Acquired severe aplastic anemia (SAA) is a rare hematologic disease associated with significant morbidity and mortality. Immune destruction of hemopoietic stem cells plays an important role in pathogenesis, as shown by successful treatment with immunosuppressive agents, leading to transfusion independence or complete recovery of peripheral blood counts in a proportion of patients. Growth factors can be combined with immunosuppressive therapy (IST) and may improve response rates, as recently shown with thrombopoietin analogs. Anabolic steroids may still play a role in combination with IST. The problem with IST is failure to respond and the development of late clonal disorders. Bone marrow transplantation (BMT) is the other therapeutic option: a matched sibling donor remains the best choice. For patients lacking a matched family donor, unrelated donors can be readily found, although mostly for patients of Caucasian origin. Other BMT options include unrelated cord blood or mismatched family donors. Acute and chronic graft-versus-host disease remain important complications of BMT. Patient age is a strong predictor of outcome for both IST and BMT, and must be considered when designing therapeutic strategies. Early diagnosis and treatment, as well as long-term monitoring, remain crucial steps for successful treatment of SAA.

Pathogenesis

Acquired SAA is regarded as the result of an immune-mediated destruction of hematopoietic cells, at least in a proportion of patients.1  The emergence of late clonal disorders in 10% to 20% of patients after immunosuppressive therapy (IST)2  raises the questions of whether some patients with SAA actually have a premalignant disease and whether IST is just postponing the inevitable.3  Support for this view has come from the identification of somatic mutations involving telomerase RNA component (TERC) and telomerase reverse transcriptase (TERT)4  and, more recently, involving myeloid cancer candidate genes in a significant proportion of patients.5-7 

Diagnosis and early intervention

The diagnosis of acquired SAA is based on the exclusion of other disorders that can cause pancytopenia and on the well-known Camitta criteria8  (Figure 1). A BM biopsy is mandatory (preceded by platelet transfusions if the platelet count is below 20 × 109/L) and will confirm an empty marrow; it should also exclude a MDS or leukemia, as well as marrow metastasis from solid tumors (Figure 1). A BM aspirate will be used for cytogenetics and/or FISH analysis to determine chromosomal abnormalities. Whether the identification of chromosomal abnormalities is compatible with the diagnosis of SAA is debated9 ; clearly, some abnormalities carry a poor prognosis (such as deletion of chromosome 7), whereas others (such as +Y and +8) are more benign and may not affect the therapeutic strategy.9  Identification of a paroxysmal nocturnal hemoglobinuria (PNH) clone by flow cytometry will help exclude an inherited form of marrow failure and may suggest this is not a hypoplastic MDS (Figure 1); a negative diepoxybutane test will exclude FA. Determination of telomere length is not mandatory, but will help exclude telomeropathies.4 

Figure 1.

Diagnostic procedures in patients with pancytopenia. ATG, antithymocyte globulin; BM, bone marrow; CsA, cyclosporine A; DEB, diepoxybutane; FA, Fanconi’s anemia; FISH, fluorescent in situ hybridization; GPI, glycosyl phosphatidyl inositol; MDS, myelodysplastic syndrome.

Figure 1.

Diagnostic procedures in patients with pancytopenia. ATG, antithymocyte globulin; BM, bone marrow; CsA, cyclosporine A; DEB, diepoxybutane; FA, Fanconi’s anemia; FISH, fluorescent in situ hybridization; GPI, glycosyl phosphatidyl inositol; MDS, myelodysplastic syndrome.

Close modal

Severity can be determined by neutrophil counts: patients with 0 to 0.2, 0.21 to 0.5, and >0.5 polymorphonuclear cells (PMNs) × 109/L are classified, respectively, as very severe, severe, and nonsevere aplastic anemia,10  and severity has been a strong predictor of survival in patients receiving IST.10,11  Patients with SAA require early diagnosis and intervention, whether IST or BMT, because the interval between diagnosis and treatment is another strong predictor of survival.12  Transfusion policies are important in the early days of diagnosis, and guidelines for supportive care have been published.13  In approximately 5% of patients, SAA will follow an episode of elevated transaminase and hyperbilirubinemia,14  although the search for hepatitis A, B, and C virus (HAV, HBV, and HCV) is typically negative, as well as the search for other viruses. Abnormal liver function test and/or elevated bilirubin levels should not stop therapeutic strategies.

Initially, one should concentrate on the diagnosis,15  with some key tests, as outlined in Figure 1. The BM biopsy is the diagnostic procedure with the highest level of accuracy. In the meantime, the patient will be transfused according to guidelines.13  Once the diagnosis has been ascertained, HLA typing of the patient and his/her family should be one of the first interventions in a patient with SAA, certainly in patients younger than 60 years of age.

BMT or IST

If an HLA-matched family donor is identified, marrow transplantation should be the first-line therapy in patients younger than 40 years (Figure 2); this is based on studies comparing HLA-identical BMT vs first-line IST.11,16  However, the advantage in failure-free survival for a young patient with a low neutrophil count declines with increasing age11  as a result of higher mortality after HLA-identical BMT in patients aged 21 to 40 years or older than 40 years.

Figure 2.

Treatment strategy in patients with acquired aplastic anemia. Patients are stratified according to whether or not they have an HLA-identical sibling. In young patients (<40 years) with a matched donor, allogeneic BMT is first-line therapy. In patients older than 40 years, and for patients without a matched sibling, ATG+CsA should be first-line therapy. In selected children with very severe disease, an unrelated donor (UD) graft may be considered first-line therapy (dashed arrow). In patients aged 0 to 60 years, nonresponders (no resp) to ATG have several choices, depending on the performance status of the patient, the availability of an HLA-matched UD, and patient age. The options are an UD BMT, a second course of IST (ATG+CsA), or an alternative donor transplant (Alt Donor Tx, indicating haploidentical transplants or cord blood [CB] transplants). At older than 60 years of age, medical treatment is preferable over BMT. d+120 = day +120; EPAG, eltrombopag; HLA = Sib, HLA-identical sibling; Sib BMT, identical sibling transplantation; UD BMT, unrelated donor BMT.

Figure 2.

Treatment strategy in patients with acquired aplastic anemia. Patients are stratified according to whether or not they have an HLA-identical sibling. In young patients (<40 years) with a matched donor, allogeneic BMT is first-line therapy. In patients older than 40 years, and for patients without a matched sibling, ATG+CsA should be first-line therapy. In selected children with very severe disease, an unrelated donor (UD) graft may be considered first-line therapy (dashed arrow). In patients aged 0 to 60 years, nonresponders (no resp) to ATG have several choices, depending on the performance status of the patient, the availability of an HLA-matched UD, and patient age. The options are an UD BMT, a second course of IST (ATG+CsA), or an alternative donor transplant (Alt Donor Tx, indicating haploidentical transplants or cord blood [CB] transplants). At older than 60 years of age, medical treatment is preferable over BMT. d+120 = day +120; EPAG, eltrombopag; HLA = Sib, HLA-identical sibling; Sib BMT, identical sibling transplantation; UD BMT, unrelated donor BMT.

Close modal

The age effect

In patients grafted from matched siblings, there is a very strong age effect, with survival of 82%, 72%, and 53% for patients aged 1 to 20, 21 to 40, and older than 40 years,17  as a result of a higher incidence of graft failure and graft-versus-host disease (GVHD).17  European Group for Blood and Marrow Transplantation (EBMT) data for patients grafted from matched siblings in the decade from 2001 to 2010 show the same age effect (Figure 3): 86%, 76%, and 55% survival at 10 years. For this reason, IST should be first-line therapy in older patients, as shown in Figure 2, and BMT should be carefully considered for selected cases with good performance status and severe disease (Figure 2, dashed arrow).

Figure 3.

A strong age effect in patients with aplastic anemia, after transplantation from an HLA identical sibling. Data from the EBMT registry.

Figure 3.

A strong age effect in patients with aplastic anemia, after transplantation from an HLA identical sibling. Data from the EBMT registry.

Close modal

The conditioning regimen

The standard conditioning regimen for matched sibling transplants is cyclophosphamide 200 mg/kg (CY 200) and ATG, as originally described.18  Although a randomized study failed to show an advantage for ATG,19  the same survival difference proved significant in a larger retrospective study20  This regimen is excellent for young patients, but CY 200 may be too toxic in older patients, although attempts have been made to reduce toxicity with fludarabine (FLU)-based regimens and lower doses of CY.21-24  Recent EBMT data (A.B., EBMT database, unpublished data) suggest that survival of patients older than 40 years can be significantly improved with a FLU-based regimen, in addition to ATG or alemtuzumab (CAMPATH), and is comparable to survival of patients in the 21- to 40-year-old age group (74% vs 75%). Current guidelines from EBMT25  and the British Society for Standards in Haematolgy26  call for a combination of FLU-CY with ATG (FCA) or alemtuzumab (CAMPATH) (FCC) for patients with SAA who are older than 30 years and receiving a matched sibling donor transplant. The CY dose to be combined with FLU is a matter of discussion, ranging from 40 to 120 mg/kg.

BM or peripheral blood

Two registry-based studies have shown that BM results in superior outcome as compared with peripheral blood (PB) in matched sibling transplants20,27  because of less acute and chronic GVHD with BM and comparable risk for rejection (2.5% for PB and 1.5% for BM). The recent British guidelines call for BM as a stem cell source in ATG-based conditioning.26  The evidence we currently have suggests BM should be the only acceptable stem cell source for transplants from HLA-identical siblings in SAA.

Case report

A 23-year-old woman was referred to us in December 2011, having failed 2 courses of ATG and CsA. The patient had developed an invasive fungal infection after the IST, with lung lesions and a left pneumothorax. She was admitted on December 20, 2010, with drainage in her left pleural cavity, high temperature, and absolute pancytopenia. In addition, the patient had developed polyneuritis with almost complete tetraplegia. Her performance status was extremely poor. She received a conditioning regimen including FLU 120 mg/m2, CY 120 mg/kg, ATG (thymoglobulin; Sanofi France) 3.75 mg/kg2, and a BMT from an 8/8 mated UD. Prophylaxis of GVHD consisted of CsA and short-course methotrexate; rituximab 200 mg was given on day +5 to prevent Epstein-Barr virus (EBV) reactivation. Treatment with voriconazole was continued. Engraftment was rapid and complete, and the pleural drainage was removed 1 month later. There was no GVHD and no EBV reactivation, but neurologic rehabilitation was slow, and the patient remained admitted in the transplant unit for several months. Antifungal treatment was continued, and lyposomial amphotericin (AmBisome) was also introduced. Two years later, the patient underwent upper left lobectomy to remove her aspergillum lesions because of their proximity to large vessels. The patients is alive and well and off immunosuppression 4 and a half years posttransplant.

This case exemplifies the infectious complications of prolonged neutropenia and immune suppression in aplastic anemia, as well as showing that an UD BMT can be performed successfully, even in very sick patients with marrow failure, in keeping with the reported improved outcome of UD grafts.28,29  This case also suggests that in the presence of a matched UD, perhaps second-line therapy should be a UD transplant, rather than a second course of ATG (Figure 2). In the absence of a matched sibling donor, I usually start a search for an UD as early as possible, certainly when the patient is younger than 50 years of age. Early identification of an 8/8 HLA-A, -B, -C, or -DRB1–matched donor is always important, especially for pediatric patients, for whom up-front UD has shown to have 96% long-term survival, which is comparable to up-front sibling BMT.28  For this reason, Figure 2 outlines a possible dashed line for patients younger than 20 years, with an option to proceed to an UD BMT as first-line therapy, although a first course of IST remains standard of care, as suggested by identical survival at 2 years for first-line UD BMT and IST.28  UD transplantation should also be considered in patients failing a first course of IST, as already mentioned (Figure 2). Increasing patient age and HLA matching remain significant predictors of survival after UD grafts29-32  (Figure 4). I consider an 8/8 HLA-matched UD the optimal choice, possibly matched with the recipient, for cytomegalovirus sero-status.33 

Figure 4.

The age effect in UD transplants: best outcome is seen for very young patients, for whom first-line UD BMT may be considered. Data from the EBMT registry.

Figure 4.

The age effect in UD transplants: best outcome is seen for very young patients, for whom first-line UD BMT may be considered. Data from the EBMT registry.

Close modal

UDs and stem cell source.

In addition, for UD grafts, BM provides a survival advantage over PB; this has been shown in studies from EBMT and from the Center for International Blood and Marrow Transplant Research.34,35  In the recent EBMT analysis, PB was the strongest negative predictor of survival in multivariate analysis.34  The British guidelines26  suggest PB can be used as a stem cell source with CAMPATH as GVHD prophylaxis, despite significant inferior survival of PB vs BM transplants (P = .03) in the British report on patients grafted with the FCC conditioning regimen.36  I personally think the data strongly suggest we should always use BM as a stem cell source; an exception could be a second transplant because of graft rejection.

UD BMT and conditioning regimens.

Several studies have explored the combination of low-dose total body irradiation (TBI) with FLU-CY29,32,37-39 ; the dosage of both TBI and cyclophosphamide has been the topic of prospective studies.40,41  The current recommendation is CY at a dose between 50 and 100 mg/kg (total dose), TBI between 2 and 4 Gy, and FLU 100–150 mg/m2.26  Registry data (EBMT) for UD transplants show survival of 85%, 77%, 66%, and 49% for patients aged 1 to 10, 11 to 30, 31 to 40, and older than 40 years (Figure 4). The donor’s age appears to be an additional prognostic factor.42 

The ideal UD is a male, HLA-matched donor at the A,B,C,DRB1 loci, younger than 30 years, cytomegalovirus matched with the recipient; the conditioning regimen would include FCA and low-dose TBI or FCC.

Unrelated cord blood transplants.

Unrelated CB transplants have been reported in 71 patients who received a single or double unrelated CB transplant,43  with 45% survival for patients receiving a total nucleated cell dose greater than 3.9 × 107/kg. The conditioning regimen for CB transplants is identical to the regimen proposed for UD transplants, although methotrexate is omitted from GVHD prophylaxis of CB grafts; 1 dose rituximab 150 mg is recommended on day +5 to prevent EBV lymphoproliferative disorders.43  Patients who do not have a suitable UD could be eligible for a CB graft if the nucleated cell dose is adequate (Figure 2).

Haploidentical transplants.

Young patients who do not have a matched UD or a suitable CB unit and who have failed at least 1 course of IST or have rejected a previous UD or CB graft are eligible for a transplant from HLA haploidentical family donors (HAPLO). Small series of patients have been reported with different conditioning regimens, many using high-dose posttransplant CY.44-49  A total of 84 patients were reported in these studies, and the average 1-year survival was 74%, which is very encouraging. Nevertheless, HAPLO grafts are still in the experimental stage and should be considered only after having failed at least 1 course of IST in the absence of a suitable UD (Figure 2, identified as Alt Donor Tx).

EBV lymphoproliferative disorders.

Patients with SAA undergoing alternative donor transplants, especially with an ATG-based regimen, are at high risk of developing EBV-related lymphoproliferative disorders. We have been using a small dose of rituximab (200 mg, fixed dose) on day +5 after transplantation50  for all patients receiving an alternative donor graft and ATG in the conditioning regimen, and this has eliminated the issue of EBV-related lymphoproliferative disorders.50 

Rejection and autologous recovery.

Rejection is seen in 5% to 15% of patients with SAA undergoing a BMT and calls for immediate action with the attempt of rescuing the patient with a second transplant. The same donor can be used, usually collecting granulocyte colony-stimulating factor (G-CSF)-mobilized PB cells, but haploidentical grafts have also been shown to be effective in a proportion of patients.44-49  Measures to completely prevent rejection in SAA have not been identified: however, the risk will be reduced with the use of ATG, low-dose radiation, and FLU for UD grafts and a marrow cell dose greater than 2 × 108/kg. Some patients rejecting their graft can undergo autologous hematologic recovery51 ; this is a rare event, with an overall rate of 4.2%, but when it occurs, survival is excellent (84%).

ATG+CsA

The standard regimen for first-line IST remains ATG and CsA, with hematological recovery in 50% to 70% of cases and excellent long-term survival among responders.15,52-58  First-line therapy is recommended for all patients without a matched sibling donor (Figure 2), but also for patients with a matched sibling who are older than 40 years.26  There is a strong age effect also after IST, as shown in Figure 5, with survival of 82% and 58% for patients younger than 20 or older than 40 years (A.B., EBMT database, unpublished data); this figure outlines survival after first-line IST and includes salvage therapy with a stem cell transplant for nonresponders. Failure-free or transplant-free survival would be clearly inferior16  and would favor transplantation, but only in younger patients and only when compared with transplants from matched sibling donors.11  For this reason, current guidelines recommend IST first-line therapy for all patients without a matched sibling donor.26  After failing a first course on IST, patients with a matched UD should undergo a transplant if clinically fit; the choice of a transplant vs a second course of IST will need to be patient-oriented.

Figure 5.

The age effect in patients receiving first-line IST. Data from the EBMT registry.

Figure 5.

The age effect in patients receiving first-line IST. Data from the EBMT registry.

Close modal

Duration of CsA therapy

CsA should be given for a minimum of 6 months, but I tend to prolong CsA for more than a year, although at low doses. Once a response has been obtained, discontinuation should be very, very slow, with frequent evaluation of peripheral blood counts to identify early signs of relapse.59  I tend to reduce CsA until I give it 2 to 3 times/week, rather than daily: We are treating an autoimmune disorder, and these patients are really never cured of the disease.

Source of ATG

Horse ATG (ATGAM; Pfizer) has been compared with rabbit ATG (rATG) (Thymoglobulin; Sanofi France) in a prospective randomized trial and has been shown to be superior in terms of response at 6 months (68% vs 37%) and 3-year survival (96% vs 76%).60  Similar results have been obtained in a matched-pair analysis of the EBMT61  and in a retrospective pediatric Japanese study.62  One hypothesis to explain this difference has been a stronger immunosuppressive effect of rATG, leading to more early infections in patients receiving this product.60-62  Other studies, although not prospective, have failed to show significant differences between horse ATG and rATG.57,63,64  A large study on more than 800 patients receiving first-line therapy with rATG (Thymoglobulin) is currently being completed.

G-CSF

The combination of ATG+CsA with G-CSF has shown very good response rates and survival.65  However, 3 randomized studies comparing ATG+CsA with or without G-CSF have failed to show a survival advantage, for patients receiving G-CSF.66-68  In the most recent study,68  the neutrophil count on day +30 predicted response and survival in G-CSF patients. This is in keeping with our initial study,65  suggesting ATG+CsA+ G-CSF identifies early nonresponders, and perhaps may indicate an urgent transplant approach. The 3 randomized studies also confirmed that there was no increased incidence of malignant clonal disorders for G-CSF patients, disproving the reported increased risk for patients receiving prolonged G-CSF treatment shown in retrospective studies.69  I personally believe ATG+CsA+G-CSF remains an interesting first-line therapy because it allows for a rapid neutrophil recovery and identifies early nonresponders. There is no place for G-CSF as single-drug therapy.

Eltrombopag

The new player in the field of nontransplant therapy for SAA is EPAG: After initial encouraging results in refractory patients,70  EPAG has been used together with ATG +CsA as initial treatment in 88 patients.71  The overall response at 6 months was 85%, with survival on the order of 90%. Cytogenetic abnormalities and clonal evolution to myelodysplasia occurred at a similar frequency compared with standard IST, although follow-up is still short.71  The EBMT Working Party for Severe Aplastic Anemia is running 2 prospective randomized studies: 1 compares ATG+CsA with or without EPAG in SAA, and the other compares CsA with or without EPAG in non-SAA.

Case report

A 62-year-old man presented in October 2012 with severe pancytopenia (hemoglobin, 8.3 g/dL after transfusions; white blood cells, 0.8 × 109/L; PMNs, 0.3 × 109/L; platelets, 8 × 109/L), which had gradually developed over the course of a year.

The patient’s medical history revealed a liver transplant at the age of 50 for HCV+ HBV+ cirrhosis, and treatment with CsA since then. In 2011, with declining peripheral blood counts, a BM aspirate showed an empty marrow with trisomy 8 and −Y. The patient was diagnosed with aplastic anemia and received a first course of horse ATG in December 2011. Because of a lack of response, the patient received a second course of ATG (rATG) in July 2012. Four months later, a BM aspirate was unsuccessful, and a marrow biopsy showed complete aplasia. The patient continued to be cytopenic and dependent on red blood cells and platelet transfusions. An unrelated transplant was ruled out because of age and the lack of an 8/8 matched donor. The patient was continued on CsA; testosterone 40 mg by mouth was added 3 times a week, together with EPAG 50 mg every other day. The dose was kept low because of potential liver toxicity, and G-CSF was also given 3 times a week. PB counts improved gradually, and the patient was transfusion-independent after 3 months. In 2013, his counts were as follows: hemoglobin, 11.3 g/dL; white blood cells, 4.8 × 1099/L; PMNs, 3.7 × 109/L; platelets, 71 × 109/L. The patient was started on sofosbuvir and simeprevir for his HCV, and CsA was changed to tacrolimus. He was able to complete his anti-HCV therapy and is now 5 years post-ATG, HCV-negative, receiving low-dose tacrolimus, EPAG 25 mg thrice weekly, low-dose testosterone, and deferoxamine for iron overload, with ferritin reduced from 13.000 to 409 ng/mL. He has developed a small PNH clone (16% CD14, 3% CD16, 0.4% CD59).

Androgens

This patient is an example of an older adult with severe marrow failure, cytogenetic abnormalities, and comorbidities in whom combined treatment with growth factors, CsA, and androgens was successful. I often use this combination in frail patients. Androgens have shown encouraging results both in the animal model72  and in patients with marrow failure.73  A first randomized trial comparing ATG with or without androgens failed to show improved response and survival in the ATG+androgens group.74  However, in a second randomized trial, women receiving ATG+androgens had significantly superior response rates, although comparable survival, as with ATG without androgens.75  Intensified IST with ATG and androgens produced a 77% response rate and 78% survival at 5 years in a single-center study.76  We have shown a 59% response rate with testosterone 40 mg /day (5 days/week) in patients with persistent cytopenia after IST.77 

Recently, sex hormones have been shown to induce telomere elongation in vitro,78  and also in vivo.79  Taken altogether, these studies point to a significant effect of androgens in patients with marrow failure, including increased telomerase activity. Perhaps we should reconsider their role in nontransplant strategies with ATG, CsA, and EPAG.

High-dose cyclophosphamide

The Baltimore group has reported encouraging results with the use of high-dose CY instead of ATG+CsA in 67 patients, with a response rate of 77% and a 10-year survival of 88%80 ; nevertheless, a prospective study has failed to show an advantage of CY over ATG and has highlighted an increased risk for infections.81  For these reasons, high-dose CY cannot be recommended as a nontransplant therapy outside a clinical trial.

Problems with IST

Patients treated with IST are not cured of their disease and are at risk for 3 major complications: no response, relapse, and clonal evolution. Patients not responding to a first course of ATG should be considered for an UD transplant if younger than 40 years (Figure 2); above the age of 40 years, a transplant from an unrelated or alternative donor is one possibility, but a second course of IST, or the addition of EPAG, are valid alternative options (Figure 2), and the choice should be made on the basis of the age of the patients, the clinical conditions, and the severity of the disease. Relapse is the second problem: it occurs in approximately 30% of responders, but can be successfully treated in the majority of patients with a second course of ATG or transplantation if a suitable donor is available. Clonal evolution is the third problem1-3 : the emergence, or existence from diagnosis, of a small GPI negative clone, as determined by flow cytometry, may be involved in the pathogenesis82  and may be a favorable prognostic indicator of response to IST83,84 ; a large GPI negative erythrocyte clone may lead to hemolysis and clinical PNH.85  The most worrisome problem is cytogenetic evolution or a myelodysplastic syndrome, which has been reported to occur in 18% of patients86  and calls for a transplant strategy for eligible patents.87  For this reason, morphologic and cytogenetic examination of marrow cells should be performed in patients with SAA after immunosuppressive treatment, possibly at yearly intervals.15,26 

Treatment of patient older than 60 years

Transplantation is rarely offered to patients older than 60 years because of high transplant-related mortality. Patients between 60 and 70 years of age should be treated with conventional ATG+CsA, and I also use G-CSF because of faster neutrophil recovery. In a study looking at elderly patients, the EBMT has shown that response to IST also can be achieved in older patients.88  Above the age of 70 or 80 years, patients can be rather frail, and I often use CsA plus androgens and reserve CsA+ATG for fit patients, as suggested also by Scheinberg and Young15 .

Acquired aplastic anemia remains a difficult disease, with problems of diagnosis and treatment, and patients should be treated, preferentially, in experienced centers, and best in the context of clinical trials. Significant progress has been made in understanding the pathogenesis of the disease, and new treatment options are now available. Improved outcome of UD transplants has produced survival comparable to that of matched sibling donor grafts, although with increased risk for GVHD. Haploidentical transplants should be considered experimental, and are being used in severe cases lacking a suitable matched donor or CB unit; early results seem promising. EPAG could become a relevant actor in the field of nontransplant therapy and may change our treatment strategies if response and survival rates, recently reported, are confirmed. I often use androgens in patients who fail to achieve hematopoietic recovery after IST, and have seen a significant number of responses. In the seventies, the mortality of acquired SAA was on the order of 80% to 90%: we are currently looking at 80% to 90% survival, which is a fantastic result of worldwide research in this field.

This work was supported in part by Fondazione Ricerca Trapianto Midollo Osseo, Genoa, Italy.

Contribution: A.B. wrote this review.

Conflict-of-interest disclosure: A.B. reports being on the speakers bureau for Sanofi, Pierre Fabre, Therakos, Miltenyi, ADIENNE, Gilead, and Novartis.

Correspondence: Andrea Bacigalupo, Istituto di Ematologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Gemelli, Largo Agostino Gemelli 1, Rome, Italy; e-mail: andrea.bacigalupo@unicatt.it.

1.
Young
NS
,
Calado
RT
,
Scheinberg
P
.
Current concepts in the pathophysiology and treatment of aplastic anemia
.
Blood
.
2006
;
108
(
8
):
2509
-
2519
.
2.
de Planque
MM
,
Bacigalupo
A
,
Würsch
A
, et al
;
Severe Aplastic Anaemia Working Party of the European Cooperative Group for Bone Marrow Transplantation (EBMT)
.
Long-term follow-up of severe aplastic anaemia patients treated with antithymocyte globulin
.
Br J Haematol
.
1989
;
73
(
1
):
121
-
126
.
3.
Moore
MA
,
Castro-Malaspina
H
.
Immunosuppression in aplastic anemia--postponing the inevitable?
N Engl J Med
.
1991
;
324
(
19
):
1358
-
1360
.
4.
Townsley
DM
,
Dumitriu
B
,
Young
NS
.
Bone marrow failure and the telomeropathies
.
Blood
.
2014
;
124
(
18
):
2775
-
2783
.
5.
Lane
AA
,
Odejide
O
,
Kopp
N
, et al
.
Low frequency clonal mutations recoverable by deep sequencing in patients with aplastic anemia
.
Leukemia
.
2013
;
27
(
4
):
968
-
971
.
6.
Yoshizato
T
,
Dumitriu
B
,
Hosokawa
K
, et al
.
Somatic mutations and clonal hematopoiesis in aplastic anemia
.
N Engl J Med
.
2015
;
373
(
1
):
35
-
47
.
7.
Kulasekararaj
AG
,
Jiang
J
,
Smith
AE
, et al
.
Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome
.
Blood
.
2014
;
124
(
17
):
2698
-
2704
.
8.
Camitta
BM
,
Rappeport
JM
,
Parkman
R
,
Nathan
DG
.
Selection of patients for bone marrow transplantation in severe aplastic anemia
.
Blood
.
1975
;
45
(
3
):
355
-
363
.
9.
Maciejewski
JP
,
Risitano
A
,
Sloand
EM
,
Nunez
O
,
Young
NS
.
Distinct clinical outcomes for cytogenetic abnormalities evolving from aplastic anemia
.
Blood
.
2002
;
99
(
9
):
3129
-
3135
.
10.
Bacigalupo
A
,
Hows
J
,
Gluckman
E
, et al
.
Bone marrow transplantation (BMT) versus immunosuppression for the treatment of severe aplastic anaemia (SAA): a report of the EBMT SAA working party
.
Br J Haematol
.
1988
;
70
(
2
):
177
-
182
.
11.
Bacigalupo
A
,
Brand
R
,
Oneto
R
, et al
.
Treatment of acquired severe aplastic anemia: bone marrow transplantation compared with immunosuppressive therapy--The European Group for Blood and Marrow Transplantation experience
.
Semin Hematol
.
2000
;
37
(
1
):
69
-
80
.
12.
Locasciulli
A
,
Oneto
R
,
Bacigalupo
A
, et al
;
Severe Aplastic Anemia Working Party of the European Blood and Marrow Transplant Group
.
Outcome of patients with acquired aplastic anemia given first line bone marrow transplantation or immunosuppressive treatment in the last decade: a report from the European Group for Blood and Marrow Transplantation (EBMT)
.
Haematologica
.
2007
;
92
(
1
):
11
-
18
.
13.
Höchsmann
B
,
Moicean
A
,
Risitano
A
,
Ljungman
P
,
Schrezenmeier
H
.
Supportive care in severe and very severe aplastic anemia
.
Bone Marrow Transplant
.
2013
;
48
(
2
):
168
-
173
.
14.
Locasciulli
A
,
Bacigalupo
A
,
Bruno
B
, et al
;
Severe Aplastic Anemia Working Party of the European Blood and Marrow Transplant Group (SAA-WP, EBMT)
.
Hepatitis-associated aplastic anaemia: epidemiology and treatment results obtained in Europe. A report of The EBMT aplastic anaemia working party
.
Br J Haematol
.
2010
;
149
(
6
):
890
-
895
.
15.
Scheinberg
P
,
Young
NS
.
How I treat acquired aplastic anemia
.
Blood
.
2012
;
120
(
6
):
1185
-
1196
.
16.
Yoshida
N
,
Kobayashi
R
,
Yabe
H
, et al
.
First-line treatment for severe aplastic anemia in children: bone marrow transplantation from a matched family donor versus immunosuppressive therapy
.
Haematologica
.
2014
;
99
(
12
):
1784
-
1791
.
17.
Gupta
V
,
Eapen
M
,
Brazauskas
R
, et al
.
Impact of age on outcomes after bone marrow transplantation for acquired aplastic anemia using HLA-matched sibling donors
.
Haematologica
.
2010
;
95
(
12
):
2119
-
2125
.
18.
Storb
R
,
Etzioni
R
,
Anasetti
C
, et al
.
Cyclophosphamide combined with antithymocyte globulin in preparation for allogeneic marrow transplants in patients with aplastic anemia
.
Blood
.
1994
;
84
(
3
):
941
-
949
.
19.
Champlin
RE
,
Perez
WS
,
Passweg
JR
, et al
.
Bone marrow transplantation for severe aplastic anemia: a randomized controlled study of conditioning regimens
.
Blood
.
2007
;
109
(
10
):
4582
-
4585
.
20.
Bacigalupo
A
,
Socié
G
,
Schrezenmeier
H
, et al
;
Aplastic Anemia Working Party of the European Group for Blood and Marrow Transplantation (WPSAA-EBMT)
.
Bone marrow versus peripheral blood as the stem cell source for sibling transplants in acquired aplastic anemia: survival advantage for bone marrow in all age groups
.
Haematologica
.
2012
;
97
(
8
):
1142
-
1148
.
21.
Srinivasan
R
,
Takahashi
Y
,
McCoy
JP
, et al
.
Overcoming graft rejection in heavily transfused and allo-immunised patients with bone marrow failure syndromes using fludarabine-based haematopoietic cell transplantation
.
Br J Haematol
.
2006
;
133
(
3
):
305
-
314
.
22.
George
B
,
Mathews
V
,
Shaji
RV
,
Srivastava
V
,
Srivastava
A
,
Chandy
M
.
Fludarabine-based conditioning for allogeneic stem cell transplantation for multiply transfused patients with Fanconi’s anemia
.
Bone Marrow Transplant
.
2005
;
35
(
4
):
341
-
343
.
23.
Resnick
IB
,
Aker
M
,
Shapira
MY
, et al
.
Allogeneic stem cell transplantation for severe acquired aplastic anaemia using a fludarabine-based preparative regimen
.
Br J Haematol
.
2006
;
133
(
6
):
649
-
654
.
24.
Maury
S
,
Bacigalupo
A
,
Anderlini
P
, et al
;
Severe Aplastic Anemia Working Party, European Group for Blood and Marrow Transplantation (EBMT-SAAWP)
.
Improved outcome of patients older than 30 years receiving HLA-identical sibling hematopoietic stem cell transplantation for severe acquired aplastic anemia using fludarabine-based conditioning: a comparison with conventional conditioning regimen
.
Haematologica
.
2009
;
94
(
9
):
1312
-
1315
.
25.
Aljurf
M
,
Al-Zahrani
H
,
Van Lint
MT
,
Passweg
JR
.
Standard treatment of acquired SAA in adult patients 18-40 years old with an HLA-identical sibling donor
.
Bone Marrow Transplant
.
2013
;
48
(
2
):
178
-
179
.
26.
Killick
SB
,
Bown
N
,
Cavenagh
J
, et al
;
British Society for Standards in Haematology
.
Guidelines for the diagnosis and management of adult aplastic anaemia
.
Br J Haematol
.
2016
;
172
(
2
):
187
-
207
.
27.
Schrezenmeier
H
,
Passweg
JR
,
Marsh
JC
, et al
.
Worse outcome and more chronic GVHD with peripheral blood progenitor cells than bone marrow in HLA-matched sibling donor transplants for young patients with severe acquired aplastic anemia
.
Blood
.
2007
;
110
(
4
):
1397
-
1400
.
28.
Dufour
C
,
Veys
P
,
Carraro
E
, et al
.
Similar outcome of upfront-unrelated and matched sibling stem cell transplantation in idiopathic paediatric aplastic anaemia. A study on behalf of the UK Paediatric BMT Working Party, Paediatric Diseases Working Party and Severe Aplastic Anaemia Working Party of EBMT
.
Br J Haematol
.
2015
;
171
(
4
):
585
-
594
.
29.
Kojima
S
,
Matsuyama
T
,
Kato
S
, et al
.
Outcome of 154 patients with severe aplastic anemia who received transplants from unrelated donors: the Japan Marrow Donor Program
.
Blood
.
2002
;
100
(
3
):
799
-
803
.
30.
Devillier
R
,
Dalle
JH
,
Kulasekararaj
A
et al
.
Unrelated alternative donor transplantation for severe acquired aplastic anemia: a study from the French Society of Bone Marrow Transplantation and Cell Therapies and the Severe Aplastic Anemia Working Party of EBMT
.
Haematologica. 2016;101(7):884-890
.
31.
Horan
J
,
Wang
T
,
Haagenson
M
, et al
.
Evaluation of HLA matching in unrelated hematopoietic stem cell transplantation for nonmalignant disorders
.
Blood
.
2012
;
120
(
14
):
2918
-
2924
.
32.
Deeg
HJ
,
O’Donnell
M
,
Tolar
J
, et al
.
Optimization of conditioning for marrow transplantation from unrelated donors for patients with aplastic anemia after failure of immunosuppressive therapy
.
Blood
.
2006
;
108
(
5
):
1485
-
1491
.
33.
Ljungman
P
,
Brand
R
,
Hoek
J
, et al
;
Infectious Diseases Working Party of the European Group for Blood and Marrow Transplantation
.
Donor cytomegalovirus status influences the outcome of allogeneic stem cell transplant: a study by the European group for blood and marrow transplantation
.
Clin Infect Dis
.
2014
;
59
(
4
):
473
-
481
.
34.
Bacigalupo
A
,
Socié
G
,
Hamladji
RM
, et al
;
Aplastic Anemia Working Party of the European Group for Blood Marrow Transplantation
.
Current outcome of HLA identical sibling versus unrelated donor transplants in severe aplastic anemia: an EBMT analysis
.
Haematologica
.
2015
;
100
(
5
):
696
-
702
.
35.
Eapen
M
,
Le Rademacher
J
,
Antin
JH
, et al
.
Effect of stem cell source on outcomes after unrelated donor transplantation in severe aplastic anemia
.
Blood
.
2011
;
118
(
9
):
2618
-
2621
.
36.
Marsh
JC
,
Gupta
V
,
Lim
Z
, et al
.
Alemtuzumab with fludarabine and cyclophosphamide reduces chronic graft-versus-host disease after allogeneic stem cell transplantation for acquired aplastic anemia
.
Blood
.
2011
;
118
(
8
):
2351
-
2357
.
37.
Bacigalupo
A
,
Socie’
G
,
Lanino
E
, et al
;
Severe Aplastic Anemia Working Party of the European Group for Blood and Marrow Transplantation
.
Fludarabine, cyclophosphamide, antithymocyte globulin, with or without low dose total body irradiation, for alternative donor transplants, in acquired severe aplastic anemia: a retrospective study from the EBMT-SAA Working Party
.
Haematologica
.
2010
;
95
(
6
):
976
-
982
.
38.
Okuda
S
,
Terasako
K
,
Oshima
K
, et al
.
Fludarabine, cyclophosphamide, anti-thymocyteglobulin, and low-dose total body irradiation conditioning enables 1-HLA-locus-mismatched hematopoietic stem cell transplantation for very severe aplastic anemia without affecting ovarian function
.
Am J Hematol
.
2009
;
84
(
3
):
167
-
169
.
39.
Koh
LP
,
Koh
MB
,
Ng
HY
, et al
.
Allogeneic hematopoietic stem cell transplantation for patients with severe aplastic anemia following nonmyeloablative conditioning using 200-cGy total body irradiation and fludarabine
.
Biol Blood Marrow Transplant
.
2006
;
12
(
8
):
887
-
890
.
40.
Deeg
HJ
,
Amylon
ID
,
Harris
RE
, et al
.
Marrow transplants from unrelated donors for patients with aplastic anemia: minimum effective dose of total body irradiation
.
Biol Blood Marrow Transplant
.
2001
;
7
(
4
):
208
-
215
.
41.
Anderlini
P
,
Wu
J
,
Gersten
I
, et al
.
Cyclophosphamide conditioning in patients with severe aplastic anaemia given unrelated marrow transplantation: a phase 1-2 dose de-escalation study
.
Lancet Haematol
.
2015
;
2
(
9
):
e367
-
e375
.
42.
Arai
Y
,
Kondo
T
,
Yamazaki
H
, et al
;
Japan Society for Hematopoietic Cell Transplantation
.
Allogeneic unrelated bone marrow transplantation from older donors results in worse prognosis in recipients with aplastic anemia
.
Haematologica
.
2016
;
101
(
5
):
644
-
652
.
43.
Peffault de Latour
R
,
Purtill
D
,
Ruggeri
A
, et al
.
Influence of nucleated cell dose on overall survival of unrelated cord blood transplantation for patients with severe acquired aplastic anemia: a study by eurocord and the aplastic anemia working party of the European group for blood and marrow transplantation
.
Biol Blood Marrow Transplant
.
2011
;
17
(
1
):
78
-
85
.
44.
Wu
Y
,
Cao
Y
,
Li
X
, et al
.
Cotransplantation of haploidentical hematopoietic and umbilical cord mesenchymal stem cells for severe aplastic anemia: successful engraftment and mild GVHD
.
Stem Cell Res (Amst)
.
2014
;
12
(
1
):
132
-
138
.
45.
Esteves
I
,
Bonfim
C
,
Pasquini
R
, et al
.
Haploidentical BMT and post-transplant Cy for severe aplastic anemia: a multicenter retrospective study
.
Bone Marrow Transplant
.
2015
;
50
(
5
):
685
-
689
.
46.
Clay
J
,
Kulasekararaj
AG
,
Potter
V
, et al
.
Nonmyeloablative peripheral blood haploidentical stem cell transplantation for refractory severe aplastic anemia
.
Biol Blood Marrow Transplant
.
2014
;
20
(
11
):
1711
-
1716
.
47.
Li
XH
,
Gao
CJ
,
Da
WM
, et al
.
Reduced intensity conditioning, combined transplantation of haploidentical hematopoietic stem cells and mesenchymal stem cells in patients with severe aplastic anemia
.
PLoS One
.
2014
;
9
(
3
):
e89666
.
48.
Gao
L
,
Li
Y
,
Zhang
Y
, et al
.
Long-term outcome of HLA-haploidentical hematopoietic SCT without in vitro T-cell depletion for adult severe aplastic anemia after modified conditioning and supportive therapy
.
Bone Marrow Transplant
.
2014
;
49
(
4
):
519
-
524
.
49.
Dezern
AE
,
Luznik
L
,
Fuchs
EJ
,
Jones
RJ
,
Brodsky
RA
.
Post-transplantation cyclophosphamide for GVHD prophylaxis in severe aplastic anemia
.
Bone Marrow Transplant
.
2011
;
46
(
7
):
1012
-
1013
.
50.
Dominietto
A
,
Tedone
E
,
Soracco
M
, et al
.
In vivo B-cell depletion with rituximab for alternative donor hemopoietic SCT
.
Bone Marrow Transplant
.
2012
;
47
(
1
):
101
-
106
.
51.
Piccin
A
,
McCann
S
,
Socié
G
, et al
;
Aplastic Anaemia Working Party of the European Group for Blood and Marrow Transplantation
.
Survival of patients with documented autologous recovery after SCT for severe aplastic anemia: a study by the WPSAA of the EBMT
.
Bone Marrow Transplant
.
2010
;
45
(
6
):
1008
-
1013
.
52.
Frickhofen
N
,
Kaltwasser
JP
,
Schrezenmeier
H
, et al
;
The German Aplastic Anemia Study Group
.
Treatment of aplastic anemia with antilymphocyte globulin and methylprednisolone with or without cyclosporine
.
N Engl J Med
.
1991
;
324
(
19
):
1297
-
1304
.
53.
Rosenfeld
SJ
,
Kimball
J
,
Vining
D
,
Young
NS
.
Intensive immunosuppression with antithymocyte globulin and cyclosporine as treatment for severe acquired aplastic anemia
.
Blood
.
1995
;
85
(
11
):
3058
-
3065
.
54.
Führer
M
,
Burdach
S
,
Ebell
W
, et al
;
German/Austrian Pediatric Aplastic Anemia Working Group
.
Relapse and clonal disease in children with aplastic anemia (AA) after immunosuppressive therapy (IST): the SAA 94 experience
.
Klin Padiatr
.
1998
;
210
(
4
):
173
-
179
.
55.
Kojima
S
,
Hibi
S
,
Kosaka
Y
, et al
.
Immunosuppressive therapy using antithymocyte globulin, cyclosporine, and danazol with or without human granulocyte colony-stimulating factor in children with acquired aplastic anemia
.
Blood
.
2000
;
96
(
6
):
2049
-
2054
.
56.
Afable
MG
2nd
,
Shaik
M
,
Sugimoto
Y
, et al
.
Efficacy of rabbit anti-thymocyte globulin in severe aplastic anemia
.
Haematologica
.
2011
;
96
(
9
):
1269
-
1275
.
57.
Vallejo
C
,
Montesinos
P
,
Polo
M
, et al
;
Bone Marrow Failure Spanish Study Group (Pethema-GETH)
.
Rabbit antithymocyte globulin versus horse antithymocyte globulin for treatment of acquired aplastic anemia: a retrospective analysis
.
Ann Hematol
.
2015
;
94
(
6
):
947
-
954
.
58.
Deyell
RJ
,
Shereck
EB
,
Milner
RA
,
Schultz
KR
.
Immunosuppressive therapy without hematopoietic growth factor exposure in pediatric acquired aplastic anemia
.
Pediatr Hematol Oncol
.
2011
;
28
(
6
):
469
-
478
.
59.
Saracco
P
,
Quarello
P
,
Iori
AP
, et al
;
Bone Marrow Failure Study Group of the AIEOP (Italian Association of Paediatric Haematology Oncology)
.
Cyclosporin A response and dependence in children with acquired aplastic anaemia: a multicentre retrospective study with long-term observation follow-up
.
Br J Haematol
.
2008
;
140
(
2
):
197
-
205
.
60.
Scheinberg
P
,
Nunez
O
,
Weinstein
B
, et al
.
Horse versus rabbit antithymocyte globulin in acquired aplastic anemia
.
N Engl J Med
.
2011
;
365
(
5
):
430
-
438
.
61.
Marsh
JC
,
Bacigalupo
A
,
Schrezenmeier
H
, et al
;
European Blood and Marrow Transplant Group Severe Aplastic Anaemia Working Party
.
Prospective study of rabbit antithymocyte globulin and cyclosporine for aplastic anemia from the EBMT Severe Aplastic Anaemia Working Party
.
Blood
.
2012
;
119
(
23
):
5391
-
5396
.
62.
Jeong
DC
,
Chung
NG
,
Cho
B
, et al
.
Long-term outcome after immunosuppressive therapy with horse or rabbit antithymocyte globulin and cyclosporine for severe aplastic anemia in children
.
Haematologica
.
2014
;
99
(
4
):
664
-
671
.
63.
Chang
MH
,
Kim
KH
,
Kim
HS
, et al
.
Predictors of response to immunosuppressive therapy with antithymocyte globulin and cyclosporine and prognostic factors for survival in patients with severe aplastic anemia
.
Eur J Haematol
.
2010
;
84
(
2
):
154
-
159
.
64.
Chen
C
,
Xue
HM
,
Xu
HG
, et al
.
Rabbit-antithymocyte globulin combined with cyclosporin A as a first-line therapy: improved, effective, and safe for children with acquired severe aplastic anemia
.
J Cancer Res Clin Oncol
.
2012
;
138
(
7
):
1105
-
1111
.
65.
Bacigalupo
A
,
Bruno
B
,
Saracco
P
, et al
;
European Group for Blood and Marrow Transplantation (EBMT) Working Party on Severe Aplastic Anemia and the Gruppo Italiano Trapianti di Midolio Osseo (GITMO)
.
Antilymphocyte globulin, cyclosporine, prednisolone, and granulocyte colony-stimulating factor for severe aplastic anemia: an update of the GITMO/EBMT study on 100 patients
.
Blood
.
2000
;
95
(
6
):
1931
-
1934
.
66.
Gluckman
E
,
Rokicka-Milewska
R
,
Hann
I
, et al
;
European Group for Blood and Marrow Transplantation Working Party for Severe Aplastic Anemia
.
Results and follow-up of a phase III randomized study of recombinant human-granulocyte stimulating factor as support for immunosuppressive therapy in patients with severe aplastic anaemia
.
Br J Haematol
.
2002
;
119
(
4
):
1075
-
1082
.
67.
Teramura
M
,
Kimura
A
,
Iwase
S
, et al
.
Treatment of severe aplastic anemia with antithymocyte globulin and cyclosporin A with or without G-CSF in adults: a multicenter randomized study in Japan
.
Blood
.
2007
;
110
(
6
):
1756
-
1761
.
68.
Tichelli
A
,
Schrezenmeier
H
,
Socié
G
, et al
.
A randomized controlled study in patients with newly diagnosed severe aplastic anemia receiving antithymocyte globulin (ATG), cyclosporine, with or without G-CSF: a study of the SAA Working Party of the European Group for Blood and Marrow Transplantation
.
Blood
.
2011
;
117
(
17
):
4434
-
4441
.
69.
Kaito
K
,
Kobayashi
M
,
Katayama
T
, et al
.
Long-term administration of G-CSF for aplastic anaemia is closely related to the early evolution of monosomy 7 MDS in adults
.
Br J Haematol
.
1998
;
103
(
2
):
297
-
303
.
70.
Desmond
R
,
Townsley
DM
,
Dumitriu
B
, et al
.
Eltrombopag restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on discontinuation of drug
.
Blood
.
2014
;
123
(
12
):
1818
-
1825
.
71.
Townsley
DM
,
Dumitriu
B
,
Scheinberg
P
, et al
.
Eltrombopag added to standard immunosuppression for aplastic anemia accelerates count recovery and increases response rates [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract LBA-2
.
72.
Najean
Y
,
Chedeville
A
,
Eberlin
A
,
Balitrand
N
.
A study of the efficacy of 5 alpha- and 5 beta-androstanes in chronic experimental aplastic anemia in mice
.
Nouv Rev Fr Hematol
.
1984
;
26
(
6
):
391
-
396
.
73.
Najean
Y
,
Haguenauer
O
;
The Cooperative Group for the Study of Aplastic and Refractory Anemias
.
Long-term (5 to 20 years) Evolution of nongrafted aplastic anemias
.
Blood
.
1990
;
76
(
11
):
2222
-
2228
.
74.
Champlin
RE
,
Ho
WG
,
Feig
SA
,
Winston
DJ
,
Lenarsky
C
,
Gale
RP
.
Do androgens enhance the response to antithymocyte globulin in patients with aplastic anemia? A prospective randomized trial
.
Blood
.
1985
;
66
(
1
):
184
-
188
.
75.
Bacigalupo
A
,
Chaple
M
,
Hows
J
, et al
.
Treatment of aplastic anaemia (AA) with antilymphocyte globulin (ALG) and methylprednisolone (MPred) with or without androgens: a randomized trial from the EBMT SAA working party
.
Br J Haematol
.
1993
;
83
(
1
):
145
-
151
.
76.
Leleu
X
,
Terriou
L
,
Duhamel
A
, et al
.
Long-term outcome in acquired aplastic anemia treated with an intensified dose schedule of horse antilymphocyte globulin in combination with androgens
.
Ann Hematol
.
2006
;
85
(
10
):
711
-
716
.
77.
Giammarco
S
,
Van Lint
MT
,
Lamparelli
T
, et al
.
Androgens may boost responses to anti-thymocyte globulin in acquired aplastic anemia [abstract]
.
Blood
.
2016
;
128
(
22
).
Abstract 3900
.
78.
Calado
RT
,
Yewdell
WT
,
Wilkerson
KL
, et al
.
Sex hormones, acting on the TERT gene, increase telomerase activity in human primary hematopoietic cells
.
Blood
.
2009
;
114
(
11
):
2236
-
2243
.
79.
Townsley
DM
,
Dumitriu
B
,
Liu
D
, et al
.
Danazol treatment for telomere diseases
.
N Engl J Med
.
2016
;
374
(
20
):
1922
-
1931
.
80.
Brodsky
RA
,
Chen
AR
,
Dorr
D
, et al
.
High-dose cyclophosphamide for severe aplastic anemia: long-term follow-up
.
Blood
.
2010
;
115
(
11
):
2136
-
2141
.
81.
Scheinberg
P
,
Townsley
D
,
Dumitriu
B
, et al
.
Moderate-dose cyclophosphamide for severe aplastic anemia has significant toxicity and does not prevent relapse and clonal evolution
.
Blood
.
2014
;
124
(
18
):
2820
-
2823
.
82.
Boccuni
P
,
Del Vecchio
L
,
Di Noto
R
,
Rotoli
B
.
Glycosyl phosphatidylinositol (GPI)-anchored molecules and the pathogenesis of paroxysmal nocturnal hemoglobinuria
.
Crit Rev Oncol Hematol
.
2000
;
33
(
1
):
25
-
43
.
83.
Sugimori
C
,
Chuhjo
T
,
Feng
X
, et al
.
Minor population of CD55-CD59- blood cells predicts response to immunosuppressive therapy and prognosis in patients with aplastic anemia
.
Blood
.
2006
;
107
(
4
):
1308
-
1314
.
84.
Narita
A
,
Muramatsu
H
,
Sekiya
Y
, et al
;
Japan Childhood Aplastic Anemia Study Group
.
Paroxysmal nocturnal hemoglobinuria and telomere length predicts response to immunosuppressive therapy in pediatric aplastic anemia
.
Haematologica
.
2015
;
100
(
12
):
1546
-
1552
.
85.
Young
NS
,
Abkowitz
JL
,
Luzzatto
L
.
New insights into the pathophysiology of acquired cytopenias
.
Hematology Am Soc Hematol Educ Program
.
2000
;
2000
:
18
-
38
.
86.
Socié
G
,
Henry-Amar
M
,
Bacigalupo
A
, et al
;
European Bone Marrow Transplantation-Severe Aplastic Anaemia Working Party
.
Malignant tumors occurring after treatment of aplastic anemia
.
N Engl J Med
.
1993
;
329
(
16
):
1152
-
1157
.
87.
Kim
SY
,
Le Rademacher
J
,
Antin
JH
, et al
.
Myelodysplastic syndrome evolving from aplastic anemia treated with immunosuppressive therapy: efficacy of hematopoietic stem cell transplantation
.
Haematologica
.
2014
;
99
(
12
):
1868
-
1875
.
88.
Tichelli
A
,
Socié
G
,
Henry-Amar
M
, et al
;
European Group for Blood and Marrow Transplantation Severe Aplastic Anaemia Working Party
.
Effectiveness of immunosuppressive therapy in older patients with aplastic anemia
.
Ann Intern Med
.
1999
;
130
(
3
):
193
-
201
.
Sign in via your Institution