Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2018

Open Access 01.12.2018 | Short report

Induction of anti-leukemic responses by stimulation of leukemic CD3+ cells with allogeneic stimulator cells

verfasst von: Alejandro Pando, John L. Reagan, Martha Nevola, Loren D. Fast

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2018

Abstract

Background

Immunotherapeutic protocols have focused on identification of stimuli that induce effective anti-leukemic immune responses. One potent immune stimulus is the encounter with allogeneic cells. Our group previously showed that the infusion of haploidentical donor white blood cells (1–2 × 108 CD3+ cells/kg) into patients with refractory hematological malignancies induced responses of varying magnitude in over half of the patients. Because donor cells were eliminated within 2 weeks in these patients, it is presumed that the responses of recipient lymphocytes were critically important in achieving prolonged anti-leukemic responses.

Methods

The role of patient CD3+ cells in anti-leukemic responses was examined by isolating peripheral blood mononuclear cells from newly diagnosed leukemic patients. Immunophenotyping was performed on these peripheral blood mononuclear cells. CD3+ cells were isolated from the peripheral blood mononuclear cells and tested for their ability to proliferate and lyse autologous leukemic cells when stimulated with unrelated allogeneic cells.

Results

Allostimulated CD3+ cells effectively generated cytolytic responses to autologous CD3-cells in 11/21 patients. Increased numbers of CD4+ cells expressing high levels of granzyme A, B and perforin and CD8+CD39+ cells were found in nonresponsive CD3+ cells.

Conclusions

These results indicate that CD3+ cells from leukemic patients are capable of generating anti-leukemic responses when stimulated with unrelated allogeneic cells. This model can be used to identify approaches using alloreactive responses by patient lymphocytes to enhance in vivo anti-leukemic responses.
Abkürzungen
CRS
cytokine release syndrome
CTL
cytolytic T lymphocyte
GVHD
graft-versus-host disease
GVL
graft-versus-leukemia
MHC
major histocompatibility complex
MLC
mixed lymphocyte culture
PBMC
peripheral blood mononuclear cells

Background

Allogeneic cells are one of the more potent stimuli of the immune system, as 100–1000 fold more cells respond to an allogeneic major histocompatibility complex (MHC) molecules than to a foreign peptide presented by self MHC molecules [1]. Alloreactivity is the driving force for deleterious responses such as graft-versus-host disease (GVHD) and organ rejection seen in transplantation, but is also linked to beneficial graft versus leukemia (GVL) responses which has led to efforts to separate GVL responses from GVHD responses [2]. Within our group a nonengraftment donor leukocyte infusion protocol, in which infusion of large numbers of haploidentical cells at a dose of 1–2 × 108 CD3+ cells/kg into patients with refractory hematological malignancies, resulted in responses of varying magnitude in over half of the patients tested (14/26 overall responses with 5 complete responses) including three complete responses in patients with refractory acute myeloid leukemia (AML) [3]. These responses occurred in the absence of durable engraftment as donor cells were no longer detected within 2 weeks of infusion. This protocol was characterized by the rapid development of a cytokine release syndrome (CRS) marked by high plasma levels of proinflammatory cytokines and fevers that remitted with corticosteroid administration. Anti-leukemic responses may potentially be mediated by donor lymphocytes for a limited period of time but, given the lack of durable chimerism, we postulate that extended responses were more likely mediated by recipient lymphocytes [4]. The intense alloreactive immune response generated by this protocol could facilitate anti-leukemic responses by a variety of mechanisms, such as the reactivation of memory anti-T cells from leukemic patients [5, 6] or the cross reactivity of patient alloreactive effector cells toward patient cancer cells [1, 7]. Herein we describe the ability of leukemic patient CD3+ lymphocytes to generate cytolytic responses against the syngeneic CD3-fraction, which includes leukemia cells, in vitro following stimulation with allogeneic donor peripheral blood mononuclear cells (PBMC).

Methods

Patient samples

The patient peripheral blood mononuclear (PBMC) samples included newly diagnosed acute lymphoid, acute myeloid, chronic myeloid, and chronic myelomonocytic leukemic patients categorized based on WHO criteria. Stimulator PBMCs were obtained from unrelated normal volunteers who were employees at Rhode Island Hospital. HLA typing was not performed on patient or donor samples. Patient and donor samples were collected following informed consent through a Rhode Island Hospital IRB approved protocol.
After isolation of PBMC using Ficoll-Hypaque discontinuous centrifugation, CD3+ and CD3-cells were isolated from the patient’s PBMC using CD3 immunomagnetic particles according to manufacturer’s instructions (Miltenyi Biotec, Inc, San Diego, CA, USA). Aliquots of the CD3-fraction of the patient’s cells, containing the leukemic cells, were frozen for later use. Blood from two unrelated normal donors were obtained for each patient. The PBMC obtained from the blood of each normal control along with the remainder of the CD3-cells from the leukemic patient were treated with Mitomycin C (Sigma, St. Louis, MO, USA).

Immunophenotyping

Immunophenotyping was conducted on the freshly isolated PBMC from the leukemic patients. The CD3+ and CD3-cells were stained with antibody panels which defined T cell subsets and NK cells (APC-H7 anti-CD3, V500 anti-CD4, V450 anti-CD8, FITC anti-TCRγδ, PerCP-Cy5.5 anti-CD56), differentiation and activation (FITC anti-CD27, PE anti-CD45RA, FITC anti-CD25, APC anti-CD39, PerCP-Cy5.5 FoxP3, APC anti-CD152, FITC anti-CD279, PE anti-Tim3,) and cytolytic activity (FITC anti-granzyme A, Alexa 647 anti-granzyme B, PE anti-perforin, FITC anti-CD107a, PE anti-TRAIL, Alexa 647 anti-Fas ligand). All antibodies were purchased from BDBiosciences (San Jose, CA).

Assays

Mixed lymphocyte cultures (MLC) were set up using RPMI medium containing 5% Human AB serum (Valley Biomedical, Winchester, VA, USA) and 1% penicillin and streptomycin (Life Technologies, Grand Island, NY, USA) [8]. 1 × 106 patient CD3+ cells or 2 × 106 control PBMC were added to 2 × 106 mitomycin C treated PBMC from controls or CD3-leukemic fraction in 2 mL per well in a 24 well plate. In addition, triplicate wells containing tenfold fewer cells/medium were prepared in a 96 well plate. The response of T cells from leukemic patient samples to bound anti-CD3/CD28 in a prepared 96 well plate was tested by plating 1 × 105 T cells from leukemic patients/well or 2 × 105 PBMC (normal controls)/well, each in triplicate. All plates were incubated at 37 °C under 10% CO2 for the designated period of time.
Proliferation of cells was determined by measuring 3H-thymidine incorporation after a 4 h pulse on day 3 for cells activated with anti-CD3/28 or day 5 for MLC cultures. The data is presented as stimulation index (S.I.), equal to the ratio of the mean counts per minute (cpm) for the response to allogeneic stimulators divided by the sum of responder vs. medium and stimulator vs. medium cpm. Effector cells generated by 7 day MLC cultures were collected and tested for their ability to lyse 51Cr labeled target cells (both CD3-leukemic cells and normal donor blasts) using varying effector target cell ratios. Donor blasts were generated by culturing PBMC in 125U/mL rhuIL-2 (Zeptometrix, Buffalo, NY, USA) and 2 ug/mL concanavalin A (Sigma, St. Louis, MO, USA) in RPMI 1640 containing 5% FCS and streptomycin/penicillin for 7 days at 37 °C. Leukemic CD3-cells were thawed on day 6 and held overnight in 20% FCS medium. The results are presented as lytic units/106 cells in which 1 lytic unit (LU) is defined as the number of effector cells required to achieve 30% lysis of 1 × 104 target cells.

Statistical analysis

Statistical analysis was performed utilizing Graphpad Prism 7.0 (GraphPad Software). The Mann–Whitney non-parametric test (Mann–Whitney U = 248, n1 = n2 = 42, P < 0.05 two-tailed) and unpaired t-tests were used to determine the P values indicated in the figures. Significance was defined to have been reached at P values from < 0.05.

Results

The characteristics of 21 newly diagnosed leukemic patients studied are described in Table 1. Cytogenetic and molecular classifications were based on NCCN criteria [9]. The median age was 60.5 years (range, 29–86), the median number of absolute lymphocyte count collected was 2.7 × 106/mL, and the median percentage of peripheral blasts in the CD3-cells was 23%. Disease types were (AML, 14 patients), chronic myeloid leukemia (CML, 1 patient), chronic myelomonocytic leukemia (CMML, 3 patients), and acute lymphocytic leukemia (ALL, 3 patients).
Table 1
Characteristics of newly diagnosed leukemic patients
Pt#
Sex
Age
Diagnosis
WBC
ALC
Peripheral blasts (%)
Cytogenetics molecular markers
1
Female
74
AML
17.1
6.2
6
XX
2
Female
65
AML
7.4
3.6
9
t (4;11), Tetrasomy 8
3
Male
26
CML
336.5
20.2
4
t (9;22)
4
Male
31
AML
9.2
2.7
39
inv (3), Monosomy 7
5
Female
86
AML
54.4
8.2
32
Trisomy 13
6
Male
32
AML
7.1
0.9
12
del (17), t (16;21)
7
Male
84
CMML
39.6
2.4
11
XY
8
Male
62
AML
19.4
2.5
23
inv (16), del (7)
9
Female
29
AML
8.6
4
47
t (11;20), FLT3 ITD+
10
Male
64
AML
138.3
16.6
80
XY, FLT3 ITD+
11
Female
70
AML
12.8
2.4
33
t (11;19)
12
Male
56
CMML
14.4
2.2
14
XY
13
Female
48
CMML
16.3
2.6
19
XX
14
Male
59
ALL
5.1
2.7
45
t (9;22)
15
Female
34
ALL
341.8
23.9
88
XX
16
Male
46
AML
18.5
2.3
73
t (15;17)
17
Male
64
AML
1.2
0.2
3
XY, FLT3 ITD+
18
Female
57
AML
34.5
1.7
9
XX inv (16)
19
Female
32
ALL
58.1
7.3
86
XX Ph+
20
Male
62
AML
28.1
8
9
XY
21
Female
77
AML
84.4
4.2
81
Not sent
CD3+ cells were either incubated with mitomycin C-treated healthy unrelated donor allogeneic PBMC (allostimulated CD3+ cells) or syngeneic CD3-cells (autologous stimulated CD3+ cells). Proliferation was detected for all combinations with similar levels of proliferation between those cells stimulated with allogeneic cells (S.I. 13.2 ± 19.3) or autologous CD3-cells (S.I. 15.3 ± 21.1). On day 7, allostimulated CD3+ cells and CD3+ cells stimulated with syngeneic CD3-cells were examined for cytolytic killing of CD3-cells. Eleven of the twenty-one patient allostimulated CD3+ cells were able to lyse CD3-cells (Fig. 1a). CD3+ cells from seven of the responding leukemic patients lysed CD3-cells when stimulated with either allogeneic stimulator. CD3+ cells from four patients were able to lyse CD3-target cells when stimulated with syngeneic CD3-cells. These CD3+ cells also proliferated strongly to the syngeneic CD3-cells (S.I. of 3.9, 20.9, 24.2, 79.6) and three out of four also generated strong alloreactive CTL when stimulated with allogeneic cells. These findings contrasted to the lack of proliferation seen when CD3+ cells obtained from normal donors were stimulated with syngeneic CD3-cells (SI = 0.9 ± 0.3, n = 4). In addition no detectable lysis of normal syngeneic CD3-cells was seen when CD3+ cells from normal donors were stimulated with allogeneic PBMNC (n = 9), syngeneic PBMNC (n = 4) or syngeneic CD3-cells (n = 4).
T CD3-cell killing was induced via allogeneic stimulation for AML and CML/CMML but not in ALL samples (Fig. 1b). Patient samples with peripheral blasts greater than 50% had an increase in the stimulation index compared to those with less than 20% peripheral blasts. Additionally, lytic effector function in greater then 50% blasts had a P value of 0.08 compared to those with less than 20%. Together this suggests that candidates with more peripheral blasts may respond better to allostimulation than those with less peripheral blasts.
Analysis of factors that were associated with the ability or inability of the alloreactive CD3+ cells to generate cytolytic activity toward syngeneic CD3-cells containing leukemic cells identified several immunophenotypes associated with the CD3+ cells which did not generate cytolytic activity toward the CD3-cells. The first was an increased number of CD4+ cells expressing high levels of perforin, granzyme A and granzyme B (Fig. 1c). A second finding was an increased number of CD8+ cells expressing CD39, a phenotype found in terminally exhausted CD8+ cells. A nonsignificant increase of CD4+CD39+cells was also observed in this set of CD3+ cells. (Figure 1d). In addition the number of CD4+ cells expressing gamma delta T cell receptor (TCRγδ) was significantly upregulated on in the unresponsive CD3+ cells (Fig. 1d). No significant differences in the number of FoxP3+ cells or the expression of inhibitory receptors was detected between the patients that lysed leukemic cells and those that did not. Staining freshly isolated cells with antibodies detecting cytolytic activity or pathways including CD107a, TRAIL and Fas Ligand did not demonstrate any predictive value for identifying which CD3+ cells would generate anti-leukemic responses (data not shown). Interestingly, all three AML patients who harbored FLT-3 internal tandem duplication (ITD) mutations, a prognostic marker in AML with poor prognosis [10], were able to lyse leukemia containing CD3-cell fractions following allogeneic stimulation.

Discussion

In summary, in our screening of 21 newly diagnosed leukemia samples approximately half of the patients’ CD3+ cells were able to generate lysis of syngeneic cells containing leukemic cells after allostimulation. The ability of alloreactive effector cells to lyse syngeneic leukemic cells was similar to the findings previously reported in mice [11, 12]. It was even reported that stimulation with syngeneic tumor cells induced alloreactive effector cells [13]. These findings would suggest that cancer cells express unique peptides on their MHC molecules that allow them to be recognized by alloreactive effector cells [14]. Previously, when the peptides bound to HLA B7 molecules isolated from normal cells and leukemic cells were analyzed and compared, it was observed that there were many more unique peptides present on the HLA B7 molecules isolated from the leukemic cells [15]. Many of these unique peptides were phosphopeptides. Stimulation of PBMC from normal donors with a pool of these unique peptides resulted in T cell responses while the same pool of peptides did not induce responses in PBMC from cancer patients. This suggested that those effector cells had been inhibited or eliminated in the cancer bearing patients.
While one possibility is that alloreactive T cells could cross react with tumor cells, another possibility is that the alloreactive responses activate T cells specific for tumor associated antigens. For example, T cells reactive with Wilms tumor-1 (WT1) antigen have been identified in many individuals [1620]. Guo et al. [17] found increased numbers of Wilm tumor-1 (WT1) antigen reactive CD8+ T cells in 33 out of 39 patients that received infusions of HLA-mismatched donor cells in their protocol. Although the source of these antigen reactive CD8+ cells was unknown in many of these cases, in 6 of these cases the WT1 reactive CD8+ cells could be determined to be of host origin. Several studies have also indicated that vaccination with WT1 vaccines facilitated or prolonged responses [21, 22]. A study in which patients received G-CSF mobilized haploidentical donor cells after receiving 100 cGy total body irradiation, a patient who went into complete remission was found to have high levels of CD8+ cells reactive with proteinase-3, a tumor associated antigen, with no detectable donor cells [23]. These examples indicate that the expansion of T cells reactive with antigens expressed by the leukemic cells could distinguish between those individuals whose cells responded in our study and those that did not.
Phenotypic studies identified an increased number of CD4+ cells expressing cytolytic effector molecules and increased number of recipient CD8+ cells expressing CD39, a marker of exhaustion as a characteristic of the CD3+ cells that did not generate anti-leukemic responses when stimulated with allogeneic cells. The appearance of cytolytic CD4+ cells can be linked to repeated antigen exposure in chronic infection and in cancer bearing individuals reflecting a senescent immune system [24]. It has been shown that these cells can mediate cytolytic activity either via the granzyme perforin pathway or via Fas/Fas ligand signaling. In patients with chronic hepatitis B infection and hepatocellular carcinoma it was found that the CD4+ CTL also produced IL-10 which inhibited the function of CD8+ CTL [25]. Adding anti-IL-10 antibody reversed the inhibition of CD8+ cell function mediated by the CD4+ CTL. This mechanism could provide a possible explanation for the association between increased numbers of CD4+ CTL and decreased CD8+ cell activity.
In our experiments, the increased numbers of CD4+ expressing the γδTCR was associated with lack of response. Usually γδ T cells have an advantage over αβ T cells in cancer because antigen processing and presentation is not required but can instead directly recognize cancer molecules [26] although some γδ subsets may facilitate cancer growth [27]. There is a recent report of CD39+ γδ T cells that were induced by TGF-β and served as potent immunoregulatory cells in colorectal cancer [28]. Although it cannot be determined directly from this data because antibodies to CD39 and γδ TCR were included in different antibody panels, increased numbers of both CD4+ γδ T cells and CD39+CD4+ cells were detected in the CD3+ cells from the unresponsive patients. These findings would be consistent with an increased number of these CD39+ γδ T regulatory cells in this group of patients that are inhibiting other T cell responses and this possibility will be examined more closely in future studies. The CD8+CD39+ phenotype is associated with the exhausted phenotype and increased numbers of these cells suggests that the cancer reactive CD8+ cells could be exhausted and unable to respond to allostimulators [29].
In future clinical trials of nonengraftment donor leukocyte infusion protocols, it will be interesting to test if the expression of these particular phenotypes will identify those patients unresponsive to this protocol. These in vivo trials will further define which factors allow for reproducible induction of an anti-leukemic response by allostimulated leukemic patient CD3+ cells.

Conclusions

Patient’s T lymphocytes are frequently able to attack syngeneic hematological malignancies when stimulated with allogeneic cells. Nonresponders can be defined using the phenotypic distinctions based on the presence of CD4+ CTL and CD8+CD39+ cells. Understanding the basis for the ability of alloreactive cells to lyse syngeneic cancer cells will allow for the development of protocols which harness these responses to achieve effective anti-cancer responses in all patients.

Authors’ contributions

All authors performed various aspects of the research. AP, JLR and LDF analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Not applicable.
These studies were approved by the Rhode Island Hospital Institutional Review Board.

Funding

These studies were funded by funds provided by the Division of Hematology/Oncology, Rhode Island Hospital.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat D’Orsogna LJ, Nguyen TH, Claas FH, Witt C, Mifsud NA. Endogenous-peptide-dependent alloreactivity: new scientific insights and clinical implications. Tissue Antigens. 2013;81(6):399–407.CrossRef D’Orsogna LJ, Nguyen TH, Claas FH, Witt C, Mifsud NA. Endogenous-peptide-dependent alloreactivity: new scientific insights and clinical implications. Tissue Antigens. 2013;81(6):399–407.CrossRef
2.
Zurück zum Zitat Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112(12):4371–83.CrossRef Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112(12):4371–83.CrossRef
3.
Zurück zum Zitat Colvin GA, Berz D, Ramanathan M, Winer ES, Fast L, Elfenbein GJ, et al. Nonengraftment haploidentical cellular immunotherapy for refractory malignancies: tumor responses without chimerism. Biol Blood Marrow Transplant. 2009;15(4):421–31.CrossRef Colvin GA, Berz D, Ramanathan M, Winer ES, Fast L, Elfenbein GJ, et al. Nonengraftment haploidentical cellular immunotherapy for refractory malignancies: tumor responses without chimerism. Biol Blood Marrow Transplant. 2009;15(4):421–31.CrossRef
4.
Zurück zum Zitat Dey BR, McAfee S, Colby C, Cieply K, Caron M, Saidman S, et al. Anti-tumour response despite loss of donor chimaerism in patients treated with non-myeloablative conditioning and allogeneic stem cell transplantation. Br J Haematol. 2005;128(3):351–9.CrossRef Dey BR, McAfee S, Colby C, Cieply K, Caron M, Saidman S, et al. Anti-tumour response despite loss of donor chimaerism in patients treated with non-myeloablative conditioning and allogeneic stem cell transplantation. Br J Haematol. 2005;128(3):351–9.CrossRef
5.
Zurück zum Zitat Freeman BE, Hammarlund E, Raue HP, Slifka MK. Regulation of innate CD8+ T-cell activation mediated by cytokines. Proc Natl Acad Sci U S A. 2012;109(25):9971–6.CrossRef Freeman BE, Hammarlund E, Raue HP, Slifka MK. Regulation of innate CD8+ T-cell activation mediated by cytokines. Proc Natl Acad Sci U S A. 2012;109(25):9971–6.CrossRef
6.
Zurück zum Zitat Raue HP, Beadling C, Haun J, Slifka MK. Cytokine-mediated programmed proliferation of virus-specific CD8(+) memory T cells. Immunity. 2013;38(1):131–9.CrossRef Raue HP, Beadling C, Haun J, Slifka MK. Cytokine-mediated programmed proliferation of virus-specific CD8(+) memory T cells. Immunity. 2013;38(1):131–9.CrossRef
7.
Zurück zum Zitat Morris GP, Uy GL, Donermeyer D, Dipersio JF, Allen PM. Dual receptor T cells mediate pathologic alloreactivity in patients with acute graft-versus-host disease. Sci Transl Med. 2013;5(188):188ra74.CrossRef Morris GP, Uy GL, Donermeyer D, Dipersio JF, Allen PM. Dual receptor T cells mediate pathologic alloreactivity in patients with acute graft-versus-host disease. Sci Transl Med. 2013;5(188):188ra74.CrossRef
8.
Zurück zum Zitat Fast LD, Dileone G, Li J, Goodrich R. Functional inactivation of white blood cells by Mirasol treatment. Transfusion. 2006;46(4):642–8.CrossRef Fast LD, Dileone G, Li J, Goodrich R. Functional inactivation of white blood cells by Mirasol treatment. Transfusion. 2006;46(4):642–8.CrossRef
9.
Zurück zum Zitat O’Donnell MR, Tallman MS, Abboud CN, Altman JK, Appelbaum FR, Arber DA, et al. Acute myeloid leukemia version 3.2017 NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(7):926–57.CrossRef O’Donnell MR, Tallman MS, Abboud CN, Altman JK, Appelbaum FR, Arber DA, et al. Acute myeloid leukemia version 3.2017 NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(7):926–57.CrossRef
10.
Zurück zum Zitat Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89.CrossRef Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89.CrossRef
11.
Zurück zum Zitat Paciucci PA, Macphail S, Zarling JM, Bach FH. Lysis of syngeneic solid tumor cells by alloantigen stimulated mouse T and non-T cells. J Immunol. 1980;124(1):370–5.PubMed Paciucci PA, Macphail S, Zarling JM, Bach FH. Lysis of syngeneic solid tumor cells by alloantigen stimulated mouse T and non-T cells. J Immunol. 1980;124(1):370–5.PubMed
12.
Zurück zum Zitat Sensi ML, Parenza M, Parmiani G. Alloreactivity and tumor antigens: generation of syngeneic antilymphoma killer lymphocytes by alloimmunization of mice with normal cells. J Natl Cancer Inst. 1983;70(2):291–7.PubMed Sensi ML, Parenza M, Parmiani G. Alloreactivity and tumor antigens: generation of syngeneic antilymphoma killer lymphocytes by alloimmunization of mice with normal cells. J Natl Cancer Inst. 1983;70(2):291–7.PubMed
13.
Zurück zum Zitat Schirrmacher V, Hubsch D, Garrido F. Syngeneic tumor cells can induce alloreactive T killer cells: a biological role for transplantation antigens. Proc Natl Acad Sci U S A. 1980;77(9):5409–13.CrossRef Schirrmacher V, Hubsch D, Garrido F. Syngeneic tumor cells can induce alloreactive T killer cells: a biological role for transplantation antigens. Proc Natl Acad Sci U S A. 1980;77(9):5409–13.CrossRef
14.
Zurück zum Zitat Kumari S, Walchli S, Fallang LE, Yang W, Lund-Johansen F, Schumacher TN, et al. Alloreactive cytotoxic T cells provide means to decipher the immunopeptidome and reveal a plethora of tumor-associated self-epitopes. Proc Natl Acad Sci U S A. 2014;111(1):403–8.CrossRef Kumari S, Walchli S, Fallang LE, Yang W, Lund-Johansen F, Schumacher TN, et al. Alloreactive cytotoxic T cells provide means to decipher the immunopeptidome and reveal a plethora of tumor-associated self-epitopes. Proc Natl Acad Sci U S A. 2014;111(1):403–8.CrossRef
15.
Zurück zum Zitat Cobbold M, De La Pena H, Norris A, Polefrone JM, Qian J, English AM, et al. MHC class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci Transl Med. 2013;5(203):203ra125.CrossRef Cobbold M, De La Pena H, Norris A, Polefrone JM, Qian J, English AM, et al. MHC class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci Transl Med. 2013;5(203):203ra125.CrossRef
16.
Zurück zum Zitat 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):1633–46.CrossRef 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):1633–46.CrossRef
17.
Zurück zum Zitat Guo M, Hu KX, Liu GX, Yu CL, Qiao JH, Sun QY, et al. HLA-mismatched stem-cell microtransplantation as postremission therapy for acute myeloid leukemia: long-term follow-up. J Clin Oncol. 2012;30(33):4084–90.CrossRef Guo M, Hu KX, Liu GX, Yu CL, Qiao JH, Sun QY, et al. HLA-mismatched stem-cell microtransplantation as postremission therapy for acute myeloid leukemia: long-term follow-up. J Clin Oncol. 2012;30(33):4084–90.CrossRef
18.
Zurück zum Zitat Rezvani K, Brenchley JM, Price DA, Kilical Y, Gostick E, Sewell AK, et al. T-cell responses directed against multiple HLA-A*0201-restricted epitopes derived from Wilms’ tumor 1 protein in patients with leukemia and healthy donors: identification, quantification, and characterization. Clin Cancer Res. 2005;11(24 Pt 1):8799–807.CrossRef Rezvani K, Brenchley JM, Price DA, Kilical Y, Gostick E, Sewell AK, et al. T-cell responses directed against multiple HLA-A*0201-restricted epitopes derived from Wilms’ tumor 1 protein in patients with leukemia and healthy donors: identification, quantification, and characterization. Clin Cancer Res. 2005;11(24 Pt 1):8799–807.CrossRef
19.
Zurück zum Zitat Rezvani K, Grube M, Brenchley JM, Sconocchia G, Fujiwara H, Price DA, et al. Functional leukemia-associated antigen-specific memory CD8+ T cells exist in healthy individuals and in patients with chronic myelogenous leukemia before and after stem cell transplantation. Blood. 2003;102(8):2892–900.CrossRef Rezvani K, Grube M, Brenchley JM, Sconocchia G, Fujiwara H, Price DA, et al. Functional leukemia-associated antigen-specific memory CD8+ T cells exist in healthy individuals and in patients with chronic myelogenous leukemia before and after stem cell transplantation. Blood. 2003;102(8):2892–900.CrossRef
20.
Zurück zum Zitat Rezvani K, Yong AS, Savani BN, Mielke S, Keyvanfar K, Gostick E, et al. Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood. 2007;110(6):1924–32.CrossRef Rezvani K, Yong AS, Savani BN, Mielke S, Keyvanfar K, Gostick E, et al. Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood. 2007;110(6):1924–32.CrossRef
21.
Zurück zum Zitat Maeda T, Hosen N, Fukushima K, Tsuboi A, Morimoto S, Matsui T, et al. Maintenance of complete remission after allogeneic stem cell transplantation in leukemia patients treated with Wilms tumor 1 peptide vaccine. Blood Cancer J. 2013;3:e130.CrossRef Maeda T, Hosen N, Fukushima K, Tsuboi A, Morimoto S, Matsui T, et al. Maintenance of complete remission after allogeneic stem cell transplantation in leukemia patients treated with Wilms tumor 1 peptide vaccine. Blood Cancer J. 2013;3:e130.CrossRef
22.
Zurück zum Zitat Van Driessche A, Berneman ZN, Van Tendeloo VF. Active specific immunotherapy targeting the Wilms’ tumor protein 1 (WT1) for patients with hematological malignancies and solid tumors: lessons from early clinical trials. Oncologist. 2012;17(2):250–9.CrossRef Van Driessche A, Berneman ZN, Van Tendeloo VF. Active specific immunotherapy targeting the Wilms’ tumor protein 1 (WT1) for patients with hematological malignancies and solid tumors: lessons from early clinical trials. Oncologist. 2012;17(2):250–9.CrossRef
23.
Zurück zum Zitat Medina DJ, Gharibo M, Savage P, Cohler A, Kuriyan M, Balsara B, et al. A pilot study of allogeneic cellular therapy for patients with advanced hematologic malignancies. Leuk Res. 2008;32(12):1842–8.CrossRef Medina DJ, Gharibo M, Savage P, Cohler A, Kuriyan M, Balsara B, et al. A pilot study of allogeneic cellular therapy for patients with advanced hematologic malignancies. Leuk Res. 2008;32(12):1842–8.CrossRef
24.
Zurück zum Zitat Brown DM, Lampe AT, Workman AM. The differentiation and protective function of cytolytic CD4 T cells in influenza infection. Front Immunol. 2016;7:93.CrossRef Brown DM, Lampe AT, Workman AM. The differentiation and protective function of cytolytic CD4 T cells in influenza infection. Front Immunol. 2016;7:93.CrossRef
25.
Zurück zum Zitat Meng F, Zhen S, Song B. HBV-specific CD4+ cytotoxic T cells in hepatocellular carcinoma are less cytolytic toward tumor cells and suppress CD8+ T cell-mediated antitumor immunity. APMIS. 2017;125(8):743–51.CrossRef Meng F, Zhen S, Song B. HBV-specific CD4+ cytotoxic T cells in hepatocellular carcinoma are less cytolytic toward tumor cells and suppress CD8+ T cell-mediated antitumor immunity. APMIS. 2017;125(8):743–51.CrossRef
26.
Zurück zum Zitat Chien YH, Konigshofer Y. Antigen recognition by gammadelta T cells. Immunol Rev. 2007;215:46–58.CrossRef Chien YH, Konigshofer Y. Antigen recognition by gammadelta T cells. Immunol Rev. 2007;215:46–58.CrossRef
27.
Zurück zum Zitat Ye J, Ma C, Hsueh EC, Eickhoff CS, Zhang Y, Varvares MA, et al. Tumor-derived gammadelta regulatory T cells suppress innate and adaptive immunity through the induction of immunosenescence. J Immunol. 2013;190(5):2403–14.CrossRef Ye J, Ma C, Hsueh EC, Eickhoff CS, Zhang Y, Varvares MA, et al. Tumor-derived gammadelta regulatory T cells suppress innate and adaptive immunity through the induction of immunosenescence. J Immunol. 2013;190(5):2403–14.CrossRef
28.
Zurück zum Zitat Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, et al. Tumor-infiltrating CD39(+)gammadeltaTregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 2017;6(2):e1277305.CrossRef Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, et al. Tumor-infiltrating CD39(+)gammadeltaTregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 2017;6(2):e1277305.CrossRef
29.
Zurück zum Zitat Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog. 2015;11(10):e1005177.CrossRef Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog. 2015;11(10):e1005177.CrossRef
Metadaten
Titel
Induction of anti-leukemic responses by stimulation of leukemic CD3+ cells with allogeneic stimulator cells
verfasst von
Alejandro Pando
John L. Reagan
Martha Nevola
Loren D. Fast
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2018
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-018-0118-5

Weitere Artikel der Ausgabe 1/2018

Experimental Hematology & Oncology 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.