ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Biology of the graft-versus-tumor effect following hematopoietic cell transplantation

Biology of the graft-versus-tumor effect following hematopoietic cell transplantation
Author:
Robert S Negrin, MD
Section Editor:
Nelson J Chao, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Jan 2024.
This topic last updated: May 04, 2022.

INTRODUCTION — The majority of patients with malignancy who undergo hematopoietic cell transplantation (HCT) are effectively treated, thereby resulting in minimal residual disease. However, this response is frequently not maintained since relapse ultimately occurs in 40 to 75 percent of patients who undergo an autologous transplant and 10 to 40 percent of those who undergo an allogeneic transplant. Further, with the development of non-myeloablative or reduced intensity allogeneic transplantation there is increased reliance on immune-mediated effects to control the underlying disease.

The rationale for using immunotherapy to prevent and/or treat the reemergence of malignancy is based upon the following observations:

Evidence indicates that the graft-versus-tumor (GVT) effect plays a major role in reducing the risk of relapse following an allogeneic transplant.

Significant advances have been made in our basic understanding of both the cellular populations responsible for potential antitumor activity and the cellular interactions and cytokines required for their activation and expansion.

The cell populations capable of recognizing and lysing malignant targets can be divided into two broad categories based upon the mechanism of cellular recognition: cytotoxic T cells (CTLs) and natural killer (NK) cells. Significant insights have been made into the functional mechanisms of these two populations.

This topic review will discuss the basic principles underlying the pathogenesis of the GVT effect after HCT. An overview of some of the clinical trials examining immunotherapy to prevent and treat relapse following HCT is presented separately. (See "Immunotherapy for the prevention and treatment of relapse following allogeneic hematopoietic cell transplantation".)

CYTOTOXIC T CELLS — Before discussing how cytotoxic T cells (CTLs) recognize and become activated in response to malignant cells, it is helpful to first briefly review the major histocompatibility complex (MHC), and how T cells recognize and are activated by any antigen. Genes of the MHC (called HLA in humans) encode two distinct classes of cell surface molecules, I and II. Class I MHC molecules are expressed on the surfaces of virtually all nucleated cells at varying densities, while class II MHC molecules are more restricted to cells of the immune system, primarily B lymphocytes, monocytes, and dendritic cells.

Since a major function of the immune system is to distinguish self from non-self, HLA molecules provide the crucial surface upon which the antigen receptors on T lymphocytes (T cell receptors or TCRs) recognize foreign (non-self) antigens. On antigen presenting cells (APCs), such as macrophages and dendritic cells, class II MHC molecules present antigenic fragments (in the form of linear peptides) to the CD4+ inducer (or helper) T cells, while class I MHC molecules function at the effector phase of immunity by presenting antigens to CD8+ T cells. This process of antigen presentation consists of the binding of a single T cell receptor to a complex on the surface of an antigen-presenting cell consisting of the MHC molecule and a peptide fragment derived from the host or foreign antigen. Due to the central role of MHC molecules in initiating an immune response, it is not surprising that matching of these genes is the major predictor of graft-versus-host disease in the setting of allogeneic transplantation. (See "Major histocompatibility complex (MHC) structure and function" and "Pathogenesis of graft-versus-host disease (GVHD)".)

In addition to engagement of the T cell receptor with the MHC molecule-antigenic peptide complex, several other signals and/or functions are required for full T cell activation and expansion:

APCs must express costimulatory molecules that provide a mandatory second signal to the T cell. The major pathways are an interaction of the B7 family of molecules (B7-1 [CD80] or B7-2 [CD86]) or of CD58 (LFA-3) on antigen presenting cells with CD28 or CD2 on T cells (figure 1) [1,2]. Other co-stimulatory molecules (eg, CD30:CD30L, OX40) have also been identified. Anergy, a state in which T cells are unable to become activated in response to their normal target antigen, develops in the absence of costimulation [2-4].

Helper T cells are also necessary for a normal immune response. They are primarily CD3+CD4+CD8-, compared with the majority of CTLs which are CD3+CD8+CD16-. Helper T cells release cytokines and provide cellular interactions (via costimulatory molecules) necessary for the appropriate stimulation of APCs (figure 1) [5].

Traditional understanding dictated that CD4+ T cells were required to produce cytokines (principally IL-2) in the local environment of the CD8+ T cells, thereby allowing for clonal expansion. This hypothesis required two populations of effector cells to be present and bind to an APC. However, studies indicate that CD4+ T cells are capable of activating APCs via engagement of CD40, which primes the APCs for more effective stimulation of CD8+ cells. These observations simplify the role of helper T cells and do not require a physical presence at the time of CD8+ T cell activation.

Regulatory T cells — Other populations of T cells regulate immune responses. The best characterized population is that of regulatory T (Treg) cells, which express CD4 and CD25 as well as the transcription factor FoxP3 [6]. Treg cells reduce T cell proliferation and may play a major role in controlling graft-versus-host disease (GVHD) in murine model systems [7] as well as in man [8]. Importantly, Treg cells did not inhibit graft-versus-tumor (GVT) reactions in mouse models [9,10]. Further, following the adoptive transfer of regulatory T cells in the setting of haploidentical transplantation, relapse rates remain low [11].  

Invariant natural killer T (iNKT) cells — A unique subset of natural killer like T cells that express an invariant T cell receptor have also been recognized that have both cytotoxic and immunoregulatory properties. Several groups have indicated that this population of cells is capable of exerting an anti-tumor effect and also regulate immune responses such as the suppression of GVHD [12,13]. Subsets of iNKT cells exist that have been shown in murine models to suppress GVHD on the one hand (iNKT2, and iNKT17 subsets) or have cytotoxic function (iNTK1) [14]. These cells may also stimulate endogenous immunity which could be enhanced by the introduction of a CAR construct [15]. Clinical translation of these concepts are underway.

Natural killer (NK) T cells also play an important immunoregulatory role under certain biological conditions, as discussed below. (See 'Natural killer cells' below.)

Recognition of malignant cells — For a donor CTL to recognize and lyse a malignant host target cell, antigen(s) expressed by the tumor must be recognized as foreign. Based upon the type of transplant, this identification principally occurs via the following two mechanisms:

Donor CTLs recognize alloantigens, possibly minor histocompatibility antigens, on host cells. This is of principal importance with allogeneic transplants.

CTLs also react against unique antigens expressed by the malignant host cells [16]. This can occur with any donor-recipient pair, as well as outside the transplant setting.

Self versus non-self — With nonidentical transplants, donor CTLs may recognize neoplastic (and normal) cells as foreign due to the expression of epitopes unique to the host. CTLs may subsequently become activated, lysing such cells.

This mechanism for a GVT effect is very similar to that which underlies GVHD. When the recipient and nonidentical donor are not matched at the MHC (eg, with mismatched related or unrelated, haploidentical or cord blood donors) GVHD (or GVT) may result because of differences within the major MHC antigens. However, when the donor-recipient pair are matched for MHC antigens, GVHD (or GVT) is initiated and propagated via recognition by the T cell and its receptor of additional antigens called minor histocompatibility antigens [17]. A major question in the field is whether the repertoire of T cells responsible for GVHD is the same as those responsible for GVT, and if not how much overlap between these two populations exists. Another question is whether there are truly disease-specific determinants that can be recognized by donor derived T cells. The use of high throughput sequencing of the TCR may help clarify how these different T cell populations interact in these complex biological reactions [18]. (See "Pathogenesis of graft-versus-host disease (GVHD)".)

The importance of this effect for the development of GVT is suggested by the different outcomes with grafts from identical twin and nonidentical donors. Patients who have an identical twin donor do not develop GVHD. However, they are at a higher risk of relapse of the underlying malignant disease than similar patients transplanted with HLA-matched but nonidentical sibling donors [19]. This is presumably due to the lack of a GVT reaction [20].

These relationships were illustrated in a review from 163 transplant centers that compared the results of 103 identical twin (syngeneic) and 1030 HLA-identical sibling (allogeneic) transplants for leukemia [21]. The three-year probability of relapse of leukemia was substantially higher in the syngeneic than allogeneic transplants in acute myeloid leukemia (52 versus 16 percent) and chronic myeloid leukemia (40 versus 7 percent).

The importance of the pathogenic link between GVT and GVHD is also suggested by the finding that GVHD can be induced by donor lymphocyte infusions. With this technique, lymphocytes from the original donor are administered to patients with relapsing disease to attempt to elicit a GVT reaction; this benefit is tempered by the enhanced risk of developing GVHD. (See "Immunotherapy for the prevention and treatment of relapse following allogeneic hematopoietic cell transplantation".)

However, the observation that some patients clearly have GVT without GVHD suggests that these two processes may result via different mechanisms (see 'Possible separation of GVT from GVHD' below).

Expression of tumor specific antigens — CTLs, independent of origin, can also recognize malignant cells as foreign when unique antigens are expressed by the tumor [22,23]. Potential antigenic targets include the following (table 1):

Unique idiotypes or TCRs, which result from rearranged immunoglobulin or TCR genes, respectively; this may occur in some B and T cell malignancies [24].

Foreign proteins encoded by the viral genome in malignancies that occur in association with viral infections, such as EBV-associated B cell lymphomas.

Specific peptides derived from proteins expressed on tumor cells such as the her-2 neu protein, a product expressed by a subset of breast cancers and neuroblastomas [25].

Products of specific oncogenes (the importance of which is unclear). Peptides derived from breakpoint regions of bcr/abl, an oncogene found in the majority of patients with chronic myeloid leukemia, have been used to stimulate a productive T cell immune response in vitro [26,27].

A variety of lineage-specific antigens, including minor histocompatibility antigens (which are found on hematopoietic tumors), the MAGE proteins (on melanomas), proteinase III, WT-1 and Wnt (on certain leukemias), and carcinoembryonic antigen (on some solid tumors).

Ongoing investigations are focusing on those characteristics of the antigen that would best enable CTLs to recognize and lyse malignant targets. No consensus has emerged concerning the optimal nature and source of these antigens, particularly whether they should be in the form of peptide(s), a whole protein, DNA, or cDNA, or whether they should be introduced into dendritic cells by gene transfer [28].

Other therapeutic maneuvers include the following:

Donor lymphocyte infusion (DLI). (See "Immunotherapy for the prevention and treatment of relapse following allogeneic hematopoietic cell transplantation", section on 'Donor lymphocyte infusion (DLI)'.)

Expansion of dendritic cells from leukemic precursors, based upon the assumption that these cells will present leukemic specific antigens [29].

Adoptive T cell transfer by infusion of activated/expanded T cells, which may promote a cytolytic response against tumor cells and enhance vaccination strategies, especially in the lymphopenic environment early after transplantation [30,31].

Introduction of a chimeric antigen receptor (CAR) construct into the donor T cells have been shown to have clinical activity in those patients who relapse following allogeneic HCT [15].

Malignant cells and costimulation — Tumor cells may lack the costimulatory molecules necessary for such an interaction [32], which, as noted above, may lead to anergy [2-4]. A variety of strategies have been employed in an attempt to overcome this problem:

In one study, follicular lymphoma cells were treated with CD40L, a protein known to upregulate the expression of costimulatory molecules of the B7 family [33]. Using this approach, autologous tumor-infiltrating CTLs could be generated in the presence of interleukin (IL)-2; they could be further expanded in vitro with IL-4, IL-7 and interferon gamma (IFNg).

Another approach stimulates malignant B cells through the toll-like receptor complex using CpG or another immunostimulant to induce more effective antigen presentation and then to re-inject these cells following irradiation as a therapeutic vaccine.

Another strategy has been to transfect tumor cells with costimulatory genes. In animal models, injection of transfected tumor cells resulted in an effective immune response against both the transfected and wild type tumor cells [34,35].

The identification of dendritic cells and their role in stimulating an immune response is a major scientific advance. Clearly, much work is required to identify not only the appropriate clinical situation but also to develop the necessary approach that is effective and technically feasible.

Checkpoint inhibitors — T cell activation, proliferation, and function are influenced by a number of different receptor ligand interactions. Some of these interactions downregulate T cell responses and can be broadly considered physiologic "brakes" or "checkpoints" on the T cell activation that is normally triggered by antigen-presenting cells. Release of this brake with checkpoint inhibitors may augment the native or graft T cell anti-tumor response. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) are examples of checkpoints that have been targeted clinically. (See "Principles of cancer immunotherapy", section on 'Checkpoint inhibitor immunotherapy'.)

Checkpoint inhibitors have demonstrated efficacy in solid tumors, and initial studies in hematologic malignancies (eg, Hodgkin lymphoma) have demonstrated responses in highly pretreated patients. (See "Treatment of relapsed or refractory classic Hodgkin lymphoma", section on 'PD-1 blockade'.)

There are limited data regarding the use of checkpoint inhibitors in the transplant setting. A phase 1 study evaluated the CTLA-4 inhibitor ipilimumab in 28 patients with relapsed hematologic malignancy after allogeneic HCT [36]. Administration was feasible, and durable responses were observed in some patients. Six patients (21 percent) had immune-related adverse events, including one death. These included GVHD, immune thrombocytopenia, colitis, and pneumonitis.

Further studies are underway regarding the use of checkpoint inhibitors to stimulate a more effective GVT response. A significant concern in the setting of allogeneic transplantation is whether this approach could result in worsening of GVHD.

NATURAL KILLER CELLS — Natural killer (NK) cells are commonly referred to as nonspecific or major histocompatibility complex (MHC)-nonrestricted. However, these terms are misnomers, since NK cells express receptors, which recognize MHC molecules.

The cell surface phenotype of NK cells is CD3-CD56+CD16+, although other cell populations have been described. These cells can be activated by a variety of different cytokines, particularly IL-2 [37,38].

Cell receptors — The mechanism of target cell recognition by NK cells is not completely understood [39]. Several groups have identified cell surface receptors expressed by NK cells, which recognize MHC molecules and, upon productive interaction, initiate an inhibitory signal that prevents lysis.

In humans, two distinct families of inhibitory receptors have been characterized:

One group of proteins is referred to as the killer inhibitory receptors (KIRs); they are composed of either two or three extracellular immunoglobulin-like domains, and recognize specific class I HLA molecules [40-43]. The role of peptides in class I recognition by KIRs has been controversial, although it appears that some peptide specificity exists.

A second structurally distinct class of receptors (CD94/NKG2A) has a C-lectin domain and recognizes peptides derived from certain class I alleles in the context of HLA-E [44,45].

NK cells also express activation receptors, which, upon triggering, engage the cytolytic machinery. The best characterized activation receptor is a molecule termed NKG2D. Ligands for NKG2D include molecules such as MICA, MICB and the ULB binding proteins [46]. NKG2D ligands are upregulated by cellular stress, including viral infection and transformation. Other natural cytotoxicity receptors (NCRs) have been identified that, upon stimulation, result in NK cell activation and more effective cytolysis.

In the setting of bone marrow transplantation, donor NK cells are inhibited upon interaction with an appropriate MHC class I molecule expressed by a normal host cell. As a result, all donor NK cells tolerate normal (eg, not malignant or infected) host cells expressing normal levels of MHC class I molecules. By comparison, tumor or virally infected cells, which either downregulate class I expression or have altered peptide expression, are recognized and killed [47].

Several aspects of NK function are unclear, including the exact requirements for an interaction to occur and what role activator receptors play [48]. Adhesion molecules appear to be required since monoclonal antibodies directed against LFA-1 and intercellular adhesion molecule (ICAM)-1 inhibit NK cytolytic activity.

NK cells also express the Fc-gamma receptor (CD16); this allows cells bound by immunoglobulin to be lysed by antibody dependent cellular cytotoxicity mechanisms. This mechanism of tumor cell removal is thought to play an important role in the mechanism of therapeutic antibodies. Individuals who express different polymorphisms of the Fc-gamma receptor may differ in their response to rituximab and presumably other therapeutic monoclonal antibodies [49,50].

Autologous and MHC-matched transplants — The mechanisms by which tumor cells are recognized by NK effector cell populations with autologous and MHC-matched transplants are not completely understood. As previously mentioned, recognition and killing of such cells may occur if the associated KIRs or CD94/NKG2A molecules are not bound by the appropriate HLA-class I molecules. By comparison, nonmalignant autologous or MHC-matched cells presumably express HLA class I molecules with the appropriate peptide; this permits binding to the inhibitory receptor expressed on the surface of the NK cell, thereby inhibiting NK cell function.

This hypothesis assumes that HLA class I molecules on the surface of tumor cells are either downregulated or alternative peptides are displayed, characteristics that block the ability of HLA class I molecules to signal through the appropriate inhibitory receptor. Experimental evidence for the lack of HLA class I expression has been found in a number of different tumor types. Alteration in peptide expression is more difficult to evaluate, but it has been observed following certain viral infections. Alternatively, expression of NKG2D ligands has been observed on a number of solid tumors and hematopoietic cell malignancies which may allow for appropriate cell recognition and lysis.

MHC-mismatched transplants — A low incidence of relapse has been observed with MHC-mismatched transplants, and NK cell-mediated cytotoxicity may contribute to GVT in MHC-mismatched transplants without associated GVHD.

Depending upon the donor-recipient pair, host cells may lack the appropriate HLA class I molecule for binding and inhibiting inhibitory molecules expressed on donor NK and NK-like T cells. Such alloreactive NK cells, which recognize recipient tumor cells based upon the lack of inhibition through the appropriate KIR molecules, have been observed both before and after mismatched transplantation [51]. Alloreactive NK cells that do not induce (and may even suppress) GVHD are observed in animal models of adoptive transfer and in human transplantation with HLA-mismatched NK cells, including haploidentical transplants [52-56].

Cytokine production — NK cells secrete a variety of cytokines, such as interferon-gamma, tumor necrosis factor (TNF), granulocyte macrophage colony-stimulating factor (GM-CSF), and transforming growth factor (TGF)-beta. These cytokines may have some direct effects on the tumor cells and may promote recruitment of other populations of effector cells.

Activation and expansion — A number of strategies have been employed to activate and expand cells with NK cell function. These cells appear to be activated with cytokines, particularly IL-2, IL-12, and IL-15 [57,58]. To enhance the antitumor function of NK cells, different methods of administering IL-2, both ex vivo and in vivo, have been used:

Many groups have used IL-2 for the ex vivo activation of NK cells, thereby resulting in cells with enhanced anti-tumor cell activity (termed lymphokine activated killer [LAK] cells). However, since NK cells do not expand effectively ex vivo, it is difficult to obtain the numbers of cells required for treatment, thereby resulting in limited efficacy.

The direct administration to the patient of high-dose IL-2 is associated with significant morbidity and limited enhancement in NK cell antitumor efficacy [59].

A randomized study of IL-2 following autologous transplantation did not improve outcomes [60], perhaps because lower doses of IL-2 expand regulatory T cells, which may negatively impact anti-tumor responses [61,62]. In the absence of more definitive data such approaches should be used only in the context of a clinical trial.

Cytokine-induced killer (CIK) cells — The active expansion of cytolytic cells derived from T cells which share functional similarities with NK cells may be obtained with the use of specific cytokines [63,64]. These NK-like T cells, also called cytokine-induced killer (CIK) cells, express both T cell specific antigens (such as the T cell receptor and CD3 complex) and NK cell markers (such as CD56 or neural cell adhesion molecule) [65]. These ex vivo treated CD3+CD56+ cells rapidly expand following mitogenic stimulation through the T cell receptor (similar to T cells) and recognize tumor cell targets without prior exposure (similar to NK cells) [66]. They also produce cytokines such as GM-CSF, interferon, and tumor necrosis factor, but do not produce IL-4. NKG2D is an activating receptor on NK cells and CD8 T cells that allows for the recognition of tumor cell by these CIK cells [67,68].

Clinical trials using these cells have suggested that this approach is tolerable following autologous and allogeneic transplantation [69,70]. In one study using this methodology, improvement in recurrence-free survival was noted in patients undergoing hepatic tumor resection at high risk of relapse where those individuals who received cellular therapy had improved DFS compared with untreated controls [71]. CIK cells infected with the oncolytic vaccinia virus were particularly effective in animal models [15,72].

The different properties of these effector cell populations that may prove useful in immunotherapy posttransplantation are summarized in the table (table 2).

CYTOTOXIC CELL LINES — Several cell lines with cytotoxic activity have been developed which have phenotypic and biological characteristics very similar to NK cells and NK-like T cells. The cell line TALL-104 originated from a child with acute T cell lymphoblastic leukemia. These cells lyse a broad panel of tumor cell lines, but not normal tissues, and have in vivo activity in animal models [73,74]. The use of irradiated TALL-104 cells is in early phase clinical trials. Other NK-like cell lines have also been developed, such as NK-92, with intriguing biological activity [75-79].

Engineered T cells — A number of groups have investigated the use of engineered T cells, which express novel receptors that redirect their cytotoxicity. An example that has demonstrated efficacy is through the use of chimeric antigen receptor T cells (CAR-T) where the T cells are capable of recognizing tumor-associated antigens, such as CD19 and CD22. Early studies had limited success, primarily thought to be related to the poor survival of these engineered T cells. With novel constructions, including co-stimulatory molecules that improve survival of the adoptively transferred T cells, dramatic responses have been noted [80,81]. Use of engineered cells with defined and desired properties is an area of intense investigation.

MECHANISMS OF CYTOLYSIS — Upon appropriate stimulation, all effector cells use similar cytotoxic machinery. Two major cytolytic pathways have been described [82]:

One involves the exocytosis of perforin and granzymes into the extracellular space at the site of target cell adhesion. Perforin induces pore formation in the membrane of the target cell, causing osmotic lysis and introducing granzymes and granulysin, which induce apoptosis [82-84]. The importance of these molecules in CTL and NK cell function has been demonstrated by the finding of greatly impaired effector cell function in mice deficient in these molecules [85,86].

The second pathway involves the expression of fas ligand (fasL); this receptor binds to fas on the target cell, directly inducing apoptosis in a calcium-independent manner [87,88].

MECHANISMS OF TUMOR CELL ESCAPE — Insights into tumor cell recognition have identified a number of potential mechanisms by which tumor cells can escape immunologic recognition (table 3). These include:

The absence of donor- or tumor-specific antigens or the downregulation of HLA class I molecules, preventing recognition by CTLs.

The lack of costimulatory molecules, resulting in anergy.

The expression of both fas and fasL, thereby inactivating T cells due to fas-mediated apoptosis [89,90]. In animal models, for example, fasL expressing tumors progress more rapidly in wild type mice than in animals with defective fas [89].

The heterogeneity of human tumor cells and the possibility of ongoing mutations.

The appropriate expression of self-MHC class I molecules, thereby inactivating NK cells.

The expression of soluble factors that inhibit NK cells, such as the NKG2D ligands MICA and MICB [91]. These factors have been described in vitro, but their clinical significance is unclear.

An additional mechanism that may undermine immune surveillance is the presence of factors that impair the ability of effector cells to survive or to successfully home to malignant cells.

These dynamic interactions complicate immunotherapy applications. However, with increased understanding of these biological processes, more effective approaches will be developed which are based upon the underlying biology of the tumor cell and the immune system. Since patients harbor a state of minimal disease and relapse rates are predictable following hematopoietic cell transplantation, such individuals are ideal candidates to test novel immunotherapeutic approaches.

POSSIBLE SEPARATION OF GVT FROM GVHD — With nonidentical transplants, donor cytotoxic T cells (CTLs) may recognize neoplastic cells as foreign due to the expression of epitopes unique to the host (and the underlying malignancy). CTLs may subsequently become activated, lysing such cells. This graft-versus-tumor (GVT) effect is very similar to that which underlies graft-versus-host disease (GVHD). This theory is supported by evidence that the timing and intensity of immunosuppressive treatment directed at preventing and treating GVHD influences the risk of recurrent malignancy [92].

However, some patients have developed a GVT response without GVHD [93]; additionally, epidemiologic studies from the European Group for Blood and Marrow Transplantation suggest that these two effects may be separable [94].

An experimental approach involves trying to enhance GVT but not GVHD. In this model, recipients were immunized against their tumor (tumor cell vaccine) one month after HCT [95]. The immunized recipients showed improved survival and protection against tumor growth; the immune response was tumor-specific as no exacerbation of GVHD was observed.

Administration of defined doses of regulatory T cells, natural killer cells, or cytokine-induced killer cells has also resulted in control of GVHD and maintenance of GVT responses in murine models of leukemia and lymphoma [9,10,96,97]. Early phase clinical trials support these observations [98-100].

Memory T cells are also under investigation as they appear to not cause GVHD and may have anti-tumor properties [101-104].

This separation of GVHD from GVT, which has been accomplished in animal models, remains an attractive clinical goal [9,66,105-117]. The successful application of these principles to patients will be a major advance in the field of hematopoietic cell transplantation.

SUMMARY

Most patients with malignancy who undergo hematopoietic cell transplantation (HCT) achieve an initial complete remission. However, relapse ultimately occurs in 40 to 75 percent of patients who undergo an autologous HCT and 10 to 40 percent of those who undergo an allogeneic HCT.

The rationale for using immunotherapy to prevent and/or treat the reemergence of malignancy (ie, relapse after HCT) is based upon evidence that the graft-versus-tumor (GVT) effect plays a major role in reducing the risk of relapse following an allogeneic HCT. (See "Immunotherapy for the prevention and treatment of relapse following allogeneic hematopoietic cell transplantation".)

When the recipient and nonidentical HCT donor are not matched at the major histocompatibility complex (MHC), GVT may result because of differences within the major MHC antigens. (See 'Recognition of malignant cells' above.)

When the donor-recipient pair is matched for MHC antigens, GVT is initiated and propagated via recognition by the T cell and its receptor of additional antigens called minor histocompatibility antigens. (See "Pathogenesis of graft-versus-host disease (GVHD)" and 'Cytotoxic T cells' above and 'Natural killer cells' above.)

  1. Clark EA, Ledbetter JA. How B and T cells talk to each other. Nature 1994; 367:425.
  2. Jenkins MK. The ups and downs of T cell costimulation. Immunity 1994; 1:443.
  3. Sayegh MH, Turka LA. T cell costimulatory pathways: promising novel targets for immunosuppression and tolerance induction. J Am Soc Nephrol 1995; 6:1143.
  4. Schwartz RH. Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy. Cell 1992; 71:1065.
  5. Lanzavecchia A. Immunology. Licence to kill. Nature 1998; 393:413.
  6. Shevach EM. Certified professionals: CD4(+)CD25(+) suppressor T cells. J Exp Med 2001; 193:F41.
  7. Hoffmann P, Ermann J, Edinger M, et al. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med 2002; 196:389.
  8. Rezvani K, Mielke S, Ahmadzadeh M, et al. High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT. Blood 2006; 108:1291.
  9. Edinger M, Hoffmann P, Ermann J, et al. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med 2003; 9:1144.
  10. Trenado A, Charlotte F, Fisson S, et al. Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J Clin Invest 2003; 112:1688.
  11. Martelli MF, Di Ianni M, Ruggeri L, et al. "Designed" grafts for HLA-haploidentical stem cell transplantation. Blood 2014; 123:967.
  12. Schneidawind D, Baker J, Pierini A, et al. Third-party CD4+ invariant natural killer T cells protect from murine GVHD lethality. Blood 2015; 125:3491.
  13. Schneidawind D, Pierini A, Alvarez M, et al. CD4+ invariant natural killer T cells protect from murine GVHD lethality through expansion of donor CD4+CD25+FoxP3+ regulatory T cells. Blood 2014; 124:3320.
  14. Maas-Bauer K, Lohmeyer JK, Hirai T, et al. Invariant natural killer T-cell subsets have diverse graft-versus-host-disease-preventing and antitumor effects. Blood 2021; 138:858.
  15. Simonetta F, Lohmeyer JK, Hirai T, et al. Allogeneic CAR Invariant Natural Killer T Cells Exert Potent Antitumor Effects through Host CD8 T-Cell Cross-Priming. Clin Cancer Res 2021; 27:6054.
  16. Ofran Y, Ritz J. Targets of tumor immunity after allogeneic hematopoietic stem cell transplantation. Clin Cancer Res 2008; 14:4997.
  17. Randolph SS, Gooley TA, Warren EH, et al. Female donors contribute to a selective graft-versus-leukemia effect in male recipients of HLA-matched, related hematopoietic stem cell transplants. Blood 2004; 103:347.
  18. Meyer EH, Hsu AR, Liliental J, et al. A distinct evolution of the T-cell repertoire categorizes treatment refractory gastrointestinal acute graft-versus-host disease. Blood 2013; 121:4955.
  19. Thomas ED, Clift RA, Fefer A, et al. Marrow transplantation for the treatment of chronic myelogenous leukemia. Ann Intern Med 1986; 104:155.
  20. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 1990; 75:555.
  21. Gale RP, Horowitz MM, Ash RC, et al. Identical-twin bone marrow transplants for leukemia. Ann Intern Med 1994; 120:646.
  22. Boon T, van der Bruggen P. Human tumor antigens recognized by T lymphocytes. J Exp Med 1996; 183:725.
  23. Nishida T, Hudecek M, Kostic A, et al. Development of tumor-reactive T cells after nonmyeloablative allogeneic hematopoietic stem cell transplant for chronic lymphocytic leukemia. Clin Cancer Res 2009; 15:4759.
  24. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med 1996; 2:52.
  25. Van den Eynde BJ, van der Bruggen P. T cell defined tumor antigens. Curr Opin Immunol 1997; 9:684.
  26. Bocchia M, Korontsvit T, Xu Q, et al. Specific human cellular immunity to bcr-abl oncogene-derived peptides. Blood 1996; 87:3587.
  27. Bosch GJ, Joosten AM, Kessler JH, et al. Recognition of BCR-ABL positive leukemic blasts by human CD4+ T cells elicited by primary in vitro immunization with a BCR-ABL breakpoint peptide. Blood 1996; 88:3522.
  28. Engleman EG. Dendritic cells in the treatment of cancer. Biol Blood Marrow Transplant 1996; 2:115.
  29. Choudhury A, Gajewski JL, Liang JC, et al. Use of leukemic dendritic cells for the generation of antileukemic cellular cytotoxicity against Philadelphia chromosome-positive chronic myelogenous leukemia. Blood 1997; 89:1133.
  30. Laport GG, Levine BL, Stadtmauer EA, et al. Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation. Blood 2003; 102:2004.
  31. Rapoport AP, Stadtmauer EA, Aqui N, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med 2005; 11:1230.
  32. Zheng Z, Takahashi M, Aoki S, et al. Expression patterns of costimulatory molecules on cells derived from human hematological malignancies. J Exp Clin Cancer Res 1998; 17:251.
  33. Schultze JL, Seamon MJ, Michalak S, et al. Autologous tumor infiltrating T cells cytotoxic for follicular lymphoma cells can be expanded in vitro. Blood 1997; 89:3806.
  34. Dunussi-Joannopoulos K, Weinstein HJ, Nickerson PW, et al. Irradiated B7-1 transduced primary acute myelogenous leukemia (AML) cells can be used as therapeutic vaccines in murine AML. Blood 1996; 87:2938.
  35. Townsend SE, Allison JP. Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science 1993; 259:368.
  36. Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N Engl J Med 2016; 375:143.
  37. Phillips JH, Lanier LL. Dissection of the lymphokine-activated killer phenomenon. Relative contribution of peripheral blood natural killer cells and T lymphocytes to cytolysis. J Exp Med 1986; 164:814.
  38. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med 1982; 155:1823.
  39. Gumperz JE, Parham P. The enigma of the natural killer cell. Nature 1995; 378:245.
  40. Döhring C, Scheidegger D, Samaridis J, et al. A human killer inhibitory receptor specific for HLA-A1,2. J Immunol 1996; 156:3098.
  41. Litwin V, Gumperz J, Parham P, et al. NKB1: a natural killer cell receptor involved in the recognition of polymorphic HLA-B molecules. J Exp Med 1994; 180:537.
  42. Moretta A, Vitale M, Bottino C, et al. P58 molecules as putative receptors for major histocompatibility complex (MHC) class I molecules in human natural killer (NK) cells. Anti-p58 antibodies reconstitute lysis of MHC class I-protected cells in NK clones displaying different specificities. J Exp Med 1993; 178:597.
  43. Pende D, Biassoni R, Cantoni C, et al. The natural killer cell receptor specific for HLA-A allotypes: a novel member of the p58/p70 family of inhibitory receptors that is characterized by three immunoglobulin-like domains and is expressed as a 140-kD disulphide-linked dimer. J Exp Med 1996; 184:505.
  44. Boyington JC, Riaz AN, Patamawenu A, et al. Structure of CD94 reveals a novel C-type lectin fold: implications for the NK cell-associated CD94/NKG2 receptors. Immunity 1999; 10:75.
  45. Brooks AG, Borrego F, Posch PE, et al. Specific recognition of HLA-E, but not classical, HLA class I molecules by soluble CD94/NKG2A and NK cells. J Immunol 1999; 162:305.
  46. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 2003; 3:781.
  47. Kärre K. Express yourself or die: peptides, MHC molecules, and NK cells. Science 1995; 267:978.
  48. Uhrberg M, Valiante NM, Shum BP, et al. Human diversity in killer cell inhibitory receptor genes. Immunity 1997; 7:753.
  49. Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol 2003; 21:3940.
  50. Weng WK, Negrin RS, Lavori P, Horning SJ. Immunoglobulin G Fc receptor FcgammaRIIIa 158 V/F polymorphism correlates with rituximab-induced neutropenia after autologous transplantation in patients with non-Hodgkin's lymphoma. J Clin Oncol 2010; 28:279.
  51. Ruggeri L, Capanni M, Casucci M, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 1999; 94:333.
  52. Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N Engl J Med 1998; 339:1186.
  53. Olson JA, Leveson-Gower DB, Gill S, et al. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood 2010; 115:4293.
  54. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005; 105:3051.
  55. Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002; 295:2097.
  56. Hsu KC, Keever-Taylor CA, Wilton A, et al. Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes. Blood 2005; 105:4878.
  57. Perussia B, Chan SH, D'Andrea A, et al. Natural killer (NK) cell stimulatory factor or IL-12 has differential effects on the proliferation of TCR-alpha beta+, TCR-gamma delta+ T lymphocytes, and NK cells. J Immunol 1992; 149:3495.
  58. Kobayashi M, Fitz L, Ryan M, et al. Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J Exp Med 1989; 170:827.
  59. Siegel JP, Puri RK. Interleukin-2 toxicity. J Clin Oncol 1991; 9:694.
  60. Thompson JA, Fisher RI, Leblanc M, et al. Total body irradiation, etoposide, cyclophosphamide, and autologous peripheral blood stem-cell transplantation followed by randomization to therapy with interleukin-2 versus observation for patients with non-Hodgkin lymphoma: results of a phase 3 randomized trial by the Southwest Oncology Group (SWOG 9438). Blood 2008; 111:4048.
  61. Yao X, Ahmadzadeh M, Lu YC, et al. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 2012; 119:5688.
  62. Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 2006; 107:2409.
  63. Ochoa AC, Gromo G, Alter BJ, et al. Long-term growth of lymphokine-activated killer (LAK) cells: role of anti-CD3, beta-IL 1, interferon-gamma and -beta. J Immunol 1987; 138:2728.
  64. Schmidt-Wolf IG, Negrin RS, Kiem HP, et al. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med 1991; 174:139.
  65. Lu PH, Negrin RS. A novel population of expanded human CD3+CD56+ cells derived from T cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency. J Immunol 1994; 153:1687.
  66. Baker J, Verneris MR, Ito M, et al. Expansion of cytolytic CD8(+) natural killer T cells with limited capacity for graft-versus-host disease induction due to interferon gamma production. Blood 2001; 97:2923.
  67. Verneris MR, Karimi M, Baker J, et al. Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8+ T cells. Blood 2004; 103:3065.
  68. Karimi M, Cao TM, Baker JA, et al. Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+ T cells and NK cells. J Immunol 2005; 175:7819.
  69. Leemhuis T, Wells S, Scheffold C, et al. A phase I trial of autologous cytokine-induced killer cells for the treatment of relapsed Hodgkin disease and non-Hodgkin lymphoma. Biol Blood Marrow Transplant 2005; 11:181.
  70. Laport GG, Sheehan K, Baker J, et al. Adoptive immunotherapy with cytokine-induced killer cells for patients with relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2011; 17:1679.
  71. Takayama T, Sekine T, Makuuchi M, et al. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet 2000; 356:802.
  72. Thorne SH, Negrin RS, Contag CH. Synergistic antitumor effects of immune cell-viral biotherapy. Science 2006; 311:1780.
  73. Cesano A, Visonneau S, Pasquini S, et al. Antitumor efficacy of a human major histocompatibility complex nonrestricted cytotoxic T-cell line (TALL-104) in immunocompetent mice bearing syngeneic leukemia. Cancer Res 1996; 56:4444.
  74. Cesano A, Visonneau S, Jeglum KA, et al. Phase I clinical trial with a human major histocompatibility complex nonrestricted cytotoxic T-cell line (TALL-104) in dogs with advanced tumors. Cancer Res 1996; 56:3021.
  75. Gong JH, Maki G, Klingemann HG. Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia 1994; 8:652.
  76. Klingemann HG, Wong E, Maki G. A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood. Biol Blood Marrow Transplant 1996; 2:68.
  77. Klingemann HG, Miyagawa B. Purging of malignant cells from blood after short ex vivo incubation with NK-92 cells. Blood 1996; 87:4913.
  78. Hambach L, Vermeij M, Buser A, et al. Targeting a single mismatched minor histocompatibility antigen with tumor-restricted expression eradicates human solid tumors. Blood 2008; 112:1844.
  79. Arai S, Meagher R, Swearingen M, et al. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial. Cytotherapy 2008; 10:625.
  80. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365:725.
  81. Porter DL, Kalos M, Zheng Z, et al. Chimeric Antigen Receptor Therapy for B-cell Malignancies. J Cancer 2011; 2:331.
  82. Henkart PA. Lymphocyte-mediated cytotoxicity: two pathways and multiple effector molecules. Immunity 1994; 1:343.
  83. Chung WH, Hung SI, Yang JY, et al. Granulysin is a key mediator for disseminated keratinocyte death in Stevens-Johnson syndrome and toxic epidermal necrolysis. Nat Med 2008; 14:1343.
  84. Nagasawa M, Isoda T, Itoh S, et al. Analysis of serum granulysin in patients with hematopoietic stem-cell transplantation: its usefulness as a marker of graft-versus-host reaction. Am J Hematol 2006; 81:340.
  85. Heusel JW, Wesselschmidt RL, Shresta S, et al. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 1994; 76:977.
  86. Kägi D, Ledermann B, Bürki K, et al. Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice. Nature 1994; 369:31.
  87. Rouvier E, Luciani MF, Golstein P. Fas involvement in Ca(2+)-independent T cell-mediated cytotoxicity. J Exp Med 1993; 177:195.
  88. Oshimi Y, Oda S, Honda Y, et al. Involvement of Fas ligand and Fas-mediated pathway in the cytotoxicity of human natural killer cells. J Immunol 1996; 157:2909.
  89. Hahne M, Rimoldi D, Schröter M, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science 1996; 274:1363.
  90. Walker PR, Saas P, Dietrich PY. Role of Fas ligand (CD95L) in immune escape: the tumor cell strikes back. J Immunol 1997; 158:4521.
  91. Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002; 419:734.
  92. Inamoto Y, Flowers ME, Lee SJ, et al. Influence of immunosuppressive treatment on risk of recurrent malignancy after allogeneic hematopoietic cell transplantation. Blood 2011; 118:456.
  93. Claret EJ, Alyea EP, Orsini E, et al. Characterization of T cell repertoire in patients with graft-versus-leukemia after donor lymphocyte infusion. J Clin Invest 1997; 100:855.
  94. Ringdén O, Labopin M, Gorin NC, et al. Is there a graft-versus-leukaemia effect in the absence of graft-versus-host disease in patients undergoing bone marrow transplantation for acute leukaemia? Br J Haematol 2000; 111:1130.
  95. Anderson LD Jr, Savary CA, Mullen CA. Immunization of allogeneic bone marrow transplant recipients with tumor cell vaccines enhances graft-versus-tumor activity without exacerbating graft-versus-host disease. Blood 2000; 95:2426.
  96. Nishimura R, Baker J, Beilhack A, et al. In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood 2008; 112:2563.
  97. Choi J, Ritchey J, Prior JL, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood 2010; 116:129.
  98. Martelli MF, Di Ianni M, Ruggeri L, et al. HLA-haploidentical transplantation with regulatory and conventional T-cell adoptive immunotherapy prevents acute leukemia relapse. Blood 2014; 124:638.
  99. Brunstein CG, Miller JS, McKenna DH, et al. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood 2016; 127:1044.
  100. Meyer EH, Laport G, Xie BJ, et al. Transplantation of donor grafts with defined ratio of conventional and regulatory T cells in HLA-matched recipients. JCI Insight 2019; 4.
  101. Muffly L, Sheehan K, Armstrong R, et al. Infusion of donor-derived CD8+ memory T cells for relapse following allogeneic hematopoietic cell transplantation. Blood Adv 2018; 2:681.
  102. Dutt S, Baker J, Kohrt HE, et al. CD8+CD44(hi) but not CD4+CD44(hi) memory T cells mediate potent graft antilymphoma activity without GVHD. Blood 2011; 117:3230.
  103. Berger C, Jensen MC, Lansdorp PM, et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest 2008; 118:294.
  104. Dutt S, Narla A, Lin K, et al. Haploinsufficiency for ribosomal protein genes causes selective activation of p53 in human erythroid progenitor cells. Blood 2011; 117:2567.
  105. Sprangers B, Van Wijmeersch B, Fevery S, et al. Experimental and clinical approaches for optimization of the graft-versus-leukemia effect. Nat Clin Pract Oncol 2007; 4:404.
  106. Schmaltz C, Alpdogan O, Horndasch KJ, et al. Differential use of Fas ligand and perforin cytotoxic pathways by donor T cells in graft-versus-host disease and graft-versus-leukemia effect. Blood 2001; 97:2886.
  107. Fontaine P, Roy-Proulx G, Knafo L, et al. Adoptive transfer of minor histocompatibility antigen-specific T lymphocytes eradicates leukemia cells without causing graft-versus-host disease. Nat Med 2001; 7:789.
  108. Epperson DE, Margolis DA, McOlash L, et al. In vitro T-cell receptor V beta repertoire analysis may identify which T-cell V beta families mediate graft-versus-leukaemia and graft-versus-host responses after human leucocyte antigen-matched sibling stem cell transplantation. Br J Haematol 2001; 114:57.
  109. André-Schmutz I, Le Deist F, Hacein-Bey-Abina S, et al. Immune reconstitution without graft-versus-host disease after haemopoietic stem-cell transplantation: a phase 1/2 study. Lancet 2002; 360:130.
  110. Guimond M, Balassy A, Barrette M, et al. P-glycoprotein targeting: a unique strategy to selectively eliminate immunoreactive T cells. Blood 2002; 100:375.
  111. Morris ES, MacDonald KP, Hill GR. Stem cell mobilization with G-CSF analogs: a rational approach to separate GVHD and GVL? Blood 2006; 107:3430.
  112. Zeiser R, Nguyen VH, Beilhack A, et al. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood 2006; 108:390.
  113. Chakraverty R, Sykes M. The role of antigen-presenting cells in triggering graft-versus-host disease and graft-versus-leukemia. Blood 2007; 110:9.
  114. Barrett AJ. Understanding and harnessing the graft-versus-leukaemia effect. Br J Haematol 2008; 142:877.
  115. Boni A, Muranski P, Cassard L, et al. Adoptive transfer of allogeneic tumor-specific T cells mediates effective regression of large tumors across major histocompatibility barriers. Blood 2008; 112:4746.
  116. Cai SF, Cao X, Hassan A, et al. Granzyme B is not required for regulatory T cell-mediated suppression of graft-versus-host disease. Blood 2010; 115:1669.
  117. Goodyear OC, Dennis M, Jilani NY, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood 2012; 119:3361.
Topic 3546 Version 18.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟