Abstract
Cell therapy currently performs an important role in the treatment of patients with various hematological malignancies. The response to the cell therapy is regulated by multiple factors including the patient's immune system status, genetic profile, stage at diagnosis, age, and underlying disease. Cell therapy that does not require genetic manipulation can be mediated by donor lymphocyte infusion strategies, selective depletion in the post-transplant setting and the ex vivo expansion of antigen-specific T cells. For hematologic malignancies, cell therapy is contributing to enhanced clinical responses and overall survival and the immune response to cell therapy is predictive of response in multiple cancer types. In this review we summarize the available T cell therapeutics that do not rely on gene engineering for the treatment of patients with blood cancers.
Keywords: Adoptive T Cell Therapy, Tumor Associated Antigens, EBV, leukemia, lymphoma
Introduction
The burgeoning field of immunotherapy for hematological malignancies under active clinical investigation includes adoptive transfer of antigen-specific and specificity-augmented genetically-engineered T cells; T regulatory cell suppression; co-stimulation agonists; and vaccines that enhance antigen processing and presentation effectiveness. Early phase clinical trials utilizing T cells are releasing promising preliminary data. The FDA approval of CD19-directed chimeric antigen receptor (CAR) T cells, which have demonstrated spectacular responses in salvage therapy settings, has revolutionized treatment of CD19+ B cell malignancies. Nevertheless, severe on-target, off-tumor toxicities, including healthy B cell aplasia, cytokine release syndrome and neurotoxicity, means that these studies can currently only be conducted at institutions that can provide intensive care support. While clinicans are learning to better manage these biological limits the current paucity of suitable antigenic targets further limits the broader applicability of this approach to both hematologic and solid tumors. However, while less in the public eye, effective T cell therapies which do not employ gene-engineering are also being developed. Here we review the application of these non-gene-modified T cells in the treatment of hematological malignancies.
Role of the immune system in hematological cancer and immunogenic features of current treatments
Many hematological malignancies arise in B and T lymphocytes and the tumor microenvironment comprises active interactions between tumor and immune cells. CD4+ T cells interact with antigen presenting cells (APC) exhibiting tumor peptides in their MHC Class II (HLA DR, DP, DM, DOA, DOB, DQ) molecules. CD8+ T cells can directly recognize tumor cells displaying peptides in their major histocompatibility (MHC) Class I (HLA A, B, C) molecules, stimulating activation against the neoplastic cell. Dendritic cells (DC)1–3, B cells4–7 and macrophages2,3,8–10 all have the capacity to cross-present tumor-associated peptides on MHC-I and, providing sufficient costimulation is concurrently supplied, can generate a robust T cell activation against the tumor peptide(s). The classification cytotoxic T lymphocyte (CTL) has primarily been applied to CD8+ T cells, however the data shows that more than just “helper cells” who provide “help” for B cells and CD8+ T cells, CD4+ T cells also act as “CTLs” in their own right11,12. Activated antigen-specific T cells form immunological synapses with target cells, subsequently releasing cytokines such as interferon-gamma (IFN-γ), tumor-necrosing factor alpha (TNF-α) and TNF-related apoptosis-inducing ligand (TRAIL). These cytokines, along with cyototoxic perforin and granzyme molecules and the transmembrane protein FAS ligand, lead to lysis and apoptosis of tumor cells. Our current understanding of immune surveillance posits that this T cell-mediated tumor cell killing is carried out during the elimination phase, during which time tumor growth is controlled by the immune system13.
It is now understood that beyond elimination of rapidly dividing cells, among the reasons for the success of chemotherapy and radiotherapy is their generation of non-targeted innate and adaptive immune activation by triggering immunologic tumour cell death and disrupting immune suppression and tolerance (reviewed in14,15). Chemotherapeutic tumour cell death can trigger immunoadjuvant pathways16–18 while cyclophosphamide-mediated blocking of immune suppression occurs via selective depletion of regulatory T cells (TREGs)19. Radiotherapy of cancer tissue also induces immunogenic tumor cell death and destruction of immune barriers, both of which can lead to increased immune cell infiltration, increased tumor-associated antigen (TAA) presentation and T cell activation20–24. Thus, alongside their roles as debulking and cytotoxic agents, certain chemotherapies and radiotherapy, are increasingly regarded as beneficial primers or adjuncts for cancer immunotherapy14,20,25–30. Unfortunately, chemotherapy and radiotherapy are non-specific treatments that result in substantial bystander tissue toxicities, including late sequelae. Immunotherapies have the potential to deliver on the vision for effective targeted approaches that minimize damage to healthy tissue. The immune system's vital role in control of hematological malignancies is demonstrated in the graft versus tumor (graft versus leukemia; GVT; GVL) effect that is seen in a proportion of patients following allogeneic hematopoietic stem cell transplant (allo-HSCT)30,31. But this immune-mediated response is insufficient and relapse continues to be the greatest reason for HSCT failure.
Allogeneic bone marrow or peripheral blood stem cell transplant (HSCT)
Through its potent GVL effect HSCT represents the original form of successful cell therapy for hematological malignancy. Despite a huge reduction in transplant-related mortality in this century, the well-recognized complications from the procedure, which include graft-versus-host disease (GVHD), continue to limit its use. In selecting the best treatment for the individual with a hematologic malignancy current disease-free survival estimates following HSCT must be continually balanced against improved outcomes from non-transplant treatments. Detailed risk stratification of individual diseases has improved our ability to select the best treatment approach. For acute myelogenous leukemia (AML) “good-risk” patients receive non-transplant treatments while the preferred treatment option for higher risk AML patients is an allogeneic HSCT in first complete remission32. Indications for HSCT in AML continue to change as more patients achive long-term survival with novel therapies33. In acute lymphoblastic leukemia (ALL) chemotherapy achieves high cure rates in children and young adults and HSCT is seldom indicated. However in older adults with ALL HSCT is widely indicated34. In all types of acute leukemia HSCT is generally indicated in second or subsequent remission when primary treatment has failed. HSCT is also indicated for many patients with myelodyasplastic syndrome and some patients with myeloproliferative disorders. However the role of HSCT in chronic leukemias continues to dwindle: in chronic myelogenous leukemia (CML), following successful development of tyrosine kinase inhibitors, HSCT is seldom required, except in treatment failure and for selected children with CML35,36. In chronic lymphocytic leukemia (CLL) new treatments such as bruton kinase inhibitors and novel bi-specific antibodies have also reduced the indications for HSCTin the last decade37. HSCT continues to be used usually as a second line treatment for aggressive lymphomas.
Improving the GVL effect through manipulation of donor immune cells
HSCT as a treatment for a wide range of hematological malignancies continues to improve with a continuing fall in transplant-related mortality extending acceptable transplant safety. However, there remains a substantial risk of leukemic relapse after HSCT, particularly for patients over 60 years of age who can generally only tolerate less intensive conditioning, and for those with no fully compatible donors who require additional pharmacological immunosuppression and/or graft manipulation. In 1990 Kolb and colleagues set the scene for T cell therapy as a means to augment the GVL effect. They used donor lymphocyte infusions (DLI) to achieve sustained remissions in CML patients who had relapsed after allogeneic SCT. Other investigators sought to modify the graft contents to selectively remove GVHD-reacting cells while preserving GVL-reactive cells38. The successful application of this “selective depletion” strategy can permit the infusion of large numbers of leukemia-reactive donor T cells without GVHD or the need for immunosuppression to prevent GVHD resulting in superior GVL effects. In recent years cell-mediated therapeutic approaches have increased in their sophistication, diversity and efficacy. Despite their toxicities, genetically engineered T cells such as CAR T cells and T cells with gene-modified TCRs have caused excitement because of their potency and novelty39 . However less risky but equally specific ways to create leukemia-reactive T cells have been developed following technologies first used to successfully control virus reactivation after HSCT. Furthermore the unique properties of natural killer (NK) cells in specifically targeting malignant cells without risking GVHD have led to studies of the adoptive transfer of NK cells to treat malignant disease40–42. Table 1 summarizes the various strategies used to augment the GVL effect. Table 2 lists the available cell-based therapies for hematological malignancies and Table 3 summarizes clinical trials that have utilized non- receptor engineered T cells for hematological malignancies.
Table 1. Cell transfer-based approaches to enhance GVL effect of allogeneic HSCT.
| Graft manipulation to prevent GVHD while conserving GVL |
| Selective allodepletion |
| CD8+ depletion |
| Naïve T cell depletion |
| Donor lymphocyte infusion (DLI) |
| Whole buffy coats |
| DLI + interferon/IL-2 |
| CD4-selected DLI |
| T cells genetically engineered for leukemia speficicity |
| CAR-T cells* |
| TCR modified T cells* |
| Antigen-specific T cells |
| Targeting minor histocompatibility antigens |
| Targeting tumor antigen |
| Targeting viral antigens expressed by lymphoma |
| Ex vivo selected and expanded NK cells* |
Not discussed in this review
Table 2. Non-gene-modified T cell therapies available for patients with hematological malignancies.
| Treatment Approach | Description |
|---|---|
| Unmodified donor lymphocyte infusion (DLI)30,31,43–50 | Infusion of donor lymphocytes to mount GVT response |
| Modified DLI51–56 | Depletion of DLI subpopulations, TREGs, naïve T cells, to select for most potent CTLs |
| Tumor antigen-targeted cytotoxic T lymphocytes (CTLs)57–63 | Infusion of ex vivo expanded tumor-specific allogeneic or autologous CTLs for elimination of tumor targets |
| Viral antigen targeted T-cells64–66 | Infusion of ex vivo-expanded virus-specific allogeneic or autologous T-cells to minimize viral reactivation during immunosuppressive period post transplant |
| Tumor peptide-loaded DC vaccination67–70 | Infusion of peptide loaded DCs for in vivo presentation of antigen to endogenous T cells |
| NK cells and cytokine-induced killer cells41 | Infusion of ex vivo expanded tumor-specific allogeneic or autologous NK cells or CIKs for elimination of tumor targets |
Table 3. Summary of Clinical Trials Using Non- Receptor Engineered T cells for Hematological Malignancies.
| Trial # | Intervention | Tumor Target | Primary Outcome | Secondary Outcome | Enrollment |
|---|---|---|---|---|---|
| NCT01627275 | Naive T Cell Depleted DLI | N/A | MTD | Immunological recovery; incidence of aGVHD, cGVHD & opportunistic infections | 28 |
| NCT00675831 | CD25+ Depleted DLI | N/A | Feasibility of CliniMACS to engineer CD25- product; Safety; | Clinical response; Immunological Impact | 24 |
| NCT02203903 (RESOLVE) | Tumor associated antigen lymphocytes (TAA-CTL) | PRAME, WT-1, survivin | Safety | TAA-CTL responses | 11 |
| NCT00052598 | Allogeneic CD8+CTL clones+aldesleukin (IL-2) | PR3 | Toxicity | Persistence; migration to BM; duration of response; proportion of responders | 7 |
| NCT00052520 | Allogeneic CD8+CTL clones+aldesleukin (IL-2) | WT-1 | Toxicity | Relapse | 37 |
| NCT00107354 | Allogeneic CD8+ C TL clones+aldesleukin (IL-2) | mHAg | Toxicity | Persistence; migration to BM; Antileukemic activity | NP |
| NCT02895412 (INTACT-WT1) | Allogeneic C TLs | WT-1 & multiple pathogens (CMV,Adv, EBV, VZV, IFV, BKV, fungal infections) | TRAE | N/A | 20 |
| NCT02074657 (LANK-2) | Activ ated and expanded natural killer cells (NKAEs) | N/A | Safety | Febrile neutropenia, or infection incidence; hematological recovery; ORR; immune reconstitution | 13 |
| NCT00620633 | Allogeneic C TLs | WT-1 | Toxicity | Disease progression; CTL survival & proliferation | 22 |
| NCT00460629 | Prophylactic donor CTLs | LAA | Feasibility; kinetics of BCR-ABL load; | Rates of aGVHD, cGVHD & infections | 20 |
| NCT00002663 | Allogeneic C TLs | EBV | Efficacy; expansion & duration; durability of response (time) | N/A | 84 |
| NCT01948180 (CITADEL) | Autologous T cells | EBV | ORR | CRR; Response Duration; time to response; PFS; DFS; OS; AEs | 35 |
| NCT00779337 | Autologous CTLs | EBV (AdE1- Latent Membrane Protein) | Feasibility; safety; reconstitution of EBV-specific CTL immunity with anti-viral efficacy | Optimal dose; clinical efficacy | 8 |
| NCT00005606 | Allogeneic CTLs | EBV | NP | NP | 10-20 |
| NCT01636388 | Allogeneic CTLs | EBV (LMP) | Safety, toxicity | Feasibility | 45 |
| NCT01956084 | Third party CTLs | EBV (LMP) | DLT | Survival & function of LMP-specific CTLs | 24 |
| NCT02057445 | Allogeneic CTLs | EBV | Safety; development of bank of third-party CTLs | Response rate | 18 |
| NCT01498484 | Allogeneic CTLs | EBV | Efficacy | In vivo expansion and duration of EBV-CTLs; durability of response | 112 |
| NCT01447056 | Allogeneic CTLs | EBV (LMP) | DLT; safety | Safety of dosing; immune function of CTLs; dz response | 18 |
| NCT01555892 (GRALE) | CTLs | EBV (LMP, BARF1 & EBNA1) | Toxicity | Survival and immune function of LMP/BARF1/EBNA1-specific CTLs; anti-viral and anti-tumor effects of CTLs | 136 |
| NCT00062868 | CTLs | EBV (LMP1/2) | DLT | Survival and immune function of LMP1/2-specific CTLs; anti-viral load and anti-tumor effects | 89 |
| NCT02287311 (MABEL) | Allogeneic CTLs | EBV (LMP, BARF1 And EBNA1) | DLT | Persistence; proliferation in vivo; CTL response to viral antigens; CR; PR | 42 |
| NCT00002663 | Allogeneic CTLs | EBV | Efficacy; In vivo ex pansion and duration; durability of clinical responses | N/A | 84 |
| NCT02973113 (PREVALE) | CTLs+Nivolumab | EBV | DLT | Duration of response; | 36 |
| NCT01333046 (TACTAL) | C TLs+/- 5-azacytidine | NY-ESO-1, MAGEA4, PRAME, Survivin and SSX | Safety (AEs/SAEs) | expansion, persistence and anti-tumor effects of CTLs; epitope spreading; | 74 |
TAAT: Tum or Associated Antigen –specific T cells; DLI: Donor Lymphocyte Infusion; MTD: maximum tolerated dose; BM: bone marrow; CTL: Cytotoxic T Lymphocytes; mHAg: minor histocompatibility antigen; NP: Not Provided; TRAE: Treatment-Related Adverse Events; cytomegalovirus (CMV), Adenovirus (Adv), Epstein Barr virus (EBV), Varicella-Zoster virus (VZV), Influenza, BK virus (BKV), and fungal infections; LAA: Leukemia-Associated Antigens; ORR: Overall Response Rate; CRR: Complete Response Rate; PFS: Progression Free Survival; DFS: Disease Free Survival; OS: Overall Survival; AEs: Adverse Events; DLT: Dose Limiting Toxicity;
Donor lymphocyte infusions (DLI)
Infusions of non-specific, third-party, naïve and antigen-experienced lymphocytes can mount a response against the recipient's tumor and are a common treatment for leukemic relapse after allo-HSCT. When successful DLIs undertake a GVT effect that eliminates neoplastic host cells. However relapse and GVHD continue to present significant challenges with DLI, particularly in acute leukemias and non-Hodgkin's lymphoma (NHL).
The GVT response post-DLI is observed most clearly in CML where DLI +/- the TK inhibitor Imatinib after allo-HSCT is particularly effective in treating relapse34,71. A retrospective analysis of 500 CML patients treated with DLI after post-allo-HSCT relapse identified some of the factors associated with increased overall survival (OS) without secondary GVHD43. In patients with cytogenetic, molecular and hematological relapse the probability of greater OS without GVHD was higher in patients who i) received DLIs early in CML relapse (at molecular or cytogenetic, rather than hematological diagnosis); ii) did not have chronic GVHD prior to infusion; and iii) received DLIs more than 1 year after allo-HSCT. For patients with hematological relapse the dose of CTLs, donor-recipient sex mismatch and chronic GVHD prior to DLI were independently associated with greater probability of reduced OS and secondary GVHD. While they occur at lower levels, GVT effects in the absence of clinical GVHD were also identified in this study, and have been reported elsewhere44. Thus GVHD is not a prerequisite for a successful GVT effect, though GVHD often accompanies an effective GVT response.
Whereas DLI is extremely effective in CML, this approach has more limited utility in acute leukemias. The efficacy of DLI in acute leukemias as a pre-emptive measure post-allo-HSCT (i.e. administering DLI to patients with minimal residual disease (MRD) or mixed chimerism), has been systematically investigated by the Acute Leukemia Working party of the European Society for Blood and Marrow Transplantation (EBMT)45. The results showed that DLI given preemptively was effective in 69% of patients. Reduction in MRD was achieved in 71% (15/21) of patients and increased donor chimerism in 68% (110/163). While encouraging, the authors note that further refinement is required to identify which patients may benefit from preemptive DLI as well as discussing the potential role for prophylactic DLI (i.e. DLI administered to high risk patients when in complete response [CR]). It was also noted that GVHD-related mortality was as high as 5% when administered as a pre-emptive strategy. Other groups have consolidated the knowledge that MRD and GVHD status are prognostic for patient relapse, and identify that multiple consolidation therapy and DLI can drive patients into MRD negative status31,46,47,56. Specifically, there is a robust association between cGVHD and achievement of a successful anti-tumor response to DLI suggesting that clinically manageable cGVHD may be an acceptable goal for the majority of patients with high-risk hematologic cancers post-allo-HSCT. However, evidence of GVT responses in the absence of GVHD confirms that we need to identify how to uncouple the GVT and GVHD responses, ameliorating both unnecessary patient suffering and the ongoing treatment expense of immune suppression agents.
Selective Depletion and Allodepleted DLI
Investigators have tried various approaches to enhance the GVT effect of DLI including: chemotherapy plus DLI56 (NCT02046122; NCT01390311; NCT00003145; NCT01369368); total body irradiation plus DLI (NCT00003196); GM-CSF mobilization pre-donor harvesting (NCT01158118); or accompanying peptide administration72 (NCT00270452); and administration of interferon or interleukin-2 (NCT02331706; NCT00005802) to stimulate an antitumor immune response. Several groups have however evaluated “selective depletion” DLI strategies to enhance the GVT response while limiting GHVD.
Investigators at the National Heart Lung and Blood Institute (NHLBI), National Intitutes of Health used anti-CD25 immunotoxin to selectively deplete host-reactive donor T cells from HLA-identical sibling peripheral blood stem cells. The results from that trial in elderly patients with advanced hematological malignancies suggested that elimination of alloreactive donor cells can enhance lymphocyte recovery and reduce the incidence of severe GVHD73. Those conclusions were subsequently supported by another trial from Baylor College of Medicine where they evaluated immune reconstitution after adding back two different doses of allo-depleted donor T cells in the haploidentical setting. That study similarly demonstrated low acute and chronic GVHD and enhanced T cell reconstitution following anti-CD25 immunotoxin allodepleted T cell infusion74. The group at the NHLBI also evaluated allodepletion using a novel photodepletion approach. Allo-reactive T cells that retained the photosensitizer 4, 5-dibromorhodamine 123 (TH9402) were eliminated by exposure to visible light. Results from this study in HLA-matched sibling donors likewise suggested that depletion of host-reactive T cells reduced the severity of GVHD, however, unlike the two previous studies, delayed immune recovery was observed in this cohort and clinical outomes were not improved75. A more recent study using photo-mediated allo-depletion in haploidentical transplants reduced transplant-related mortality (TRM) to 17% compared to 34% for HSCT alone76. One-year overall survival was increased to 67% in the allo-depleted group compared with 20% in the HSCT-only arm. The reduced TRM was attributed to the retention of immune protection in the group receiving allo-depleted T cells, as infections were the primary cause of TRM in the control cohort. Another promising pre-clinical study has demonstrated adenosine-mediated selective depletion of allo-reactive T cells in HLA-mismatch mixed lymphocyte reactions52. Virus- and leukemia-specific non-alloreactive naïve, central, and effector memory T cells were retained and T cells recognizing leukemia-associated antigens were efficiently generated in vitro from the adenosine-treated cell product. However these pre-clinical studies are yet to be tested in clinical trials.
A group at Dana-Farber more recently hypothesized that selective depletion of regulatory CD4+CD25+ cells (TREGs) from DLI populations prior to infusion could boost the anti-tumor effect, by removing their suppression of the GVT response51. Their Phase I trial compared a cohort of patients who received unmodified DLI with a cohort that received CD4+CD25+-depleted DLI. CD25+ cells were removed by antibody conjugated magnetic bead selection. Rates of severe GVHD were 19% and 33% at 8wks and 1 year respectively. These rates were similar to rates observed in other studies of unmodified DLI and, importantly, anti-tumor responses were not dependent upon GVHD. Improved outcomes were achieved in the CD4+CD25+-depleted DLI group (60% response rate at 8-12 weeks versus a 14% response rate in patients receiving unmodified DLI). Moreover, in patients receiving unmodified DLI, the 1 year event-free survival (EFS) was 0%, compared with 27% in the TREG-depleted group. Increased proportions of naïve (TN) and T central memory (TCM) cells, and reductions in T effector memory (TEM) and terminal effectors were observed49. Overall, the results from this small trial suggest that CD25-depleted DLI may be somewhat more effective than unmodified DLI for the management of relapsed acute leukemia post-allo-HSCT.
Finally, following the observation in mice that naïve T cells cause more severe GVHD than memory T cells, a group in Seattle evaluated naïve T cell depletion as a strategy to prevent GVHD while preserving GVL77. Positive selection of CD34+ progenitor cells was followed by depletion of CD45RA+ cells from the CD34-negative fraction by antibody-conjugated magnetic beads. In this trial, incidence of acute GVHD was not reduced but an impressive reduction in chronic GVHD was achieved (9% compared with historical rates of approximately 50%). Furthermore, T cell tolerance was dramatically faster in the TN-depleted arm: time to 60% completion of corticosteroids was around 150 days in the TN-depleted cohort compared with approximately 1400 days in the control arm. Figure 1 provides a visual illustration of the main selective depletion approaches that have been evaluated in clinical trials to date.
Figure 1. Non-gene-modified selective depletion-based T cell immunotherapeutics for hematological malignancies.

Donor peripheral blood stem cell (PBSC) populations can be enriched for cell subsets of interest or depleted of undesirable subsets such as regulatory T cells (TREGs), naïve T cells (TN) and allo-reactive T cells. A) Monoclonal antibodies (mAB) linked to metal spheres bind to cellular targets and, when passed through a magnetic column, cells attached to the spheres are retained within the column. This positive fraction, or the negative fraction, can be collected for downstream application, such as TN (CD34-CD3+CD45RO-), TREG (CD3+CD4+FoxP3+CD25+) or allo-reactive T cell (CD69+) selective depletion. B)The interleukin-2 (IL-2) receptor α chain CD25, a T cell activation marker, can also be targeted by pharmaceutical approaches. CD25 immunotoxin is a murine anti-CD25 mAb linked to deglycosylated ricin α chain. Co-culture of recipient lymphoblastoid cell lines (LCL) with donor peripheral blood mononuclear cells (PBMC) results in allo-reactivity of donor T cells. Allo-reactive cells can be removed by overnight treatment with CD25 immunotoxin, or by 72 hour culture with 2 mmol/L adenosine. Following haploidentical stem cell transplant (HSCT) the allo-depleted donor cells are also transfused. C) Allo-depletion can also be achieved by incubating mixed lymphocyte reactions (MLR) in the presence of the photo sensitizer 4,5-dibromorhodamine 123 (TH9402). Activated cells retain TH9402 and are eliminated upon exposure to visible light. Patients received infusions of autologous photo-depleted (PD) effector T cells along with CD34-selected stem cells from HLA-matched siblings.
Antigen-specific T cells
Targeting leukemia-associated antigens
In contrast to selective depletion or suicide gene DLI strategies, another approach to enhance the GVL effect while minimizing GVHD is to selectively expand the tumor antigen-specific T cells of interest. GVT responses have been attributed to CD8+, CD4+ and NK cells. Consequently tumor-specific CTLs and, to a lesser extent, NK cells targeting leukemia antigens or minor histocompatibility antigens (mHag), in relapsed patients have attracted strong attention by investigators59,61–63,78–80. (NCT02203903; NCT02074657; NCT00052598; NCT00052520; NCT00107354; NCT02895412; NCT00620633; NCT00460629). While the focus of this review is on T cells it is appropriate to mention that other lymphocytes can contribute to the anti-leukemia/lymphoma response. NK cells and cytokine-induced killer cells are also relatively prevalent in the field of immunotherapy for hematologic malignancies and are regarded as a feasible approach. Data has shown that they can provide benefit in the context of acute leukemia, particularly for AML, with little or no GVHD41.
Targeting Minor Histocompatibility Antigens
Minor histocompatibility antigens that cause graft rejection in transplant recipients and play a role in GVHD, may yet be harnessed to drive a GVT effect in acute leukemia81. mHAgs are peptides presented on the cell surface by HLA molecules. These peptides differ between individuals by just a few amino acids and can induce anti-mHAg T cell responses from mHAg-negative donors. The best-characterized human mHAg is hematopoietic tissue-restricted HA-1, which is presented by HLA-A2. HA-1's ability to drive a GVL response has been reported, however T cells specific for HA-1 could potentially induce collateral damage to surrounding tissue and thereby contribute to GVHD in the early post-HSCT period when DCs of host origin remain in recipient tissues79. In one trial Warren and Riddell targeted mHAgs that were identified on the basis of T cell clonal response, but not molecularly characterized. Five of seven patients achieved complete, though transient, remissions82 (NCT00107354). Grade 3 or 4 pulmonary toxicity occurred in three patients within 2 hours of CTL infusions (>1 × 109 cells), and was attributed to the T cell therapy. The fact that expression of some of the targeted mHAgs was not restricted to hematopoietic cells was revealed during post-trial analysis of these 3 patients when the genes encoding a number of the targeted minor H antigens were identified and tissue expression examined. To minimize damage to non-leukemic tissue it is possible to isolate mHAg-specific CTLs that do not recognize the same mHAg on surrounding epithelial, GI tract or liver tissues83. However the sophisticated nature of these time-heavy, expensive assays currently limits their availability.
Targeting Tumor-Associated Antigens
Various approaches for targeting specific leukemia-associated antigens (LAA) have been attempted. Subcutaneous administration of the peptides Wilms Tumor protein 1 (WT1) and pathogenesis-related protein 1 (PR1) in Montanide adjuvant along with GM-CSF-mediated immune stimulation resulted in detection of CD8+WT1+PR1+ CTLs in 8/8 myeloid leukemia patients and coincided with reduction of WT1 mRNA transcripts69 (NCT00270452). However, this anti-leukemia response was not durable and further boosts of peptides+Montanide failed to elicit further benefit84,85. The significant decline in vaccine-specific CD8+ T cells with further peptide+Montanide boosts was also observed by another group who cautioned against the use of Montanide and/or CpG as vaccine adjuvants in the leukemia setting86. To improve on these vaccine strategies, several groups have evaluated the use of leukemia-specific T cells. One of the first reports of ex vivo expanded leukemia-specific T cells for the treatment of patients in accelerated phase CML showed not only complete eradication of CML after the third dose, but interestingly treatment success was not accompanied by GVHD78. These results were particularly exciting as they demonstrated that separation of the GVHD and GVL responses was possible. This early trial selected leukemia-specific T cells, not by their ability to recognize known leukemia antigens, but by their ability to inhibit CML progenitor cell growth in vitro. In this simple but elegant approach three T cell products were generated that lysed CML cells but not donor hematopoietic progenitor cells.
Leukemia-associated antigen-specific T cells
Studies have attempted to enhance leukemia-specific T cell specificity by expanding T cells specific for known leukemia-associated antigens such as cancer/testis antigens (MAGE-A3, A4, PRAME; NCT02203903), transcription factors (WT187; NCT02203903; NCT00052520; NCT02895412; NCT00620633; NCT00460629), pro-survival/anti-apoptosis proteins (survivin; NCT02203903), serine proteases (PR3; NCT00052598; PR1; NCT0046062961) and oncogene products (BCR-ABL; NCT00460629) (Table 4). These approaches are achieved by priming T cells with specific LAA epitopes or mixtures of overlapping peptide sequences that span known or predicted antigenic regions. The first report of adoptively transferred WT1-specific CTLs for high risk leukemia patients post-HSCT described administration of allogeneic CD8+ T cells specific for the 9mer HLA-A*0201-restricted WT1 epitope, WT1126-134 (RMFPNAPYL)63. Seven patients that received IL-2, IL-7, IL-15-expanded WT1 CTLs relapsed, whereas 4/4 patients who received IL-2, IL-7, IL-15, IL-21-expanded WT1 CTLs were alive at 18-30 months post-infusion. The addition of IL-21 to the expansion medium appears to have improved function and persistence of the infused CTLs. A confounding variable in this study is the fact that 5 of the 7 IL-2 only patients had detectable disease at the time of transplant compared with only one of the group that received the WT1 CTLs. Thus it is unclear whether the superior persistence of the IL-21-expanded CTLs was due to the less suppressive in vivo leukemic milieu in these patients. However the impressive result in the one patient with detectable disease encouraged further study. Another Phase I trial of allogeneic WT1-specific CTLs utilized pooled peptides encompassing the full WT1 protein to expand a broader range of WT1-epitope-specific clones, that were not HLA-A*0201-restricted. These WT1-specific CTLs were also able to mediate transient reduction or elimination of WT1-expressing cells in the absence of toxicities or GVHD87. Further refinement of this approach has seen studies utilizing CTLS specific for multiple LAA. Fourteen CML patients received prophylactic PR1, WT1 and/or BCR-ABL-specific CTLs that had been expanded in the presence of IL-2 and IL-761. No toxicity or GVHD was reported and 50% obtained molecular remission (median follow-up 45 months).
Table 4. Non-gene modified T cells for leukemia.
| NCT # (ACRONYM) | Disease Target | Cell Type | Age | Inclusion Criteria | Exclusion Criteria | Phase | Sponsor |
|---|---|---|---|---|---|---|---|
| NCT01627275 | Hematological malignancies | Naive T cell depleted DLI | ≥ 18 | HLA-matched allo-transplant; ≥ 60d post-transplant; KPS 50-100%; Donor engraftement ≥ 40%; Acute GvHD ≤ grade II; No extensive cGvHD; Corticosteroids ≤ 20 mg; Mycophenolate mofetil ≤2000mg/d Cyclosporine/tacrolimus ≤ therapeutic levels; No change in immunosuppressants 2 wks before DLI. | Pregnant or lactating; Major medical or psychiatric illnesses. | I | Duke University |
|
| |||||||
| NCT00675831 | Hematological malignancies | CD25+ T cell-depleted DLI | ≥ 18 | Relapse after HLA-A, -B, -C, and-DRBI matched allo HSCT; ≥ 2 months post-HSCT; Off immunosuppressants for GVHD 2 wks prior to DLI; Donor chimerism ≥ 20% 6 wks prior to DLI; ≤ 50% BM involvement (% cellularity) and ≤ LNs 6 wks prior to DLI; ECOG score 0-2; Prior stem cell donor medically fit to undergo leukapheresis. | Chronic phase relapsed CML; Prior DLI or other immunotherapy treatment ≤ 8 wks prior to enrollment; Chemotherapy ≤ 4 wks prior to enrollment; Clinically significant and active autoimmune disease in donor or patient; Acute or cGVHD; Active infection. | I | Dana-Farber Cancer Institute |
|
| |||||||
| NCT02203903 (RESOLVE) | Relapsed/Refractor y Hematopoietic Malignancies | Autologous or allogeneic WT1, NE, PR3, PRAME, MAGE-A3, MAGE-A4, NY-ESO, and survivin-specific CTLs | 6 m-80 y | Allo-HSCT pts with high risk for relapse or residual/recurrent dz OR relapsed/refractory dz (> 2 regimens with > M1 marrow or persistent HD) with anticipated allo- HSCT pre- and/or post-HSCT; ALL, AML, Ambiguous lineage leukemia or lymphoma, CML, MDS by flow cytometry, morphology, or cytogenetic evaluation within BM or extramedullary sites; HD by morphology, PET/CT uptake in site of previous dz in absence of other etiologies; KPS/LPS > 50; ANC > 500/μL;Bilirubin < 2.5 mg/dL; AST/ALT <5× ULN; Serum creatinine < 1.0 or 2× ULN (whichever is higher); pulse oximetry > 90%; On contraception during study; LVEF > 50% or LVSF > 27% if history of TBI (within 6 mths); Steroids ≤ 0.5 mg/kg/day; | Uncontrolled infection; HIV infection; GVHD > grade 2 or bronchiolitis obliterans syndrome, sclerotic GVHD, or serositis; Pregnant or lactating; Patients on ATG, or Campath or other immunosuppressive T cell mABs within 28 days of screening for enrollment | I | Children's Research Institute, Johns Hopkins University |
|
| |||||||
| NCT00052598 | Accelerated Phase CML, AML With Multilineage Dysplasia Following MDS, Adult AML With 11q23 (MLL) Abnormalities/Del(5q)/Inv(16)(p13;q22)/t(15;17)(q22;q12)/t(16;16)(p13;q22)/t(8;21)(q22;q22), Blastic Phase CML, Childhood CML, Childhood MDS, Recurrent Adult AML, Recurrent Childhood AML, Relapsing CML, Secondary AML, | Autologous and allogeneic proteinase 3 (Myeloblastin)-specific CTLs + IL-2 | Child Adult Snr | Pts undergoing alloHSCT for CML in accelerated/blast phase, AML beyond first remission, 1° refractory AML, tx-related AML any stage, acute leukemia any stage, antecedent dx of MDS/MPS (incl CML, polycythemia vera, essential thrombocytosis, and agnogenic myeloid metaplasia with my elofibrosis); Pts and donors HLA-A2+; Able to provide blood & BM; Eligibility for prophylactic treatment with CD8+ CTL after Transplant (Highest Risk Subgroup): PR3-specific CD8+ CTL generated & QC tested; > 5% morphologic blasts in BM or PB just prior to or at time of transplant; Posttransplant ANC > 500/mm̂3 7 d prior to CTL infusions; Pts on immsupp tx for GVHD eligible if not on corticosteroids or if dose can be tapered to ≤ 0.5 mg/kg/day prednisone; pt's symptoms must remain stable and unlikely to increase to stage III or IV aGVHD; or cGVHD unlikely to progress following change in immsupp tx, after monitoring period, as deemed by the pts' physician & the PI; Morphologic/flow cytometric/cytogenic/molecular evidence of recurrent/progressive dz posttransplant | Pts for whom PR3-specific CD8+ CTL clones have not been generated before dz relapse/progression post-transplant Pts whose malignant cells do not overexpress PR3, based on BM sample with > 50% blasts or on leukemia cells; KPS/LPS ≤ 30; Stage III or IV GVHD unresponsive to tx or requiring tx with anti-CD3 mAb, prednisone > 0.5 mg/kg/day (or equivalent), or other txs that ablate or inactivate T cells (eg anti-T cell mABs); Concurrent use of cyclosporine, FK506, or my cophenolate mofetil (MMF) not strictly an exclusion criterion, but attempts should be made to discontinue; Concurrent therapy with hydroxyurea or other agents that may interfere with the function or survival of infused CTL clones; Nonhematopoietic organ toxicity; graft rejection or failure | I/II | Fred Hutchinson Cancer Research Center, NCI |
|
| |||||||
| NCT00052520 | Advanced MDS, CML, AML or ALL | Autologous and allogeneic CD8+ WT1-specific CTLs + IL-2 | Child Adult Snr |
Eligibility for Enrollment: a.i) Pre-transplant: Pts undergoing alloHSCT for RAEB, RAEB-t, CML beyond chronic phase, AML beyond 1st remission, BCR-ABL+ ALL any stage, any ALL beyond 1st remission, 1° refractory AML or ALL, tx-related AML any stage, or acute leukemia any stage in pt with antecedent dx of MDS/MPS; ii) Post-transplant: Pts relapsed after transplant (morphologic, flow cytometric, cytogenetic and molecular) may undergo tx if dz control possible while awaiting generation of study tx b. Pts and donors must both express an HLA- allele from which it is possible to generate WT1-specific clones c. Pts must be able to provide blood and BM Eligibility for Prophylactic Tx with CD8+ CTL After Transplant (Highest Risk Subgroup): a. > 5% blasts in BM or PB just prior to/at time of transplant b. post-transplant ANC > 500/mm̂3) at least 7d prior to CTL infusions c. Pts on immsupp tx for GVHD eligible if not on corticosteroids or if dose can be tapered to ≤ 0.5 mg/kg/day prednisone; pt's symptoms must remain stable and unlikely to increase to stage III or IV aGVHD; or cGVHD unlikely to progress following change in immsupp tx, after monitoring period, as deemed by the pts' physician & the PI; Eligibility for Treatment with CD8+ CTL at Time of Relapse after Transplant (All Others): a. Morphologic/flow cytometric/cytogenetic/molecular evidence of recurrent dz post- transplant b. Pts on immsupp tx for GVHD eligible if not on corticosteroids or if dose can be tapered to ≤ 0.5 mg/kg/day prednisone; pt's symptoms must remain stable and unlikely to increase to stage III or IV aGVHD; or cGVHD unlikely to progress following change in immsupp tx, after monitoring period, as deemed by the pts' physician & the PI; DONOR: undergo separate leukapheresis for PBMC ≥ 2 weeks before/after stem cell mobilization and harvest if possible; if separate leukapheresis not possible, portion of PBMC from PB stem cells may be used to generate WT1-specific CTL clones; ≥18 yrs |
Pts for whom CD8+ WT1-specific CTL clones have not been generated in time for planned infusion; Pts whose malignant cells do not over-express WT-1; KPS/LPS =< 30% Current stage III or IV GVHD unresponsive to tx or requiring tx with anti-CD3 mAb, prednisone > 0.5 mg/kg/day (or equivalent), or other txs resulting in ablation/inactivation of T cells (eg anti-T cell mABs); concurrent use of cyclosporine, FK506, or MMF not strictly an exclusion criteria, butattempts should be made to discontinue if possible; concurrent tx with hydroxyurea or other agents that interfere with function or survival of infused CTL clones; preexisting nonhematopoietic organ toxicity deemed by PI to place pt at unacceptable risk; graft rejection or failure DONOR: Medical conditions precluding either leukapheresis or blood donation: Inadequate age or weight Active infection, with or without antibiotic treatment Recent hepatitis exposure, hepatitis A or B antigenemia, or hep C antibody + Pregnancy or nursing; HIV or human T-lymphotropic virus (HTLV) infection Severe cardiovascular disease (e.g., uncontrolled hypertension, recent myocardial infarction [MI], or unstable angina) |
I/II | Fred Hutchinson Cancer Research Center, NCI |
|
| |||||||
| NCT00107354 | Leukemia, MDS | Autologous and allogeneic minor histocompatibility antigen-Specific-CTLs | ≥ 14 yrs child adult Snr | AlloHSCT from MHC-identical donor for 1° refractory AML or ALL, AML or ALL beyond 1st remission, Tx-related AML any stage, BCR- ABL+ ALL, RAEB, RAEBt Morphologic, flow cytometric/cytogenetic/molecular evidence of recurrent dz pos-t transplant No grade III or IV aGVHD at post-transplant relapse; No extensive cGVHD at post-transplant relapse KPS 60-100% at post-transplant relapse; No preexisting major nonhematopoietic organ toxicity ≥ grade 3 Concurrent immsupp steroid tx for GVHD allowed provided: Able to taper steroid dose to < 0.5 mg/kg/day No increase of > 1 grade in aGVHD OR progression of cGVHD within 14 d after dose change | I | Fred Hutchinson Cancer Research Center, NCI | |
|
| |||||||
| NCT02895412 (INTACT-WT1) | Leukemia, Myelocytic, Acute | Allogeneic WT1 and multiple pathogen specific-CTLs (cytomegalovirus (CMV), Adenovirus (Adv), Epstein Barr virus (EBV), Varicella-Zoster virus (VZV), Influenza, BK virus (BKV), and fungal infections) | Child Adult Snr | my eloablative or non-myeloablative allo transplant from HLA (A, B and DR) identical or 1-3 antigen mismatched donor; Transplant performed for AML; Leukemia blasts WT1+ by ELISA (> 50 copies/104ABL in BM or >50 copies/104ABL in PB); PB HSCT recipients; Adequate hepatic & renal function (< 3 × ULN AST, ALT, < 2 × ULN total bilirubin, serum creatinine); Estimated life expectancy ≥ 12 m | Anti-lymphocyte globulin (ALG, ATG, Campath or other broad spectrum lymphocyte antibody) in 4 wks immediately prior to infusion or planned within 4 wks after infusion; ≥ Grade II GVHD within 1 wk prior to infusion; Prednisone/methyl-prednisone > 1 mg/kg (or equivalent) within 72 hours prior to cell infusion; Prior allo HSCT; Privately insured in or outpatients | I | University of Sydney |
|
| |||||||
| NCT02074657 (LANK-2) | Relapsed/Refractor y Paediatric Acute Leukaemia | Allogeneic activated and expanded natural killer cells | Child Adult ≤23y | AML in second relapse, post-transplant relapse or refractory, or AML relapsed or refractory; Lansky index > 60%; Mild (<2) functional organ alteration (hepatic, renal, respiratory) per (NCICTCAE v 4; LVEF > 39%; compatible haploidentical donor (father or mother or sibling) | History of poor therapeutic compliance; Patients not valid after psycho-social evaluation; HIV+ serology | II | Hospital Universitario La Paz |
|
| |||||||
| NCT00620633 | Leukemia, MDS | Allogeneic WT1-specific CTLs | Child Adult Snr All ages | WT1+ leukemic or MDS blasts (IRS 4-12 considered positive); Leukemia relapse/persistent MRD following allo-HSCT; Pts with expected of risk of relapse post-transplant of >30% once MRD confirmed or relapse occurs; ALL, AML or MDS refractory to 1° induction therapy; ALL or AML any stage later than 1° relapse; CML ≥ 2° chronic phase after chemotx; CML in persistent accelerated phase or blast crisis; High risk MDS (RAEB and RAEB+) that failed to respond or recurred following induction chemotx; Donor must consent to 2 vol leukapheresis or whole blood donations obtained at one phlebotomy, approx 250 ml from which WT-1-specific T cells for adoptive transfer will be generated; KPS or LPS ≥ 40; Adequate BM, renal and hepatic function at time of recieving WT1-specific T cells; ANC ≥ 1000/mm3 +/- G-CSF (may be waived if pt relapsed or counts not yet recovered from chemotx); Platelets ≥20,000/mm3 (may be waived if pt relapsed or counts not yet recovered from chemotx); Creatinine ≤2.0mg/dL; ALT, AST <3.0 × institutional ULN; bilirubin < 2.5 × institutional ULN; Pts with CNS relapse may be treated however T cells should not be administered until ≥ 24 h after intrathecal chemotx | Active (grade 2-4) aGVHD, cGVHD or overt autoimmune dz (e.g. hemolytic anemia) requiring high dose glucocorticosteroid (>0.5 mg/kg/day prednisone or equivalent); other conditions not related to leukemic relapse (e.g. v eno-occlusive disease or uncontrolled bacterial, viral or invasive fungal infection) which are also life-threatening and which would preclude evaluation of the effects of a T cell infusion; pregnancy; DONOR ELIGIBILITY: WT1-specific T cell donor = HSCT donor; EBV+ T cells will also be generated from EBV+ donors and infused along with WT1 cells and growth and persistence compared; Initial donation of 25ml blood prior to HSCT donation, to generate EBV-APCs; Leukapheresis/whole blood for WT1-specific T cells 2 wks post transplant donation; Consent to use blood for virus-specific T cells for recipient if required Donor exclusion: HIV+ Other uncontrolled infection; Anemia (Hgb ≤10 gm/dl) persisting since original transplant donation; MI or stroke since HSCT donation; |
I | Memorial Sloan Kettering Cancer Center |
|
| |||||||
| NCT00460629 | CML | Allogeneic BCR-ABL, PR1, and WT1-specific CTLs | Adult 18-60y | Ph+ CML HLA A0201, 0301, 1101, B0801 BCR-ABL b3a2+ No significant comorbidities Resistance to or intolerance of imatinib | HIV+ Blast crisis | I/II | University Hospital Carl Gustav Carus |
Allo: allogeneic; ALL: Acute lymphoblastic Leukemia; AML: Acute Myeloid Leukemia; CML: Chronic Myeloid leukemia; MDS: Myeloid Dysplastic Syndrome; MPS: Myeloproliferative Syndrome; CTL: Cytotoxic T Lymphocyte; NCI: National Cancer Institute; KPS: Karnofsky Performance Status; LPS: Lansky Play Score; BM: bone marrow; LN: lymph nodes; ECOG: ECOG (Eastern Cooperative Oncology Group) Performance Status Score; aGVHD: acute GVHD; cGVHD: chronic GVHD; ANC: absolute neutrophil count; mABs: monoclonal antibodies; Snr: Senior; Pts: patients; dz: disease; dx: diagnosis; tx: treatment or therapy; PB: peripheral blood; immsupp: immunosuppressive; RAEB: refractory anemia with excess blasts; RAEB-t: refractory anemia with excess blasts in transformation; HLA: human leukocyte antigen; ELSA: European LeukaemiaNet standardised assay; UL: Upper limit; NCI CTCAE v4: National Cancer Institute Common Terminology Criteria for Adverse Events; LVEF: Left ventricular ejection fraction; IRS: German Immunoreactive Score; ULN: upper limit of normal; MI: myocardial infarction; EBVBLCL: EBV+ B cell line; 1°: primary; APCs: antigen-presenting cells.
Targeting multiple TAA
To circumvent the more limited applicability of HLA-restricted approaches, strategies to expand cell products with specificity for multiple TAAs, expressed by a range of hematological malignancies have been developed. Both MHC-I and MHC-II-restricted epitopes are included in the composite peptide pools, thus both CD8+ and CD4+ T cells can be primed. Trial NCT02203903 is an important step toward multi-specific T cells for post-allo-HSCT leukemia patients, an approach that could ultimately be used in a variety of hematological malignancies without the need to first identify specific tumor antigens in the patient. Targeting multiple antigens also minimizes the possibility of tumor escape since selection pressure is not applied to a single antigen.
Lymphoma antigen-specific T cells
Clinical trials for T cell-mediated lymphoma have revealed that potent immune responses can be generated against lymphoma-associated viral antigens, but that poorly immunogenic tumor antigens constitute more difficult targets. T cells specific for PRAME, SSX2, MAGE-A4, NY-ESO1 and survivin can be generated ex vivo from HL and NHL patients and their effectiveness is being assessed in two clinical trials (Table 5): (NCT01333046 (TACTAL); and NCT02203903 (RESOLVE). Table 5 summarizes lymphoma trial data.
Table 5. T cells for lymphoma.
| NCT # (ACRONYM) | Disease Target | Cell Type | Age | Inclusion Criteria | Exclusion Criteria | Phase | Sponsor |
|---|---|---|---|---|---|---|---|
| NCT00002663 | Lymphoma, leukemia, other adult and pediatric solid tumors | Allogeneic EBV-specific CTLs | Child Adult Snr | EBV+ LPD, lymphoma, or other EBV-associated malignancy OR Severely immunocompromised pts at high risk for EBV LPD(> 500 copies/ml EBV DNA in PB) following allo BMT; or allo organ transplant; or HIV-induced immunodeficiency; or congenital immune deficit; or anti-neoplastic or immune suppression tx; or non-immune-suppressed pts with EBV+ lymphoma, EBV+ HD, EBV+ NHL, EBV+ nasopharyngeal carcinoma, EBV+ hemophagocytic lymphohistiocytosis, or EBV+ leiomyosarcoma; Pregnant women | Moribund pts with heart, kidney, liver, lung, or neurologic dysfunction not related to lymphoma, who are unlikely to survive the 6-8 weeks required for in vitro generation and expansion of EBV-specific T cells to be infused and the subsequent 3 weeks required assess effects of infusions | I/II | Memorial Sloan Kettering Cancer Centre and NCI |
|
| |||||||
| NCT01948180 (CITADEL) | EBV+ extranodal NK/T cell Lymphoma | Autologous EBV-specific CTLs (CMD-003) | ≥ 18y Adult Snr | SCREENING PHASE 1. extranodal NK/T lymphoma, EBV+ by EBER or LMP1 immunostaining 2. Active dz: relapse/progression,1st/2nd relapse following at least one cycle ABC OR initial dz or 1st/2nd relapse and unable to tolerate one full cycle ABC OR High-risk dz (stage III/IV, KPI grps 3-4 or IPI intermediate-high) prior to second CR regardless of previous chemotx. Weigh ≥ 35 kg; ECOG performance score 0-2, inclusively; Negative β-hCG women of childbearing potential; TREATMENT PHASE Relapse/progression following at least one prior cycle of ABC; Active dz: based on any one of the following at baseline or within 2 wks prior to baseline: Imaging; clinical sign(s): lymphomatous skin lesions, organ dysfunction or organomegaly not attributable to other causes; or other clinical sign(s); PB/plasma ENV DNA; Chemotx completed ≥ 2 wks prior to 1st study dose; Recovery from acute hematological, hepatic and renal chemotherapy-related toxicities as defined by ≤ Grade 1 according to NCI CTCAE v4.0; Life expectancy ≥ 8 weeks. | SCREENING PHASE CNS lymphoma; NK cell leukemia; Hemophagocytic lymphohistiocytosis; HIV+, hep B+, hep C+, syphilis+, HTLV+; Systemic corticosteroids >0.5 mg/kg/day within 10 d prior to obtaining 200 mL PB starting material; Pregnant or lactating; Active second malignancy; Prior alloHSCT or solid organ transplant; ARD:Progression during initial ABC & up to 3 mth after end of initial ABC; OR failure to achieve at least PR with initial ABC. ALC <400/μL; Any previous autologous EBV-specific T cell treatment; Any uncontrolled systemic infection; Third or greater relapse; TREATMENT PHASE Use of any investigational agents within prior 4 wks; Radiotherapy within prior 3 wks; Major surgery within prior 2 wks; Systemic corticosteroids within 24 h prior to study drug administration; Hepatic dysfunction: serum bilirubin >3 × ULN or ALT >5 times ULN or AST >5 times ULN | II | Cell Medica Ltd |
|
| |||||||
| NCT00779337 | Lymphoma | Autologous AdE1-Latent Membrane Protein-specific CTLs | ≥ 18y Adult Snr | EBV+ lymphoma by in situ hybridization or equivalent (excluding Burkitts Lymphoma). ECOG performance status 1, 2 or 3 Life expectancy ≥ 6 mths; Measurable disease: relapsing/PR/refractory/PD dz by clinical examination or radiography (incl CT scans/functional imaging), or plasma EBV viral load; No chemotx/radiotx &/or antibody tx ≥ 2 wks prior to anticipated first infusion. | EBV negative tumour; Malignant cells in PB by flow cytometry or morphology; infection, positive serology for HIV I&II, H TLV1 or syphilis; Negative serology for EBV; Psychiatric, addictive or any condition which may compromise trial participation Serious infection within 28 days that has not adequately responded to tx; Pregnancy/not using contraception; Serology (within 3 mths of CTL release date) indicating active HBV or HCV | I | Queensland Institute of Medical Research, The Atlantic Philanthropies, Australian Department of Industry, Tourism and Resources, British Society Health and Medical Research Council, Australia for Haematology, National |
|
| |||||||
| NCT00005606 | Lymphoma, Leukemia, Multiple Myeloma Plasma Cell Neoplasm | Allogeneic EBV-specific CTLs | Child Adult Snr (≤ 65 yrs) | Pts who have received or will receive a solid organ transplant or T cell depleted BMT EBV DNA+and seroneg OR EBV sero+ Matched or 1 HLA mismatched sib donor HIV neg Hep B surface antigen neg Hep C antibody neg No prior 1° malignancy within past 5 yrs in donor except previously resected skin cancer | None specified | II | Northwestern University, NCI |
|
| |||||||
| NCT01636388 | HL | Allogeneic LMP-specific CTLs | Child Adult (≤ 45 yrs) | Off other investigational tx for 1 mth prior to entry in this study Adequate renal function: Serum creatinine <2.0 × ULN, or Creatinine clearance or radioisotope GFR > 40 ml/min/m2 or >60 ml/min/1.73 m2 or equivalent GFR; Adequate liver function: bilirubin <2.0 × ULN; and SGOT (AST) or SGPT (ALT) <5.0 × ULN; Adequate cardiac function: Shortening fraction of >27% by ECG; HL with: 1° induction failure (failure to achieve initial CR) and/or 1°refractory dz. 1st relapse. Early relapse (within 12 mth off tx) (excl those who received no tx or radiation tx only for initial tx); Late relapse (> 12 mths off tx). Only pts with recurrent Stage III or IV dz &/or B symptoms at relapse (all other late relapses excluded); 2nd relapse; 3rd relapse. History of prior ablative auto HSCT or ineligible for ablative auto HSCT or ≥25% RD after at least two reinduction chemo cycles. EBV sero+ IgG HLA matched family/MUD HLA matched family donor (6/6 or 5/6) MUD (7/8 or 8/8). Ideally have tissue from original dx specimen &/or relapse reviewed centrally for confirmation of HL. If no specimen avail, pathology documenting EBV+ status. Immunophenotyping to confirm dx will be performed. In situ hybridization for EBV (LMP1, and/or EBER) will be performed. All central morphologic, IHC/insitu hybridization performed in Sherrie Perkins & Rodney Miles lab at U of Utah. | HD with ≥ 4th CR, PR, and/or SD; Rapid PD unresponsive to reinduction chemo, radio, or immunotx; EBV neg HL; Patients with no eligible donor; Pregnancy | II | New York Medical College, Children's Research Institute Baylor College of Medicine M.D. Anderson Cancer Center City of Hope Medical Center Johns Hopkins University Ohio State University University of Utah University of Michigan |
|
| |||||||
| NCT01956084 | HL, NHL, SCAEBV Lymphoepithelioma, Leiomyosarcoma | Allogeneic LMP1/2-specific CTLs | Child Adult Snr | EBV+ HL/NHL or EBV-associated-T/NK-LPD or lymphoepithelioma/leiomyosarcoma or severe Chronic EBV and in remission (group A) or with detectable dz (group B) after allo SCT Life expectancy > 6 weeks Tumor tissue EBV+; KPS/LPS > 50; Donor HIV neg; ≥ 50% donor chimerism in either PB or BM; Bilirubin <2× ULN, AST <5× ULN, and Hgb >8.0; Creatinine <2× ULN; Off other investigational tx 1 mth prior to entry in this study; | HIV+ GVHD > Grade II Pregnancy | I | Children's Research Institute |
|
| |||||||
| NCT02057445 | HL, NHL, Lymphoproliferative disorder, | Allogeneic LMP-specific CTLs | Child Adult Snr (≥ 1yr) | Off other investigational tx 1 mth prior to entry in this study; Adequate renal function: Serum creatinine <2.0 × Must meet criteria as per 21 CFR 1271; ULN, or Creatinine clearance or radioisotope GFR > 40 ml/min/m2 or >60 ml/min/1.73 m2 or equivalent GFR; Adequate liver function: Total bilirubin <2.0 × ULN; and SGOT (AST) or SGPT (ALT) <5.0 × ULN; Adequate pulmonary function: Pulse oximetry >94%. LPS (< 16yr) or KPS (> 16 yrs) ≥ 50%; Life expenctancy ≥ 6 weeks; Negative urine pregnancy test; Clinical status at enrollment allows tapering steroids to < 0.5mg/kg/day prednisone at time of tx. HL/NHL LPD, SCAEBV: > 4000 genomes per μg PBMC DNA, &/or biopsy tissue EBV+ At dx unable to receive conventional chemotx or in 1st relapse AND not a candidate for HSCT PR after conventional tx. Refractory to conventional tx. In 2nd or subsequent relapse. RD after auto, syngeneic or allo HSCT; Ideally have tissue from original dx specimen &/or relapse reviewed centrally for confirmation of HL. If no specimen avail, pathology documenting EBV+ status. Immunophenotyping to confirm dx will be performed. In situ hybridization for EBV (LMP1, and/or EBER) will be performed. All central morphologic, IHC/insitu hybridization performed in Sherrie Perkins & Rodney Miles lab at U of Utah. DONOR ELIGIBILITY for LMP-CTL 3rd Party Banking: Adequate hematopoietic function: ANC > 1000/mm3, hemoglobin > 10 g/dl, platelet count >50,000/mm3 and EBV IgG sero+; ≥ 12 kg or 24 pounds; <18 years: max 3cc/kg blood in 8 wk period; min 60 cc PB × 2; For stem cell collection, PB for LMP specific CTLs collected prior to stem cell collection, no specific day specification; Must meet criteria as per 21 CFR 1271; |
Currently receiving any investigational agents or have received any tumor vaccines within previous 4 wks; Active acute grade III-IV GVHD; Severe refractory intercurrent infection other than EBV; Received alemtuzumab or other anti-Tcell antibody within 28 days. HIV sero+; Pregnancy or lactation; PTLD post solid organ transplantation eligible for the COG PTLD LMP/CTL protocol | I | New York Medical College, Children's Research Institute, Baylor College of Medicine, M.D. Anderson Cancer Center, University of Michigan, University of Utah, City of Hope Medical Center, Ohio University, Johns Hopkins University |
|
| |||||||
| NCT01498484 | NHL, EBV infection | Allogeneic EBV-specific CTLs | Child Adult Snr | EBV+ LPD, lymphoma or other EBV-assocd malignancy; OR Current or prior PB EBV DNA > 500 copies/ml by qRT PCR; OR EBV DNA > 500 copies/ml in pts treated for EBV-LPD with chemotx &/or rituximab with no clinically or radiologically evaluable dz but high risk of recurrence; Availability of EBV-specific T cells for adoptive immune cell therapy from donor; 3rd party EBV-CTLs will be: 1) matched for ≥ 2 HLA antigens & 2) restricted by an allele shared with the EBV+ malignancy (if known), or with the donor in HSCT recipients, or patient in organ transplant or immunodeficient patients; KPS/LPS ≥ 20; Life expectancy ≥ 6 weeks; Adequate BM, heart, lung, liver & kidney function at tx initiation with EBV-specificT cells, including: ANC ≥ 1,000/μL +/- GCSF support; Platelets ≥ 20,000/μL; Creatinine ≤ 2.0mg/dl; ALT, AST < 3.0× ULN; Bilirubin < 2.5× ULN; Stable BP & circulation not requiring pressor support; Adequate cardiac function (EKG &/or ECG within 30 days prior to treatment); Abnormalities of specific organs not grounds for exclusion if arising from EBV+ malignancy or its tx (e.g. renal allograft recipient with EBV LPD on dialysis due allograft rejection after immune suppression stopped); EBV+ lymphoma or LPD following allo HSCT; or allo organ transplant, or due to HIV-induced profound acquired immunodeficiency; or other EBV-assocd malignancy due congenital immune deficit or sequela of antineoplastic or immunosuppressive tx; EBV-assocd malignancies without pre-existing immune deficiency, including: EBV+ HD & NHL, EBV+ nasopharyngeal carcinoma, EBV+ hemophagocytic lymphohistiocytosis, or EBV+ leiomyosarcoma. | Active (grade 2-4) aGVHD, cGVHD or overt autoimmune disease (e.g. hemolytic anemia) requiring glucocorticosteroid (>0.5 mg/kg/day prednisone or equiv); Pregnancy; Severe comorbidities, unrelated to EBV-associated malignancy, that would be expected to preclude their survival for the 6 wks required to assess response of T cell therapy Patients eligible for MSK protocol #16-803 (EBV-CTL-201) | II | Atara Biotherapeutics, Memorial Sloan Kettering Cancer Centre |
|
| |||||||
| NCT01447056 | HL, NHL, Lymphoproliferative disease, Nasopharyngeal Carcinoma, Leiomyosarcoma, SCAEBV | Allogeneic LMP-specific CTLs | Child Adult Snr |
SCREENING EBV+ HL, NHL, LPD, Nasopharyngeal carcinoma, Leiomyosarcoma, SCAEBV, EBV load in plasma or PBMC > 4000 genomes per ug PBMC DNA &/or biopsy tissue EBV+; KPS/LPS ≥ 50%; TREATMENT EBV+ HL, NHL, LPD, Nasopharyngeal carcinoma, Leiomyosarcoma, SCAEBV, EBV load in plasma/PBMC > 4000 genomes per ug PBMC DNA &/or biopsy tissue EBV+; At dx or in 1st relapse AND unable to receive chemotx OR in 2nd or subsequent relapse OR with RD after auto, syngeneic or allo HSCT; Life expectancy ≥ 6 wks; Tumor tissue EBV+; KPS/LPS score ≥ 50%; Bilirubin < 3 × ULN; AST < 5 × ULN; Hgb > 8.0 g/dL; Serum creatinine < 3 less × ULN; Pulse oximetry > 90%; If post allo HSCT, patient ≥50% donor chimerism in either PB or BM; Clinical status at enrollment allows tapering steroids to < 0.5 mg/kg/day prednisone at tx; Willing to utilize birth control during study and 3 mths after study conclusion. |
SCREENING HIV+; TREATMENT Currently receiving any investigational agents or have received any tumor vaccines within previous 4 wks; Active acute grade III-IV GVHD; Severe intercurrent infection; Alemtuzumab or other anti-T-cell antibody within 28 days; HIV seropositivity; Pregnancy or lactation; Tumor in location where enlargement could cause airway obstruction |
I | Baylor College of Medicine |
|
| |||||||
| NCT01555892 (GRALE) | HL, NHL, T/NK lymphoproliferative Disease Lymphoma | Autologous or syngeneic donor LMP1/2, EBNA-1 and BARF-specific CTLs | Child Adult Snr | EBV+ HL/NHL or EBV-assocd T/NK-LPD or SCAEBV ≥ 12 kg In 2nd or subsequent relapse (or 1st relapse or with active dz if immunosuppressive chemotx contraindicated; or multiply relapsed patients in remission who have a high risk of relapse); OR any patient with 1° dz or in 1st remission if immunosuppressive chemotx contraindicated, e.g. HD after solid organ transplant or if Lymphoma is a 2nd malignancy e.g. Richter's transformation of CLL. (Group A) OR In remission or MRD after auto or syngeneic SCT (Group B); Life expectancy ≥ 6 wks; Bilirubin ≤ 3× ULN; AST ≤ 5× ULN; Hgb > 8.0; creatinine ≤2× ULN; Pulse oximetry > 90%; Off other investigational therapy 4 wks prior to entry in this study. KPS/LPS ≥ 50; Willing to utilize birth control during study and 3 mths after study conclusion. | Active infection with HIV, HTLV, HBV, HCV; Pregnant or lactating; Severe intercurrent infection; Current systemic corticosteroids > 0.5 mg/kg/day | I | Baylor College of Medicine, NCI |
|
| |||||||
| NCT00062868 | HL, NHL, Lymphoepithelioma, Leiomyosarcoma | Autologous or allogeneic LMP1/2-specific CTLs | Child Adult Snr | In 2nd or subsequent relapse (or 1st relapse or with active dz if immunosuppressive chemotx contraindicated; or multiply relapsed patients in remission who have a high risk of relapse); OR any patient with 1° dz or in 1st remission if immunosuppressive chemotx contraindicated, e.g. HD after solid organ transplant or if Lymphoma is a 2nd malignancy e.g. Richter's transformation of CLL. (Group A) OR In remission or MRD after autol or syngeneic SCT. (Group B); OR detectable dz after alloSCT (Group C); Life expectancy ≥ 6 wks; KPS/LPS ≥ 50; ≥ 50% donor chimerism in either PB or BM if post alloSCT; Bilirubin ≤ 3× ULN; AST ≤ 5× ULN; Hgb > 8.0; creatinine ≤2× ULN; Off other investigational tx 1 mth prior to entry into study | Severe intercurrent infection; HIV+ donors or HIV+ pts if autologous product to be used > Grade II GVHD Pregnancy | I | Baylor College of Medicine |
|
| |||||||
| NCT02287311 (MABEL) | HL, NHL, SCAEBV, T/NK-lymphoproliferative Disease | LMP, BARF-1 and EBNA1- specific | Child Adult Snr | EBV+ HL; EBV+ NHL; EBV-assocd T/NK-LPD, or SCAEBV (>4000 EBV genomes per ug PBMC DNA) &/or biopsy tissue EBV+ AND in 1st or subsequent relapse (Group A); with active dz persisting despite tx (Group B); with active dz if immsupp chemotx contraindicated e.g. pts who develop HD after solid organ transplant or if lymphoma = 2nd malignancy e.g.Richter's transformation of CLL (Group C); ≥ 12kg; | Pregnant or lactating; Severe intercurrent infection; Current systemic corticosteroids > 0.5 mg/kg/day; Receiving ATG, Campath, or other immunosuppressive T cell monoclonal antibodies within 30 days. | I | Baylor College of Medicine |
|
| |||||||
| NCT00002663 | Lymphoma, Leukemia, Unspecified Adult Solid Tumor, Protocol Specific Unspecified Childhood Solid Tumor, Protocol Specific | Allogeneic EBV-specific CTLs | Child Adult Snr | EBV+ LPD, lymphoma, or other EBV-assocd malignancy OR Severely immunocompromised pts with PB levels EBV DNA > 500 copies/ml at high risk for EBV LPD; Incl pts with/at risk for EBV lymphomas or LPDpost allo BMT or allo organ transplant; AIDS pts w/EBV lymphomas/LPD due HIV-induced acquired immunodeficiency; EBV lymphomas/LPD due congenital imunodeficiencies or acquired as asequela of anti-neoplastic or immsupp tx; EBV+ HD & NHD, EBV+ nasopharyngeal carcinoma, EBV+ hemophagocytic lymphohistiocytosis, or EBV+ leiomyosarcoma in immune compentent pts; Pregnancy not contraindicated for infusions of EBV-specific T cells | Moribund patients who due heart, kidney, liver, lung, or neurologic dysfunction unrelated to lymphoma, are unlikely to survive the 6-8 wks required for in vitro generation and expansion of EBV-specific T cells to be used for tx & the subsequent 3 wks required to assess effects of infusions of EBV-specific T cells. | I/II | Atara Biotherapeutics, Memorial Sloan Kettering Cancer Centre, NCI |
|
| |||||||
| NCT02973113 (PREVALE) | HL, NHL, Lymphoproliferative Disorders, EBV-related Lymphoma, EBV-Related PTLD, EBV-Related NHL EBV-Related HL | Autologous EBV-specific CTLs+nivolumab (anti-PD-1 mAB) | Child Adult Snr | PROCUREMENT EBV+ HL or NHL or EBV-assoc T/NK/B cell LPD. 1st 3 pts will be adults. Pts <18 years of age eligible if 1st 3 pts don't experience DLT considered to be primarily related to EB-VST or Nivolumab; Relapsed or refractory lymphoma who failed or are ineligible for autoHSCT; ≥12kg; Life expectancy ≥ 6 weeks. TREATMENT As above AND HL in 2nd relapse or 1st relapse & refractory to at least 2 lines salvage chemotx incl Brentuximab Vedotin or 1° refractory dz after at least 2 lines tx OR NHL in 1st relapse and/or refractory to at least one salvage chemotx or with 1° refractory disease after at least 2 lines of tx or in 2nd or subsequent relapse OR T/NK- or B LPD in 1st relapse &/or refractory to at least one salvage chemotx or with 1° refractory dz after at least 2 lines of tx or in 2nd or subsequent relapse; Life expectancy > 6 weeks; Bilirubin ≤ 3× ULN; AST ≤ 5× ULN; Hgb > 8.0; creatinine ≤2× ULN; pulse oximetry > 90%; Off other investigational therapy 4 wks prior to entry in this study; KPS/LPS > 60; Recovered from acute toxic effects of chemotx at ≥ 1 week before entering study; Willing to utilize birth control during study and 6 mths after study conclusion. | PROCUREMENT Active HIV, HTLV, HBV, HCV infection; Hx of solid organ transplant; TREATMENT Pregnant or lactating; Severe active intercurrent infection; Current systemic corticosteroids >0.5 mg/kg/day; Current investigational agents or radiotx within 4 wks prior to entering study; CNS involvement; Hx of allergic rx n attributed to nivolumab or other checkpoint inhibitors; Uncontrolled autoimmune dz needing systemic steroids or steroid sparing agents except for hypothyroidism or type I diabetes. Uncontrolled intercurrent illness including, but not limited to, ongoing or active infection, symptomatic congestive heart failure, unstable angina pectoris, cardiac arrhy thmia, or psychiatric illness/social situations that would limit compliance with study requirements. Lipase > 70U/ml | I | Baylor College of Medicine |
|
| |||||||
| NCT01333046 (TACTAL) | HL, NHL | 5-azacytadine+ autologous NY-ESO-1, MAGEA4, PRAME, Survivin and SSX-specific CTLs | Adult Snr ≥ 18y | PROCUREMENT HL or NHL Life expectancy ≥ 6 weeks; Hgb > 8.0 TREATMENT: Group A: ≥ 18 years old with active dz: in 2nd or subsequent relapse; in 1st relapse for indolent lymphoma after first-line tx for relapse; or 1st relapse if immsupp chemotx contraindicated; 1° refractory dz or persistent dz after 1st-line tx of relapse; multiply relapsed patients in remission at high risk of relapse; lymphoma is a 2nd malignancy e.g. Richters transformation of CLL after failing front line tx. Group B: ≥ 18 years old after auto or syngeneic SCT (as adjuvant tx) Group C: 5-azacytidine plus multiTAA-T cells. ≥18 years old with active dz in: 2nd or subsequent relapse; 1st relapse for indolent lymphoma after 1st line tx for relapse; 1st relapse if immsupp chemotx contraindicated; with 1° refractory dz or PD after 1st line tx of relapse; or lymphoma as 2nd malignancy e.g. Richters transformation of CLL after failing front line therapy GROUP D: ≤18 yrs old with active disease in: 2nd or subsequent relapse; 1st relapse for indolent lymphoma after 1st line tx for relapse; 1st relapse if immsupp chemotx contraindicated; with 1°refractory dz or persistent dz after 1st line tx of relapse; lymphoma as 2nd malignancy e.g. Richters transformation of CLL after failing front line tx; Life expectancy ≥ 6 weeks; Pulse oximetry ≥95 percent having previously received radiation tx; KPS/LPS > 50; Bilirubin ≤ 2× ULN; AST ≤ 3× ULN; Hgb > 8.0; platelets > 25,000 (Group C only); Creatinine ≤ 2× ULN; Off other investigational tx for 1 mth prior to entry in this study; Off conventional tx ≥ 1 wk prior to entry in this study, including rituximab; Females of child-bearing potential: at least 2 forms contraception unless hx of hysterectomy or tubal ligation; | PROCUREMENT: Severe intercurrent infection; Active HIV infection; Systemic corticosteroids. TREATMENT: Severe intercurrent infection; Systemic corticosteroids; Pregnant or breastfeeding; Abnormal coagulation parameters (PT > 15 s, PTT > 40 s &/or INR > 1.5) Significant active cardiac dz within previous 6 mths incl: NYHA class 4 CHF; Unstable angina; MI; Active HIV or hepatitis B or C infection defined PB reactive for Hep B, C and/or HIV confirmed with qPCR; Known or suspected hypersensitivity to azacitidine or mannitol; Advanced malignant hepatic tumors | I | Baylor College of Medicine, NCI |
|
| |||||||
| NCT02203903 (RESOLVE) | Relapsed/Refractor y Hematopoietic Malignancies | Autologous or allogeneic WT1, NE, PR3, PRAME, MAGE-A3, MAGE-A4, NY-ESO, and survivin-specific CTLs | Child Adult 6mo-65y | PROCUREMENT Allo-HSCT with high risk for relapse or residual/recurrent dz OR relapsed/refractory dz (> 2 regimens with > M1 marrow or persistent HD) with anticipated allo-HSCT pre- and/or post-HSCT; ALL, AML, Ambiguous lineage leukemia or lymphoma, CML, MDS: Evidence of active leukemia or lymphoma by flow cytometry/morphology/cytogenetic evaluation in BM or extramedullary sites; HD by morphology, PET/CT uptake in site of previous dz in absence of other etiologies; KPS/LPS > 50; ANC > 500/μL +/- GCSF; Bilirubin < 2.5 mg/dL, AST/ALT <5× ULN, Serum creatinine < 1.0 or 2× ULN (whichever is higher); Pulse oximetry > 90%; LVEF > 50% or LVSF > 27% if hx of TBI; Agree to contraception during study protocol participation (age appropriate) TREATMENT Steroids < 0.5 mg/kg/day prednisone or equivalent KPS/LPS > 50 Pulse oximetry > 90% | PROCUREMENT Uncontrolled infections; HIV infection; Current GVHD > grade 2 or bronchiolitis obliterans syndrome, sclerotic GVHD, or serositis; Pregnancy or lactating TREATMENT Receiving ATG, or Campath or other immsupp T cell mABs within 28 days of screening for enrollment. Uncontrolled infections Acute GVHD > grade 2 or cGVHD manifestations: bronchiolitis obliterans syndrome, sclerotic GVHD, or serositis. | I | Children's Research Institute, Johns Hopkins University |
HD: Hodgkin Disease; HL: Hodgkin lymphoma; NHL: Non-Hodgkins Lymphoma; SCAEBV: severe Chronic Active EBV Infection Syndrome; CTL: cytotoxic T lymphocyte; NCI: National Cancer Institute; DZ: disease; HTLV: human T Cell leukemia virus; ARD: Asparaginase refractory disease; ABC: asparaginase-based chemotherapy; ALC: Absolute lymphocyte count; EBV: Epstein Barr virus; EBER: EBV encoded RNA; ECOG: Eastern Cooperative Oncology Group; NCI CTCAE v4.0: National Cancer Institute Common Terminology Criteria for Adverse Events; PR: partially responsive/partial response; PD: progressive disease; incl: including/includes; sib: sibling; ECG: echocardiogram; neg: negative; 1°: primary; 2°: secondary; RD: residual dz; MUD: or matched unrelated adult donor; SCAEBV: Severe chronic active EBV infection syndrome;qRT-PCR: quantitative real time PCR; Immsupp: immunosuppressive; DLT: dose limiting toxicity; Hx: history; rxn: reaction; MI: Myocardial infarction; TBI: total body irradiation; LVEF: left ventricular ejection fraction; LVSF: left ventricular shortening fraction; mAB: monoclonal antibodies;
Targeting viral antigens on lymphomas
The “right” antigens for lymphoma are currently unknown. Epstein Barr virus (EBV) remains dormant in around 1% of B cells following infection resolution and is highly associated with approximately 40% of cases in immune-competent HL and NHL. EBV is also >90% associated with post-transplant lymphoproliferative disorder (PTLD), thus it has been a valuable target for T cell therapy of EBV+ malignancies. In contrast, for patients with EBV-negative lymphomas, identification of lymphoma-specific tumor antigens has been vital for generating lymphoma-specific therapies. Both EBV-specific and LAA-specific T cells are under current investigation by multiple institutions to identify which are able to most effectively clear lymphoma.
EBV-specific T cells for PTLD
EBV-mediated lymphomas that occur in patients post-HSCT have not been exposed to the influence of immune-selection and as such are exceptionally sensitive to immune cell-mediated therapy. Rituximab, a monoclonal antibody (mAB) targeting CD20 is prescribed for EBV-mediated lymphoproliferative disease in immune-compromised transplant patients. In a proportion of patients Rituximab alone can constrain B cell proliferation throughout the post-transplant immune cell regeneration stage. Thereafter the patient's immune system can resume regulation of EBV-infected B cells, quelling their proliferation. T cells specific for EBV antigens have proven highly beneficial for regaining control of EBV-driven proliferation in patients with PTLD for whom Rituximab is not successful. This approach elucidated the mechanism of action of non-specific DLI, which was first demonstrated to reestablish EBV immunity and suppress lymphoproliferative disease in the mid-1990s - although this was often accompanied by severe GVHD88. The approach was later refined with the generation of EBV-specific T cells designed to eliminate EBV+ tumors and reduce the risk of GVHD89–91. In a trial of 114 HSCT patients, from 3 study sites, none of the patients who received prophylactic infusion of third party, EBV-primed CTLs developed lymphoproliferative disease91 establishing the robustness of this approach.
EBV-specific T cells for HL and NHL
EBV+ Hodgkin's lymphoma (HL) and NHL arise in an immune-competent setting. These tumors have been exposed to immune editing and display minimal expression of EBV antigens. Consequently these tumors are less immunogenic and more resistant to immune control. Nevertheless a 20% response rate, and persistence up to a year, has been observed following treatment with autologous EBV-CTLs in these patients65. Further studies with antigen presenting cells (APC) genetically modified to express LMP1/2 proteins for expanding both autologous and allogeneic CTLs demonstrated safety and efficacy of these EBV-CTLs64.
The limits of success for adoptive T cell therapy
Transferred T cells are also subject to endogenous immune suppression in the host tumor microenvironment (TME). Clinicians can evaluate the effectiveness of cell-based therapeutics by monitoring adoptively transferred cells' capacity to home to the tumor site(s), engage with the target(s) in vivo, expand and survive. Unfortunately, in the absence of gene “marking” definitive determination of whether such responses are related to the adoptive T cell transfer can be difficult to achieve. However, more complete knowledge of where and when inhibition of infused cells occurs will enable design of sound tactics to thwart immune-restricting factors in vivo. Lymphodepletion pre-T cell infusion is one approach employed to overcome the tumor-induced immune suppressive microenvironment and to promote homeostatic lymphoproliferation. Proliferation of donor T cells in the host is also fostered by administering exogenous cytokines, such as interleukin-2 (IL-2) and IL-15, that promote T cell survival and expansion. Ex vivo expansion of T cells outside of the inhibitory elements of the malignant microenvironment in vivo can produce highly activated, antigen-specific, cytotoxic T cells. Nevertheless following adoptive transfer these cells remain susceptible to the actions of steroids administered to control GVHD post-HSCT, which can inhibit the function of the infused cells and stimulate apoptosis.
Tumors can avoid direct CD8+ T cell-mediated lysis by downregulating their HLA Class I molecules. Thus treatments that induce tumors to upregulate MHC-I can reestablish antigenicity, and the efficacy of some chemotherapies lies in their capacity to stimulate MHC-I expression92. Further inhibitory mechanisms include ownregulation of costimulatory molecules thus limiting CTL activation, and secretion of immune-modulating factors. TGF-β, for example, promotes regulatory functions in dendritic cells and macrophages and directly inhibits T cell function93. Restoration of anti-tumor immunity via engineering antigen-specific T cells that are resistant to TGF-β is one hypothesis that is currently being explored (NCT00368082)94. Tumor-secreted factors also contribute to the accumulation of myeloid-derived suppressor cells (MDSC) that in turn inhibit T cell function and drive CD4+ T cells down the regulatory pathway into TREGs. Strategies for overcoming the pro-tumor effects of these immature myeloid cells include targeted depletion, deactivation, induction of maturation and blocking their development95,96.
The existence of tumor stem cells that are ignored by the immune system may provide a reservoir of MRD cells that remain undetected after an apparently successful treatment and cause disease relapse. The bone marrow, which is a source of hematopoietic progenitors, may provide this leukemia-favoring niche. Hence, strategies designed to eliminate leukemia stem cells are focused on survival and self-renewal pathways (NF-κB, PI3K/Akt/mTOR, Wnt/β-catenin, Hedgehog); cell surface antigens (CD123, CLL-1, CD25, CD47, CD33); and the bone marrow microenvironment (adhesion and homing [CXCR4/SDF-1, VLA-4, CD44, MUC1-C] and hypoxia [HIF-1a, VEGF, hypoxia-activated prodrugs])97.
Finally, targeting immune checkpoints has seen impressive results in the treatment of patients with some solid tumors and hematological malignancies. CTLA4 is upregulated on activated T cells to regulate contraction of the expanded clonal population. CTLA4 binding to CD80/86 on antigen presenting cells or tumor cells triggers ‘off’ signaling pathways in the T cell, hence anti-CTLA4 mABs are utilized in cancer patients to block CTLA4 binding with its ligand and thus maintain T cell activity98. Likewise the PD-1–PD-L1 interaction between T cells and tumor cells inhibits T cell proliferation, and anti-PD-1/PD-L1 mABs are able to rescue this inhibition, particularly in HL99.
However not all patients respond to PD-1/PD-L1 blockade and the degree to which T cell exhaustion can be reversed is unknown. Nevertheless, exhaustion reversal has been observed after DLI and is associated with disease responses in patients with relapsed CML50. Therefore, the consensus is that single immune or cell-based therapeutics probably will not work in isolation and combination approaches (checkpoint inhibition with cell-based therapies) may be the best way forward for a potent anti-tumoral response in vivo (NCT02973113 (PREVALE); NCT02846376; NCT02733042).
Conclusion
T cell therapeutics for hematological malignancies are making steady progress towards more efficacious treatments but are currently restrained by certain limitations: Donor Lymphocyte Infusions have achieved very positive results in CML but lymphoma and acute leukemia patients remain less responsive to this approach; and the means to disengage the GVT effect from GVHD is one “Moon Shot” that still awaits discovery. Researchers are elucidating various means by which heightened cell-mediated anti-tumor effects can be achieved, including selective depletion of certain T cell subsets, and ex vivo production of T cells that target tumor- or viral-associated antigens. More finely-tuned T cell-based immunotherapies, combined with: (i) rational immune checkpoint blockade, (ii) genetically engineered TCRs or chimeric antigen receptors, and/or (iii) highly precise pharmacological molecular targeting, will likely lead to effective, durable and less toxic treatments for patients.
Practice Points.
Bone Marrow Transplant represents the original form of successful cell therapy for hematological malignancy.
Donor lymphocyte infusions administered to patients after BMT can achieve sustained remissions especially in patients with CML
Selective depletion strategies can permit the infusion of large numbers of leukemia-reactive donor T cells without GVHD, or the need for immunosuppression to prevent GVHD, leading to superior GVL effects in patients after BMT
Selective expansion of tumor antigen-specific T cells has shown efficacy in patients with leukemias and lymphomas without the toxicity profile of gene engineered T cells
Research Agenda.
Collaborative studies are required to extend the tumor antigen-specific T cell field beyond boutique centers
Combining antigen-specific T cells with other immunotherapeutic modalities such as checkpoint inhibitors may be required to enhance efficacy
Footnotes
Disclosures: The authors report no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.van Mierlo GJD, Boonman ZFHM, Dumortier HMH, den Boer AT, Fransen MF, Nouta J, et al. Activation of Dendritic Cells That Cross-Present Tumor-Derived Antigen Licenses CD8+ CTL to Cause Tumor Eradication. J Immunol. 2004;173(11):6753–6759. doi: 10.4049/jimmunol.173.11.6753. [DOI] [PubMed] [Google Scholar]
- 2.Pozzi LAM, Maciaszek JW, Rock KL. Both dendritic cells and macrophages can stimulate naive CD8 T cells in vivo to proliferate, develop effector function, and differentiate into memory cells. J Immunol Baltim Md 1950. 2005;175(4):2071–2081. doi: 10.4049/jimmunol.175.4.2071. [DOI] [PubMed] [Google Scholar]
- 3.Barrio MM, Abes R, Colombo M, Pizzurro G, Boix C, Roberti MP, et al. Human macrophages and dendritic cells can equally present MART-1 antigen to CD8(+) T cells after phagocytosis of gamma-irradiated melanoma cells. PloS One. 2012;7(7):e40311. doi: 10.1371/journal.pone.0040311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hon H, Oran A, Brocker T, Jacob J. B Lymphocytes Participate in Cross-Presentation of Antigen following Gene Gun Vaccination. J Immunol. 2005;174(9):5233–5242. doi: 10.4049/jimmunol.174.9.5233. [DOI] [PubMed] [Google Scholar]
- 5.de Wit J, Souwer Y, Jorritsma T, Klaasse Bos H, ten Brinke A, Neefjes J, et al. Antigen-Specific B Cells Reactivate an Effective Cytotoxic T Cell Response against Phagocytosed Salmonella through Cross-Presentation. PLoS One. 2010;5(9):e13016. doi: 10.1371/journal.pone.0013016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mariño E, Tan B, Binge L, Mackay CR, Grey ST. B-cell cross-presentation of autologous antigen precipitates diabetes. Diabetes. 2012;61(11):2893–2905. doi: 10.2337/db12-0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Heit A, Huster KM, Schmitz F, Schiemann M, Busch D, Wagner H. CpG-DNA Aided Cross-Priming by Cross-Presenting B Cells. J Immunol. 2004;172(3):1501–1507. doi: 10.4049/jimmunol.172.3.1501. [DOI] [PubMed] [Google Scholar]
- 8.Bernhard CA, Ried C, Kochanek S, Brocker T. CD169+ macrophages are sufficient for priming of CTLs with specificities left out by cross-priming dendritic cells. Proc Natl Acad Sci. 2015;112(17):5461–5466. doi: 10.1073/pnas.1423356112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Martinez-Pomares L, Gordon S. CD169+ macrophages at the crossroads of antigen presentation. Trends Immunol. 2012;33(2):66–70. doi: 10.1016/j.it.2011.11.001. [DOI] [PubMed] [Google Scholar]
- 10.Asano K, Nabeyama A, Miyake Y, Qiu CH, Kurita A, Tomura M, et al. CD169-positive macrophages dominate antitumor immunity by crosspresenting dead cell-associated antigens. Immunity. 2011;34(1):85–95. doi: 10.1016/j.immuni.2010.12.011. [DOI] [PubMed] [Google Scholar]
- 11.Mauri D, Wyss-Coray T, Gallati H, Pichler WJ. Antigen-presenting T cells induce the development of cytotoxic CD4+ T cells. I. Involvement of the CD80-CD28 adhesion molecules. J Immunol. 1995;155(1):118–127. [PubMed] [Google Scholar]
- 12.Haabeth OAW, Tveita AA, Fauskanger M, Schjesvold F, Lorvik KB, Hofgaard PO, et al. How Do CD4+ T Cells Detect and Eliminate Tumor Cells That Either Lack or Express MHC Class II Molecules? Front Immunol. 2014;5 doi: 10.3389/fimmu.2014.00174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dunn GP, Old LJ, Schreiber RD. The Three Es of Cancer Immunoediting. Annu Rev Immunol. 2004;22(1):329–360. doi: 10.1146/annurev.immunol.22.012703.104803. [DOI] [PubMed] [Google Scholar]
- 14.Emens LA. Chemoimmunotherapy. Cancer J Sudbury Mass. 2010;16(4):295–303. doi: 10.1097/PPO.0b013e3181eb5066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8(1):59–73. doi: 10.1038/nri2216. [DOI] [PubMed] [Google Scholar]
- 16.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13(9):1050–1059. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
- 17.Apetoh L, Tesniere A, Ghiringhelli F, Kroemer G, Zitvogel L. Molecular Interactions between Dying Tumor Cells and the Innate Immune System Determine the Efficacy of Conventional Anticancer Therapies. Cancer Res. 2008;68(11):4026–4030. doi: 10.1158/0008-5472.CAN-08-0427. [DOI] [PubMed] [Google Scholar]
- 18.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2006;13(1):54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
- 19.Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother CII. 2007;56(5):641–648. doi: 10.1007/s00262-006-0225-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Derer A, Frey B, Fietkau R, Gaipl US. Immune-modulating properties of ionizing radiation: rationale for the treatment of cancer by combination radiotherapy and immune checkpoint inhibitors. Cancer Immunol Immunother CII. 2015 doi: 10.1007/s00262-015-1771-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10(7):718–726. doi: 10.1016/S1470-2045(09)70082-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14(10):1014–1022. doi: 10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Golden EB, Apetoh L. Radiotherapy and Immunogenic Cell Death. Semin Radiat Oncol. 2015;25(1):11–17. doi: 10.1016/j.semradonc.2014.07.005. [DOI] [PubMed] [Google Scholar]
- 24.Park B, Yee C, Lee KM. The Effect of Radiation on the Immune Response to Cancers. Int J Mol Sci. 2014;15(1):927–943. doi: 10.3390/ijms15010927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kalbasi A, June CH, Haas N, Vapiwala N. Radiation and immunotherapy: a synergistic combination. J Clin Invest. 2013;123(7):2756–2763. doi: 10.1172/JCI69219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hennel R, Brix N, Seidl K, Ernst A, Scheithauer H, Belka C, et al. Release of monocyte migration signals by breast cancer cell lines after ablative and fractionated γ-irradiation. Radiat Oncol. 2014;9(1):1. doi: 10.1186/1748-717X-9-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rubner Y, Muth C, Strnad A, Derer A, Sieber R, Buslei R, et al. Fractionated radiotherapy is the main stimulus for the induction of cell death and of Hsp70 release of p53 mutated glioblastoma cell lines. Radiat Oncol. 2014;9(1):1. doi: 10.1186/1748-717X-9-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
- 29.Scheithauer H, Belka C, Lauber K, Gaipl US. Immunological aspects of radiotherapy. Radiat Oncol. 2014;9(1):185. doi: 10.1186/1748-717X-9-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112(12):4371–4383. doi: 10.1182/blood-2008-03-077974. [DOI] [PubMed] [Google Scholar]
- 31.Jedlickova Z, Schmid C, Koenecke C, Hertenstein B, Baurmann H, Schwerdtfeger R, et al. Long-term results of adjuvant donor lymphocyte transfusion in AML after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51(5):663–667. doi: 10.1038/bmt.2015.234. [DOI] [PubMed] [Google Scholar]
- 32.Horan JT, Alonzo TA, Lyman GH, Gerbing RB, Lange BJ, Ravindranath Y, et al. Impact of Disease Risk on Efficacy of Matched Related Bone Marrow Transplantation for Pediatric Acute Myeloid Leukemia: The Children's Oncology Group. J Clin Oncol. 2008;26(35):5797–5801. doi: 10.1200/JCO.2007.13.5244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Creutzig U, Reinhardt D. current controversies: which patients with acute myeloid leukaemia should receive a bone marrow transplantation? – A European view. Br J Haematol. 2002;118(2):365–377. doi: 10.1046/j.1365-2141.2002.03697.x. [DOI] [PubMed] [Google Scholar]
- 34.Bierings M, Nachman JB, Zwaan CM. Stem cell transplantation in pediatric leukemia and myelodysplasia: state of the art and current challenges. Curr Stem Cell Res Ther. 2007;2(1):53–63. doi: 10.2174/157488807779317035. [DOI] [PubMed] [Google Scholar]
- 35.Ottmann OG, Druker BJ, Sawyers CL, Goldman JM, Reiffers J, Silver RT, et al. A phase 2 study of imatinib in patients with relapsed or refractory Philadelphia chromosome–positive acute lymphoid leukemias. Blood. 2002;100(6):1965–1971. doi: 10.1182/blood-2001-12-0181. [DOI] [PubMed] [Google Scholar]
- 36.Shah NP. Loss of Response to Imatinib: Mechanisms and Management. ASH Educ. Program Book. 2005;2005(1):183–187. doi: 10.1182/asheducation-2005.1.183. [DOI] [PubMed] [Google Scholar]
- 37.Weisberg E, Manley P, Mestan J, Cowan-Jacob S, Ray A, Griffin JD. AMN107 (nilotinib): a novel and selective inhibitor of BCR-ABL. Br J Cancer. 2006;94(12):1765–1769. doi: 10.1038/sj.bjc.6603170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kolb HJ, Mittermuller J, Clemm C, Holler E, Ledderose G, Brehm G, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76(12):2462–2465. [PubMed] [Google Scholar]
- 39.Ertl HCJ, Zaia J, Rosenberg SA, June CH, Dotti G, Kahn J, et al. Considerations for the Clinical Application of Chimeric Antigen Receptor (CAR) T Cells: Observations from a Recombinant DNA Advisory Committee (RAC) Symposium June 15, 2010. Cancer Res. 2011;71(9):3175–3181. doi: 10.1158/0008-5472.CAN-10-4035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Miller JS. Therapeutic applications: natural killer cells in the clinic. Hematol Am Soc Hematol Educ Program. 2013;2013:247–253. doi: 10.1182/asheducation-2013.1.247. [DOI] [PubMed] [Google Scholar]
- 41.Pittari G, Filippini P, Gentilcore G, Grivel JC, Rutella S. Revving up Natural Killer Cells and Cytokine-Induced Killer Cells Against Hematological Malignancies. Front Immunol. 2015;6 doi: 10.3389/fimmu.2015.00230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Smith A, Khuu H, Betters DM, Cook L, Ramos C, Grasmeder S, et al. Adoptive Transfer of Escalating Doses of Ex Vivo Expanded Autologous Natural Killer (NK) Cells In Patients with Advanced Malignancies Following Bortezomib Treatment to Sensitize to NK-TRAIL Cytotoxicity. Blood. 2010;116(21):4296–4296. [Google Scholar]
- 43.Radujkovic A, Guglielmi C, Bergantini S, Iacobelli S, van Biezen A, Milojkovic D, et al. Donor Lymphocyte Infusions for Chronic Myeloid Leukemia Relapsing after Allogeneic Stem Cell Transplantation: May We Predict Graft-versus-Leukemia Without Graft-versus-Host Disease? Biol Blood Marrow Transplant. 2015;21(7):1230–1236. doi: 10.1016/j.bbmt.2015.03.012. [DOI] [PubMed] [Google Scholar]
- 44.Baron F, Beguin Y. Prophylactic donor lymphocyte infusion in patients with high-risk acute myeloid leukemia: ready for prime time? Bone Marrow Transplant. 2016;51(5):640–642. doi: 10.1038/bmt.2016.38. [DOI] [PubMed] [Google Scholar]
- 45.Schmid C, Labopin M, Veelken H, Schaap NPM, Schleuning M, Stadler M, et al. Efficacy, Safety and Long Term Results of Prophylactic and Preemptive Donor Lymphocyte Infusion after Allogeneic Stem Cell Transplantation for Acute Leukemia: A Registry-Based Evaluation on 343 Patients By the Acute Leukemia Working Party of EBMT. Blood. 2015;126(23):863–863. [Google Scholar]
- 46.Yan CH, Liu DH, Liu KY, Xu LP, Liu YR, Chen H, et al. Risk stratification–directed donor lymphocyte infusion could reduce relapse of standard-risk acute leukemia patients after allogeneic hematopoietic stem cell transplantation. Blood. 2012;119(14):3256–3262. doi: 10.1182/blood-2011-09-380386. [DOI] [PubMed] [Google Scholar]
- 47.Yan CH, Wang Y, Wang JZ, Chen YH, Chen Y, Wang FR, et al. Minimal residual disease- and graft-vs.-host disease-guided multiple consolidation chemotherapy and donor lymphocyte infusion prevent second acute leukemia relapse after allotransplant. J Hematol Oncol J Hematol Oncol. 2016;9:87. doi: 10.1186/s13045-016-0319-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Eefting M, de Wreede LC, Halkes CJM, von dem Borne PA, Kersting S, Marijt EWA, et al. Multi-state analysis illustrates treatment success after stem cell transplantation for acute myeloid leukemia followed by donor lymphocyte infusion. Haematologica. 2016;101(4):506–514. doi: 10.3324/haematol.2015.136846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Barber LD, Rakhayes MA, Golding J, Feliu J, Perez-Abellan P, Floro L, et al. Pre-Emptive Donor Lymphocyte Infusion Corrects Deficiencies in the T-Cell Repertoire after Allogeneic Haematopoietic Stem Cell Transplantation with Lymphocyte-Depletion for Acute Myeloid Leukaemia and Myelodysplastic Syndromes. Biol Blood Marrow Transplant. 2016;22(3):S150. [Google Scholar]
- 50.Bachireddy P, Hainz U, Rooney M, Pozdnyakova O, Aldridge J, Zhang W, et al. Reversal of in situ T-cell exhaustion during effective human antileukemia responses to donor lymphocyte infusion. Blood. 2014;123(9):1412–1421. doi: 10.1182/blood-2013-08-523001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nikiforow S, Kim HT, Daley H, Reynolds C, Jones KT, Armand P, et al. A phase I study of CD25/regulatory T-cell-depleted donor lymphocyte infusion for relapse after allogeneic stem cell transplantation. Haematologica. 2016;101(10):1251–1259. doi: 10.3324/haematol.2015.141176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Whitehill GD, Amarnath S, Muranski P, Keyvanfar K, Battiwalla M, Barrett A, et al. Adenosine Selectively Depletes Alloreactive T Cells to Prevent GVHD While Conserving Immunity to Viruses and Leukemia. Mol Ther. 2016;24(9):1655–1664. doi: 10.1038/mt.2016.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rådestad E, Destad E, Wikell H, Engstrom M, Watz E, Thunberg S, et al. Alpha/Beta T-Cell Depleted Grafts as an Immunological Booster to Treat Graft Failure after Hematopoietic Stem Cell Transplantation with HLA -Matched Related and Unrelated Donors. J Immunol Res. 2014;2014:e578741. doi: 10.1155/2014/578741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Meyer RG, Britten CM, Wehler D, Bender K, Hess G, Konur A, et al. Prophylactic transfer of CD8-depleted donor lymphocytes after T-cell–depleted reduced-intensity transplantation. Blood. 2007;109(1):374–382. doi: 10.1182/blood-2006-03-005769. [DOI] [PubMed] [Google Scholar]
- 55.Zorn E, Wang KS, Hochberg EP, Canning C, Alyea EP, Soiffer RJ, et al. Infusion of CD4+ Donor Lymphocytes Induces the Expansion of CD8+ Donor T Cells with Cytolytic Activity Directed against Recipient Hematopoietic Cells. Clin Cancer Res. 2002;8(7):2052–2060. [PubMed] [Google Scholar]
- 56.Yan CH, Wang JZ, Liu DH, Xu LP, Chen H, Liu KY, et al. Chemotherapy followed by modified donor lymphocyte infusion as a treatment for relapsed acute leukemia after haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion: superior outcomes compared with chemotherapy alone and an analysis of prognostic factors. Eur J Haematol. 2013;91(4):304–314. doi: 10.1111/ejh.12168. [DOI] [PubMed] [Google Scholar]
- 57.Cruz CR, Bollard CM. T-cell and natural killer cell therapies for hematologic malignancies after hematopoietic stem cell transplantation: enhancing the graft-versus-leukemia effect. Haematologica. 2015;100(6):709–719. doi: 10.3324/haematol.2014.113860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Shafer JA, Cruz CR, Leen AM, Ku S, Lu A, Rousseau A, et al. Antigen-Specific Cytotoxic T Lymphocytes can Target Chemoresistant Side-Population Tumor Cells in Hodgkin's Lymphoma. Leuk Lymphoma. 2010;51(5):870–880. doi: 10.3109/10428191003713968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Weber G, Caruana I, Rouce RH, Barrett AJ, Gerdemann U, Leen AM, et al. Generation of tumor antigen-specific T cell lines from pediatric patients with acute lymphoblastic leukemia--implications for immunotherapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2013;19(18):5079–5091. doi: 10.1158/1078-0432.CCR-13-0955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Leen A, Tzannou I, Bilgi M, Liu H, Vera JF, Gerdemann U, et al. Immunotherapy for Lymphoma Using T Cells Targeting Multiple Tumor Associated Antigens. Blood. 2015;126(23):186–186. [Google Scholar]
- 61.Bornhäuser M, Thiede C, Platzbecker U, Kiani A, Oelschlaegel U, Babatz J, et al. Prophylactic transfer of BCR-ABL–, PR1-, and WT1-reactive donor T cells after T cell–depleted allogeneic hematopoietic cell transplantation in patients with chronic myeloid leukemia. Blood. 2011;117(26):7174–7184. doi: 10.1182/blood-2010-09-308569. [DOI] [PubMed] [Google Scholar]
- 62.Mohamed YS, Bashawri LA, Vatte C, Abu-Rish EY, Cyrus C, Khalaf WS, et al. The in vitro generation of multi-tumor antigen-specific cytotoxic T cell clones: Candidates for leukemia adoptive immunotherapy following allogeneic stem cell transplantation. Mol Immunol. 2016;77:79–88. doi: 10.1016/j.molimm.2016.07.012. [DOI] [PubMed] [Google Scholar]
- 63.Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci TranslMed. 2013;5(174):174ra27. doi: 10.1126/scitranslmed.3004916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, et al. Sustained Complete Responses in Patients With Lymphoma Receiving Autologous Cytotoxic T Lymphocytes Targeting Epstein-Barr Virus Latent Membrane Proteins. J Clin Oncol. 2014;32(8):798–808. doi: 10.1200/JCO.2013.51.5304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bollard CM, Aguilar L, Straathof KC, Gahn B, Huls MH, Rousseau A, et al. Cytotoxic T Lymphocyte Therapy for Epstein-Barr Virus+ Hodgkin's Disease. J Exp Med. 2004;200(12):1623–1633. doi: 10.1084/jem.20040890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bollard CM, Gottschalk S, Leen AM, Weiss H, Straathof KC, Carrum G, et al. Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer. Blood. 2007;110(8):2838–2845. doi: 10.1182/blood-2007-05-091280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Saito S, Yanagisawa R, Yoshikawa K, Higuchi Y, Koya T, Yoshizawa K, et al. Safety and tolerability of allogeneic dendritic cell vaccination with induction of Wilms tumor 1–specific T cells in a pediatric donor and pediatric patient with relapsed leukemia: a case report and review of the literature. Cytotherapy. 2015;17(3):330–335. doi: 10.1016/j.jcyt.2014.10.003. [DOI] [PubMed] [Google Scholar]
- 68.Tendeloo VFV, de Velde AV, Driessche AV, Cools N, Anguille S, Ladell K, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci. 2010;107(31):13824–13829. doi: 10.1073/pnas.1008051107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Shah NN, Loeb DM, Khuu H, Stroncek D, Ariyo T, Raffeld M, et al. Induction of Immune Response after Allogeneic Wilms' Tumor 1 Dendritic Cell Vaccination and Donor Lymphocyte Infusion in Patients with Hematologic Malignancies and Post-Transplantation Relapse. Biol Blood Marrow Transplant. 2016;22(12):2149–2154. doi: 10.1016/j.bbmt.2016.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kitawaki T, Kadowaki N, Kondo T, Ishikawa T, Ichinohe T, Teramukai S, et al. Potential of dendritic cell immunotherapy for relapse after allogeneic hematopoietic stem cell transplantation, shown by WT1 peptide - and keyhole limpet hemocyanin-pulsed, donor-derived dendritic cell vaccine for acute myeloid leukemia. Am J Hematol. 2008;83(4):315–317. doi: 10.1002/ajh.21127. [DOI] [PubMed] [Google Scholar]
- 71.Hess G, Bunjes D, Siegert W, Schwerdtfeger R, Ledderose G, Wassmann B, et al. Sustained complete molecular remissions after treatment with imatinib-mesylate in patients with failure after allogeneic stem cell transplantation for chronic myelogenous leukemia: results of a prospective phase II open-label multicenter study. J Clin Oncol Off J Am Soc Clin Oncol. 2005;23(30):7583–7593. doi: 10.1200/JCO.2005.01.3110. [DOI] [PubMed] [Google Scholar]
- 72.Rezvani K, Yong ASM, Mielke S, Savani BN, Musse L, Superata J, et al. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood. 2008;111(1):236–242. doi: 10.1182/blood-2007-08-108241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Solomon SR, Mielke S, Savani BN, Montero A, Wisch L, Childs R, et al. Selective depletion of alloreactive donor lymphocytes: a novel method to reduce the severity of graft-versus-host disease in older patients undergoing matched sibling donor stem cell transplantation. Blood. 2005;106(3):1123–1129. doi: 10.1182/blood-2005-01-0393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Amrolia PJ, Muccioli-Casadei G, Huls H, Adams S, Durett A, Gee A, et al. Adoptive immunotherapy with allodepleted donor T-cells improves immune reconstitution after haploidentical stem cell transplantation. Blood. 2006;108(6):1797–1808. doi: 10.1182/blood-2006-02-001909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Mielke S, McIver Z, Shenoy A, Fellowes V, Khuu H, Stroncek D, et al. Selectively T cell depleted allografts from HLA-matched sibling donors followed by low-dose post transplant immunosuppression to improve transplant outcome in patients with hematological malignancies. Biol Blood Marrow Transplant J Am Soc Blood Marrow Transplant. 2011;17(12):1855–1861. doi: 10.1016/j.bbmt.2011.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Roy DC, Lachance S, Roy J, Walker I, Maertens J, Cohen S, et al. Donor Lymphocytes Depleted of Alloreactive T-Cells (ATIR101) Reduce Transplant Related Mortality and Improve Overall Survival in Haploidentical HSCT for Patients with AML and ALL, Using an Immunosuppressant-Free Transplant Regimen. Blood. 2015;126(23):4391–4391. [Google Scholar]
- 77.Bleakley M, Heimfeld S, Loeb KR, Jones LA, Chaney C, Seropian S, et al. Outcomes of acute leukemia patients transplanted with naive T cell–depleted stem cell grafts. J Clin Invest. 2015;125(7):2677–2689. doi: 10.1172/JCI81229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Falkenburg JHF, Wafelman AR, Joosten P, Smit WM, van Bergen CAM, Bongaerts R, et al. Complete Remission of Accelerated Phase Chronic Myeloid Leukemia by Treatment With Leukemia-Reactive Cytotoxic T Lymphocytes. Blood. 1999;94(4):1201–1208. [PubMed] [Google Scholar]
- 79.Falkenburg JHF, van de Corput L, Marijt EWA, Willemze R. Minor histocompatibility antigens in human stem cell transplantation. Exp Hematol. 2003;31(9):743–751. doi: 10.1016/s0301-472x(03)00190-5. [DOI] [PubMed] [Google Scholar]
- 80.Stumpf AN, van der Meijden ED, van Bergen CAM, Willemze R, Falkenberg JHF, Griffioen M. Identification of 4 new HLA-DR–restricted minor histocompatibility antigens as hematopoietic targets in antitumor immunity. Blood. 2009;114(17):3684–3692. doi: 10.1182/blood-2009-03-208017. [DOI] [PubMed] [Google Scholar]
- 81.Mutis T, Brand R, Gallardo D, van Biezen A, Niederwieser D, Goulmy E. Graft-versus-host driven graft-versus-leukemia effect of minor histocompatibility antigen HA-1 in chronic myeloid leukemia patients. Leukemia. 2010;24(7):1388–1392. doi: 10.1038/leu.2010.115. [DOI] [PubMed] [Google Scholar]
- 82.Warren EH, Fujii N, Akatsuka Y, Chaney C, Mito JK, Loeb KR, et al. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood. 2010;115(19):3869–3878. doi: 10.1182/blood-2009-10-248997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bleakley M, Otterud BE, Richardt JL, Mollerup AD, Hudecek M, Nishida T, et al. Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by in vitro stimulation of naive CD8+ T cells. Blood. 2010;115(23):4923–4933. doi: 10.1182/blood-2009-12-260539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Rezvani K, Yong ASM, Mielke S, Jafarpour B, Savani BN, Le RQ, 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(3):432–440. doi: 10.3324/haematol.2010.031674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Di Stasi A, Jimenez AM, Minagawa K, Al-Obaidi M, Rezvani K. Review of the Results of WT1 Peptide Vaccination Strategies for Myelodysplastic Syndromes and Acute Myeloid Leukemia from Nine Different Studies. Front Immunol. 2015;6 doi: 10.3389/fimmu.2015.00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kuball J, de Boer K, Wagner E, Wattad M, Antunes E, Weeratna RD, et al. Pitfalls of vaccinations with WT1-, Proteinase3- and MUC1-derived peptides in combination with MontanideISA51 and CpG7909. Cancer Immunol Immunother. 2011;60(2):161–171. doi: 10.1007/s00262-010-0929-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Doubrovina E, Carpenter T, Pankov D, Selvakumar A, Hasan A, O'Reilly RJ. Mapping of novel peptides of WT-1 and presenting HLA alleles that induce epitope-specific HLA-restricted T cells with cytotoxic activity against WT-1+ leukemias. Blood. 2012;120(8):1633–1646. doi: 10.1182/blood-2011-11-394619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.O'Reilly RJ, Lacerda JF, Lucas KG, Rosenfield NS, Small TN, Papadopoulos EB. Adoptive cell therapy with donor lymphocytes for EBV-associated lymphomas developing after allogeneic marrow transplants. Important Adv Oncol. 1996:149–166. [PubMed] [Google Scholar]
- 89.Rooney CM, Smith CA, Ng CYC, Loftin SK, Sixbey JW, Gan Y, et al. Infusion of Cytotoxic T Cells for the Prevention and Treatment of Epstein-Barr Virus–Induced Lymphoma in Allogeneic Transplant Recipients. Blood. 1998;92(5):1549–1555. [PubMed] [Google Scholar]
- 90.Bollard CM, Rooney CM, Heslop HE. T-cell therapy in the treatment of post-transplant lymphoproliferative disease. Nat Rev Clin Oncol. 2012;9(9):510–519. doi: 10.1038/nrclinonc.2012.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010;115(5):925–935. doi: 10.1182/blood-2009-08-239186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wan S, Pestka S, Jubin RG, Lyu YL, Tsai YC, Liu LF. Chemotherapeutics and Radiation Stimulate MHC Class I Expression through Elevated Interferon-beta Signaling in Breast Cancer Cells. PLoS One. 2012;7(3):e32542. doi: 10.1371/journal.pone.0032542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hargadon KM. Dysregulation of TGFβ1 Activity in Cancer and Its Influence on the Quality of Anti -Tumor Immunity. J Clin Med. 2016;5(76) doi: 10.3390/jcm5090076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Foster AE, Dotti G, Lu A, Khalil M, Brenner MK, Heslop HE, et al. Antitumor Activity of EBV-specific T Lymphocytes Transduced With a Dominant Negative TGF-β Receptor. J Immunother Hagerstown Md. 2008;199731(5):500–505. doi: 10.1097/CJI.0b013e318177092b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.De Veirman K, Van Valckenborgh E, Lahmar Q, Geeraerts X, De Bruyne E, Menu E, et al. Myeloid-Derived Suppressor Cells as Therapeutic Target in Hematological Malignancies. Front Oncol. 2014;4 doi: 10.3389/fonc.2014.00349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Giallongo C, Parrinello N, Brundo MV, Raccuia SA, Di Rosa M, La Cava P, et al. Myeloid Derived Suppressor Cells in Chronic Myeloid Leukemia. Front Oncol. 2015;5 doi: 10.3389/fonc.2015.00107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Gojo I, Karp JE. New Strategies in Acute Myelogenous Leukemia: Leukemogenesis and Personalized Medicine. Clin Cancer Res Off J Am Assoc Cancer Res. 2014;20(24):6233–6241. doi: 10.1158/1078-0432.CCR-14-0900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.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):1581–1588. doi: 10.1182/blood-2008-07-168468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Armand P. Immune checkpoint blockade in hematologic malignancies. Blood. 2015;125(22):3393–3400. doi: 10.1182/blood-2015-02-567453. [DOI] [PubMed] [Google Scholar]
