Next Article in Journal
Aptamer Cell-Based Selection: Overview and Advances
Next Article in Special Issue
A Critical Appraisal of Extracorporeal Photopheresis as a Treatment Modality for Acute and Chronic Graft-Versus-Host Disease
Previous Article in Journal
Estrogen Repression of MicroRNAs Is Associated with High Guanine Content in the Terminal Loop Sequences of Their Precursors
Previous Article in Special Issue
Mesenchymal Stromal Cells: What Is the Mechanism in Acute Graft-Versus-Host Disease?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Hitting the Holy Grail of Hematopoietic Cell Transplantation with Naive T-Cell Depleted Allografts—Graft Engineered Hematopoietic Stem Cell Transplant

1
Department of Adult Hematology and Stem Cell Transplant, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
2
Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2017, 5(3), 48; https://doi.org/10.3390/biomedicines5030048
Submission received: 18 July 2017 / Revised: 8 August 2017 / Accepted: 11 August 2017 / Published: 14 August 2017
(This article belongs to the Special Issue Cell Therapy for the Treatment of GVHD)

Abstract

:
Hematopoietic cell transplant is a potentially curative procedure for many benign and malignant conditions. The efficacy of allogeneic transplant relies in part on the cytotoxicity of the conditioning regimen and the graft versus tumor effect mediated by alloreactive donor T cells; the same cells are also implicated in the development of graft versus host disease (GVHD). Selective identification and depletion of the T cells implicated in GVHD, while preserving the T cells responsible for graft versus tumor effect has been the focus of many research groups in the recent years. Here we briefly review the physiology of T cells in transplantation, and comment on a recent clinical trial published by Bleakly et al. using a novel way of graft engineered allograft via naïve T cell depletion.

1. Introduction

Hematopoietic cell transplant (HCT) is considered potentially curable for many conditions; however, it comes with the price of a devastating side effect: graft-versus-host disease (GVHD), which is the leading cause of non-relapse mortality and morbidity. Pioneering work done by many investigators in 1970s and 1980s indicated that T-cells mediate potent graft-versus-tumor (GVT) effects, a concept that was later consolidated by large database registry studies in which relapse was found to be higher in identical twins compared to matched donors [1]. Thus, the obvious question, which is still considered to be the Holy Grail in the sixth decade of the HCT field, is how can we preserve GVT without causing GVHD? A recent study by Bleakly et al. in the Journal of Clinical Investigation depicts another novel way of deciphering optimum outcomes in HCT [2].
The preparative regimen for a HCT exerts direct cytotoxicity, whereas the harvested allograft, which is composed of progenitor cells (CD34+), T-cells (CD3+, both αβ and γδ T-cells), B-cells (CD19+), NK cells (CD56+), and dendritic cells (CD14+), can exert potent GVT or cause GVHD [3]. The T-cells exist in various states of maturation and activation; the memory T-cells are CD45RO positive, the naïve T-cells are CD45RA positive [3,4,5,6]. When activated, the naïve T-cells (CD45RA+, CD45RO−, CD62L+) differentiate into short-lived effector T-cells, and long-lived memory cells (central memory CD45RO+CD62L+, effector memory CD45RO+CD62L− and tissue-resident memory CD45RO+ CD62L−CD69+) [6,7,8]. In mouse models, the naïve T-cells were found to be associated with severe GVHD development, while memory T-cells played a central role in immune reconstitution and graft-versus-leukemia (GVL) effect without inducing significant GVHD [7,8,9,10,11,12,13]. Non-selective depletion of T-cells, in vivo or in vitro, has been associated with reduced risk of GVHD at a cost of more relapses, graft failure and delayed immune reconstitution, resulting in life-threatening infections [13,14,15,16]. Graft engineering, and selective T-cell depletion aiming to deplete naïve T-cells to decrease the risk of GVHD while preserving memory T-cells to allow immune reconstitution and preserve GVL is a relatively new clinical concept.

2. Discussion

The paper by Bleakley et al. is the first in human trial to apply this concept with respect to naïve T-cell depletion [2]. The authors report the outcomes of 35 patients with acute myeloid leukemia (AML) and myelodysplastic syndrome treated with a myeloablative conditioning regimen followed by matched related donor (MRD) transplant with selective in vitro naïve T-cell depletion. With a median follow up of 2.5 years, they reported no graft rejections, 66% grade II–IV acute GVHD (aGVHD), 9% chronic GVHD (cGVHD) (3/35 cases; one mild, one moderate, and one severe), 9% TRM at 2 years (3 patients died; all older than 46 years), 78% and 70% overall survival [OS] and disease-free survival [DFS], respectively, at 2 years. The recovery of blood lymphocytes was comparable to T-cell replete MRD HCT with no excess in EBV or CMV reactivation or BK virus cystitis, which goes along with adequate immune reconstitution. The trial met one of the primary end points (engraftment), but did not meet the second primary end point (aGVHD); thus, in a statistical sense, it can be considered a negative trial; however, apart from aGVHD, for the rest of the clinically relevant variables—i.e., secondary end points (cGVHD, transplant related mortality (TRM), OS, DFS, and immune reconstitution)—the intervention arm compared favorably to T-cell replete MRD HCT. Why the acute GVHD was not reduced remains elusive, and reflects our superficial understanding of the immune system complex function and the complicated physiology of aGVHD (cytokines, conditioning intensity, GVHD prophylaxis). The significant decrease in cGVHD incidence, and the fact that all aGVHD cases were steroid responsive, remain significant advancements in this approach, in addition to the results of all the other secondary end points.
Some issues besides failure to meet the co-primary end point are worthy of mentioning. The intensity of the regimen used in this trial limits its applicability to young patients only, while the AML/MDS is mainly a disease of older populations; therefore, investigating less intense regimens with the same concept is required for external validity. For HCTs in malignancies, two newer end points—cGVHD/relapse free survival (CRFS) and GVHD/relapse or progression free survival (GRFS)—have become standard end points for clinical trials; thereby, we hope that if this concept is taken to a phase III study, the next step would be CRFS or GRFS as the primary end point. Another limitation (a real-world issue), is that the feasibility of selective T-cell depletions by sophisticated techniques is limited to a few transplant centers; another method of T-cell depletion utilizing post-transplant cyclophosphamide is logistically easier, economic, and widely available, so not surprisingly, its use is on the rise globally. Thus, comparing these two approaches would be required eventually to test which approach would confer the optimum GVL without increasing GVHD.
While the results of this trial are impressive in opening new avenues in transplantation (matched and mismatched HCT), these need to be confirmed in larger randomized trials. Ideally, one would compare two similar transplant platforms with identical conditioning regimens and GVHD prophylaxis, with or without naïve T-cell depletion. For separating out GVL from GVHD, many investigators have embarked on various avenues of graft manipulations. The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) conducted a TCD allograft approach with selective CD34+ selection, and compared the results with T-cell replete allografts plus immunosuppressive therapy (IST) with impressive results, indicating significantly less 2-year rates of cGVHD with TCD grafts than the IST group (19% vs. 50%, respectively; p < 0.001) [14]. There were no differences in rates of graft rejection, leukemia relapse, DFS, TRM or OS rates. On the same lines of dissociating GVT from GVHD, Miller et al. have embarked on clinical trials focusing on manipulating NK cells to confer optimum GVL in various donor sources [15]. Dendritic cell engineering approaches (particularly vaccination post-HCT) are also becoming important avenues, and are currently being tested [16]. “HLA-Mismatched stem cell microtransplantation”, as a post-remission therapy for AML, has been tested in a phase II fashion by a Chinese group with impressive results, yielding a 6-year OS and DFS of >80% with no acute or chronic GVHD [17]. Lastly, many cellular-therapy/gene-editing maneuvers (including inducible safety switch for adoptive cellular therapy), myriad of epigenetic targeting (micro-RNAs, kinases, JAK/Stat pathway, and notch signaling pathway at the time of HCT or pre-HCT), and chimeric antigen receptor immunotherapy (particularly with knockout via gene editing of αβ T-cell receptor to reduce GVHD) studies are on the horizon. Thus, in the presence of various novel techniques translating from animal models to clinical trials in the current era, how can one prioritize the novelty into a large randomized prospective trial, taking into consideration the costs and logistical issues associated with large randomized trials?

3. Conclusions

We believe that the approach by Bleakly et al. and colleagues has shown that outcomes, which are clinically meaningful to both the clinicians and patients, are promising enough to further expand this avenue into a randomized trial. Although acute GVHD was not low, it was easily treatable and the mortality was not increased compared to a historical cohort of T-cell replete HCT. Most importantly, it is clear that the risk of cGVHD was much lower with naïve T-cell depleted allografts, without increasing the risk of relapses as indicated by the results of this trial. Compared to other novel strategies, this method is safe, and mature enough (this was a phase II trial) to lead to a multicenter randomized clinical trial in the hope of finally resolving the issue of the holy grail of HCTs for the benefit of mankind.

Author Contributions

Riad El Fakih and Shahrukh K. Hashmi wrote the manuscript. Both authors contributed substantially to the conception, acquisition, and interpretation of the data for the work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Horowitz, M.M.; Gale, R.P.; Sondel, P.M.; Goldman, J.M.; Kersey, J.; Kolb, H.J.; Rimm, A.A.; Ringden, O.; Rozman, C.; Speck, B. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 1990, 75, 555–562. [Google Scholar] [PubMed]
  2. Bleakley, M.; Heimfeld, S.; Loeb, K.R.; Jones, L.A.; Chaney, C.; Seropian, S.; Gooley, T.A.; Sommermeyer, F.; Riddell, S.R.; Shlomchik, W.D. Outcomes of acute leukemia patients transplanted with naive T cell–depleted stem cell grafts. J. Clin. Investig. 2015, 125, 2677. [Google Scholar] [CrossRef] [PubMed]
  3. Shlomchik, W.D. Graft-versus-host disease. Nat. Rev. Immunol. 2007, 7, 340–352. [Google Scholar] [CrossRef] [PubMed]
  4. Palathumpat, V.; Dejbakhsh-Jones, S.; Holm, B.; Strober, S. Different subsets of T cells in the adult mouse bone marrow and spleen induce or suppress acute graft-versus-host disease. J. Immunol. 1992, 149, 808–817. [Google Scholar] [PubMed]
  5. Aversa, G.; Waugh, J.A.; Hall, B.M. A monoclonal antibody (A6) recognizing a unique epitope restricted to CD45RO and RB isoforms of the leukocyte common antigen family identifies functional T cell subsets. Cell. Immunol. 1994, 158, 314–328. [Google Scholar] [CrossRef] [PubMed]
  6. Conlon, K.; Osborne, J.; Morimoto, C.; Ortaldo, J.R.; Young, H.A. Comparison of lymphokine secretion and mRNA expression in the CD45RA+ and CD45RO+ subsets of human peripheral blood CD4+ and CD8+ lymphocytes. Eur. J. Immunol. 1995, 25, 644–648. [Google Scholar] [CrossRef] [PubMed]
  7. Pihlgren, M.; Lightstone, L.; Mamalaki, C.; Rimon, G.; Kioussis, D.; Marvel, J. Expression in vivo of CD45RA, CD45RB and CD44 on T cell receptor-transgenic CD8+ T cells following immunization. Eur. J. Immunol. 1995, 25, 1755–1759. [Google Scholar] [CrossRef] [PubMed]
  8. Lanzavecchia, A.; Sallusto, F. Dynamics of T lymphocyte responses: Intermediates, effectors, and memory cells. Science 2000, 290, 92–97. [Google Scholar] [CrossRef] [PubMed]
  9. Anderson, B.E.; McNiff, J.; Yan, J.; Doyle, H.; Mamula, M.; Shlomchik, M.J.; Shlomchik, W.D. Memory CD4+ T cells do not induce graft-versus-host disease. J. Clin. Investig. 2003, 112, 101–108. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, B.J.; Cui, X.; Sempowski, G.D.; Liu, C.; Chao, N.J. Transfer of allogeneic CD62Lmemory T cells without graft-versus-host disease. Blood 2004, 103, 1534–1541. [Google Scholar] [CrossRef] [PubMed]
  11. Zheng, H.; Matte-Martone, C.; Li, H.; Anderson, B.E.; Venketesan, S.; Tan, H.S.; Jain, D.; McNiff, J.; Shlomchik, W.D. Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood 2008, 111, 2476–2484. [Google Scholar] [CrossRef] [PubMed]
  12. Zheng, H.; Matte-Martone, C.; Jain, D.; McNiff, J.; Shlomchik, W.D. Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia. J. Immunol. 2009, 182, 5938–5948. [Google Scholar] [CrossRef] [PubMed]
  13. Li, N.; Matte-Martone, C.; Zheng, H.; Cui, W.; Venkatesan, S.; Tan, H.S.; McNiff, J.; Demetris, A.J.; Roopenian, D.; Kaech, S.; et al. Memory T cells from minor histocompatibility antigen vaccinated and virus-immune donors improve GVL and immune reconstitution. Blood 2011, 118, 5965–5976. [Google Scholar] [CrossRef] [PubMed]
  14. Pasquini, M.C.; Devine, S.; Mendizabal, A.; Baden, L.R.; Wingard, J.R.; Lazarus, H.M.; Appelbaum, F.R.; Keever-Taylor, C.A.; Horowitz, M.M.; Carter, S.; et al. Comparative outcomes of donor graft CD34+ selection and immune suppressive therapy as graft-versus-host disease prophylaxis for patients with acute myeloid leukemia in complete remission undergoing HLA-matched sibling allogeneic hematopoietic cell transplantation. J. Clin. Oncol. 2012, 30, 3194–3201. [Google Scholar] [PubMed]
  15. Wiernik, A.; Foley, B.; Zhang, B.; Verneris, M.R.; Warlick, E.; Gleason, M.K.; Ross, J.A.; Luo, X.; Weisdorf, D.J.; Walcheck, B.; et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16× 33 bispecific killer cell engager and ADAM17 inhibition. Clin. Cancer Res. 2013, 19, 3844–3855. [Google Scholar] [CrossRef] [PubMed]
  16. Plantinga, M.; de Haar, C.; Nierkens, S.; Boelens, J.J. Dendritic cell therapy in an allogeneic-hematopoietic cell transplantation setting: An effective strategy toward better disease control? Front. Immunol. 2014, 5, 218. [Google Scholar] [CrossRef] [PubMed]
  17. Guo, M.; Hu, K.X.; Liu, G.X.; Yu, C.L.; Qiao, J.H.; Sun, Q.Y.; Qiao, J.X.; Dong, Z.; Sun, W.J.; Sun, X.D.; et al. HLA-mismatched stem-cell microtransplantation as postremission therapy for acute myeloid leukemia: Long-term follow-up. J. Clin. Oncol. 2012, 30, 4084–4090. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

El Fakih, R.; Hashmi, S.K. Hitting the Holy Grail of Hematopoietic Cell Transplantation with Naive T-Cell Depleted Allografts—Graft Engineered Hematopoietic Stem Cell Transplant. Biomedicines 2017, 5, 48. https://doi.org/10.3390/biomedicines5030048

AMA Style

El Fakih R, Hashmi SK. Hitting the Holy Grail of Hematopoietic Cell Transplantation with Naive T-Cell Depleted Allografts—Graft Engineered Hematopoietic Stem Cell Transplant. Biomedicines. 2017; 5(3):48. https://doi.org/10.3390/biomedicines5030048

Chicago/Turabian Style

El Fakih, Riad, and Shahrukh K. Hashmi. 2017. "Hitting the Holy Grail of Hematopoietic Cell Transplantation with Naive T-Cell Depleted Allografts—Graft Engineered Hematopoietic Stem Cell Transplant" Biomedicines 5, no. 3: 48. https://doi.org/10.3390/biomedicines5030048

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop