• No results found

Adoptive immunotherapy after HLA mismatched stem cell transplantation Oosten, L.E.M.

N/A
N/A
Protected

Academic year: 2021

Share "Adoptive immunotherapy after HLA mismatched stem cell transplantation Oosten, L.E.M."

Copied!
7
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Adoptive immunotherapy after HLA mismatched stem cell

transplantation

Oosten, L.E.M.

Citation

Oosten, L. E. M. (2007, November 21). Adoptive immunotherapy after HLA

mismatched stem cell transplantation. Retrieved from

https://hdl.handle.net/1887/12446

Version: Corrected Publisher’s Version

License: Licence agreement concerning inclusion of doctoral

thesis in the Institutional Repository of the University

of Leiden

Downloaded from: https://hdl.handle.net/1887/12446

Note: To cite this publication please use the final published version (if

applicable).

(2)

123

Summary

(3)
(4)

Summary

125 Allogeneic stem cell transplantation (SCT) is a well established treatment for a

variety of hematological malignancies. After SCT, donor-derived T cells present in the stem cell graft elicit immune responses that mediate both beneficial graft-versus- leukemia effects (GvL) and detrimental graft-versus-host disease (GvHD). The best stem cell donor in terms of minimal GvHD is a human leukocyte antigen (HLA) -identical sibling, but for many patients such donors are not available. SCT across HLA mismatches requires T cell depletion to minimize GvHD. However, because T cell depletion also reduces GvL, the risk of leukemia relapse after SCT is significantly increased. Leukemia relapse after HLA-matched SCT can be treated with donor lymphocyte infusions (DLI). Unfortunately, DLI therapy is often accompanied by GvHD as the donor T cells have not been selected for preferential reactivity against the patient’s malignant cells. The use of DLI after HLA-mismatched SCT is associated with severe GvHD-related morbidity and mortality.

An attractive strategy to treat relapsed leukemia after HLA-mismatched SCT comprises adoptive immunotherapy with ex vivo generated donor T cells that display reactivity limited to the patient’s residual leukemic or hematopoietic cells. This may be achieved by employing the patient’s mismatched HLA molecules as antigen-presenting molecules for hematopoietic system-specifi c antigens to generate non-self- (allo) HLA-restricted T cells from the stem cell donor. The minor histocompatibility antigens (mHags) HA-1 and HA-2 are suitable target antigens as they display hematopoietic system-restricted tissue distribution and relevant expression on leukemic cells and their progenitors.

In theory, alloHLA-restricted mHag-specifi c cytotoxic T cells should lyse the patient’s hematopoietic cells, including leukemic cells, but not non-hematopoietic cells that do not express the relevant mHag. Likewise, graft-derived donor hematopoietic cells, which lack the appropriate HLA restriction molecule, will not be affected. Current protocols for the generation of alloHLA-restricted cytotoxic T cells are complex and associated with a high risk of adventitious expansion of broad alloHLA-specifi c T cells that are potentially harmful to the patient. In this thesis, we explored the alloHLA-reactive T cell repertoire and investigated several new strategies to generate HA-1- and HA-2-specifi c alloHLA-A2- restricted T cells.

In chapter 2, we investigated whether artifi cial antigen-presenting constructs (aAPCs) could replace professional antigen-presenting cells (APCs) for the generation of mHag-specifi c T cells. To that end, we coated cell-sized latex beads with recombinant HLA-A2/mHag complexes and the costimulatory molecules CD80 and CD54. These aAPCs effectively stimulated mHag-specifi c T cell clones in a ligand-density dependent manner and could be used to enrich polyclonal HA-1-specifi c T cell lines without affecting T cell phenotype or cytolytic activity. In addition, these aAPCs did not stimulate broad alloHLA-A2-reactive T cells (data incorporated in chapter 4). Thus, HLA-A2/mHag complex-coated aAPCs can be exploited in vitro to promote selective outgrowth of mHag-specifi c alloHLA-restricted T cells present in a pool of alloHLA-reactive T cells.

(5)

Adoptive Immunotherapy after HLA-mismatched Stem Cell Transplantation

126

In chapter 3, we examined whether APCs transduced with a transporter associated with antigen processing (TAP) -inhibiting protein could serve as antigen-specifi c stimulators for mHag-specifi c T cells. Inhibition of TAP signifi cantly reduces endogenous antigen presentation but does not impair exogenous peptide loading. APCs transduced with the herpesvirus-derived proteins US6, ICP47, or UL49.5, that are known to inhibit TAP, exhibited a stable decrease in cell surface HLA class I expression and were protected from lysis by mHag-specifi c T cells. Exogenous addition of mHag peptide fully restored target cell recognition. UL49.5 showed the most pronounced inhibitory effect, reducing cell- surface HLA class I expression and mHag-specifi c lysis by 99%. UL49.5 also signifi cantly diminished alloHLA-reactivities mediated by alloHLA-specifi c T cells. However, reduction of alloHLA-specifi c lysis was not complete. Therefore, inhibition of TAP alone does not reduce an APC’s alloHLA-antigenicity to the extent that adventitious generation of broad alloHLA-reactive T cells would be prevented.

In chapter 4, we aimed to determine whether alloHLA-restricted T cells generated after repeated stimulation with alloHLA-expressing APCs are predominantly peptide-specifi c or peptide-independent. Hereto, we generated alloHLA-A2-reactive T cell populations from HLA-A2neg donors. Peptide-specifi c alloHLA-A2 recognition was determined using a panel of HLA-A2 tetramers representing peptides that are derived from ubiquitously expressed self-proteins. Each alloHLA-A2-reactive T cell population contained distinct T cell subsets binding a single specifi c tetramer. Using the aAPC described in chapter 2, we observed that none of these T cell populations could be stimulated by aAPCs coated with HLA-A2 complexes representing irrelevant peptides, whereas aAPCs coated with a relevant HLA-A2/peptide complex provided adequate stimulation. Thus, the majority of alloHLA- A2-reactive T cell lines, generated using a conventional stimulation protocol, displayed a peptide-specifi c and not a peptide-independent recognition pattern.

In chapter 5, we describe the results of two multimer-based strategies to generate alloHLA- restricted mHag-specifi c T cells. Both methods introduced recombinant HLA-A2/mHag complexes as the sole allogeneic target antigens. In the one strategy, we explored the feasibility of generating mHag-specifi c T cells from HLA-A2neg donors using autologous HLA-A2neg dendritic cells coated with monomeric HLA-A2/mHag complexes. In the other strategy, alloHLA-A2/HA-2-specifi c T cells were directly isolated from the peripheral blood of HLA-A2neg parous females with HLA-A2pos offspring using tetrameric HLA-A2/HA-2 complexes. Both methods yielded T cells with selective tetramer-staining profi les. However, these T cells were either of low avidity or displayed selective, but not solely mHag-specifi c, cytolytic activity. Apparently, alloHLA-restricted T cells tend to recognize HLA/peptide complexes in a peptide-dependent but not peptide-specifi c manner.

In chapter 6, we discuss these fi ndings and conclude that alloHLA-recognition by T cells is inherently crossreactive. A degree of crossreactivity is required to ensure effective pathogen surveillance. AlloHLA-reactivity seems to be the (unwanted) by-product of a crossreactive T cell pool originally selected for reactivity to self-HLA molecules. Consequently, T cell crossreactivity is far more prominent in the HLA-mismatched setting than in the HLA-

(6)

Summary

127 matched setting. Unfortunately, as we show in chapter 5, currently available technologies

can not readily distinguish between crossreactive and antigen-specifi c alloHLA-restricted T cells. We therefore conclude that generation of alloHLA-restricted leukemia- or hematopoietic system antigen-specifi c T cells is as yet not feasible in a clinical setting.

(7)

Referenties

GERELATEERDE DOCUMENTEN

Artifi cial antigen-presenting constructs effi ciently stimulate minor 35 histocompatibility antigen-specifi c cytotoxic T lymphocytes.

Generation of minor histocompatibility antigen HA-1-specifi c cytotoxic T cells restricted by nonself HLA molecules: a potential strategy to treat relapsed leukemia after

Proliferation (A) and IFN-γ production (B) of various HA-1-specifi c CTL clones (2.12, 3HA15, 5W38) and HA-2-specifi c CTL clones (1.7, 1.9) incubated for 48 hours with medium only,

EFFECTS OF US6, ICP47, AND UL49.5 ON M HAG-SPECIFIC TARGET CELL RECOGNITION To determine whether the downregulation of HLA class I cell surface expression resulted in a decrement

Alloreactive CD8 + T cell populations were shown to contain small distinct subsets of T cells that exhibit precise specifi city for peptides bound endogenously by foreign HLA class

SPECIFICITY OF IN VITRO GENERATED ALLO HLA-A2-RESTRICTED HA-1-SPECIFIC T CELLS To further analyze the specifi cities of the alloHLA-A2-restricted HA-1-specifi c T cells generated

Interestingly, one of the isolated T cell clones bound all HLA-A2 tetramers tested with high intensity but showed no lytic activity to HLA-A2 pos target cells whatsoever..

Het bloedcel-specifi eke peptide komt wel tot expressie op de bloedcellen van de patiënt (want die hebben het juiste HLA-molecuul), maar niet op de bloedcellen van de donor (die