Elsevier

Blood Reviews

Volume 32, Issue 6, November 2018, Pages 449-456
Blood Reviews

Review
Recipient and donor cells in the graft-versus-solid tumor effect: It takes two to tango

https://doi.org/10.1016/j.blre.2018.04.002Get rights and content

Abstract

Allogeneic hematopoietic stem cell transplantation (alloHSCT) produces –similar to the long-established graft-versus-leukemia effect– graft-versus-solid-tumor effects. Clinical trials reported response rates of up to 53%, occurring mostly but not invariably in association with full donor chimerism and/or graft-versus-host disease. Although donor-derived T cells are considered the principal effectors of anti-tumor immunity after alloHSCT or donor leukocyte infusion (DLI), growing evidence indicate that recipient-derived immune cells may also contribute. Whereas the role of recipient-derived antigen-presenting cells in eliciting graft-versus-host reactions and priming donor T cells following DLI is well known, resulting inflammatory responses may also break tolerance of recipient effector cells towards the tumor. Additionally, mouse studies indicated that post-transplant recipient leukocyte infusion produces anti-leukemia and anti-solid-tumor effects that were exclusively mediated by recipient-type effector cells, without graft-versus-host disease. Here, we review current preclinical and clinical evidence on graft-versus-solid-tumor effects and growing evidence on the effector role of recipient-derived immune cells in the anti-tumor effect of alloHSCT.

Introduction

Allogeneic hematopoietic stem cell transplantation (alloHSCT) is recognized as one of the most effective strategies in the treatment of advanced hematological cancers, particularly in combination with donor leukocyte infusion (DLI) [[1], [2], [3], [4]]. Whereas treatment with hematopoietic stem cell transplantation was initially conceived to reconstitute the hematopoietic system after high-dose chemo-radiotherapy, soon it became evident that the curative potential of alloHSCT can be attributed to an immune-based response of the transplanted and reconstituting donor cells against the malignant cells, also referred to graft-versus-tumor (GvT) effect [5,6]. Over the last decade, the field of alloHSCT has continued to evolve, with an emerging role for alloHSCT in the treatment of advanced solid tumors [7], and growing evidence on the potential contribution of recipient immune cells in the anti-tumor reactivity following alloHSCT.

Section snippets

AlloHSCT and the existence of a graft-versus-tumor effect

In early alloHSCT regimens, myeloablative conditioning including high-dose chemo-radiotherapy was used to maximally eradicate cancer cells to suppress the recipient immune system and allow donor engraftment. However, such high-dose regimens were associated with high treatment-related toxicities [8] and induction of chemo-resistance [9] while disease recurrence still occurred indicating that high-dose cytotoxic therapy was unable to eliminate tumor cells completely [10]. Moreover, the notion of

AlloHSCT in the treatment of advanced solid tumors

With currently available therapies, several types of advanced-stage solid tumors still present with a bleak prognosis. The observation that GvL effects can produce durable remissions in hematologic cancer patients, led researchers to investigate whether alloHSCT holds potential for solid tumors. Since inflammation was shown to facilitate trafficking of alloreactive T cells from the lymphohematopoietic system into epithelial target cells [24,25], it was postulated that the same mechanism would

A role for recipient immune cells in the anti-tumor effects of alloHSCT

The very first to speculate on a role for recipient immune cells in the anti-tumor effect of donor cell therapy were Alexander et al. in 1966, in a study showing that xenogeneic lymphocytes from tumor-immunized sheep reduced fibrosarcoma growth in immunocompetent rats. The authors postulated that the observed anti-tumor effect was not mediated via direct anti-tumor reactivity of donor T cells as these were rapidly rejected in the xenogeneic setting, but indirectly through what they called a

Conclusion

The finding that alloHSCT can generate immune anti-tumor effects against hematologic as well as solid tumors, is one of the most important advances in the field of HSCT. Although donor T cells are considered the principal mediators of the anti-tumor effect following alloHSCT, recent observations also indicate a key role for recipient-derived immune cells. The presence of recipient APC was shown to be crucial for effective anti-tumor responses as a means to increase the immunocompetence and to

Practice points

  • There is growing preclinical and clinical evidence to indicate that alloHSCT can provide an immune anti-solid tumor effect.

  • Clinical observations indicate that full donor chimerism and GvHD are not prerequisites for effective anti-solid tumor immunity after alloHSCT.

  • Whereas the importance of pre-existing recipient chimerism is well known in anti-tumor responses provided by post-transplant DLI, evidence has emerged indicating that recipient immune cells can also contribute as effectors to the

Research agenda

  • Prospective monitoring of recipient immune cell activation in clinical trials with alloHSCT for advanced solid tumors would contribute to a better understanding of the emerging role of recipient effector cells in the anti-tumor effect.

  • Experimental studies should further unravel the interactions between the different effector immune cells thought to be involved in the anti-solid tumor effect of alloHSCT. Experimental models should be expanded to include other common and difficult-to-treat solid

Conflict of interest statement

The authors declare that they have no conflict of interest.

Funding

Isabelle Dierckx de Casterlé is supported by the Olivia Hendrickx Research Fund (www.Olivia.be). This funding source supported the work to obtain the documented findings on the role of recipient effector cells following post-transplant challenge with RLI in the preclinical neuroblastoma model, but had no involvement in the collection, analysis or interpretation of the data, nor in the writing of the manuscript.

References (82)

  • S. Servais et al.

    Allogeneic hematopoietic stem cell transplantation (HSCT) after reduced intensity conditioning

    Transfus Apher Sci

    (2011)
  • I.F. Khouri et al.

    Nonablative allogeneic hematopoietic transplantation as adoptive immunotherapy for indolent lymphoma: low incidence of toxicity, acute graft-versus-host disease, and treatment-related mortality

    Blood

    (2001)
  • F. Baron et al.

    Kinetics of engraftment following allogeneic hematopoietic cell transplantation with reduced-intensity or nonmyeloablative conditioning

    Blood Rev

    (2005)
  • S. Bhatia et al.

    Late mortality after allogeneic hematopoietic cell transplantation and functional status of long-term survivors: report from the Bone Marrow Transplant Survivor Study

    Blood

    (2007)
  • V.T. Ho et al.

    The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation

    Blood

    (2001)
  • W.R. Drobyski et al.

    Salvage immunotherapy using donor leukocyte infusions as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation: efficacy and toxicity of a defined T-cell dose

    Blood

    (1993)
  • B.D. Johnson et al.

    Delayed infusion of immunocompetent donor cells after bone marrow transplantation breaks graft-host tolerance allows for persistent antileukemic reactivity without severe graft-versus-host disease

    Blood

    (1995)
  • A.D. Billiau et al.

    Crucial role of timing of donor lymphocyte infusion in generating dissociated graft-versus-host and graft-versus-leukemia responses in mice receiving allogeneic bone marrow transplants

    Blood

    (2002)
  • B. Eibl et al.

    Evidence for a graft-versus-tumor effect in a patient treated with marrow ablative chemotherapy and allogeneic bone marrow transplantation for breast cancer

    Blood

    (1996)
  • M.R. Bishop

    Nonmyeloablative allogeneic hematopoietic stem cell transplantation for metastatic breast cancer

    Clin Breast Cancer

    (2003)
  • M. Bregni et al.

    Nonmyeloablative conditioning followed by hematopoietic cell allografting and donor lymphocyte infusions for patients with metastatic renal and breast cancer

    Blood

    (2002)
  • N.T. Ueno et al.

    Rapid induction of complete donor chimerism by the use of a reduced-intensity conditioning regimen composed of fludarabine and melphalan in allogeneic stem cell transplantation for metastatic solid tumors

    Blood

    (2003)
  • L. Barkholt et al.

    Allogeneic haematopoietic stem cell transplantation for metastatic renal carcinoma in Europe

    Ann Oncol

    (2006)
  • D. Blaise et al.

    Reduced-intensity preparative regimen and allogeneic stem cell transplantation for advanced solid tumors

    Blood

    (2004)
  • M. Aglietta et al.

    Reduced-intensity allogeneic hematopoietic stem cell transplantation in metastatic colorectal cancer as a novel adoptive cell therapy approach. The European group for blood and marrow transplantation experience

    Biol Blood Marrow Transplant

    (2009)
  • B. Omazic et al.

    Long-term follow-up of allogeneic hematopoietic stem cell transplantation for solid cancer

    Biol Blood Marrow Transplant

    (2016)
  • E. Orsini et al.

    Conversion to full donor chimerism following donor lymphocyte infusion is associated with disease response in patients with multiple myeloma

    Biol Blood Marrow Transplant

    (2000)
  • M.R. Bishop et al.

    Establishment of early donor engraftment after reduced-intensity allogeneic hematopoietic stem cell transplantation to potentiate the graft-versus-lymphoma effect against refractory lymphomas

    Biol Blood Marrow Transplant

    (2003)
  • P. Alexander et al.

    The effect of lymphoid cells from the lymph of specifically immunised sheep on the growth of primary sarcomata in rats

    Lancet

    (1966)
  • H.J. Symons et al.

    The allogeneic effect revisited: exogenous help for endogenous, tumor-specific T cells

    Biol Blood Marrow Transplant

    (2008)
  • L. Schwarzenberg et al.

    Attempted adoptive immunotherapy of acute leukaemia by leucocyte transfusions

    Lancet

    (1966)
  • D.J. Medina et al.

    A pilot study of allogeneic cellular therapy for patients with advanced hematologic malignancies

    Leuk Res

    (2008)
  • G.A. Colvin et al.

    Nonengraftment Haploidentical cellular immunotherapy for refractory malignancies: tumor responses without chimerism

    Biol Blood Marrow Transplant

    (2009)
  • M. Guo et al.

    Infusion of HLA-mismatched peripheral blood stem cells improves the outcome of chemotherapy for acute myeloid leukemia in elderly patients

    Blood

    (2011)
  • L. Luznik et al.

    Successful therapy of metastatic cancer using tumor vaccines in mixed allogeneic bone marrow chimeras

    Blood

    (2003)
  • M. Sykes

    Mixed chimerism and transplant tolerance

    Immunity

    (2001)
  • O. Alpdogan et al.

    Immune tolerance and transplantation

    Semin Oncol

    (2012)
  • M.Y. Mapara et al.

    Donor lymphocyte infusions mediate superior graft-versus-leukemia effects in mixed compared to fully allogeneic chimeras: a critical role for host antigen-presenting cells

    Blood

    (2002)
  • P.V. O'Donnell et al.

    Nonmyeloablative bone marrow transplantation from partially HLA-mismatched related donors using posttransplantation cyclophosphamide

    Biol Blood Marrow Transplant

    (2002)
  • M.T. Rubio et al.

    Antitumor effect of donor marrow graft rejection induced by recipient leukocyte infusions in mixed chimeras prepared with nonmyeloablative conditioning: critical role for recipient-derived IFN-gamma

    Blood

    (2003)
  • M.T. Rubio et al.

    Role of indirect Allo- and autoreactivity in anti-tumor responses induced by recipient leukocyte infusions (RLI) in mixed chimeras prepared with nonmyeloablative conditioning

    Clin Immunol

    (2006)
  • Cited by (8)

    View all citing articles on Scopus
    1

    Shared last authorship

    View full text