple Myeloma

Multiple myeloma (MM) is a clonally heterogeneous hematologic malignancy derived from terminally differentiated B cells known as plasma cells which secrete clonal monoclonal immunoglobulin paraproteins and whose effects in the bone marrow tumor microenvironment cause systemic multifactorial cellular and humoral immunodeficiency. The MM bone marrow tumor microenvironment is characterized by profound immunosuppression related to inhibitory cytokines3640, suppressive cells such as myeloid-derived suppressor cells41,42, and regulatory T cells4345, and known active PD-1/PD-L1 signaling12. In contrast to plasma cells in premalignant states, MM cells express PD-L1 driven by IFN-gamma, toll-like receptor, Akt, and Ras signaling12,46, and its expression is functionally associated with reduced susceptibility to effector T cell killing12. PD-1 is expressed on marrow infiltrating T cells, and PD-1 ligands are expressed and upregulated on malignant plasma cells in MM47. Multifactorial immunosuppression within the MM tumor microenvironment inhibits MM-specific tumor infiltrating T cells, which retain the capacity for recognizing and killing MM cells ex vivo, suggestive of a potential benefit to reversing local immunosuppression in the tumor microenvironment47. A large retrospective study recently found that MM patients with a high mutational and neoantigen burden in MM have inferior PFS and outcomes48, a potentially provocative result given that sensitivity to immune checkpoint blockade was observed among patients with high mutation burden solid tumors.

Despite evidence suggesting importance of the PD-1 pathway in MM, there was no clinical activity seen in the 27 patients treated with nivolumab on the CheckMate 039 study30. Pre-clinical data supported a synergistic role for immunomodulatory drugs (IMiDs) with PD-1 pathway blockade in patients with myeloma, as IMiDs polarize T cells toward a Th1 phenotype and promote NK cell cytotoxicity42,49, inhibit regulatory T cells (Treg)50, and downregulate PD-L1 on tumor cells42,49. Preliminary results of PD-1 pathway blockade combined with IMiDs were encouraging. The phase I study of pembrolizumab with lenalidomide and dexamethasone (KEYNOTE-023, NCT02036502) showed a response to therapy in 20/40 evaluable patients (ORR 50%) with 5 patients reaching VGPR, and 14 patients achieving PR51. In the lenalidomide-refractory patients, the ORR to pembrolizumab plus lenalidomide and dexamethasone was 38% (11/29)51. In patients with detectable bone marrow plasma cells by flow cytometry, 100% expressed PD-L1, while PD-L2 expression was variable51. The phase II study of pembrolizumab plus pomalidomide and dexamethasone showed an ORR of 60%, with 8% of patients achieving a strict complete remission. In patients previously refractory to both proteasome inhibitors and IMiDs the response rate was 68%52.

Building on these results, two phase III studies of pembrolizumab in combination with lenalidomide and dexamethasone (KEYNOTE-185, NCT02579863) or pomalidomide and dexamethasone (KEYNOTE-183, NCT02576977) and one phase III study of pomalidomide and dexamethasone vs. nivolumab, pomalidomide, and dexamethasone vs. nivolumab, elotuzumab, pomalidomide and dexamethasone (CheckMate 602, NCT02726581) were initiated. However, on June 12, 2017, following a review of data by the studies’ data monitoring committee, the FDA requested a stop to new accrual to these studies due to increased patient deaths on the trial arms receiving experimental therapy with pembrolizumab in combination with immunomodulatory drugs lenalidomide or pomalidomide53. An investigation of the nature of these deaths are underway, and on July 3, 2017, the FDA required that all patients on these trials be discontinued from further treatment54. Interim analysis of the KEYNOTE-183 data using a data cutoff date of June 2 showed a higher relative risk of death for patients in these studies receiving pembrolizumab plus pomalidomide and dexamethasone over pomalidomide and dexamethasone (HR 1.61, (5% CI 0.91–2.85), and an objective response rate of 34% compared to 40% in the control arm55. In the KEYNOTE-185 study, the hazard ratio of the pembrolizumab-containing investigational arm was 2.06 (95% CI: 0.93–4.55), and an ORR of 64% in the investigational arm compared with ORR 62% in the control arm55. Until further analysis can be performed other studies including PD-1 and PD-L1 blocking antibodies in myeloma have been placed on hold by the FDA as a precaution54. In addition, durvalumab trials in combination with immunomodulatory agents in patients with chronic lymphocytic leukemia (CLL) or lymphoma were also placed on hold56. The pembrolizumab phase III results in MM highlight a critical need to explore immune regulation at the interface between malignant plasma cells and the tumor microenvironment. Further analysis and followup of patients accrued to both the pembrolizumab and other IMiD/anti-PD-1 or anti-PD-L1 trials will hopefully elucidate if there are patient or disease factors that predict for benefit or toxicity of this therapy in MM.

Leukemia

PD-1 pathway blockade is currently under exploration for treatment of myeloid hematologic cancers, often in combination with other epigenetic and immunomodulatory agents. In contrast to lymphoma, there have been limited clinical results reported to date of PD-1 pathway blockade in patients with leukemia. However, pre-clinical data suggest a potential role for the PD-1 pathway in patients with myeloid malignancies. In myelodysplastic syndromes (MDS), PD-L1 is expressed by CD34+ blasts in a significant subset of patient samples (~36%), while stroma and non-blast cellular components express PD-157. In acute myeloid leukemia (AML), PD-L1 expression on AML cells is negatively correlated with outcome5759. Although PD-L1 expression is low on most cases of de novo AML, interferon-induced PD-L1 expression increases on AML blast cells during treatment, and PD-L1 expression is higher in patients with relapsed AML than in de novo patients11,58,60. Pre-clinical mouse models also point towards the PD-1/PD-L1 pathway in immune evasion by AML.6164 Hypomethylating agents (HMAs) upregulate immune checkpoints57, further suggesting an appealing treatment strategy of combination epigenetic therapy and immunotherapy for myeloid malignancies. HMAs also trigger expression of endogenous retroviruses in multiple human tumor types, activating a cellular viral defense program mediated by interferon-β, and strongly potentiating checkpoint blockade in mouse models65,66. A phase II study is underway in MDS evaluating nivolumab or ipilimumab combined with 5-azacitidine67. Nivolumab as a single agent had no activity in MDS, but early data showed an overall response rate of 69% for combination azacitidine and nivolumab in patients previously failing azaciditine67. Preliminary clinical data for nivolumab in combination with azacitidine in patients with relapsed or refractory AML also suggest promising overall response rates (ORR) of 34% (N=53) in this high risk patient population.68 There are several trials underway evaluating nivolumab in combination with other agents in precursor B acute lymphoblastic leukemia (B-ALL) (NCT02879695) and acute myeloid leukemia (AML) (NCT 02532231). Pembrolizumab trials are underway in B-ALL and T acute lymphoblastic leukemia (T-ALL) (NCT02767934), AML (transplant-ineligible) (NCT02771197), and R/R CLL (NCT02332980).

Toxicity and Immune Related Adverse Events

Immune checkpoint blockade has been clearly associated with unique immune mediated toxicities. With single agent therapy, the side effect profile in hematologic malignancies has been similar to that seen in solid tumor patients; and side effects can be readily reversed with established algorithms for immunosuppression. Specific consideration to overlapping toxicities in combination studies of PD-1 pathway blockade and drugs routinely used in hematologic malignancies are beyond the scope of this review. Nevertheless, therapies that cause pneumonitis, such as radiation, brentuximab vedotin, bleomycin for treatment of Hodgkin disease, and IMiDs for treatment of MM highlight some of the potential challenges of discerning causality in the face of toxicity. Another specific scenario is in the setting of stem cell transplantation, a therapy unique to hematologic cancer. Several preclinical studies demonstrated augmentation of graft-versus-tumor (GVT) effects69,70, as well as graft-versus-host-disease (GVHD)71. After four treatment related deaths were noted among 39 patients receiving PD-1 blockade before allogeneic stem cell transplant, the FDA added a warning to the package insert for nivolumab72, urging vigilance for early evidence of GVH after allogeneic stem cell transplant such as severe acute GVHD, hepatic veno-occlusive disease, steroid-responsive febrile syndromes that my precede GVHD, and other immune-related phenomena47. There are limited data to support use of PD-1 antibodies after allogeneic stem cell transplant and off-label use in this manner is increasing in frequency73. Three retrospective series report that the high response rate to treatment with PD-1 blockade after allo-HSCT (ORR 77–95%) was accompanied by a rate of severe treatment emergent GVHD of 30–55%7375. A prospective trial of PD-1 blockade as a maintenance therapy after allo-HSCT is ongoing (NCT02985554).

Conclusions

Successes in cHL and subtypes of NHL have led to a surge of investigation into the potential benefits of PD-1 pathway blockade in distinct cohorts of patients with lymphoma. Development of PD-1/PD-L1 blockade in myeloma has been halted for the moment by safety issues that demand thoughtful scrutiny to protect patients and inform future clinical trials. Drug development of PD-1 in leukemias is underway with limited results to date, though preliminary data suggest that combination epigenetic therapy and PD-1 blockade may have some efficacy in myeloid malignancies. Safety issues have also been raised in the context of allogeneic SCT. These observations highlight a multitude of disease specific challenges facing investigators seeking to treat cancers of immune cell origins with therapies that potently modulate the immune response.

Successes in immune checkpoint blockade have also fueled enthusiasm for other novel immunotherapies such as adoptive T cell therapy with chimeric antigen receptor T cells, novel antibody engineering products such as bispecific T cell engager proteins (BiTEs), and tumor vaccines or immunotherapies directed at shared tumor antigens or patient-specific neoantigens. Development of novel biomarkers predictive of response and failure of these therapies in hematologic malignancies is critical to expanding the number of patients who derive durable benefits and to evaluate for rationale to combine with immune checkpoint blockade in search of durable remissions or cures for a greater number of patients.

Acknowledgments

Funding Support

This work was supported in part by the Memorial Sloan Kettering Cancer Center (MSKCC) NCI core grant [P30 CA008748]. M.J.P. receives support from the MSKCC Mortimer J. Lacher Fellowship supported by the Lymphoma Foundation, the MSK Sawiris Foundation, and the NIH/National Center for Advancing Translational Sciences (UL1TR00457), administered by the Clinical and Translational Science Center at Weill Cornell Medical Center and MSKCC. A.D.G. receives support from the Young Investigator Award from the Conquer Cancer Foundation. A.M.L. is a member of the Parker Institute for Cancer Immunotherapy. A.M.L. and also receives support from the MSK Sawiris Foundation and from Mr. William H. Goodwin and Mrs. Alice Goodwin and the Commonwealth Foundation for Cancer Research and The Center for Experimental Therapeutics at Memorial Sloan Kettering Cancer Center.

Footnotes

Disclosures: M.J.P.: No disclosures

A.D.G.: Research: ADC Therapeutics, Genentech, Pfizer. Consultancy: Celgene.

A.M.L.: Research: BMS, Novartis, Celgene, Genentech, Merck. Consultancy: BMS, Novartis.

References

  • 1.Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: Enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med. 2015;7:280sr1. doi: 10.1126/scitranslmed.3010274. [DOI] [PubMed] [Google Scholar]
  • 2.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;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 3.Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13:227–42. doi: 10.1038/nri3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Flies DB, Sandler BJ, Sznol M, Chen L. Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy. The Yale journal of biology and medicine. 2011;84:409–21. [PMC free article] [PubMed] [Google Scholar]
  • 5.Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell immunity. Curr Opin Immunol. 2007;19:309–14. doi: 10.1016/j.coi.2007.04.012. [DOI] [PubMed] [Google Scholar]
  • 6.Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–9. doi: 10.1038/ni.2035. [DOI] [PubMed] [Google Scholar]
  • 7.Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8:467–77. doi: 10.1038/nri2326. [DOI] [PubMed] [Google Scholar]
  • 8.Curiel TJ, Wei S, Dong H, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9:562–7. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • 9.Wu K, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppression of CD8+ T cells in human hepatocellular carcinoma is mediated by B7-H1/programmed death-1 interactions. Cancer Res. 2009;69:8067–75. doi: 10.1158/0008-5472.CAN-09-0901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nazareth MR, Broderick L, Simpson-Abelson MR, Kelleher RJ, Jr, Yokota SJ, Bankert RB. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. J Immunol. 2007;178:5552–62. doi: 10.4049/jimmunol.178.9.5552. [DOI] [PubMed] [Google Scholar]
  • 11.Kronig H, Kremmler L, Haller B, et al. Interferon-induced programmed death-ligand 1 (PD-L1/B7-H1) expression increases on human acute myeloid leukemia blast cells during treatment. Eur J Haematol. 2014;92:195–203. doi: 10.1111/ejh.12228. [DOI] [PubMed] [Google Scholar]
  • 12.Liu J, Hamrouni A, Wolowiec D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110:296–304. doi: 10.1182/blood-2006-10-051482. [DOI] [PubMed] [Google Scholar]
  • 13.Garcia-Diaz A, Shin DS, Moreno BH, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017;19:1189–201. doi: 10.1016/j.celrep.2017.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012;18:1611–8. doi: 10.1158/1078-0432.CCR-11-1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bi XW, Wang H, Zhang WW, et al. PD-L1 is upregulated by EBV-driven LMP1 through NF-kappaB pathway and correlates with poor prognosis in natural killer/T-cell lymphoma. J Hematol Oncol. 2016;9:109. doi: 10.1186/s13045-016-0341-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Marzec M, Zhang Q, Goradia A, et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1) Proc Natl Acad Sci U S A. 2008;105:20852–7. doi: 10.1073/pnas.0810958105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116:3268–77. doi: 10.1182/blood-2010-05-282780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Roemer MG, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J Clin Oncol. 2016;34:2690–7. doi: 10.1200/JCO.2016.66.4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372:311–9. doi: 10.1056/NEJMoa1411087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ansell S, Armand P, Timmerman JM, et al. Nivolumab in Patients (Pts) with Relapsed or Refractory Classical Hodgkin Lymphoma (R/R cHL): Clinical Outcomes from Extended Follow-up of a Phase 1 Study (CA209-039) Blood. 2015;126:583. [Google Scholar]
  • 21.Armand P, Shipp M, Ribrag V, et al. Pembrolizumab in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Long-Term Efficacy from the Phase 1b Keynote-013 Study. Blood. 2016;128 [Google Scholar]
  • 22.Chen R, Zinzani PL, Fanale MA, et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J Clin Oncol. 2017:JCO2016721316. doi: 10.1200/JCO.2016.72.1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Herrera AF, Moskowitz AJ, Bartlett NL, et al. INTERIM RESULTS FROM a PHASE 1/2 STUDY OF BRENTUXIMAB VEDOTIN IN COMBINATION WITH NIVOLUMAB IN PATIENTS WITH RELAPSED OR REFRACTORY HODGKIN LYMPHOMA. Hematological Oncology. 2017;35:85–6. [Google Scholar]
  • 24.Zinzani PL, Ribrag V, Moskowitz CH, et al. Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood. 2017;130:267–70. doi: 10.1182/blood-2016-12-758383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Nayak L, Iwamoto FM, LaCasce A, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood. 2017;129:3071–3. doi: 10.1182/blood-2017-01-764209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Berghoff AS, Ricken G, Widhalm G, et al. PD1 (CD279) and PD-L1 (CD274, B7H1) expression in primary central nervous system lymphomas (PCNSL) Clin Neuropathol. 2014;33:42–9. doi: 10.5414/np300698. [DOI] [PubMed] [Google Scholar]
  • 27.Kim WY, Jung HY, Nam SJ, et al. Expression of programmed cell death ligand 1 (PD-L1) in advanced stage EBV-associated extranodal NK/T cell lymphoma is associated with better prognosis. Virchows Arch. 2016;469:581–90. doi: 10.1007/s00428-016-2011-0. [DOI] [PubMed] [Google Scholar]
  • 28.Kiyasu J, Miyoshi H, Hirata A, et al. Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood. 2015;126:2193–201. doi: 10.1182/blood-2015-02-629600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Rossille D, Gressier M, Damotte D, et al. High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia. 2014;28:2367–75. doi: 10.1038/leu.2014.137. [DOI] [PubMed] [Google Scholar]
  • 30.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;34:2698–704. doi: 10.1200/JCO.2015.65.9789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gravelle P, Burroni B, Pericart S, et al. Mechanisms of PD-1/PD-L1 expression and prognostic relevance in non-Hodgkin lymphoma: a summary of immunohistochemical studies. Oncotarget. 2017 doi: 10.18632/oncotarget.16680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Khodadoust M, Rook AH, Porcu P, et al. Pembrolizumab for Treatment of Relapsed/Refractory Mycosis Fungoides and Sezary Syndrome: Clinical Efficacy in a Citn Multicenter Phase 2 Study. Blood. 2016;128 [Google Scholar]
  • 33.Yang ZZ, Grote DM, Ziesmer SC, Xiu B, Novak AJ, Ansell SM. PD-1 expression defines two distinct T-cell sub-populations in follicular lymphoma that differentially impact patient survival. Blood Cancer J. 2015;5:e281. doi: 10.1038/bcj.2015.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yang ZZ, Price-Troska T, Novak AJ, Ansell S. The Exhausted Intratumoral T Cell Population in B-Cell Non-Hodgkin Lymphoma Is Defined By LAG-3, PD-1 and Tim-3 Expression. Blood. 2015;126 [Google Scholar]
  • 35.Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375:819–29. doi: 10.1056/NEJMoa1604958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cook G, Campbell JD, Carr CE, Boyd KS, Franklin IM. Transforming growth factor beta from multiple myeloma cells inhibits proliferation and IL-2 responsiveness in T lymphocytes. J Leukoc Biol. 1999;66:981–8. doi: 10.1002/jlb.66.6.981. [DOI] [PubMed] [Google Scholar]
  • 37.Filella X, Blade J, Guillermo AL, Molina R, Rozman C, Ballesta AM. Cytokines (IL-6, TNF-alpha, IL-1alpha) and soluble interleukin-2 receptor as serum tumor markers in multiple myeloma. Cancer Detect Prev. 1996;20:52–6. [PubMed] [Google Scholar]
  • 38.Brimnes MK, Svane IM, Johnsen HE. Impaired functionality and phenotypic profile of dendritic cells from patients with multiple myeloma. Clin Exp Immunol. 2006;144:76–84. doi: 10.1111/j.1365-2249.2006.03037.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu ZY, Zhang XG, Rodriguez C, et al. Interleukin-10 is a proliferation factor but not a differentiation factor for human myeloma cells. Blood. 1995;85:2521–7. [PubMed] [Google Scholar]
  • 40.Caux C, Massacrier C, Vanbervliet B, Barthelemy C, Liu YJ, Banchereau J. Interleukin 10 inhibits T cell alloreaction induced by human dendritic cells. Int Immunol. 1994;6:1177–85. doi: 10.1093/intimm/6.8.1177. [DOI] [PubMed] [Google Scholar]
  • 41.Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. 2006:2691–702. doi: 10.1084/jem.20061104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gorgun GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121:2975–87. doi: 10.1182/blood-2012-08-448548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Beyer M, Kochanek M, Giese T, et al. In vivo peripheral expansion of naive CD4+CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood. 2006;107:3940–9. doi: 10.1182/blood-2005-09-3671. [DOI] [PubMed] [Google Scholar]
  • 44.Feyler S, von Lilienfeld-Toal M, Jarmin S, et al. CD4(+)CD25(+)FoxP3(+) regulatory T cells are increased whilst CD3(+)CD4(-)CD8(-)alphabetaTCR(+) Double Negative T cells are decreased in the peripheral blood of patients with multiple myeloma which correlates with disease burden. Br J Haematol. 2009;144:686–95. doi: 10.1111/j.1365-2141.2008.07530.x. [DOI] [PubMed] [Google Scholar]
  • 45.Mills KH, Cawley JC. Abnormal monoclonal antibody-defined helper/suppressor T-cell subpopulations in multiple myeloma: relationship to treatment and clinical stage. Br J Haematol. 1983;53:271–5. doi: 10.1111/j.1365-2141.1983.tb02021.x. [DOI] [PubMed] [Google Scholar]
  • 46.Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5:ra46. doi: 10.1126/scisignal.2002796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pianko MJ, Moskowitz AJ, Lesokhin AM. Facts and Hopes in Immunotherapy of Lymphoma and Myeloma. Clin Cancer Res. 2017 doi: 10.1158/1078-0432.CCR-17-0539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Miller A, Asmann Y, Cattaneo L, et al. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer Journal. 2017;7:e612. doi: 10.1038/bcj.2017.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gorgun G, Samur MK, Cowens KB, et al. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin Cancer Res. 2015;21:4607–18. doi: 10.1158/1078-0432.CCR-15-0200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Galustian C, Meyer B, Labarthe MC, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother. 2009;58:1033–45. doi: 10.1007/s00262-008-0620-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ocio EM, Mateos MV, Orlowski R, et al. J Clin Oncol. Chicago, IL: 2017. Pembrolizumab (Pembro) plus lenalidomide (Len) and low-dose dexamethasone (Dex) for relapsed/refractory multiple myeloma (RRMM): Efficacy and biomarker analyses. [Google Scholar]
  • 52.Badros A, Hyjek E, Ma N, et al. Pembrolizumab, pomalidomide and low dose dexamethasone for relapsed/refractory multiple myeloma. Blood. 2017 doi: 10.1182/blood-2017-03-775122. [DOI] [PubMed] [Google Scholar]
  • 53.Merck provides further update on three multiple myeloma studies evaluation PEMBROLIZUMAB (pembrolizumab) in combination with pomalidomide or lenalidomide [news release] 2017 (Accessed September 2017., at http://investors.merck.com/news/press-release-details/2017/Merck-Provides-Further-Update-on-Three-Multiple-Myeloma-Studies-Evaluating-PEMBROLIZUMAB-pembrolizumab-in-Combination-with-Pomalidomide-or-Lenalidomide/default.aspx.)
  • 54.Administration FaD. Keytruda (pembrolizumab) in Patients with Multiple Myeloma: FDA Statement - Two Clinical Trials on Hold. 2017 [Google Scholar]
  • 55.Administration USFD. FDA Alerts Healthcare Professionals and Oncology Clinical Investigators about Two Clinical Trials on Hold Evaluating KEYTRUDA® (pembrolizumab) in Patients with Multiple Myeloma. 2017 [Google Scholar]
  • 56.Bunt SK, Clements VK, Hanson EM, Sinha P, Ostrand-Rosenberg S. Inflammation enhances myeloid-derived suppressor cell cross-talk by signaling through Toll-like receptor 4. J Leukoc Biol. 2009 doi: 10.1189/jlb.0708446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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;28:1280–8. doi: 10.1038/leu.2013.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.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 biology & therapy. 2008;7:622–7. doi: 10.4161/cbt.7.5.5689. [DOI] [PubMed] [Google Scholar]
  • 59.Salih HR, Wintterle S, Krusch M, et al. The role of leukemia-derived B7-H1 (PD-L1) in tumor-T-cell interactions in humans. Experimental hematology. 2006;34:888–94. doi: 10.1016/j.exphem.2006.03.006. [DOI] [PubMed] [Google Scholar]
  • 60.Krupka C, Kufer P, Kischel R, et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia. 2016;30:484–91. doi: 10.1038/leu.2015.214. [DOI] [PubMed] [Google Scholar]
  • 61.Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114:1545–52. doi: 10.1182/blood-2009-03-206672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ahmad SM, Svane IM, Andersen MH. The stimulation of PD-L1-specific cytotoxic T lymphocytes can both directly and indirectly enhance antileukemic immunity. Blood cancer journal. 2014;4:e230. doi: 10.1038/bcj.2014.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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;117:4501–10. doi: 10.1182/blood-2010-10-310425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhou Q, Munger ME, Highfill SL, et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010;116:2484–93. doi: 10.1182/blood-2010-03-275446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell. 2015;162:974–86. doi: 10.1016/j.cell.2015.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Roulois D, Loo Yau H, Singhania R, et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell. 2015;162:961–73. doi: 10.1016/j.cell.2015.07.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.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 [Google Scholar]
  • 68.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) Am Soc Hematology. 2016 [Google Scholar]
  • 69.Michonneau D, Sagoo P, Breart B, Garcia Z, Celli S, Bousso P. The PD-1 Axis Enforces an Anatomical Segregation of CTL Activity that Creates Tumor Niches after Allogeneic Hematopoietic Stem Cell Transplantation. Immunity. 2016;44:143–54. doi: 10.1016/j.immuni.2015.12.008. [DOI] [PubMed] [Google Scholar]
  • 70.Koestner W, Hapke M, Herbst J, et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood. 2011;117:1030–41. doi: 10.1182/blood-2010-04-283119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Asakura S, Hashimoto D, Takashima S, et al. Alloantigen expression on non-hematopoietic cells reduces graft-versus-leukemia effects in mice. J Clin Invest. 2010;120:2370–8. doi: 10.1172/JCI39165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Opdivo(R) [package nsert] Bristol-Myers Squibb Company; Princeton, NJ, USA: 04/2017. Accessed May 9, 2017., at https://packageinserts.bms.com/pi/pi_opdivo.pdf. [Google Scholar]
  • 73.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 doi: 10.1182/blood-2017-01-761346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Herbaux C, Gauthier J, Brice P, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood. 2017;129:2471–8. doi: 10.1182/blood-2016-11-749556. [DOI] [PubMed] [Google Scholar]
  • 75.El Cheikh J, Massoud R, Abudalle I, et al. Nivolumab salvage therapy before or after allogeneic stem cell transplantation in Hodgkin lymphoma. Bone Marrow Transplant. 2017 doi: 10.1038/bmt.2017.69. [DOI] [PubMed] [Google Scholar]

RESOURCES