+ All Categories
Home > Documents > Mesenchymal Stromal Cells || MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment and...

Mesenchymal Stromal Cells || MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment and...

Date post: 18-Dec-2016
Category:
Upload: armand
View: 213 times
Download: 0 times
Share this document with a friend
12
443 P. Hematti and A. Keating (eds.), Mesenchymal Stromal Cells: Biology and Clinical Applications, Stem Cell Biology and Regenerative Medicine, DOI 10.1007/978-1-4614-5711-4_24, © Springer Science+Business Media New York 2013 Abstract Engraftment failure is a rare but life-threatening complication of hematopoietic cell transplantation (HCT). Newer approaches to HCT, including use of haploidentical donors, umbilical cord blood (UCB) transplant, nonmyeloablative and reduced-intensity conditioning regimens, appear to have an increased risk of graft failure. Multipotent mesenchymal stromal cells (MSCs) are essential bone marrow components that have the potential to differentiate in vitro into tissues along mesenchymal lineages, including bone marrow stroma. This regenerative potential, coupled with the capability to secrete cytokines and growth factors, suggests that MSCs would facilitate and promote hematopoiesis. Moreover, MSCs have immu- noregulatory properties and thus could have an additional application in the setting of HCT by reducing both graft rejection and graft-versus-host disease. Initial trials have demonstrated the safety and feasibility of infusion of ex vivo-expanded autolo- gous and allogeneic MSCs. Results from these early trials suggested MSCs may enhance hematopoiesis when infused at the time of HCT; however, subsequent trials have not yet provided confirmation that MSCs accelerate hematopoietic recovery when given shortly after HCT. Ongoing research initiatives include use of MSC infusions for patients who have some evidence of regenerating marrow but have delayed or incomplete hematopoiesis. P.F. Caimi, M.D. • H.M. Lazarus, M.D., F.A.C.P . (*) Department of Medicine, University Hospitals Case Medical Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA e-mail: [email protected] Chapter 24 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment and Poor Graft Function Paolo F. Caimi and Hillard M. Lazarus
Transcript

443P. Hematti and A. Keating (eds.), Mesenchymal Stromal Cells: Biology and Clinical Applications, Stem Cell Biology and Regenerative Medicine, DOI 10.1007/978-1-4614-5711-4_24, © Springer Science+Business Media New York 2013

Abstract Engraftment failure is a rare but life-threatening complication of hematopoietic cell transplantation (HCT). Newer approaches to HCT, including use of haploidentical donors, umbilical cord blood (UCB) transplant, nonmyeloablative and reduced-intensity conditioning regimens, appear to have an increased risk of graft failure. Multipotent mesenchymal stromal cells (MSCs) are essential bone marrow components that have the potential to differentiate in vitro into tissues along mesenchymal lineages, including bone marrow stroma. This regenerative potential, coupled with the capability to secrete cytokines and growth factors, suggests that MSCs would facilitate and promote hematopoiesis. Moreover, MSCs have immu-noregulatory properties and thus could have an additional application in the setting of HCT by reducing both graft rejection and graft-versus-host disease. Initial trials have demonstrated the safety and feasibility of infusion of ex vivo-expanded autolo-gous and allogeneic MSCs. Results from these early trials suggested MSCs may enhance hematopoiesis when infused at the time of HCT; however, subsequent trials have not yet provided con fi rmation that MSCs accelerate hematopoietic recovery when given shortly after HCT. Ongoing research initiatives include use of MSC infusions for patients who have some evidence of regenerating marrow but have delayed or incomplete hematopoiesis.

P. F. Caimi , M.D. • H. M. Lazarus , M.D., F.A.C.P . (*) Department of Medicine , University Hospitals Case Medical Center, Case Comprehensive Cancer Center, Case Western Reserve University , Cleveland , OH 44106 , USA e-mail: [email protected]

Chapter 24 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment and Poor Graft Function

Paolo F. Caimi and Hillard M. Lazarus

444 P.F. Caimi and H.M. Lazarus

Introduction

Hematopoietic cell transplantation (HCT) is a potentially curative therapy for a variety of benign and malignant hematologic diseases. In benign disorders, such as bone marrow failure syndromes, hemoglobinopathies, immunode fi ciencies, and enzyme de fi ciencies, infusion of the normal donor-derived hematopoietic progeni-tor cells (HPCs) can reconstitute normal hematopoiesis and provide immunocom-petent and metabolically intact progenitor cells. In malignant disorders, the autologous or allogeneic HPCs rescue patients from the myeloablative effects of escalated doses of chemotherapy; further, allogeneic immunocompetent cells may confer the added bene fi t of a “graft-versus-malignancy” (allogeneic) effect. The full clinical effectiveness of HCT is hampered by several barriers that relate both to the choice of the graft source as well as the type of conditioning regimen. In the autolo-gous HCT setting, delayed, poor, or absent engraftment may result from infusion of insuf fi cient numbers of HPCs. Umbilical cord blood (UCB) represents a valuable source of HPCs in patients who do not have available sibling-matched or matched unrelated donors, but slow engraftment of red cells, neutrophils, and platelets often is the norm. Other strategies include use of haploidentical donors (parents or chil-dren of the patient) that may be associated with an increased risk of engraftment failure [ 1 ] . In these situations, prolonged neutropenia carries a markedly increased risk of opportunistic infection. A prolonged time to restore production of red blood cells and platelets is associated with the dangers of increased alloimmunization, iron overload, and transmission of infections, in addition to the inconvenience of frequently receiving blood products and the associated signi fi cant fi nancial cost.

Myeloablative conditioning regimens are fraught with added danger when administered to frail or elderly patients and to those with signi fi cant comorbidities. Further, graft-versus-host disease (GVHD) with or without opportunistic infection remains a signi fi cant complication of the posttransplant state. Reduced-intensity conditioning (RIC) and nonmyeloablative conditioning have been used with increased frequency to facilitate allogeneic HCT in patients who cannot tolerate the signi fi cantly higher-dose myeloablative chemotherapy. These approaches rely pre-dominantly upon the donor effector cells in the graft to exert the therapeutic alloge-neic or immune effect. Graft failure, however, is an important complication of RIC and nonmyeloablative allogeneic HCT, with rates reported between 18 and 42%, depending on the regimen used [ 2 ] . Finally, in vitro T cell depletion of the graft, for a variety of reasons, may be associated with engraftment failure states [ 3 ] .

Multipotent Mesenchymal Stromal Cells (MSCs)

Multipotent mesenchymal stromal cells (MSCs) are essential components of the bone marrow microenvironment. These cells provide a supporting physical matrix and elaborate a variety of cytokines and other factors that have been shown in vitro and in vivo to support hematopoiesis [ 4 ] . As a result, over the last two decades,

44524 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment…

MSCs have been the object of intensive research [ 5, 6 ] . These cells have the capacity to differentiate in vitro along mesenchymal lineages and give rise to multiple tis-sues, including bone, cartilage, adipose tissue, and bone marrow stroma; they also exhibit intense paracrine activity, secreting bioactive molecules with trophic and immunomodulatory capacity [ 7 ] . When infused, MSCs home to tissue sites of active in fl ammation [ 8 ] and participate in tissue repair [ 7 ] . The tissue regenerative poten-tial has prompted interest in using MSCs to provide stability and restore function in organs such as the heart and nervous system, using intravenous as well as intrathe-cally administered MSCs [ 9– 11 ] ; the immunomodulatory or immunosuppressive activity of MSCs has led to novel research initiatives in autoimmune conditions [ 12 ] and in the treatment of GVHD. The combination of these characteristics, e.g., the capacity to enhance regeneration of the hematopoietic process and the bone marrow stroma, the immunomodulatory properties [ 13 ] , and the secretion of bioactive agents [ 14 ] , has led to the study of the role of MSCs in facilitating HPC engraftment and the prevention and treatment of graft failure after HCT.

Biologic Properties of MSCs

First identi fi ed by Friedenstein and colleagues in postnatal bone marrow more than 40 years ago [ 5, 15 ] , MSCs have been isolated from a wide variety of adult organs, including periosteum, muscle connective tissue, perichondrium, and adipose tissue [ 16– 19 ] . These cells also are present in fetal bone marrow, lung, liver, and spleen [ 20, 21 ] . Amniotic fl uid and placenta are rich sources of MSCs, the latter containing both maternal and fetal MSCs [ 22, 23 ] . Their isolation from UCB has been reported with variable success [ 24 ] . The proportion of MSCs in bone marrow is small and varies among species, representing 0.001–0.01% of all nucleated marrow cells. Moreover, their number decreases with age, ranging from 1 MSC per 10,000 nucle-ated marrow cells in newborns to 1 MSC per 250,000 nucleated marrow cells in adults [ 25 ] . This paucity of MSCs in bone marrow and other tissues, along with the lack of speci fi c markers, has made study of directly isolated cells dif fi cult, and thus, little is known about the primary progenitor cell in vivo. On the other hand, MSCs have a remarkable capacity to undergo expansion in ex vivo culture settings, which has led to two important developments. First, most of the information regarding MSC function and phenotype is based on cells expanded in vitro. Secondly, as a small bone marrow sample can be expanded to yield hundreds of millions of cells, clinical use of culture-expanded MSCs has been made possible, and thus, clinical studies have been performed before a reliable preclinical animal model was readily available [ 26 ] .

The wide variety of tissues from which MSCs can be harvested and the multiple available methods for isolation and expansion of cells prompted the International Society for Cellular Therapy (ISCT) to de fi ne minimal criteria for the de fi nition of MSCs [ 6 ] . These criteria include the adherence to plastic under standard culture conditions; a speci fi c phenotype ( ³ 95% of the cells expressing CD105, CD73, and

446 P.F. Caimi and H.M. Lazarus

CD90 and £ 2% of cells expressing CD45, CD34, CD14, CD11b, CD79a, or CD19 or HLA Class II); and the capacity to differentiate into osteoblasts, chondrocytes, or adipocytes under standard in vitro differentiating conditions. The population of plastic-adherent cells, however, is heterogeneous and only a small proportion of these cells can generate fi broblast colonies in vitro [ 27 ] . In 2005 the ISCT clari fi ed the nomenclature for “mesenchymal stem cells,” a term popularized in early 1990s by Caplan [ 25 ] , to the current “multipotent mesenchymal stromal cells.” This des-ignation conveys the multipotentiality and tissue-regenerating capacity without ascribing to them the homogeneous quality of stem cells [ 25 ] .

The range and mechanisms of the immunomodulatory properties of MSCs have not yet been fully elucidated. MSCs have been shown to suppress T cell proliferation and cytokine production in response to alloantigens and nonspeci fi c mitogens [ 28, 29 ] . These cells appear to exert their in fl uence both by soluble fac-tors (IFN-gamma and nitric oxide appear to have a central role [ 30, 31 ] ) as well as by direct cell-cell interactions [ 32 ] . MSCs also inhibit the proliferation, differen-tiation, and cytokine secretion of dendritic cells [ 33– 35 ] and natural killer (NK) cells leading to decreased cell-mediated cytotoxic activity [ 36 ] . B cell prolifera-tion, chemotaxis, antibody production, and terminal differentiation also are sup-pressed by MSCs [ 37– 39 ] .

In addition to their immunomodulatory properties, MSCs appear to be immuno-logically privileged cells [ 40 ] . Culture-expanded human MSCs express major histo-compatibility complex (MHC) class I proteins, are negative for MHC class II proteins, and appear to lack expression of the costimulatory molecules CD80, CD86, and CD40 [ 41– 44 ] . MHC class I expression and upregulation in the presence of increased interferon-gamma make MSCs less susceptible to NK cell lysis. MSCs can suppress the proliferation of CD4+ and CD8+ T lymphocytes [ 32 ] while selec-tively promoting proliferation of CD4+ CD25+ T regulatory lymphocytes [ 45 ] . When cocultured with allogeneic lymphocytes, MSCs did not induce lymphocyte proliferation [ 46 ] ; and in animal models, infusion of allogeneic mismatched MSCs did not induce an immune response [ 8 ] . Moreover, the immunosuppressive effect of MSCs appears to be independent from MHC compatibility status [ 29 ] . Such data provide a scienti fi c basis for undertaking investigations of infusing MSCs obtained from unrelated or mismatched donors for immunomodulation and promotion of engraftment in the setting of HCT.

Clinical Studies

Early Studies

As a result of differences in the immunoregulatory properties of MSCs between species, no adequate preclinical animal models are available to predict the in vivo function of MSCs in the HCT setting [ 47 ] . The establishment of human MSC expan-sion methods, however, has facilitated their clinical use. The consequence has been

44724 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment…

a unique situation in which clinical trials of MSC infusion were initiated before complete understanding of the in vivo properties of these cells [ 48 ] .

In 1995 Lazarus and colleagues [ 49 ] published the fi rst clinical trial using MSCs. This pilot study examined collection, ex vivo expansion, and reinfusion of autolo-gous bone marrow-derived MSCs obtained from 23 hematologic malignancy patients. The procedure, demonstrated to be safe and feasible, was followed by a phase I–II trial of autologous, culture-expanded bone marrow-derived MSC infu-sion after high-dose chemotherapy and autologous peripheral blood progenitor cell transplantation in 28 breast cancer patients [ 50 ] . No toxic effects directly attribut-able to MSC infusion were observed and hematologic recovery was rapid. Blood neutrophil engraftment (neutrophils > 500/ m L) occurred at a median of 8 days and untransfused platelet count exceeded 20,000/ m L in a median of 8.5 days. Unfortunately, no fi rm conclusions regarding the bene fi t in hematopoietic engraft-ment could be drawn due to the nonrandomized nature of this trial.

Frassoni and coworkers [ 51 ] retrospectively examined a matched-pair analysis of 31 hematologic malignancy patients undergoing HLA-identical, sibling-matched HCT procedures at the US and European centers. Patients received culture-expanded bone marrow-derived MSCs harvested from the HLA-identical HPC donors. Compared with historic controls, study patients had a statistically signi fi cant lower incidence of acute and chronic GVHD and superior survival rates after 6 months of follow-up [ 51 ] . No observations were reported regarding engraftment rates. These initial trials spearheaded the use of human MSCs, demonstrating the safety and feasibility of their use, both in the autologous and HLA-identical allogeneic setting, and suggested these cells may have therapeutic potential in the HCT setting, as engraftment enhancers or prophylaxis of GVHD.

Infusion of MSCs for Enhancement of Hematopoietic Engraftment

Recently, several trials have been undertaken to evaluate the coadministration of bone marrow-derived, culture-expanded MSCs along with varied sources of HPCs during allogeneic HCT for the purpose of correcting or preventing engraftment failure. The Karolinska University clinical group [ 52 ] reported seven patients who underwent cotransplantation of HPCs and MSCs. Three patients were treated for a previous graft failure or rejection. The remaining four were part of a pilot study designed to enhance hematopoietic engraftment in which haploidentical MSCs were given. Despite remarkable variability in the patient population, source of HPCs and MSCs, and HLA compatibility status of donors, all patients had hematopoietic engraftment, with median time to neutrophil engraftment (ANC > 0.5 × 10 9 /L) of 12 days (range 10–28 days) and median time to platelet engraftment (platelets > 30 × 10 9 /L) of 12 days (range 8–36 days). While these are encouraging results, the patient popula-tion, underlying diagnoses and transplant settings were markedly heterogeneous, making dif fi cult a generalization of the fi ndings on the effects of MSCs.

448 P.F. Caimi and H.M. Lazarus

Ball and colleagues [ 53 ] cotransplanted MSCs and HPCs obtained from haploidentical related donors into 14 children who had hematologic malignancies, immune de fi ciencies, and nonmalignant disorders. HPC engraftment was demon-strated in all patients. Mean MSC dose was 1.6 × 10 6 MSC/kg (range 1–3.3 × 10 6 MSC/kg). When compared to historic controls ( n = 47), the 14 study patients had comparable platelet and neutrophil recoveries yet faster attainments of a total blood leukocyte count > 1 × 10 9 /L. Acute and chronic GVHD rates were comparable with historic controls.

Macmillan and colleagues [ 54 ] infused ex vivo-expanded MSCs in conjunction with an UCB transplant. Fifteen children with hematologic malignancies received UCB grafts; 8 subjects also received parental haploidentical MSCs infusions on the day of UCB transplant, while 3 received repeat MSC infusions 21 days later. A second MSC infusion could not be given to the remaining fi ve patients secondary to insuf fi cient growth of parental MSCs. The median MSC dose on day 0 was 2.1 × 10 6 MSC/kg (range 0.9–5 × 10 6 MSC/kg). Haploidentical MSC infusions at the time of UCB transplant were shown to be safe, but tempo to recovery of neutrophil and platelet engraftment as well as the rates of acute GVHD were similar to those of historic controls.

A recent multicenter European trial explored the coinfusion of culture-expanded parental MSCs in 13 children who received UCB grafts obtained from related or unrelated donors [ 55 ] . Median MSC dose was 1.9 × 10 6 MSC/kg (range 1–3.9 × 10 6 MSC/kg). The incidence of graft failure or the rate of neutro-phil and platelet engraftment did not differ statistically from that observed in 39 historic controls matched for the diagnosis and type of UCB donor. Overall sur-vival rates also did not differ. On the other hand, there were no cases of severe acute GVHD (grades III–IV) in patients who received MSC coinfusion, com-pared to 10 (grade III, 7 cases and grade IV, 3 cases) of 39 patients in the historic controls. The authors postulated that in the UCB transplant setting, graft failure is more a function of the low numbers of infused HPCs rather than an immune-mediated mechanism; hence, MSC coinfusion may not be justi fi ed in the UCB transplant setting. However, MSCs may enhance hematopoietic engraftment via additional, non-immune- mediated mechanisms, such as cytokine release and reconstitution of the bone marrow stroma, effects that may be of particular importance in cases with borderline hematopoiesis.

Gonzalo-Daganzo et al. reported [ 56 ] a phase I–II study in which nine hemato-logic malignancy patients received third-party MSCs 1–24 h after coinfusion of UCB grafts and third-party HPCs (UCB/HPC). The median (range) MSC dose was 1.18 (1.04–2.22) × 10 6 MSC/kg. No adverse effects of MSC infusion were observed. Hematopoietic engraftment and achievement of full UCB chimerism appeared to be delayed in the study group when compared with 46 controls that received UCB/HPC coinfusion alone, although the differences were not statistically signi fi cant. The incidence of acute GVHD also did not differ statistically from that observed in control patients. Two patients who received additional subsequent MSC infusions for treatment of corticosteroid-refractory GVHD attained complete responses. This study showed that MSC infusion at the time of cotransplantation of UCB/HPC is

44924 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment…

safe and well tolerated, but the small number of patients included precludes further conclusions from being drawn regarding the effect of MSCs on engraftment and GVHD incidence.

Finally, Baron and colleagues [ 57 ] reported a recent study using cotransplan-tation of MSCs and HLA-mismatched MSCs after nonmyeloablative condition-ing. Twenty hematologic malignancy patients were compared to 16 historic controls who also received nonmyeloablative conditioning and HLA-mismatched HCT. MSC cotransplantation was associated with a decrease in 1-year nonre-lapse mortality (HR = 0.2, 95% CI 0.04–0.9, p = 0.03) and 1-year overall mortal-ity (HR = 0.4, 95% CI 0.1–0.9, p = 0.03). Severe, acute GVHD was low in the cotransplantation group, without an increased relapse risk, suggesting that GVHD was ameliorated without abrogating the graft-versus-tumor effect. Engraftment was prompt in both groups but there was no discernible enhance-ment of engraftment with MSC coinfusion; one patient receiving MSCs experi-enced primary graft failure. This study shows that MSC cotransplantation is feasible in the setting of nonmyeloablative HLA-mismatched HCT, where the risk of GVHD and graft rejection is higher, but no clear engraftment bene fi t was reported.

In summary, studies of MSC infusion at the time of HCT aimed at enhancement of engraftment, and prevention of HPC graft failure have not yielded conclusive results. While safety of MSC infusion largely has been demonstrated, the studies assessing the effect of MSC infusion on engraftment have been limited to small, nonrandomized studies that utilize historic controls. Results have been variable and several studies have failed to demonstrate engraftment enhancement with MSCs. Larger, randomized studies are needed to settle whether the use of these cells is justi fi ed.

Infusion of MSCs for Treatment of Graft Failure

Primary graft failure after HCT represents a rare but serious, life-threatening com-plication of the HCT procedure. Management strategies include use of recombinant hematopoietic growth factors, modi fi cations of immunosuppressant regimens, infu-sion of “backup” autologous HPCs, or second allogeneic HCT. Outcomes remain dismal, even after second allogeneic HCT, as evidenced by a recent observational study from the Center for International Blood and Marrow Transplant Research (CIBMTR) [ 58 ] . Such patients with prolonged bone marrow failure are at high risk of succumbing to infection and hemorrhage.

In addition to development of strategies for early identi fi cation of those patients at high risk of engraftment failure, new therapeutic alternatives are needed for this complication. MSCs may prove of value in this setting, with their bone marrow stromal regenerative potential and the additional capacity to ameliorate graft rejec-tion without signi fi cantly increasing risk of infection.

450 P.F. Caimi and H.M. Lazarus

Fouillard and colleagues [ 59 ] reported a case of a 40-year-old woman with AML in complete remission who exhibited primary graft failure after autologous HCT. Partial recovery of her counts was achieved with administration of granulocyte colony-stimulating factor and erythropoietin therapy administered three times a week. Three years after HCT she received culture-expanded MSCs harvested from her HLA-mismatched brother. MSC dose was 2.78 × 10 6 /kg. Rapid and sustained recovery of her neutrophil and platelet counts was observed, while no effect on hemoglobin concentration was observed. Granulocyte-macrophage colony-forming units (CFU-GM) and colony-forming unit fi broblasts (CFU-F) were increased 1 month and 1 year after MSC infusion. There were no side effects or adverse reac-tions to the MSC infusion and no GVHD was observed. Studies of MSC engraft-ment in the recipient 1 month post MSC infusion showed male DNA was detected at a frequency of 10 −5 per cell, whereas 1 year after infusion, it was no longer detect-able, results that are compatible with very low levels of MSC engraftment observed in animal studies. This report suggests that MSCs can potentially be used for treat-ment of engraftment failure, although the mechanism mediating their bene fi t is not yet elucidated.

Fang and colleagues [ 60 ] reported two pediatric patients with severe aplastic anemia (SAA) who presented with graft failure after receiving HLA-identical sibling peripheral blood HCT. The fi rst case, an 11-year-old girl, had relapse of SAA after her second HLA-identical sibling HCT. Haploidentical culture-expanded adipose tissue MSCs harvested from the patient’s mother were infused after a third infusion of HPCs from her HLA-identical sister. Neutrophil count reached 0.6 × 10 9 /L by 16 days after HCT, while the platelet count apparently recovered by 20 days, although the details were not provided by the authors. No acute or chronic GVHD was observed. The second case was a 12-year-old boy with SAA in relapse after HLA-identical sibling HCT. A second HCT from the same donor was followed by infusion of culture-expanded adipose tissue MSCs harvested from his mother. Neutrophil recovery occurred by 15 days while self-sustaining platelet count was achieved by 19 days. Two months after transplant the patient experienced “grade alpha” (apparently grade I) acute GVHD of the skin that completely responded to corticosteroid therapy. Both patients had sus-tained hematopoietic function beyond 2 years at the time of publication. Several reports have documented the safety and feasibility of infusion of culture-expanded MSCs harvested from adipose tissue [ 61, 62 ] . These reports suggest that coinfusion with HPCs may improve the rates of graft failure. Although referred to as equivalent to marrow-derived MSCs, experience with this source of cells is still limited and it is possible that there are subtle biologic and immu-nologic differences not yet identi fi ed. These positive results warrant proceeding with larger studies of MSC infusion and coinfusion with HPCs for treatment of engraftment failure.

Meuleman and coauthors [ 63 ] recently reported the results of a pilot clinical trial of infusion of culture-expanded MSCs without coinfusion of HPCs. Patients included had received HCT and achieved full donor chimerism but with poor engraftment. The authors de fi ned the latter as persistent posttransplant

45124 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment…

pancytopenia, with blood neutrophils < 1 × 10 9 /L and platelets < 50 × 10 9 /L at 30 days after HCT and despite treatment with a minimum of 10 days of granulo-cyte colony-stimulating factor. Patients with complete engraftment failure or rejection were excluded from this study. Six patients were included, all received myeloablative, allogeneic, mobilized peripheral blood HCT; three patients received HLA-identical related donor and three haploidentical related donor hematopoietic grafts. Although full donor chimerism was observed in all cases, bone marrow examination showed hypoplasia and all patients had varying degrees of pancytopenia. MSCs were obtained from bone marrow aspiration of the origi-nal HPC donors. The MSC dose was 1 × 10 6 MSC/kg. No acute side effects to MSC infusion were observed. Two patients manifested hematologic recovery after MSC infusion; both had received HLA-identical sibling HCTs and were in fi rst complete remission, in comparison to the more heavily pretreated other sub-jects. One patient presented early CMV infection (day 12) and subsequently died several months later from repeat CMV infection. The relationship between the viral infection and MSC infusion is unclear, as MSCs have not been shown to affect virus-speci fi c T cell function [ 64, 65 ] . This study demonstrates the feasibil-ity of MSC infusion without HPCs and suggests MSCs may aid in hematopoietic recovery in those patients who have residual hematopoiesis, likely through a com-bination of stromal reconstruction, cytokines, and modulation of rejection. Therefore, heavily pretreated patients or those with profound pancytopenia may bene fi t from larger doses of MSCs and possibly with coinfusion with HPCs as other investigators [ 52 ] have observed.

Conclusions

Although MSCs have been the subject of intensive laboratory and clinical research over the last 15 years, their in vivo properties and effects after administration in the clinical setting of HCT have not yet been fully established. Their biologic proper-ties, including constitutive secretion of bioactive molecules, capacity to differenti-ate into bone marrow stroma, and remarkable immunomodulatory capacity, have suggested that these cells may have a role to facilitate hematopoietic engraftment and prevent development of GVHD.

Small pilot studies demonstrated the safety of infusion of HLA-compatible MSCs and suggested a possible bene fi t in hematopoietic engraftment rates. Subsequent studies have demonstrated the feasibility of HLA-haploidentical and HLA-unmatched MSCs in a variety of settings, including coinfusion with HLA-identical and haploidentical peripheral blood HPCs as well as with UCB grafts. These studies, however, have not conclusively demonstrated that MSCs provide an advantage in terms of hematopoietic engraftment rates. Larger, randomized studies have been performed to evaluate the effect of MSCs for treatment and prophylaxis of GVHD. In the next few years, we anticipate the design and conduct of trials to evaluate the effect of MSC infusion on hematopoietic engraftment.

452 P.F. Caimi and H.M. Lazarus

References

1. Copelan EA (2006) Hematopoietic stem-cell transplantation. N Engl J Med 354:1813–1826 2. Leung AY, Kwong YL (2010) Haematopoietic stem cell transplantation: current concepts and

novel therapeutic strategies. Br Med Bull 93:85–103 3. Ho VT, Soiffer RJ (2001) The history and future of T-cell depletion as graft-versus-host dis-

ease prophylaxis for allogeneic hematopoietic stem cell transplantation. Blood 98:3192–3204 4. Maitra B, Szekely E, Gjini K et al (2004) Human mesenchymal stem cells support unrelated donor

hematopoietic stem cells and suppress T-cell activation. Bone Marrow Transplant 33:597–604 5. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP (1968) Heterotopic of bone marrow.

Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 6:230–247 6. Dominici M, Le Blanc K, Mueller I et al (2006) Minimal criteria for de fi ning multipotent

mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317

7. Bernardo ME, Locatelli F, Fibbe WE (2009) Mesenchymal stromal cells. Ann N Y Acad Sci 1176:101–117

8. Devine SM, Cobbs C, Jennings M et al (2003) Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 101:2999–3001

9. Hare JM, Traverse JH, Henry TD et al (2009) A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 54:2277–2286

10. Hu SL, Luo HS, Li JT et al (2010) Functional recovery in acute traumatic spinal cord injury after transplantation of human umbilical cord mesenchymal stem cells. Crit Care Med 38:2181–2189

11. Karussis D, Karageorgiou C, Vaknin-Dembinsky A et al (2010) Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyo-trophic lateral sclerosis. Arch Neurol 67:1187–1194

12. Liang J, Zhang H, Hua B et al (2010) Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study. Ann Rheum Dis 69:1423–1429

13. Tolar J, Le Blanc K, Keating A, Blazar BR (2010) Concise review: hitting the right spot with mesenchymal stromal cells. Stem Cells 28:1446–1455

14. Caplan AI (2009) Why are MSCs therapeutic? New data: new insight. J Pathol 217:318–324 15. Friedenstein AJ, Deriglasova UF, Kulagina NN et al (1974) Precursors for fi broblasts in differ-

ent populations of hematopoietic cells as detected by the in vitro colony assay method. Exp Hematol 2:83–92

16. Nakahara H, Goldberg VM, Caplan AI (1991) Culture-expanded human periosteal-derived cells exhibit osteochondral potential in vivo. J Orthop Res 9:465–476

17. Lee JY, Qu-Petersen Z, Cao B et al (2000) Clonal isolation of muscle-derived cells capable of enhancing muscle regeneration and bone healing. J Cell Biol 150:1085–1100

18. Arai F, Ohneda O, Miyamoto T et al (2002) Mesenchymal stem cells in perichondrium express activated leukocyte cell adhesion molecule and participate in bone marrow formation. J Exp Med 195:1549–1563

19. Zuk PA, Zhu M, Mizuno H et al (2001) Multilineage cells from human adipose tissue: implica-tions for cell-based therapies. Tissue Eng 7:211–228

20. Campagnoli C, Roberts IA, Kumar S et al (2001) Identi fi cation of mesenchymal stem/progeni-tor cells in human fi rst-trimester fetal blood, liver, and bone marrow. Blood 98:2396–2402

21. In ’t Anker PS, Noort WA, Scherjon SA et al (2003) Mesenchymal stem cells in human sec-ond-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica 88:845–852

22. In ’t Anker PS, Scherjon SA, Kleijburg-van der Keur C et al (2003) Amniotic fl uid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102:1548–1549

23. Miao Z, Jin J, Chen L et al (2006) Isolation of mesenchymal stem cells from human placenta: comparison with human bone marrow mesenchymal stem cells. Cell Biol Int 30:681–687

45324 MSCs for Enhancement of Hematopoietic Progenitor Cell Engraftment…

24. Lee OK, Kuo TK, Chen WM et al (2004) Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood 103:1669–1675

25. Caplan AI (2007) Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 213:341–347

26. Caimi PF, Reese J, Lee Z, Lazarus HM (2010) Emerging therapeutic approaches for multipo-tent mesenchymal stromal cells. Curr Opin Hematol 17:505–513

27. Horwitz EM, Le Blanc K, Dominici M et al (2005) Clari fi cation of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy 7:393–395

28. Rasmusson I, Ringden O, Sundberg B, Le Blanc K (2005) Mesenchymal stem cells inhibit lymphocyte proliferation by mitogens and alloantigens by different mechanisms. Exp Cell Res 305:33–41

29. Le Blanc K, Tammik L, Sundberg B et al (2003) Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompat-ibility complex. Scand J Immunol 57:11–20

30. Ren G, Zhang L, Zhao X et al (2008) Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2:141–150

31. Polchert D, Sobinsky J, Douglas G et al (2008) IFN-gamma activation of mesenchymal stem cells for treatment and prevention of graft versus host disease. Eur J Immunol 38:1745–1755

32. Krampera M, Glennie S, Dyson J et al (2003) Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-speci fi c T cells to their cognate peptide. Blood 101:3722–3729

33. Jiang XX, Zhang Y, Liu B et al (2005) Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 105:4120–4126

34. Nauta AJ, Kruisselbrink AB, Lurvink E et al (2006) Mesenchymal stem cells inhibit genera-tion and function of both CD34+−derived and monocyte-derived dendritic cells. J Immunol 177:2080–2087

35. Zhang B, Liu R, Shi D et al (2009) Mesenchymal stem cells induce mature dendritic cells into a novel Jagged-2-dependent regulatory dendritic cell population. Blood 113:46–57

36. Spaggiari GM, Capobianco A, Abdelrazik H et al (2008) Mesenchymal stem cells inhibit natu-ral killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-diox-ygenase and prostaglandin E2. Blood 111:1327–1333

37. Asari S, Itakura S, Ferreri K et al (2009) Mesenchymal stem cells suppress B-cell terminal differentiation. Exp Hematol 37:604–615

38. Corcione A, Benvenuto F, Ferretti E et al (2006) Human mesenchymal stem cells modulate B-cell functions. Blood 107:367–372

39. Chabannes D, Hill M, Merieau E et al (2007) A role for heme oxygenase-1 in the immunosup-pressive effect of adult rat and human mesenchymal stem cells. Blood 110:3691–3694

40. Sundin M, Barrett AJ, Ringden O et al (2009) HSCT recipients have speci fi c tolerance to MSC but not to the MSC donor. J Immunother 32:755–764

41. Majumdar MK, Keane-Moore M, Buyaner D et al (2003) Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci 10:228–241

42. Le Blanc K, Tammik C, Rosendahl K et al (2003) HLA expression and immunologic proper-ties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 31:890–896

43. Chan JL, Tang KC, Patel AP et al (2006) Antigen-presenting property of mesenchymal stem cells occurs during a narrow window at low levels of interferon-gamma. Blood 107:4817–4824

44. Parekkadan B, Milwid JM (2010) Mesenchymal stem cells as therapeutics. Annu Rev Biomed Eng 12:87–117

45. English K, Ryan JM, Tobin L et al (2009) Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25(high) forkhead box P3+ regulatory T cells. Clin Exp Immunol 156:149–160

46. Klyushnenkova E, Mosca JD, Zernetkina V et al (2005) T cell responses to allogeneic human mesenchymal stem cells: immunogenicity, tolerance, and suppression. J Biomed Sci 12:47–57

47. Le Blanc K, Ringden O (2007) Immunomodulation by mesenchymal stem cells and clinical experience. J Intern Med 262:509–525

454 P.F. Caimi and H.M. Lazarus

48. Cohen JL, Sudres M (2009) A role for mesenchymal stem cells in the control of graft-versus-host disease. Transplantation 87:S53–S54

49. Lazarus HM, Haynesworth SE, Gerson SL et al (1995) Ex vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Transplant 16:557–564

50. Koc ON, Gerson SL, Cooper BW et al (2000) Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol 18:307–316

51. Frassoni F, Labopin M, Bacigalupo A et al (2002) Expanded mesenchymal stem cells (MSC) co-infused with HLA identical stem cell transplants, reduce acute and chronic graft versus host disease: a matched pair analysis. Bone Marrow Transplant 29:S2

52. Le Blanc K, Samuelsson H, Gustafsson B et al (2007) Transplantation of mesenchymal stem cells to enhance engraftment of hematopoietic stem cells. Leukemia 21:1733–1738

53. Ball LM, Bernardo ME, Roelofs H et al (2007) Cotransplantation of ex vivo expanded mesen-chymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 110:2764–2767

54. Macmillan ML, Blazar BR, DeFor TE, Wagner JE (2009) Transplantation of ex-vivo culture-expanded parental haploidentical mesenchymal stem cells to promote engraftment in pediatric recipients of unrelated donor umbilical cord blood: results of a phase I–II clinical trial. Bone Marrow Transplant 43:447–454

55. Bernardo ME, Ball LM, Cometa AM et al (2011) Co-infusion of ex vivo-expanded, parental MSCs prevents life-threatening acute GVHD, but does not reduce the risk of graft failure in pediatric patients undergoing allogeneic umbilical cord blood transplantation. Bone Marrow Transplant 46:200–207

56. Gonzalo-Daganzo R, Regidor C, Martin-Donaire T et al (2009) Results of a pilot study on the use of third-party donor mesenchymal stromal cells in cord blood transplantation in adults. Cytotherapy 11:278–288

57. Baron F, Lechanteur C, Willems E et al (2010) Cotransplantation of mesenchymal stem cells might prevent death from graft-versus-host disease (GVHD) without abrogating graft-versus-tumor effects after HLA-mismatched allogeneic transplantation following nonmyeloablative conditioning. Biol Blood Marrow Transplant 16:838–847

58. Schriber J, Agovi MA, Ho V et al (2010) Second unrelated donor hematopoietic cell transplan-tation for primary graft failure. Biol Blood Marrow Transplant 16:1099–1106

59. Fouillard L, Chapel A, Bories D et al (2007) Infusion of allogeneic-related HLA mismatched mesenchymal stem cells for the treatment of incomplete engraftment following autologous haematopoietic stem cell transplantation. Leukemia 21:568–570

60. Fang B, Li N, Song Y et al (2009) Cotransplantation of haploidentical mesenchymal stem cells to enhance engraftment of hematopoietic stem cells and to reduce the risk of graft failure in two children with severe aplastic anemia. Pediatr Transplant 13:499–502

61. Fang B, Song Y, Liao L et al (2007) Favorable response to human adipose tissue-derived mesenchy-mal stem cells in steroid-refractory acute graft-versus-host disease. Transplant Proc 39:3358–3362

62. Fang B, Song Y, Zhao RC et al (2007) Using human adipose tissue-derived mesenchymal stem cells as salvage therapy for hepatic graft-versus-host disease resembling acute hepatitis. Transplant Proc 39:1710–1713

63. Meuleman N, Tondreau T, Ahmad I et al (2009) Infusion of mesenchymal stromal cells can aid hematopoietic recovery following allogeneic hematopoietic stem cell myeloablative trans-plant: a pilot study. Stem Cells Dev 18:1247–1252

64. Sundin M, Orvell C, Rasmusson I et al (2006) Mesenchymal stem cells are susceptible to human herpesviruses, but viral DNA cannot be detected in the healthy seropositive individual. Bone Marrow Transplant 37:1051–1059

65. Karlsson H, Samarasinghe S, Ball LM et al (2008) Mesenchymal stem cells exert differential effects on alloantigen and virus-speci fi c T-cell responses. Blood 112:532–541


Recommended