Therapy-Related Myelodysplastic Syndromes


 

 

Therapy-Related Myelodysplastic Syndromes

Therapy-related myelodysplastic syndromes are increasing in frequency as the utilization of intensive chemotherapy and radiation increases in other solid tumors and lymphoma.36–43 These cases have a poor prognosis and are not included in the International Prognostic Scoring System (IPSS). Cellular abnormalities of chromosomes 5, 7, and 8 are common in these cases.317 MDS following breast cancer is associated with older age, presence of other cancers, and multiple first-degree relatives with cancer.318 As compared to patients with myeloma or germ cell tumors, patients with lymphoma undergoing autologous stem cell transplantation have a higher incidence of treatment-related MDS. In this group, pretransplantation therapy, total-body irradiation, and other transplantation-related factors play a role, as do inherited polymorphisms in genes governing drug metabolism and DNA repair.319 Therapy-related MDS has been reported after high-dose melphalan for myeloma treatment, but the risk is relatively low.320 Accelerated telomere shortening precedes development of therapy-related myelodysplasia after autologous transplantation for lymphoma.321 Treatment-related MDS is managed as are de novo cases of MDS, but are very refractory to treatment. Allogeneic hematopoietic stem cell transplantation can result in long-term disease-free survival, but most patients with therapy-related MDS are not candidates because of advanced age, comorbidities, or the inability to control the primary cancer.322

Treatment of Myelodysplastic Syndrome Based on Prognostic ScoreTherapeutic decisions in MDS patients can be based on the category of disease, such as 5q– syndrome or clonal anemia. These approaches are outlined after each category of specific syndrome. Because the syndromes are a continuum and have irregular manifestations, an IPSS was devised to assign patients at the time of diagnosis to a category that estimates the likelihood of early progression, the average time by which patients with those characteristics will evolve to AML, and incorporates information beyond the MDS subtype.277,279,309 Multivariate analysis combines the impact of (1) percentage of marrow blasts, (2) three cytogenetic subgroups (favorable, unfavorable, intermediate), and (3) number of cytopenias (Table 88–3). In large numbers of patients, the following frequency distribution of patients has been observed: low-risk group (i.e., longest time to evolve to AML) in 15 to 30 percent of patients; intermediate-1 risk group in 30 to 40 percent of patients; intermediate-2 risk group in 20 to 25 percent of patients; and high-risk group in 5 to 10 percent of patients.277,279,323

Table 88–3. International Prognostic Scoring System for Myelodysplastic Syndromes277
Prognostic Variable Score Value
0 0.5 1.0 1.5
Marrow Blast (%) <5 5–10 11–20
Karyotype Good Intermediate Poor
Cytopenias 0, 1 2, 3
Risk groups: Low, 0; INT-1, 0.5–1.0; INT-2, 1.5–2.0; High, ≥2.5.Karyotype: Good score, -Y, del(5q); poor score, complex abnormalities and chromosome 7 abnormalities; intermediate score, other abnormalities. See “Marrow: Cytogenetics” above for further details.

Using the IPSS to classify patients, survival is worse as risk category increases in four defined groups from low-risk to high-risk (Table 88–4), and this effect is influenced by age at diagnosis within the same prognostic category (Table 88–5).

Table 88–4. Survival of Patients with Clonal Cytopenias and Oligoblastic Myelogenous Leukemia Based on the International Prognostic Scoring System
IPSS Score at Diagnosis No. of Patients 2-Year Survival 5-Year Survival 10-Year Survival 15-Year Survival
Low 267 85% 55% 28% 20%
Intermediate-1 314 70% 35% 17% 12%
Intermediate-2 179 30% 8% 0
High 56 5% 0
IPSS, International Prognostic Scoring System.These data were extrapolated from curves in Figure 6 of reference 277. Data are expressed as percent of all patients in that risk category surviving at the time interval shown.
Table 88–5. Survival of Patients with Clonal Cytopenias and Oligoblastic Myelogenous Leukemia Based on the International Prognostic Scoring System Stratified by Age
IPSS Score and Age 2-Year Survival 5-Year Survival 10-Year Survival 15-Year Survival
Low ≤60 years 95% 80% 65% 30%
Low >60 years 80% 45% 18% 18%
Intermediate-1 ≤60 years 85% 50% 37% 18%
Intermediate-1 >60 years 62% 30% 12% ND
Intermediate-2 ≤60 years 50% 15% ND
Intermediate-2 >60 years 25% 5% 0
High ≤60 years 0
High >60 years 7% 0
IPSS, International Prognostic Scoring System; ND, no data.These data were extrapolated from curves in Figure 7 of reference 277. Data are expressed as percent of all patients in that risk category surviving at the time interval shown.

The prognostic score should not be the sole guide to treatment because many patients deviate from the average expectation of disease behavior. Unexpected progression may necessitate changes in treatment approach and in the case of patients who are candidates for allogeneic hematopoietic stem cell transplantation; their course may require recommending the procedure. Furthermore, several refinements to the original IPSS score have been proposed to incorporate factors such as ALIP and CD34 expression,324 duration of MDS and prior therapy,325 and lactate dehydrogenase.326,327 Also, other time-dependent prognostic scoring systems have been proposed, including the WHO Prognositc scoring System.328 Although these systems have benefit in defining populations and allowing comparisons between defined groups, they are often of little aid in making treatment decisions in individual patients and in understanding the biology of MDS.329

Treatments based on the IPSS can be considered (1) supportive care, (2) low-intensity therapy, or (3) high-intensity treatment.330–332 Treatment response is judged based on the MDS subtype and IPSS score of the patient and the presence of treatment-induced (secondary) MDS.333

Therapy for Patients with Low and Low-Intermediate (Int-1) Prognostic Scores

Supportive care consists of improving quality of life with specific treatment of cytopenias or their complications and providing psychosocial support, while monitoring the patient’s clinical status at intervals.334,335

Management of Anemia

Red Cell Transfusion

Red cell transfusions should be administered for symptomatic anemia. Often patients will tolerate hemoglobin levels as low as 8 g/dL, but the level at which symptoms develop varies from patient to patient. Higher thresholds have been suggested to prevent cardiac consequences of prolonged anemia.336

Erythropoiesis-Stimulating Agents

Red cell transfusion dependency may have a negative impact on clinical outcomes in MDS probably related to more severe marrow failure in those cases, increased iron overload, and possibly correlation with increased risk of transformation to AML.337 Some studies found that neither the serum ferritin nor the number of red blood cell transfusions impacted survival in clonal sideroblastic anemia.338 Recombinant erythropoietin can be used to treat anemia in patients who are transfusion-dependent, if the serum erythropoietin level is low for the hemoglobin level. Responses are best with low erythropoietin levels, normal blast counts, lower IPSS scores,339 normal cytogenetics,340 and in patients who do not require transfusion.341 Hemolysis, or iron, vitamin B12, or folate deficiency should be ruled out as a cause of anemia before erythropoietin therapy is started. Iron stores should be kept replete during erythropoietin therapy. Erythropoietin 150 to 300 U/kg per day or single weekly doses of 40,000 U are effective.340 Darbepoetin alpha in various schedules of administration has also been found effective in increasing hemoglobin levels and in enhancing quality of life.343,344 The probability of a response increases with duration of therapy; for example, optimal at 26 weeks compared to 12 weeks.345 Meta-analysis has confirmed erythropoietic response rates are similar for those treated either with epoetin alfa or with the longer-acting darbepoetin alfa.346 Unlike the case in patients with solid tumors or renal failure, there is no evidence that erythropoietic agents increase thromboembolic disease or accelerate progress to leukemia, but followup in these studies has been short.347 G-CSF combined with erythropoietin may produce a response more frequently.348,349 This combination does not appear to affect the risk of leukemic transformation and may have a positive impact on survival in those with low transfusion needs.350 This approach is not recommended for those with intermediate-2 risk or high-risk IPSS scores.351 There is evidence that marrow erythroid cells of MDS patients who respond to erythropoietin have a different gene expression pattern than do those of nonresponders.352

Iron-Chelation Therapy

Chelation may be necessary to prevent iron overload in patients receiving frequent transfusions. Numerous consensus guidelines have been published regarding the treatment of iron overload in myelodysplastic syndromes.353–355 These emphasize that there is no prospectively validated threshold for (1) the number of units of transfused blood or (2) the level of serum ferritin that should trigger iron chelation.355 Several of these guidelines use a serum ferritin >1000 mcg/L as a threshold for starting iron-chelation therapy. They also take into account the patient’s candidacy for allogeneic stem cell transplantation, life expectancy, and evidence of iron-related organ damage.355 Both deferoxamine given subcutaneously or intravenously and deferasirox given orally are available for chelation therapy in MDS patients.356 Cardiac magnetic resonance imaging may provide more reliable means of assessing myocardial iron overload than does measurement of serum ferritin.357

Low-Dose Cytarabine

Low-dose cytarabine 5 to 20 mg/m2 per day by subcutaneous injection every 12 hours for up to 8 to 16 weeks or by continuous intravenous infusion has been used in lieu of intensive chemotherapy.358,359 Although this approach led to remission in approximately 20 percent of patients with oligoblastic leukemia, the median duration of remission is approximately 10 months, and survival has not been prolonged compared with supportive care alone. Moreover, low-dose cytosine arabinoside usually is cytotoxic, inducing marrow hypoplasia and worsening cytopenias. Although occasional reports of remission following low-dose cytarabine have been consistent with an effect on leukemia cell maturation, most patients experience suppression of the malignant cell clone, leading to marrow repopulation with polyclonal hemopoiesis.304,309,330 This treatment approach is now utilized less often since the advent of other FDA-approved agents for MDS, but may still have a role in some patients, especially when combined with G-CSF.360

Immunotherapy

Cyclosporine and Antithymocyte Globulin

In some patients with MDS, T-lymphocyte–mediated inhibition of hematopoiesis occurs and contributes to cytopenias. The cytopenias can be ameliorated by treatment with immunosuppressive agents.361 In patients who recovered effective hematopoiesis after treatment, the V (T-cell receptor-) spectra-type representative of clonal or oligoclonal T-cell populations reverted to normal patterns.361 A nonclonal X-chromosome inactivation pattern in the marrow, as assessed by the human androgen receptor gene assay and the phosphoglycerated kinase-1 assay, was associated with a response to ATG. This finding was attributed to incomplete clonal expansion, with ATG improving normal hematopoiesis by relieving the immunologic pressure on the remaining normal progenitors.362 Others have postulated that responses may result from suppression of interferon- secretion by CD4+ T cells.363 Some series have reported response rates to ATG of 15 to 60 percent364–366 and longer survival times in patients who respond. Human leukocyte antigen (HLA)-DR15 (DR2) is overrepresented in MDS and predicts a response to immunosuppressive therapy.367 In one series of 60 patients treated with ATG and cyclosporine, 60 percent had hematologic improvement, and more responders had good karyotype or DRB1 1501.368 Most of the patients in this series had refractory anemia and an IPSS score of intermediate-1. Most, but not all, responses have occurred in patients with hypocellular marrows.369,370 In a series of 129 patients who were treated with immunosuppressive therapy at a single institution, 30 percent had either a complete or partial response, and younger age and intermediate or low IPSS score favored survival.371 Other groups have reported lack of response to ATG and prednisone. One study was stopped early because of lack of efficacy and development of adverse reactions.372 Other studies also have reported lack of efficacy of single-agent cyclosporine.373

Other Treatment Options in Low- or Intermediate-1–Risk Patients

For those patients not likely to respond to supportive care measures alone or for those who are not likely to respond to immune suppressive therapies, azacytidine, decitabine, or lenalidomide can be used. In those not responding to these approved agents, in appropriate patients, allogeneic stem cell transplantation or other investigational options can be considered (see Hematopoietic Stem Cell Transplantation).

Therapy for Patients with High-Intermediate (Int-2) Prognostic Scores

Demethylating Agent and Histone Deacetylation Inhibitor Therapy

Oligoblastic and secondary myelogenous leukemias have a high prevalence of tumor suppressor gene hypermethylation.374 5-Azacytidine is a pyrimidine analogue that inhibits DNA methyltransferase, reduces cytosine methylation, and induces maturation of some leukemic cell lines. It also is an antiproliferative drug. Administration of the drug and its congener decitabine has resulted in improvement of some patients with oligoblastic leukemia.267,375 5-Azacytidine at a dose of 75 mg/m2 once per day given subcutaneously for 7 consecutive days each month provided significantly more frequent benefit to two-thirds of patients than did supportive care. Quality of life was enhanced, and disease progression was delayed.267,275,376 Complete responses were seen in approximately 15 percent of 5-azacytidine–treated patients, and up to 36 percent had hematologic improvement.375 Ninety percent of responses were seen by cycle 6. In another series,377 subclasses of MDS did not predict for response to 5-azacytidine. A decrease in the white blood count during the initial cycle correlated with a higher response rate. 5-Azacytidine was approved by the FDA in 2004 for treatment of all subtypes of myelodysplastic syndrome. Treatment with this agent can usually be accomplished on an outpatient basis,378 and intravenous379 and oral formulations have been examined.380 Other schedules of administration to accommodate outpatient therapy have been reported to have benefit but have not been directly compared to the 75 mg/m2 daily dose for 7 days every 4 weeks.381

5-Aza-2′-deoxycytidine (decitabine) is also FDA approved for all MDS risk categories. Seventeen percent of patients in one series had a major cytogenetic response on an intention-to-treat basis after a median of three courses. The median duration of cytogenetic response was 7.5 months in all IPSS groups.382 Patients who responded had improved survival compared with patients in whom the cytogenetically abnormal clone persisted.383,384 A 5-day intravenous schedule was found to be optimal in one series, which examined several schedules of administration385; examination of optimal doses and schedules continues.386 Decitabine probably works partly through demethylation, as it has resulted in demethylation of a hypermethylated p15/INK4B gene in patients.383 Demethylation was associated with clinical responses.384 Oligodeoxynucleotide antisense approaches to DNA methyltransferase-1 inhibition are also being explored in MDS.387

Inhibitors of histone deacetylation may have activity in MDS and are under investigation. Numerous agents are being studied and include depsipeptide, butyrate derivatives, suberoylanilide hydroxamic acid, and valproic acid.388 Phase I trials have been completed in MDS with LBH589, a cinnamic hydroxamic acid analogue389 and with MGCD0103.390 There is interest in combining histone deacetylation inhibitors with DNA methyltransferase inhibitors.391

Management of Thrombocytopenia

Thrombocytopenia is common in MDS and has a higher prevalence in higher-risk IPSS categories.392 Furthermore, many therapies used in MDS may exacerbate thrombocytopenia. Platelet transfusions may be required if the platelet count falls below 10 x 109 cells/L or in support of chemotherapeutic-induced thrombocytopenia. Antifibrinolytic agents such as aminocaproic acid can be used in patients who have bleeding despite platelet transfusion or to decrease the need of platelet transfusions.393 Low-dose IL-11 is being studied as a means of increasing the platelet count in patients with symptomatic thrombocytopenia as are thrombopoietin-receptor agonists. AMG-531 (Romiplostim) and Eltrombopag may increase platelet counts in a subset of MDS patients394–396 and are under investigation for this purpose.

Neutropenia, Fever, and Infections

Granulocyte-Stimulating Factors

Randomized, double-blind studies have not shown that any cytokine prolongs survival or reduces morbidity in oligoblastic leukemia. GM-CSF and G-CSF397–399 increase neutrophil counts and functions in some patients. G-CSF receptor expression may be low in some patients with MDS and prevents a good response to endogenous or administered G-CSF.400 Complete remissions have been reported in hypoplastic AML/MDS with G-CSF alone.401 Granulocyte transfusions are rarely used in MDS.402 Rare serious complications, such as splenic rupture, have been reported with use of G-CSF.403 Cytokines do not delay progression to acute leukemia; however, they increase the percentage of blasts in the blood in a proportion of patients, an event that is not always reversible with cessation of cytokine.397,398 In one review, 22 of 83 reported cases of myelodysplasia treated with G-CSF or GM-CSF had an increase in marrow blast percentage, and AML evolved in 12 of 69 patients. An increased percentage of abnormal macrophages has been reported.404 Use of these agents without chemotherapy in oligoblastic leukemias carries a risk of promoting expansion of leukemic blast cells.405 Combinations of growth factors alone or coupled with maturing agents have not significantly improved response or survival rates.406,407

Antibiotics

Febrile events are common in higher-risk syndromes because of the frequency of moderately severe neutropenia and functional disorders of neutrophils and monocytes. Also, chemotherapy is more likely to be used in these situations, inducing severe neutropenia. Careful cultures and use of broad-spectrum antibiotics until and if a specific organism are found is important (see Chap. 20).

Therapy for Patients with High-Intermediate (Int-2) or High-Risk Prognostic Scores

Demethylating Agent Therapy

Patients in higher-risk IPSS categories can be treated with demethylating agents if they are not suitable candidates for allogeneic hematopoietic stem cell transplantation. In one study of higher-risk MDS patients where 5-azacytidine was compared to conventional care regimens that included supportive care, low-dose cytarabine treatment, or intensive induction chemotherapy, 5-azacytidine increased survival as compared to standard care regimens.408 With the exception of allogeneic hematopoietic stem cell transplantation, however, all of these treatments are palliative.409

Acute Myelogenous Leukemia Chemotherapy

Chemotherapeutic regimens containing standard doses of cytarabine, an anthracycline antibiotic, and/or etoposide (see Chap. 89) result in remission in fewer than 20 percent of patients with high-risk MDS. Moreover, a proportion of patients become worse with intensive chemotherapy. The advanced age and the high frequency of cardiac, renal, immunologic, and other organ system impairment in most patients with oligoblastic leukemia are largely responsible for the poor outcome. Patients who are younger than age 60 years have higher remission rates of up to 50 percent410 and can be considered for intensive therapy. Patients older than age 60 years have a median survival of only 9.5 months with this approach and the survival is reduced to 4 months in those with unfavorable karyotypes, indicating a lack of benefit in this group.411 In addition to the standard combination of anthracycline and cytarabine, other regimens, such as liposomal daunorubicin and topotecan with or without thalidomide, did not result in clinical benefit in patients with AML or high-risk MDS.412 The so-called FLAG-Ida regimen (fludarabine, cytarabine, idarubicin, and G-CSF) resulted in 53 percent complete remissions and 11 percent improvement in 45 patients with high-risk myeloid malignancies, 13 of whom had MDS.413 Gemtuzumab ozogamicin (Mylotarg), which is approved for treatment of relapsed AML in elderly patients, has not been useful for treatment of MDS.414,415

Hematopoietic Stem Cell Transplantation

Allogeneic Stem Cell Transplantation

This approach has been used to treat various MDS in patients ranging in age from 1 month to older than 70 years.416–418 It remains the only treatment with curative potential for MDS. Conditioning regimens have consisted of cyclophosphamide plus irradiation or busulfan plus cyclophosphamide. Most patients have received transplants from histocompatible sibling donors, although some experience with partially mismatched, related, and unrelated donors has been reported. A good representation of the results of this traditional approach using marrow stem cells is a study of 93 patients (age range: 1 month to older than 60 years; median age: 30 years).419 The most favorable results were seen in patients younger than age 40 years with shorter duration of disease and with less than 5 percent blast cells in the marrow at the time of transplant. These patients may have a disease-free survival of 60 percent at 4 years and an overall disease-free survival estimated at 40 percent. Older patients had higher peritransplantation mortality and relapse rates. Actuarial relapse probability at 4 years was 30 percent for the entire group and 50 percent for patients with greater than 5 percent marrow blasts. Cytogenetic abnormalities did not predict outcome in this study, but adverse cytogenetics were an important prognostic factor in other studies. With targeted busulfan therapy, stem cell transplantation can be successfully performed in patients as old as 66 years of age.420,421 Numerous factors such as disease stage, patient age, comorbidities, prior therapies, type of donor, and source of stem cells need to be weighed when recommending stem cell transplantation to MDS patients.

An International Bone Marrow Transplant Registry report of 452 patients with MDS who received allogeneic transplantation found that young age and platelet counts greater then 100 x 109/L prior to transplantation were associated with lower transplantation mortality, higher disease-free survival, and overall survival. Patients with higher percentage of blasts and high IPSS scores had higher relapse rates.422 Blood or marrow stem cell sources can be utilized. One report showed superior results with mobilized blood versus marrow stem cells.423

The National Marrow Donor Program transplantation experience in MDS included 510 patients. Median age was 38 years, and the probability of disease-free survival at 2 years was 29 percent (confidence interval [CI] 25–33%). The 2-year incidence of treatment-related mortality was 54 percent, which was the major barrier to success in this patient population.424 Unrelated cord blood transplantation for adult and pediatric patients with MDS has been successfully performed,425 but results with unrelated marrow donors are inferior to the results of matched sibling transplants. It is anticipated that these results will improve in the era of high-resolution HLA matching between donor and recipient.426

Stem cell transplantation for MDS should be performed before the disease progresses to AML.427 When T-cell depletion is used to prevent graft-versus-host disease, the best outcomes occur in those who are transplanted while in remission.428 Poor cytogenetics may impact risk of relapse but not nonrelapse mortality.429 In one retrospective series, blast percentage less than 5 percent at time of transplantation was the best predictor of improved disease-free survival, and myeloablative conditioning was associated with lower relapse risk but could not overcome increased disease burden.430 Patients with secondary MDS have comparable outcomes after stem cell transplantation as those with de novo MDS when high-risk cytogenetics are considered.431,432 Pretransplantation neutropenia is also associated with inferior outcomes as a result of infection-related mortality.433 Prior therapy with demethylating agents does not appear to increase the toxicity of transplantation and whether it will improve outcomes by decreasing disease burden remains to be studied systematically.434 The morbidity and mortality of various transplantation approaches remain high, and some patients are not candidates for ablative transplantation because of age or comorbid conditions.435

Reduced-intensity conditioning with allogeneic hematopoietic stem cell transplantation from HLA-identical family members or unrelated donors has been examined for MDS treatment.436 In one series of 16 patients (median age: 54 years) receiving a conditioning regimen of fludarabine and cyclophosphamide, no day 100 transplantation-related mortality was observed, and the 2-year actuarial event-free survival was 56 percent (CI 30–68%). Other fludarabine-containing conditioning regimens have been reported.437,438 One series compared reduced intensity to standard transplantation in MDS patients and noted similar 2-year overall and disease-free survival with different patterns of toxicity.439 In some series, patients older than 70 years of age have undergone transplantation440; the future role this will play in therapy of older MDS patients is under investigation.441 For those patients who relapse after reduced-intensity stem cell transplantation, salvage therapy with donor lymphocyte infusions, second transplantations, or chemotherapy may be feasible.442,443

Autologous Stem Cell Infusion

Patients with oligoblastic leukemia have been infused with their own stem cells after intensive chemotherapy.444 The approach may be limited by contamination of the stem cell product with a repopulating leukemic cell and the absence of a graft-versus-leukemia effect. The absence of a graft-versus-host reaction makes the approach more applicable to the age group usually affected. In selected patients, peritransplantation mortality with intensive therapy and stem cell rescue has been approximately 10 percent, and approximately 50 percent of selected patients had extended survivals.445 The more advanced the disease at the time of treatment, the worse the outcome. With the increasing use of reduced-intensity allogeneic transplantation, autologous stem cell transplantation has been used less often. Interestingly, when autologous transplantations for AML are performed in patients with antecedent myelodysplasia, no impact on stem cell mobilization or hematopoietic recovery has been noted.446

Other Therapies in Use or under Study in MDS

Other Single-Agent Cytotoxic Drugs

Hydroxyurea and low-dose etoposide are useful in controlling leukemic cell proliferation but usually produce only partial responses and do not influence survival duration.331 Occasional patients have achieved remissions with etoposide (50 mg as a 2-hour infusion, two to seven times weekly for 4 weeks; or 100 mg/day orally for 3 days and then 50 mg twice weekly).447 Low-dose melphalan,448 gemcitabine,449 CPT-11, a DNA topoisomerase I inhibitor,450 troxacitabine, an enantiomer of cytarabine,451 and weekly doses of oral idarubicin452 have each resulted in responses in some patients. Clofarabine, a purine nucleoside analogue, has activity in MDS.453 Oral topotecan has only modest activity in MDS.454 ABT-751, a microtubule inhibitor, is being studied in MDS.455

Antiangiogenesis Agents

Thalidomide has shown effectiveness in MDS therapy.456 Patients receiving 100 to 400 mg/day for 12 or more weeks had no cytogenetic or complete responses, but 16 patients had hematologic improvement.457 In another series of 34 patients in whom 400 mg/day was the median dose tolerated, 6 patients had progressive disease, 4 patients had stable disease, and 11 patients had partial remissions (5 major responses and 6 minor responses), accounting for a 56 percent response rate. Hematologic improvement was not noted until after a median of 2 months.458 Cytogenetic responses have been seen in cases of monosomy 7, the 5q– syndrome, and with complex karyotypic abnormalities. Although thalidomide has antiangiogenesis activity, it also decreases vascular endothelial growth factor and basic fibroblast growth factor levels.459 The drug may have a particular role in patients with marrow fibrosis.460 Thromboembolic events have occurred in patients receiving thalidomide in combination with darbepoietin-.461

The thalidomide derivative lenalidomide (Revlimid) lowers levels of proangiogenic cytokines, inhibits attachment of stromal cells, promotes cell-cycle arrest and apoptosis, and affects function of natural killer and cytotoxic T lymphocytes. In patients who have chromosome 5q deletion, lenalidomide has been found to reduce transfusion requirements and reverses cytogenetic abnormalities.462 In patients without deletion 5q, reduction in transfusion requirements occur in approximately 43 percent with some patients becoming independent of transfusions. This trial included only low- or Int-1–risk MDS,463 whereas 5q– patients who have high-risk MDS can show responses. Additional cytogenetic abnormalities limit the responses.464 Unlike the case with thalidomide, dose reduction for myelosuppression often was required, and myelosuppression appears to be lenalidomide’s primary toxicity.465 In those patients with 5q deletion, cytopenias during therapy may be indicative of a response.466

Anti-Tumor Necrosis Factor Therapy

The soluble TNF receptor fusion protein etanercept (p75 TNFR:Fc) has produced mixed results in MDS. In one pilot series, moderate improvement in cytopenias was noted,467 whereas in another trial, no responses were noted in 10 patients.468 In another pilot study of 3 months duration, one patient became transfusion independent temporarily, but overall efficacy was low.469 The chimeric anti–TNF- monoclonal antibody infliximab resulted in two sustained erythroid responses (one major and one minor), and a decreased percentage of apoptotic cells in the marrow.470 It is anticipated that therapies such as TNF inhibitors, which inhibit apoptosis, might be useful in low-grade MDS, whereas in high-grade MDS, therapies that promote apoptosis might be more effective.471

Agents that Alter Oxidation State

Amifostine has had minimal activity in MDS.472 TLK199, a glutathione analogue inhibitor of glutathione S-transferase has resulted in hematologic improvement in early phase studies473 and is still undergoing evaluation.

Retinoids, Vitamin D Derivatives, Arsenic Trioxide, and Other Potentially Maturation-Enhancing Agents

Glucocorticoids, vitamin A analogues (retinoids), vitamin D analogues (dihydroxyvitamin D3), pyrimidine analogues (cytarabine), hexamethylene bisacetamide, and interferon are among other agents that can induce in vitro maturation of mouse and human leukemic cells.474–476 Use of cis-retinoic acid, 20 to 100 mg/m2, isotretinoin, 25 mg/m2, or ATRA, 45 mg/ m2, orally given daily for up to 3 months, has produced only slight, transient (few weeks) improvement in a very small proportion of patients with oligoblastic leukemia.477,478

A combination of low-dose cytarabine, retinoic acid, and 1,25-dihydroxyvitamin D3 in 44 patients with oligoblastic leukemias produced 50 percent response rates, with longer survival in responders than in nonresponders.479 Hexamethylene bisacetamide given intravenously at a dosage of 20 to 24 g/m2 per day for 10 days, followed by an 18- to 75-day observation period, resulted in increased neutrophil counts and reduced marrow blasts in 4 of 16 patients with oligoblastic leukemia.480 In another study, no responses were observed.475 Sodium phenylbutyrate is an agent that has shown some activity against oligoblastic leukemia and is in clinical trials.481

Arsenic trioxide, used as a single agent, results in responses in approximately 20 percent of cases.482 Low-risk MDS patients are most likely to show benefit.483 Whether the drug affects cell maturation, apoptosis, or proliferation in this disease remains to be determined.484–486

Tyrosine Kinase and Other Cell-Signaling Inhibitors

Imatinib mesylate, the tyrosine kinase inhibitor of ABL, KIT, and platelet-derived growth factor receptor, has not resulted in clinical responses in patients with MDS.487,488 Inhibitors of RAF protein kinase such as sorafenib,489 and inhibitors of farnesyltransferase such as lonafarnib,490 tipifarnib,491,492 and BMS-214662,493 have been examined in MDS. Statins that inhibit geranylgeranylation are being studied for treatment of AML and MDS.494 Agents such as bortezomib, which indirectly target nuclear factor-B, are being examined in MDS,495–497 as are agents which target mTOR (mammalian target of rapamycin).498 Progenitors involved in MDS rarely express FLT-3 mutations,499 so FLT-3 inhibitors are not thought to be useful in the treatment of MDS.

Uncommon Acquired Syndromes with Increased Risk of Acute Myelogenous LeukemiaAmegakaryocytic Thrombocytopenia

Amegakaryocytic thrombocytopenia may be congenital, associated with MPL gene mutations, or acquired,500,501 and are both very uncommon preleukemic syndromes (<1%), although bona fide cases have transformed into AML months or years after diagnosis.502,503 Among 1220 cases of MDS, 11 cases of isolated thrombocytopenia were associated with clonal chromosome abnormalities, usually involving chromosome 3, 5, 8, or 20. Antiplatelet antibodies were not present, and glucocorticoids were ineffective. Five of the 11 patients progressed to acute myelogenous leukemia (Table 88–6; see Chap. 119).502

Table 88–6. Hypocellular Marrow Syndromes that May Precede Onset of Acute Myelogenous Leukemia
Amegakaryocytic thrombocytopenia (Chap. 110)
Chronic hypoplastic neutropenia (Chap. 65)
Apparent aplastic anemia with evidence of clonal hematopoiesis (Chap. 34)
Paroxysmal nocturnal hemoglobinuria–aplastic anemia syndrome (Chaps. 34 and 40)

Isolated Neutropenia

Acquired, isolated, chronic neutropenic states are very rare antecedents of AML. Congenital neutropenia can evolve into AML.504 The latter evolution is associated with mutations in the G-CSF-receptor (CSF3R) gene (see Chap. 65). Evolution of Shwachman-Diamond syndrome (neutropenia and exocrine pancreatic insufficiency) into oligoblastic or overt acute leukemia has been documented.505 The related disorder, Pearson syndrome (sideroblastic anemia, neutropenia, and exocrine pancreatic insufficiency), is a preleukemia disorder in children (see Chap. 34).506

Chronic Monocytosis

In a small proportion of patients, unexplained persistent monocytosis may be the most striking blood cell abnormality for months or years before development of acute leukemia.122–124

Aplastic Anemia, Paroxysmal Nocturnal Hemoglobinuria, and Eosinophilic Fasciitis

AML or MDS occurs in a proportion of patients with acquired aplastic anemia.507,508 Since the advent of immunotherapy, the propensity to myelodysplasia and leukemia has increased, partly because of the greater longevity of patients and the often incomplete restitution of hematopoiesis. Patients who initially responded to immunosuppressive therapy have later developed MDS (see Chap. 34 for a discussion of the interrelationship among aplastic anemia, MDS, and paroxysmal nocturnal hemoglobinuria).509

Paroxysmal nocturnal hemoglobinuria is a clonal stem cell disease that often is associated with marrow hypoplasia (see Chap. 40). AML may ensue in approximately 0.5 percent of patients. It is a clonally derived syndrome with a low incidence of leukemic transformation relative to other clonal myeloid diseases. All chronic clonal hemopoietic stem cell disorders (e.g., polycythemia vera, essential thrombocythemia, idiopathic myelofibrosis, chronic myelogenous leukemia) have a propensity to undergo clonal evolution to AML (see Chap. 85). Patients with indolent myeloid clonal disorders may have a paroxysmal nocturnal hemoglobinuria-like defect of their blood cell membranes.

Eosinophilic fasciitis mimics the cutaneous manifestations of scleroderma. Symmetrical swelling and induration of arms and legs, sparing the hands and feet, are common.510,511 Eosinophilia and hypergammaglobulinemia are frequent. Immune cytopenias, aplastic anemia, myelodysplasia, AML, and lymphoma have been associated with the disease.512 An immune mechanism has been postulated for all the disease manifestations. The risk of developing AML is greatly increased compared with healthy individuals.510–512 Marrow transplantation has been used to treat the aplastic anemia.513

Prodromal Syndromes Antedating Lymphocytic LeukemiaThe indolent clonal disorders usually imply conditions that are an antecedent of myelogenous leukemia. A significant proportion of cases of AML are preceded by MDS. Even in de novo AML many cases have protracted periods of symptoms and signs before onset. ALL usually begins explosively, and symptoms rarely are present for more than a few weeks prior to diagnosis (see Chap. 93). Intermediate syndromes (e.g., smoldering or oligoblastic lymphocytic leukemia or prodromal anemias) are rare, but the latter have been reported, occasionally in children,514 but especially in adults who develop ALL.515–521

Apparent aplastic anemia522–526 or erythroid hypoplasia527 has been described as an antecedent to ALL in approximately 2 percent of childhood cases and much less commonly in adult cases. The aplasia is promptly improved by glucocorticoids, and ALL ensues soon, usually within 1 to 8 months. The brief interval between remission of aplastic anemia and onset of leukemia suggests the leukemia, although inapparent on marrow biopsy, in some way initiates the aplasia.522,528 Remission of aplasia followed shortly by ALL has occurred in the absence of glucocorticoid or other specific therapy in several cases. The aplastic marrow prodrome of ALL may be distinguishable by its very high prevalence in females (approximately 90%), high prevalence of fibrosis on marrow biopsy (approximately 90%), frequent marrow lymphocytosis (approximately 60%), and spontaneous, temporary recovery (>90%).529

Indolent Clonal Myeloid Disorders or Oligoblastic (Myelogenous) Leukemia Preceding or Emerging in Lymphoid Malignancies Other Than Acute Lymphocytic LeukemiaSideroblastic anemia sometimes associated with qualitative disorders of other blood cell lines (such as thrombopathy) has developed in patients who had, or later developed, a lymphoproliferative disease, such as hairy cell leukemia, lymphocytic lymphoma, myeloma, chronic lymphocytic leukemia, or Hodgkin lymphoma.530–538 The sideroblastic anemia in these cases was not preceded by cytotoxic therapy. Similar associations have been reported in patients who received chemotherapy or radiotherapy for a lymphoproliferative disease or a solid tumor and who later developed a preleukemic syndrome presumed to result from the prior treatment. Other types of myelodysplasia can occur concurrent with B- or T-lymphocyte–derived tumors.530–539
References

1. Lichtman MA: Myelodysplasia or myeloneoplasia: Thoughts on the nosology of the clonal myeloid disorders. Blood Cells Mol Dis 26:572, 2000. [PMID: 11112390]
2. Brunning RD, Porwit A, Orazi A, et al: Myelodysplastic syndromes, in WHO Classification of Tumors; Tumors of Haematopoietic and Lymphoid Tissues, edited by SH Swerdlow, E Campo, NL Harris, ES Jaffe, SA Pileri, H Stein, J Thiele, JW Vardiman, p 87. IARC Press, Lyon, 2008.
3. Dreyfus B, Rochant H, Sultan C, et al: Les anémies réfractaires avec excès de myeloblastes dans la moelle. Etude de onze observations. Presse Med 78:359, 1970. [PMID: 5264816]
4. Layton DM, Mufti GJ: Myelodysplastic syndromes: Their history, evolution, and relation to acute myeloid leukemia. Blut 53:423, 1986. [PMID: 3542084]
5. Chevallier P: Sur la terminologie des leucoses et des affection frontières. Les odo-leucoses. Sang 15:587, 1942–43.
6. Hamilton-Paterson JL: Preleukaemic anemia. Acta Haematol 2:309, 1949. [PMID: 15390487]
7. Block M, Jacobson LO, Bethard WJ: Preleukemic acute human leukemia. JAMA 152:1018, 1953. [PMID: 13052490]
8. Vilter RW, Jarrold T, Will JJ, et al: Refractory anemia with hyperplastic bone marrow. Blood 15:1, 1960. [PMID: 13842324]
9. Schiller M, Rachmilewitz EA, Izak G: Pancytopenia with hypercellular hemopoietic tissue. Isr J Med Sci 5:69, 1969. [PMID: 5796282]
10. Saarni MI, Linman JW: Preleukemia. Am J Med 55:38, 1973. [PMID: 4515079]
11. Linman JW, Saarni MI: The preleukemic syndrome. Semin Hematol 11:93, 1974. [PMID: 4520530]
12. Pierre RV: Preleukemic states. Semin Hematol 11:73, 1974. [PMID: 4809725]
13. Dreyfus B: Preleukemic states. I. Definition and classification. II. Refractory anemia with an excess of myeloblasts in the bone marrow (smoldering acute leukemia) Blood Cells 2:33, 1976.
14. Linman JW, Bagby GC Jr: The preleukemic syndrome: Clinical and laboratory features, natural course and management. Blood Cells 2:11, 1976.
15. Linman JW, Bagby GC Jr: The preleukemic syndrome (hemopoietic dysplasia). Cancer 42:854, 1978. [PMID: 356956]
16. Izrael V, Jacquillat C, Chastang C, et al: New data about oligoblastic leukemias. Apropos of an analysis of 120 cases. Nouv Presse Med 4:947, 1975. [PMID: 1144035]
17. Bernard J, Izrael V, Jacquillat C: Oligoblastic leukemias. Nouv Presse Med 4:943, 1975. [PMID: 1144034]
18. Bessis M, Bernard J: Hematopoietic dysplasias. Blood Cells 2:5, 1976.
19. WHO Classification of Tumors of Hematopoietic and Lymphoid Tissues, 4th ed., edited by SH Swerdlow, E Campo, NL Harris, ES Jaffe, SA Pileri, H Stein, J Thiele, JW Vardiman. WHO Press, Lyons, 2008.
20. Maynadie M, Picard F, Husson B, et al: Immunophenotypic clustering of myelodysplastic syndromes. Blood 100:2349, 2002. [PMID: 12239142]
21. Stetler-Stevenson M, Arthur DC, Jabbour N, et al: Diagnostic utility of flow cytometric immunophenotyping in myelodysplastic syndrome. Blood 98:979, 2001. [PMID: 11493442]
22. Groupe Francais de Morphologie Hématologique: French registry of acute leukemia and myelodysplastic syndromes. Cancer 60:1385, 1987.
23. Aul C, Gatterman N, Schneider W: Age-related incidence and other epidemiologic aspects of myelodysplastic syndrome. Br J Haematol 82:358, 1992. [PMID: 1419819]
24. McNally RJO, Rowland D, Roman E, Cartwright RA: Age and sex distributions of hematological malignancies in the U.K. Hematol Oncol 15:173, 1997. [PMID: 9722889]
25. Luna-Fineman S, Shannon KM, Atwater SK, et al: Myelodysplastic and myeloproliferative disorders of childhood: A study of 167 patients. Blood 93:459, 1999. [PMID: 9885207]
26. Novitzky N, Prindull G, for the European Society of Paediatric Haematology and Immunology: Myelodysplastic syndromes in children. Am J Hematol 63:212, 2000. [PMID: 10706766]
27. Hasle H, Niemeyer CM, Chessells JM, et al: A pediatric approach to the WHO classification of myelodysplastic and myeloproliferative diseases. Leukemia 17:277, 2003. [PMID: 12592323]
28. Sasaki H, Manabe A, Kojima S, et al: Myelodysplastic syndrome in childhood. Leukemia 15:713, 2001.
29. Kardos G, Baumann I, Passmore SJ, et al: Refractory anemia in childhood: A retrospective analysis of 67 patients with particular reference to monosomy 7. Blood 102:1997, 2003. [PMID: 12763938]
30. Ma X, Does M, Raza A, Mayne ST: Myelodysplastic syndromes: Incidence and survival in the United States. Cancer 109:1536, 2007. [PMID: 17345612]
31. Rollison DE, Howlader N, Smith MT, et al: Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the United States, 2001–2004, using data from the NAACCR and SEER programs. Blood 112:45, 2008. [PMID: 18443215]
32. West RR, Stafford DA, White DT, et al: Cytogenetic abnormalities in the myelodysplastic syndromes and occupational or environmental exposure. Blood 95:2093, 2000. [PMID: 10706879]
33. Nisse C, Haguenoer JM, Grandbastien B, et al: Occupational and environmental risk factors of the myelodysplastic syndromes in the North of France. Br J Haematol 112:927, 2001. [PMID: 11298587]
34. Yin SN, Hayes RB, Linet MS, et al: A cohort study of cancer among benzene-exposed workers in China: Overall results. Am J Ind Med 29:227, 1996. [PMID: 8833775]
35. Snyder R: Benzene and leukemia. Crit Rev Toxicol 32:155, 2002. [PMID: 12071572]
36. Park DJ, Koeffler HP: Therapy-related myelodysplastic syndromes. Semin Hematol 33:256, 1996. [PMID: 8819235]
37. Rigolin GM, Cuneo A, Roberti MG, et al: Exposure to myelotoxic agents and myelodysplasia: Case-control study and correlation with clinicobiological findings. Br J Haematol 103:189, 1998. [PMID: 9792307]
38. Sterkers Y, Preudhomme C, Lai JL, et al: Acute myeloid leukemia and myelodysplastic syndromes following essential thrombocythemia treated with hydroxyurea: High proportion of cases with 17p deletion. Blood 91:616, 1998. [PMID: 9427717]
39. Van Den Neste E, Louviaux I, Michaux JL, et al: Myelodysplastic syndrome with monosomy 5 and/or 7 following therapy with 2-chloro-2′-deoxyadenosine. Br J Haematol 105:268, 1999.
40. Krishnan A, Bhatia S, Slovak ML, et al: Predictors of therapy-related leukemia and myelodysplasia following autologous transplantation for lymphoma. Blood 95:1588, 2000. [PMID: 10688812]
41. Abruzzese E, Radford JE, Miller JS, et al: Detection of abnormal pretransplant clones in progenitor cells of patients who developed myelodysplasia after autologous transplantation. Blood 94:1814, 2000.
42. Smith SH, Le Beau MM, Huo D, et al: Clinical-cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: The University of Chicago series. Blood 102:43, 2003. [PMID: 12623843]
43. Lobe I, Rigal-Huguet F, Vekhoff A, et al: Myelodysplastic syndrome after acute promyelocytic leukemia: The European APL group experience. Leukemia 17:1600, 2003. [PMID: 12886249]
44. Nakanishi M, Tanaka K, Shintani T, et al: Chromosomal instability in acute myelocytic leukemia and myelodysplastic syndrome patients among atomic bomb survivors. J Radiat Res (Tokyo) 40:159, 1999. [PMID: 10494147]
45. Finch SC: Myelodysplasia and radiation. Radiat Res 161:603, 2004. [PMID: 15161362]
46. Alter BP: Cancer in Fanconi’s anemia 1923–2001. Cancer 97:425, 2003. [PMID: 12518367]
47. Segel GB, Lichtman MA: Familial (inherited) leukemia, lymphoma, and myeloma. Blood Cells Mol Dis 32:2004.
48. Horwitz M, Sabath DE, Smithson WA, Radich J: A family inheriting different subtypes of acute myelogenous leukemia. Am J Hematol 52:295, 1996. [PMID: 8701948]
49. Pradhan A, Mijovic A, Mills K, et al: Differentially expressed genes in adult familial myelodysplastic syndromes. Leukemia 18:449, 2004. [PMID: 14737073]
50. Kumar T, Mandla SG, Greer WL: Familial myelodysplastic syndrome with early age onset. Am J Hematol 64:53, 2000. [PMID: 10815788]
51. Griffith DP, Liff DA, Ziegler TR, et al: Acquired copper deficiency: A potentially serious and preventable complication following gastric bypass surgery. Obesity (Silver Spring) 17:827, 2009. [PMID: 19148115]
52. Fong T, Vij R, Vijayan A, et al: Copper deficiency: An important consideration in the differential diagnosis of myelodysplastic syndrome. Haematologica 92:1429, 2007. [PMID: 18024379]
53. Abkowitz JL, Fialkow PJ, Niebrugge DJ, et al: Pancytopenia as a clonal disorder of a multipotent hemopoietic stem cell. J Clin Invest 73:258, 1984. [PMID: 6690481]
54. Rasking WH, Tirumali N, Jacobson R, et al: Evidence for a multistep pathogenesis of a myelodysplastic syndrome. Blood 63:1318, 1984.
55. Mongkonsritragoon W, Letendre L, Li CY: Multiple lymphoid nodules in bone marrow have the same clonality as underlying myelodysplastic syndrome recognized with fluorescent in situ hybridization technique. Am J Hematol 59:252, 1998. [PMID: 9798667]
56. Janssen JWG, Buschle M, Layton M, et al: Clonal analysis of myelodysplastic syndromes: Evidence of multipotent stem cell origin. Blood 73:248, 1989. [PMID: 2562924]
57. Boultwood J, Weainscot JS: Clonality in the myelodysplastic syndromes. Int J Hematol 73:411, 2001. [PMID: 11503954]
58. Gerritsen WR, Donohue J, Bauman J, et al: Clonal analysis of myelodysplastic syndrome: Monosomy 7 is expressed in the myeloid lineage but not in the lymphoid lineage as detected by fluorescent in situ hybridization. Blood 80:217, 1992. [PMID: 1611087]
59. Anastasi J, Fang J, LeBeau MM, et al: Cytogenetic clonality in myelodysplastic syndromes studied with fluorescence in situ hybridization: Lineage, response to growth factor therapy, and clone expansion. Blood 81:1580, 1993. [PMID: 8453104]
60. Culligan DJ, Cachia P, Whittaker A, et al: Clonal lymphocytes are detectable in only some cases of MDS. Br J Haematol 81:346, 1992. [PMID: 1390207]
61. Abrahamson G, Boultwod J, Madden J, et al: Clonality of cell population in refractory anaemia using combined approach of gene loss and X-linked restricting fragment length polymorphism–methylation analysis. Br J Haematol 79:550, 1991. [PMID: 1685326]
62. Delforge M, Demuynck H, Verhoef G, et al: Patients with high risk myelodysplastic syndrome can have polyclonal or clonal haemopoiesis in complete haematological remission. Br J Haematol 102:486, 1998. [PMID: 9695963]
63. Lawrence HJ, Broudy VC, Magenis RE, et al: Cytogenetic evidence for involvement of B-lymphocytes in acquired idiopathic sideroblastic anemia. Blood 70:1003, 1982.
64. Meers S, Vandenberghe P, Boogaerts M, et al: The clinical significance of activated lymphocytes in patients with myelodysplastic syndromes: A single centre study of 131 patients. Leuk Res 32:1026, 2008. [PMID: 18006057]
65. Epling-Burnette PK, Painter JS, Rollison DE, et al: Prevalence and clinical association of clonal T-cell expansions in myelodysplastic syndrome. Leukemia 21:659, 2007. [PMID: 17301813]
66. Flores-Figueroa E, Montesinos JJ, Flores-Guzm·n P, et al: Functional analysis of myelodysplastic syndromes-derived mesenchymal stem cells. Leuk Res 32:1407, 2008. [PMID: 18405968]
67. Nakagawa T, Saitoh S, Imoto S, et al: Multiple point mutation of N-ras and K-ras oncogenes in myelodysplastic syndrome and acute myelogenous leukemia. Oncology 49:114, 1992. [PMID: 1574246]
68. VanKamp H, DePijper C, Verlaan-de Vries M, et al: Longitudinal analysis of point mutations of the N-ras protooncogene in patients with myelodysplasia using archival blood smears. Blood 79:1266, 1992.
69. Paquette RL, Landau EM, Pierre RV, et al: N-ras mutations are associated with poor prognosis and increased risk of leukemia in myelodysplastic syndrome. Blood 82:590, 1993. [PMID: 8329714]
70. Bartram CR: Molecular genetic aspects of myelodysplastic syndromes. Semin Hematol 33:139, 1996. [PMID: 8722684]
71. Parker J, Mufti GJ: Ras and myelodysplasia: Lessons from the last decade. Semin Hematol 33:206, 1996. [PMID: 8819231]
72. Padua RA, Guinn BA, Al-Sabah AI, et al: RAS, FMS and p53 mutations and poor clinical outcome in myelodysplasias: A 10-year follow-up. Leukemia 12:887, 1998. [PMID: 9639416]
73. Plata E, Viniou N, Abazis D, et al: Cytogenetic analysis and RAS mutations in primary myelodysplastic syndromes. Cancer Genet Cytogenet 111:124, 1999. [PMID: 10347548]
74. Quesnel B, Guillerm G, Vereecque R, et al: Methylation of the p15 (INK4b) gene in myelodysplastic syndromes is frequent and acquired during disease progression. Blood 91:2985, 1998. [PMID: 9531610]
75. Miyazato A, Ueno S, Ohmine K, et al: Identification of myelodysplastic syndrome-specific genes by DNA microarray analysis with purified hematopoietic stem cell fraction. Blood 98:422, 2001. [PMID: 11435312]
76. Fadilah SAW, Cheong SK, Roslan H, et al: GATA-1 and GATA-2 gene expression is related to the severity of dysplasia in myelodysplastic syndrome. Leukemia 16:1563, 2002. [PMID: 12145700]
77. Wulfert M, Küpper AC, Tapprich C, et al: Analysis of mitochondrial DNA in 104 patients with myelodysplastic syndromes. Exp Hematol 36:577, 2008. [PMID: 18439489]
78. Greenberg PL: Apoptosis and its role in the myelodysplastic syndromes: Implications for disease natural history and treatment. Leuk Res 22:1123, 1998. [PMID: 9922076]
79. Van de Loosdrecht AA, Vellenga E: Myelodysplasia and apoptosis. Med Oncol 17:16, 2000.
80. Huh YO, Jilani I, Estey E, et al: More cell death in refractory anemia with excess blasts in transformation than in acute myeloid leukemia. Leukemia 16:2249, 2002. [PMID: 12399969]
81. Raza A, Alvi S, Broady-Robinson L, et al: Cell cycle kinetic studies in 68 patients with myelodysplastic syndromes following intravenous iodo- and/or bromodeoxyuridine. Exp Hematol 25:530, 1997. [PMID: 9197332]
82. Gersuk GM, Beckham C, Loken MR, et al: A role for tumour necrosis factor-alpha, Fas and Fas-ligand in marrow failure associated with myelodysplastic syndrome. Br J Haematol 103:176, 1998. [PMID: 9792306]
83. Mundle SD, Ali A, Cartlidge JD, et al: Evidence for involvement of tumor necrosis factor-alpha in apoptotic death of bone marrow cells in myelodysplastic syndromes. Am J Hematol 60:36, 1999. [PMID: 9883804]
84. Parker JE, Fishlock KL, Mijovic A, et al: “Low-risk” myelodysplastic syndrome is associated with excessive apoptosis and an increased ratio of pro- versus anti-apoptotic bcl-2-related proteins. Br J Haematol 103:1075, 1998. [PMID: 9886323]
85. Gyan E, Frisan E, Beyne-Rauzy O, et al: Spontaneous and Fas-induced apoptosis of low-grade MDS erythroid precursors involves the endoplasmic reticulum. Leukemia 22:1864, 2008. [PMID: 18615109]
86. Meers S, Kasran A, Boon L, et al: Monocytes are activated in patients with myelodysplastic syndromes and can contribute to bone marrow failure through CD40-CD40L interactions with T helper cells. Leukemia 21:2411, 2007. [PMID: 17805323]
87. Amin HM, Jilani I, Estey EH, et al: Increased apoptosis in bone marrow B lymphocytes but not T lymphocytes in myelodysplastic syndrome. Blood 102:1866, 2003. [PMID: 12730116]
88. Baumann I, Scheid C, Koref MS, et al: Autologous lymphocytes inhibit hemopoiesis in long-term culture in patients with myelodysplastic syndrome. Exp Hematol 30:1045, 2002.
89. Moldrem J, Jiang Y, Stetler-Stevenson M, et al: Haematologic response of patients with myelodysplastic syndrome to antithymocyte globulin is associated with a loss of lymphocyte-mediated inhibition of CFU-GM and alterations in T cell receptor V profiles. Br J Haematol 102:1314, 1998.
90. Epperson D, Nakamura R, Saunthararajah Y, et al: Oligoclonal T cell expansion in myelodysplastic syndrome: Evidence for an autoimmune process. Leuk Res 25:1075, 2001. [PMID: 11684279]
91. Marcondes AM, Mhyre AJ, Stirewalt DL, et al: Dysregulation of IL-32 in myelodysplastic syndrome and chronic myelomonocytic leukemia modulates apoptosis and impairs NK function. Proc Natl Acad Sci U S A 105:2865, 2008. [PMID: 18287021]
92. Rosenfeld C, List A: A hypothesis for the pathogenesis of myelodysplastic syndromes: Implications for new therapies. Leukemia 14:2, 2000. [PMID: 10637470]
93. Bagby GC: The preleukemic syndrome (hematopoietic dysplasia). Blood Rev 2:194, 1988. [PMID: 3052665]
94. Noel P, Solberg LA Jr: Myelodysplastic syndromes: Pathogenesis, diagnosis and treatment. Crit Rev Oncol Hematol 12:193, 1992. [PMID: 1379818]
95. Ahmad YH, Kiehl R, Papac RJ: Myelodysplasia. The clinical spectrum of 51 patients. Cancer 76:869, 1995. [PMID: 8625191]
96. Hebbar M, Hebbar-Savean K, Fenaux P: Systemic diseases in myelodysplastic syndromes. Rev Med Interne 16:897, 1995. [PMID: 8570952]
97. Saif MW, Hopkins JL, Gore SD: Autoimmune phenomena in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia. Leuk Lymphoma 43:2409, 2002.
98. de la Chapelle A, Lahtinen R: Monosomy 7 predisposes to diabetes insipidus in leukaemia and myelodysplastic syndrome. Eur J Haematol 39:404, 1987.
99. Nakamura F, Kishimoto Y, Handa T, et al: Diabetes insipidus manifesting hypodipsic hypernatremia and dehydration. Am J Hematol 75:213, 2004. [PMID: 15054812]
100. Soppi E, Nousiainen T, Seppa A, et al: Acute febrile neutrophilic dermatosis (Sweet’s syndrome) in association with myelodysplastic syndromes: A report of three cases and a review of the literature. Br J Haematol 73:43, 1989. [PMID: 2679862]
101. Arbetter KR, Hubbard KW, Markovic SN, et al: Case of granulocyte colony-stimulating factor-induced Sweet’s syndrome. Am J Hematol 61:126, 1999. [PMID: 10367792]
102. Avi I, Rosenbaum H, Levy Y, Rowe J: Myelodysplastic syndrome and associated skin lesions: A review of the literature. Leuk Res 23:323, 1999.
103. Weber RFA, Geraedts JPM, Kerkhofs H, Leeksma CHW: The preleukemic syndrome. Acta Med Scand 207:391, 1980. [PMID: 7386231]
104. Ohno E, Ohtsuka E, Watanabe K, et al: Behçet’s disease associated with myelodysplastic syndromes. A case report and a review of the literature. Cancer 79:262, 1997. [PMID: 9010099]
105. Komatsuda A, Miura I, Ohtani H, et al: Crescentic glomerulonephritis accompanied by myeloperoxidase-antineutrophil cytoplasmic antibodies in a patient having myelodysplastic syndrome with trisomy 7. Am J Kidney Dis 31:336, 1998. [PMID: 9469507]
106. Saitoh T, Murakami H, Uchiumi H, et al: Myelodysplastic syndromes with nephrotic syndrome. Am J Hematol 60:200, 1999. [PMID: 10072111]
107. Harewood GC, Loftus EV Jr, Tefferi A, et al: Concurrent inflammatory bowel disease and myelodysplastic syndromes. Inflamm Bowel Dis 5:98, 1999. [PMID: 10338378]
108. Lesprit P, Piette AM, Baumelou E, et al: Panniculitis and myelodysplasia: Report of 2 cases. Eur J Med 2:500, 1993. [PMID: 8258054]
109. Saif MW, Hopkins JL, Gore SD: Autoimmune phenomena in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia. Leuk Lymphoma 43:2083, 2002. [PMID: 12533032]
110. Clark RE, Payne HE, Jacobs A: Primary myelodysplastic syndrome and cancer. Br Med J 294:937, 1987. [PMID: 3107665]
111. Sans-Sabrafen J, Buxó-Costa J, Woessner S, et al: Myelodysplastic syndromes and malignant solid tumors. Am J Hematol 41:1, 1992. [PMID: 1503093]
112. Florensa L, Vallespi T, Woessner S, et al: Incidence and characteristics of lymphoid malignancies in untreated myelodysplastic syndromes. Leuk Lymphoma 23:609, 1996. [PMID: 9031093]
113. Park S, Merlat A, Guesnu M, et al: Pure red cell aplasia associated with myelodysplastic syndromes. Leukemia 14:1709, 2000. [PMID: 10995022]
114. Choi JW, Kim Y, Fujino M, Ito M: Significance of fetal hemoglobin-containing erythroblasts (F blasts) and the F blast/F cell ratio in myelodysplastic syndromes. Leukemia 19:1478, 2002.
115. Kornberg A, Goldfarb A: Preleukemia manifested by hemolytic anemia with pyruvate-kinase deficiency. Arch Intern Med 146:785, 1986. [PMID: 3457556]
116. Harris JW, Koscick R, Lazarus HM, et al: Leukemia arising out of paroxysmal nocturnal hemoglobinuria. Leuk Lymphoma 32:401, 1999. [PMID: 10048414]
117. Lopez M, Bonnet-Gajdos M, Reviron M, et al: Acute leukemia augured before clinical signs by blood group antigen abnormalities and low levels of A and H blood group transferase activities in erythrocytes. Br J Haematol 63:535, 1986. [PMID: 3089272]
118. Anagnou NP, Ley TJ, Chesbro B, et al: Acquired -thalassemia in pre-leukemia is due to decreased expression of all four -globin genes. Proc Natl Acad Sci U S A 80:6051, 1983. [PMID: 6136971]
119. Helder J, Deisseroth A: S1 nuclease analysis of -globin gene expression in preleukemic patients with acquired hemoglobin H disease after transfer to mouse erythroleukemia cells. Proc Natl Acad Sci U S A 84:2387, 1987. [PMID: 3031681]
120. Steensma DP, Higgs DR, Fisher CA, Gibbons RJ: Acquired somatic ATRX mutations in myelodysplastic syndrome associated with -thalassemia (ATMDS) convey a more severe hematological phenotype than germline ATRX mutations. Blood 103:2019, 2004. [PMID: 14592816]
121. Group Française de Morphologie Hématologique: French registry of acute leukemia and myelodysplastic syndromes. Cancer 60:1385, 1987.
122. Jaworkowsky LI, Solovey DY, Rhausova LY, Udris OY: Monocytosis as a sign of subsequent leukemia in patients with cytopenias (preleukemia). Folia Haematol (Frankf) 110:395, 1983. [PMID: 6194073]
123. Friedland ML, Ward H, Wittels EG, Arlin ZA: A monocytic leukemoid reaction: A manifestation of preleukemia. R I Med J 68:173, 1985. [PMID: 3858941]
124. Economopoulos T, Stathakis N, Maragoyannis Z, et al: Myelodysplastic syndrome. Clinical significance of monocyte concentration, degree of blastic infiltration and ring sideroblasts. Acta Haematol 65:97, 1981. [PMID: 6785968]
125. Shetty VT, Mundle SD, Raza A: Pseudo Pelger-Huët anomaly in myelodysplastic syndrome: Hyposegmented or apoptotic neutrophil? Blood 98:1273, 2001. [PMID: 11510471]
126. Langenhuijsen MM: Neutrophils with ring-shaped nuclei in myeloproliferative disease. Br J Haematol 58:227, 1984. [PMID: 6591944]
127. Clark RE, Smith SA, Jacobs A: Myeloid surface antigen abnormalities in myelodysplasia: Relation to prognosis and modification by 13-cis retinoic acid. J Clin Pathol 40:652, 1987. [PMID: 3475292]
128. Cech P, Markert M, Perrin LH: Partial myeloperoxidase deficiency in preleukemia. Blut 47:21, 1983. [PMID: 6305446]
129. Schofield KP, Stone PCW, Kelsey P, et al: Quantitative cytochemistry of blood neutrophils in myelodysplastic syndromes and chronic granulocytic leukaemia. Cell Biochem Funct 1:92, 1983. [PMID: 6090032]
130. Elghetany MT, Peterson B, MacCallum J, et al: Deficiency of neutrophilic granule membrane glycoproteins in the myelodysplastic syndromes: A common deficiency in 216 patients studied by the Cancer and Leukemia Group B. Leuk Res 21:801, 1997. [PMID: 9393593]
131. Ruutu P: Granulocyte function in myelodysplastic syndromes. Scand J Haematol 36(Suppl 45):66, 1986.
132. Prodan M, Tulissi P, Perticarari S, et al: Flow cytometric assay for the evaluation of phagocytosis and oxidative burst of polymorphonuclear leukocytes and monocytes in myelodysplastic disorders. Haematologica 80:212, 1995. [PMID: 7672714]
133. Piva E, De Toni S, Caenazzo A, et al: Neutrophil NADPH oxidase activity in chronic myeloproliferative and myelodysplastic diseases by microscopic and photometric assays. Acta Haematol 94:16, 1995. [PMID: 7653208]
134. Carulli G, Sbrana S, Minnucci S, et al: Actin polymerization in neutrophils from patients affected by myelodysplastic syndromes—A flow cytometric study. Leuk Res 21:513, 1997. [PMID: 9279362]
135. Nakaseko C, Asai T, Wakita H, et al: Signaling defect in FMLP-induced neutrophil respiratory burst in myelodysplastic syndromes. Br J Haematol 95:482, 1996. [PMID: 8943888]
136. Pamphilon DH, Aparicio SR, Roberts BE, et al: The myelodysplastic syndromes—A study of haemostatic function and platelet ultrastructure. Scand J Haematol 33:486, 1984. [PMID: 6515331]
137. Payne CM, Glasser L: An ultrastructural morphometric analysis of platelet grant and fusion granules. Blood 67:299, 1986. [PMID: 3942827]
138. Rasi V, Lintula R: Platelet-function in the myelodysplastic syndromes. Scand J Haematol 36(Suppl 45):71, 1986.
139. Hamblin TJ: Immunological abnormalities in myelodysplastic syndromes. Semin Hematol 33:150, 1996. [PMID: 8722685]
140. Anderson RW, Volsky DJ, Greenberg B, et al: Lymphocyte abnormalities in preleukemia: I. Decreased NK activity, anomalous immunoregulatory cell subsets and deficient EBV receptors. Leuk Res 7:389, 1983. [PMID: 6310273]
141. Kerndrup G, Meyer K, Ellegaard J, Hokland P: Natural killer (NK)-cell activity and antibody-dependent cellular cytotoxicity (ADCC) in primary preleukemic syndrome. Leuk Res 8:239, 1984. [PMID: 6717064]
142. Takagi S, Kitagawa S, Takeda A, et al: Natural killer–interferon system in patients with preleukaemic states. Br J Haematol 58:71, 1984. [PMID: 6466573]
143. Volsky DJ, Anderson RW: Deficiency in Epstein-Barr virus receptors on B lymphocytes of preleukemia patients. Cancer Res 43:3923, 1983. [PMID: 6602654]
144. Knox SJ, Greenberg BR, Anderson RW, Rosenblatt LS: Studies of T lymphocytes in preleukemic disorders and acute nonlymphocytic leukemia: In vitro radiosensitivity, mitogenic responsiveness, colony formation, and enumeration of lymphocytic subpopulations. Blood 61:449, 1983. [PMID: 6600631]
145. Baumann MA, Milson TJ, Patrick CW, et al: Immunoregulatory abnormalities in myelodysplastic disorders. Am J Hematol 22:17, 1986. [PMID: 3953564]
146. Economopoulos T, Economidou J, Giannopoulos G, et al: Immune abnormalities in myelodysplastic syndromes. J Clin Pathol 38:908, 1985. [PMID: 3928701]
147. Mufti GJ, Figes A, Hamblin TJ, et al: Immunological abnormalities in myelodysplastic syndromes. Br J Haematol 63:143, 1986. [PMID: 3707859]
148. Gatto S, Ball G, Onida F, et al: Contribution of -2 microglobulin levels to the prognostic stratification of survival in patients with myelodysplastic syndrome (MDS). Blood 102:1622, 2003. [PMID: 12738677]
149. Takagi S, Tanaka O, Miura Y: Magnetic resonance imaging of femoral marrow in patients with myelodysplastic syndromes or leukemia. Blood 86:316, 1995. [PMID: 7795239]
150. Delacretaz F, Schmidt PM, Piguet D, et al: Histopathology and myelodysplastic syndromes: The FAB classification (proposals) applied to bone marrow biopsy. Am J Clin Pathol 87:180, 1987. [PMID: 3812349]
151. Yue G, Hao S, Fadare O, et al: Hypocellularity in myelodysplastic syndrome is an independent factor which predicts a favorable outcome. Leuk Res 32:553, 2008. [PMID: 17888511]
152. Fohlmeister I, Fischer R, Modder B, et al: Aplastic anemia and hypocellular myelodysplastic syndrome. J Clin Pathol 38:1218, 1985. [PMID: 4066981]
153. Kuriyama K, Tomonaga M, Matsuo T, et al: Diagnostic significance of pseudo Pelger Huët anomalies and micro-megakaryocytes in myelodysplastic syndrome. Br J Haematol 63:665, 1986. [PMID: 3460627]
154. Tricot G, DeWolf-Peeters C, Vlietinck R, Verwilghen RL: Bone marrow histology in myelodysplastic syndromes. II. Prognostic value of ALIP in MDS. Br J Haematol 58:217, 1984. [PMID: 6477833]
155. Mangi MH, Mufti GJ: Primary myelodysplastic syndromes: Diagnostic and prognostic significance of immunohistochemical assessment of bone marrow biopsies. Blood 79:198, 1992. [PMID: 1370203]
156. Della Porta MG, Malcovati L, Boveri E, et al: Clinical relevance of bone marrow fibrosis and cd34-positive cell clusters in primary myelodysplastic syndromes. J Clin Oncol 27:754, 2009.
157. Bellamy WT, Richter L, Sirjani D, et al: Vascular endothelial cell growth factor (VEGF) is an autocrine promoter of abnormal localized immature precursors (ALIP) and leukemia progenitor formation in myelodysplastic syndromes. Blood 97:1427, 2001. [PMID: 11222390]
158. Matsushima T, Handa H, Yokohama A, et al: Prevalence and clinical characteristics of myelodysplastic syndrome with bone marrow eosinophilia or basophilia. Blood 101:3386, 2003. [PMID: 12506028]
159. Smith WB, Ablin A, Goodman JR, Brecher J: Atypical megakaryocytes in the preleukemic phase of AML. Blood 42:535, 1973. [PMID: 4521565]
160. Queisser W, Queisser U, Ansmann M, et al: Megakaryocyte polyploidization in acute leukemia and preleukemia. Br J Haematol 28:261, 1974. [PMID: 4139968]
161. Bartl R, Frisch B, Baumgart R: Morphologic classification of the myelodysplastic syndromes (MDS): Combined utilization of bone marrow aspirates and trephine biopsies. Leuk Res 16:15, 1992. [PMID: 1732665]
162. Maschek H, Georgii A, Kaloutsi V, et al: Myelofibrosis in primary myelodysplastic syndromes: A retrospective study of 352 patients. Eur J Haematol 148:208, 1992.
163. Moehler TM, Ho AD, Goldschmidt H, Barlogie B: Angiogenesis in hematological malignancies. Crit Rev Oncol Hematol 45:227, 2003. [PMID: 12633837]
164. Ribatti D, Polimeno G, Vacca A, et al: Correlation of bone marrow angiogenesis and mast cells with tryptase activity in myelodysplastic syndromes. Leukemia 16:1680, 2002. [PMID: 12200681]
165. Della Porta MG, Malcovati L, Rigolin GM, et al: Immunophenotypic, cytogenetic and functional characterization of circulating endothelial cells in myelodysplastic syndromes. Leukemia 22:530, 2008.
166. Greenberg PL: Biologic and clinical implications of marrow culture studies in the myelodysplastic syndromes. Semin Hematol 33:163, 1996. [PMID: 8722686]
167. Chui DHK, Clarke BJ: Abnormal erythroid progenitor cells in human preleukemia. Blood 60:362, 1982. [PMID: 6980031]
168. Senn JS, Messner HA, Pinkerton PH, et al: Peripheral blood blast cell progenitors in human preleukemia. Blood 59:106, 1982. [PMID: 7053757]
169. Juvonen E, Partanen S, Knuutila S, Ruutu T: Megakaryocyte colony formation by bone marrow progenitors in myelodysplastic syndrome. Br J Haematol 63:331, 1986. [PMID: 3718873]
170. Lidbeck J: In vitro colony and cluster growth haemopoietic dysplasia (the preleukaemic syndrome): I. Clinical correlations. Scand J Haematol 24:412, 1980.
171. Raymakers R, DeWitte T, Joziasse J, et al: In vitro growth pattern and differentiation predict for progression of myelodysplastic syndromes to acute nonlymphocytic leukemia. Br J Haematol 78:35, 1991. [PMID: 2043479]
172. Konwalinka G, Peschel C, Schmalzl F, et al: CFU-GM assay, cytochemical and electron microscopic studies in agar in patients with preleukemia syndrome and aplastic anemia. Int J Cell Cloning 3:367, 1985. [PMID: 4067359]
173. Cambier N, Baruchel A, Schlageter MH, et al: Chronic myelomonocytic leukemia: From biology to therapy. Hematol Cell Ther 39:41, 1997. [PMID: 9168299]
174. Aul C, Gatterman N, Schneider W: Comparison of in vitro growth characteristics of blast cell progenitors (CFU-BL) in patients with myelodysplastic syndromes and acute myeloid leukemia. Blood 80:625, 1992. [PMID: 1638020]
175. Flores-Figueroa E, Gutierrez-Espindola G, Guerrero-Rivera S, et al: Hematopoietic progenitor cells from patients with myelodysplastic syndromes: In vitro colony growth and long-term proliferation. Leuk Res 23:385, 1999. [PMID: 10229325]
176. Sato T, Kim S, Selleri C, et al: Measurement of secondary colony formation after 5 weeks in long-term cultures in patients with myelodysplastic syndrome. Leukemia 12:1187, 1998. [PMID: 9697872]
177. Aizawa S, Nakano M, Iwase O, et al: Bone marrow stroma from refractory anemia of myelodysplastic syndrome is defective in its ability to support normal CD34-positive cell proliferation and differentiation in vitro. Leuk Res 23:239, 1999. [PMID: 10071075]
178. Janowska-Wieczorek A, Bilch AR, Jacobs A: Increased circulating colony-stimulating factor-1 in patients with preleukemia, leukemia, and lymphoid malignancies. Blood 77:1796, 1991. [PMID: 2015402]
179. Verhoef GEG, DeSchouder P, Ceuppens JL: Measurement of serum cytokine levels in patients with myelodysplastic syndromes. Leukemia 6:1268, 1992. [PMID: 1280751]
180. Bowen D, Yancik S, Bennett L, et al: Serum stem cell factor concentration in patients with myelodysplastic syndromes. Br J Haematol 85:63, 1993. [PMID: 7504511]
181. Zwierzina H, Anderson JE, Rollinger-Holzinger I, et al: Endogenous FLT-3 ligand serum levels are associated with disease stage in patients with myelodysplastic syndromes. Leukemia 13:553, 1999. [PMID: 10214861]
182. Tamura H, Ogata K, Luo S, et al: Plasma thrombopoietin (TPO) levels and expression of TPO receptor on platelets in patients with myelodysplastic syndromes. Br J Haematol 103:778, 1998. [PMID: 9858230]
183. Mecucci C, La Starza R: Cytogenetics of myelodysplastic syndromes. Forum (Genova) 9:4, 1999. [PMID: 10101207]
184. Haase D: Cytogenetic features in myelodysplastic syndromes. Ann Hematol 87:515, 2008. [PMID: 18414863]
185. Solé F, Luño E, Sanzo C, et al: Identification of novel cytogenetic markers with prognostic significance in a series of 968 patients with primary myelodysplastic syndromes. Haematologica 90:1168, 2005. [PMID: 20177619]
186. Estey E, Trujillo JM, Cork A, et al: AML-associated cytogenetic abnormalities inv ((16), del (16), t(8;21)) in patients with myelodysplastic syndromes. Hematol Pathol 6:43, 1992. [PMID: 1601822]
187. Block AW, Carroll AJ, Hagemeijer A, et al: Rare recurring balanced chromosome abnormalities in therapy-related myelodysplastic syndromes and acute leukemia. Genes Chromosomes Cancer 33:401, 2002. [PMID: 11921274]
188. Rossi G, Pelizzari AM, Bellotti D, et al: Cytogenetic analogy between myelodysplastic syndrome and acute myeloid leukemia of elderly patients. Leukemia 14:636, 2000. [PMID: 10764149]
189. Zhang L, Rothman N, Wang Y, et al: Increased aneusomy and long arm deletion of chromosomes 5 and 7 in the lymphocytes of Chinese workers exposed to benzene. Carcinogenesis 19:1955, 1998. [PMID: 9855009]
190. Olney HJ, Le Beau MM: The cytogenetics and molecular biology of myelodysplastic syndromes, in The Myelodysplastic Syndromes, edited by JM Bennett, p 89. Marcel Dekker, New York, 2002.
191. Wong AK, Fang B, Zhang L, et al: Loss of the Y chromosome: An age-related or clonal phenomenon in acute myelogenous leukemia/myelodysplastic syndrome? Arch Pathol Lab Med 132:1329, 2008. [PMID: 18684036]
192. Xie D, Hofmann W-K, Mori N, et al: Allelotype analysis of the myelodysplastic syndrome. Leukemia 14:805, 2000. [PMID: 10803510]
193. Hoffman W-K, De Vos S, Komor M, et al: Characterization of gene expression of CD34+ cells from normal and myelodysplastic bone marrow. Blood 100:3553, 2002.
194. Bernasconi P: Molecular pathways in myelodysplastic syndromes and acute myeloid leukemia: Relationships and distinctions: A review. Br J Haematol 142:695, 2008. [PMID: 18540941]
195. Cesana C, Klersy C, Brando B, et al: Prognostic value of circulating CD34+ cells in myelodysplastic syndromes. Leuk Res 32:1715, 2008. [PMID: 18456323]
196. Nilsson L, EdÈn P, Olsson E, et al: The molecular signature of MDS stem cells supports a stem-cell origin of 5q myelodysplastic syndromes. Blood 110:3005, 2007. [PMID: 17616640]
197. Van den Berghe H, Cassiman JJ, David G, et al: Distinct haematological disorder with deletion of long arm of no. 5 chromosome. Nature 251:437, 1974.
198. Boultwood J, Lewis S, Wainscoat JS, et al: The 5q– syndrome. Blood 84:3253, 1994. [PMID: 7949083]
199. Washington LT, Doherty D, Glassman A, et al: Myeloid disorders with deletion 5q as the sole karyotypic abnormality: The clinical spectrum and pathological spectrum. Leuk Lymphoma 43:761, 2002. [PMID: 12153162]
200. Van den Berghe H, Michaux L: 5q-, twenty-five years later: A synopsis. Cancer Genet Cytogenet 94:1, 1997.
201. Bigoni R, Cuneo A, Milani R, et al: Multilineage involvement in the 5q– syndrome: A fluorescent in situ hybridization study on bone marrow smears. Haematologica 86:375, 2001. [PMID: 11325642]
202. Anderson K, Arvidsson I, Jacobsson B, Hast R: Fluorescence in situ hybridization for the study of cell lineage involvement in myelodysplastic syndromes with chromosome 5 anomalies. Cancer Genet Cytogenet 136:101, 2002. [PMID: 12237232]
203. Jaju RJ, Jones M, Boultwood J, et al: Combined immunophenotyping and FISH identifies the involvement of B-C cells in 5q– syndrome. Genes Chromosomes Cancer 29:276, 2000. [PMID: 10992302]
204. Nilsson L, Astrand-Grundastrom I, Arvidsson I, et al: Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: Evidence for involvement at the hematopoietic stem cell level. Blood 96:2012, 2000. [PMID: 10979941]
205. Cermak J, Michalova K, Brezinova J, Zemanova Z: A prognostic impact of separation of refractory cytopenia with multilineage dysplasia and 5q– syndrome from refractory anemia in primary myelodysplastic syndrome. Leuk Res 27:221, 2003. [PMID: 12537974]
206. Giagounidis AAN, Germing U, Haase S, et al: Clinical, morphological, cytogenetics, and prognostic features of patients with myelodysplastic syndrome and del(5q) including band q31. Leukemia 18:113, 2004. [PMID: 14586479]
207. Washington LT, Dherty D, Glassman A, et al: Myeloid disorders with deletion of 5q as sole karyotypic abnormality: The clinical and pathological spectrum. Leuk Lymphoma 43:761, 2002. [PMID: 12153162]
208. Pedersen B: Anatomy of the 5q– deletion: Different sex ratios and deleted 5q bands in MDS and AML. Leukemia 10:1883, 1996. [PMID: 8946926]
209. Mohamedali A, Mufti GJ: Van-den Berghe’s 5q– syndrome in 2008. Br J Haematol 144:157, 2009. [PMID: 19016715]
210. Ebert BL, Pretz J, Bosco J, et al: Identification of RPS14 as a 5q– syndrome gene by RNA interference screen. Nature 451:335, 2008. [PMID: 18202658]
211. Mauro VP, Edelman GM: The ribosome filter redux. Cell Cycle 6:2246, 2007. [PMID: 17890902]
212. Pellagatti A, Hellström-Lindberg E, Giagounidis A, et al: Haploinsufficiency of RPS14 in 5q– syndrome is associated with deregulation of ribosomal- and translation-related genes. Br J Haematol 142:57, 2008. [PMID: 18477045]
213. Joslin JM, Fernald AA, Tennant TR, et al: Haploinsufficiency of EGR1, a candidate gene in the del(5q), leads to the development of myeloid disorders. Blood 110:719, 2007. [PMID: 17420284]
214. Kelaidi C, Eclache V, Fenaux P: The role of lenalidomide in the management of myelodysplasia with del 5q. Br J Haematol 140:267, 2008. [PMID: 18217896]
215. Melchert M, Kale V, List A: The role of lenalidomide in the treatment of patients with chromosome 5q deletion and other myelodysplastic syndromes. Curr Opin Hematol 14:123, 2007. [PMID: 17255789]
216. Mohr B, Oelschlaegel U, Thiede C, et al; The response to lenalidomide of myelodysplastic syndrome patients with deletion del(5q) can be sequentially monitored in CD34+ progenitor cells. Haematologica 94:430, 2009. [PMID: 19181778]
217. Bernstein R, Philip P, Ueshima Y: Fourth international workshop on chromosomes in leukemia 1982. Abnormalities of chromosome 7 resulting in monosomy 7 or in deletion of the long arm (7q–): Review of translocations, breakpoints, and associated abnormalities. Cancer Genet Cytogenet 11:300, 1984. [PMID: 6704933]
218. Michiels JJ, Mallios-Zorbala H, Prins MEF, et al: Simple monosomy 7 and myelodysplastic syndrome in thirteen patients without previous cytostatic treatment. Br J Haematol 64:425, 1986. [PMID: 3466639]
219. Pasquali F, Bernasconi P, Cosalone R, et al: Pathogenetic significance of “pure” monosomy 7 in myeloproliferative disorders. Analysis of 14 cases. Hum Genet 62:40, 1982. [PMID: 6961098]
220. Kardos G, Baumann I, Passmore SJ, et al: Refractory anemia in childhood: A retrospective analysis of 67 patients with particular reference to monosomy 7. Blood 102:1997, 2003. [PMID: 12763938]
221. Brozek I, Babinska M, Kardas I, et al: Cytogenetic analysis and clinical significance of chromosome 7 aberrations in acute leukemia. J Appl Genet 44:401, 2003. [PMID: 12923315]
222. Dohner K, Brown J, Hehmann U, et al: Molecular cytogenetic characterization of a critical region in bands 7q35-q36 commonly deleted in malignant myeloid disorders. Blood 92:4031, 1998. [PMID: 9834205]
223. Sessarego M, Fugazza G, Gobbi M, et al: Complex structural involvement of chromosome 7 in primary myelodysplastic syndromes determined by fluorescence in situ hybridization. Cancer Genet Cytogenet 106:110, 1998. [PMID: 9797774]
224. Hayashi Y, Egushi M, Sugita K, et al: Cytogenetic findings and clinical features in acute leukemia and transient myeloproliferation disorder in Down’s syndrome. Blood 72:15, 1988. [PMID: 2968822]
225. Berger R, LeConiat M, Schaison G: Chromosome abnormalities in bone marrow of Fanconi anemia patients. Cancer Genet Cytogenet 65:47, 1993. [PMID: 8431915]
226. Minelli A, Maserati E, Giudici G, et al: Familial partial monosomy 7 and myelodysplasia: Different parental origin of monosomy 7 suggests action of a mutator gene. Cancer Genet Cytogenet 124:147, 2001. [PMID: 11172908]
227. Smadja N, Krulik M, DeGramont A, et al: Translocation 1;7 in preleukemic states. Cancer Genet Cytogenet 18:189, 1985. [PMID: 4052979]
228. Woloschak GF, Dewald GW, Gahn RS, et al: Amplification of RNA and DNA specific for erb B in unbalanced 1;7 chromosomal translocation associated with myelodysplastic syndrome. J Cell Biochem 32:23, 1986. [PMID: 3464613]
229. Ueda H, Tashiro S, Kojima S, et al: Instability of chromosome 7 in colony forming cells of patients with aplastic anemia. Int J Hematol 70:13, 1999. [PMID: 10446489]
230. Kaito K, Kobayashi M, Katayama T, et al: Long-term administration of G-CSF for aplastic anemia is closely related too the early evolution of monosomy 7 MDS in adults. Br J Haematol 103:297, 1998. [PMID: 9827895]
231. Bjorkman SE: Chronic refractory anemia with sideroblastic bone marrow. A study of four cases. Blood 11:250, 1956. [PMID: 13293201]
232. Kushner JP, Lee GR, Wintrobe MM, et al: Idiopathic refractory sideroblastic anemia. Medicine (Baltimore) 50:139, 1971. [PMID: 5286053]
233. Boultwood J, Pellagatti A, Nikpour M, et al: The role of the iron transporter ABCB7 in refractory anemia with ring sideroblasts. PLoS ONE 3:e1970, 2008.
234. Chang KL, O’Donnell MR, Slovak ML, et al: Primary myelodysplasia occurring in adults under 50 years old: A clinicopathologic study of 52 patients. Leukemia 16:623, 2002. [PMID: 11960342]
235. Kardos G, Veerman AJ, De Waal FC, et al: Familial sideroblastic anemia with emergence of monosomy 5 and myelodysplastic syndrome. Med Pediatr Oncol 26:54, 1996. [PMID: 7494512]
236. Garand R, Gardars J, Bizet M, et al: Heterogeneity of acquired idiopathic sideroblastic anemia (AISA). Leuk Res 16:463, 1992. [PMID: 1625471]
237. Bowen DT, Jacobs A: Primary acquired sideroblastic erythropoiesis in non-anaemic and minimally anaemic subjects. J Clin Pathol 42:56, 1989. [PMID: 2921346]
238. Antilla P, Thalainen J, Salo A, et al: Idiopathic macrocytic anaemia in the aged: Molecular and cytogenetic findings. Br J Haematol 90:797, 1995.
239. Steensma DP, Dewald GW, Hodnfield JM, et al: Clonal cytogenetic abnormalities in bone marrow specimens without clear morphologic evidence of dysplasia: A form fruste of myelodysplasia? Leuk Res 27:235, 2003. [PMID: 12537976]
240. Yoshida Y, Oguma S, Tohyama K, et al: Diagnostic and biological significance of sideroblastic erythropoiesis in the myelodysplastic syndromes. Int J Hematol 67:137, 1998. [PMID: 9631580]
241. Beris PH, Graf J, Miescher PA: Primary acquired sideroblastic and primary acquired refractory anemia. Semin Hematol 20:101, 1983. [PMID: 6348954]
242. Mecucci C, Van Orshoven A, Vermaelen K, et al: 11q– chromosome is associated with abnormal iron stores in myelodysplastic syndromes. Cancer Genet Cytogenet 27:39, 1987. [PMID: 3472648]
243. Parlier V, Van Melle G, Beris PH, et al: Hematological, clinical, and cytogenetic analysis in 109 in patients with primary myelodysplastic syndrome. Cancer Genet Cytogenet 78:219, 1994. [PMID: 7828157]
244. Berrebi A, Bruck R, Shtalrid M, Chemke J: Philadelphia chromosome in idiopathic acquired sideroblastic anemia. Acta Haematol 72:343, 1984. [PMID: 6441415]
245. Carroll AJ, Poon M-C, Robinson NC, Christ WM: Sideroblastic anemia associated with thrombocytosis and a chromosome 3 abnormality. Cancer Genet Cytogenet 22:183, 1986. [PMID: 3458520]
246. Bennett DD, Stanley WS, Johnson CB: Combined phenotypic and genotypic analysis of ringed sideroblasts in acquired idiopathic sideroblastic anemia. Acta Haematol 73:235, 1985. [PMID: 3933247]
247. DeWald GW, Brecher M, Travis LB, Stupea PJ: Twenty-six patients with hematologic disorders and X-chromosome abnormalities. Cancer Genet Cytogenet 42:173, 1989. [PMID: 2790752]
248. Remacha AF, Nomdedéu JF, Puget G, et al: Occurrence of the JAK2 V617F mutation in the WHO provisional entity: Myelodysplastic/myeloproliferative disease, unclassifiable-refractory anemia with ringed sideroblasts associated with marked thrombocytosis. Haematologica 91:719, 2006. [PMID: 16670082]
249. Szpurka H, Tiu R, Murugesan G, et al: Refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T), another myeloproliferative condition characterized by JAK2 V617F mutation. Blood 108:2173, 2006. [PMID: 16741247]
250. Schmitt-Graeff AH, Teo SS, Olschewski M, et al: JAK2V617F mutation status identifies subtypes of refractory anemia with ringed sideroblasts associated with marked thrombocytosis. Haematologica 93:34, 2008. [PMID: 18166783]
251. Chabannori G, Molina L, Pegouri-Bandelier B, et al: A review of 76 patients with myelodysplastic syndromes treated with danazol. Cancer 73:3073, 1994.
252. Chan G, DiVenuti G, Miller K: Danazol for the treatment of thrombocytopenia in patients with myelodysplastic syndrome. Am J Hematol 71:166, 2002. [PMID: 12410570]
253. Sadek I, Zayed E, Hayne O, Fernandez L: Prolonged complete remission of myelodysplastic syndrome treated with danazol, retinoic acid and low-dose prednisone. Am J Hematol 64:306, 2000. [PMID: 10911385]
254. Dreyfus F: The deleterious effects of iron overload in patients with myelodysplastic syndromes. Blood Rev 22(Suppl 2):S29, 2008.
255. Steensma DP: Myelodysplasia paranoia: Iron as the new radon. Leuk Res 33:1158, 2009. [PMID: 19036443]
256. Messa E, Cilloni D, Messa F, et al: Deferasirox treatment improved the hemoglobin level and decreased transfusion requirements in four patients with the myelodysplastic syndrome and primary myelofibrosis. Acta Haematol 120:70, 2008. [PMID: 18827475]
257. Bennett JM: MDS Foundation’s Working Group on Transfusional Iron Overload. Consensus statement on iron overload in myelodysplastic syndromes. Am J Hematol 83:858, 2008. [PMID: 18767130]
258. Yang LP, Keam SJ, Keating GM: Deferasirox: A review of its use in the management of transfusional chronic iron overload. Drugs 67:2211, 2007. [PMID: 17927285]
259. Hellström-Lindberg E, Gulbrandsen N, Lindberg G, et al: A validated decision model for treating the anaemia of myelodysplastic syndromes with erythropoietin + granulocyte colony-stimulating factor: Significant effects on quality of life. Br J Haematol 120:1037, 2003. [PMID: 11415271]
260. Casadevall N, Durieux P, Dubois S, et al: Health, economic, and quality-of-life effects of erythropoietin and granulocyte colony-stimulating factor for the treatment of myelodysplastic syndromes: A randomized, controlled trial. Blood 104:321, 2004. [PMID: 15054036]
261. Balleari E, Rossi E, Clavio M, et al: Erythropoietin plus granulocyte colony-stimulating factor is better than erythropoietin alone to treat anemia in low-risk myelodysplastic syndromes: Results from a randomized single-centre study. Ann Hematol 85:174, 2006. [PMID: 16408206]
262. Jädersten M, Malcovati L, Dybedal I, et al: Erythropoietin and granulocyte-colony stimulating factor treatment associated with improved survival in myelodysplastic syndrome. J Clin Oncol 26:3607, 2008. [PMID: 20177949]
263. Silverman LR, McKenzie DR, Peterson BL, et al: Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: Studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B. J Clin Oncol 24:3895, 2006. [PMID: 16921040]
264. Müller-Thomas C, Schuster T, Peschel C, Götze KS: A limited number of 5-azacitidine cycles can be effective treatment in MDS. Ann Hematol 88:213, 2009. [PMID: 20178103]
265. Kaminskas E, Farrell A, Abraham S, et al: Approval summary: Azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 11:3604, 2005. [PMID: 15897554]
266. Gore SD, Hermes-DeSantis ER: Future directions in myelodysplastic syndrome: Newer agents and the role of combination approaches. Cancer Control 15(Suppl):40, 2008.
267. Kornblith AB, Herndon JE II, Silverman EP, et al: Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized phase III trial. J Clin Oncol 15:2441, 2002.
268. Cazzola M, Barosi G, Gobbi PG, et al: Natural history of idiopathic refractory sideroblastic anemia. Blood 71:305, 1988. [PMID: 3337899]
269. Lewy RI, Kansu E, Gabuzda T: Leukemia in patients with acquired idiopathic sideroblastic anemia. Am J Hematol 6:323, 1979. [PMID: 294823]
270. Cheng DS, Kushner JP, Wintrobe MM: Idiopathic refractory sideroblastic anemia. Incidence and risk factors for leukemic transformation. Cancer 44:724, 1979. [PMID: 383260]
271. Hast R, Reizenstein P: Sideroblastic anemia and development of leukemia. Blut 42:203, 1981. [PMID: 7225579]