1,851
Views
3
CrossRef citations to date
0
Altmetric
Review

Myelodysplastic syndrome and immunotherapy novel to next in-line treatments

&
Pages 2602-2616 | Received 07 Dec 2020, Accepted 27 Feb 2021, Published online: 03 May 2021

ABSTRACT

Patients with Myelodysplastic syndromes (MDS) have few therapy options for sustainable responses in the frontline setting, and even less after hypomethylating agent (HMA) failure in relapsed and refractory setting. The only potential cure is an allogeneic hematopoietic stem cell transplant which is an unrealistic option for the majority of MDS patients. Immunotherapy with checkpoint inhibition, CAR-T cells, and vaccine therapy few have shown promise in a variety cancer and have now been tested in patients with MDS. Most trials have focused on AML patients and included small numbers of MDS patients. Until now, a dedicated review of immunotherapy outcomes in MDS patients has been lacking. Thus, herein we review outcomes of MDS patients after immunotherapies on a variety of clinical trials reported to date.

Introduction

The myelodysplastic syndrome(s) (MDS) are a heterogenous group of neoplasms arising from myeloid progenitor cellsCitation1 with consequential ineffective hematopoiesis, dysplasia and peripheral blood cytopenias.Citation2,Citation3 The pathogenesis is complex involving epigenetic alterations, splicing changes, etc. that lead to maturation arrest. Hence, clinical outcomes across the MDS disease spectrum can be highly variable.

The International Prognostic Scoring System (IPSS) and the revised IPSS (IPSS-R) differentiates categories of MDS based on prognosis (estimated survival and time to leukemia progression).Citation4,Citation5 There are four prognostic categories per IPSS-R, in descending order of estimated survival these are: Low, Intermediate-1, Intermediate-2 and High-risk MDS (HR-MDS).Citation6

The only cure for MDS is with an allogeneic hematopoietic stem cell transplant (HSCT) which is the ultimate example of immunotherapy wherein a graft versus tumor (GVT) effect eliminates the MDS clone.Citation7,Citation8 However, the toxicities of this procedure limit HSCT to only a subset of young and fit patients (~15%).Citation9

Hypomethylating agents (HMA) such as azacitidine (AZA) or decitabine (DEC) are the recommended frontline line therapy for transplant ineligible intermediate, high or transfusion dependent MDS.Citation10 Indeed, randomized phase III trials with AZA vs best supportive care or low dose cytarabine (ara-c) demonstrated a survival benefit in higher-risk (IPSS) MDS patients (21–24 months in AZA arm vs 11.5 months in BSC vs 15.3 months in ara-c).Citation11–13 DEC similarly has improved PFS and reduced AML transformation.Citation11,Citation14

The prognosis after HMA failure is poor with a median OS of 5.6 months, and currently there are no standard therapeutic options for such patients.Citation15 Elderly patients or those with comorbidities are significantly vulnerable, and there is a dire need for alternate salvage regimens for high risk MDS patients after HMA refractoriness.Citation16,Citation17

Therapeutic manipulation of the immune system with newly introduced agents such as checkpoint inhibitors or adoptive T cell transfer has shown promise in several clinical trials for hematologic malignancies such as lymphomas but has yet to demonstrate efficacy in myeloid malignancies. Indeed, several approaches have been attempted in the context of acute myeloid leukemia (AML) with reported successes. However, MDS patients often make up only a small subset of those trials. Therefore, the purpose of this review, is to detail the clinical outcomes of exclusively MDS patients enrolled on immunotherapy clinical trials reported to date.

Immune checkpoint inhibitors

The natural function of immune checkpoints is to restore antigenic self-tolerance through multiple mechanisms such as modulating co-stimulation or direct immune cell inhibition.Citation18 Checkpoint receptors are reliant on cognate ligands to facilitate interaction and these are variably expressed by a variety of cell-types.Citation19,Citation20 For example, expression of the inhibitory ligand CD47 on myeloid leukemia cell acts as an anti-apoptotic “don’t eat me” molecule that also increases functioning of immune-suppressor T- regulatory cells (Treg) resulting in the death of antigen-specific effector T-cells (Teff).Citation19,Citation20When inhibitory ligand expression is low or the checkpoint is pharmacologically blocked, tumor cell apoptosis can occur in addition to T cell receptor (TCR) mediated cytolysis induced by Teff cells. Indeed, this occurs naturally in normal responses to foreign antigens such as viral, intracellular bacterial pathogens and cancers.

Neoplastic cells of myeloid origin have taken advantage of these immune checkpoints with upregulation of inhibitory ligands such as PD-L1, B7-1, B7-2, CD47 etc. at baseline.Citation19 MDS cells have demonstrated the capacity to harness some of these immunosuppressive effects to facilitate their survival, longevity, and proliferation. The latter is most marked in higher risk MDS (HR- MDS) and RAEB (refractory anemia with excess blasts).Citation19,Citation20 Conversely, higher expression of immuno-inhibitory receptors of these ligands such as CTLA4, TIM3, PD-1, and ICOS were found on T-cells of patients with treatment refractory MDS patients compared with healthy donors.Citation18,Citation19 In summary, sufficient evidence exists that patients with MDS have altered immune response to aberrant myeloid clones partly mediated by immune checkpoint modulation. Moreover, treatment with hypomethylating agents (HMA) has been shown to upregulate several of these immune checkpoints such as PD-L1, TIM-3 and CD47 on MDS cells. Therefore, it is conceivable that checkpoint inhibitors (CPI) that have been FDA approved for various solid tumors, lymphomas could be effective in MDS with or without concurrent HMA therapy yet reported outcomes to date have been unimpressive and colored by high rates of toxicities.Citation21 CPIs have induced serious immune-related adverse events in MDS patients which can be particularly problematic if used soon before or after an HSCT. This next section will focus on safety and efficacy outcomes reported from CPI-based clinical trials. Only select CPI trials with preliminary or completed data exclusively enrolling MDS patients and/or data for MDS patients that are distinctly reported separately in multi-disease/AML trials are represented in ().

Table 1. Select MDS studies with immune check-point inhibitors

Programmed death ligand-1 & receptor (PD-L1/PD-1) inhibitors

PD-L1 (B7H1) is universally expressed on antigen presenting cells (APC) as an inhibitory ligand for PD-1 on Teff. PDL-1 expression on MDS cells was found in some patients to be increased at baseline and sometime greater than two times normal in those with HMA refractory disease, suggesting a possible mechanism for immune evasion that would benefit from inhibition of the PD-1/PD-L1 interaction.Citation18

Pembrolizumab

The monoclonal antibody, Pembrolizumab, is a PD-1 blocking agent.Citation22 In 2016, Garcia-Manero et al presented early phase I data on 28 relapsed/refractory MDS patients treated with PEM after failure of response to prior epigenetic therapy.Citation23 Enrolled patients were IPSS high 25%, intermediate-2 (int-2) 32% and intermediate-1 (int-1) in 36%.

At a median follow-up of 5.6 months ORR to PEM monotherapy was: 1 (3.5%) partial response (PR), 3 (11%) marrow complete response (mCR) and 14 (52%) stable disease (SD). The 6-month OS was 49% with dismal probability of survival at 29% and 22% for IPSS high and int-2 patients respectively. Int-1 patients had a 1-year OS of 89%. Overall, monotherapy with pembrolizumab was well tolerated (7% treatment related adverse events), the most notable treatment related events were 2 cases of tumor lysis syndrome in those with excess blasts. To build upon the efficacy demonstrated in this trial, the same group, Chien et al. combined PEM with AZA in a phase II trial of 35 high risk MDS patients. Patients were stratified as therapy naïve MDS or had previously failed HMA therapy.Citation24 The overall best response rate amongst the HMA refractory MDS cohort (n = 20) was 30%, including 2 CR, 2 mCR, and 2 hematologic improvement (HI). The HMA naive cohort (n = 10) had an impressive 70% best response rate (2 CR, 2 mCR, 2 mCR + HI and 1 with HI alone). The 13 responding patients had complex cytogenetics (n = 2), or mutations in ASXL1(n = 4) and TP53 (n = 3) frequently associated with poor outcomes in MDS.Citation25 However, overall survival estimates of combination PEM + AZA combination in the HMA refractory cohort remained at a lowly 5.9 and 12.9 months for HMA naïve patients at a follow-up of nearly 11 months, thus not much different from what would be expected with HMAs alone. The combination appeared safe and the most frequent AE’s of any grade were neutropenia, arthralgia and myalgia.

Durvalumab

Durvalumab is a monoclonal antibody targets the ligand PDL-1.Citation26 Zaiden et alCitation27 evaluated responses in 84 treatment naïve IPSS-R higher risk MDS patients in an open label phase II study with equal randomization to durvalumab + AZA vs. AZA alone. Notably, this is one of the few randomized trials comparing an HMA alone to a combination of HMA and immunotherapy as front-line therapy. The trial ended early due to higher-than-expected rates of disease progression, death and lack of response in the treatment cohort. Yet, the AZA only arm had twice the number of censored patients (n = 19) compared to the combination (n = 10) and a lower ORR (47%) compared with the combination (61%) that was not statistically significant. There were no differences in PFS or OS (see ) but the combination treatment was well tolerated with 7 grade ≤ 2 immune-related adverse events (iRAEs).Citation27

Nivolumab

Nivolumab is a humanized monoclonal antibody that binds PD-1.Citation21 Nivolumab has been tested as part of cytarabine-based induction regimens for AML and MDS-EB as preclinical models demonstrated upregulation of PD-1 ligands.Citation21,Citation28 However, its use pre- HSCT may be limited due to immune-related AE’s (irAE’s) and the severity of GVHD post-HSCT.Citation29,Citation30 Nivolumab has been evaluated as an agent to enhance “graft-versus-tumor” effect to treat post-HSCT relapse of MDS. In this phase I trial, of 28 patients with various hematologic malignancies which included 7 (25%) with HR-MDS, 3 of them attained a PR.Citation31 However, induction and worsening of graft-versus-host disease (GVHD) after nivolumab remains a major safety concern in this setting. Indeed, there were two fatalities from GVHD and two more experienced serious dose limiting toxicities in the entire treated cohort. In summary, use of checkpoint inhibitors pre- or post-HSCT maybe have beneficial anti-MDS effects, but this should be weighed against GVHD in individual patients and should only be offered in the context of a clinical trial.

As part of a large multi-arm phase II trial, Garcia-Manero et. al reported preliminary data on 90 MDS patients with relapsed/refractory or therapy naïve disease divided into 6 treatment cohorts.

There were 76 patients that received checkpoint inhibitor monotherapy in 4 cohortsCitation32,Citation33 vs. double checkpoint inhibitors (n = 14) in 2 cohorts.Citation34 All patients in treatment naïve cohorts received AZA in addition to immunotherapy.

We will discuss 2 cohorts treated that were treated with nivolumab monotherapy here (see section on CTLA4 inhibitors for the remaining 4 cohorts).

Thirty-five patients were treated with nivolumab with or without HMA in two cohorts.Citation32,Citation33 The group of RR MDS patients (n = 15) received single agent nivolumab with 6 individuals having ≧ grade 3 AE, most frequently pulmonary infection (40%) and febrile neutropenia (33%).Citation32 As it lacked clinical activity this arm closed prior to completed enrollment. The therapy naive cohort (n = 20) received nivolumab with the addition of AZA with 50% CR/CRp responses.Citation33 This suggests a synergistic effect with HMA induced PD-1/PD-L1 upregulation with simultaneous PD-1 blockade on Teff cells with nivolumab.Citation18

Atezolizumab

The PD-L1 inhibitor, atezolizumab,Citation35 was evaluated in a phase Ib trial with IPSS-R higher risk MDS who were older (median age: 76; range: 63–89) and stratified by whether they had failed prior HMA (where they received only atezolizumab or in combination with an HMA) or were treatment naïve (where they received combination HMA and atezolizumab).Citation36 This trial much like the durvalumab trial was halted early because of a high incidence of grade ≥ 3 adverse events (71%) in both arms of the study (71% and 57%, respectively) as well as excessive early deaths (29%) within the naïve cohort compared with historical controls. In contrast, atezolizumab in combination with Guadecitabine, a next-generation HMA, was well tolerated in 9 relapsed and refractory MDS patients.Citation37 There were no DLT’s after a median of 5 treatment cycles and 94% of grade 3/4 AE’s were related to peripheral cytopenias.

Cytotoxic T-lymphocyte-associated protein 4 (CTLA4) inhibition

Ipilimumab

The T cell expressed surface protein CTLA4, dampens T cell response after binding its ligands: CD80 (B7-1) and CD86 (B7-2) which are typically expressed by antigen presenting cells. Ipilimumab (IPI) effectively blocks CTLA4 function thereby activating Teff resulting in clonal proliferation of antigen-specific T cells.Citation38 Simultaneously, IPI has been shown to downregulate immunosuppressive functioning of Treg cells through its action within lymph nodes and within the cancer micro-environment.Citation39

Much like PD-L1 expression, MDS cells from untreated patients have been shown to overexpress CTLA4 which can be enhanced by HMA therapy.Citation18

A multicenter phase I trial of IPI established a maximum tolerated dose of 3 mg/kg at which no grade ≥ 2 immune-related adverse events were seen in 29 HMA-refractory MDS patients.Citation40 In this refractory cohort, IPI as a single agent induced a 21% clinical benefit rate (defined as stable disease for over 12 months) of which 2 (9%) were mCRs. Furthermore, no early excessive deaths were observed (death rate of 15%) and in five who went onto get an HSCT, there was no increased incidence of GVHD post-HSCT either. Responding patients had significantly higher expression of inducible T cell costimulatory molecule (ICOS) on CD4+ (p = .05) and CD8+ (p = .01) T cells in comparison with those who failed therapy.Citation41

Garcia-Manero reported phase II safety and preliminary responses for 41 MDS patients treated with ipilimumab in 2 cohorts.Citation32,Citation33 Relapsed and refractory patients received single agent IPI with manageable side effectsCitation32 and ORR of 35% (n = 7).Citation33 Therapy naïve patients received IPI + AZA and had ORR 71% with 38% of responders having CR/CRp.Citation33

In conclusion, the 4 treatment cohorts (including those in the section on PD-1 inhibitors) that received checkpoint inhibitors with or without AZA had a favorable safety profile and tolerable toxicities, with rash and fatigue being the most common AE of any grade. Responses in the combination groups with HMAs were encouraging and perhaps higher than would be expected with HMAs alone, yet confirmatory trials are pending.

CTLA4/PD-1 combined inhibition: ipilimumab with nivolumab

In the Garcia-Manero multi-cohort phase II trial, 8 patients were treated on an arm that received a combination of ipilimumab and nivolumab to treat RR MDS while 6 others were treated on another arm that included treatment naïve patients who received combination CPIs with an HMA. The RR group of 8 MDS patients had an ORR of 29% with 1 CR and 1 HI while 3 of 6 in the treatment naïve cohort achieved CRs.Citation34

Taken together, checkpoint inhibitors of CTLA4 and PD-1 pathways that have been highly effective in select lymphomas and refractory solid malignancies, have yet to demonstrate similar results in patients with MDS. Combination approaches with HMAs have been encouraging but direct comparisons with HMA therapy alone is still in progress. The reasons for minimal single-agent efficacy in MDS patients might be multifold including lack of immunogenicity of MDS cells, defective T cell function in patients with MDS etc, all of which are under preclinical investigation.

T cell immunoglobulin and mucin domain-3 (TIM-3) inhibition

The immune checkpoint receptor T-cell Immunoglobulin and mucin domain 3 (TIM-3) is expressed on monocytes, dendritic cells, and Teff. Interaction with its ligand, galectin-9, leads to suppression of T cell activation. Like all previously described checkpoints, Tim-3 is expressed at a higher level on exhausted T cells.Citation42 The ligand is overexpressed preferentially on leukemic stem cells and myeloid blasts compared to normal hematopoietic stem cells and has been recognized as a potential target for AML/MDS immunotherapy.Citation42

MBG453

MBG453 is an anti-TIM-3 monoclonal antibody that has been tested in the clinic for its dual capacity to induce antibody-dependent cytotoxicity as well as its ability to enhance T cell activation.

Borate et al.Citation43,Citation44 reported separate outcomes for therapy naive MDS patients () that were included in a phase I treatment arm with MBG453 + HMA using AZA or DEC. The MBG453 + DEC cohort has treated 19 HR-MDS in its most recent report, in whom at ~5 months post-treatment, 58% (11/19) of patients achieved a clinical response including 5 CRs. Ten of the 13 patients that received MBG453 + AZA were evaluable for responses with 1PR and 6 mCR.

Longer follow-up and comparison with standard of care cohorts will determine if this response rate while promising is different from HMAs alone. Immune-related adverse events with MBG453 were reassuringly low and only 1 patient experienced elevated liver enzymes (at grade 3). MBG453 added to HMA appeared to have a favorable safety profile, with response durations that might be longer than would be expected for DEC or AZA alone, however, the full report is eagerly awaited.Citation43,Citation44

CD-47 inhibition

The CD47 ligand SIRP-α is universally expressed on human cells and serves as a “don’t eat me” signal for the innate immune system, specifically macrophages.Citation45,Citation46 Reproducibly, MDS cells have been shown to greatly overexpress CD47 possibly to evade macrophage mediated killing while enhancing antiapoptotic and proliferative signals.Citation39,Citation47 Indeed, its overexpression correlates with higher risk MDS as well as poor overall survival.Citation47,Citation48 So, strategies to block the CD47-SIRP-α interaction may enhance macrophage mediated phagocytosis and have now been exploited in the clinic with potential agents in clinical development discussed below.Citation47,Citation49,Citation50

CC-90002

CC-90002 is an anti-CD47 humanized IgG4 monoclonal antibody, which was tested in a phase 1 trial of relapsed or refractory high risk MDS (n = 4) and AML (n = 24).Citation51 Unfortunately, treatment resulted in excessive serious treatment related AE’s (SAE’s) in 82% of all dose escalation study patients (0.1 mg/kg up to 4 mg/kg) CC-90002 and 16 on-study deaths. Four patients experienced a serious DLT with grade 4 disseminated intravascular coagulation and grade 4 cerebral hemorrhage, grade 3 purpura, grade 4 congestive cardiac failure and grade 4 acute respiratory failure, and grade 4 sepsis. Perhaps the advanced blast burden or the dosing schema as well as the immunogenicity of the monoclonal antibody used may have contributed to the observed hyperinflammatory/autoimmune process.Citation52–54 The drug had poor preliminary efficacy in all study patients with best responses in those with MDS patients having SD (n = 2), causing the sponsor to terminate the trial.

Magrolimab (Hu5F9-G4 or 5F9)

By contrast, the humanized and thus probably less immunogenic anti-CD47 monoclonal antibody, Magrolimab (aka Hu5F9-G4 or 5F9) did not demonstrate similar toxicitiesCitation55,Citation56 in a phase Ib dose escalation study of IPSS-R Int-2 or high risk MDS. All patients received a priming dose of 5F9 (1–30 mg/kg weekly) that was done to mitigate hemolysisCitation52 which was reported in a prior study with an unclear mechanism of action.Citation53 This “prime” may have also potentially offset severe AE’s seen with CC-90002. MDS patients enrolled were either treatment naïve (n = 39)Citation57 or relapsed/refractory (n = 4).Citation55,Citation56 In combination with HMA, the investigators reported an impressive ORR of 100% with 54% CR and 39% mCR + HI. Two of these patients had a baseline tp53 mutations and both achieved an MRD negative CR. The responses were rapid with a median 1.9 months to response which is much faster than expected for AZA alone (up to 4 cycles of 4 months).Citation56 Indeed most of these responses lasted for longer than 4 months, as reported from an update presented in 2020, where ORR at ~6 months remains high at 91% (30 of 33 still in response) with no deaths reported (100% OS). Those who had failed HMA, received monotherapy with 5F9, where no responses were seen.Citation55 However, both arms tolerated treatment with 5F9 well (MTD not reached) and the most common adverse events were cytopenias (<25% developed any cytopenia).

Based on the results of the MDS cohort a phase III National Cancer Institute (NCI) clinical trial has started recruiting 9/2020 registered at NCT04313881 as a double-blind placebo controlled comparing magrolimab in combination with AZA vs. the standard of care AZA + placebo in therapy naïve higher risk MDS patients. Results from this and other ongoing efficacy-based trials for this combination are eagerly awaited.CPI have demonstrated objective responses in early phase trials with higher-risk MDS patients however not to the extent observed with lymphomas or select solid tumors. With the caveat of cross trial comparisons, the CR+PR and ORR of patients treated with CPI + HMA is safe and at least similar to standard of care therapy in the frontline setting with some indications of additive benefit.Citation11,Citation14 Unfortunately, MDS patients that have previously failed HMA therapy, remain an unmet need and CPIs like many other treatments for this cohort have been largely unsuccessful at inducing high response rates.

There is a trade-off with increasing severity and incidence of adverse events particularly with utilization of two CPI drugs. Most irAE’s were reversible with steroid administration and drug holding with safe resumption of the CPI. However, in the real world this may limit its widespread use in elderly patients with poor PS and other comorbidities.

As with other malignancies, the search for biomarkers that may predict response to check-point inhibitors for MDS/AML is ongoing and might be necessary before accepting CPIs as a bonafide treatment modality for MDS.

Adoptive T-cell transfer therapy

In malignancy, adoptive cellular therapy is the direct killing of tumor cells by specific effector lymphocytes. In its simplest form, adoptive T-cell transfer is the infusion of T-lymphocytes into a patient’s body to directly target an antigen, usually for cytotoxicity. It can be autologous or allogeneic T-cell transfer, which can be unmanipulated (as in simple donor lymphocyte infusion) or manipulated such as CAR (Chimeric Antigen Receptor) T-cell constructs, modified T-Cell receptors, or ex-vivo antigen exposure priming with or without genetic modification.

The myeloid malignancies have not had the same success to date as their lymphocytic counterparts, as there are many shared antigens with progenitor stem cells. However, we are finding unique ways to manipulate lymphocytes and target myeloid antigens in a more streamlined fashion without the toxicities seen in other immunotherapies to date, although this may be related to their lack of engraftment and/or expansion in vivo.

Here we will discuss select therapies with CAR-T and manipulated DLI for patients with MDS.

Chimeric antigen receptor

Autologous lymphocytes can be engineered to recognize and target antigens completely independent of MHC or HLA restriction. In other words, they are continuously programmed to bind and recognize the target without needing TCR nor co-stimulation in second generation formats, as activating receptors (such as CD28) can already be fused to the intracellular CD3 TCR complex.Citation58

Natural killer group 2 (NKG2) receptors

The natural killer group 2 receptors (NKG2D)Citation59 are a group of C-type lectin immunomodulatory proteins found on natural killer (NK) and CD8 + T cells. In response to intracellular stressors such as DNA damage, toxins, infection, inflammation and cancer, its ligands: MHC I chain-related proteins (MIC) MIC-A, MIC-B molecules and the activating unique long 16-binding proteins (ULBP 1–6) are upregulated on affected cells (eg. cancer cells).Citation60

Upon activation there is simultaneous rapid NK-mediated cytotoxicity as well adaptive T cell immune response that leads to marked increases in circulating levels of IFNγ and TNFα.Citation61 The same ligands that activate NKG2D receptors are cleaved into soluble forms forming of negative feedback to downregulate receptor expression and dampen the inflammatory process.Citation60

As NKG2D ligands are only expressed under pathologic inflammatory and not in the normal state, it makes an attractive target for CAR-T cell therapy. However, its expression is dynamic and fluctuates considerably. Nevertheless, T cells expressing CARs that mimic NKG2D receptors have been clinically tested with reported successes.

NKG2D-CAR T cells

T cells genetically engineered to express NKG2D have demonstrated the potential to target and specifically kill not just cancer cells but also modulate Tregs and myeloid-derived suppressor cells within the tumor microenvironment.Citation62 Furthermore, in these preclinical models, genetic overexpression of the receptor overcomes the natural inhibition mediated by cleaved soluble ligands like MIC-A.Citation61,Citation63,Citation64 NKG2D-CAR T cells rely on DNAX-activating protein of 10kDa (DAP10) endogenous expression to stabilize the NKG2D receptor which is in contrast with CD28 or 4–1BB co-stimulation required for CD19 CAR T cells.Citation58,Citation65–67

Clinical trials of NKG2D CAR-T cells have included patients with MDS because MDS cells, much like AML or myeloma cells have also been shown to greatly overexpress the ligands for NKG2D.Citation62,Citation68 We will review the available data for two investigational NKG2D CAR-T therapies that include MDS patients on protocol. Only select Adoptive Cellular therapy trials with preliminary/completed data exclusively enrolling MDS patients and/or data for MDS patients that are distinctly reported separately in multi-disease/AML trials are represented in .

Table 2. Select MDS studies with adoptive cellular therapies

CM-CS1

Baumeister et. al. published the first phase I trial of a NKG2D CAR T cell (called CM-CS1) with single dose administration of fresh product in 4 escalation cohorts with R/R MDS/AML (n = 6) and MM (n = 5) patients.Citation67,Citation69 The highest infused dose was 3 × 107 T cells. Compared with other autologous CAR-T cell products, CM-CS1 was safe and well tolerated, administered without lymphodepletion. Levels of CM-CS1 CAR-T cells were undetectable after 1–2 weeks in most patients. Unfortunately, there were no responses per disease specific response criteria at day 28 post infusion, and 9 evaluable patients had PD.Citation67

CYAD-01

Celyad pharmaceuticals manufactured the NKG2D-CAR (NKR-2) T-cell product, named CYAD, for use in the multi-national phase I THINK (THerapeutic Immunotherapy with NKR-2) trial with various cancer types.Citation61 In contrast to CM-CS1, CYAD-01 requires cryopreservation for multiple NKR-2 injections from single patient apheresis.Citation70

Sallman et al.Citation71 presented outcomes in 25 relapsed and refractory patients (3 MM, 19 AML, 3 MDS) receiving CYAD-01 CAR-T cells without prior lymphodepleting CT (THINK trial). All 3 MDS patients (ages 80–85) had HMA refractory disease. The most common grade ≥3 adverse event was CRS which occurred in 24% of all treated subjects (6 of 25) and resolved completely post-tocilizumab. One out of 2 MDS who were available for response assessment had a transient morphological CR after three infusions of CYAD-01 at the highest DL3 (3 x 109 cells). However, after week 12 despite a second infusion cycle the patient experienced disease progression. Overall, in the entire cohort, the duration of response in all 5 responding patients was brief (~4-8 weeks in the majority). There was no correlation between the dose levels of CAR-T cells infused on obtaining a response.

Of note, the THINK trial was conducted without the administration of lymphodepleting chemotherapy. Preclinical models have since demonstrated that cytotoxic chemotherapy upregulates NKG2D ligand expression, decreases Treg activity and thus enhances NKG2D CAR-T cell expansion.Citation62,Citation68,Citation72,Citation73 This led to the DEPLETHINK phase 1 trialCitation74 for patients with relapsed/refractory AML or MDS where patients were treated with lymphodepleting conditioning with cyclophosphamide and fludarabine prior to a single dose of CYAD-01 CAR-T cells. When comparing expansion metrics of the THINK and DEPLETHINK participants, the expansion and perhaps even persistence was superior in those that received lymphodepleting chemotherapy, however, this did not translate into clinical response as 2 patients with MDS on DEPLETHINK had no objective response by week 4 post-infusion. The higher CAR-T cell expansion resulted in higher frequencies of serious toxicities that were dose dependent. Indeed, 2 out 3 patients that received chemotherapy experienced grade 3 or higher CRS and both experienced encephalopathy syndromes consistent with immune effector cell-associated neurotoxicity. Novel strategies during manufacture of NKG2D CAR-T cells as well as new constructs (CYAD-02) have demonstrated superior efficacy to CYAD-01 in preclinical models because they address some major barriers such as fratricide or target specificity of NKG2D-CAR T cells are currently under investigation. Results from clinical trials of these new NKG2D CAR-T cell products are eagerly awaited.

Donor directed lymphocytes

Relapse after allotransplant for higher risk MDS patients has a dismal outlook with poor survival.Citation75 Consideration of unmanipulated donor cell infusion (DLI)Citation76 or second allo-HSCT is dependent on the duration of the previous remission period.Citation77,Citation78 However, neither of these options have durable responses and DLI is unpredictable with life-threatening GVHD.

Patient tailored DLI with tumor-associated antigen (TAA) stimulation ex vivo selects for an enriched, polyclonal CD4+ and CD8+ specifically directed against myeloid malignancies, such as AML and MDS. Our group has been able to mitigate GVHD with non-manipulated DLI by selecting T cells from DLIs that are specific for AML/MDS antigens typically not present on normal host cells, such as WT1, PRAME, Survivin and NYESO1.Citation79

Data presented thus far have shown this approach to be safe in 19 patients with high-risk AML/MDS (of which 2 had MDS). These patients had either relapsed after HSCT or had an unacceptably high risk of relapse due to the nature of their underlying disease. The latter group of patients that received TAA-T cell prophylaxis (n = 12) had 4 relapses <1 year from the infusion, but 2 were subsequently re-treated and one went onto another line of therapy. In summary, 11 of 12 patients that received the donor-derived TAA-T cells remain alive and in CR.

The responders had measurable expansion of leukemia antigen-specific T-cells that persisted for >9 months and exhibited durable memory. Impressively, there was no GVHD related to the infusions which was likely offset by selecting for “leukemic-specific” targets that are not present on normal cells. Confirmatory comparator trials are now underway (NCT04511130).

The use of preprogramed direct cytotoxic T-cells makes adoptive cellular therapy with autologous CAR T-cells and TAA- DLI attractive options for RR MDS.

Donor derived killer T cells are primed ex vivo with a selective immune signature tailored to the patients’ MDS cells. Thus far, TAA-DLI rapidly achieves disease control and appears to be safer, with respect to GVHD, than unselected donor T-cell infusion in the post-transplant setting. Of course, this may not be produced on a large scale and is highly dependent on having donor availability.

The utilization of autologous CAR T cell therapy in myeloid malignancies and particularly in MDS remains uncertain. There are many obstacles that must be overcome to be a successful therapy option for RR high-risk MDS, and their inability to persist with memory is problematic. Certainly, the CAR T cell expansion process is improved with lymphodepleting chemotherapy prior to infusion. This further reinforces the central dogma that within the bone marrow microenvironment of MDS there is profound immunosuppressive effects and impaired immune surveillance. Key players such as myeloid-derived suppressor and regulatory T-cells must be intervened upon for to have any meaningful therapeutic effect.Citation80,Citation81

On the horizon are constructs with multiple MDS antigen targets such as compound CAR T’s cells (cCAR) and Natural killer CAR’s, and NK-cell based CAR therapies.

Therapeutic vaccines

For more than 2 decades, various myeloid leukemia antigens have been identified as targets for vaccine-based T cell immunotherapies. The intention of preclinically and clinically tested vaccines is to stimulate a “tumor-specific” pool of T cells with specificity mediated through their native T cell receptor (TCR). Strategies have also focused on recruiting CD4+ helper T-cells by presenting peptides in the context of major histocompatibility class (MHC) II molecules by professional antigen presenting cells (APCs). This would not only give a robust delayed type hypersensitivity reaction with enduring memory, but it could also reactivate the exhausted T-cells with immunologic dormancy. Here-in we will discuss therapeutic MDS vaccine strategies with various platforms including peptide tumor associated antigens (TAA), dendritic cell based and autologous tumor cells. Only select vaccine trials with preliminary/completed data exclusively enrolling MDS patients and/or data for MDS patients that are distinctly reported separately in multi-disease/AML trials are represented in .

Table 3. Select MDS studies with vaccines

Peptide vaccines

Wilms’ tumor-1 (WT1)

Wilm’s tumor (WT1) gene encodes various regulatory functions involved in cellular differentiation and proliferation.Citation82 It has oncogenic potential with overexpression and association with bcl-2 and p53 genes, amongst others. In patients with MDS, there is a strict direct correlation between high WT1 mRNA levels and marrow blast percentages.Citation83,Citation84 Furthermore, higher levels of WT1 in peripheral blood correlate with poor response to HMA therapy and thus an inferior OS.Citation85 WT1 is a suitable immunotherapeutic target, as WT1 expression is significantly higher in MDS patients compared with healthy controls and is relatively immunogenic to T cells.Citation83,Citation86–88 Pilot studies of peptide vaccines of WT1 to date have demonstrated the capacity to induce clonal expansion of WT1 reactive T cells (predominantly CD8 + T cells).Citation89–91 Indeed, >50% of all vaccine recipients across the tested peptide vaccines develop WT1-peptide reactive T cell expansions. In all the trials reported to date the expanding WT1 reactive T cells in response to MHC I or II bound peptides did not induce any significant adverse events or autoreactivity (). For example, Suzuki et al.Citation92 demonstrated no grade ≥3 events at the highest tested therapeutic dose of their peptide vaccine and similarly, the vaccine DSP-7888 which contains both Class I and class II peptides did not demonstrate any grade ≥3 toxicities at the highest tested doses.Citation92 There were 2 SAE’s with myocarditis and fever, unrelated to the dose. The predominant response-type reported among MDS patients to date has been disease stability. There have been select cases of exceptional responders with Suzuki et al. reporting 4 of 22 obtaining an objective response (1 mCR and 3 HIs). Brayer et al.Citation91 reported an elderly MDS patient who was HMA refractory but achieved long lasting (for 22 months) transfusion independence post vaccination and Keilholz et al.Citation93 reported 1 MDS patient out of 2 with complete recovery of neutrophil counts post-vaccination that persisted for 10 months. In all trials, authors reported longer than expected disease stability and survival which might reflect an alteration in the natural course of the disease induced by T cell pressure. So future strategies that can expand the breadth of target peptides (as a combination with targeting other TAAs, see below), combinatorial approaches with complementary agents such as HMAs (to enhance expression of WT1) or checkpoint inhibitors might be necessary to induce deeper and longer lasting clinical responses.

Proteinase-3+ neutrophil elastase (PR-1)

Proteinase 3 (PR3) is present in high concentrations within the cytosol of myeloid leukemic blasts. By contrast normal granulocytes express PR3 only within the granules with minimal processing in the cytosol. Indeed, PR3 peptides have been shown to be presented naturally by only leukemic blasts and not normal granulocytes in the context of HLA-A2.Citation94 Endogenous responses to PR-3 have been shown to exist in HLA-A2+ patients with AML or MDS but it has been hypothesized that the chronic overexpression may lead to immune tolerance and exhaustion with selective clonal apoptosis of PR3-specific memory T cells.Citation95 Thus PR3 is considered an attractive target for vaccine immunotherapy of MDS. The largest trial of a PR3 vaccine was reported by Qazilbash et al.Citation96 where in 4 of 11 patients with MDS who received HLA-A2 restricted peptide-based PR3 in combination with neutrophil elastase, called the PR-1 vaccineCitation96 had a clinical response (1 PR and 3 HIs). As with tumor-based vaccine trials, the vaccine was safe at all tested doses despite demonstrating >2-fold increase in peripheral PR3-specific CD8+ specific T cells in 2 of the 4 responding patients. Similar to the WT1 vaccine approaches, this trial demonstrated that PR3 vaccines can be used in older MDS patients unfit for aggressive therapies as well as a safe immunotherapeutic approach in the post-HCT settings.

PR1 and WT1 combination vaccine

In an attempt to broaden the immune response to MDS clones, Rezvani et al.Citation90 administered a HLA-A2 restricted PR1 as well as a WT1 peptide vaccine to 2 patients with MDS. While neither patient had an objective response, both mounted PR1- and WT1-specific T cells post-vaccine that correlated in a decline in mRNA transcripts of WT1 and PR1 and this translated into long-term disease stability lasting for >2 years in both cases.

NY-ESO-1

NY-ESO-1 is one of several cancer testes antigens (CTA) expressed in trophoblastic tissue, spermatocytes and germline cells, but not in normal post-meiotic cells.Citation97–100 Germline cells naturally lack MHC I expression to promote an immunoprotective environment against potential self-destruction. Therefore, CD8+ T-cells cannot recognize CTA expression on these immune privileged cells.Citation98

These groups of antigens, especially NY-ESO-1 are highly immunogenic, and able to elicit spontaneous humoral and cell mediated immunity when exposed. Naturally this makes an attractive target for immunotherapy in treating cancers, and CTA’s have been found expressed on some solid cancers.Citation101 However, their expression is low or completely absent in AML and MDS as the CTA genes are silenced through promotor hypermethylation.Citation102,Citation103

Preclinical models in AML/MDS cells treated with HMA had an increase in NY-ESO-1 mRNA through a time and dose-dependent manner through recognition by antigen-specific CD8+ CTL. Circulating myeloid blasts in patients that received HMA also demonstrated de novo NY-ESO-1 expression.Citation104 Clearly treating MDS patients with epigenetic therapy enhance the expression of proteins once exempt from the immune system. So Griffiths et al. administered an NY-ESO-1 therapeutic peptide vaccine to 7 patients with MDS after 4 cycles of decitabine and observed clinical responses in 5 (3 CR and 2 HI). Since they were all HMA naïve any assertions on efficacy was impossible, however, the authors did observe serially measured immune responses to the vaccine with measurable IFN-γ levels. The immune responses were the most robust in 3 MDS (2CR and 1 SD) patients with detectable baseline NY-ESO-1 specific CD4+ T-cell clones.Citation104,Citation105

Targeting of antigens exclusive to myelodysplastic progenitor stem cells presents a challenge as many are non-immunogenic within the HSC compartment niche.

The tumor-associated antigens selected for use in therapeutic peptide vaccines described thus far are not unique to MDS. For example, WT-1 is expressed in CLL, CML, AML, primary peritoneal carcinoma, malignant mesothelioma, and many solid cancers to name a few.Citation82

Therefore, WT-1 antigen is not specific to MDS/AML. Rather, it is the disproportionate overexpression of these TAA antigens within their IPSS/IPSSR higher-risk native diseased state relative to their “lower risk” MDS counterparts.

Indeed, across MDS prognostic categories, CD34+ stem cells have WT-1 overexpression with a strict correlation between high mRNA levels and higher blast percentages and IPSS risk.Citation83,Citation84 Even within the risk subgroups a strong association exists between higher levels of WT-1 mRNA on having increased bone marrow blast percentages and adverse cytogenetics.Citation83

A retrospective study found higher expression of WT1 in the peripheral blood of MDS patients independently predicted a reduced likelihood to obtain a response with HMA, and was associated with an inferior OS that was independent from IPSS-R.Citation85

Though the expression of PR3 in HR-MDS has not been fully described in the literature, RNA levels in AML patients were found to be highest with persistent disease rather than adjusted blast counts. Interestingly, PR-3 expression was the highest in favorable ELN risk de novo AML. In contrast, it was the lowest in unfavorable risk ELN cytogenetic risk and in secondary AML from MDS.Citation106

Whole cell vaccines

Gene transduced tumor cell vaccine: GVAX

Another therapeutic vaccine strategy for MDS used patient derived whole tumor cell (K562) lines transfected with plasmid encoded Granulocyte Macrophage–Colony-Stimulating Factor (GM-CSF) secreting capability.Citation107–109 The autologous administration of irradiated K562/GM-CSF (GVAX) whole tumor cells primed native dendritic cells to present a panel of malignant myeloid epitopes in an HLA unrestricted manner to activate multiple facets of the adoptive and innate immune system.

A phase I pilot study with GVAX was given as five individual injections to 1 CMML and 4 MDS patients over the course of 3–4 months (). It was well tolerated with the only grade 1 injection site reactions. There was only 1 major HI without INF gamma production and 1 patient with achieved transfusion independence. Clinical activity was not seen in the three other patients did not have clinical activity though the study was too small to comment on any preliminary efficacy.Citation110

Taken together, therapeutic vaccines for MDS have demonstrated an excellent safety profile but reproducible effectiveness has not yet been achieved despite induction of robust MDS-TAA reactivity in vivo. For a vaccine to be therapeutic it must illicit a strong cytotoxic immune response for stem cell eradication. The initial recognition and processing of vaccine antigen is dependent on innate immune cells and functional antigen presenting cells. A potential issue with relying on the MDS patients’ native APC’s, is the dendritic cell dysfunction inherent to advanced disease. Patients with higher risk MDS have been reported to not only have lower DC counts, but also reduced functionality.Citation111

Larger confirmatory and preferably comparator arm trials are necessary to confirm any clinical benefit with vaccination approaches.

In conclusion, several immunotherapeutic approaches that have been effective in other cancers have been attempted in MDS with reported successes in select early phase trials. To date, HSCT remains the only proven effective immunotherapy for MDS, however, the identification and therapeutic modulation of MDS-specific targets such as CD47, TIM-3, WT1, etc. along with combination approaches are likely to be the future of MDS immunotherapy.

HMA-refractory MDS has no available effective treatments and remains the most challenging cohort of MDS patients. Findings from CD47 inhibitor and TIM-3 inhibitor trials are most impressive in MDS patients to date and have outperformed conventional checkpoint inhibitor therapies (PD-1 and CTLA4 inhibition). Single agent and combinatorial approaches with these drugs are now in confirmatory trials and an FDA-approval in this space is closer than ever before.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

Dr. Premal Lulla would like to thank the Edward P. Evans MDS Discovery research grant (PI: Premal Lulla), and The Cancer prevention and research institute of Texas (CPRIT) Early Career Clinical Investigator Award (RP200584).

References

  • Pang WW, Pluvinage JV, Price EA, Sridhar K, Arber DA, Greenberg PL, Schrier SL, Park CY, Weissman IL. Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes. Proc Natl Acad Sci USA. 2013;110:3011–16. doi:10.1073/pnas.1222861110.
  • Garcia-Manero G. Myelodysplastic syndromes: 2012 update on diagnosis, risk-stratification, and management. Am J Hematol. 2012;87:692–701. doi:10.1002/ajh.23264.
  • Valent P, Horny H, Bennett JM, Fonatsch C, Germing U, Greenberg P, Haferlach T, Haase D, Kolb HJ, Krieger O, et al. Definitions and standards in the diagnosis and treatment of the myelodysplastic syndromes: consensus statements and report from a working conference. Leuk Res. 2007;31:727–36. doi:10.1016/j.leukres.2006.11.009.
  • Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–88.
  • Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Solé F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120:2454–65. doi:10.1182/blood-2012-03-420489.
  • Nachtkamp K, Stark R, Strupp C, Kündgen A, Giagounidis A, Aul C, Hildebrandt B, Haas R, Gattermann N, Germing U, et al. Causes of death in 2877 patients with myelodysplastic syndromes. Ann Hematol. 2016;95:937–44. doi:10.1007/s00277-016-2649-3.
  • Stern M, De Wreede LC, Brand R, Van Biezen A, Dreger P, Mohty M, De Witte TM, Kröger N, Ruutu T. Sensitivity of hematological malignancies to graft-versus-host effects: an EBMT megafile analysis. Leukemia. 2014;28:2235–40. doi:10.1038/leu.2014.145.
  • Anderson JE, Appelbaum FR, Fisher LD, Schoch G, Shulman H, Anasetti C, Bensinger WI, Bryant E, Buckner CD, Doney K, et al. Allogeneic bone marrow transplantation for 93 patients with myelodysplastic syndrome. Blood. 1993;82:677–81. doi:10.1182/blood.V82.2.677.677.
  • Alessandrino EP, Della Porta MG, Bacigalupo A, Van Lint MT, Falda M, Onida F, Bernardi M, Iori AP, Rambaldi A, Cerretti R, et al. WHO classification and WPSS predict posttransplantation outcome in patients with myelodysplastic syndrome: a study from the Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Blood. 2008;112:895–902. doi:10.1182/blood-2008-03-143735.
  • Anonymous The National Comprehensive Cancer Network. ® Myelodysplastic syndromes. (Version 1.2021); 2020 Nov 27.
  • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, Schoch R, Gattermann N, Sanz G, List A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:223–32. doi:10.1016/S1470-2045(09)70003-8.
  • Cheson BD, Greenberg PL, Bennett JM, Lowenberg B, Wijermans PW, Nimer SD, Pinto A, Beran M, De Witte TM, Stone RM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108:419–25. doi:10.1182/blood-2005-10-4149.
  • Seymour JF, Fenaux P, Silverman LR, Mufti GJ, Hellström-Lindberg E, Santini V, List AF, Gore SD, Backstrom J, et al. Effects of azacitidine compared with conventional care regimens in elderly (≥ 75 years) patients with higher-risk myelodysplastic syndromes. Crit Rev Oncol Hematol. 2010;76:218–27. doi:10.1016/j.critrevonc.2010.04.005.
  • Lübbert M, Suciu S, Baila L, Rüter BH, Platzbecker U, Giagounidis A, Selleslag D, Labar B, Germing U, Salih HR, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the european organisation for research and treatment of cancer leukemia group and the german MDS study group. JCO. 2011;29:1987–96.
  • Prébet T, Gore SD, Esterni B, Gardin C, Itzykson R, Thepot S, Dreyfus F, Rauzy OB, Recher C, Adès L, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. JCO. 2011;29:3322–27. doi:10.1200/JCO.2011.35.8135.
  • Kantarjian H, O’brien S, Cortes J, Giles F, Faderl S, Jabbour E, Garcia-Manero G, Wierda W, Pierce S, Shan J, et al. Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome. Cancer. 2006;106:1090–98. doi:10.1002/cncr.21723.
  • Platzbecker U. Treatment of MDS. Blood. 2019;133:1096–107. doi:10.1182/blood-2018-10-844696.
  • Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, Fang Z, Nguyen M, Pierce S, Wei Y, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2013;28:1280–88. doi:10.1038/leu.2013.355.
  • Williams P, Basu S, Garcia-Manero G, Hourigan CS, Oetjen KA, Cortes JE, Ravandi F, Jabbour EJ, Al‐Hamal Z, Konopleva M, et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer. 2019;125:1470–81. doi:10.1002/cncr.31896.
  • Blank C, Mackensen A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion. Cancer Immunol Immunother. 2007;56:739–45. doi:10.1007/s00262-006-0272-1.
  • Bristol-Myers Squibb Company. OPDIVO (nivolumab). FDA; 2014 [accessed 2020 Oct 21]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125554s078lbl.pdf
  • FDA package insert. Pembrolizumab prescribing information. 2020. <https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125514s084lbl.pdf>
  • Garcia-Manero G, Tallman MS, Martinelli G, Ribrag V, Yang H, Balakumaran A, Chlosta S, Zhang Y, Smith BD. Pembrolizumab, a PD-1 inhibitor, in patients with myelodysplastic syndrome (MDS) after failure of hypomethylating agent treatment. Blood. 2016;128:345–345. doi:10.1182/blood.V128.22.345.345.
  • Chien KS, Borthakur GM, Naqvi K, Daver NG, Cortes JE, DiNardo CD, Jabbour E, Andreeff M, Alvarado Y, Bose P, et al. Updated preliminary results from a phase II study combining azacitidine and pembrolizumab in patients with higher-risk myelodysplastic syndrome. Blood. 2019;134:4240–4240.
  • Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, Kantarjian H, Raza A, Levine RL, Neuberg D. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011;364:2496–506. doi:10.1056/NEJMoa1013343.
  • AstraZeneca. Durvalumab prescribing information. FDA; 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761069s018lbl.pdf
  • Zeidan AM, Cavenagh J, Voso MT, Taussig D, Tormo M, Boss I, Copeland WB, Gray VE, Previtali A, O’ Connor T, et al. Efficacy and safety of azacitidine (AZA) in combination with the anti-PD-L1 durvalumab (durva) for the front-line treatment of older patients (pts) with Acute Myeloid Leukemia (AML) who are unfit for intensive chemotherapy (IC) and pts with higher-risk myelodysplastic syndromes (HR-MDS): results from a large, international, randomized phase 2 study. Blood. 2019;134:829–829.
  • Vereecque R, Saudemont A, Quesnel B. Cytosine arabinoside induces costimulatory molecule expression in acute myeloid leukemia cells. Leukemia. 2004;18:1223–30. doi:10.1038/sj.leu.2403391.
  • Ravandi F, Assi R, Daver N, Benton CB, Kadia T, Thompson PA, Borthakur G, Alvarado Y, Jabbour EJ, Konopleva M, et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study. Lancet Haematol. 2019;6:e480–e488. doi:10.1016/S2352-3026(19)30114-0.
  • Saha A, Aoyama K, Taylor PA, Koehn BH, Veenstra RG, Panoskaltsis-Mortari A, Munn DH, Murphy WJ, Azuma M, Yagita H, et al. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood. 2013;122:3062–73. doi:10.1182/blood-2013-05-500801.
  • Davids MS; (BCRP), on behalf of the Leukemia & Lymphoma Society Blood Cancer Research Partnership; Kim HT, Costello C, Herrera AF, Locke FL, Maegawa RO, Savell A, Mazzeo M, Anderson A, Boardman AP, et al. A multicenter phase 1 study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation. Blood. 2020;135:2182–91. doi:10.1182/blood.2019004710.
  • Garcia-Manero G, Daver NG, Montalban-Bravo G, Jabbour EJ, DiNardo CD, Kornblau SM, Bose P, Alvarado Y, Ohanian M, Borthakur 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;128:344–344. doi:10.1182/blood.V128.22.344.344.
  • Garcia-Manero G, Sasaki K, Montalban-Bravo G, Daver NG, Jabbour EJ, Alvarado Y, DiNardo CD, Ravandi F, Borthakur G, Bose P, et al. A phase II study of nivolumab or ipilimumab with or without azacitidine for patients with myelodysplastic syndrome (MDS). Blood. 2018;132:465–465.
  • Garcia-Manero G, Montalban-Bravo G, Sasaki K, Daver NG, Jabbour EJ, Alvarado Y, DiNardo CD, Ravandi F, Borthakur G, Bose P, et al. Double immune checkpoint inhibitor blockade with nivolumab and ipilimumab with or without azacitidine in patients with myelodysplastic syndrome (MDS). Blood. 2018;132:1831–1831. doi:10.1182/blood-2018-99-118948.
  • Genentech Inc. Atezolizumab prescribing information. FDA; 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761034s018lbl.pdf
  • Gerds AT, Scott BL, Greenberg PL, Khaled SK, Lin TL, Pollyea DA, Verma A, Dail M, Green C, Ma C, et al. PD-L1 blockade with atezolizumab in higher-risk myelodysplastic syndrome: an initial safety and efficacy analysis. Blood. 2018;132:466–466. doi:10.1182/blood-2018-99-118577.
  • O’Connell CL, Kropf PL, Punwani N, Rogers D, Sposto R, Grønbæk K. Phase I results of a multicenter clinical trial combining guadecitabine, a DNA methyltransferase inhibitor, with atezolizumab, an immune checkpoint inhibitor, in patients with relapsed or refractory myelodysplastic syndrome or chronic myelomonocytic leukemia. Blood. 2018;132:1811–1811.
  • Bristol-Myers Squibb Company. Ipilimumab prescribing information. 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125377s110lbl.pdf
  • Kokkaliaris KD, Scadden DT. Cell interactions in the bone marrow microenvironment affecting myeloid malignancies. Blood Adv. 2020;4:3795–803. doi:10.1182/bloodadvances.2020002127.
  • Zeidan AM, Knaus H, Robinson TM, Zeidner JF, Blackford AL, Rizzieri D, Frattini MG, Levy MY, Schroeder MA, Ferguson AK, et al. A phase I trial of ipilimumab (ipi) in patients (pts) with myelodysplastic syndromes (MDS) after hypomethylating agent (HMAs) failure. JCO. 2017;35:7010. doi:10.1200/JCO.2017.35.15_suppl.7010.
  • Robinson TM, Knaus H, Smith BD, Towlerton AM, Warren EH, Zeidner JF, Blackford A, Duffield AS, Rizzieri D, Frattini MG, et al. Immunological correlates of treatment with the CTLA-4 inhibitor ipilimumab in patients with refractory myelodysplastic syndromes (MDS). Blood. 2017;130:1699–1699. doi:10.1182/blood-2017-04-778225.
  • Banerjee H, Kane LP. Immune regulation by tim-3. F1000Res. 2018;7:316. doi:10.12688/f1000research.13446.1.
  • Borate U, Esteve J, Porkka K, Knapper S, Vey N, Scholl S, Garcia-Manero G, Wermke M, Janssen J, Traer E, et al. Phase Ib study of the anti-TIM-3 antibody MBG453 in combination with decitabine in patients with high-risk myelodysplastic syndrome (MDS) and Acute Myeloid Leukemia (AML). Blood. 2019;134:570–570. doi:10.1182/blood-2019-128178.
  • Borate U, Esteve J, Porkka K. Anti-TIM-3 antibody MBG453 in combination with hypomethylating agents in patients with high-risk myelodysplastic syndrome and acute myeloid leukemia: a phase 1 study. 25th European Hematology Association Congress. 2020.
  • Sick E, Jeanne A, Schneider C, Dedieu S, Takeda K, Martiny L. CD47 update: a multifaceted actor in the tumour microenvironment of potential therapeutic interest. Br J Pharmacol. 2012;167:1415–30. doi:10.1111/j.1476-5381.2012.02099.x.
  • Chao MP, Takimoto CH, Feng DD, McKenna K, Gip P, Liu J, Volkmer JP, Weissman IL, Majeti R. Therapeutic targeting of the macrophage immune checkpoint CD47 in myeloid malignancies. Front Oncol. 2020;9. doi:10.3389/fonc.2019.01380.
  • Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD, Van Rooijen N, Weissman IL. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286–99. doi:10.1016/j.cell.2009.05.045.
  • Boasman K, Simmonds MJ, Rinaldi CR. Expression of CD47 and calr in myeloproliferative neoplasms and myelodysplastic syndrome: potential new therapeutical targets. Blood. 2019;134:5031–5031. doi:10.1182/blood-2019-128422.
  • Jaiswal S, Chao MP, Majeti R, Weissman IL. Macrophages as mediators of tumor immunosurveillance. Trends Immunol. 2010;31:212–19.
  • Feng D, Gip P, McKenna KM, Zhao F, Mata O, Choi TS, Duan J, Sompalli K, Majeti R, Weissman IL, et al. Combination treatment with 5F9 and azacitidine enhances phagocytic elimination of acute myeloid leukemia. Blood. 2018;132:2729–2729. doi:10.1182/blood-2018-99-120170.
  • Zeidan AM, DeAngelo DJ, Palmer JM, Seet CS, Tallman MS, Wei X, Li YF, Hock N, Burgess MR, Hege K, et al. A phase I study of CC-90002, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory (R/R) acute myeloid leukemia (AML) and high-risk myelodysplastic syndromes (MDS): final results. Blood. 2019;134:1320–1320. doi:10.1182/blood-2019-125363.
  • Brierley CK, Staves J, Roberts C, Johnson H, Vyas P, Goodnough LT, Murphy MF. The effects of monoclonal anti-CD47 on RBCs, compatibility testing, and transfusion requirements in refractory acute myeloid leukemia. Transfusion. 2019;59(7):2248–54. doi:10.1111/trf.15397.
  • Chen JY, McKenna KM, Choi TS, Duan J, Brown L, Stewart JJ, Sompalli K, Vyas P, Schrier S, Majeti R, et al. RBC-specific CD47 pruning confers protection and underlies the transient anemia in patients treated with anti-CD47 antibody 5F9. Blood. 2018;132:2327–2327. doi:10.1182/blood-2018-99-115674.
  • Kidder K, Bian Z, Shi L, Liu Y. Inflammation unrestrained by SIRPα induces secondary hemophagocytic lymphohistiocytosis independent of IFN-γ. J Immunol. 2020;205:2821–33. doi:10.4049/jimmunol.2000652.
  • Sallman DA, Donnellan WB, Asch AS, Lee DJ, Al Malki M, Marcucci G, Pollyea DA, Kambhampati S, Komrokji RS, Van Elk J, et al. The first-in-class anti-CD47 antibody Hu5F9-G4 is active and well tolerated alone or with azacitidine in AML and MDS patients: initial phase 1b results. JCO. 2019;37:7009. doi:10.1200/JCO.2019.37.15_suppl.7009.
  • Sallman DA, Asch AS, Al Malki MM, Lee DJ, Donnellan WB, Marcucci G, Kambhampati S, Daver NG, Garcia-Manero G, Komrokji RS, et al. The first-in-class anti-CD47 antibody magrolimab (5F9) in combination with azacitidine is effective in MDS and AML patients: ongoing phase 1b results. Blood. 2019;134:569–569. doi:10.1182/blood-2019-126271.
  • Sallman DA, Al Malki M, Asch AS, Lee DJ, Kambhampati S, Donnellan WB, Bradley TJ, Vyas P, Jeyakumar D, Marcucci G, et al. Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in MDS and AML patients: phase ib results. JCO. 2020;38:7507. doi:10.1200/JCO.2020.38.15_suppl.7507.
  • Lonez C, Verma B, Hendlisz A, Aftimos P, Awada A, Van Den Neste E, Catala G, Machiels JP, Piette F, Brayer JB, et al. Study protocol for THINK: a multinational open-label phase I study to assess the safety and clinical activity of multiple administrations of NKR-2 in patients with different metastatic tumour types. BMJ Open. 2017;7:e017075. doi:10.1136/bmjopen-2017-017075.
  • Houchins JP, Yabe T, McSherry C, Miyokawa N, Bach FH. Isolation and characterization of NK cell or NK/T cell-specific cDNA clones. J Mole Cell Immunol. 1990;4:295–304. discussion 305.
  • Carapito R, Bahram S. Genetics, genomics, and evolutionary biology of NKG2D ligands. Immunol Rev. 2015;267:88–116.
  • Murad JM, Baumeister SH, Werner L, Daley H, Trébéden-Negre H, Reder J, Sentman CL, Gilham D, Lehmann F, Snykers S, et al. Manufacturing development and clinical production of NKG2D chimeric antigen receptor-expressing T cells for autologous adoptive cell therapy. Cytotherapy. 2018;20:952–63. doi:10.1016/j.jcyt.2018.05.001.
  • Spear P, Wu M, Sentman M, Sentman CL. NKG2D ligands as therapeutic targets. Cancer Immun. 2013;13:8.
  • Giuliani M, Poggi A. Checkpoint inhibitors and engineered cells: new weapons for natural killer cell arsenal against hematological malignancies. Cells. 2020;9:1578. doi:10.3390/cells9071578.
  • Zhang T, Barber A, Sentman CL. Generation of antitumor responses by genetic modification of primary human T cells with a chimeric NKG2D receptor. Cancer Res. 2006;66:5927–33. doi:10.1158/0008-5472.CAN-06-0130.
  • Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther. 2011;18:509–16. doi:10.1038/gt.2010.174.
  • Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol. 2002;3:1150–55. doi:10.1038/ni857.
  • Baumeister SH, Murad J, Werner L, Daley H, Trebeden-Negre H, Gicobi JK, Schmucker A, Reder J, Sentman CL, Gilham DE, et al. Phase I trial of autologous CAR T cells targeting NKG2D ligands in patients with AML/MDS and multiple myeloma. Cancer Immunol Res. 2019;7:100–12. doi:10.1158/2326-6066.CIR-18-0307.
  • Bert NL, Gasser S. Advances in NKG2D ligand recognition and responses by NK cells. Immunol Cell Biol. 2014;92:230–36. doi:10.1038/icb.2013.111.
  • Nikiforow S, Werner L, Murad J, Jacobs M, Johnston L, Patches S, White R, Daley H, Negre H, Reder J, et al. Safety data from a first-in-human phase 1 trial of NKG2D chimeric antigen receptor-T cells in AML/MDS and multiple Myeloma. Blood. 2016;128:4052–4052. doi:10.1182/blood.V128.22.4052.4052.
  • Anonymous Pioneering innovative therapies for patients with life-threatening diseases. ASH 2019 Presentation Update on r/r AML and MDS Program; 2019 Dec 9 [accessed Nov 27]. https://celyad.com/wp-content/uploads/2020/08/Celyad-ASH-Symposium-Presentation_December-9_vF.pdf
  • Sallman DA, Brayer JB, Poire X, Havelange V, Awada A, Lewalle P, Odunsi K, Wang ES, Lonez C, Lequertier T, et al. Results from the completed dose-escalation of the hematological arm of the phase I think study evaluating multiple infusions of NKG2D-based CAR T-cells as standalone therapy in relapse/refractory acute myeloid leukemia and myelodysplastic syndrome patients. Blood. 2019;134:3826–3826. doi:10.1182/blood-2019-128020.
  • Liu H, Wang S, Xin J, Wang J, Yao C, Zhang Z. Role of NKG2D and its ligands in cancer immunotherapy. Am J Cancer Res. 2019;9:2064–78.
  • Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature (London). 2005;436:1186–90. doi:10.1038/nature03884.
  • Al-Homsi AS, Purev E, Lewalle P, Abdul-Hay M, Pollyea DA, Salaroli A, Lequertier T, Alcantar-Orozco E, Borghese F, Lonez C, et al. Interim results from the phase I deplethink trial evaluating the infusion of a NKG2D CAR T-cell therapy post a non-myeloablative conditioning in relapse or refractory acute myeloid leukemia and myelodysplastic syndrome patients. Blood. 2019;134:3844–3844. doi:10.1182/blood-2019-128267.
  • Hong S, Rybicki L, Corrigan D, Hamilton BK, Sobecks R, Kalaycio M, Dean RM, Hill BT, Pohlman B, Jagadeesh D, et al. Targeted treatment and survival following relapse after allogeneic hematopoietic cell transplantation for acute leukemia and MDS in the contemporary era. Blood. 2019;134:4567–4567. doi:10.1182/blood-2019-124533.
  • Krishnamurthy P, Potter VT, Barber LD, Kulasekararaj AG, Lim ZY, Pearce RM, De Lavallade H, Kenyon M, Ireland RM, Marsh JCW, et al. Outcome of donor lymphocyte infusion after T cell-depleted allogeneic hematopoietic stem cell transplantation for acute myelogenous leukemia and myelodysplastic syndromes. Biol Blood Marrow Transplant. 2013;19:562–68. doi:10.1016/j.bbmt.2012.12.013.
  • Claiborne J, Bandyopathyay D, Roberts C, Hawks K, Aziz M, Simmons G, Wiedl C, Chung H, Clark W, McCarty J, et al. Managing post allograft relapse of myeloid neoplasms: azacitidine and donor lymphocyte infusions as salvage therapy. Leuk Lymphoma. 2019;60:2733–43. doi:10.1080/10428194.2019.1605066.
  • Zeiser R, Beelen DW, Bethge W, Bornhäuser M, Bug G, Burchert A, Christopeit M, Duyster J, Finke J, Gerbitz A, et al. Biology-driven approaches to prevent and treat relapse of myeloid neoplasia after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2019;25:e128–e140. doi:10.1016/j.bbmt.2019.01.016.
  • Lulla P, Naik S, Tzannou I, Mukhi S, Kuvalekar M, Robertson C, Ramos CA, Carrum G, Kamble RT, Gee AP, et al. Administering leukemia-directed donor lymphocytes to patients with AML or MDS to prevent or treat post-allogeneic HSCT relapse. Biol Blood Marrow Transplant. 2019;25:S10–S11. doi:10.1016/j.bbmt.2018.12.075.
  • Epperly R, Gottschalk S, Velasquez MP. A bump in the road: how the hostile AML microenvironment affects CAR T cell therapy. Front Oncol. 2020;10:262. doi:10.3389/fonc.2020.00262.
  • Sarhan D, Wang J, Sunil Arvindam U, Hallstrom C, Verneris MR, Grzywacz B, Warlick E, Blazar BR, Miller JS. Mesenchymal stromal cells shape the MDS microenvironment by inducing suppressive monocytes that dampen NK cell function. JCI Insight. 2020;5:5. doi:10.1172/jci.insight.130155.
  • Bergmann L, Maurer U, Weidmann E. Wilms tumor gene expression in acute myeloid leukemias. Leuk Lymphoma. 1997;25:435–43. doi:10.3109/10428199709039030.
  • Cilloni D, Gottardi E, Messa F, Fava M, Scaravaglio P, Bertini M, Girotto M, Marinone C, Ferrero D, Gallamini A, et al. Significant correlation between the degree of WT1 expression and the international prognostic scoring system score in patients with myelodysplastic syndromes. JCO. 2003;21:1988–95. doi:10.1200/JCO.2003.10.503.
  • Rautenberg C, Germing U, Pechtel S, Lamers M, Fischermanns C, Jäger P, Geyh S, Haas R, Kobbe G, Schroeder T, et al. Prognostic impact of peripheral blood WT1- mRNA expression in patients with MDS. Blood Cancer J. 2019;9:1–8. doi:10.1038/s41408-019-0248-y.
  • Jo T, Sakai K, Muranushi H, Okamoto Y, Tsukamoto T, Sugiura H, Matsui H, Ueda T, Okada K, Maeda T, et al. Pre-treatment WT1 mRNA expression level in peripheral blood predicts response and overall survival of myelodysplastic syndrome patients in the azacitidine era. Blood. 2013;122:1528–1528. doi:10.1182/blood.V122.21.1528.1528.
  • Gao L, Bellantuono I, Elsasser A, Marley SB, Gordon MY, Goldman JM, Stauss HJ. Selective elimination of leukemic CD34+ progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 2000;95:2198–203. doi:10.1182/blood.V95.7.2198.
  • Pinilla-Ibarz J, May RJ, Korontsvit T, Gomez M, Kappel B, Zakhaleva V, Zhang RH, Scheinberg DA. Improved human T-cell responses against synthetic HLA-0201 analog peptides derived from the WT1 oncoprotein. Leukemia. 2006;20:2025–33. doi:10.1038/sj.leu.2404380.
  • Ohminami H, Yasukawa M, Fujita S. HLA class I-restricted lysis of leukemia cells by a CD8(+) cytotoxic T-lymphocyte clone specific for WT1 peptide. Blood. 2000;95:286–93. doi:10.1182/blood.V95.1.286.
  • Rezvani K, Yong ASM, Mielke S, Jafarpour B, Savani BN, Le RQ, Eniafe R, Musse L, Boss C, Kurlander R, et al. Repeated PR1 and WT1 peptide vaccination in montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies. Haematologica. 2011;96:432–40. doi:10.3324/haematol.2010.031674.
  • Rezvani K, Yong ASM, Mielke S, Savani BN, Musse L, Superata J, Jafarpour B, Boss C, Barrett AJ. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood. 2008;111:236–42. doi:10.1182/blood-2007-08-108241.
  • Brayer J, Lancet JE, Powers J, List A, Balducci L, Komrokji R, Pinilla-Ibarz J. WT1 vaccination in AML and MDS: a pilot trial with synthetic analog peptides. Am J Hematol. 2015;90:602–07. doi:10.1002/ajh.24014.
  • Suzuki T, Ueda Y, Ogura M, Uchida T, Ozawa K, Miyakoshi S, Naoe T, Murata M, Kizaki M, Uike N, et al. A phase 1/2 study of WT1 peptide cancer vaccine WT4869 in patients with myelodysplastic syndromes (MDS). Blood. 2015;126:2868–2868. doi:10.1182/blood.V126.23.2868.2868.
  • Keilholz U, Letsch A, Busse A, Asemissen AM, Bauer S, Blau IW, Hofmann WK, Uharek L, Thiel E, Scheibenbogen C, et al. A clinical and immunologic phase 2 trial of wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood. 2009;113:6541–48. doi:10.1182/blood-2009-02-202598.
  • Molldrem J, Dermime S, Parker K, Jiang YZ, Mavroudis D, Hensel N, Fukushima P, Barrett AJ. Targeted T-cell therapy for human leukemia: cytotoxic T lymphocytes specific for a peptide derived from proteinase 3 preferentially lyse human myeloid leukemia cells. Blood. 1996;88:2450–57. doi:10.1182/blood.V88.7.2450.bloodjournal8872450.
  • Molldrem JJ, Lee PP, Kant S, Wieder E, Jiang W, Lu S, Wang C, Davis MM. Chronic myelogenous leukemia shapes host immunity by selective deletion of high-avidity leukemia-specific T cells. J Clin Invest. 2003;111:639–47. doi:10.1172/JCI200316398.
  • Qazilbash MH, Wieder E, Thall PF, Wang X, Rios R, Lu S, Kanodia S, Ruisaard KE, Giralt SA, Estey EH, et al. PR1 peptide vaccine induces specific immunity with clinical responses in myeloid malignancies. Leukemia. 2017;31:697–704. doi:10.1038/leu.2016.254.
  • Almstedt M, Blagitko-Dorfs N, Duque-Afonso J, Karbach J, Pfeifer D, Jäger E, Lübbert M. The DNA demethylating agent 5-aza-2ʹ-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells. Leuk Res. 2010;34:899–905. doi:10.1016/j.leukres.2010.02.004.
  • Satie A, Meyts ER, Spagnoli GC, Henno S, Olivo L, Jacobsen GK, Rioux-Leclercq N, Jégou B, Samson M. The cancer-testis gene, NY-ESO-1, is expressed in normal fetal and adult testes and in spermatocytic seminomas and testicular carcinoma. Lab Invest. 2002;82:775–80. doi:10.1097/01.LAB.0000017169.26718.5F.
  • Zendman AJW, Ruiter DJ, Van Muijen GNP. Cancer/testis-associated genes: identification, expression profile, and putative function. J Cell Physiol. 2003;194:272–88. doi:10.1002/jcp.10215.
  • Scanlan MJ, Simpson AJG, Old LJ. The cancer/testis genes: review, standardization, and commentary. Cancer Immun. 2004;4:1.
  • Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, Decock J. NY-ESO-1 based immunotherapy of cancer: current perspectives. Front Immunol. 2018;9:947. doi:10.3389/fimmu.2018.00947.
  • De Smet C, De Backer O, Faraoni I, Lurquin C, Brasseur F, Boon T. The activation of human gene MAGE-1 in tumor cells is correlated with genome-wide demethylation. Proc Natl Acad Sci USA. 1996;93:7149–53. doi:10.1073/pnas.93.14.7149.
  • Atanackovic D, Luetkens T, Kloth B, Fuchs G, Cao Y, Hildebrandt Y, Meyer S, Bartels K, Reinhard H, Lajmi N, et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am J Hematol. 2011;86:918–22. doi:10.1002/ajh.22141.
  • Srivastava P, Paluch BE, Matsuzaki J, James SR, Collamat-Lai G, Blagitko-Dorfs N, Ford LA, Naqash R, Lübbert M, Karpf AR, et al. Induction of cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monotherapy. Oncotarget. 2016;7:12840–56. doi:10.18632/oncotarget.7326.
  • Griffiths EA, Srivastava P, Matsuzaki J, Brumberger Z, Wang ES, Kocent J, Miller A, Roloff GW, Wong HY, Paluch BE, et al. NY-ESO-1 vaccination in combination with decitabine induces antigen-specific T-lymphocyte responses in patients with myelodysplastic syndrome. Clin Cancer Res. 2018;24:1019–29. doi:10.1158/1078-0432.CCR-17-1792.
  • Steger B, Floro L, Amberger DC, Kroell T, Tischer J, Kolb H-J, Schmetzer HM. WT1, PRAME, and PR3 mRNA expression in acute myeloid leukemia (AML). J Immunother. 2020;43:204–15. doi:10.1097/CJI.0000000000000322.
  • Borrello I, Sotomayor EM, Cooke S, Levitsky HI. A universal granulocyte-macrophage colony-stimulating factor-producing bystander cell line for use in the formulation of autologous tumor cell-based vaccines. Hum Gene Ther. 1999;10:1983–91. doi:10.1089/10430349950017347.
  • Lozzio CB, Lozzio BB. Human chronic myelogenous leukemia cell-line with positive philadelphia chromosome. Blood. 1975;45:321–34. doi:10.1182/blood.V45.3.321.321.
  • Smith BD, Kasamon YL, Kowalski J, Gocke C, Murphy K, Miller CB, Garrett-Mayer E, Tsai H-L, Qin L, Chia C, et al. K562/GM-CSF immunotherapy reduces tumor burden in chronic myeloid leukemia patients with residual disease on imatinib mesylate. Clin Cancer Res. 2010;16:338–47. doi:10.1158/1078-0432.CCR-09-2046.
  • Robinson TM, Prince GT, Thoburn C, Warlick E, Ferguson A, Kasamon YL, Borrello IM, Hess A, Smith BD. Pilot trial of K562/GM-CSF whole-cell vaccination in MDS patients. Leuk Lymphoma. 2018;59:2801–11. doi:10.1080/10428194.2018.1443449.
  • Saft L, Björklund E, Berg E, Hellström-Lindberg E, Porwit A. Bone marrow dendritic cells are reduced in patients with high-risk myelodysplastic syndromes. Leuk Res. 2013;37:266–73. doi:10.1016/j.leukres.2012.10.010.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.