Elsevier

Blood Reviews

Volume 22, Issue 2, March 2008, Pages 53-63
Blood Reviews

Review
Advances in the allogeneic transplantation for thalassemia

https://doi.org/10.1016/j.blre.2007.10.001Get rights and content

Summary

Hematopoietic stem cell transplantation (HSCT) remains the only curative option for patients with thalassemia. Current results of transplantation in patients aged less than 17 years from matched related donors offer 80% to 87% probability of cure according to risk classes. Adult thalassemics treated with myeloablative conditioning continue to have inferior results because of their advanced stage of disease. With the introduction of high-resolution tissue typing techniques transplant centres worldwide are able to offer allogeneic HSCT to a much larger cohort of patients who could not benefit from transplantation because of lack of matched family donor. Although limited number of patients treated, results of transplant from unrelated matched donors are comparable to those obtained using sibling donors. Graft failure or rejection remains a significant cause of transplant failure in patients with thalassemia making difficult to perform reduced intensity conditioning regimens. Mixed chimerism is a common phenomenon after transplantation and is a risk factor for rejection. Ex-thalassemics still carry the clinical complications acquired during years of transfusion and chelation therapy. Longer follow-up and management of these complications in ex-thalassemics are essential.

Introduction

The thalassemias refer to a diverse group of hemoglobin disorders characterized by a reduced synthesis of one or more of the globin chains (α, β, γ, δβ, γδβ, δ and εγδβ) and are the most common monogenic disorders to cause a major public health problem in the world.1 It is estimated that there are 270 million carriers of haemoglobin disorders, of which 80 million are carriers of β thalassemia. Worldwide about 60, 000 children with a major thalassaemia and 250, 000 with a sickle cell disorder are born annually, giving a rate of more than 2.4 affected children per 1,000 births.2 Though originally endemic to the tropics and subtropics, the thalassemias are now found worldwide as a result of migration and have become an important part of clinical practice in Europe, the US and Australasia.

In patients with β-thalassemia major the absent or extremely reduced production of the β chain of hemoglobin causes severe ineffective erythropoiesis, massive erythroid hyperplasia in the bone marrow and extramedullary sites, and hemolytic anemia necessitating chronic transfusions. Both ineffective erythropoiesis and chronic transfusion therapy inevitably lead to iron overload resulting in progressive multiple organ damage which cause endocrine deficiencies, liver disease, and cardiac disease leading to poor quality of life and increased mortality. Regular blood transfusion and iron chelation have improved both survival and quality of life of patients with thalassemia and have changed a previously fatal disease with early death to a chronic, although progressive disease compatible with prolonged survival.2, 3, 4 Despite the prolonged life expectancy, a recent study from U.K. Thalassemia Registry showed a steady decline in survival starting from the second decade, with fewer that 50% of patients remaining alive beyond 35 years mainly because of poor compliance with chelation therapy.5 In the developing world where thalassemia is more common, most children die before the age of 20 years because of the unavailability of safe blood products and/or expensive iron chelating drugs. Two recently developed oral iron-chelators (deferiprone and deferasirox) could allow better compliance with chelation, and therefore could have a favorable impact on survival of patients with thalassemia although their efficacy compared to desferrioxamine in the long-term has not yet been determined. However, even an ideal iron- chelator with rigorous adherence only substantially reduces, but does not eliminate, the iron overload of patients on lifelong transfusions.

Stem cell gene therapy for β-thalassemia requires gene transfer into autologous hematopoietic stem cells using integrating vectors that direct regulated expression of β globin at therapeutic levels. Despite promising results of gene-therapy in animal models, their clinical potential remains uncertain and the safety of these vectors to use for gene therapy of hemoglobinopathies remains to be seen. Early attempts using conventional oncoretroviral vectors carrying the human β globin gene and portions of the locus control region (LCR) have suffered from problems of vector instability, low titers and variable expression.6 Recently human immunodefficiency virus-based lentiviral (LV) vectors were shown to stably transmit the human β globin gene and a large LCR element, resulting in correction of the mouse thalassemia intermedia phenotype, with variable levels of β globin expression.7 The levels of β globin expression achieved from insulated self-inactivating LV vectors were sufficient to phenotypically correct the thalassemia phenotype from 4 patients with thalassemia major in vitro, and this correction persisted long term for up to 4 months, in xeno-transplanted mice in vivo.8 Accumulating preclinical and clinical data show that gene therapy may induce several unexpected side effects such as: 1) insertional mutagenesis; 2) interference of transgene product with cellular signaling networks; 3) loss of homing potential of genetically modified cells; 4) fusiogenic properties of viral envelop proteins; 5) elimination of modified transgenic cells by antibodies; 6) cytotoxic T cells directed against transgene-encoded antigens; and 7) side effects of regimen used to selectively engraft or expand manipulated cells.9

Hematopoietic stem cell transplantation (SCT) has been used in attempts at curing thalassemia. The first two transplant procedures for the treatment of thalassemia with marrow from matched related donors were performed in December 1981, in Seattle, WA, and in Pesaro, Italy. The Seattle approach was based on the assumption that the risks associated with BMT would be increased by the iron overload and by sensitization to human leukocyte antigens (HLAs) induced by hypertransfusion. Therefore, it was decided that early clinical studies would be conducted in very young patients who had received very few transfusions. On December 3, 1981 a 14-month-old child with β-thalassemia major who had been transfused with a total of 250 mL of packed RBCs received BMT from his HLA-identical sister in Seattle with successful outcome.10

The Pesaro approach was based on an assessment that restricting transplants to untransfused patients was impracticable. On December 17, 1981 the Pesaro team performed a transplant in a 16-year-old thalassemic patient who had received 150 RBC transfusions, using marrow from his HLA identical brother. This patient rejected the graft and was the first of an extensive series of transplants for thalassemia performed by the Pesaro Team.11, 12

The acute toxicities of transplantation are challenged by the observation that in the developed world patients with thalassemia are well served by medical treatment, and in some countries patients receiving regular blood transfusion and chelation could have a better survival rate.3, 13 Unfortunately, these same patients with increasing age currently experience poor outcome on conventional treatment, even in well-resourced countries with universal access to good medical treatment.5, 14

The purpose of allogeneic SCT for hemoglobinopathies is to correct the basic genetic defect by repleting genes essential for normal hematopoiesis through allogeneic stem cells as vectors following conditioning to overcome the immunological barrier. Therefore allogeneic SCT in these diseases could be considered as allogeneic stem cell gene therapy.

Advances in hematopoietic SCT, supportive care and tissue typing techniques have steadily led to consider this curative approach also for patients who lack matched related donors using alternative donors such as matched unrelated and mismatched related donors. In this review we provide our experience of transplantation in thalassemia and summarise the published data on SCT in this disease.

Section snippets

Preparatory regimens

Preparatory regimens for BMT of patients with thalassemia must achieve two objectives: elimination of the disordered marrow and establishment of a tolerant environment that will permit transplanted marrow to survive and thrive. Total body irradiation (TBI) can satisfy both these objectives, but there are many reasons to avoid the use of this marrow-ablative modality. These include the known growth-retarding effects of TBI in young children and the increased risk of secondary malignancies which

Class 1 and Class 2 patients

Between October 1985 and August 2007 five hundred and fifteen class 1 and class 2 patients with median age of 7 years (range 1 to 16 years) were given bone marrow transplantation following conditioning with Bu 3,5 mg/kg/day for 4 consecutive days and CY 50 mg/kg/day for subsequent 4 days. Since 2003 patients aged less than 4 years were also given thiotepa 10 mg/kg/day in addition to BUCY. Four hundred and seventy eight of these patients were given transplant at the Hospital of Pesaro12, 28 and

Alternative related donors

Approximately 25% to 30% of patients with thalassemia could have an HLA matched related donor. As HSCT is the only cure for thalassemia there is need to develop alternative stem cells donations. Our past experience with BMT from alternative donors for 29 patients with β-thalassemia major who received phenotypically matched grafts or haploidentical grafts mismatched for one, two, or three antigens was characterized by higher graft failure (55%), and low thalassemia-free survival (21%).45

Graft failure or rejection

Patients with engraftment failure and without functioning marrow have a bleak prospect because an early second transplant with a second course of conditioning is usually not a reasonable option. However, occasionally patients have late graft failure without thalassemia recurrence and in this situation second transplant attempts with intensive conditioning may provide the only treatment option to offer a chance of prolonged survival.

Patients who reject their grafts and have a return of host

Mixed chimerism

Mixed hematopoietic chimerism (MC) is a common phenomenon after myeloablative transplantation for thalassemia. In fact the incidence of MC at 2 months was 32.2%.53 While none of the patients with complete chimerism at 2 months rejected their grafts, 35 of 108 patients with MC determined at the same time lost the graft suggesting that MC after BMT for thalassemia is a risk factor for rejection. The percentages of residual host hematopoietic cells (RHCs) after transplant were predictive for graft

Reduced intensity hematopoietic stem cell transplantation

Unlike hematological malignancies where mixed chimerism may predict relapse, in nonmalignant disorders, the development of stable mixed chimerism has a potential for ameliorative effect that was well documented for β-thalassemia major and sickle cell anemia.54, 56 Stable mixed chimerism is not uncommon in thalassemia major patients after transplantation. It appears that when stable mixed chimerism is established, even a minority of donor cells is sufficient to overcome an underlying genetic

Management of the ex-thalassemics

After a successful transplantation, ex-thalassemic patients still carry the clinical complications acquired during years of transfusion and chelation therapy. Among the issues requiring long-term management in such patients are iron overload, chronic hepatitis, liver fibrosis and endocrine dysfunction. Management of these complications in ex-thalassemics is an important issue. Iron stores after transplantation remain elevated in most patients receiving HSCT in advanced stage of disease (class 2

Conclusion

Hematopoietic stem cell transplantation offers the only chance of cure and the return of both life expectancy and quality of life to normal. Current results of HSCT clearly demonstrate that 80% to 87% of patients with thalassemia could be cured if transplants are given from HLA-matched related donors. Improvements in molecular tissue typing have allowed allogeneic transplantation to be expanded to a much wider cohort of patients lacking a matched related donor with very encouraging results.

Acknowledgements

We thank all physicians, nurses, and support personnel at the Hospital of Pesaro and at the Mediterranean Institute of Hematology for their dedicated care of patients on this study.

References (67)

  • P. Di Bartolomeo et al.

    Long-term results of bone marrow transplantation for thalassemia major in Pescara

    Blood

    (2004)
  • S. Hongeng et al.

    Outcomes of transplantation with related and unrelated-donor stem cells in children with severe thalassemia

    Biol Blood Marrow Transplant

    (2006)
  • G. La Nasa et al.

    Unrelated donor bone marrow transplantation for thalassemia: the effect of extended haplotypes

    Blood

    (2002)
  • J.E. Wagner et al.

    Transplantation of unrelated donor unblical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival

    Blood

    (2002)
  • M. Andreani et al.

    Persistence of mixed chimerism in patients transplanted for the treatment of thalassemia

    Blood

    (1996)
  • M. Walters et al.

    Multicenter investigation of bone marrow transplantation for sickle cell disease. Stabel mixed haematopoietic chimerism after bone marrow transplantation for sickle cell anemia

    Biol Blood Marrow Transplant

    (2001)
  • R. Iannone et al.

    Results of minimally toxic nonmyeloablative transplantation in patients with sickle cell anemia and beta-thalassemia

    Biol Blood Marrow Transplant

    (2003)
  • G. Lucarelli et al.

    Fate of iron stores in thalassemia after bone marrow transplantation

    Lancet

    (1993)
  • E. Angelucci et al.

    Effects of iron overload and hepatitis C virus positivity in determining progression of liver fibrosis inthalassemia following bone marrow transplantation

    Blood

    (2002)
  • D.J. Weatheral et al.

    The Thalassemia Syndromes

    (2001)
  • B. Modell et al.

    A national register for surveillance of inherited disorders: b thalassaemia in the United Kingdom

    Bull World Health Organ

    (2001)
  • A. Piga et al.

    Mortality and morbidity in thalassemia with conventional treatment

    Bone Marrow Transplant

    (1977)
  • C. Borgna- Pignatti et al.

    Survival and complications in patients with thalassemia major treated with transfusions and deferrioxamine

    Haematologica

    (2004)
  • I. Pavlec et al.

    A human beta-globin gene fused to the human beta globin locus control region is expressed at high levels in erythroid cells of mice engrafted with retrovirus-transduced hematopoietic stem cells

    Blood

    (1993)
  • C. May et al.

    Therapeutic haemoglobin synthesis in beta –thalassemic mice expressing lentovirus-encoded human beta-globin

    Nature

    (2000)
  • P. Malik et al.

    Gene therapy for β-thalassemia

    Hematology

    (2005)
  • E.D. Thomas et al.

    Marrow transplantation for thalassaemia

    Lancet

    (1982)
  • G. Lucarelli et al.

    Marrow transplantation in patients with advanced thalassemia

    N Engl J Med

    (1987)
  • G. Lucarelli et al.

    Bone marrow transplantation in patients with thalassemia

    N Engl J Med

    (1990)
  • N.F. Olivieri et al.

    Survival in medically treated patients with homogygous beta-thalassemia

    N Engl J Med

    (1994)
  • M.J. Cunningham et al.

    Thalassemia Clinical Research Network. Complications of beta –thalassemia major in North America

    Blood

    (2002)
  • R.P. Witherspoon et al.

    Secondary cancers after bone marrow transplantation for leukemia or aplastic anemia

    N Engl J Med

    (1989)
  • R. Parkman et al.

    Complete correction of the Wiskott-Aldrich syndrome by allogeneic bone-marrow transplantation

    N Engl J Med

    (1978)
  • Cited by (0)

    1

    Tel.: +39 06 20661307; fax: +39 06 20661302.

    View full text