Biology Reference
In-Depth Information
method for the generation of cytokine-polarized human CD4 + T cells in
rapamycin, as shown in Figure 11.3 . This method has several potential
advantages relative to our initial method. First, T cells are generated after a
single round of costimulation, which greatly increases the feasibility of the
culture method; after only 12 days in culture, CD4 + T cells are rendered rapa-
mycin-resistant and cytokine polarized, with an increase in Th2 relative to
Th1 composition, as defined by an approximate 5:1 ratio of T cells express-
ing GATA-3 and T-bet [99] ( Figure 11.3 ); of interest, manufactured T-cell
expression of FOXP3 was minimal, which is a result that stands somewhat
in contrast to other observations that rapamycin can promote the expan-
sion of Treg cells [76] . In addition, because of the reduced time in culture
and the influence of rapamycin, manufactured T cells express a minimally
differentiated phenotype, as indicated by low levels of cytokine secretion
at completion of T-cell manufacturing. However, as shown in Figure 11.3 ,
further expansion of the T-cell product without polarizing cytokines or
rapamycin demonstrates the significant cytokine secretion potential of the
manufactured product; because both the transcription factor analysis and
the cytokine secretion profile indicate a mix of Th2 and Th1 cells, we have
used the term “T-Rapa” cells to describe the manufactured product.
238
The ex vivo manufacturing of mixed Th2/Th1 cells in rapamycin carries the
feasibility disadvantage of limited T-cell expansion during culture, with a
typical culture yielding only a few-fold increase in CD4 cell number relative
to input cell number. Nonetheless, CD4 cell harvest from a 5- to 10-liter
apheresis product will allow T-Rapa cell therapy at the intermediate dose
established in our initial clinical trial, 2.5 × 10 7 manufactured cells/kg. This
second-generation clinical trial (NCT00074490), in addition to evaluating a
new method of T-cell manufacturing, is also evaluating a new post-trans-
plant GVHD chemoprophylaxis regimen consisting of cyclosporin plus
a short course of Sirolimus (through day 14 post-transplant) ( Figure 11.3
schema). In our murine studies [78] , we found that rapamycin-resistant T
cells did not necessarily exhibit resistance to rapamycin in vivo; we specu-
late that these observations may relate to the inhibitory effect of rapamy-
cin on host or donor APCs [76] (see Chapter 9) that drive T-cell expansion
in vivo. In addition, this immune-suppression strategy places dual-agent
suppression on the unmanipulated T cells contained in the hematopoietic
cell allograft and single-agent cyclosporin suppression on the manufac-
tured product, which is administered at day 14 post-transplant as a preemp-
tive donor lymphocyte infusion (DLI), thereby creating a selective pressure
favoring immune responses generated from the T-Rapa cells.
In addition, as outlined in Figure 11.3 , we have evaluated the day 14 post-
transplant T-Rapa cell DLI in the context of a low-intensity chemotherapy
regimen that uses a cyclophosphamide dose that is 75% reduced relative
to our first-generation clinical trial [97] . Use of this low-intensity regimen
offers both potential therapeutic advantages (reduced toxicity in heavily
pretreated patients with comorbid conditions and chemotherapy refractory
disease) and a new opportunity to evaluate the biologic activity of T-Rapa
cells (ability to convert mixed chimerism). Indeed, using this platform, we
found that each of 40 transplant recipients had mixed donor/host chime-
rism in both T-cell and myeloid cell lineages at day 14 post-transplant prior
to T-Rapa cell infusion [100] . Then, within 2 weeks of T-Rapa cell infusion,
Search WWH ::




Custom Search