20
ORIGINAL ARTICLE In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping Susanne Reuther Helga Schmetzer Friedhelm R. Schuster Pina Krell Christine Grabrucker Anja Liepert Tanja Kroell Hans-Jochem Kolb Arndt Borkhardt Raymund Buhmann Received: 19 August 2011 / Accepted: 28 February 2012 Ó Springer-Verlag 2012 Abstract Myeloid leukemic cells can be induced to dif- ferentiate into leukemia-derived dendritic cells (DC leu ) regaining the stimulatory capacity of professional DCs while presenting the leukemic antigen repertoire. But so far, the induced antileukemic T-cell responses are variable both in specificity and in efficacy. In an attempt to elucidate the underlying causes of different T-cell response patterns, T-cell receptor (TR) Vb chain rearrangements were corre- lated with the T cells corresponding immunophenotypic profile, as well as their proliferative response and cytolytic capacities. In three different settings, donor T cells, either human leukocyte antigen matched or mismatched (haplo- identical), or autologous T cells were repeatedly stimulated with myeloid blasts or leukemia-derived DC/DC leus from the corresponding patients diseased from acute myeloid leukemia (AML). Although no significant differences in T-cell proliferation were observed, the T-cell-mediated cytolytic response pattern varied considerably and even caused blast proliferation in two cases. Spectratyping revealed a remarkable restriction ( [ 75 % of normal level) of the CD4 ? or CD8 ? -TR repertoire of blast- or DC/DC leu - stimulated T cells. Although in absolute terms, DC/DC leu stimulation induced the highest grade of restriction in the CD8 ? T-cell subset, the CD4 ? T-cell compartment seemed to be relatively more affected. But most importantly, in vitro stimulation with DC/DC leu resulted into an identical TR restriction pattern (b chain) that could be identified in vivo in a patient sample 3 months after allo-SCT. Thus, in vitro tests combining functional flow cytometry with spectratyping might provide predictive information about T cellular response patterns in vivo. Keywords Acute myeloid leukemia Dendritic cells T-cell response Immunoscope spectratyping Immunotherapy Introduction Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are clonal disorders of hematopoietic stem cells, characterized by an impaired normal cell dif- ferentiation [1]. About 70 % of successfully chemothera- peutically treated AML patients relapse soon [2], indicating the need for additional therapeutic strategies in order to maintain stable remissions. So far, allogeneic stem cell transplantation (SCT) is the only curative treatment option [3] with donor T cells mediating the antileukemic reaction [4, 5]. But although transfusion of donor T cells (DLT) can re-induce complete remissions in relapsed patients after Susanne Reuther, Helga Schmetzer, Friedhelm R. Schuster contributed equally. S. Reuther F. R. Schuster P. Krell A. Borkhardt Faculty of Medical, Department of Paediatric Oncology, Haematology and Immunology, University Dusseldorf, 40225 Dusseldorf, Germany H. Schmetzer C. Grabrucker A. Liepert T. Kroell H.-J. Kolb R. Buhmann Department of Medicine III, University of Munich-Grosshadern, Munich, Germany P. Krell Faculty of Technology and Institute for Bioinformatics, Center for Biotechnology, Bielefeld University, Bielefeld, Germany H.-J. Kolb R. Buhmann (&) Helmholtz Center Munich, German Research Center for Environmental Health, CCG-HCT, Marchioninistr. 25, 81377 Munich, Germany e-mail: [email protected] 123 Clin Exp Med DOI 10.1007/s10238-012-0180-y

In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

  • Upload
    raymund

  • View
    213

  • Download
    0

Embed Size (px)

Citation preview

Page 1: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

ORIGINAL ARTICLE

In vitro-induced response patterns of antileukemic T cells:characterization by spectratyping and immunophenotyping

Susanne Reuther • Helga Schmetzer • Friedhelm R. Schuster •

Pina Krell • Christine Grabrucker • Anja Liepert • Tanja Kroell •

Hans-Jochem Kolb • Arndt Borkhardt • Raymund Buhmann

Received: 19 August 2011 / Accepted: 28 February 2012

� Springer-Verlag 2012

Abstract Myeloid leukemic cells can be induced to dif-

ferentiate into leukemia-derived dendritic cells (DCleu)

regaining the stimulatory capacity of professional DCs

while presenting the leukemic antigen repertoire. But so far,

the induced antileukemic T-cell responses are variable both

in specificity and in efficacy. In an attempt to elucidate the

underlying causes of different T-cell response patterns,

T-cell receptor (TR) Vb chain rearrangements were corre-

lated with the T cells corresponding immunophenotypic

profile, as well as their proliferative response and cytolytic

capacities. In three different settings, donor T cells, either

human leukocyte antigen matched or mismatched (haplo-

identical), or autologous T cells were repeatedly stimulated

with myeloid blasts or leukemia-derived DC/DCleus from

the corresponding patients diseased from acute myeloid

leukemia (AML). Although no significant differences in

T-cell proliferation were observed, the T-cell-mediated

cytolytic response pattern varied considerably and even

caused blast proliferation in two cases. Spectratyping

revealed a remarkable restriction ([75 % of normal level)

of the CD4? or CD8?-TR repertoire of blast- or DC/DCleu-

stimulated T cells. Although in absolute terms, DC/DCleu

stimulation induced the highest grade of restriction in the

CD8? T-cell subset, the CD4? T-cell compartment seemed

to be relatively more affected. But most importantly, in

vitro stimulation with DC/DCleu resulted into an identical

TR restriction pattern (b chain) that could be identified in

vivo in a patient sample 3 months after allo-SCT. Thus, in

vitro tests combining functional flow cytometry with

spectratyping might provide predictive information about T

cellular response patterns in vivo.

Keywords Acute myeloid leukemia � Dendritic cells �T-cell response � Immunoscope spectratyping �Immunotherapy

Introduction

Acute myeloid leukemia (AML) and myelodysplastic

syndrome (MDS) are clonal disorders of hematopoietic

stem cells, characterized by an impaired normal cell dif-

ferentiation [1]. About 70 % of successfully chemothera-

peutically treated AML patients relapse soon [2], indicating

the need for additional therapeutic strategies in order to

maintain stable remissions. So far, allogeneic stem cell

transplantation (SCT) is the only curative treatment option

[3] with donor T cells mediating the antileukemic reaction

[4, 5]. But although transfusion of donor T cells (DLT) can

re-induce complete remissions in relapsed patients after

Susanne Reuther, Helga Schmetzer, Friedhelm R. Schuster

contributed equally.

S. Reuther � F. R. Schuster � P. Krell � A. Borkhardt

Faculty of Medical, Department of Paediatric Oncology,

Haematology and Immunology, University Dusseldorf,

40225 Dusseldorf, Germany

H. Schmetzer � C. Grabrucker � A. Liepert � T. Kroell �H.-J. Kolb � R. Buhmann

Department of Medicine III, University of Munich-Grosshadern,

Munich, Germany

P. Krell

Faculty of Technology and Institute for Bioinformatics, Center

for Biotechnology, Bielefeld University, Bielefeld, Germany

H.-J. Kolb � R. Buhmann (&)

Helmholtz Center Munich, German Research Center for

Environmental Health, CCG-HCT, Marchioninistr. 25,

81377 Munich, Germany

e-mail: [email protected]

123

Clin Exp Med

DOI 10.1007/s10238-012-0180-y

Page 2: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

SCT, there are still numerous patients who do not respond.

Moreover, appearance of graft-versus-host (GvH) reactions

can impair the efficacy of SCT or relapse therapy [3, 6].

The reasons for these varying T-cell effects have to be

elucidated. An insufficient expression of costimulatory

antigens, major histocompatibility complex (MHC) mole-

cules and tumor-associated antigens (TAA) on the surface

of cancer cells as well as disturbed mechanisms of apop-

tosis might be the main reasons for an ineffective immune

response in malignant diseases [7]. As dendritic cells (DCs)

are known to stimulate T-effector cells, especially tumor

cytotoxic T cells [8, 9], they are of potential interest for

antitumor or antileukemic vaccination strategies [10, 11].

In contrast to solid tumor cells, myeloid leukemic cells can

be converted in leukemia-derived DCs (DCleu) expressing

DC-typical antigens (e.g., CD40, CD86, CD80, CD1a and

CD83), thereby regaining the stimulatory capacity of

mature professional dendritic cells [12–14]. Thus, the

cumbersome identification and subsequent pulsing with

tumor-specific antigens probably can be overcome because

the leukemia-derived DCs might still present the complete

leukemic antigen repertoire [15–21]. In most cases, stim-

ulation with DCleu containing DC fractions (DC/DCleu)

resulted in a very effective cytotoxic T-cell response. But

there are a few cases, where opposite T-cell response

patterns have been observed, in terms of T cells mediating

anergy or even supporting blast proliferation [20–24]. So

far, these different T-cell response patterns are not pre-

dictable, but it might be expected that different ‘qualities’

of stimulators may result into different clonal compositions

and functional diversities of T-cell subsets.

Thus, in this study, we sought to determine the impact of

blast- versus DC/DCleu-mediated stimulation on T-cell

differentiation and correlated the induced T-cell receptor

(TR) b chain rearrangements with the T cells correspond-

ing immunophenotypic profile, as well as their proliferative

response and cytolytic capacities. Thereby, T-cell receptor

rearrangement analysis and functional T-cell assays might

contribute not only to the understanding of antileukemia

directed immunoreactions in myeloid leukemia, but could

also help to identify the DNA regions of T-cell receptor

loci involved in these reactions, to further isolate leukemia-

specific T cells for adoptive immunotherapy.

Materials and methods

Patient characteristics, sample collection and diagnosis

After informed consent, peripheral blood samples were

obtained from three patients with AML at the time of

diagnosis or after treatment. Diagnosis was based on the

revised World Health Organization (WHO) classification

of myeloid neoplasms and acute leukemia [25]. Cytoge-

netic analysis was performed according to standard proto-

cols and criteria defined by the International System for

Human Cytogenetic Nomenclature [26]. Patient #502 had

presented with persisting AML-M4eo after chemotherapy,

with 95 % CD34?, CD117?, CD65? and CD15? myeloid

blasts and a complex aberrant karyotype. Patient #546 had

presented with an AML-M2 at first diagnosis and 50 %

CD34?, CD117? and CD65? myeloid blasts. Patient #538

had presented with relapsed AML-M0 after SCT, normal

karyotype and 93 % CD34? and CD65? myeloid blasts

(Table 1).

DC generation

Mononuclear cells (MNC) from peripheral blood (PB) (PB-

MNC) samples were isolated by centrifugation on a Ficoll-

Hypaque (Biochrom, Berlin, Germany) density gradient,

washed and suspended in PBS without Ca2? and Mg2?

(Biochrom, Berlin, Germany). Subsequently, DCs were

generated using 3 different DC-differentiating methods

(MCM-Mimic, Picibanil and Ca-Ionophore) in parallel as

described previously [15–18, 21, 22]. The method resulting

in the best DC-counts was chosen for the quantitative

generation of DC. In brief, MNCs obtained from AML

Table 1 Sample characteristics

a Proportions of positive cells

in the mononuclear cell fraction

(MNC)

Sample no. #502 #546 #538

FAB type AML M4eo AML M2 AML MO

Karyotype 46,XY,t(2;20)qq, t(5;5)qp,

del(5p),r7, t(12;16)pq

45,X,-Y,t(8;21)qq Normal

Stage of disease Persistance First diagnosis Relapse after SCT

Leukemic blasts (%)a 95 50 93

Blast phenotype (CD) CD34, 117, 13, 33, 65, 15 CD34, 117, 33, 65, 13 CD34, 65, 33

Monocytes (%)a 2 4 2

B cells (%)a n.a. 17 3

T cells (%)a 3 8 4

NK cells (%)a 2 9 4

Clin Exp Med

123

Page 3: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

patients in an active, blast-rich phase of the disease were

incubated in 12-well multiwell tissue culture plates in

X-Vivo 15 (BioWhittaker, Verviers, Belgium) serum-free

medium. (1) DCs were generated from 2.5 9 106 MNC/mL

in ‘MCM-Mimic’ medium containing 800 U/mL granulo-

cyte macrophage colony-stimulating factor (GM-CSF), 500

U/mL interleukin (IL)-4 and 40 ng/mL FMS-related tyro-

sine kinase 3 ligand (FLT-3). After 5 days, the same

cytokines were added to the culture again. Half medium

exchange was performed on day 8. Again GM-CSF, IL-4

and FLT-3 were added together with 150 ng/mL IL-6,

5 ng/mL IL-1b, 1 lg/mL prostaglandin PGE2 and 5 ng/mL

tumor necrosis factor TNFa. After 12 days in culture, the

DCs were harvested for subsequent experiments. (2) DCs

were generated from 7 9 105 MNC/mL in ‘Ca-Ionophore’

and were cultured in the presence of 375 ng/mL A23187

and 250 U/mL IL-4 and harvested after 3 days. (3) DCs

were generated with ‘Picibanil,’ a lysis product of Strep-

tococcus pyogenes, which has nonspecific immunmodula-

tory effects from 1 to 1.25 9 106 MNC/mL in the presence

of 500 U/mL GM-CSF and 250 U/mL IL-4. After 7 days in

culture, 5 lL/mL OK-432 (Picibanil) and 1 lg/mL PGE2

were added. The cells were harvested after 10 days in

culture. All of the substances used for DC generation are

approved for human treatment.

Flow cytometry

Flow cytometric analysis with a panel of mouse mono-

clonal antibodies (moAbs) directly conjugated with fluo-

rescein isothiocyanate (FITC), phycoerythrin (PE), tandem

Cy7-PE-conjugation (PC7) or allophycocyanine (APC)

was performed to evaluate and quantify the percentage and

phenotypes of the leukemic cells, B, T and NK cells and

DC in the PB/BM (bone marrow) samples analyzed. The

antibodies were purchased from Becton–Dickinsona (Hei-

delberg, Germany), Immunotech/Beckmann Coulterb

(Krefeld, Germany) and Caltagc (Hamburg, Germany).

CD1b(a), CD3(b), CD4(a), CD28(b), CD33(b), CD45RO(b),

CD65(b), CD83(b) and CD86(c) were used as FITC-con-

jugated moAbs. CD1a(b), CD8(a), CD56(b), CD80(b),

CD154(b) and CD206 (b) were used as PE-conjugated

moAbs. CD3(b), CD14(a), CD19(b), CD25(a), CD34(b) and

CD117(b) were chosen as the PC7-conjugated moAbs.

CD3(b), CD4(b), CD13(b), CD14(b), CD33(b), CD34(b),

CD15(a), CD45RA(a) and CD71(a) were used as APC-

conjugated moAbs. The MNC or cultured cells were sus-

pended in PBS with 20 % FCS (Biochrom, Berlin, Ger-

many) and incubated with moAbs according to the

manufacturer’s instructions. Appropriate isotype controls

were used. At least 5,000 events were evaluated on a BD

FACSCalibur flow cytometer (Becton–Dickinson, Heidel-

berg, Germany) using CellQuest data acquisition and

analysis software (Becton–Dickinson, Heidelberg, Ger-

many). For analysis and quantification of the lymphocytes,

monocytes and leukemic cells before culture the total MNC

fractions were gated. An AML sample was considered as

‘positive’ for a surface marker, if the percentage of positive

events in a gate surrounding the blasts, lymphocytes and

monocytes was more than 20 %, as described [21–23].

Proportions of positive events in defined gates compared

with the isotype controls were calculated using CellQuest

software.

Quantification and characterization of DC

For analysis and quantification of DCs and leukemia-derived

DCs, a refined gating strategy was applied and DCs were

quantified by flow cytometry as already described [20–22,

27]. In brief, for quantification, DC markers were selected,

which were not expressed on naı̈ve blasts. A refined gating

strategy was applied for an exact quantification of DC sub-

sets. A ‘blast gate’ was set surrounding the blast population

and residual lymphocytes or monocytes. The remaining cells

were gated in a ‘DC gate.’ Cells displaying the typical scatter

of dendritic cells and expressing typical DC antigens (e.g.,

CD86, CD80, CD40, CD1a and CD83) could be counted.

Moreover, ‘blasts’ could be quantified using blast-specific

markers. Blasts converted to leukemia-derived DC (DCleu)

could be quantified estimating those blast cells that had

gained a DC antigen. In addition, also blasts could be

quantified with this gating strategy that had not been con-

verted to DCleu or DC of non-leukemic origin. We also

quantified DCopt, the DC marker with highest expression

rates on DC gained after culture being not expressed on naı̈ve

blasts before. DC co-expressing CD83 was defined as

‘mature DC.’ In addition, viable DC (7AAD-/DC?) and

CCR7? DC were quantified. In AML cases with an immu-

nophenotypically detectable blast population in MNC frac-

tions, amounts of blasts being converted to DC co-expressing

specific blast antigens (e.g., CD56 and CD117) were quan-

tified. Microscopical controls were regularly performed and

revealed the DC-typical morphology of large cells with

irregular shapes and cytoplasmic projections.

Mixed lymphocyte cultures (MLC): generation

of leukemia-cytotoxic T cells

Positively selected CD3 T cells (1 9 106 cells/well) (Mil-

teney Biotech, Bergisch-Gladbach, Germany) from the

healthy stem cell donor of patient #502 and patient #538 or

autologous T cells prepared from patient #546 were cocul-

tured with irradiated (20 Gy) cell suspensions containing

25,000 DCs generated from the patient’s MNC or the same

amount of irradiated MNC as a control in 1 mL RPMI 1640

medium (Biochrom, Berlin, Germany) containing 15 %

Clin Exp Med

123

Page 4: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

human serum (PAA Laboratories, Pasching, Austria) and 50

U/mL IL-2 (Proleukin R5, Chiron, Munich, Germany) as

described. The cells were harvested after 10 days of cocul-

ture and twofold restimulation with 5 9 104 irradiated DC/

DCleu (T*DC/DCleu) or 5x104 irradiated blasts (T*blasts) and

supplementation of IL-2, as described previously [20]. Half

medium exchange was carried out every 3–4 days. Six days

after the last restimulation, the cells were harvested and the

cytotoxicity assay was carried out.

The antigen expressions on the transferred CD3? T cells

were evaluated by FACS analyses comparing their co-

expression of CD4, CD8, CD45RA, CD45RO, CCR7 and

CD71 before and after blast and DC/DCleu stimulation.

This contributed to evaluate proportions of proliferating

(CD71?), naı̈ve, non-naive, central memory, CD4?, CD8?

or CCR7? T cells before or after blast or DC/DCleu

coculture [19, 28, 29].

Cytotoxicity assay (Fluorolysis)

The lytic activity of effector T cells was measured by a

fluorolysis assay counting viable target cells, labeled with

specific fluorochrome-labeled antibodies, before and after

effector cell (E) contact. In brief, DC/DCleu- or blast-stim-

ulated and unstimulated donor or patient-derived T cells

were cocultured in 1.5-mL Eppendorf tubes with thawed

blasts as target cells. The E:T ratio was adjusted to 1:1, and

cells were incubated for 3 h and overnight at 37 �C and 5 %

CO2. Before culture, target cells were stained for 15 min

with two FITC- and/or PE-conjugated ‘blast-’ or ‘DC-’

specific antibodies. As a control, target and effector cells

were cultured separately and mingled shortly before FACS

analysis. To evaluate amounts of viable (7AAD-) target T

cells and to quantify the cell loss after the corresponding

incubation time, cells were harvested, washed in PBS and

resuspended in a FACS flow solution containing 7AAD

(BD, Biosciences Pharmingen, Heidelberg, Germany) and a

defined number of fluorosphere beads (Becton–Dickinson,

Heidelberg, Germany). Viable, 7AAD negative cells co-

expressing specific blast marker (combinations) were

quantified taking into account defined counts of calibration

beads as described [29–31]. Cells were analyzed in a BD

FACSCalibur flow cytometer using CellQuest software

(Becton Dickinson, Heidelberg, Germany). The percentage

of lysis was the difference between proportions of viable

blasts before and after effector cell contact.

CDR3 immunoscope spectratyping of TR-Vbtranscripts

After CD4- and CD8-positive T-cell selection of the

unstimulated, or blast- or DC/DCleu-stimulated T-cell sus-

pensions on day 10 (Dynabeads; Dynal Biotech ASA, Oslo,

Norway), the total RNA was prepared from 1 9 105 cells

by using the TRIzol reagent (Invitrogen, Carlsbad, CA,

USA). Complementary DNA (cDNA) was synthesized by

random priming, using the cDNA synthesis kit (Quanti-

Tect; Qiagen, Hilden, Germany) and subjected to TR-Vbgene family-specific polymerase chain reaction (PCR) in

26 separate reactions, each containing one of the 26 Vbfamily primers in combination with a universal Cb-specific

primer [32]. In a second step, run-off products were gen-

erated from every Vb-specific PCR product by each of the

13 fluorescence-labeled, Jb-specific oligonucleotides [33].

The fragments were separated on an automated 48-capil-

lary DNA sequencer (Applied Biosystems 3730 Genetic

Analyzer; ABI, Foster City, CA, USA). The length and

fluorescence intensities of the complementary determining

region 3 (CDR3) of the different Vb–Jb combinations were

determined using Genescan 500LIZ (Applied Biosystems,

Warrington, UK) as standard and analyzed by using

GeneMapper software (Applied Biosystems, Warrington,

UK).

Cloning and sequencing of CD8-Vb13.1-Jb2.7 T-cell

receptor b chains

To sequence the CDR3 region, the cDNA derived from

CD4? and CD8? T cells was amplified by PCR in 50-lL

reaction volumes containing 19 Phusion Master Mix

(Finnzymes, Oslo, Norway) and 0.2 pmol of Vb13.1 and

Jb2.7 primer [32]. The PCR procedure included 35 cycles

of annealing at 60 �C. PCR products were purified with the

QIAquick PCR Purification Kit (Qiagen, Hilden, Ger-

many). For subsequent cloning into the TOPO TA vector

system (Invitrogen, Carlsbad, CA, USA), a 30 A-overhang

to PCR products was added to create sticky ends using

0.1–1.5 pmol of purified of PCR product, 0.2 mM dATP

(Invitrogen, Carlsbad, CA, USA) and 1 unit of Taq poly-

merase in a 50-lL reaction volume. The reaction was

incubated for 10 min at 72 �C, then placed on ice. The

TOPO TA cloning reaction and the transformation in

TOP10 chemically competent E. coli cells were performed

according to the manufacturer0s instructions. Briefly,

0.5–4 lL of the cDNA PCR product was mixed with 1 lL

of TOPO TA vector, 1 lL of salt solution (Invitrogen,

Carlsbad, CA, USA) in a 5-lL reaction volume and incu-

bated for 5 min at room temperature and then placed on ice

as preparing for the transformation in competent TOP10 E.

coli cells. 2 lL of each TOPO cloning reaction was added

to a vial of chemically competent E. coli TOP10 cells and

incubated for 10 min on ice. The cells were heat-shocked

for 30 s at 42 �C and placed on ice for 2 min. 250 lL of

S.O.C. medium was added to each vial of cells and was

shaken (200 rpm) at 37 �C for 1 h. 50 lL of each trans-

formation vial was spread out on pre-warmed culture

Clin Exp Med

123

Page 5: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

dishes containing LB agar (Sigma-Aldrich, St. Louis, MO,

USA), 50 lg/mL Ampicillin (Sigma-Aldrich, St. Louis,

MO, USA) and 50 mg/mL 5-bromo-4-chloro-3-indolyl-b-

D-galactopyranoside (X-gal) (Promega, Madison, WI,

USA). Plates were incubated overnight at 37 �C. Trans-

formants were selected by X-gal-blue-white-screening.

Colonies containing transformants are white or light blue

colored. Colonies containing transformants were picked and

cultivated each in 3 mL LB medium (Sigma-Aldrich, St.

Louis, MO) and 3 lL Ampicillin (50 lg/mL) (Sigma-

Aldrich, St. Louis, MO, USA) overnight at 37 �C. The

Plasmid-Mini-Preparation was carried out with the Fast-

Plasmid Mini Kit (Eppendorf, Hamburg, Germany)

according to the manufacturer 0s instructions. To control the

TOPO TA cloning result, a restriction digest with EcoRI

(FastDigest, Fermentas, Leon-Rot, Germany) was per-

formed, which was analyzed by agarose gel electrophoresis.

Plasmids including the cDNA PCR product were purified

with the QIAquick PCR Purification Kit (Qiagen, Hilden,

Germany) according to the manufacturer 0s instructions.

Sequencing

The cDNA PCR products were sequenced by Sanger

reaction containing 400 ng of cDNA, 10 pmol M13 For-

ward Primer (Invitrogen, Carlsbad, CA, USA) and 4 lL of

Big Dye Sequencing RR-100 (Applied Biosytems, Foster

City, CA, USA) in a total reaction volume of 20 lL. The

PCR conditions included a first step at 96 �C (30 s), 26

cycles at 55 �C (30 s) and 60 �C (4 min). Each reaction

was purified with the DyeEx 2.0 Spin Kit (Qiagen, Hilden,

Germany) according to the manufacturer 0s instructions and

was sequenced on an Applied Biosystems 3130 genetic

analyzer (Applied Biosystems, Foster City, CA, USA).

Sequences were aligned using the VectorNTI software

(Invitrogen, Carlsbad, CA, USA).

Quantitative analysis

Spectratyping was analyzed in two ways. First, the grade of

restriction (normal and restricted) and the size of restricted

peaks were described by visual evaluation. The complexity

of restriction within the Vb families was determined by

counting the number of discrete peaks [34]. In a normal

population of T cells, CDR3 length analysis produces

approximately 5–10 identifiable or discrete peaks spaced by

3 nucleotides, with fluorescence intensity following a

Gaussian-like distribution. If discrete peaks were given but

not shown as Gaussian-like distribution, the spectratyping

profiles were determined as skewed. In cases with no discrete

peaks presented, they were scored as absent. For better

comparability, results were given also in percentages.

Taken into account that visual determination of spectra-

typing profiles suffice merely for semi-quantitative estima-

tions of clonal DNA in distinct peak positions, we performed

a second, quantitative analysis of the spectratyping data, in

order to obtain an objective and sensitive determination of

the magnitude of restriction or skewing of two given Vbfamily CDR3 profiles from a Gaussian-like distribution, the

control sample and the T-cell sample stimulated with blasts

or DC/DCleu. Therefore, the Vb-CDR3 profiles data were

analyzed using the generalized Hamming distance (HD)

analysis as introduced by Currier et al. [35, 36]. Employing a

so-called HD score, the degree of skewing of given Vb-

CDR3 Gaussian-like distributions can be analyzed, and

different distributions can be compared to one another. To

obtain the HD score for a Vb distribution with respect to a

control distribution, for each peak i (of the corresponding

Vb-CDR3 length Si), its area under the peak (Ai) relative to the

area under the whole Vb distribution (Atotal) was compared to

the corresponding value of the control, giving the degree

of skewing at each Vb-CDR3 length, Di = (Ai/Atotal) -

(Acontroli =Acontrol

total ). Summing up all absolute differences of

the frequencies of all CDR3 peaks and dividing the result

by two gives the HD score for the Vb distribution:

HD = � Ri |Di|. A Vb-CDR3 profile identical to the

control has an HD score of 0 %, while a discordant profile

obtains an HD score of 100 %. Two randomly drawn

profiles will have an HD score of 50 %.

Results

Repeated stimulation of allogeneic or autologous

T cells with myeloid blasts or blast-derived DC/DCleu

cause different T-cell response patterns

In most cases, repeated stimulation of T cells with leuke-

mic blasts or blast-derived dendritic cells result in effective

tumor cell lysis. But in some cases, T cells are generated

mediating anergy or even tumor cell growth [20, 31, 37].

To further elucidate the underlying cause for this opposite

T-cell response pattern, we studied the impact of blast- or

DC/DCleu-mediated stimulation on T-cell differentiation in

context of an autologous (#546), human leukocyte antigen

(HLA)-identical (#502) and haplo-identical (#538) experi-

mental setting. Leukemia-derived DC/DCleu from three

different patient samples (#502, #546 and #538; Table 1)

were generated as described. The results are summarized in

Table 2. In all samples tested, CD34 was identified to be

the selected marker with highest expression on blast cells

and CD206 identified to be the selected marker for the

quantification of DC since it was not expressed on naı̈ve

blasts and highest expressed on DC after culture. In patient

Clin Exp Med

123

Page 6: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

sample #502, the Picibanil method turned out to provide

the best results: quantitative DC preparation resulted in

20 % DC, displaying in 70 % a DCleu, in 71 % a mature

and in 48 % a CCR7? DC phenotype. In patient sample

#546, we could generate 21 % DC with Picibanil display-

ing in 45 % DCleu, in 45 % a mature and in 2 % a CCR7?

DC phenotype. In patient sample #538, we succeeded to

generate 15 % DC with MCM-Mimic being the best

method, displaying in 62 % a DCleu, in 48 % a mature and

in 61 % a CCR7? DC phenotype.

To determine the impact of these different stimulator

cells (blasts or DC/DCleu) on the T-cell response pattern,

MLC were set up with the corresponding donor T cells

(#502, HLA-identical (sister); #538, haplo-identical

(mother); #546, (autologous). After a 10-day stimulation

period with blasts or DC/DCleu, respectively, T cells were

harvested and phenotypically characterized as shown in

Fig. 1. In all three cases, more than 95 % viable, 7AAD-

negative T cells were found after stimulation. T-cell pro-

liferation, as to be measured by co-expression of CD71,

increased in all cases during the blast and DC/DCleu

stimulation period. With the exception of patient #546,

where CD45RO? T cells declined to 15 % after blast

stimulation, CD45RO? non-naive T cells increased after

blast and even to a higher extent after DC/DCleu stimula-

tion. Moreover, in the allogeneic setting (patient #502,

patient #538), stimulation with myeloid blasts resulted in

increased proportions of CD8? T cells, whereas stimulation

with DC/DCleu seemed to favor the increase in CD4? T

cells. Although the percentage of T cells expressing che-

mokine receptors varied in the settings compared, stimu-

lation with DC/DCleu caused increased CCR4 expression

levels (Fig. 1). In an attempt to compare the induced T-cell

activities, cytotoxicity assays were performed. In patient

#502, we observed that both, blast- and DC/DCleu-stimu-

lated T cells mediated blast lysis. But interestingly, stim-

ulation with DC/DCleu resulted not only in a more

effective, but also in a more prolonged lytic activity, as

measured after a period of 3 and 24 h of incubation with

target cells (Fig. 2). However, in marked contrast, in

patient #546 and #538, we observed that both, blast- and

DC/DCleu-stimulated T cells resulted in blast proliferation

(Fig. 2). In previous experiments, we found that a

CD4:CD8 ratio of [1 after DC/DCleu stimulation corre-

lated with a cytotoxic activity of the effector cell popula-

tion [24]. In case #502, the calculated CD4:CD8 ratio was

3.1 after DC/DCleu stimulation, and in cases #546 and

#538, the calculated ratio was 1.1 and 0.9, further sup-

porting this previous observation. No correlations could be

established for unstimulated or blast-stimulated T cells.

Blast or DC/DCleu stimulation results into distinctive

TR-Vb profiles

To further dissect the stimulatory impact on the effector

cells as well as to evaluate changes in the amounts of gene

products in Vb subunits in different settings, TR-Vb gene

profiles were analyzed via immunoscope spectratyping.

Results obtained in cases #502, #546 and #538 are sum-

marized in Table 3 and Fig. 3. The restricted repertoires

are highlighted with red circles (Fig. 3). In all three cases,

CD8? T cells showed a more restricted TR repertoire than

CD4? T cells after stimulation, independently of blast or

DC/DCleu used as stimulators. Moreover, with exception of

Table 2 Generation of

leukemia-derived dendritic cells

(DCleu)

DCleu/MNC: percentage of

DCleu in the MNC fraction

DCleu/DC: percentage of DCleu

in the DC fraction

Sample no. #502 #546 #538

A) Determination of the best DCleu conversion conditions

Selected marker profile

Blast CD34 34 34 34

DC CD206 206 206 206

DC pre-culture

(% DC generated) MCM-Mimic 13 15 15

Picibanil 24 21 n.a.

Ca-Ionophore 5 19 7

B) Quantitative results of the DCleu preparation

DC/MNC (%) 20 21 15

Blasts converted/MNC (%) 25 25 27

DCleu/MNC (%) 10 5 5

DCleu/DC (%) 70 45 62

Viable DC/DC (%) 88 75 75

Mature DC/DC (%) 71 45 48

Migratory DC/DC (%) 48 2 61

Clin Exp Med

123

Page 7: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

DC/DCleu-stimulated CD4? T cells from patient #538,

stimulation with DC/DCleu resulted in a higher grade of

restriction than stimulation with blasts (Table 3).

In patient #502, the unstimulated CD4? and CD8? T

lymphocytes derived from the corresponding HLA-matched

donor displayed a normal, non-restricted Gaussian-like

Stimulation with myeloid blasts Stimulation with DC/DCleu

patient #502

patient #546

patient #538

Fig. 1 Positively selected CD3 T cells from the healthy stem cell

donors of patient #502 and patient #538 or autologous T cells

prepared from patient #546 were cocultured for 10 days with

irradiated blasts or DC/DCleu. Subsequently, the antigen expressions

of the CD3 T cells were evaluated by flow cytometry and compared

for their co-expression of CD4, CD8, CD45RA, CD45RO, CCR7 and

CD71 before and after blast and DC/DCleu stimulation. This

contributed to evaluate distinct proportions of proliferating

(CD71?), naı̈ve, non-naı̈ve and central memory T cells in context

of the respective stimulation

Clin Exp Med

123

Page 8: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

distribution of the CDR3 region of all Vb–Jb gene com-

binations of the TR, indicating a normal TR repertoire

(Fig. 3a). After blast or DC/DCleu stimulation, both the

CD4? and the CD8? T-cell subset displayed a significant

TR restriction, but more pronounced for the CD8? T-cell

subset (56/30 peaks in the CD8*blast/CD8*DC/DCleu fraction

vs. 88/36 peaks in the CD4*blast/CD4*DC/DCleu fraction).

Interestingly, the DC/DCleu stimulation induced an even

more skewed TR pattern, affecting both the CD4? and

CD8? T-cell subsets (88 peaks in the CD4*blast fraction vs.

36 peaks in the CD4*DC/DCleu fraction, and 56 peaks in the

CD8*blast fraction versus 30 peaks in the CD8*DC/DCleu

fraction). Moreover, the percental distribution of clonal

DNA showed highest amounts of Vb5.2 DNA (13.9 %)

and 8.3 % of Vb2, Vb5.1, Vb8 and Vb18 DNA in the

CD4*DC/DCleu fractions, whereas the percentages of clonal

DNA stayed B6.8 % in the CD4*blast-stimulated T-cell

subsets (Table 3). Concerning the percental distribution of

clonal DNA in the CD8*DC/DCleu fractions, highest amounts

were found in the Vb18 DNA (13.3 %) and each 10 % in

the Vb5.2, Vb10 and Vb23 DNA, whereas the values

stayed B8.9 % in the CD8*blast fraction (Table 3).

Cytotoxicity after 3 hours Cytotoxicity after 24 hours

patient #502

patient #546

patient #538

Fig. 2 Cytolytic activity of unstimulated versus blast- or DC/DCleu-

stimulated allogeneic (#502; #538) or autologous (#546) T cells.

Antileukemic activity was evaluated via a non-radioactive fluorolysis

assay after 3-and 24-h periods of co-incubation of the corresponding

effector cells and blasts. Positive bars indicate blast lysis, and

negative values display blast growth

Clin Exp Med

123

Page 9: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

Ta

ble

3V

bp

eak

sas

det

ecte

db

yim

mu

no

sco

pe

spec

trat

yp

ing

wit

hin

the

TR

gen

elo

cus

bef

ore

and

afte

rst

imu

lati

on

of

Tce

lls

wit

hm

yel

oid

bla

sts

or

DC

/DC

leu

Vb

1V

b2

Vb

3V

b4

Vb5

.1V

b5

.2V

b6

Vb

7V

b8

Vb

9V

b1

0V

b1

1V

b1

2V

b13

.1

Pat

ien

t#

50

2

CD

4d

ay0

3(2

.1)

7(5

.0)

5(3

.6)

5(3

.6)

4(2

.9)

7(5

.0)

6(4

.3)

6(4

.3)

6(4

.3)

7(5

.0)

3(2

.1)

7(5

.0)

4(2

.9)

7(5

.0)

CD

4b

last

day

10

2(2

.3)

5(5

.7)

3(3

.4)

3(3

.4)

4(4

.6)

5(5

.7)

5(5

.7)

4(4

.6)

3(3

.4)

3(3

.4)

2(2

.3)

3(3

.4)

2(2

.3)

6(6

.8)

CD

4D

C/D

Cle

ud

ay1

00

3(8

.3)

2(5

.6)

2(5

.6)

3(8

.3)

5(1

3.9

)2

(5.6

)0

3(8

.3)

1(2

.8)

1(2

.8)

1(2

.8)

1(2

.8)

1(2

.8)

CD

8d

ay0

4(3

.4)

7(5

.9)

5(4

.2)

4(3

.4)

1(0

.8)

5(4

.2)

5(4

.2)

5(4

.2)

6(5

.1)

6(5

.1)

1(0

.8)

5(4

.1)

4(3

.4)

6(5

.1)

CD

8b

last

day

10

3(5

.4)

4(7

.1)

2(3

.6)

2(3

.6)

1(1

.8)

3(5

.4)

4(7

.1)

2(3

.6)

2(3

.6)

3(5

.4)

2(3

.6)

2(3

.6)

1(1

.8)

4(7

.1)

CD

8D

C/D

Cle

ud

ay1

01

(3.3

)1

(3.3

)1

(3.3

)1

(3.3

)1

(3.3

)3

(10

.0)

1(3

.3)

00

1(3

.3)

3(1

0.0

)0

01

(3.3

)

Pa

tien

t#

54

6

CD

4d

ay0

1(3

.7)

01

(3.7

)0

00

2(7

.4)

1(3

.7)

2(7

.4)

2(7

.4)

01

(3.7

)0

2(7

.4)

CD

4b

last

day

10

3(3

.1)

3(3

.1)

6(6

.1)

2(2

.0)

3(3

.1)

6(6

.1)

5(5

.1)

4(4

.1)

4(4

.1)

6(6

.1)

1(1

.0)

2(2

.0)

3(3

.1)

5(5

.1)

CD

4D

C/D

Cle

ud

ay1

01

(2.6

)2

(5.3

)1

(2.6

)1

(2.6

)3

(7.9

)1

(2.6

)2

(5.3

)1

(2.6

)3

(7.9

)1

(2.6

)3

(7.9

)0

02

(5.3

)

CD

8d

ay0

02

(5.0

)1

(2.5

)1

(2.5

)0

2(5

.0)

3(7

.5)

4(1

0.0

)5

(12

.5)

2(5

.0)

1(2

.5)

00

1(2

.5)

CD

8b

last

day

10

1(1

.9)

1(1

.9)

2(3

.9)

1(1

.9)

03

(5.8

)4

(7.7

)5

(9.6

)5

(9.6

)2

(3.9

)2

(3.9

)0

03

(5.8

)

CD

8D

C/D

Cle

ud

ay1

01

(2.9

)2

(5.9

)1

(2.9

)2

(5.9

)1

(2.9

)1

(2.9

)3

(8.8

)2

(5.9

)2

(5.9

)4

(11

.8)

00

04

(11

.8)

Pat

ien

t#

53

8

CD

4d

ay0

5(1

7.8

)1

(3.6

)2

(7.1

)2

(7.1

)0

01

(3.6

)5

(17

.8)

03

(10

.7)

3(1

0.7

)0

00

CD

4b

last

day

10

1(3

.1)

3(9

.4)

2(6

.3)

00

04

(12

.5)

4(1

2.5

)4

(12

.5)

00

00

3(9

.4)

CD

4D

C/D

Cle

ud

ay1

00

1(2

.1)

1(2

.1)

00

2(4

.3)

4(8

.5)

1(2

.1)

1(2

.1)

3(6

.4)

04

(8.5

)4

(8.5

)3

(6.4

)

CD

8d

ay0

1(8

.3)

2(1

6.6

)2

(16

.6)

00

2(1

6.6

)2

(16

.6)

01

(8.3

)0

00

00

CD

8b

last

day

10

00

2(1

2.5

)2

(12

.5)

00

01

(6.3

)0

01

(6.3

)0

05

(31

.3)

CD

8D

C/D

Cle

ud

ay1

00

02

(28

.6)

00

00

01

(14

.3)

00

00

2(2

8.6

)

Vb

13

.2V

b1

4V

b1

5V

b1

6V

b1

7V

b1

8V

b19

Vb

20

Vb

21

Vb

22

Vb

23

Vb

24

Sco

reS

core

(%)

Rat

io

Pat

ien

t#

50

2

CD

4d

ay0

6(4

.3)

6(4

.3)

5(3

.6)

6(4

.3)

6(4

.3)

5(3

.6)

1(0

.7)

4(2

.9)

6(4

.3)

7(5

.0)

5(3

.6)

6(4

.3)

14

01

00

CD

4b

last

day

10

5(5

.7)

5(5

.7)

3(3

.4)

3(3

.4)

5(5

.7)

4(4

.6)

1(1

.1)

02

(2.3

)5

(5.7

)4

(4.6

)1

(1.1

)8

86

32

.4

CD

4D

C/D

Cle

ud

ay1

00

1(2

.8)

01

(2.8

)1

(2.8

)3

(8.3

)0

01

(2.8

)2

(5.6

)1

(2.8

)1

(2.8

)3

62

6

CD

8d

ay0

5(4

.1)

7(5

.9)

3(2

.5)

6(5

.1)

6(5

.1)

3(2

.5)

5(4

.1)

1(0

.8)

3(2

.5)

6(5

.1)

5(4

.2)

5(4

.1)

11

91

00

CD

8b

last

day

10

2(3

.6)

5(8

.9)

01

(1.8

)5

(8.9

)0

00

2(3

.6)

3(5

.4)

3(5

.4)

05

64

71

.9

CD

8D

C/D

Cle

ud

ay1

01

(3.3

)0

2(6

.7)

01

(3.3

)4

(13

.3)

00

2(6

.7)

1(3

.3)

3(1

0.0

)2

(6.7

)3

02

5

Pat

ien

t#

54

6

CD

4d

ay0

04

(14

.8)

00

3(1

1.1

)1

(3.7

)2

(7.4

)1

(3.7

)0

2(7

.4)

1(3

.7)

1(3

.7)

27

10

0

CD

4b

last

day

10

)5

(5.1

)s

(5.1

)4

(4.1

)3

(3.1

)3

(3.1

)3

(3.1

)1

(1.0

)4

(4.1

)1

(1.0

)6

(6.1

)5

(5.1

)5

(5.1

)9

83

62

2.6

CD

4D

C/D

Cle

ud

ay1

00

5(1

3.2

)0

01

(2.6

)2

(5.3

)1

(2.6

)2

(5.3

)0

2(5

.3)

2(5

.3)

2(5

.3)

38

14

0

CD

8d

ay0

02

(5.0

)1

(2.5

)1

(2.5

)3

(7.5

)2

(5.0

)1

(2.5

)2

(5.0

)0

2(5

.0)

3(7

.5)

1(2

.5)

40

10

0

CD

8b

last

day

10

3(5

.8)

1(1

.9)

1(1

.9)

3(5

.8)

2(3

.9)

3(5

.8)

02

(3.9

)0

1(1

.9)

4(7

.7)

3(5

.8)

52

13

01

.5

Clin Exp Med

123

Page 10: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

The autologous T lymphocytes of case #546 were col-

lected at first diagnosis and presented with a restricted TR-

Vb repertoire in CD4? and CD8? T-cell subset repertoire

(Fig. 3b). After stimulation an increase of the TR reper-

toire was observed. But again, stimulation with DC/DCleu

correlated with a higher grade of TR restriction than

stimulation with blasts and affected to a higher extend

the CD8? T-cell subset (98 peaks in the CD4*blast frac-

tion versus 38 peaks in the CD4*DC/DCleu fraction, and 52

peaks in the CD8*blast fraction versus 34 peaks in the

CD8*DC/DCleu fraction). The percental distribution of the

clonal DNA showed 13.2 % of Vb14 and high amounts of

Vb5.1, Vb8 and Vb10 (7.9 % each) after DC/DCleu stim-

ulation of CD4? T cells, whereas the percentages of clonal

DNA stayed B6.1 % in the blast-stimulated subset. Perc-

ental distributions of clonal DNA in the CD8? T-cell

fraction showed 9.6 % in Vb7 and Vb8 DNA after blast

stimulation. After DC/DCleu stimulation, the CD8? T-cell

fraction was found to have highest restricted DNA clones

in Vb9 and Vb13.1 (11.8 % each) and Vb6 and Vb24

(8.8 % each).

Unstimulated CD4? and CD8? T lymphocytes derived

from the HLA haplo-identical donor of patient #538

showed in both T-cell subsets a normal Gaussian-like

distribution of the Vb family TR repertoire (Fig. 3c). After

blast stimulation, the CD4? T-cell subset revealed a

slightly more restricted Vb-TR pattern as compared to the

CD4? T-cell subset stimulated with DC/DCleu (32 peaks in

the CD4*blast fraction vs. 47 peaks in the CD4*DC/DCleu

fraction). The CD8? T-cell subset, however, showed again

a more restricted Vb repertoire after DC/DCleu stimulation

(16 peaks in the CD8*blast fraction vs. 7 peaks in the

CD8*DC/DCleu fraction). The percental distribution in the

CD4 subset after blast stimulation showed highest amounts

of clonal DNA for Vb16 (15.6 %) and Vb6, Vb7 and Vb8

DNA (12.5 % each). After DC/DCleu stimulation, DNA

was distributed to Vb14 (10.6 %), and Vb6, Vb11, Vb12

and Vb24 (8.5% each). Within the CD8*blast fraction,

clonal DNA was highly distributed to Vb13.1 (31.3 %),

and Vb3, Vb4 and Vb18 (12.5 % each). After DC/DCleu

stimulation, Vb3 and Vb13.1 are presented with 28.6 %

and Vb8, Vb18 and Vb22 with 14.3 % of the clonal DNA

(Table 3). In summary, the strongest Vb-TR restriction

was found within the CD8? T-cell subset after stimulation

with DC/DCleu, independent of the functional activity of

the involved T cells in all three patients.

Hamming distance displays disparity of the Vb profile

restriction in dependence of blast or DCleu stimulation

To quantify the degree of Vb-TR restriction also in an

objective and sensitive manner, Hamming distance (HD)

scores were calculated for the determined spectratypeTa

ble

3co

nti

nu

ed

Vb

13

.2V

b1

4V

b1

5V

b1

6V

b1

7V

b1

8V

b19

Vb

20

Vb

21

Vb

22

Vb

23

Vb

24

Sco

reS

core

(%)

Rat

io

CD

8D

C/D

Cle

ud

ay1

00

1(2

.9)

02

(5.9

)0

01

(2.9

)0

02

(5.9

)2

(5.9

)3

(8.8

)3

48

5

Pat

ien

t#

53

8

CD

4d

ay0

1(3

.6)

1(3

.6)

00

01

(3.6

)0

03

(10

.7)

00

01

36

10

0

CD

4b

last

day

10

00

1(3

.1)

5(1

5.6

)0

01

(3.1

)1

(3.1

)0

02

(6.3

)1

(3.1

)3

22

40

.7

CD

4D

C/D

Cle

ud

ay1

03

(6.4

)5

(10

.6)

00

1(2

.1)

03

(6.4

)3

(6.4

)0

3(6

.4)

1(2

.1)

4(8

.5)

47

35

CD

8d

ay0

00

01

(8.3

)0

1(8

.3)

00

00

00

12

91

00

CD

8b

last

day

10

1(6

.3)

00

1(6

.3)

02

(12

.5)

00

00

1(6

.3)

01

61

22

.3

CD

8D

C/D

Cle

ud

ay1

00

00

00

1(1

4.3

)0

00

1(1

4.3

)0

07

5

Pea

kco

un

tsw

ith

inV

bfa

mil

ies

of

po

siti

vel

yse

lect

edC

D4

?an

dC

D8

?T

cell

sar

ep

rese

nte

db

efo

re(‘

day

0’)

or

afte

rst

imu

lati

on

for

10

day

s(‘

day

10

’).P

eak

so

fea

chV

bfa

mil

yar

ep

rese

nte

din

nu

mb

ers,

resp

ecti

vel

y,

by

cou

nti

ng

all

pea

ks

for

each

Vb

fam

ily

inb

rack

ets

asp

erce

nta

ge

refe

rrin

gto

the

tota

ln

um

ber

of

pea

ks.

Th

eo

ver

all

cou

nts

of

the

pea

ks

are

sum

mar

ized

asa

sco

re.

Th

e

rati

oo

fb

last

and

DC

/DC

leu

stim

ula

tio

nex

pre

sses

the

rela

tiv

ere

stri

ctio

no

fth

eT

Rre

per

toir

ew

ith

inth

eco

rres

po

nd

ing

T-c

ell

sub

set

Th

eb

old

face

dv

alu

esin

dic

ate

the

Vb

fam

ilie

sw

ith

the

hig

hes

tp

eak

cou

nts

and

corr

esp

on

din

gp

erce

nta

ge

val

ues

wit

hin

the

spec

ific

frac

tio

ns

Clin Exp Med

123

Page 11: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

(A) patient #502

CD4+ T cell fractions day 0 (w/o stimulation) day 10 (stimulation blast) d10 (DC/DCleustimulation

base pair position →

CD8+ T cell fractions

day 0 (w/o stimulation) day 10 (stimulation blast) day 10 (DC/DCleustimulation

base pair position →

fluo

resc

ence

inte

nsit

y →

fluo

resc

ence

inte

nsit

y →

Fig. 3 T-cell receptor Vb families 3 and 13.1 with the corresponding

Jb families (1.1–2.7), as determined by immunoscope spectratyping,

are shown for the selected CD4? and CD8? T cells derived from

patients #502, #546 and #538, before and after 10 days of in vitro

stimulation with either blasts or DC/DCleu. The line on the top shows

both Vb3 (first Gaussian-like distribution) and Vb13.1 (second

Gaussian-like distribution) families. The Vb3 family peaks are

located between the 160 and 200 base pair position, whereas the

Vb13.1 family exhibits a base pair length of 220–240. The lines

below show the corresponding Jb families labeled with three different

colors: The second line presents the Jb families 1.1 (green), 1.2 (blue)

and 1.3 (black). In the third line, the Jb families 1.4 (green), 1.5 (blue)

and 1.6 (black) are presented. The forth line involves the Jb families

2.1 (green), 2.2 (blue) and 2.3 (black). In the fifth line, the Jb families

2.4 (green), 2.5 (blue) and 2.6 (black) are shown, and in the bottom

line, the Jb family 2.7 (green) is presented. The red circles indicate

the development of a highly restricted repertoire after both stimula-

tion settings, either with blasts or DCleu (color figure online)

Clin Exp Med

123

Page 12: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

profiles after blast and DCleu stimulation, respectively, and

compared with the corresponding internal control distri-

bution (day 0 w/o stimulation) spanning the total range of

CDR3 lengths.

As exemplarily shown in Fig. 4a (patient #502), HD

scores for the CD4? TR-Vb3 and Vb13.1 family were

calculated to be 38 % and 13 % after blast stimulation, and

46 % and 47 % after DCleu stimulation, respectively,

indicating a higher degree of restriction after DCleu stim-

ulation. Within the CD8? T-cell fraction, HD scores for the

corresponding families were determined to be 51 and 36 %

after blast stimulation, and 50 and 72 % after DCleu stim-

ulation. According to the calculation formula, HD scores of

50 % indicate absence of the Vb profile as compared with

Patient #546 CD4+ T cell fractions

day 0 (w/o stimulation) day 10 (stimulation blast) day 10 (DC/DCleustimulation)

base pair position →

CD8+ T cell fractions day 10 (stimulation blast)day 0 (w/o stimulation) day 10 (DC/DCleustimulation)

base pair position →

fluo

resc

ence

inte

nsit

y →

fluo

resc

ence

inte

nsit

y →

(B)

Fig. 3 continued

Clin Exp Med

123

Page 13: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

the internal control, whereas HD scores of 36 % indicate

intermediate, 51 % strong and 72 % very strong restriction

of the corresponding Vb profiles.

In patient #546 (Fig. 4b), the HD scores for the Vb3 and

Vb13.1 families within the CD4? fraction were calculated

to be 70 and 40 % after stimulation with blasts, and 57 and

16 % after DCleu stimulation. For the CD8? subpopulation,

HD scores for the corresponding families were 31 and

48 % after blast stimulation and 0 and 16 % after DCleu

stimulation, whereas according to calculation formula, an

HD score of 0 % is indicating an identical Vb3 profile, as

compared to the internal control (day 0 w/o stimulation).

Although in this case, the internal control displayed already

a skewed Vb repertoire, further stimulation with DCleu

Patient #538

CD4+ T cell fractions

base pair position →CD8+ T cell fractions

day 0 (w/o stimulation) day 10 (stimulation blast) leustimulation) day 10 (DC/DC

day 0 (w/o stimulation) day 10 (stimulation blast) day 10 (DC/DCleustimulation)

base pair position →

fluo

resc

ence

inte

nsit

y →

fluo

resc

ence

inte

nsit

y →

(C)

Fig. 3 continued

Clin Exp Med

123

Page 14: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

resulted in a more restricted TR-Vb profile as compared to

stimulation with blasts.

For patient #538 (Fig. 4c), the corresponding HD scores

for the CD4? TR-Vb3 and Vb13.1 family were calculated

to be 33 % and 32 % after blast stimulation, and 64 % and

58 % after DCleu stimulation, respectively, indicating a

more skewed repertoire after DCleu stimulation. Within the

CD8? compartment, HD scores for the corresponding

Patient #502day 0 (w/o stimulation) day 10 (stimulation blast) d10 (DC/DCleustimulation)

Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1

CD4+

HD score control control 38 % 13 % 46 % 47 %

CD8+

HD score control control 51 % 36 % 50 % 72 %

Patient #546

day 0 (w/o stimulation) day 10 (stimulation blast) d10 (DC/DCleustimulation)

Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1

CD4+

HD score control control 70 % 40 % 57 % 16 %

CD8+

HD score control control 31 % 48 % 0 % 16 %

Patient #538day 0 (w/o stimulation) day 10 (stimulation blast) d10 (DC/DCleustimulation)

Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1 Vβ 3 Vβ 13.1

CD4+

HD score control control 33 % 32 % 64% 58 %

CD8+

HD score control control 88 % 68 % 32 % 38 %

(A)

(B)

(C)

Fig. 4 Hamming distance scores were calculated for the Vb3 and

Vb13.1 Gaussian-like distributions of all three patients to obtain the

extent of perturbation or skewing at each CDR3 length. The HD score

for each Vb distribution with respect to a control distribution, for each

peak i (of the corresponding Vb-CDR3 length Si), its area under the

peak (Ai) relative to the area under the whole Vb distribution (Atotal)

was compared to the corresponding value of the control, giving the

degree of skewing at each Vb-CDR3 length, Di = (Ai/Atotal) -

(Acontroli =Acontrol

total ). Summing up all absolute differences of the

frequencies of all CDR3 peaks and dividing the result by two gives

the HD score for the Vb distribution: HD = � Ri |Di|

Clin Exp Med

123

Page 15: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

families were found to be 88 and 68 % after blast stimu-

lation, and 32 and 38 % after DCleu stimulation, indicating

a more pronounced TR restriction for the Vb3 and Vb13.1

family after blast stimulation. However, HD score analysis

of the additional Vb families of the CD8 compartment

(data not shown) again supported the observation that

DCleu stimulation resulted in a more restricted Vb profile

than blast stimulation.

Blast or DC/DCleu stimulation results into distinctive

TR-Jb profiles

Additionally, TR-Jb gene profiles were analyzed via im-

munoscope spectratyping. The Jb peak score ranges for an

unstimulated T-cell population between 1,000 and 1,400 Jbpeaks. In all three cases, a restricted Jb repertoire was

observed after blast or DC/DCleu stimulation as summa-

rized in Table 4. With exception of case #538 (337 peaks

in the CD4*blast fraction vs. 569 peaks in the CD4*DC/DCleu

fraction and 344 peaks in the CD8*blast fraction vs. 528

peaks in the CD8*DC/DCleu fraction), again the CD8? T-cell

subset displayed a higher grade of TR restriction as com-

pared to the CD4? T-cell subset. Moreover, stimulation

with DC/DCleu was superior to stimulation with blasts and

caused a more restrictive Jb repertoire in both T-cell sub-

sets. Regarding case #502, the percental distribution of

clonal Jb DNA families showed highest amounts of Jb2.3

and 2.7 in all CD4? and CD8? T-cell fractions. In case

#546, it is shown that the Jb2.3 and Jb2.7 DNA families

were often recombined within their TR b chains in all

CD4? and CD8? T-cell fractions as well. Concerning case

#538, it is shown that the Jb family 2.7 is recombined in all

CD4? and CD8? T-cell fractions for the most part.

Donor T cells stimulated in vitro with DC/DCleu give

rise to one T-cell clone that could also be found in vivo

after allogeneic transplantation

In an attempt to evaluate whether in vitro stimulation might

provide predictive information about potential TR restric-

tion patterns evolving after transplantation, T cells derived

3 months after SCT from patient #538 were analyzed and

compared with the Vb profiles of CD4? and CD8? T cells

as obtained after in vitro stimulation with DC/DCleu. As

shown and exemplified for the Vb3 and Vb13.1 families in

Fig. 5, spectratyping of T cells obtained 96 days after stem

cell transplantation displayed restricted peaks located at the

same base pair positions as found after in vitro stimulation

with DC/DCleu. To ascertain clonal identity, specific

Vb13.1/Jb2.7 PCR products were subcloned and sequence

analysis performed. Out of 220 different CD8? Vb-rear-

ranged T-cell clones generated, screening for identical

sequences revealed the same sequence once in an in vivo

and tenfold in in vitro CD8? T-cell clones after DC/DCleu

stimulation (Fig. 6). Thus, T cells stimulated in vitro with

DC/DCleu gave rise to identical, specifically TR rearranged

T-cell clones as found in vivo after allogeneic stem cell

transplantation.

Discussion

Allogeneic SCT provides a potential curative treatment

option in myeloid leukemia, taking advantage of a graft-

versus-leukemia (GvL) effect mediated by donor lympho-

cytes [3]. Even in recurrent myeloid leukemia after allo-

geneic SCT, subsequent transfusion of donor lymphocytes

can induce sustained remissions in some patients [4, 6]. But

so far, the prerequisite conditions to induce a productive,

leukemia-directed immune response in vivo are not well

understood.

Taken the central role of dendritic cells in adaptive

immunity, we already demonstrated the potential of leu-

kemia-derived dendritic cells (DCleu) to put forward an

effective antileukemic T-cell response. However, in a few

cases, T-cell clones mediating anergy or even blast pro-

liferation could be observed and it already became obvious

that both quantity and quality of the generated DCleus as

well as the induced microenvironment might be crucial for

the induction of a protective T-cell response [20, 31, 37].

In the present report, we tried to further characterize and

delineate the impact of blast- versus DCleu-mediated

stimulation on the evolving T-cell response in context of an

autologous or allogeneic setting. Myeloid blasts were

derived from three patients with active disease before or

after SCT and successfully induced to differentiate in

myeloid leukemia-derived dendritic cells as published

previously [18, 21, 37]. Quantitative conversion of blasts

resulted in 15–21 % DCs, with 45–70 % of these DCs

being characterized to be of leukemic origin (DC/DCleu).

Stimulation with DC/DCleu did not further extend the

T-cell proliferation (range 43–49 %) in the allogeneic

context; however, it was found to be strikingly superior in

the autologous system (#546) as compared to stimulation

with blasts (41 vs. 16 %; Fig. 1), indicating the gain of

immunostimulatory competence of myeloid blasts via

conversion. Thereby, stimulation with DC/DCleu favored

the expansion of CD4?, CCR4? and CD45RO? T cells. In

previous publications, it was hypothesized that a CD4:CD8

T cell ratio [1 after DC/DCleu stimulation might be pre-

dictive and permissive for the induction of an effective

tumor cell lysis [23, 24]. In line with this hypothesis, case

#502 displayed a significant tumor cell lysis. But interest-

ingly, both DC/DCleu- and blast-stimulated T cells resulted

in effective blast lysis. However, DC/DCleu-stimulated T

cells seemed to mediate not only a more efficient lysis than

Clin Exp Med

123

Page 16: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

Ta

ble

4Jb

pea

ks

det

ecte

db

yim

mu

no

sco

pe

spec

trat

yp

ing

of

CD

4?

and

CD

8?

Tce

lls

wer

eco

un

ted

for

each

Jbfa

mil

y,

resp

ecti

vel

y,

bef

ore

(‘d

ay0

’)an

daf

ter

stim

ula

tio

n(‘

day

10

’)w

ith

my

elo

idb

last

so

rD

C/D

Cle

uto

ob

tain

the

sem

i-q

uan

tita

tiv

ean

aly

sis

asp

erfo

rmed

for

the

Vb

fam

ilie

s

Jb1.1

Jb1.2

Jb1.3

Jb1.4

Jb1.5

Jb1.6

Jb2.1

Jb2.2

Jb2.3

Jb2.4

Jb2.5

Jb2.6

Jb2.7

Sco

reS

core

(%)

Rat

io

Pat

ient

#502

CD

4day

099

(9.1

)57

(5.1

)92

(8.2

)22

(2.1

)63

(5.2

)41

(3.2

)114

(10.2

)97

(9.1

)154

(14.1

)26

(2.1

)114

(10.2

)65

(6.1

)127

(11.2

)1071

100

CD

4bla

stday

10

49

(4.2

)45

(4.1

)37

(3.2

)36

(3.1

)45

(4.1

)28

(2.2

)38

(3.2

)23

(2.1

)69

(6.1

)26

(2.1

)51

(4.2

)19

(1.2

)76

(7.1

)542

51

3

CD

4D

C/D

Cle

uday

10

22

(2.1

)6

(0.2

)21

(1.2

)7

(0.2

)3

(0.1

)9

(0.2

)18

(1.2

)0

24

(2.1

)14

(1.1

)15

(1.1

)7

(0.2

)17

(1.2

)163

15

CD

8day

081

(7.2

)57

(5.1

)88

(8.1

)45

(4.1

)57

(5.1

)43

(4.1

)119

(11.1

)50

(4.2

)132

(12.1

)40

(3.2

)26

(2.1

)15

(1.1

)74

(6.2

)827

100

CD

8bla

stday

10

18

(1.2

)21

(1.2

)19

(1.2

)11

(1.1

)9

(0.2

)13

(1.1

)47

(4.1

)16

(1.1

)49

(4.2

)10

(0.2

)17

(1.2

)9

(0.2

)25

(2.1

)264

32

1.9

CD

8D

C/D

Cle

uday

10

1(1

.1)

1(1

.1)

8(0

.2)

1(0

.1)

1(0

.1)

8(0

.2)

21

(1.2

)12

(1.1

)23

(2.1

)10

(0.2

)17

(1.2

)2

(0.1

)15

(1.1

)140

17

Pat

ient

#546

CD

4day

089

(6.2

)112

(8.2

)105

(7.2

)72

(5.1

)86

(6.2

)93

(7.1

)117

(8.2

)83

(6.1

)97

(7.1

)113

(8.2

)127

(9.2

)75

(5.2

)148

(11.1

)1317

100

CD

4bla

stday

10

91

(6.2

)71

(5.1

)88

(6.2

)55

(4.1

)56

(4.1

)59

(4.1

)78

(5.1

)46

(3.1

)96

(7.1

)35

(2.2

)102

(7.2

)54

(4.1

)21

(1.2

)852

65

4.2

CD

4D

C/D

Cle

uday

10

17

(1.1

)26

(1.2

)20

(1.2

)16

(1.1

)9

(0.2

)18

(1.1

)15

(1.1

)4

(0.1

)22

(1.2

)10

(0.2

)19

(1.1

)5

(0.1

)24

(1.2

)205

16

CD

8day

067

(5.1

)94

(7.1

)99

(7.2

)86

(6.2

)75

(5.2

)121

(9.1

)145

(11.1

)69

(5.1

)156

(11.2

)93

(7.1

)83

(6.1

)69

(5.1

)119

(9.1

)1276

100

CD

8bla

stday

10

23

(1.2

)28

(2.1

)28

(2.1

)15

(1.1

)14

(1.1

)21

(1.2

)27

(2.1

)7

(0.2

)41

(3.1

)14

(1.1

)21

(1.2

)14

(1.1

)47

(3.2

)300

23

1.5

CD

8D

C/D

Cle

uday

10

10

(0.2

)18

(1.1

)18

(1.1

)16

(1.1

)12

(0.2

)21

(1.2

)19

(1.1

)1

(0.1

)18

(1.1

)11

(0.2

)18

(1.1

)11

(0.2

)32

(2.1

)205

16

Pat

ient

#538

CD

4day

0107

(7.2

)108

(7.2

)112

(8.1

)93

(6.2

)109

(7.2

)99

(7.1

)127

(9.1

)89

(6.1

)73

(5.1

)90

(6.1

)125

(9.1

)98

(7.1

)157

(11.1

)1387

100

CD

4bla

stday

10

29

(2.1

)24

(1.2

)33

(2.1

)17

(1.1

)10

(0.2

)21

(1.2

)38

(2.2

)13

(0.2

)47

(3.1

)14

(1.1

)26

(1.2

)13

(0.2

)52

(3.2

)337

24

0.6

CD

4D

C/D

Cle

uday

10

44

(3.1

)21

(1.2

)42

(3.1

)21

(1.2

)31

(2.1

)100

(7.1

)43

(3.1

)22

(1.2

)83

(5.2

)3

(0.1

)40

(2.2

)26

(1.2

)93

(6.2

)569

41

CD

8day

093

(6.2

)97

(6.2

)113

(8.1

)108

(7.2

)126

(9.1

)78

(5.2

)114

(8.1

)118

(8.2

)114

(8.1

)107

(7.2

)114

(8.1

)73

(5.1

)121

(8.2

)1376

100

CD

8bla

stday

10

31

(2.1

)19

(1.1

)36

(2.2

)23

(1.2

)17

(1.1

)24

(1.1

)41

(2.2

)7

(0.2

)50

(3.1

)13

(0.2

)19

(1.1

)20

(1.1

)44

(3.1

)344

25

0.7

CD

8D

C/D

Cle

uday

10

58

(4.1

)39

(2.2

)38

(2.2

)27

(1.2

)23

(1.2

)41

(2.2

)46

(3.1

)18

(1.1

)80

(5.2

)15

(1.1

)41

(2.2

)22

(1.2

)80

(5.2

)528

38

Counti

ng

and

sum

min

gup

all

Jbfa

mil

ypea

ks,

resp

ecti

vel

y,

resu

lted

inth

eto

tal

Jbpea

ksc

ore

sfo

rea

chfr

acti

on.

Acc

ord

ing

toth

eto

tal

Jbpea

ksc

ore

s,th

eper

centa

ges

refe

rrin

gto

the

tota

lpea

knum

ber

are

show

nin

bra

cket

s.T

he

rest

rict

ion

of

the

TR

reper

toir

eaf

ter

bla

stan

dD

Cle

ust

imula

tion

isex

pre

ssed

asra

tio

Clin Exp Med

123

Page 17: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

the blast-stimulated T cells (74 vs. 57 %), but also dis-

played a more sustained killing activity (83 vs. 45 %), as

measured after a 3- and 24-h co-incubation with leukemic

target cells (Fig. 2). The importance of cytotoxic CD4?

cells to suppress leukemia colony formation in vitro has

been discussed previously [38, 39] and might be in line

CD4+ T cell fraction in vivo: in vitro:

leu

base pair position →

CD8+ T cell fraction

3 months after SCT 10 days after stimulation with DC/DC

in vivo: in vitro:

leu3 months after SCT 10 days after stimulation with DC/DC

base pair position →

fluo

resc

ence

inte

nsit

y →

fluo

resc

ence

inte

nsit

y →

Fig. 5 Comparison of Vb3 and Vb13.1 T-cell receptor repertoires as

derived from patient samples (#538) 3 months after allogeneic

transplantation (in vivo) and T-cell receptor repertoires as derived

after 10-day in vitro stimulation of the corresponding donor

lymphocytes with DCleu generated from the blasts of the same

patient. The line on the top shows both Vb3 (first Gaussian-like

distribution) and Vb13.1 (second Gaussian-like distribution) families.

The Vb3 family peaks are located between the 160 and 200 base pair

position, whereas the Vb13.1 family exhibits a base pair length of

220–240. The lines below show the corresponding Jb families labeled

with three different colors: The second line presents the Jb families

1.1 (green), 1.2 (blue) and 1.3 (black). In the third line, the Jb families

1.4 (green), 1.5 (blue) and 1.6 (black) are presented. The forth line

involves the Jb families 2.1 (green), 2.2 (blue) and 2.3 (black). In the

fifth line, the Jb families 2.4 (green), 2.5 (blue) and 2.6 (black) are

shown, and in the bottom line, the Jb family 2.7 (green) is presented.

After in vivo and in vitro stimulation identical rearrangements of Vband Jb families with the same CDR3 length could be detected (redcircles). Within the CD8 T-cell fraction, the highlighted circlesdisplay the rearrangement of the Jb family 2.7 with the corresponding

Vb13.1 family, and within the CD4 fraction the Jb family 2.2 with the

corresponding Vb3 family (color figure online)

Clin Exp Med

123

Page 18: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

with the observation that CD8? T cells can be depleted

from the DLT without jeopardizing the GvL effect, while

concurrently diminishing GvHD [40]. So far, it is under

investigation whether in vivo CD4? cells additionally have

to recruit leukemia-reactive, minor H-antigen-specific

CD8? cells to exert effective cytolytic activity [41]. In

cases #546 and #538, the induced effector cell population

displayed a more balanced CD4:CD8 ratio resulting in

anergy or even blast growth. But so far, it is still unclear

and remains speculative whether the failure to mediate

protective immunity was due to a relative increase in CD8?

regulatory T cells controlling an outgrowth of antigen-

triggered CD4? T cells or a relative increase in CD4?

regulatory T cells dominating the CD4? T-cell subset [42,

43]. In marked contrast, the stimulatory DC/DCleu fractions

generated in cases #546 and #538 consisted of a lesser

proportion of mature DCs as well as DCleus as compared to

case #502. This was in line with previous observations,

where decreased proportions of these cells were correlated

with a reduced cytotoxicity of the induced effector cell

population [23, 24].

To further delineate these different response patterns,

the T-cell receptor repertoire of the induced CD4? and

CD8? T-cell subsets were analyzed. Thereby, immuno-

scope spectratyping of the Vb gene families revealed that

stimulation with both blast and DC/DCleu caused a signif-

icant clonal restriction of the TR repertoire within both

T-cell subsets, whereas stimulation with DC/DCleu seemed

to have more pronounced effects. Although the CD8?

T-cell compartment per se displayed the highest grade of

restriction, the CD4? T-cell subset seemed to be relatively

more affected by stimulation with DC/DCleu (Tables 3, 4).

Additional analysis of the Jb gene families displayed a

comparable restriction picture. Again, stimulation with

DC/DCleu fractions induced a relatively higher grade of TR

restriction within the CD4? T-cell subset (Table 4; #502

and #546). This was not the case for patient #538. In this

context, DC/DCleu stimulation displayed in both T-cell

subsets a lesser grade of restriction of the Jb gene families.

This might indicate that in case #538, defined Vb gene

segments were rearranged with a higher diversity of Jbgene segments. Although the frequency of the expressed Jbgene families appeared to be very individual, in both T-cell

subsets rearrangements with Jb2.1, Jb2.3 and Jb2.7 seg-

ments seemed to be preferred [44]. So far, no favored Vb/

Jb TR combinations could be established.

To enable comparability of the various TR-Vb profiles,

spectratype data were additionally converted to frequency

distributions and Hamming distance (HD) scores calcu-

lated, thus resulting in a more objective and sensitive data

interpretation. Interestingly, although in patient #538, HD

scores at first glance suggested blast simulation to be

associated with a more restricted profile than DCleu stim-

ulation within the CD8 compartment, as exemplified in

Fig. 4c for the Vb3 and Vb13.1 families, calculation of the

HD scores for all Vb families further confirmed superiority

of DCleu stimulation in line with visual peak evalution.

However technically, immunoscope spectratyping does

not provide any sequence information about the restricted

TR clones subsumed by the displayed peaks. Thus to

demonstrate the occurrence of genetically identical TR bchain clones, the respective clones have to be sequenced.

Thereby in case #538, a CDR3 sequence pattern rearranged

by the Vb13.1 and Jb2.7 genes was found to be identical

within the CD8? T-cell compartment as generated in vitro

after DC/DCleu-mediated stimulation or derived from the

patient material isolated 3 months after haploidentical

transplantation. In vitro, this specific b chain sequence was

found to be tenfold enriched and might indicate the

potential of this approach to predict TR restriction patterns

in vivo. Unfortunately, due to restricted patient material, no

defined antigen specificity of this identified clone could be

established.

Taken together, our results demonstrate that in vitro

stimulation with DC/DCleu fractions result in TR restriction

patterns that might anticipate evolving T-cell repertoires in

vivo. Subsequent analysis via spectratyping in combination

with additional functional assays will further specify the

induced T-cell response pattern. Thus, Vb/Jb T-cell

selection strategies utilizing clonal TR sequence informa-

tion might pave the way for adoptive, leukemia-directed

T-cell therapy or provide the opportunity to anticipate the

risk of relapse at an early stage while monitoring the

defined T-cell clones during a clinical course.

Acknowledgments This work was supported by grants of the

Deutsche Forschungsgesellschaft (SFB/TR 36) to Hans-Jochem Kolb,

Susanne Reuther and Arndt Borkhardt; R. Buhmann was supported by

Fig. 6 Sequencing of the Vb13.1/Jb2.7 recombination was per-

formed for the potential cytotoxic CD8 fraction according to an

identical restriction pattern found 3 months after transplantation and

after in vitro stimulation with DCleu as shown in Fig. 5. The

nucleotide sequences are translated into amino acid sequences

Clin Exp Med

123

Page 19: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

grants of the EU (Stemdiagnostics/LSHB-CT-2007037703). We are

indebted to many nurses and physicians for their unconditional

assistance in patient care, referral of patient material and data col-

lection. For extended bioinformatic assistance, we are cordially

indebted to Jens Stoye and Nils Hoffmann. Parts of the results pre-

sented in this manuscript were worked out in the doctoral thesis of

Susanne Reuther.

Conflict of interest None.

References

1. Giles FJ, Keating A, Goldstone AH, Avivi I, Willman CL,

Kantarjian HM (2002) Acute myeloid leukemia. Hematol Am

Soc Hematol Educ Program 73–110

2. Buchner T, Hiddemann W, Berdel WE, Wormann B, Schoch C,

Fonatsch C, Loffler H, Haferlach T, Ludwig WD, Maschmeyer G,

Staib P, Aul C, Gruneisen A, Lengfelder E, Frickhofen N, Kern

W, Serve HL, Mesters RM, Sauerland MC, Heinecke A (2003)

6-Thioguanine, cytarabine, and daunorubicin (TAD) and high-

dose cytarabine and mitoxantrone (HAM) for induction, TAD for

consolidation, and either prolonged maintenance by reduced

monthly TAD or TAD-HAM-TAD and one course of intensive

consolidation by sequential HAM in adult patients at all ages with

de novo acute myeloid leukemia (AML): a randomized trial of

the German AML cooperative group. J Clin Oncol 21(24):4496–

4504. doi:10.1200/JCO.2003.02.133

3. Kolb HJ, Schmid C, Barrett AJ, Schendel DJ (2004) Graft-versus-

leukemia reactions in allogeneic chimeras. Blood 103(3):767–

776. doi:10.1182/blood-2003-02-0342

4. Kolb HJ, Mittermuller J, Clemm C, Holler E, Ledderose G,

Brehm G, Heim M, Wilmanns W (1990) Donor leukocyte

transfusions for treatment of recurrent chronic myelogenous

leukemia in marrow transplant patients. Blood 76(12):2462–2465

5. Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B, Jacobsen

N, Arcese W, Ljungman P, Ferrant A, Verdonck L, Niederwieser

D, van Rhee F, Mittermueller J, de Witte T, Holler E, Ansari H

(1995) Graft-versus-leukemia effect of donor lymphocyte trans-

fusions in marrow grafted patients. Blood 86(5):2041–2050

6. Schmid C, Schleuning M, Schwerdtfeger R, Hertenstein B,

Mischak-Weissinger E, Bunjes D, Harsdorf SV, Scheid C, Hol-

tick U, Greinix H, Keil F, Schneider B, Sandherr M, Bug G,

Tischer J, Ledderose G, Hallek M, Hiddemann W, Kolb HJ

(2006) Long-term survival in refractory acute myeloid leukemia

after sequential treatment with chemotherapy and reduced-

intensity conditioning for allogeneic stem cell transplantation.

Blood 108(3):1092–1099. doi:10.1182/blood-2005-10-4165

7. Karp SE, Farber A, Salo JC, Hwu P, Jaffe G, Asher AL, Shiloni

E, Restifo NP, Mule JJ, Rosenberg SA (1993) Cytokine secretion

by genetically modified nonimmunogenic murine fibrosarcoma.

Tumor inhibition by IL-2 but not tumor necrosis factor. J Immu-

nol 150(3):896–908

8. Brossart P (2002) Dendritic cells in vaccination therapies of

malignant diseases. Transfus Apher Sci 27(2):183–186

9. Cella M, Sallusto F, Lanzavecchia A (1997) Origin, maturation

and antigen presenting function of dendritic cells. Curr Opin

Immunol 9(1):10–16

10. Nestle FO, Banchereau J, Hart D (2001) Dendritic cells: on the

move from bench to bedside. Nat Med 7(7):761–765. doi:

10.1038/89863

11. Claxton DF, McMannis J, Champlin R, Choudhury A (2001)

Therapeutic potential of leukemia-derived dendritic cells:

preclinical and clinical progress. Crit Rev Immunol 21(1–3):

147–155

12. Stripecke R, Levine AM, Pullarkat V, Cardoso AA (2002)

Immunotherapy with acute leukemia cells modified into antigen-

presenting cells: ex vivo culture and gene transfer methods.

Leukemia 16(10):1974–1983. doi:10.1038/sj.leu.2402701

13. Caux C, Massacrier C, Vanbervliet B, Dubois B, de Saint-Vis B,

Dezutter-Dambuyant C, Jacquet C, Schmitt D, Banchereau J

(1997) CD34 ? hematopoietic progenitors from human cord

blood differentiate along two independent dendritic cell pathways

in response to GM-CSF ? TNF alpha. Adv Exp Med Biol

417:21–25

14. Choudhury BA, Liang JC, Thomas EK, Flores-Romo L, Xie QS,

Agusala K, Sutaria S, Sinha I, Champlin RE, Claxton DF (1999)

Dendritic cells derived in vitro from acute myelogenous leukemia

cells stimulate autologous, antileukemic T-cell responses. Blood

93(3):780–786

15. Lee AW, Truong T, Bickham K, Fonteneau JF, Larsson M, Da

Silva I, Somersan S, Thomas EK, Bhardwaj N (2002) A clinical

grade cocktail of cytokines and PGE2 results in uniform matu-

ration of human monocyte-derived dendritic cells: implications

for immunotherapy. Vaccine 20(suppl 4):A8–A22

16. Houtenbos I, Westers TM, Stam AG, de Gruijl TD, Scheper RJ,

Ossenkoppele GJ, van de Loosdrecht AA (2003) Serum-free

generation of antigen presenting cells from acute myeloid leu-

kaemic blasts for active specific immunisation. Cancer Immunol

Immunother 52(7):455–462. doi:10.1007/s00262-003-0389-4

17. Westers TM, Stam AG, Scheper RJ, Regelink JC, Nieuwint AW,

Schuurhuis GJ, van de Loosdrecht AA, Ossenkoppele GJ (2003)

Rapid generation of antigen-presenting cells from leukaemic

blasts in acute myeloid leukaemia. Cancer Immunol Immunother

52(1):17–27. doi:10.1007/s00262-002-0316-0

18. Sato M, Takayama T, Tanaka H, Konishi J, Suzuki T, Kaiga T,

Tahara H (2003) Generation of mature dendritic cells fully

capable of T helper type 1 polarization using OK-432 combined

with prostaglandin E(2). Cancer Sci 94(12):1091–1098

19. Kufner S, Fleischer RP, Kroell T, Schmid C, Zitzelsberger H,

Salih H, de Valle F, Treder W, Schmetzer HM (2005) Serum-free

generation and quantification of functionally active Leukemia-

derived DC is possible from malignant blasts in acute myeloid

leukemia and myelodysplastic syndromes. Cancer Immunol Im-

munother 54(10):953–970. doi:10.1007/s00262-004-0657-y

20. Kremser A, Dressig J, Grabrucker C, Liepert A, Kroell T, Scholl

N, Schmid C, Tischer J, Kufner S, Salih H, Kolb HJ, Schmetzer H

(2010) Dendritic cells (DCs) can be successfully generated from

leukemic blasts in individual patients with AML or MDS: an

evaluation of different methods. J Immunother 33(2):185–199.

doi:10.1097/CJI.0b013e3181b8f4ce

21. Dreyssig J, Kremser A, Liepert A, Grabrucker C, Freudenreich

M, Schmid C, Kroell T, Scholl N, Tischer J, Kufner S, Salih H,

Kolb HJ, Schmetzer HM (2011) Various ‘dendritic cell antigens’

are already expressed on uncultured blasts in acute myeloid

leukemia and myelodysplastic syndromes. Immunotherapy

3(9):1113–1124. doi:10.2217/imt.11.108

22. Schmetzer HM, Kremser A, Loibl J, Kroell T, Kolb HJ (2007)

Quantification of ex vivo generated dendritic cells (DC) and

leukemia-derived DC contributes to estimate the quality of DC, to

detect optimal DC-generating methods or to optimize DC-medi-

ated T-cell-activation-procedures ex vivo or in vivo. Leukemia

21(6):1338–1341. doi:10.1038/sj.leu.2404639

23. Grabrucker C, Liepert A, Dreyig J, Kremser A, Kroell T, Freu-

denreich M, Schmid C, Schweiger C, Tischer J, Kolb HJ, Sch-

metzer H (2010) The quality and quantity of leukemia-derived

dendritic cells from patients with acute myeloid leukemia and

myelodysplastic syndrome are a predictive factor for the lytic

Clin Exp Med

123

Page 20: In vitro-induced response patterns of antileukemic T cells: characterization by spectratyping and immunophenotyping

potential of dendritic cells-primed leukemia-specific T cells. J Im-

munother 33(5):523–537. doi:10.1097/CJI.0b013e3181d87ffd

24. Liepert A, Grabrucker C, Kremser A, Dreyssig J, Ansprenger C,

Freudenreich M, Kroell T, Reibke R, Tischer J, Schweiger C,

Schmid C, Kolb HJ, Schmetzer H (2010) Quality of T-cells after

stimulation with leukemia-derived dendritic cells (DC) from

patients with acute myeloid leukemia (AML) or myeloid dys-

plastic syndrome (MDS) is predictive for their leukemia cytotoxic

potential. Cell Immunol 265(1):23–30. doi:10.1016/j.cellimm.

2010.06.009

25. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ,

Porwit A, Harris NL, Le Beau MM, Hellstrom-Lindberg E,

Tefferi A, Bloomfield CD (2009) The 2008 revision of the World

Health Organization (WHO) classification of myeloid neoplasms

and acute leukemia: rationale and important changes. Blood

114(5):937–951. doi:10.1182/blood-2009-03-209262

26. Mitelman F (ed) (1995) ISCN 1995 guidelines for cancer cyto-

genetics, supplement to: an international system for human

cytogenetic nomenclature. S Karger, Basel, Switzerland

27. Schuster FR, Buhmann R, Reuther S, Hubner B, Grabrucker C,

Liepert A, Reibke R, Lichtner P, Yang T, Kroell T, Kolb HJ,

Borkhardt A, Schmetzer H (2008) Improved effector function of

leukemia-specific T-lymphocyte clones trained with AML-derived

dendritic cells. Cancer Genomics Proteomics 5(5):275–286

28. Lanzavecchia A, Sallusto F (2002) Progressive differentiation

and selection of the fittest in the immune response. Nat Rev

Immunol 2(12):982–987. doi:10.1038/nri959

29. Nguyen XD, Eichler H, Dugrillon A, Piechaczek C, Braun M,

Kluter H (2003) Flow cytometric analysis of T cell proliferation

in a mixed lymphocyte reaction with dendritic cells. J Immunol

Methods 275(1–2):57–68

30. Kienzle N, Olver S, Buttigieg K, Kelso A (2002) The fluorolysis

assay, a highly sensitive method for measuring the cytolytic

activity of T cells at very low numbers. J Immunol Methods

267(2):99–108

31. Kufner S, Zitzelsberger H, Kroell T, Pelka-Fleischer R, Salem A,

de Valle F, Schweiger C, Nuessler V, Schmid C, Kolb HJ, Sch-

metzer HM (2005) Leukemia-derived dendritic cells can be

generated from blood or bone marrow cells from patients with

acute myeloid leukaemia: a methodological approach under

serum-free culture conditions. Scand J Immunol 62(1):86–98.

doi:10.1111/j.1365-3083.2005.01630.x

32. Monteiro J, Hingorani R, Peroglizzi R, Apatoff B, Gregersen PK

(1996) Oligoclonality of CD8 ? T cells in multiple sclerosis.

Autoimmunity 23(2):127–138

33. Puisieux I, Even J, Pannetier C, Jotereau F, Favrot M, Kourilsky

P (1994) Oligoclonality of tumor-infiltrating lymphocytes from

human melanomas. J Immunol 153(6):2807–2818

34. Lu J, Basu A, Melenhorst JJ, Young NS, Brown KE (2004)

Analysis of T-cell repertoire in hepatitis-associated aplastic

anemia. Blood 103(12):4588–4593

35. Currier JR, Robinson MA (2001) Spectratype/immunoscope

analysis of the expressed TCR repertoire. Curr Protoc Immunol

Chapter 10 Unit 10 28. doi:10.1002/0471142735.im1028s38

36. Currier JR, Stevenson KS, Kehn PJ, Zheng K, Hirsch VM, Rob-

inson MA (1999) Contributions of CD4 ? , CD8 ? , and

CD4 ? CD8 ? T cells to skewing within the peripheral T cell

receptor beta chain repertoire of healthy macaques. Hum Immunol

60(3):209–222. doi:org/10.1016/S0198-8859(98)00109-8

37. Schmetzer H, Liepert A, Grabrucker C, Kremser A, Loibl J,

Schmid C, Buhmann R, Yang T, Kroell T, Treder W, Kolb H

(2007) Role of the quality and quantity of leukemia-derived de-

dritic cells and anti-leukemia-directed T cells to predict the

course and success of immunotherapy. Bone Marrow Transpl

39(1):O302

38. Childs R, Chernoff A, Contentin N, Bahceci E, Schrump D,

Leitman S, Read EJ, Tisdale J, Dunbar C, Linehan WM, Young

NS, Barrett AJ (2000) Regression of metastatic renal-cell carci-

noma after nonmyeloablative allogeneic peripheral-blood stem-

cell transplantation. N Engl J Med 343(11):750–758

39. Childs RW (2000) Nonmyeloablative allogeneic peripheral blood

stem-cell transplantation as immunotherapy for malignant dis-

eases. Cancer J 6(3):179–187

40. Giralt S, Hester J, Huh Y, Hirsch-Ginsberg C, Rondon G, Seong

D, Lee M, Gajewski J, Van Besien K, Khouri I, Mehra R, Prz-

epiorka D, Korbling M, Talpaz M, Kantarjian H, Fischer H,

Deisseroth A, Champlin R (1995) CD8-depleted donor lympho-

cyte infusion as treatment for relapsed chronic myelogenous

leukemia after allogeneic bone marrow transplantation. Blood

86(11):4337–4343

41. Zorn E, Wang KS, Hochberg EP, Canning C, Alyea EP, Soiffer

RJ, Ritz J (2002) Infusion of CD4 ? donor lymphocytes induces

the expansion of CD8 ? donor T cells with cytolytic activity

directed against recipient hematopoietic cells. Clin Cancer Res

8(7):2052–2060

42. Chess L, Jiang H (2004) Resurrecting CD8 ? suppressor T cells.

Nat Immunol 5(5):469–471. doi:10.1038/ni0504-469ni0504-469

43. Jiang H, Chess L (2006) Regulation of immune responses by T cells.

N Engl J Med 354(11):1166–1176. doi:10.1056/NEJMra055446

44. Freeman JD, Warren RL, Webb JR, Nelson BH, Holt RA (2009)

Profiling the T-cell receptor beta-chain repertoire by massively

parallel sequencing. Genome Res 19(10):1817–1824. doi:

10.1101/gr.092924.109

Clin Exp Med

123