Abstract
Immune checkpoint blockade has driven a revolution in modern oncology, and robust drug development of immune checkpoint inhibitors is underway in both solid tumors and hematologic malignancies. High response rates to programmed cell death 1 (PD-1) blockade using nivolumab or pembrolizumab in classical Hodgkin lymphoma (cHL) and several variants of non-Hodgkin lymphoma (NHL) revealed an intrinsic biologic sensitivity to this approach, and work is ongoing exploring combinations with immune checkpoint inhibitors in both cHL and NHL. There are also preliminary data suggesting antitumor efficacy of PD-1 inhibitors used in combination with immunomodulatory drugs in multiple myeloma (MM), and effects of novel monoclonal antibody therapies on the tumor microenvironment may lead to synergy with checkpoint blockade. Although immune checkpoint inhibitors are generally well-tolerated, clinicians must use caution and remain vigilant when treating patients with these agents in order to identify immune related toxicities and prevent treatment-related morbidity and mortality. Autologous stem cell transplant is a useful tool for treatment of hematologic malignancies and has potential as a platform for use of immune checkpoint inhibitors. An important safety signal has emerged surrounding the risk of graft-versus-host-disease (GVHD) associated with use of PD-1 inhibitors before and after allogeneic stem cell transplant. We aim to discuss the facts known to date in the use of immune checkpoint inhibitors for patients with lymphoid malignancies, and discuss our hopes for expanding the benefits of immunotherapy to patients in the future.
Introduction
Blocking inhibitory surface receptor-ligand pairs, which function to limit T cell activation and autoimmunity has revealed a critical role for immune checkpoints in aiding cancer’s evasion of host immunity (1–3). Blockade of immune checkpoints cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) is revolutionizing treatment in many types of solid tumors by stimulating endogenous antitumor immune responses(4). Immune checkpoint blockade therapy (CBT) is also under development in several subtypes of hematologic malignancies, with impressive responses seen in relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) and recent promising results seen in multiple myeloma (MM) combining CBT with immunomodulatory drugs (IMiDs). Herein, we will review the development of CBT for the treatment of lymphoid cancers to date and discuss opportunities for future progress.
Immune checkpoint blockade in lymphoma
Hodgkin lymphoma can be cured in the majority of cases, however despite optimal therapy, salvage autologous stem cell transplant (auto-HSCT), and brentuximab vedotin (BV), additional treatment options are needed for a subset of relapsing patients. cHL is characterized by the presence of an inflammatory immune infiltrate surrounding the malignant Hodgkin Reed Sternberg (HRS) cell and near universal genetic amplification of the 9p24.1 locus that encodes the PD-1 ligands as well as JAK2, which in a dose-dependent fashion can further upregulate PD-L1 expression via JAK2-STAT signaling(5). These observations formed the rationale for exploring CBT in this patient population. Patients with cHL treated with anti-PD-1 experienced objective response rates that were higher than expected, suggesting a potential intrinsic sensitivity to PD-1 blockade directly correlated with the degree of 9p24.1 amplification(5–11).
Patients with R/R cHL after auto-HSCT and BV receiving nivolumab on the phase I CheckMate039 study(7) had an 87% overall response rate (ORR), with 17% reaching a complete response (CR) and 70% achieving partial response (PR). The phase II CheckMate205 study (9) demonstrated an overall response rate of 66%, with 7 reaching CR, and 26 patients reaching PR. The phase I study of pembrolizumab (KEYNOTE-013) showed an ORR of 58%, CRR of 19%, and 12% of patients reached PR(12,13). In the phase II study (KEYNOTE-087) exploring pembrolizumab among three cohorts defined by history of auto-HSCT and exposure to BV, there was an ORR of 65.4–68.3%, CRR of 21.7–20%, and 93.7% of patients had a reduction in their tumor burden by radiographic assessment(14). In patients with R/R cHL after auto-HSCT and BV, the landmark clinical trials of immune checkpoint blockade led to accelerated approval of nivolumab and pembrolizumab by the Food and Drug Administration (FDA) for this indication.
Beyond its use in relapsed/refractory patients, PD-1 pathway blockade in combination therapies are being rapidly explored in other cHL populations, including newly diagnosed patients, autologous transplant in the salvage setting, transplant-ineligible patients, brentuximab-naïve patients, and patients with localized early stage disease with unfavorable characteristics. (Table 1). Early data is encouraging. Interim results from the phase I/II study of nivolumab combined with brentuximab vedotin as first salvage therapy after frontline chemotherapy before auto-HSCT showed a complete response rate (CRR) of 63% among the 59 evaluable patients, a rate significantly higher than expected with use of either agent alone(15). In R/R cHL, early data from the phase I ECOG-ACRIN E4412 study presented recently showed a CRR of 61% in 18 evaluable patients among 19 treated with the combination of nivolumab plus one of two dose levels of brentuximab vedotin (n=10 with1.2 mg/kg and n=9 with 1.8 mg/kg)(16). In the brentuximab plus ipilimumab arms, patients treated with brentuximab vedotin 1.8 mg/kg plus one of two dose levels of ipilimumab (1mg/kg or 3mg/kg) responded at a rate of 67%, with 5/12 (42%) achieving CR, with responses seen at both dose levels(17). Brentuximab vedotin plus nivolumab will be further evaluated in a pending phase III clinical trial in auto-HSCT-ineligible or R/R patients (CheckMate 812, NCT03138499).
Table 1.
Disease Setting | Regimen | Target | Phase | Status | Est. Study Completion Date | NCT |
---|---|---|---|---|---|---|
Newly Diagnosed, untreated cHL | ||||||
Newly diagnosed cHL (age <60 with HR features, age >60) | A(B)VD +Nivo | PD-1 | I | Recruiting | 01/2020 | NCT03033914 |
Early stage, unfavorable risk, no prior treatment. | Nivo + AVD -> IFRT vs. Nivo × 4 cycles -> Nivo + AVD × 2 cycles -> AVD × 2 cycles -> IFRT | PD-1 | II | Recruiting | 12/2020 | NCT03004833 |
Age>60, ineligible for or declined conventional chemotherapy | Nivo + BV vs BV + Benda vs. BV + dacarbazine vs BV | PD-1 | II | Recruiting | 10/2018 | NCT01716806 |
Untreated, transplant ineligible | Nivo + BV | PD-1 | II | Recruiting | 05/2024 | NCT02758717 |
Relapsed/Refractory cHL | ||||||
Early stage Relapsed or Primary Refractory cHL | Pembro + ISRT | PD-1 | II | Recruiting | 06/2020 | NCT03179917 |
R/R cHL (No prior BV, IO agent, or transplant) | Nivo + BV | PD-1 | I/II | Recruiting | 05/2020 | NCT02572167 |
R/R cHL (2nd line only) | Nivo + ICE | PD-1 | II | Recruiting | 04/2019 | NCT03016871 |
R/R cHL, no prior SCT (allo or auto) | Pembro + ICE | PD-1 | II | Recruiting | 02/2020 | NCT03077828 |
R/R cHL, prior auto or allo HSCT allowed, BV, and IO agent allowed | Ipi + Nivo + BV vs Ipi + BV vs. Ipi + Nivo + BV | CTLA-4, PD-1 | I | Recruiting | 06/2018 | NCT01896999 |
R/R cHL (no prior allo-HSCT) | Avelumab | PDL1 | Ib | Recruiting | 09/2017 | NCT02603419 |
R/R cHL (no prior allo-HSCT) | Ibrutinib + Nivo | PD-1 | II | Recruiting | 05/2020 | NCT02940301 |
Children/Adolescents/Young Adults (≥1 line of therapy, no prior HSCT) | Nivo + BV, followed by BV+Benda in suboptimal responders (CheckMate 744) | PD-1 | II | Recruiting | 03/2022 | NCT02927769 |
R/R (after SCT or transplant-ineligible) | ||||||
R/R cHL (prior auto-HSCT, or transplant-ineligible) | Nivo + BV vs. BV (CheckMate 812) | PD-1 | III | NYO | 04/2024 | NCT03138499 |
R/R cHL in BV-naive (failed auto-HSCT or transplant ineligible) | Pembro vs. BV (KEYNOTE-204) | PD-1 | III | Recruiting | 12/2019 | NCT02684292 |
R/R cHL after auto-HSCT and BV, or chemo-refractory with or without prior auto-HSCT) | Pembro | PD | II | Active, not recruiting | 04/2021 | NCT02453594 |
Post auto-HSCT | Pembro | PD-1 | II | Recruiting | 12/2018 | NCT02362997 |
R/R HR cHL | Nivo + BV, to start within 30–60 days of auto-HSCT stem cell infusion | PD-1 | II | Recruiting | 04/2019 | NCT03057795 |
R/R cHL (transplant ineligible) | Pembro + lenalidomide | PD-1 | I | Recruiting | 08/2023 | NCT02875067 |
R/R cHL with prior auto-HSCT or R/R transplant-ineligible | Nivo + lenalidomide | PD-1 | Ib | Recruiting | 04/2020 | NCT03015896 |
Relapse after allo-HSCT | Ipi or Nivo | CTLA-4, PD-1 | I | Recruiting | 12/2018 | NCT01822509 |
Relapse after allo-HSCT | Pembro | PD-1 | I | Recruiting | 02/2020 | NCT02981914 |
Legend: Allo = Allogeneic, AVD = Adriamycin + Vinblastine + Dacarbazine, Benda = Bendamustine, BV = Brentuximab vedotin, Dex = Dexamethasone, Durva = Durvalumab (anti-PDL1), HD = High Dose, HR = High Risk, IFRT = Involved field radiotherapy, ISRT = Involved Site Radiotherapy, IO = immuno-oncology, Ipi = Ipilimumab (anti-CTLA-4), Len = Lenalidomide, MM = Multiple Myeloma, Nivo = Nivolumab (anti-PD-1), NR = Not Reported, NYO = Not Yet Open, Pembro = Pembrolizumab (anti-PD-1), Pom = Pomalidomide, SCT = Stem Cell Transplant, Treme = Tremelimumab (anti-CTLA-4). Status as reported by http://clinicaltrials.gov, accessed 6/12/2017
Among the non-Hodgkin lymphomas (NHL), PDL1 overexpression is observed in many entities, including primary mediastinal large B-cell lymphoma (PMBL), primary CNS lymphoma, primary testicular lymphoma, plasmablastic lymphoma, HHV-8 associated primary effusion lymphoma, T-cell/histiocyte-rich B-cell lymphoma, both Epstein Barr Virus (EBV) -positive and EBV-negative post-transplant lymphoproliferative disorders and EBV-associated diffuse large B cell lymphoma and extranodal NK/T cell lymphoma (ENKL) (18,19). Some NHL subtypes, such as PMBL, derive PDL1 overexpression from 9p24.1 mutations or copy number alterations. (5,19). In other entities EBV drives PD-L1 overexpression through a mechanism independent of 9p24.1 amplification through effects of the EBV-encoded latent membrane protein-1 (LMP-1) which promotes AP1 and JAK-STAT signaling and increases PD-L1 expression via an AP-1 dependent enhancer (Figure 1)(19–21). Recent studies have focused on entities with PDL1 expression and promising activity was observed in the phase Ib study with PMBL (ORR 41% among 17 patients) as well as a phase II study in mycosis fungoides/Sézary syndrome (ORR 38% among 24 patients)(10,22,23). In addition, impressive activity was reported in small retrospective series of patients with ENKL and CNS lymphoma(20,21). Building upon this data, a prospective study in CNS lymphoma is underway (clinicaltrials.gov NCT02857426) and further analysis in ENKL is certainly warranted. Apart from PD-1, markers of immune exhaustion LAG-3 and TIM-3 are co-expressed in T cell infiltrates in NHL and represent potential additional targets for checkpoint blockade with in vitro data supporting this approach(24,25).
Despite remarkable activity of anti-PD-1 in cHL and several variants of NHL, a subset of patients experience progressive disease after an initial response, or are primary refractory to PD-1 blockade underscoring the importance of elucidating mechanisms of response and resistance beyond 9p24.1 amplification. Studies from solid tumors highlight a need for tumor cell recognition by T cells for efficacy of CBT, a process that requires relevant antigens and antigen presentation machinery(26). A retrospective series found decreased or absent expression of β2M and/or MHC I in 80% and decreased or absent MHC class II in 70% of cHL patients; β2M is the most frequently mutated gene in cHL(27). A retrospective analysis of 108 newly diagnosed cHL patients treated with conventional chemotherapy plus modified involved field radiotherapy found that those with reduced or absent β2M or MHC class I expression on HRS cells had poor outcomes independent of 9p24.1 status(28). Loss of MHC-II expression on HRS cells is also found more commonly in patients with relapsed cHL compared with newly diagnosed patients(29). Although the relationship between β2M mutations and response to CBT has not yet been described in cHL, β2M mutations and loss of MHC-I in melanoma have been described in patients with progressive disease and resistance to PD-1 blockade(30). Identification of tumor antigens in cHL is complicated by the relative rarity of HRS cells in the tumor microenvironment and requires enrichment techniques such as laser-capture microdissection or cell-sorting using flow cytometry(27). As such associations between antigen specific immune response against either shared or mutation-derived neo-antigens and efficacy of PD-1 blockade are not known. Additional research is needed to better define mechanisms of resistance to PD-1 blockade in cHL in order to inform design of rational clinical trials aimed toward achieving durable remissions in a larger proportion of patients.
Immunotherapy for multiple myeloma: combinations offer a path forward
Preclinical data support a role for the PD-1/PDL1 pathway in myeloma via expression of the PD-1 receptor on T and NK cells in patients with MM and expression of PD-1 ligands on malignant plasma cells(31). T cells have been shown to recognize abnormal plasma cells (PC), as supported by detection of marrow-infiltrating T cells in MGUS capable of mounting anti-PC immune responses and presence of immunity against shared antigens is associated with prolonged progression to over symptomatic MM. However, once symptomatic MM develops, marrow T cell responses have not been observed without ex vivo expansion steps(32–34). The reasons for the loss of antigen-specific T cell activity in vivo in MM compared with precursor disease is not well understood, but could be due to progressive immunosuppression by the tumor microenvironment during disease progression from MGUS to MM, in contrast to the pro-inflammatory milieu present in the cHL tumor microenvironment (Figure 1). Perhaps the relative paucity of antigen-specific T cells is one reason that anti-PD-1 monotherapy using nivolumab had limited clinical activity(10). Interestingly, lenalidomide administration appeared to have transient efficacy immediately following nivolumab during a period of where prolonged receptor occupancy of the PD-1 receptor was expected(35).
IMiD drugs (thalidomide, lenalidomide, and pomalidomide) enhance T cell responsiveness to APC and polarize T cells toward a Th1 phenotype, inhibit myeloid derived suppressor cells (MDSC) and regulatory T cells (Treg), and down-regulate PD-L1 on tumor cells (36–39). These observations suggested the hypothesis that IMiD and PD-1 blockade combinations could result in clinically relevant antimyeloma immune responses in relapsed, refractory MM (Table 2). The KEYNOTE-023 study evaluating pembrolizumab, lenalidomide, and dexamethasone demonstrated an ORR of 44% (n=50), with sCR 4%, VGPR 12%, and PR of 28%. Lenalidomide-refractory patients responded to pembrolizumab plus lenalidomide and dexamethasone at a rate of 35%, with 5.4% achieving sCR, 8.1% reaching VGPR, and 21.6% achieving a PR(40). A phase II study of pembrolizumab, pomalidomide, and dexamethasone demonstrated an overall response rate of 60% (29/48), with 4(8%) reaching sCR/CR, 9(19%) reaching VGPR, and 16 (33%) reaching PR. Although limited by a small sample size, correlative analyses of pre-treatment tissue biopsies demonstrated that presence of CD3+/PD-1+ marrow-infiltrating lymphocytes was associated with shorter progression free survival(41). Patients expressing PD-L1 in the bone marrow before treatment had a trend toward a higher rate of responses of VGPR or better(41). An alternative hypothesis for the failure of PD-1 monotherapy in MM proposes that clonal bone marrow T cells expressing PD-1 in MM exhibit a telomere-independent senescence phenotype and are unable to respond to reinvigoration with immune checkpoint blockade(42). Additional biomarker studies are needed to better understand the association between response and PDL1 expression in MM marrow; and if PD-1+ T cells in MM are senescent T cells or can be re-invigorated (43).
Table 2.
Phase | Subgroup | Patients | ORR (≥PR) (%) |
CR or sCR n(%) |
VGP R n(%) |
PR n(% ) |
SD n(%) |
PD n(%) |
Median Follow Up (95% CI) |
DOR (mo) | Median PFS (95%CI) |
Median OS (95% CI) |
Ref | |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Nivolumab CheckMate-039 NCT01592370 |
Ib | NA | 27 | 0% | 0 | 0 | 0 | 17 (63%) | 10 (37%) | NR | NA | 10 | NR | (51) |
Pembrolizumab plus Len/Dex KEYNOTE-023 NCT02036502 |
Ib: | All patients* | 50 | 44% | 2(4%) | 6 (12%) | 14 (28%) | 25 (50%) | 1 (2%) | 18.9 mo (0.8–36) | 18.7mo (0.7–30.4)† | 7.2 mo (3.9–12.3) | NR (22.4-NR) | (40) |
Len-Refractory population | 37 | 13(35.1%) | 2(5.4%) | 2 (8.1%) | 8 (21.6%) | 22 (59.5%) | 1 (3.3%) | U | 24.9mo (0.7–24.9)†† | 6.3 mo (2.8–8.5 | 26.3 mo (22.4-NR) | |||
Double or More Refractory | 30 | 13(33.3%) | 1(3.3%) | 13 (33.3%) | 5 (16.7%) | 18 (60%) | 1 (3.3%) | U | U | U | U | |||
Pembrolizumab plus Pom/Dex NCT02289222 |
II | All patients | 48 (3 NE) | 60% | 4(8%) | 9 (19%) | 16 (33%) | 11 (23%) | 2(4%) | 15.6 (9.2–17.5) | 14.7# | 17.4mo (11.7–18.8) | NR (18.9-NR) ## | (41) |
Double refractory (PI/IMiD) | 32 (73%) | 66% | 1(4%) | 6 (18%) | 14 (44%) | U | U | U | U | U | U | |||
HR CG | 27 (56%) | 56% | 3(11%) | 1 (4%) | 11 (41%) | U | U | U | U | 15.1mo (9.1–17.9)** | U |
ORR = Overall Response Rate, CR = complete response, sCR = strict complete response, VGPR = very good partial response, PR = partial response, SD = Stable disease, PD = Progressive disease, DOR = duration of response, PFS = progression free survival, OS = overall response, Ref = reference, CG = cytogenetics, NA = not applicable, NE = Not Evaluable, NR = Not reached, U = Unknown.
2 (4%) of patients were not yet assessable.,
vs 19 mo (16-NR). for Standard Risk CG,
= for patients meeting objective response criteria (PR or better),
as of cut-off date 11/1/2016; 22 (49%) had PD, 9(20%) died, and 23 continue to receive treatment,
n=22,
n=13 patients
Additional combination strategies in MM: shifting the balance in the microenvironment
Encouraging clinical activity observed with the IMiDs and anti-PD-1 combinations has spurred evaluation of agents capable of shifting the tumor microenvironment toward immune activation while inducing myeloma cell killing. In this regard, CD38 has emerged as an interesting target in MM due to high-levels of expression on plasma cells, a contribution to T cell anergy through ectoenzyme function that leads to adenosine production and expression on inhibitory cell populations such as MDSC and Treg (44,45). Targeting CD38 with daratumumab kills malignant plasma cells through traditional antibody dependent cellular cytotoxic (ADCC) mechanisms. In responding patients, daratumumab also appears to not only deplete subpopulations of Treg and MDSC in the myeloma microenvironment, but also result in T cell expansion and increased T cell clonality suggestive of an immune mechanism of myeloma disease control (46). These observations have provided rationale for investigation of daratumumab in combination with PD-1/PDL1 blockade with or without IMiD drugs (NCT01592370, NCT03000452, NCT02431208).
Radiation may also be an effective combination partner with PD-1 blockade by taking advantage of in situ vaccination caused by immunogenic cell death. Radiation has been shown to result in epitope spreading and augmented antigen-presentation by local APC. These effects have been associated with abscopal (distant) clinical effects in a variety of diseases (47–49). Temporal upregulation of PDL1 in the irradiated tumor suggests intrinsic mechanisms that inhibit immune responses after radiation, and provides rationale for blockade of PDL1 in combination with radiation(50) to overcome this mechanism. Several reported cases of systemic responses in patients with MM and plasmacytomas irradiated while receiving anti-PD-1 suggest potential induction of abscopal effects (41,51), which previously have been reported to occur spontaneously in very rare instances (52–54). We have recently begun enrollment of a combination trial using radiation plus PD-1 pathway blockade in patients with solitary bone plasmacytoma and limited clonal bone marrow plasmacytosis (NCT03196401) with the aim to elicit systemic immunity and the abscopal effect.
Hematopoietic Cell Transplantation: Risks, Rewards, and Potential
Both autologous and allogeneic HSCT are commonly used for treatment of patients with hematologic malignancies. In addition to antitumor responses produced by immunological graft versus tumor (GVT) effects after allo-HSCT, immune responses by the donor immune system against non-tumor host tissue can result in acute graft-versus-host disease (aGVHD) and chronic GVHD (cGVHD), leading to morbidity and treatment-related mortality (TRM). The normal function of immune checkpoints limit T-cell mediated immune responses against host tissues. Relapse after allo-HSCT represents a significant clinical dilemma and CBT is also being explored in this patient population. Preclinical studies examining PD-1 axis blockade after allo-HSCT demonstrated not only potentiation of GVT effects(55,56), but also evidence supporting exacerbation of GVHD(57).
The feasibility of immune checkpoint inhibition for treatment of hematologic cancers relapsing after allo-HSCT was first explored using CTLA-4 blockade with ipilimumab in two studies with responses observed in both lymphoid and myeloid malignancies without high rates of treatment-emergent GVHD(58,59). Several series further elaborate on efficacy and toxicity of PD-1 inhibitor use before or after allo-HSCT (Table 3). Based on early reports suggesting a toxicity signal of hyperacute, severe acute and chronic GVHD, and 4 treatment-related deaths observed among 39 patients who received PD-1 blockade before allo-HSCT (60), a warning was added to the FDA package insert for nivolumab(61). The FDA recommends that patients receiving allo-HSCT after PD-1 blockade be closely monitored for early evidence of transplant-related complications, such as hyperacute GVHD, severe acute GVHD, steroid-requiring febrile syndrome (as a potential harbinger of severe acute GVHD), hepatic veno-occlusive disease, and other immune mediated reactions.
Table 3.
PD-1 blockade Pre Allo | PD-1 blockade Post Allo | CTLA-4 Blockade Post Allo | ||||
---|---|---|---|---|---|---|
Merryman RW et al 2017 (60) | El-Cheikh J et al 2017 (64) | Haverkos et al 2017(62) | Herbaux et al 2017(63) | El Cheikh J et al 2017 (64) | Davids et al 2016(59) | |
Patients | 31 HL, 2 DLBCL, 2FL, 2 PMBCL, 1 EATL, 1 MCL | 9 HL | 30 HL 1 NHL |
20 HL | 2 HL | n=28, 7 HL(25%), 4 NHL (14%), 1 MM Myeloid = 20 |
Med. Time from CBT to allo (range) | 62d (7–260) | 44d (23–100) | NA | NA | NA | NA |
Med. Time allo to CBT (range) | NA | NA | 26.4mo (4.8–108) | 23 mo(2–111) | 10–28 mo | 675d (198–1830) |
CBT agent n(%) | Nivo 28 (72), Pembro 11 (28) | Nivo 9(100) | 28 Nivo 3 Pembro |
Nivo (100%) | Nivo 2 (100%) | Ipilimumab 28 (100%) |
ORR n(%) | 89% HL74%, NHL 13%) | 7 (77.8%) | 77% (95%CI: 58–90) | 95% | 2 (100%) | 1/12 (lymphoid cancers) |
BR to aPD-1 n(%) | CR 14 (36%) PR 10 (26%) SD 7(18%), PD 8 (21%) |
CR 4 (44%), PR 3 (33%) SD 0 (0%) PD 2 (22%) |
CR: 15(48.4%), PR: 8 (25.8%) SD: 3 (9.7%) PD: 4 |
CR: 8(42%) PR: 10 (52%) |
CR: 2 (100%) | PR: 1/7 in HL |
IrAE pre/post allo-HSCT | 4(11%), colitis 2(6%), pneumonitis 2 (6%), | NR | 17 TE-GVHD (55%) | NR | NR | 6 patients (21%) n=1 death Gr5 n=3 pneumonitis (2 Gr2, 1 Gr4) n=1 ITP Gr 2 n=1 diarrhea, Gr 2 |
Treatment Emergent Gr. 2–4 aGVHD (%, 95%CI) | 44% | 9(100%) | 10 (32%) | 6 (30%) | 100% | NR, Gut n=1 Gr2 |
Treatment Emergent Gr. 3–4 aGVHD (%, 95%CI) | 23(11–37) | 6(66%) | 6 (19%) | 5 (25%) | 2(100%) | 0 |
1 year cGVHD (%, 95%CI) | 41(22–60) | NR† | NR | NR | NR | 3 cases cGVHD liver (not graded) |
1 year TRM (%) | 10%* | 1 (11%; VOD) | 8 (4 aGVHD, 4 cGVHD) | 2 (10%) | NR | 1 (3.6%) |
Median Follow Up (range) | 12 (2–33) | 10 mo (5–19) | 428 (133–833) | 370 days (24–486) | 1.3–8 | 15mo (8–27) |
1 year OS (%, 95%CI) | 89(74–96) | NR | NR (21/31 patients alive at study conclusion; mean 400 days) | 78% at 16 mo | NR | 49% |
PFS (%, 95% CI) | 76(56–87) at 1 year | NR | Median PFS 591 days (95%CI: 400–644) | median not reached | NR | 17.9% |
aGVHD = acute graft versus host disease, CBT = checkboint blockade therapy cGVHD = chronic graft versus host disease, IrAE = immune related adverse event, ITP = immune thrombocytopenia purpura, Nivo = Nivolumab (anti-PD-1), Pembro = Pembrolizumab (anti-PD-1), Ipi = ipilimumab (antiCTLA-4), NA = not applicable, NR = Not Reported, PFS = progression free survival, TRM = treatment related mortality, VOD = Veno-occlusive disease. *One patient with enteropathy-associated T cell lymphoma received ipilimumab concurrently with anti-PD-1 therapy who died with grade 4 aGVHD, the remained had HL. †two patients had ‘mild chronic’ GVHD, and one patient had ‘severe chronic’ GVHD.
For patients with relapsed cHL after allo-HSCT, limited treatment options have led to increased off-label usage of PD-1 inhibitors. These data suggest that patients can achieve objective responses to PD-1 blockade after allo-HSCT (ORR: 77–95%), but this is complicated by a significant risk of developing severe treatment-emergent GVHD in up to 30–55% of patients. Among 53 total patients with publicly reported outcomes following receipt of PD-1 inhibitors after allo-HSCT, the observed rate of treatment-emergent GVHD was 47.2%, with 30.2% of treated patients developing grade 3–4 acute or severe chronic GVHD (62–64).
Given the potential risk involved with the use of PD-1 inhibitors before or after allogeneic stem cell transplant, this approach should only be pursued in the context of a clinical trial. PD-1 blockade is being formally explored in prospective studies after allo-HSCT as maintenance therapy (NCT02985554). Perhaps these studies will provide greater insight into predictors of GVH risk versus GVT benefit of this approach and define appropriate patient populations in which clinicians can safely harness the potential of PD-1 blockade to maintain a meaningful GVT response while minimizing the risk of developing treatment-emergent GVHD after CBT.
Auto-HSCT avoids the challenges of GVHD, but absence of GVT is thought to be a limitation to durability of responses. Nevertheless, the dynamics of immune reconstitution early after autologous stem cell transplant alter the immune regulatory network to favor autologous graft versus tumor response that may be further augmented by immune checkpoint inhibition(65). For example, Treg populations decline as CD8+ T cells expand during early lymphocyte recovery after autologous stem cell transplant. Seeking to harness this potentially favorable immune phenotype, a trial testing autologous lymphocyte infusions and combined CTLA-4 and PD-1 pathway blockade in concert with auto-HSCT for MM is ongoing (NCT02716805). In addition, recent studies showing that T cells produced by the autograft are able to respond to APC and develop into antigen-specific CTLs as early as 12 days after auto-HSCT support vaccine strategies in this setting as well(65). Several ongoing studies aiming to improve durability of disease control following auto-HSCT via induction of MM-directed immune responses include a DC-tumor cell fusion vaccine (NCT02728102), a WT1-directed vaccine (NCT01827137), and an RNA-electroporated DC vaccine (NCT01995708). Future combinations trials incorporating vaccines with CBT in the post-autologous transplant space are a logical extension of these studies.
Immune-related toxicities of checkpoint blockade in hematologic malignancies
Immune checkpoint blockade is well tolerated in many patients, but immune mediated toxicities do develop. Three phase I studies in hematologic malignancy trials reported a drug-related grade 3 adverse event (AE) rate ranging from 18–20%, a small number of grade 4, and a single case of fatal pneumonitis (7,10,12).
The phase II studies of pembrolizumab and nivolumab in R/R cHL demonstrated acceptable safety profiles consistent with prior PD-1 inhibitor phase I studies. In the phase II study of nivolumab, 13/210 (5.4%) of patients had a treatment-related grade 3 AE, and there were no treatment-related grade 4 or 5 AEs reported. In the phase II study of pembrolizumab 22/80 patients had grade 3 AEs by investigator assessment, 2 patients had grade 4 increased lipase and one patient developed grade 4 neutropenia and there were no reported treatment related deaths(9,14).
In MM, pembrolizumab plus pomalidomide and dexamethasone did not appear to result in additive toxicity greater than that seen in solid tumors(66). Six patients developed immune mediated pneumonitis, the majority of which were grade 1–2 in severity, only one patient developed grade 3 pneumonitis(41), despite pomalidomide’s association with pneumonitis (67). Of note, a hold on accrual of subjects to the phase III KEYNOTE-183 and KEYNOTE-185 studies evaluating the additive benefit of pembrolizumab to lenalidomide and dexamethasone or pomalidomide and dexamethasone was instituted by Merck in June 2017 due to excess deaths in the pembrolizumab treatment arm. Further evaluation of this safety signal is pending. In our experience, early detection and treatment of immune-related AEs is critical as the severity of an these events seems to be inversely proportional to the time from onset of symptoms to treatment. As clinicians become accustomed to the patterns of toxicities seen with CBT, it is expected the severity of toxicities should diminish.
Future Directions and Conclusions
Clinical successes with blockade of the PD-1 pathway in cHL have lead to regulatory approvals and significant excitement among clinicians in evaluating the utility of these treatments earlier in disease natural history. Genetically driven increases in the 9p24.1 locus in HRS cells appear to have a clear association with depth of response underscoring an intrinsic sensitivity to PD-1 blockade in cHL. However, absence of antigen presentation machinery in most HRS cells highlights that additional study is needed to understand precise mechanisms of activity of PD-1 blockade in this disease. A broad range of combination trials currently ongoing will undoubtedly define how to best use PD-1 blockade within the landscape of cHL therapy over the coming years. It is hoped that further study of mechanisms of activity in cHL will enable tailoring of better patient selection for specific combination approaches and perhaps addressing emergent resistance. Beyond cHL, PD1 blockade is active in several virally-driven NHL subtypes and entities with 9p24.1 abnormalities; prospective clinical studies of immune checkpoint inhibitors are ongoing to follow up these observations. In subtypes of lymphoma with limited response to checkpoint blockade, development of reliable biomarkers to predict which subsets of patients might respond to these agents are needed.
In contrast, single agent PD-1 pathway blockade in MM was underwhelming. Fortunately, rationally designed combination trials with IMiDs in MM have had encouraging results and opened the door to pivotal phase III trials whose results are eagerly awaited. Additional immunotherapeutic interventions in MM, including monoclonal antibody therapy with daratumumab or elotuzumab, vaccine strategies and highly encouraging early data from chimeric antigen receptor modified T cell therapies form unique opportunities to rapidly evaluate rational combination strategies.
Numerous additional questions remain on the use of immune checkpoint blockade therapy in these two distinct diseases. Can stem cell transplant, radiation, and other chemotherapies routinely used in cHL and MM combinations result in a favorable efficacy/safety profile? Will evaluating PDL1 expression, T cell clonality, or other biomarkers derived from studies in solid tumor malignancies have applicability in cHL, MM, and other lymphoid malignancies? What is the role of antigen specific immunity in these diseases in the context of checkpoint blockade and will shared or neo-antigens emerge as potential predictors of activity? Are there additional immune checkpoints or agonists whose modulation will also be therapeutically effective for these diseases? The emerging paradigm has been to evaluate combinations on a PD-1 blockade backbone, but perhaps this approach will mask unique biology or augment toxicity of other immune modulatory pathways.
Partnership of immune checkpoint antibodies with other immune-based approaches such as adoptive cellular immunotherapy such as chimeric antigen receptor modified T (CAR-T) cells or antibody engineering products such as bispecific T cell engagers (BiTEs) might exhibit synergistic activity. Vaccine-based approaches aimed at stimulating antigen-specific immunity to shared tumor antigens or neoantigens potentially through dendritic cell based platforms could also be rationally combined with immune checkpoint blockade to amplify antitumor immune responses.
Tumor immunotherapy originated more than 120 years ago by William Coley and his induction of inflammation by direct tumor inoculation of bacterial products at the turn of the 20th century. Years of basic science investigations since that time have delineated pathways of immune activation and regulation; and ultimately have yielded the realization that negative regulators of immune activation are dominant pathways of cancer immune evasion. As such, checkpoint blockade has in effect re-invigorated the entire field of tumor immunotherapy.
Acknowledgments
Funding:
This work was supported in part by the Memorial Sloan Kettering Cancer Center (MSKCC) NCI core grant [P30 CA008748] (A.M.L., PI: Craig B. Thompson, MD).
M.J.P. is supported in part by the MSKCC Mortimer J. Lacher Fellowship supported by the Lymphoma Foundation, the MSK Sawiris Foundation, and is supported in part by a grant from 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.M.L. is a member of the Parker Institute for Cancer Immunotherapy, which supported the MSKCC Cancer Immunotherapy Program. A.M.L. also receives support from the MSK Sawiris Foundation and Cycle For Survival.
Footnotes
Disclosures: Matthew J. Pianko: no relevant disclosures.
Alison J. Moskowitz: Honoraria: Seattle Genetics, Bristol-Myers Squibb, Takeda, Merck. Research funding: Seattle Genetics, Bristol-Myers Squibb.
Alexander M. Lesokhin: Honoraria: Bristol-Myers Squibb, Aduro, Juno; Consulting or advisory role: Bristol-Myers Squibb, Research funding: Bristol-Myers Squibb, Serametrix, Celgene, Genentech; Patents, royalties, other intellectual property: Serametrix.
References
- 1.Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734–6. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
- 2.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–7. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2(3):261–8. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 4.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(280):280sr1. doi: 10.1126/scitranslmed.3010274. [DOI] [PubMed] [Google Scholar]
- 5.Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O’Donnell E, 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(17):3268–77. doi: 10.1182/blood-2010-05-282780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Roemer MG, Advani RH, Ligon AH, Natkunam Y, Redd RA, Homer H, et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J Clin Oncol. 2016;34(23):2690–7. doi: 10.1200/JCO.2016.66.4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–9. doi: 10.1056/NEJMoa1411087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ansell S, Armand P, Timmerman JM, Shipp MA, Bradley Garelik MB, Zhu L, 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(23):583. [Google Scholar]
- 9.Younes A, Santoro A, Shipp M, Zinzani PL, Timmerman JM, Ansell S, et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. The lancet oncology. 2016;17(9):1283–94. doi: 10.1016/S1470-2045(16)30167-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol. 2016;34(23):2698–704. doi: 10.1200/JCO.2015.65.9789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Goodman A, Patel SP, Kurzrock R. PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nature reviews Clinical oncology. 2016 doi: 10.1038/nrclinonc.2016.168. [DOI] [PubMed] [Google Scholar]
- 12.Moskowitz CH, Ribrag V, Michot J, Martinelli G, Zinzani PL, Gutierrez M, et al. PD-1 Blockade with the Monoclonal Antibody Pembrolizumab (MK-3475) in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Preliminary Results from a Phase 1b Study (KEYNOTE-013) American Society for Hematology Annual Meeting. Volume 124. San Francisco, CA. Blood. 2014:290. [Google Scholar]
- 13.Armand P, Shipp M, Ribrag V, Michot J, Zinzani P, Kuruvilla J, 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 ASH Abstract #1108. [Google Scholar]
- 14.Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, 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]
- 15.Herrera AF, Moskowitz AJ, Bartlett NL, Vose JM, Ramchandren R, Feldman TA, 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. doi: 10.1002/hon.2437_73. [DOI] [Google Scholar]
- 16.Diefenbach CS, Hong F, David K, Cohen J, Roberston M, Advani R, et al. SAFETY AND EFFICACY OF COMBINATION OF BRENTUXIMAB VEDOTIN AND NIVOLUMAB IN RELAPSED/REFRACTORY HODGKIN LYMPHOMA: a TRIAL OF THE ECOG-ACRIN CANCER RESEARCH GROUP (E4412) Hematological Oncology. 2017;35:84–5. doi: 10.1002/hon.2437_72. [DOI] [Google Scholar]
- 17.Diefenbach CS, Hong F, Cohen JB, Robertson MJ, Ambinder RF, Fenske TS, et al. Preliminary Safety and Efficacy of the Combination of Brentuximab Vedotin and Ipilimumab in Relapsed/Refractory Hodgkin Lymphoma: A Trial of the ECOG-ACRIN Cancer Research Group (E4412) American Society of Hematology 2015; Orlando, FL, USA: 2015. [Google Scholar]
- 18.Chapuy B, Roemer MG, Stewart C, Tan Y, Abo RP, Zhang L, et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016;127(7):869–81. doi: 10.1182/blood-2015-10-673236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen BJ, Chapuy B, Ouyang J, Sun HH, Roemer MG, Xu ML, et al. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res. 2013;19(13):3462–73. doi: 10.1158/1078-0432.CCR-13-0855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Nayak L, Iwamoto FM, LaCasce A, Mukundan S, Roemer MGM, Chapuy B, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood. 2017;129(23):3071–3. doi: 10.1182/blood-2017-01-764209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kwong YL, Chan TS, Tan D, Kim SJ, Poon LM, Mow B, et al. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing L-asparaginase. Blood. 2017 doi: 10.1182/blood-2016-12-756841. [DOI] [PubMed] [Google Scholar]
- 22.Zinzani PL, Ribrag V, Moskowitz CH, Michot JM, Kuruvilla J, Balakumaran A, et al. Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood. 2017;130(3):267–70. doi: 10.1182/blood-2016-12-758383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Khodadoust M, Rook AH, Porcu P, Foss FM, Moskowitz AJ, Shustov AR, 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 American Society of Hematology 2016 Annual Meeting Abstracts, #181. [Google Scholar]
- 24.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]
- 25.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(2661) [Google Scholar]
- 26.Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell. 2017;168(4):707–23. doi: 10.1016/j.cell.2017.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Reichel J, Chadburn A, Rubinstein PG, Giulino-Roth L, Tam W, Liu Y, et al. Flow-sorting and exome sequencing reveals the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood. 2014 doi: 10.1182/blood-2014-11-610436. [DOI] [PubMed] [Google Scholar]
- 28.Roemer MG, Advani RH, Redd RA, Pinkus GS, Natkunam Y, Ligon AH, et al. Classical Hodgkin Lymphoma with Reduced beta2M/MHC Class I Expression is Associated with Inferior Outcome Independent of 9p24.1 Status. Cancer immunology research. 2016 doi: 10.1158/2326-6066.CIR-16-0201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Vardhana SA, Ozkaya N, Matsuki E, Pichardo J, Moskowitz AJ, Straus DJ, et al. Association of MHC-II, PD-L1, and FoxP3 with Disease Status and Outcomes in Patients with Hodgkin Lymphoma. American Society of Hematology 2016 Annual Meeting; San Diego, CA. 2016. [Google Scholar]
- 30.Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375(9):819–29. doi: 10.1056/NEJMoa1604958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pianko MJ, Liu Y, Bagchi S, Lesokhin AM. Immune checkpoint blockade for hematologic malignancies: a review. Stem Cell Investig. 2017;4:32. doi: 10.21037/sci.2017.03.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dhodapkar MV, Krasovsky J, Olson K. T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells. Proc Natl Acad Sci U S A. 2002;99(20):13009–13. doi: 10.1073/pnas.202491499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Noonan KA, Huff CA, Davis J, Lemas MV, Fiorino S, Bitzan J, et al. Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci Transl Med. 2015;7(288):288ra78. doi: 10.1126/scitranslmed.aaa7014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Spisek R, Kukreja A, Chen LC, Matthews P, Mazumder A, Vesole D, et al. Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J Exp Med. 2007;204(4):831–40. doi: 10.1084/jem.20062387. doi jem.20062387 [pii] 10.1084/jem.20062387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pianko MJ, Funt SA, Page DB, Cattry D, Scott EC, Ansell SM, et al. Efficacy and toxicity of therapy immediately after treatment with nivolumab in relapsed multiple myeloma. Leuk Lymphoma. 2017:1–4. doi: 10.1080/10428194.2017.1320713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Luptakova K, Rosenblatt J, Glotzbecker B, Mills H, Stroopinsky D, Kufe T, et al. Lenalidomide enhances anti-myeloma cellular immunity. Cancer Immunol Immunother. 2013;62(1):39–49. doi: 10.1007/s00262-012-1308-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Galustian C, Meyer B, Labarthe MC, Dredge K, Klaschka D, Henry J, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother. 2009;58(7):1033–45. doi: 10.1007/s00262-008-0620-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gorgun GT, Whitehill G, Anderson JL, Hideshima T, Maguire C, Laubach J, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121(15):2975–87. doi: 10.1182/blood-2012-08-448548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gorgun G, Samur MK, Cowens KB, Paula S, Bianchi G, Anderson JE, et al. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin Cancer Res. 2015;21(20):4607–18. doi: 10.1158/1078-0432.CCR-15-0200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ocio EM, Mateos MV, Orlowski R, Siegel D, Reece DE, Moreau P, et al. J Clin Oncol. Vol. 35. 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]
- 41.Badros A, Hyjek E, Ma N, Lesokhin A, Dogan A, Rapoport AP, 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]
- 42.Suen H, Brown R, Yang S, Ho PJ, Gibson J, Joshua D. The failure of immune checkpoint blockade in multiple myeloma with PD-1 inhibitors in a phase 1 study. Leukemia. 2015;29(7):1621–2. doi: 10.1038/leu.2015.104. [DOI] [PubMed] [Google Scholar]
- 43.Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–9. doi: 10.1038/ni.2035. [DOI] [PubMed] [Google Scholar]
- 44.Feng X, Zhang L, Acharya C, An G, Wen K, Qiu L, et al. Targeting CD38 Suppresses Induction and Function of T Regulatory Cells to Mitigate Immunosuppression in Multiple Myeloma. Clin Cancer Res. 2017 doi: 10.1158/1078-0432.CCR-16-3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Young A, Mittal D, Stagg J, Smyth MJ. Targeting cancer-derived adenosine: new therapeutic approaches. Cancer discovery. 2014;4(8):879–88. doi: 10.1158/2159-8290.CD-14-0341. [DOI] [PubMed] [Google Scholar]
- 46.Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128(3):384–94. doi: 10.1182/blood-2015-12-687749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58(3):862–70. doi: 10.1016/j.ijrobp.2003.09.012. [DOI] [PubMed] [Google Scholar]
- 48.Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105(4):256–65. doi: 10.1093/jnci/djs629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. The lancet oncology. 2015;16(7):795–803. doi: 10.1016/S1470-2045(15)00054-6. [DOI] [PubMed] [Google Scholar]
- 50.Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74(19):5458–68. doi: 10.1158/0008-5472.CAN-14-1258. [DOI] [PubMed] [Google Scholar]
- 51.Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol. 2016 doi: 10.1200/JCO.2015.65.9789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Saba R, Saleem N, Peace D. Long-term survival consequent on the abscopal effect in a patient with multiple myeloma. BMJ Case Rep. 2016;2016 doi: 10.1136/bcr-2016-215237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Robin HI, AuBuchon J, Varanasi VR, Weinstein AB. The abscopal effect: demonstration in lymphomatous involvement of kidneys. Med Pediatr Oncol. 1981;9(5):473–6. doi: 10.1002/mpo.2950090510. [DOI] [PubMed] [Google Scholar]
- 54.Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–31. doi: 10.1056/NEJMoa1112824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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(1):143–54. doi: 10.1016/j.immuni.2015.12.008. [DOI] [PubMed] [Google Scholar]
- 56.Koestner W, Hapke M, Herbst J, Klein C, Welte K, Fruehauf 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(3):1030–41. doi: 10.1182/blood-2010-04-283119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Asakura S, Hashimoto D, Takashima S, Sugiyama H, Maeda Y, Akashi K, et al. Alloantigen expression on non-hematopoietic cells reduces graft-versus-leukemia effects in mice. J Clin Invest. 2010;120(7):2370–8. doi: 10.1172/JCI39165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113(7):1581–8. doi: 10.1182/blood-2008-07-168468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N Engl J Med. 2016;375(2):143–53. doi: 10.1056/NEJMoa1601202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Merryman RW, Kim HT, Zinzani PL, Carlo-Stella C, Ansell SM, Perales MA, et al. Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood. 2017;129(10):1380–8. doi: 10.1182/blood-2016-09-738385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Opdivo(R) [package nsert] Bristol-Myers Squibb Company; Princeton, NJ, USA: 04/2017 < https://packageinserts.bms.com/pi/pi_opdivo.pdf>. Accessed Accessed May 9, 2017. [Google Scholar]
- 62.Haverkos BM, Abbott D, Hamadani M, Armand P, Flowers ME, Merryman R, 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]
- 63.Herbaux C, Gauthier J, Brice P, Drumez E, Ysebaert L, Doyen H, et al. Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. Blood. 2017;129(18):2471–8. doi: 10.1182/blood-2016-11-749556. [DOI] [PubMed] [Google Scholar]
- 64.El Cheikh J, Massoud R, Abudalle I, Haffar B, Mahfouz R, Kharfan-Dabaja MA, 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]
- 65.Chung DJ, Pronschinske KB, Shyer JA, Sharma S, Leung S, Curran SA, et al. T-cell Exhaustion in Multiple Myeloma Relapse after Autotransplant: Optimal Timing of Immunotherapy. Cancer immunology research. 2016;4(1):61–71. doi: 10.1158/2326-6066.CIR-15-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Nishino M, Ramaiya NH, Awad MM, Sholl LM, Maattala JA, Taibi M, et al. PD-1 Inhibitor-Related Pneumonitis in Advanced Cancer Patients: Radiographic Patterns and Clinical Course. Clin Cancer Res. 2016;22(24):6051–60. doi: 10.1158/1078-0432.CCR-16-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Geyer HL, Viggiano RW, Lacy MQ, Witzig TE, Leslie KO, Mikhael JR, et al. Acute lung toxicity related to pomalidomide. Chest. 2011;140(2):529–33. doi: 10.1378/chest.10-2082. [DOI] [PubMed] [Google Scholar]