Allogeneic Hematopoietic Stem-Cell Transplantation for Genetic Diseases and Acquired Anemias - CAM 80122HB

Description
A number of inherited and acquired conditions have the potential for severe and/or progressive disease. For some conditions, allogeneic hematopoietic cell transplantation (allo-HCT) has been used to alter the natural history of the disease or potentially offer a cure.

For individuals who have a hemoglobinopathy, bone marrow failure syndrome, primary immunodeficiency, inherited metabolic syndrome disease (specifically those other than Hunter, Sanfilippo, or Morquio syndromes), or a genetic disorder affecting skeletal tissue who receive allo-HCT, the evidence includes mostly case series, case reports, and registry data. Relevant outcomes are overall survival, disease-specific survival, symptoms, quality of life, and treatment-related morbidity. The evidence has shown that, for most of these disorders, there is a demonstrable improvement in overall survival and other disease-specific outcomes. Allo-HCT is likely to improve health outcomes in select patients with certain inherited and acquired diseases. The evidence is sufficient to determine that the technology results in a meaningful improvement in the net health outcome.

For individuals who have an inherited metabolic syndrome disease (specifically those including Hunter, Sanfilippo, and Morquio syndromes) who receive allo-HCT, the evidence includes case reports. Relevant outcomes are overall survival, disease-specific survival, symptoms, quality of life, and treatment-related morbidity. Use of allo-HCT to treat patients with Hunter, Sanfilippo, or Morquio syndromes does not result in improvements in neurologic, neuropsychologic, and neurophysiologic function. The evidence is insufficient to determine the effects of the technology on health outcomes.

Background     
Genetic Diseases and Acquired Anemias
Hemoglobinopathies
Thalassemias result from variants in the globin genes, resulting in reduced or absent hemoglobin production, thereby reducing oxygen delivery. The supportive treatment of b-thalassemia major requires life-long red blood cell transfusions that lead to progressive iron overload and the potential for organ damage and impaired cardiac, hepatic, and endocrine function. Sickle cell disease is caused by a single amino acid substitution in the beta chain of hemoglobin and, unlike thalassemia major, has a variable course of clinical severity.1 Sickle cell disease typically manifests clinically with anemia, severe painful crises, acute chest syndrome, stroke, chronic pulmonary and renal dysfunction, growth retardation, neurologic deficits, and premature death. The mean age of death for patients with sickle cell disease has been demonstrated as 42 years for men and 48 for women.

Treatment
The only definitive cure for thalassemia is to correct the genetic defect with allogeneic hematopoietic cell transplantation (allo-HCT).

Three major therapeutic options are available for sickle cell disease: chronic blood transfusions, hydroxyurea, and allo-HCT, the latter being the only possibility for cure.1

Bone Marrow Failure Syndromes
Aplastic anemia in children is rare; most often, it is idiopathic and, less commonly, due to a hereditary disorder. Inherited syndromes include Fanconi anemia, a rare, autosomal recessive disease characterized by genomic instability, with congenital abnormalities, chromosome breakage, cancer susceptibility, and progressive bone marrow failure leading to pancytopenia and severe aplastic anemia. Frequently, this disease terminates in myelodysplastic syndrome or acute myeloid leukemia. Most patients with Fanconi anemia succumb to the complications of severe aplastic anemia, leukemia, or solid tumors, with a median survival of 30 years of age.2

Dyskeratosis congenita is characterized by marked telomere dysregulation with clinical features of reticulated skin hyperpigmentation, nail dystrophy, and oral leukoplakia.3 Early mortality is associated with bone marrow failure, infections, pulmonary complications, or malignancy.

Variants affecting ribosome assembly and function are associated with Shwachman-Diamond syndrome and Diamond-Blackfan syndrome.3 Shwachman-Diamond has clinical features that include pancreatic exocrine insufficiency, skeletal abnormalities, and cytopenias, with some patients developing aplastic anemia. As with other bone marrow failure syndromes, patients are at increased risk of myelodysplastic syndrome and malignant transformation, especially acute myeloid leukemia. Diamond-Blackfan anemia is characterized by absent or decreased erythroid precursors in the bone marrow, with 30% of patients also having a variety of physical anomalies.3

Treatment
In Fanconi anemia, HCT is currently the only treatment that definitively restores normal hematopoiesis. Excellent results have been observed with the use of human leukocyte antigen (HLA)-matched sibling allo-HCT, with cure of the marrow failure and amelioration of the risk of leukemia.

Primary Immunodeficiencies
The primary immunodeficiencies are a genetically heterogeneous group of diseases that affect distinct components of the immune system. More than 120 gene defects have been described, causing more than 150 disease phenotypes.4 The most severe defects (collectively known as severe combined immunodeficiency) cause an absence or dysfunction of T lymphocytes and sometimes B lymphocytes and natural killer cells.4

Treatment
Without treatment, patients with severe combined immunodeficiency usually die by 12 to 18 months of age. With supportive care, including prophylactic medication, the lifespan of these patients can be prolonged, but long-term outlook is still poor, with many dying from infectious or inflammatory complications or malignancy by early adulthood.4 Bone marrow transplantation is the only definitive cure, and the treatment of choice for severe combined immunodeficiency and other primary immunodeficiencies, including Wiskott-Aldrich syndrome and congenital defects of neutrophil function.5

Inherited Metabolic Diseases
Lysosomal storage disorders consist of many different rare diseases caused by a single gene defect, and most are inherited as an autosomal recessive trait.6 Lysosomal storage disorders are caused by specific enzyme deficiencies that result in defective lysosomal acid hydrolysis of endogenous macromolecules that subsequently accumulate as a toxic substance. Peroxisomal storage disorders arise due to a defect in a membrane transporter protein that leads to defects in the metabolism of long-chain fatty acids. Lysosomal storage disorders and peroxisomal storage disorders affect multiple organ systems, including the central and peripheral nervous systems. These disorders are progressive and often fatal in childhood due to both the accumulation of toxic substrate and a deficiency of the product of the enzyme reaction.6 Hurler syndrome usually leads to premature death by 5 years of age.

Treatment
Exogenous enzyme replacement therapy is available for a limited number of the inherited metabolic diseases; however, these drugs do not cross the blood-brain barrier, which results in the ineffective treatment of the central nervous system. Stem cell transplantation provides a constant source of enzyme replacement from the engrafted donor cells, which are not impeded by the blood-brain barrier.6 The donor-derived cells can migrate and engraft in many organ systems, giving rise to different types of cells (e.g., microglial cells in the brain and Kupffer cells in the liver).6

Allo-HCT has been primarily used to treat the inherited metabolic diseases that belong to the lysosomal and peroxisomal storage disorders, as listed in Table 1.6 The first stem cell transplant for an inherited metabolic disease was performed in 1980 in a patient with Hurler syndrome. Since that time, more than 1,000 transplants have been performed worldwide.6

Table 1. Lysosomal and Peroxisomal Storage Disorders

Category

Diagnosis

Other Names

Mucopolysaccharidosis

Mucopolysaccharidosis I H or H/S

Mucopolysaccharidosis II

Mucopolysaccharidosis III A-D

Mucopolysaccharidosis IV A-B

Mucopolysaccharidosis VI

Mucopolysaccharidosis VII

Hurler syndrome or Hurler-Scheie syndrome

Hunter syndrome

Sanfilippo syndrome A-D

Morquio syndrome A-B

Maroteaux-Lamy syndrome

Sly syndrome

Sphingolipidosis

Fabry disease

Farber disease

Gaucher disease types 1 and 3

GM1 gangliosidosis

Niemann-Pick disease A and B

Tay-Sachs disease

Sandhoff disease

Globoid cell leukodystrophy

Metachromatic leukodystrophy

 

Lipogranulomatosis

 

 

 

 

 

Krabbe disease

MLD

Glycoproteinosis

Aspartylglucosaminuria

Fucosidosis

Alpha-mannosidosis

Beta-mannosidosis

Mucolipidosis III and IV

 

 

 

 

Sialidosis

Other lipidoses

Niemann-Pick disease C

Wolman disease

Ceroid lipofuscinosis type III

 

 

Batten disease

Glycogen storage

Glycogen storage disease type II

Pompe disease

Multiple enzyme deficiency

Galactosialidosis

Mucolipidosis type II

 

I-cell disease

Lysosomal transport defects

Cystinosis

Sialic acid storage disease

Salla disease

 

Peroxisomal storage disorders

Adrenoleukodystrophy

Adrenomyeloneuropathy

ALD

AMN

Genetic Disorders Affecting Skeletal Tissue
Osteopetrosis is a condition caused by defects in osteoclast development and/or function. The osteoclast (the cell that functions in the breakdown and resorption of bone tissue) is known to be part of the hematopoietic family and shares a common progenitor with the macrophage in the bone marrow.7 Osteopetrosis is a heterogeneous group of heritable disorders, resulting in several different types of variable severity. The most severely affected patients are those with infantile malignant osteopetrosis (Albers-Schonberg disease or marble bone disease). Patients with infantile malignant osteopetrosis suffer from dense bone, including a heavy head with frontal bossing, exophthalmos, blindness by approximately 6 months of age, and severe hematologic malfunction with bone marrow failure.7 Seventy percent of these patients die before the age of 6 years, often of recurrent infections.7

Treatment
HCT is the only curative therapy for this fatal disease.

Hematopoietic Cell Transplantation
HCT is a procedure in which hematopoietic stem cells are intravenously infused to restore bone marrow and immune function in cancer patients who receive bone marrow-toxic doses of cytotoxic drugs with or without whole-body radiotherapy. Hematopoietic stem cells may be obtained from the transplant recipient (autologous HCT) or a donor (allo-HCT). They can be harvested from bone marrow, peripheral blood, or umbilical cord blood shortly after delivery of neonates. Cord blood transplantation is discussed in detail in evidence review 7.01.50.

Immunologic compatibility between infused hematopoietic stem cells and the recipient is not an issue in autologous HCT. In allogeneic stem cell transplantation, immunologic compatibility between donor and patient is a critical factor for achieving a successful outcome. Compatibility is established by typing ofHLA using cellular, serologic, or molecular techniques. HLA refers to the gene complex expressed at the HLA-A, -B, and -DR (antigen-D related) loci on each arm of chromosome 6. An acceptable donor will match the patient at all or most of the HLA loci.

Conditioning for Hematopoietic Cell Transplantation
Conventional Conditioning
The conventional (“classical”) practice of allo-HCT involves administration of cytotoxic agents (e.g., cyclophosphamide, busulfan) with or without total body irradiation at doses sufficient to cause bone marrow ablation in the recipient. The beneficial treatment effect of this procedure is due to a combination of the initial eradication of malignant cells and subsequent graft-versus-malignancy effect mediated by non-self-immunologic effector cells. While the slower graft-versus-malignancy effect is considered the potentially curative component, it may be overwhelmed by existing disease in the absence of pretransplant conditioning. Intense conditioning regimens are limited to patients who are sufficiently medically fit to tolerate substantial adverse effects. These include opportunistic infections secondary to loss of endogenous bone marrow function and organ damage or failure caused by cytotoxic drugs. Subsequent to graft infusion in allo-HCT, immunosuppressant drugs are required to minimize graft rejection and graft-versus-host disease, which increases susceptibility to opportunistic infections.

The success of autologous HCT is predicated on the potential of cytotoxic chemotherapy, with or without radiotherapy, to eradicate cancerous cells from the blood and bone marrow. This permits subsequent engraftment and repopulation of the bone marrow with presumably normal hematopoietic stem cells obtained from the patient before undergoing bone marrow ablation. Therefore, autologous HCT is typically performed as consolidation therapy when the patient’s disease is in complete remission. Patients who undergo autologous HCT are also susceptible to chemotherapy-related toxicities and opportunistic infections before engraftment, but not GVH disease.

Reduced-Intensity Conditioning Allogeneic Hematopoietic Cell Transplantation
RIC refers to the pretransplant use of lower doses of cytotoxic drugs or less intense regimens of radiotherapy than are used in traditional full-dose myeloablative conditioning treatments. Although the definition of RIC is variable, with numerous versions employed, all regimens seek to balance the competing effects of relapse due to residual disease and non-relapse mortality. The goal of RIC is to reduce disease burden and to minimize associated treatment-related morbidity and non-relapse mortality in the period during which the beneficial graft-versus-malignancy effect of allogeneic transplantation develops. RIC regimens range from nearly total myeloablative to minimally myeloablative with lymphoablation, with intensity tailored to specific diseases and patient condition. Patients who undergo RIC with allo-HCT initially demonstrate donor cell engraftment and bone marrow mixed chimerism. Most will subsequently convert to full-donor chimerism. In this review, the term reduced-intensity conditioning will refer to all conditioning regimens intended to be non-myeloablative.

Regulatory Status
The U.S. Food and Drug Administration regulates human cells and tissues intended for implantation, transplantation or infusion through the Center for Biologics Evaluation and Research, under the Code of Federal Regulation title 21, parts 1270 and 1271.8 Hematopoietic stem cells are included in these regulations. 

Policy:
Allogeneic hematopoietic stem cell transplantation is considered MEDICALLY NECESSARY for selected patients with the following disorders:

Hemoglobinopathies

  • Sickle cell anemia for children or young adults with either a history of prior stroke or at increased risk of stroke or end-organ damage
  • Homozygous beta-thalassemia (i.e., thalassemia major)

Bone Marrow Failure Syndromes

  • Aplastic anemia including hereditary (including Fanconi anemia, dyskeratosis congenita, Shwachman-Diamond, Diamond-Blackfan) or acquired (e.g., secondary to drug or toxin exposure) forms

Primary Immunodeficiencies

  • Absent or defective T-cell function (e.g., severe combined immunodeficiency, Wiskott-Aldrich syndrome, X-linked lymphoproliferative syndrome)
  • Absent or defective natural killer function (e.g., Chediak-Higashi syndrome)
  • Absent or defective neutrophil function (e.g., Kostmann syndrome, chronic granulomatous disease, leukocyte adhesion defect)

(See Policy Guideline # 1.)

Inherited Metabolic Disease

  • Lysosomal and peroxisomal storage disorders except Hunter, Sanfilippo and Morquio syndromes

(See Policy Guideline # 2.)

Genetic Disorders Affecting Skeletal Tissue

  • Infantile malignant osteopetrosis (Albers-Schonberg disease or marble bone disease)

Policy Guidelines
1. The following lists the immunodeficiencies that have been successfully treated by allogeneic hematopoietic stem-cell transplantation (HSCT) (4):

Lymphocyte Immunodeficiencies

  • Adenosine deaminase deficiency
  • Artemis deficiency
  • Calcium channel deficiency
  • CD 40 ligand deficiency
  • Cernunnos/X-linked lymphoproliferative disease deficiency
  • CHARGE syndrome with immune deficiency
  • Common gamma chain deficiency
  • Deficiencies in CD 45, CD3, CD8
  • DiGeorge syndrome
  • DNA ligase IV
  • Interleukin-7 receptor alpha deficiency
  • Janus-associated kinase three (JAK3) deficiency
  • Major histocompatibility class II deficiency
  • Omenn syndrome
  • Purine nucleoside phosphorylase deficiency
  • Recombinase-activating gene (RAG) 1/2 deficiency
  • Reticular dysgenesis
  • Winged helix deficiency
  • Wiskott-Aldrich syndrome
  • X-linked lymphoproliferative disease
  • Zeta-chain-associated protein-70 (ZAP-70) deficiency

Phagocytic Deficiencies

  • Chediak-Higashi syndrome
  • Chronic granulomatous disease
  • Hemophagocytic lymphohistiocytosis
  • Griscelli syndrome, type 2
  • Interferon-gamma receptor deficiencies
  • Leukocyte adhesion deficiency
  • Severe congenital neutropenias
  • Shwachman-Diamond syndrome

Other Immunodeficiencies

  • Autoimmune lymphoproliferative syndrome
  • Cartilage hair hypoplasia
  • CD25 deficiency
  • Hyper IgD and IgE syndromes
  • ICF syndrome
  • IPEX syndrome
  • NEMO deficiency
  • NF-κB inhibitor, alpha (IκB-alpha) deficiency
  • Nijmegen breakage syndrome

2. In the inherited metabolic disorders, allogeneic HSCT has been proven effective in some cases of Hurler, Maroteaux-Lamy and Sly syndromes, childhood onset cerebral X-linked adrenoleukodystrophy, globoid-cell leukodystrophy, metachromatic leukodystrophy, alpha-mannosidosis and aspartylglucosaminuria. Allogeneic HSCT is possibly effective for fucosidosis, Gaucher types one and 3, Farber lipogranulomatosis, galactosialidosis, GM1, gangliosidosis, mucolipidosis II (I-cell disease), multiple sulfatase deficiency, Niemann-Pick, neuronal ceroid lipofuscinosis, sialidosis and Wolman disease. Allogeneic HSCT has not been effective in Hunter, Sanfilippo or Morquio syndromes. (8)

The experience with reduced-intensity conditioning (RIC) and allogeneic HSCT for the diseases listed in this policy has been limited to small numbers of patients and has yielded mixed results, depending upon the disease category. In general, the results have been most promising in the bone marrow failure syndromes and primary immunodeficiencies. In the hemoglobinopathies, success has been hampered by difficulties with high rates of graft rejection, and in adult patients, severe graft versus host disease (GVHD). Several Phase II/III trials are ongoing examining the role of this type of transplant for these diseases, as outlined in the clinical trial section under each disease type.

Benefit Application
BlueCard®/National Account Issues
State or federal mandates (e.g., FEP) may dictate that certain U.S. Food and Drug Administration-approved devices, drugs or biologics may not be considered investigational, and, thus, these devices may be assessed only on the basis of their medical necessity.

Rationale  
Evidence reviews assess the clinical evidence to determine whether the use of technology improves the net health outcome. Broadly defined, health outcomes are the length of life, quality of life (QOL) and ability to function — including benefits and harms. Every clinical condition has specific outcomes that are important to patients and managing the courseof that condition. Validated outcome measures are necessary to ascertain whether a condition improves or worsens; and whether the magnitude of that change is clinically significant. The net health outcome is a balance of benefits and harms.

To assess whether the evidence is sufficient to draw conclusions about the net health outcome of technology, 2 domains are examined: the relevance, and quality and credibility. To be relevant, studies must represent one or more intended clinical uses of the technology in the intended population and compare an effective and appropriate alternative at a comparable intensity. For some conditions, the alternative will be supportive care or surveillance. The quality and credibility of the evidence depend on study design and conduct, minimizing bias and confounding that can generate incorrect findings. The randomized controlled trial (RCT) is preferred to assess efficacy; however, in some circumstances, nonrandomized studies may be adequate. RCTs are rarely large enough or long enough to capture less common adverse events and long-term effects. Other types of studies can be used for these purposes and to assess generalizability to broader clinical populations and settings of clinical practice.

Clinical Context and Test Purpose
The purpose of allogeneic hematopoietic cell transplantation (allo-HCT) is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with a hemoglobinopathy.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with a hemoglobinopathy?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with a hemoglobinopathy.

Interventions
The therapy being considered is allo-HCT.

Patients with a hemoglobinopathy are actively managed by hematologists and primary care providers in an outpatient clinical setting. Patients with a hemoglobinopathy undergoing HCT are actively managed in the inpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
The general outcomes of interest are overall survival (OS), disease-specific survival (DSS), symptoms, QOL, and treatment-related morbidity (TRM).

Follow-up at 3- and 12- years is of interest for allo-HCT to monitor relevant outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles: 

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Hemoglobinopathies
Systematic Reviews
Review articles summarize the experience to date with HCT and the hemoglobinopathies.8,9,10,11

β-Thalassemia
Systematic Reviews
More than 3,000 patients worldwide have been treated for β-thalassemia with allo-HCT.10 The OS rates have ranged from 65% to 98% at 5 years, up to 87% at 15 years, up to 89% at 20 years, and thalassemia-free survival has been reported to be as high as 86% at 6 years.12 The Pesaro risk stratification system classifies patients with thalassemia who plan to undergo allo-HCT into risk groups 1 through 3 based on the presence of hepatomegaly, portal fibrosis, or adequacy of chelation (class 1 having no risk factors, class 2 with 2 risk factors, and class 3 with all 3 risk factors).13 The outcome of allo-HCT in more than 800 patients with thalassemia according to risk stratification has shown OS and event-free survival (EFS) rates of 95% and 90% for Pesaro class 1, 87% and 84% for class 2, and 79% and 58% for class 3.

Noncomparative Studies
A 2015 study of 489 patients with nonmalignant hematologic disorders who underwent allo-HCT between 1997 and 2012 included 152 patients with β-thalassemia.14 Mean age at transplantation was 5.7 years (range, 1.1 – 23 years). At the time of transplantation, 26 (17%) patients had Pesaro class 1, 103 (68%) had class 2, and 23 (15%) had class 3; 132 patients received peripheral blood stem cells and 20 received bone marrow grafts. Mean times to neutrophil and platelet engraftment were 21.4 days (range, 8 – 69 days) and 32.8 days (range, 7 – 134 days), respectively. The incidence of graft rejection was significantly lower in patients who received peripheral blood stem cells than in those who received bone marrow grafts (9% vs. 25%; p = 0.036). Acute graft-versus-host-disease (GVHD) grade II, III, and IV occurred in 15% of β-thalassemia patients, and chronic GVHD occurred in 12%. The incidence of transplant-related mortality for this group was 18%. After a median follow-up period of 12 years, the OS rate for these patients was 82.4%. The disease-free survival (DFS) rate for the whole group of β-thalassemia patients was 72.4% (74% in the peripheral blood cell transplantation group vs. 64% in the bone marrow cell transplantation group; p = 0.381), which might be attributed to the higher incidence of graft rejection in bone marrow groups.

Bernardo et al. (2012) reported on the results of 60 thalassemia patients (median age, 7 years; range, 1 – 37 years) who underwent allo-HCT after a reduced-intensity conditioning (RIC) regimen based on treosulfan.15 Before the transplant, 27 children were assigned to class 1 of the Pesaro risk stratification system, 17 to class 2, and 4 to class 3; 12 patients were adults. Twenty patients were transplanted from a human leukocyte antigen (HLA)-identical sibling and 40 from an unrelated donor. The cumulative incidence of graft failure and transplantation-related mortality were 9% and 7%, respectively. Eight patients experienced grade II, III, or IV acute GVHD, the cumulative incidence being 14%. Among 56 patients at risk, 1 developed limited chronic GVHD. With a median follow-up of 36 months (range, 4 – 72 months), the 5-year probability of survival and thalassemia-free survival were 93% and 84%, respectively. Neither the class of risk nor the donor used influenced outcomes.

In a 2014 report on RIC HCT, 98 patients with class 3 thalassemia were transplanted with related or unrelated donor stemcells.16 Seventy-six of patients less than 10 years of age received a conventional myeloablative conditioning regimen (cyclophosphamide, busulfan, with or without fludarabine). The remaining 22 patients were 10 years of age or older with hepatomegaly and, in several instances, additional comorbidity problems, who underwent HCT with a novel RIC regimen (fludarabine and busulfan). Rates of EFS (86% vs. 90%, respectively), and OS (95% vs. 90%, respectively) did not differ significantly between groups. However, a higher incidence of serious treatment-related complications was observed in the group that received myeloablative conditioning. Furthermore, graft failures occurred in 6 patients in the myeloablatedgroup (8%), although none occurred in the RIC group.

Sickle Cell Disease
Systematic Reviews
A Cochrane systematic review published in 2013,17 and updated in 201618, and 202019, identified no completed RCTs that assessed risk or benefit of any method of HCT in patients with sickle cell disease.

Noncomparative Studies
Approximately 500 to 600 patients with sickle cell disease have undergone allo-HCT, and most of the experience with allo-HCT and sickle cell disease comes from 3 major clinical series.1,10 The largest series to date consists of 87 symptomatic patients, most of whom received donor allografts from siblings who are HLA-identical.20, The results from that series and the 2 others21,22 were similar, with rates of OS ranging from 92% to 94% and EFS from 82% to 86%, with a median follow-up ranging from 0.9 to 17.9 years.1

Experience with reduced-intensity preparative regimens (RIC and allo-HCT for the hemoglobinopathies) is limited to asmall number of patients. Challenges have included high rates of graft rejection (10% – 30%)8 and, in adult patients, severe GVHD, which has been observed with the use of RIC regimens.9

Hsieh et al. (2014) reported on results from 30 patients aged 16 to 65 years with severe sickle cell phenotype who were enrolled in a RIC allo-HCT study, consisting of alemtuzumab (1 mg/kg in divided doses), total body irradiation (300centigray), sirolimus, and infusion of unmanipulated filgrastim-mobilized peripheral blood stem cells from HLA-matchedsiblings.23 The primary endpoint was treatment success at 1 year after the transplant, defined as full-donor-typehemoglobin for patients with sickle cell disease and transfusion independence for patients with thalassemia. Secondary endpoints included the level of donor leukocyte chimerism; incidence of acute and chronic GVHD; and sickle cell-thalassemia DFS, immunologic recovery, and changes in organ function. Twenty-nine patients survived a median of 3.4years (range, 1 – 8.6 years), with no nonrelapse mortality. One patient died from intracranial bleeding after relapse. Normalized hemoglobin and resolution of hemolysis among engrafted patients were accompanied by stabilization in brain imaging, a reduction of echocardiographic estimates of pulmonary pressure, and allowed for phlebotomy to reduce hepatic iron. A total of 38 serious adverse events were reported: pain and related management, infections, abdominal events, and sirolimus-related toxic effects.

Section Summary: Hemoglobinopathies
The use of allo-HCT to treat patients with β-thalassemia or sickle cell disease has been shown to improve OS, EFS, or DFS.

Clinical Context and Test Purpose
The purpose of allo-HCT is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with a bone marrow failure syndrome.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with a bone marrow failure syndrome?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with bone marrow failure syndrome.

Interventions
The therapy being considered is allo-HCT.

Patients with bone marrow failure syndrome are actively managed by hematologists and primary care providers in an outpatient clinical setting. Patients with bone marrow failure syndrome undergoing HCT are actively managed in the inpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
The general outcomes of interest are OS, DSS, symptoms, QOL, and TRM.

Follow-up at 1.1- and 8-years is of interest for allo-HCT to monitor relevant outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles: 

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Bone Marrow Failure Syndromes
Systematic Reviews
Review articles summarize the experience to date on the use of HCT to treat bone marrow failure syndromes.8,24,25,26

Fanconi Anemia
Noncomparative Studies
Yabe et al. (2020) analyzed nationwide records of 163 patients with Fanconi anemia who received an allo-HCT in Japan between 1987 and 2015.27 The 5-year OS rate was 81%. The 10-year and 15-year OS rates were 77% and 72%. Of the 163 patients, 154 had a stable engraftment. Among evaluable patients (n = 154), 17% and 5.8% developed grade II-IV andgrade III – IV GVHD, respectively.

In a 2008 summary of patients with Fanconi anemia who received allo-HCT from matched related donors over 6 years (N = 103 patients), OS rates ranged from 83% to 88%, with transplant-related mortality ranging from 8% to 18.5% and average chronic GVHD of 12%.28

The outcomes in patients with Fanconi anemia and an unrelated donor allo-HCT have not been as promising. The European Group for Blood and Marrow Transplantation has analyzed the outcomes using alternative donors in 67 patients with Fanconi anemia. Median 2-year survival was 28%.3 Causes of death included infection, hemorrhage, acute and chronic GVHD, and liver veno-occlusive disease.3 The Center for International Blood and Marrow Transplant Research analyzed 98 patients transplanted with unrelated donor marrow between 1990 and 2003. Three-year OS rates were 13% and 52%, respectively, in patients who received non-fludarabine- or fludarabine-based regimens.3

Zanis-Neto et al. (2005) reported on the results of 30 patients with Fanconi anemia treated with RIC regimens, consisting of low-dose cyclophosphamide.29 Seven patients were treated with cyclophosphamide at 80 mg/kg and 23 with 60mg/kg. Grade II or III acute GVHD rates were 57% and 14% for patients who received the higher and lower doses, respectively (p = 0.001). Four of the 7 patients who received the higher dose were alive at a median of 47 months (range,44 – 58 months), and 22 of 23 given the lower dose were alive at a median of 16 months (range, 3 – 52 months). The authors concluded that a lower dose of cyclophosphamide conditioning resulted in lower rates of GVHD and was acceptable for engraftment.

In a retrospective study of 98 unrelated donor transplantations for Fanconi anemia reported to the Center for International Blood and Marrow Transplant Research, Wagner et al. (2007) reported that fludarabine-based (reduced-intensity) regimens were associated with improved engraftment, decreased treatment-related mortality, and improved 3 – year OS rates (52% vs. 13%, respectively; p < 0.001) compared with non-fludarabine-based regimens.30

Dyskeratosis Congenita
Noncomparative Studies
Results with allo-HCT in dyskeratosis congenita have been disappointing because of severe late effects, including diffuse vasculitis and lung fibrosis.3 Currently, nonmyeloablative conditioning regimens with fludarabine are being explored;however, very few results have been published.3

Outcomes after allo-HCT were reported in 2013 for 34 patients with dyskeratosis congenita who underwent transplantation between 1981 and 2009.31 Median age at transplantation was 13 years (range, 2 – 35 years).Approximately 50% of transplantations were from related donors. The day-28 probability of neutrophil recovery was 73%, and the day-100 platelet recovery was 72%. The day-100 probability of grade II, III, or IV acute GVHD and the 3-year probability of chronic GVHD were 24% and 37%, respectively. The 10-year probability of survival was 30% and 14 patients were still alive at last follow-up. Ten deaths occurred within 4 months from transplantation because of graft failure (n = 6) or other transplantation-related complications; 9 of these patients had undergone transplantation from mismatched related or unrelated donors. Another 10 deaths occurred after 4 months; 6 of which occurred more than 5 years after transplantation, and 4 deaths were attributed to pulmonary failure. Transplantation regimen intensity and transplantations from mismatched related or unrelated donors were associated with early mortality. Transplantation of grafts from HLA-matched siblings with cyclophosphamide-containing nonradiation regimens was associated with early low toxicity. Late mortality was attributed mainly to pulmonary complications and likely related to the underlying disease.

Shwachman-Diamond Syndrome
Noncomparative Studies
Experience with allo-HCT in Shwachman-Diamond syndrome is limited because very few patients have undergone allogeneic transplants for this disease.3 Cesaro et al. (2005) reported on 26 patients with Shwachman-Diamond syndrome from the European Group for Blood and Marrow Transplantation registry, who received HCT for treatment of severe aplastic anemia (n = 16); myelodysplastic syndrome-acute myeloid leukemia (n = 9); or another diagnosis (n = 1).32,Various preparative regimens were used; most included busulfan (54%) or total body irradiation (23%) followed by an HLA-matched sibling (n = 6), mismatched related (n = 1), or unrelated graft (n = 19). Graft failure occurred in 5 (19%)patients, and the incidence of grade III to IV acute and chronic GVHD were 24% and 29%, respectively. With a median follow-up of 1.1 years, the OS rate was 65%. Deaths were primarily caused by infections with or without GVHD (n = 5) or major organ toxicities (n = 3). The analysis suggested that the presence of myelodysplastic syndrome-acute myeloidleukemia or use of total body irradiation-based conditioning regimens were factors associated with a poorer outcome.

Myers et al. (2020) evaluated outcomes in 52 patients with Shwachman-Diamond syndrome who received a HCT between 2000 and 2017 for bone marrow failure (n = 39) or myelodysplastic syndrome-acute myeloid leukemia (n = 13).33 For patients with bone marrow failure, preparative regimens were myeloablative in 13 patients and reduced intensity in 26 patients. The 5-year OS in this subgroup was 72%. For patients with myelodysplastic syndrome-acute myeloid leukemia,preparative regimens were myeloablative in 8 patients and reduced intensity in 5. At the time of the study report, only 2 of 13 (15%) were alive. Most deaths in this subgroup (8/11) occurred within 8 months after transplantation.

Cesaro et al. (2020) reported on outcomes in 74 patients with Shwachman-Diamond syndrome who received a HCT between 1988 and 2016 for bone marrow failure (n = 61), myelodysplastic syndrome (n = 7), or acute myeloid leukemia (n = 6) in an updated report from the European Group for Blood and Marrow Transplantation registry.34 The preparative regimens were myeloablative and reduced intensity in 54% and 46% of the cases, respectively. After a median follow-up of 7.3 years, the 5-year OS was 63.3%. The 5-year OS was significantly higher in patients who received HCT due to bone marrow failure (70.7%) versus in patients who received HCT due to myelodysplastic syndrome/acute myeloid leukemia (28.8%; p = 0.005).The rate of graft failure was 15% and grades I-IV GVHD were reported in 55% of patients.

Diamond-Blackfan Syndrome
Noncomparative Studies
In Diamond-Blackfan syndrome, allo-HCT is an option in corticosteroid-resistant disease.3 In a report from the Diamond­ Blackfan Anemia Registry (2008), 20 of 354 registered patients underwent allo-HCT, and the 5-year survival rate was 87.5% when recipients received HLA-identical sibling grafts but was poor in recipients of alternative donors.3 Another team of investigators (2005) examined outcomes reported to the International Bone Marrow Transplant Registry between 1984 and 2000 for 61 patients with Diamond-Blackfan syndrome who underwent HCT.3,5 Sixty-seven percent of patients were transplanted with an HLA-identical sibling donor. Probability of OS after transplantation for patients transplanted from an HLA-identical sibling donor (vs. an alternative donor) was 78% versus 45% (p = 0.01) at 1 year and 76% versus 39% (p = 0.01) at 3 years, respectively.

Aplastic Anemia
Systematic Reviews
EIGohary et al. 2020 performed a systematic review and meta-analysis on 15 prospective or retrospective studies (N = 577) evaluating outcomes after HCT in patients with idiopathic aplastic anemia.36 Seven studies were retrospective in nature, deriving data from registries and records, while 8 studies had a prospective design. Across the studies, the pooled incidence of successful engraftment was 97.3% (95% confidence interval [Cl], 95.9 to 98.7). Grade II-IV GVHD was reported in 26.6% of patients. The pooled incidence of transplant-related mortality was 6.7% per year (95% Cl, 4.0 to 9.4). Patients who received reduced intensity conditioning regimens had lower rates of GVHD (18.7% vs. 29.5%) and engraftment (91.7% vs. 97.7%) compared to nonmyeloablative regimens.

Randomized Controlled Trials
A randomized phase 3 trial (2012) compared 2 conditioning regimens in patients (n = 79) with high-risk aplastic anemia who underwent allo-HCT.37 Patients in the cyclophosphamide plus anti-thymocyte globulin (ATG) arm (n = 39) received cyclophosphamide at 200 mg/kg; those in the cyclophosphamide-fludarabine-ATG arm (n = 40) received cyclophosphamide at 100 mg/kg and fludarabine at 150 mg/m2. No difference in engraftment rates was reported between arms. Infections with an identified causative organism and sinusoidal obstruction syndrome, hematuria, febrile episodes, and death from any cause tended to be more frequent among those receiving cyclophosphamide-ATG but did not differ significantly between treatment arms. For example, at 4 years, OS rates did not differ significantly between the cyclophosphamide-ATG (78%) and the cyclophosphamide-fludarabine-ATG arms (86%; p = 0.41). Although this study was underpowered to detect real differences between the conditioning regimens, the results suggested that a RIC regimen with cyclophosphamide-fludarabine-ATG appears to be as safe as a more conventional myeloablative regimen using cyclophosphamide plus ATG in allo-HCT.

Noncomparative Studies
A 2015 study analyzed outcomes reported to the European Group for Blood and Marrow Transplantation of children with idiopathic aplastic anemia, according to treatment received.38 Front-line immunosuppressive therapy (1ST) was compared with front-line HCT from an HLA-matched family donor, to evaluate the outcomes of patients who, after having failed 1ST, underwent rescue HCT, and to compare their outcomes using front-line HCT with those who did not fail 1ST (1ST with no subsequent transplant). Additional outcomes that were evaluated were the cumulative incidence of posttherapy tumors and prognostic factors that might affect the outcome of the disease. Included in the analysis were records from 563 consecutive children (313 boys, 250 girls [age range, 0 – 12 years]) diagnosed between 2000 and 2009. Geographical origin, if known, was distributed as follows: 383 patients from Europe, 51 from Africa, 51 from the Middle East, 2 from Australia, and 1 from Brazil. The median age at diagnosis was 7.8 years (range, 0.01 – 11.9 years). A total of 167 children received front-line 1ST (consisting of ATG plus cyclosporine); of these, 91 (55%) failed 1ST as front-line treatment and underwent rescue HCT (HCT post-lST failure) whereas 1ST was the only treatment received (1ST alone) for 76 patients. The 3-year probability of OS and EFS for the whole population was 90% and 86%, respectively. The 3-year OS rate was 91% after matched family donor front-line HCT and 87% after first-line 1ST (p = 0.18). The 3-year probability of OS after HCT post-IST failure was 83%, 91% after matched family donor front-line HCT, and 97% after 1ST alone (p = 0.017). A subgroup analysis showed no significant difference between 1ST alone and matched family donor front-line HCT (p = 0.21), but significantly longer OS of both matched family donor front-line HCT (p = 0.02) and 1ST alone (p = 0.047) over HCT post-lST failure.

A 2015 study (discussed earlier), which examined 489 patients with nonmalignant hematologic disorders who underwent allo-HCT, including 273 patients with severe aplastic anemia.14 Of these subjects, 212 were men, and 61 were women, and the mean age at transplantation was 19.7 years (range, 1.5 – 51 years). Mean times to neutrophil and platelet engraftment were 13.9 days (range, 10 – 26 days) and 14.1 days (range, 8 – 83 days), respectively. Graft rejection occurred in 1% of patients. Acute GVHD grade 11, 111, or IV occurred in 15%, and chronic GVHD occurred in 28% of patients. The incidence of transplant-related mortality was 22%. At 8 years, OS and DFS rates were both 74%. Conditioning regimens differed among the patients, with 181 receiving fludarabine and cyclophosphamide and 92 receiving cyclophosphamide and ATG. No statistically significant differences between conditioning groups were observed regarding mean time to neutrophil engraftment (p = 0.136) or incidence of extensive chronic GVHD (p = 0.651). Mean time to platelet engraftment was significantly longer in the cyclophosphamide plus ATG group (p = 0.016). The incidence of transplant-related mortality in the fludarabine plus cyclophosphamide group was 17%, which was significantly lower than in the cyclophosphamide plus ATG group (33%; p = 0.002). After a median follow-up of 8 years, the OS rate was statistically significantly better in the fludarabine plus cyclophosphamide group (80%) than in the cyclophosphamide plus ATG group of patients (64%; p = 0.021).

Section Summary: Bone Marrow Failure Syndromes
Use of allo-HCT to treat patients with Fanconi anemia, dyskeratosis congenital, Shwachman-Diamond syndrome, Diamond-Blackfan syndrome, and aplastic anemia have been shown to improve OS or DFS.

Primary Immunodeficiencies
Clinical Context and Test Purpose
The purpose of allo-HCT is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with a primary immunodeficiency.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with a primary immunodeficiency?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with a primary immunodeficiency.

Interventions
The therapy being considered is allo-HCT.

Patients with a primary immunodeficiency are actively managed by hematologists and primary care providers in an outpatient clinical setting. Patients with primary immunodeficiency syndrome undergoing HCT are actively managed in the inpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
general outcomes of interest are OS, DSS, symptoms, QOL, and TRM.

Follow-up at 6- and 21-months is of interest for allo-HCT to monitor relevant outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles: 

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCT. 
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review articles summarize experience the use of HCT to treat primary immunodeficiencies.39.40 Additional individual studies are reported next.

Review of Evidence
Chronic Granulomatous Disease
Noncomparative Studies
HCT outcomes were compared with those of conventional treatment in a study of 41 patients in Sweden who were diagnosed with chronic granulomatous disease (CGD) between 1990 and 2012.41 From 1997 to 2012, 14 patients (age range, 1 – 35 years) underwent HCT and received grafts either from an HLA-matched sibling donor or a matched unrelated donor. Thirteen (93%) of the 14 transplanted patients were reported alive and well in 2013. The mean age at transplantation was 10.4 years and the mean survival time was 7.7 years. In contrast, 7 of 13 men or boys with X-linked CGD who were treated conventionally died from complications of CGD at a mean age of 19 years, while the remainder suffered life-threatening infections.

A 2014 prospective study in 16 centers across 10 countries worldwide enrolled CGD patients ages O to 40 years to examine the effects of a RIC regimen before HCT, consisting of high-dose fludarabine, serotherapy, or low-dose alemtuzumab, and low-dose (50% to 72% of myeloablative dose) or targeted busulfan administration.42 Unmanipulated bone marrow or peripheral blood stem cells from HLA-matched related donors or HLA-9/10 or HLA-10/10 matched unrelated donors were infused. The primary endpoints were OS and EFS, probabilities of OS and EFS at 2 years, the incidence of acute and chronic GVHD, achievement of at least 90% myeloid donor chimerism, and incidence of graft failure after at least 6 months of follow-up. A total of 56 patients (median age 12.7 years) were included; 42 (75%) patients had high-risk features (i.e., intractable infections and auto-inflammation) and 25 (45%) were adolescents and young adults (age range, 14 – 39 years). Median time to engraftment was 19 days for neutrophils and 21 days for platelets. At a median follow-up of 21 months, the OS rate was 93%, and the EFS rate was 89%. The 2-year probability of OS was 96% (95% Cl , 86.46% to 99.09%) and of EFS was 91% (95% Cl, 79.78% to 96.17%). Graft failure occurred in 5% of patients. The cumulative incidence of acute GVHD grade Ill or IV was 4% and chronic GVHD was 7%. Stable ( 90%) myeloid donor chimerism was documented in 52 (93%) surviving patients.

Chiesa et al. (2020) conducted a retrospective, multicenter evaluation of 712 patients with CGD who received a HCT between 1993 and 2018.43 The majority of patients in the cohort were children (n = 635) and the median age at transplantation was 7 years (range, 0.1 to 48.6). Additionally, 87% of patients underwent transplantation after 2006. The 3-year OS probability was 85.7% (95% Cl, 82.8 to 88.5) and the 3-year EFS probability was 75.8% (95% Cl, 72.3 to 79.3). Donor engraftment was achieved in 88% of evaluable patients. The cumulative incidence of grades II-IV and grades Ill-IV acute GVHD was 20.1% and 9%, respectively. The cumulative incidence of chronic GVHD at 3 years was 17.8%.

Severe Combined Immunodeficiency
Noncomparative Studies
HCT using HLA-identical sibling donors can correct underlying primary immunodeficiencies, such as severe combined immunodeficiency (SCIO), Wiskott-Aldrich syndrome, and other prematurely lethal X-linked immunodeficiencies, in approximately 90% of cases.44 According to a 2008 European series of 475 patients collected between 1968 and 1999, survival rates for SCIO were approximately 80% with a matched sibling donor, 50% with a haploidentical donor, and 70% with a transplant from an unrelated donor.44 Another 2008 report found an OS rate for patients with SCIO who have undergone HCT to be 71%.4 

Hassan et al. (2012) reported on a multicenter retrospective study, which analyzed HCT outcomes in 106 patients with adenosine deaminase deficient-SCIO who received a total of 119 transplants.45 HCT using matched sibling and family donors had significantly better OS (86% and 81%, respectively) compared with HCT using matched unrelated (66%; p < 0.05) and haploidentical donors (43%; p < 0.001). Superior OS was also seen in patients who received unconditioned transplants compared with myeloablative procedures (81% vs. 54%; p < 0.003), although in unconditioned haploidentical donor HCT, nonengraftment was a major problem. Long-term immune recovery showed that, regardless of transplant type, overall T-cell counts were similar, although a faster rate of T-cell recovery was observed following matched sibling and family donor HCT. Humoral immunity and donor B-cell engraftment was achieved in nearly all evaluable surviving patients and was seen even after unconditioned HCT.

Wiskott-Aldrich Syndrome
Noncomparative Studies
For Wiskott-Aldrich syndrome, a 2001 analysis of 170 patients transplanted between 1968 and 1996 demonstrated the impact of donor type on outcomes.4,6 Fifty-five transplants were from HLA-identical sibling donors, with a 5-year survival probability of 87% (95% Cl, 74% to 93%); 48 were from other relatives, with a 5-year survival probability of 52% (95% Cl,37% to 65%); and 67 were from unrelated donors with a 5-year survival probability of 71% (95% Cl, 58% to 80%; p < 0.001).

Moratto et al. (2011) retrospectively reported on the long-term outcome and donor cell engraftment in 194 patients with Wiskott-Aldrich syndrome treated by HCT from 1980 to 2009.47 The OS rate was 84.0% and was even higher (89.1% 5- year survival) for those who had received HCT since the year 2000, reflecting the recent improvement in outcome after transplantation from mismatched family donors and for patients who received HCT from an unrelated donor older than 5 years of age. Also, patients who proceeded to transplantation in better clinical condition had a lower rate of post-HCT complications. Retrospective analysis of lineage-specific donor cell engraftment showed that stable full-donor chimerism was attained by 72.3% of the patients who survived for at least 1 year after HCT. Mixed chimerism was associated with an increased risk of incomplete reconstitution of lymphocyte counts and post-HCT autoimmunity, and myeloid donor cell chimerism less than 50% was associated with persistent thrombocytopenia.

Burroughs et al. (2020) reported outcomes in patients with Wiskott-Aldrich syndrome who received a HCT at a Primary Immune Deficiency Treatment Consortium center between 2005 through 2015.48 The most common conditioning regimens were a myeloablative busulfan-based regimen (n = 84) and a busulfan-based reduced intensity conditioning regimen (n = 25). After a median follow-up of 4.5 years, the 5-year OS probability was 91% (95% Cl, 85 to 95). Patients who received the HCT prior to age 5 years had superior 5-year OS outcomes compared to older patients (94% vs. 66%). The cumulative incidence of grades II – IV and grades Ill – IV acute GVHD were 27% and 15%, respectively.

X-Linked Lymphoproliferative Disease
Noncomparative Studies
X-linked lymphoproliferative disease (XLP1) is a rare, deadly immune deficiency caused by variants in the SH201A gene. Allo-HCT is often performed because of the morbidity and mortality associated with XLP1. There is limited experience using RIC regimens for these patients. One study (2014) reported on an 8-year single-center experience.49 Sixteen consecutive patients diagnosed with XLP1 underwent allo-HCT between 2006 and 2013 after a RIC regimen consisting of alemtuzumab, fludarabine, and melphalan. Fourteen of 16 patients received fully HLA-matched (8/8) unrelated or related bone marrow grafts, whereas 2 patients received mismatched unrelated grafts. All patients had a hematopoietic recovery. No cases of hepatic veno-occlusive disease or pulmonary hemorrhage were reported. One (6%) patient developed acute GVHD and later also developed chronic GVHD. Five (31%) patients developed mixed chimerism. One-year survival estimated by Kaplan-Meier analysis was 80%, with long-term survival estimated at 71%. There were no occurrences of lymphoma after HCT.

Other Immunodeficiencies
Noncomparative Studies
For patients with genetic immune or inflammatory disorders, such as hemophagocytic lymphohistiocytosis, the 5-year DFS rates with allo-HCT ranged from 60% to 70%.

For patients with other immunodeficiencies, reported OS rates are 74%, with even better results (90%) with well-matched donors for defined conditions, such as CGD.4

To date, studies have indicated that RIC regimens have an important role in treating patients with primary immunodeficiency.40 In the absence of prospective or larger registry studies, it is not possible to prove the superiority of RIC in more stable patients with primary immunodeficiency; however, RIC does offer the advantage that long-term sequelae (e.g., infertility, growth retardation) may be avoided or reduced. Currently, RIC HCT using unrelated donors may offer a survival advantage in patients with T-cell deficiencies, hemophagocytic lymphohistiocytosis, Wiskott-Aldrich syndrome (patients > 5 years of age), and CGD with ongoing inflammatory or infective complications. Minimal-intensity conditioning HCT may be particularly suited to unrelated donor HCT in young SCIO patients with significant comorbidities.

Section Summary: Primary Immunodeficienciesspan >
Use of allo-HCT to treat select patients with primary immunodeficiencies results in improvements in OS or DFS.

Inherited Metabolic Diseases Including Hunter, Sanfilippo, or Morquio Syndromes
Clinical Context and Test Purposes
The purpose of allo-HCT is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with inherited metabolic diseases including Hunter, Sanfilippo, or Morquio syndromes.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with an inherited metabolic disease?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with inherited metabolic diseases including Hunter, Sanfilippo, or Morquio syndromes.

Interventions
The therapy being considered is allo-HCT.

Patients with inherited metabolic diseases including Hunter, Sanfilippo, or Morquio syndromes are actively managed by metabolic specialists and primary care providers in an outpatient clinical setting. Patients with Hunter, Sanfilippo, or Morquio syndromes undergoing HCT are actively managed in the inpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
The general outcomes of interest are OS, DSS, symptoms, QOL, and TRM.

Follow-up at 7- to 17-years is of interest for allo-HCT to monitor relevant outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Hunter Syndrome
Noncomparative
Hunter syndrome is composed of 2 distinct clinical entities, a severe and an attenuated form. The attenuated form is characterized by a prolonged lifespan, minimal to no central nervous system involvement, and a slow progressions.50 Experience with allo-HCT in patients with severe Hunter syndrome has shown that it has failed to alter the disease course favorably or significantlys.50 Some have suggested that HCT would not be justifiable in the attenuated form because the risks outweigh the possible benefitss.50

Eight patients with Hunter syndrome received an allo-HCT between the ages of 3 and 16 years.51 In 6 cases, the donor as an HLA-identical sibling; in one case, an HLA-compatible unrelated donor was used, and in another, a mismatched unrelated donor was used. The severity of disease before the transplant was rated by assessing the age at diagnosis, behavior, and IQs at the time of graft and genotype. Five patients were considered to have severe central nervous system involvement (i.e., diagnosis before the age of 4 years and an IQ < 80), 2 were considered to have the attenuated form (i.e., diagnosis at 5 years of age and normal IQ), and 1 as intermediate (i.e., diagnosis after the age of 4 years and IQ between 80 and 90). After follow-up ranging from 7 to 17 years, all were still alive except 1 patient who died of unrelated causes. Successful engraftment was achieved in all patients, and cardiovascular abnormalities stabilized in all patients, hepatosplenomegaly resolved, and joint stiffness improved. Perceptual hearing defects remained stable, and transmission hearing defects improved. The neuropsychological outcome was variable: the 2 patients with the attenuated phenotype reached adulthood with normal IQ, social and scholastic development, and no language impairment. Four patients with the severe form of the syndrome deteriorated after the graft, and their IQ/developmental quotient had declined below 50 at their last evaluation. Of the patients with the severe form, 3 lost the ability to walk in their early teens, 2 lost language at 9 and 11 years of age, respectively, and 2 developed epilepsy. The remaining 2 patients with the severe form required special schooling and had poor social and language skills.

Sanfilippo Syndrome
Noncomparative Studies
Experience with allo-HCT in patients with Sanfilippo syndrome (MPS 111) has shown no alteration in the course of neuropsychologic deterioration seen in these patients.50 The literature addressing the use of HCT in Sanfilippo syndrome consists of 2 older case reports.50 Vellodi et al. (1992) reported on the outcomes of twin girls diagnosed with Sanfilippo syndrome who underwent allo-HCT and were followed for 9 yearss.52 At the time of transplant, both girls were functioning in the low-average range of intellectual development. Over the next 8 years, both girls had a steady decline in cognitive development, and both functioned in the area of significant developmental delay. The authors postulated that the continued deterioration in the twins, despite the demonstration of full chimerism, was a very low level of enzyme throughout the years after transplant. One other patient with Sanfilippo syndrome who had received allo-HCT was 5.3 years old at the time of the transplant and continued to deteriorate posttransplant.53

Morquio Syndrome
Allo-HCT has not been effective in Morquio syndromes.50

Section Summary: Inherited Metabolic Diseases Including Hunter, Sanfilippo, or Morquio Syndromes
The use of allo-HCT to treat patients with Hunter, Sanfilippo or Morquio syndromes does not result in improvements in neurologic, neuropsychologic, and neurophysiologic function.

Inherited Metabolic Diseases Excluding Hunter, Sanfilippo, or Morquio syndromes
Clinical Context and Test Purpose
The purpose of allo-HCT is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with inherited metabolic diseases excluding Hunter, Sanfilippo, or Morquio syndromes.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with an inherited metabolic disease?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with inherited metabolic diseases excluding Hunter, Sanfilippo, or Morquio syndromes.

Interventions
The therapy being considered is allo-HCT.

Patients with inherited metabolic diseases excluding Hunter, Sanfilippo, or Morquio syndromes are actively managed by metabolic specialists and primary care providers in an outpatient clinical setting. Patients with inherited metabolic diseases excluding Hunter, Sanfilippo, or Morquio syndromes undergoing HCT are actively managed in the inpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
The general outcomes of interest are OS, DSS, symptoms, QOL, and TRM.

Follow-up at 5.5- and 9.2-years is of interest for allo-HCT to monitor relevant outcomes.

Study Selection Criteria
Methodologically credible studies were selected using the following principles: 

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review articles summarize the experience using HGT to treat inherited metabolic diseases.54,55

Review of Evidence
Lysosomal Storage Disorders
Noncomparative Studies
HGT has been performed in approximately 20 of the estimated 40 known lysosomal storage disorders and peroxisomal storage disordes.6 Most instances (> 80%) have been in patients with Hurler syndrome (mucopolysaccharidosis I [MPS I]) or other MPS syndromes (Hunter syndrome [MPS II], Sanfilippo syndrome types A [MPS IIIA] and 8 [MPS 1118), Maroteaux-Lamy syndrome [MPS VI], adrenoleukodyst rophy, metachromatic leukodystrophy, and globoid cell leukodystrophy).6 Except for Hurler syndrome and globoid cell leukodystrophy, most published data are from single-case reports or small series with short follow-up.56 The benefit of allo-HCT appears to be limited to select subsets of patients with few types of lysosomal storage diseases and is not effective in patients who have developed overt neurologic symptoms or in those with aggressive infantile forms.56

Hurler syndrome is a lysosomal storage disease that, if left untreated, results in progressive multisystem morbidity including neuro-developmental deterioration, severe orthopedic manifestations, and cardiopulmonary complications leading to death in early childhood. Although enzyme replacement therapy is available, HGT remains the only treatment that delivers the deficient enzyme to the central nervous system.57 Impressive results have been observed with allo-HCT in Hurler syndrome. The benefits that have been observed include improvements in neurocognitive functioning, joint integrity, motor development, linear growth, corneal clouding, cardiac function, and others.6 Survival of engrafted Hurler syndrome patients has been radically changed from that of untransplanted patients, with long-term survival data indicating that lifespan can be extended by many decades.50 An 2007 analysis of nearly 150 transplanted patients with Hurler syndrome showed an OS rate of more than 80%.58

In 2015, an international retrospective analysis reported on long-term results of 217 patients with Hurler syndrome who successfully underwent allo-HCT between 1985 and 2011.57 Median follow-up was 9.2 years (range, 3 – 23 years), median age at diagnosis was 9 months (range, 0 – 42 months), and median age at transplant was 16 months (range, 2 – 47 months). Primary study endpoints were neurodevelopmental outcomes and growth; secondary endpoints included outcomes involving several different organ systems. Pre-HGT, 56.9% of patients showed normal neurodevelopment, and 26.6% showed only mildly impaired neurodevelopment. At last follow-up post-HGT, normal or only mildly impaired neurodevelopment was observed in 26.9% and 28.3% of the patients, respectively, and 44.9% suffered from moderately to severely impaired neurodevelopment. Predictors of better outcomes posttransplant were higher baseline developmental and IQ pretransplant, younger age at transplant, and a normal a-L-iduronidase enzyme level posttransplant.

Experience with allo-HCT and a RIC regimen was reported in 2008 for 7 patients with Hurler syndrome.59 Six of the patients received transplants from unrelated donors, and 1 received the transplant from a sibling. All patients had initial donor engraftment at 100 days, and there were no reports of severe acute GVHD. Six of the 7 children were alive at a median of 1014 days (range, 726 – 2222 days) posttransplant.

Mynarek et al. (2012) reported on the results of a retrospective, multicenter analysis of 17 patients with a-mannosidosis who underwent allo-HCT.60 Patients were diagnosed with the disease at a median age of 2.5 years (range, 1.1 – 23 years) and underwent allo-HCT at a median age of 3.6 years (1.3 – 23.1 years) . After a median follow-up of 5.5 years (range, 2.1 – 12.6 years), the OS rate was 88%. One patient died 76 days after transplantation from sepsis, GVHD, and pulmonary hemorrhage, and another patient died on day 135 posttransplant due to viral infections and multiorgan failure. Before allo­ HCT, the extent of developmental delay in the 17 patients varied over a wide range. After allo-HCT, patients made some developmental progress; however, normal development was not achieved. Hearing ability improved in some but not all patients.

Fewer than 40 patients with globoid cell leukodystrophy have undergone allo-HCT; however, there have been reports of dramatic improvements in neurologic, neuropsychologic, and neurophysiologic function.50

Many patients with metachromatic leukodystrophy who have undergone allo-HCT and had long-term engraftment have had amelioration of the disease signs and symptoms and prolonged survivai.50

The few patients with Maroteaux-Lamy syndrome (MPS VI) or Sly syndrome (MPS VII) who have received transplants have shown promising results, with clinical improvement posttransplant.50

Peroxisomal Disorders
Noncomparative Studies
Outcomes with allo-HCT have varied but promising. In boys and men with X-linked adrenoleukodystrophy, outcomes have depended on disease status at transplant and transplant-related complications,50 but reports of preservation of neuropsychologic and neurologic function have been presented.

Miller et al. (2011) reported on the results of 60 boys who underwent allo-HCT for cerebral adrenoleukodystrophy between 2000 and 2009.61 Median age at transplantation was 8.7 years; conditioning regimens and allograft sources varied. At HCT, 50% demonstrated a Loes radiographic severity score of 10 or more, and 62% showed clinical evidence of neurologic dysfunction. A total of 78% (n = 47) were alive at a median 3.7 years after allo-HCT. The 5-year survival estimate for boys with a Loes score less than 10 at HCT was 89%, whereas that for boys with a Loes score of 10 or more was 60% (p = 0.03). The 5-year survival estimate for boys without clinical cerebral disease at HCT was 91%, whereas that for boys with neurologic dysfunction was 66% (p = 0.08). The cumulative incidence of transplantation-related mortality at day 100 was 8%. Post-transplantation progression of neurologic dysfunction depended significantly on the pre-HCT Loes score and clinical neurologic status.

Section Summary: Inherited Metabolic Diseases Excluding Hunter, Sanfilippo, or Morquio Syndromes
Use of allo-HCT to treat select subsets of patients without overt neurologic symptoms or without aggressive infantile forms with lysosomal and peroxisomal storage disorders results in improvements in neurologic, neuropsychologic, and neurophysiologic function.

Genetic Disorders Affecting Skeletal Tissue
Clinical Context and Test Purpose
The purpose of allo-HCT is to provide a treatment option that is an alternative to or an improvement on existing therapies in patients with a genetic disorder affecting skeletal tissue.

The question addressed in this evidence review is: Does the use of allo-HCT improve the net health outcome in individuals with a genetic disorder affecting skeletal tissue?

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with a genetic disorder affecting skeletal tissue.

Interventions
The therapy being considered is allo-HCT.

Patients with a genetic disorder affecting skeletal tissue are actively managed by orthopedists and primary care providers in an outpatient clinical setting.

Comparators
Comparators of interest include standard of care.

Outcomes
The general outcomes of interest are OS, DSS, symptoms, QOL, and TRM.

Though not completely standardized, follow-up for a genetic disorder affecting skeletal tissue symptoms would typically occur in the months to years after starting treatment.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Noncomparative Studies
A 2010 review article has summarized the experience using HCT to treat osteopetrosis.62

The success of allo-HCT in infantile malignant osteopetrosis has depended greatly on the type of donor, with patients receiving grafts from HLA-identical siblings having 5-year DFS rates of 73% to 79% versus 13% to 45% for those requiring transplantation from an unrelated or mismatched donor.7

A 2003 retrospective analysis of 122 children who received an allo-HCT for autosomal recessive osteopetrosis between 1980 and 2001 reported 5-year DFS rates of 73% for recipients of a genotype HLA-identical HCT (n = 40); 43% for those of a phenotype HLA-identical or 1 HLA-antigen mismatch graft from a related donor (n = 21); 40% for recipients of a graft from a matched unrelated donor (n = 20); and 24% for patients who received an HLA-haplotype mismatch graft from a related donor (n = 41)63

Section Summary: Genetic Disorders Affecting Skeletal Tissues
Use of allo-HCT to treat patients with osteopetrosis results in improvements in DFS.

Summary of Evidence
For individuals who have a hemoglobinopathy, bone marrow failure syndrome, primary immunodeficiency, inherited metabolic syndrome disease (specifically those other than Hunter, Sanfilippo, or Morquio syndromes), or a genetic disorder affecting skeletal tissue who receive allo-HCT, the evidence includes mostly case series, case reports, and registry data. Relevant outcomes are overall survival, disease-specific survival, symptoms, quality of life, and treatment­ related morbidity. The evidence has shown that, for most of these disorders, there is a demonstrable improvement in overall survival and other disease-specific outcomes. Allo-HCT is likely to improve health outcomes in select patients with certain inherited and acquired diseases. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who have an inherited metabolic syndrome disease (specifically those including Hunter, Sanfilippo, and Morquio syndromes) who receive allo-HCT, the evidence includes case reports. Relevant outcomes are overall survival, disease-specific survival, symptoms, quality of life, and treatment-related morbidity. Use of allo-HCT to treat patients with Hunter, Sanfilippo, or Morquio syndromes does not result in improvements in neurologic, neuropsychologic, and neurophysiologic function. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Clinical Input From Physician Specialty Societies and Academic Medical Centers
While the various physician specialty societies and academic medical centers may collaborate with and make recommendations during this process, through the provision of appropriate reviewers, input received does not represent an endorsement or position statement by the physician specialty societies or academic medical centers, unless otherwise noted.

In response to requests, input was received from 1 physician specialty society (3 reviewers) and 3 academic medical centers while this policy was under review in 2009. There was general agreement with the policy statements. In particular, the reviewers were specifically asked to comment on the use of hematopoietic cell transplant in the inherited metabolic diseases, except for Hunter, Sanfilippo, and Morquio syndromes; 4 reviewers agreed with the current policy statement, 1 disagreed, and 1 did not address this specific question.

Practice Guidelines and Position Statements
American Society for Blood and Marrow Transplantation
In 2015, the American Society for Blood and Marrow Transplantation published consensus guidelines on the use of hematopoietic cell transplantation to treat specific conditions in and out of the clinical trial settings.64 Specific to this review Table 2 provides the allogeneic guidelines for specific indications.

Table 2. Recommendations for Use of Allogeneic HCT to Treat Genetic Diseases and Acquired Anemias

Table 2. Recommendations for Use of Allogeneic HCT to Treat Genetic Diseases and Acquired AnemiasIndications

Allogeneic HCT < 18 Years

Severe aplastic anemia, new diagnosis

S

Severe aplastic anemia, relapse/refractory

S

Fanconi anemia

R

Dyskeratosis congenita

R

Blackfan-Diamond anemia

R

Sickle cell disease

C

Thalassemia

S

Congenital amegakaryocytic thrombocytopenia

R

Severe combined immunodeficiency

R

T-cell immunodeficiency, severe combined immunodeficiency variants

R

Wiskott-Aldrich syndrome

R

Hemophagocytic disorders

R

Lymphoproliferative disorders

R

Severe congenital neutropenia

R

Chronic granulomatous disease

R

Other phagocytic cell disorders

R

Immunodysregulation polyendocrinopathy enteropathy X-linked syndrome

R

Juvenile rheumatoid arthritis

D

Systemic sclerosis

D

Other autoimmune and immune dysregulation disorders

R

Mucopolysaccharidoses (MPS-I and MPS-VI)

R

Other metabolic diseases

R

Osteopetrosis

R

Globoid cell leukodystrophy (Krabbe)

R

Metachromatic leukodystrophy

R

Cerebral X-linked adrenoleukodystrophy

R

Indications

Allogeneic HCT > 18 Years

Severe aplastic anemia, new diagnosis

S

Severe aplastic anemia, relapse/refractory

S

Fanconi anemia

R

Dyskeratosis congenita

R

Sickle cell disease

C

Thalassemia

D

Hemophagocytic syndromes, refractory

R

Mast cell diseases

R

Common variable immunodeficiency

R

Wiskott-Aldrich syndrome

R

Chronic granulomatous disease

R

Multiple sclerosis

N

Systemic sclerosis

N

Rheumatoid arthritis

N

Systemic lupus erythematosus

Crohn’s disease

Polymyositis-dermatomyositis 

C: clinical evidence available; D: developmental; HCT: hematopoietic cell transplantation; N: not generally recommended; R: standard of care, rare indication; S: standard of care.

British Committee for Standards in Haematology
In 2015, the British Committee for Standards in Haematology published guidelines on the diagnosis and management of adult aplastic anemia.65 The following key recommendations on HGT were included in the guidelines:

Matched sibling donor (allogeneic) HGT is the treatment of choice for severe aplastic anemia; however, for patients aged 35 to 50 years, patients need to be assessed for comorbidities before being considered for HGT.

For adults, unrelated donor HGT should be considered if patients fail to respond to a single course of immunosuppressive therapy.

Although there have been improvements in outcomes after alternative donor HGT, these transplants are still experimental, and expert consultation should be sought before considering their use.

European Blood and Marrow Transplantation
In 2014, the European Blood and Marrow Transplantation provided consensus-based recommendations on indications for HGT and transplant management in the hemoglobinopathies.10

Pediatric Haemato-Oncology Italian Association
In 2015, the Pediatric Haemato-Oncology Italian Association issued guidelines on the diagnosis and treatment of acquired aplastic anemia in childhood.66

U.S. Preventive Services Task Force Recommendations
Not applicable

Ongoing and Unpublished Clinical Trials
Some currently unpublished trials that might influence this review are listed in Table 3.

Table 3. Summary of Key Trials

NCT No.                     I Trial Name

Planned Enrollment

Completion Date

Ongoing

 

 

 

 

NCT02766465

A Study to Compare Bone Marrow Transplantation to Standard Care in Adolescents and Young Adults With Severe Sickle Cell Disease

 

200

 

Mar 2023

NCT02356653

Expanded Access Protocol Using CD3+/CD19+ Depleted Unrelated Donor or Partially Matched Related Donor Peripheral Stem Cells

100

Jan 2021

NCT02986698

A Single-Center, Non-Randomized Study of the Safety and Efficacy of In Utero Hematopoietic Stem Cell Transplantation for the Treatment of Fetuses With Alpha Thalassemia Major

10

Feb 2024

Unpublished

 

NCT00176826

In-vivo T-cell Depletion and Hematopoietic Stem Cell Transplantation for Life-Threatening Immune Deficiencies and Histiocytic Disorders

22

Aug 2015

(Terminated)

NCT: national clinical trial.

References  

  1. Bhatia M, Walters MC. Hematopoietic cell transplantation for thalassemia and sickle cell disease: past, present andfuture. Bone Marrow Transplant. Jan 2008; 41(2): 109-17. PMID 18059330
  2. Mehta P. Hematopoietic stem cell transplantation for inherited bone marrow failure syndromes. In: Mehta P, ed.Pediatric Stem Cell Transplantation. Sudbury, MA: Jones and Bartlett; 2004:281-316.
  3. Gluckman E, Wagner JE. Hematopoietic stem cell transplantation in childhood inherited bone marrow failuresyndrome. Bone Marrow Transplant. Jan 2008; 41(2): 127-32. PMID 18084332
  4. Gennery AR, Cant AJ. Advances in hematopoietic stem cell transplantation for primary immunodeficiency. ImmunolAllergy Clin North Am. May 2008; 28(2): 439-56, x-xi. PMID 18424341
  5. Porta F, Forino C, De Martiis D, et al. Stem cell transplantation for primary immunodeficiencies. Bone MarrowTransplant. Jun 2008; 41 Suppl 2: S83-6. PMID 18545252
  6. Prasad VK, Kurtzberg J. Emerging trends in transplantation of inherited metabolic diseases. Bone MarrowTransplant. Jan 2008; 41(2): 99-108. PMID 18176609
  7. Askmyr MK, Fasth A, Richter J. Towards a better understanding and new therapeutics of osteopetrosis. Br JHaematol. Mar 2008; 140(6): 597-609. PMID 18241253
  8. MacMillan ML, Walters MC, Gluckman E. Transplant outcomes in bone marrow failure syndromes and hemoglobinopathies. Semin Hematol. Jan 2010; 47(1): 37-45. PMID 20109610
  9. Smiers FJ, Krishnamurti L, Lucarelli G. Hematopoietic stem cell transplantation for hemoglobinopathies: currentpractice and emerging trends. Pediatr Clin North Am. Feb 2010; 57(1): 181-205. PMID 20307718
  10. Angelucci E, Matthes-Martin S, Baronciani D, et al. Hematopoietic stem cell transplantation in thalassemia majorand sickle cell disease: indications and management recommendations from an international expert panel.Haematologica. May 2014; 99(5): 811-20. PMID 24790059
  11. Mathews V, Srivastava A, Chandy M. Allogeneic stem cell transplantation for thalassemia major. Hematol OncolClin North Am. Dec 2014; 28(6): 1187-200. PMID 25459187
  12. Locatelli F, Kabbara N, Ruggeri A, et al. Outcome of patients with hemoglobinopathies given either cord blood orbone marrow transplantation from an HLA-identical sibling. Blood. Aug 08 2013; 122(6): 1072-8. PMID 23692854
  13. Mehta P. Hematopoietic stem cell transplantation for hemoglobinopathies. In: Mehta P, ed. Pediatric Stem CellTransplantation. Sudbury, MA: Jones and Bartlett; 2004:259-279.
  14. Mahmoud HK, Elhaddad AM, Fahmy OA, et al. Allogeneic hematopoietic stem cell transplantation for non-malignant hematological disorders. J Adv Res. May 2015; 6(3): 449-58. PMID 26257943
  15. Bernardo ME, Piras E, Vacca A, et al. Allogeneic hematopoietic stem cell transplantation in thalassemia major:results of a reduced-toxicity conditioning regimen based on the use of treosulfan. Blood. Jul 12 2012; 120(2): 473-6. PMID 22645178
  16. Anurathapan U, Pakakasama S, Mekjaruskul P, et al. Outcomes of thalassemia patients undergoing hematopoieticstem cell transplantation by using a standard myeloablative versus a novel reduced-toxicity conditioning regimenaccording to a new risk stratification. Biol Blood Marrow Transplant. Dec 2014; 20(12): 2066-71. PMID 25064743
  17. Oringanje C, Nemecek E, Oniyangi O. Hematopoietic stem cell transplantation for people with sickle cell disease.Cochrane Database Syst Rev. May 31 2013; (5): CD007001. PMID 23728664
  18. Oringanje C, Nemecek E, Oniyangi O. Hematopoietic stem cell transplantation for people with sickle cell disease.Cochrane Database Syst Rev. May 19 2016; (5): CD007001. PMID 27194464
  19. Oringanje C, Nemecek E, Oniyangi O. Hematopoietic stem cell transplantation for people with sickle cell disease.Cochrane Database Syst Rev. Jul 03 2020; 7: CD007001. PMID 32617981
  20. Bernaudin F, Socie G, Kuentz M, et al. Long-term results of related myeloablative stem-cell transplantation to curesickle cell disease. Blood. Oct 01 2007; 110(7): 2749-56. PMID 17606762
  21. Walters MC, Patience M, Leisenring W, et al. Bone marrow transplantation for sickle cell disease. N Engl J Med.Aug 08 1996; 335(6): 369-76. PMID 8663884
  22. Walters MC, Storb R, Patience M, et al. Impact of bone marrow transplantation for symptomatic sickle cell disease:an interim report. Multicenter investigation of bone marrow transplantation for sickle cell disease. Blood. Mar 152000; 95(6): 1918-24. PMID 10706855
  23. Hsieh MM, Fitzhugh CD, Weitzel RP, et al. Nonmyeloablative HLA-matched sibling allogeneic hematopoietic stemcell transplantation for severe sickle cell phenotype. JAMA. Jul 02 2014; 312(1): 48-56. PMID 25058217
  24. Mehta P, Locatelli F, Stary J, et al. Bone marrow transplantation for inherited bone marrow failure syndromes.Pediatr Clin North Am. Feb 2010; 57(1): 147-70. PMID 20307716
  25. Miano M, Dufour C. The diagnosis and treatment of aplastic anemia: a review. Int J Hematol. Jun 2015; 101(6):527-35. PMID 25837779
  26. Bacigalupo A. Bone marrow transplantation for acquired severe aplastic anemia. Hematol Oncol Clin North Am.Dec 2014; 28(6): 1145-55. PMID 25459184
  27. Yabe M, Morio T, Tabuchi K, et al. Long-term outcome in patients with Fanconi anemia who received hematopoieticstem cell transplantation: a retrospective nationwide analysis. Int J Hematol. Jan 2021; 113(1): 134-144. PMID32949371
  28. Dufour C, Svahn J. Fanconi anaemia: new strategies. Bone Marrow Transplant. Jun 2008; 41 Suppl 2: S90-5.PMID 18545254
  29. Zanis-Neto J, Flowers ME, Medeiros CR, et al. Low-dose cyclophosphamide conditioning for haematopoietic celltransplantation from HLA-matched related donors in patients with Fanconi anaemia. Br J Haematol. Jul 2005;130(1): 99-106. PMID 15982351
  30. Wagner JE, Eapen M, MacMillan ML, et al. Unrelated donor bone marrow transplantation for the treatment ofFanconi anemia. Blood. Mar 01 2007; 109(5): 2256-62. PMID 17038525
  31. Gadalla SM, Sales-Bonfim C, Carreras J, et al. Outcomes of allogeneic hematopoietic cell transplantation inpatients with dyskeratosis congenita. Biol Blood Marrow Transplant. Aug 2013; 19(8): 1238-43. PMID 23751955
  32. Cesaro S, Oneto R, Messina C, et al. Haematopoietic stem cell transplantation for Shwachman-Diamond disease:a study from the European Group for blood and marrow transplantation. Br J Haematol. Oct 2005; 131(2): 231-6.PMID 16197455
  33. Myers K, Hebert K, Antin J, et al. Hematopoietic Stem Cell Transplantation for Shwachman-Diamond Syndrome.Biol Blood Marrow Transplant. Aug 2020; 26(8): 1446-1451. PMID 32428734
  34. Cesaro S, Pillon M, Sauer M, et al. Long-term outcome after allogeneic hematopoietic stem cell transplantation forShwachman-Diamond syndrome: a retrospective analysis and a review of the literature by the Severe AplasticAnemia Working Party of the European Society for Blood and Marrow Transplantation (SAAWP-EBMT). BoneMarrow Transplant. Sep 2020; 55(9): 1796-1809. PMID 32203264
  35. Roy V, Perez WS, Eapen M, et al. Bone marrow transplantation for diamond-blackfan anemia. Biol Blood MarrowTransplant. Aug 2005; 11(8): 600-8. PMID 16041310
  36. ElGohary G, El Fakih R, de Latour R, et al. Haploidentical hematopoietic stem cell transplantation in aplasticanemia: a systematic review and meta-analysis of clinical outcome on behalf of the severe aplastic anemia workingparty of the European group for blood and marrow transplantation (SAAWP of EBMT). Bone Marrow Transplant.Oct 2020; 55(10): 1906-1917. PMID 32346079
  37. Kim H, Lee JH, Joo YD, et al. A randomized comparison of cyclophosphamide vs. reduced dosecyclophosphamide plus fludarabine for allogeneic hematopoietic cell transplantation in patients with aplasticanemia and hypoplastic myelodysplastic syndrome. Ann Hematol. Sep 2012; 91(9): 1459-69. PMID 22526363
  38. Dufour C, Pillon M, Socie G, et al. Outcome of aplastic anaemia in children. A study by the severe aplasticanaemia and paediatric disease working parties of the European group blood and bone marrow transplant. Br JHaematol. May 2015; 169(4): 565-73. PMID 25683884
  39. Smith AR, Gross TG, Baker KS. Transplant outcomes for primary immunodeficiency disease. Semin Hematol. Jan2010; 47(1): 79-85. PMID 20109615
  40. Szabolcs P, Cavazzana-Calvo M, Fischer A, et al. Bone marrow transplantation for primary immunodeficiencydiseases. Pediatr Clin North Am. Feb 2010; 57(1): 207-37. PMID 20307719
  41. Ahlin A, Fugelang J, de Boer M, et al. Chronic granulomatous disease-haematopoietic stem cell transplantationversus conventional treatment. Acta Paediatr. Nov 2013; 102(11): 1087-94. PMID 23937637
  42. Gungor T, Teira P, Slatter M, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-celltransplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet. Feb 012014; 383(9915): 436-48. PMID 24161820
  43. Chiesa R, Wang J, Blok HJ, et al. Hematopoietic cell transplantation in chronic granulomatous disease: a study of712 children and adults. Blood. Sep 03 2020; 136(10): 1201-1211. PMID 32614953
  44. Filipovich A. Hematopoietic cell transplantation for correction of primary immunodeficiencies. Bone MarrowTransplant. Aug 2008; 42 Suppl 1: S49-S52. PMID 18724301
  45. Hassan A, Booth C, Brightwell A, et al. Outcome of hematopoietic stem cell transplantation for adenosinedeaminase-deficient severe combined immunodeficiency. Blood. Oct 25 2012; 120(17): 3615-24; quiz 3626. PMID22791287
  46. Filipovich AH, Stone JV, Tomany SC, et al. Impact of donor type on outcome of bone marrow transplantation forWiskott-Aldrich syndrome: collaborative study of the International Bone Marrow Transplant Registry and theNational Marrow Donor Program. Blood. Mar 15 2001; 97(6): 1598-603. PMID 11238097
  47. Moratto D, Giliani S, Bonfim C, et al. Long-term outcome and lineage-specific chimerism in 194 patients withWiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980-2009: an internationalcollaborative study. Blood. Aug 11 2011; 118(6): 1675-84. PMID 21659547
  48. Burroughs LM, Petrovic A, Brazauskas R, et al. Excellent outcomes following hematopoietic cell transplantation forWiskott-Aldrich syndrome: a PIDTC report. Blood. Jun 04 2020; 135(23): 2094-2105. PMID 32268350
  49. Marsh RA, Bleesing JJ, Chandrakasan S, et al. Reduced-intensity conditioning hematopoietic cell transplantation isan effective treatment for patients with SLAM-associated protein deficiency/X-linked lymphoproliferative diseasetype 1. Biol Blood Marrow Transplant. Oct 2014; 20(10): 1641-5. PMID 24923536
  50. Mehta P. Metabolic diseases. In: Mehta P, ed. Pediatric Stem Cell Transplantation. Sudbury, MA: Jones andBartlett; 2004:233-258.
  51. Guffon N, Bertrand Y, Forest I, et al. Bone marrow transplantation in children with Hunter syndrome: outcome after7 to 17 years. J Pediatr. May 2009; 154(5): 733-7. PMID 19167723
  52. Vellodi A, Young E, New M, et al. Bone marrow transplantation for Sanfilippo disease type B. J Inherit Metab Dis.1992; 15(6): 911-8. PMID 1293388
  53. Bordigoni P, Vidailbet M, Lena M, et al. Bone marrow transplantation for Sanfilippo syndrome. In: Hobbs JR, ed.Correction of Certain Genetic Diseases by Transplantation. London: Cogent; 1989:114-119.
  54. Boelens JJ, Prasad VK, Tolar J, et al. Current international perspectives on hematopoietic stem cell transplantationfor inherited metabolic disorders. Pediatr Clin North Am. Feb 2010; 57(1): 123-45. PMID 20307715
  55. Prasad VK, Kurtzberg J. Transplant outcomes in mucopolysaccharidoses. Semin Hematol. Jan 2010; 47(1): 59-69.PMID 20109613
  56. Rovelli AM. The controversial and changing role of haematopoietic cell transplantation for lysosomal storagedisorders: an update. Bone Marrow Transplant. Jun 2008; 41 Suppl 2: S87-9. PMID 18545253
  57. Aldenhoven M, Wynn RF, Orchard PJ, et al. Long-term outcome of Hurler syndrome patients after hematopoieticcell transplantation: an international multicenter study. Blood. Mar 26 2015; 125(13): 2164-72. PMID 25624320
  58. Boelens JJ, Wynn RF, O'Meara A, et al. Outcomes of hematopoietic stem cell transplantation for Hurler's syndromein Europe: a risk factor analysis for graft failure. Bone Marrow Transplant. Aug 2007; 40(3): 225-33. PMID17529997
  59. Hansen MD, Filipovich AH, Davies SM, et al. Allogeneic hematopoietic cell transplantation (HCT) in Hurler'ssyndrome using a reduced intensity preparative regimen. Bone Marrow Transplant. Feb 2008; 41(4): 349-53. PMID18026148
  60. Mynarek M, Tolar J, Albert MH, et al. Allogeneic hematopoietic SCT for alpha-mannosidosis: an analysis of 17patients. Bone Marrow Transplant. Mar 2012; 47(3): 352-9. PMID 21552297
  61. Miller WP, Rothman SM, Nascene D, et al. Outcomes after allogeneic hematopoietic cell transplantation forchildhood cerebral adrenoleukodystrophy: the largest single-institution cohort report. Blood. Aug 18 2011; 118(7):1971-8. PMID 21586746
  62. Steward CG. Hematopoietic stem cell transplantation for osteopetrosis. Pediatr Clin North Am. Feb 2010; 57(1):171-80. PMID 20307717
  63. Driessen GJ, Gerritsen EJ, Fischer A, et al. Long-term outcome of haematopoietic stem cell transplantation inautosomal recessive osteopetrosis: an EBMT report. Bone Marrow Transplant. Oct 2003; 32(7): 657-63. PMID13130312
  64. Majhail NS, Farnia SH, Carpenter PA, et al. Indications for Autologous and Allogeneic Hematopoietic CellTransplantation: Guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood MarrowTransplant. Nov 2015; 21(11): 1863-1869. PMID 26256941
  65. Killick SB, Bown N, Cavenagh J, et al. Guidelines for the diagnosis and management of adult aplastic anaemia. BrJ Haematol. Jan 2016; 172(2): 187-207. PMID 26568159
  66. Barone A, Lucarelli A, Onofrillo D, et al. Diagnosis and management of acquired aplastic anemia in childhood.Guidelines from the Marrow Failure Study Group of the Pediatric Haemato-Oncology Italian Association (AIEOP).Blood Cells Mol Dis. Jun 2015; 55(1): 40-7. PMID 25976466

Coding Section

Codes Number Description
CPT 38204 Management of recipient hematopoietic cell donor search and cell acquisition
  38205 Blood-derived hematopoietic progenitor cell harvesting for transplantation, per collection, allogeneic
  38208 Transplant preparation of hematopoietic progenitor cells; thawing of previously frozen harvest, without washing
  38209 Thawing of previously frozen harvest, with washing
  38210 Specific cell depletion within harvest, T-cell depletion
  38212 Red blood cell removal
  38213 Platelet depletion
  38214 Plasma (volume) depletion
  38215 Cell concentration in plasma, mononuclear, or buffy coat layer
  38220  Bone marrow; aspiration only
  38221 Bone marrow; biopsy, needle or trocar
  38230 Bone marrow harvesting for transplantation; allogeneic
  38240 Bone marrow or blood-derived peripheral stem-cell transplantation; allogeneic
  38242 Allogeneic donor lymphocyte infusions
  86812-86822 Histocompatibility studies code range
ICD-9 Procedure 41.02 Allogeneic bone marrow transplant with purging
  41.03 Allogeneic bone marrow transplant without purging
  41.05 Allogeneic hematopoietic stem-cell transplant without purging
  41.08 Allogeneic hematopoietic stem-cell transplant with purging
  41.91 Aspiration of bone marrow from donor for transplant
  99.79 Other therapeutic apheresis (includes harvest of stem cells)
ICD-9 Diagnosis 272.7 Lipidoses (includes mucolipidosis)
  277.5 Mucopolysaccharidosis
  279.12 Wiskott-Aldrich syndrome
  279.2 Combined immunity deficiency
  282.41-282.49 Thalassemia code range
  282.60-282.69 Sickle-cell disease code range
  284.01-284.9 Aplastic anemia code range
  288.01 Congenital neutropenia (includes Kostmann’s syndrome)
  756.52 Osteopetrosis
HCPCS G0265

Cryopreservation, freezing and storage of cells for therapeutic use, each cell line

  G0266 Thawing and expansion of frozen cells for therapeutic use, each cell line
  G0267 Bone marrow or peripheral stem-cell harvest, modification or treatment to eliminate cell type(s) (e.g., T cells, metastatic carcinoma)
  Q0083-Q0085 Chemotherapy administration code range
  J9000-J9999 Chemotherapy drug code range
  S2140 Cord blood harvesting for transplantation, allogeneic
  S2142 Cord blood-derived stem-cell transplantation, allogeneic
  S2150 Bone marrow or blood-derived peripheral stem-cell harvesting and transplantation, allogeneic or autologous, including pheresis, high-dose chemotherapy, and the number of days of post-transplant care in the global definition (including drugs; hospitalization; medical, surgical, diagnostic and emergency services)
ICD-10-CM (effective 10/01/15) D56.1 Beta thalassemia
  D57.00-D57.819 Sickle-cell disorders code range
  D61.01-D61.89 Other aplastic anemias and other bone marrow failure syndromes code range (includes Fanconi anemia)
  D70.0 Congenital agranulocytosis (includes Kostmann’s disease)
  D71 Functional disorders of polymorphonuclear neutrophils (includes chronic granulomatous disease)
  D81.0-D81.9 Combined immunodeficiencies code range
  D82.0-D82.9 Immunodeficiency associated with other major defects code range (includes Wiskott-Aldrich and X-linked lymphoproliferative syndromes)
  E70.330 Chediak-Higashi syndrome
  E71.50-E71.548 Peroxisomal disorders code range (includes childhood cerebral X-linked adrenoleukodystrophy)
  E75.25 Metachromatic leukodystrophy
  E76.01 Hurler’s syndrome
  E76.29 Other mucopolysaccharidoses (includes Maroteaux-Lamy syndrome)
  E77.01 Defects in glycoprotein degradation (includes aspartylglucosaminuria, mannosidosis and fucosidosis)
 

Q78.2

Osteopetrosis (includes Albers-Schonberg syndrome)
ICD-10-PCS (effective 10/01/15)   ICD-10-PCS codes are only used for inpatient services.
  30243G1, 30243X1, 30243Y1 Percutaneous transfusion, central vein, bone marrow or stem cells, nonautologous, code list
  07DQ0ZZ, 07DQ3ZZ, 07DR0ZZ, 07DR3ZZ, 07DS0ZZ, 07DS3ZZ

Surgical, lymphatic and hemic systems, extraction, bone marrow, code list

Type of Service Therapy  
Place of Service Inpatient/Outpatient  

Procedure and diagnosis codes on Medical Policy documents are included only as a general reference tool for each policy. They may not be all-inclusive. 

This medical policy was developed through consideration of peer-reviewed medical literature generally recognized by the relevant medical community, U.S. FDA approval status, nationally accepted standards of medical practice and accepted standards of medical practice in this community, Blue Cross Blue Shield Association technology assessment program (TEC) and other nonaffiliated technology evaluation centers, reference to federal regulations, other plan medical policies and accredited national guidelines.

"Current Procedural Terminology © American Medical Association. All Rights Reserved" 

History From 2024 Forward     

07/10/2024 Annual review, no change to policy intent 

01/01/2024 NEW POLICY

Complementary Content
${loading}