Woman and man using natural therapies for myelodysplastic syndromes (mds)

Myelodysplastic Syndromes (MDS)

Myelodysplastic Syndromes (MDS)

Last Section Update: 11/2019

Contributor(s): Shayna Sandhaus, PhD

1 Introduction

Summary and Quick Facts for Myelodysplastic Syndromes

  • Myelodysplastic syndromes (MDS) encompass a group of disorders caused by a disruption in the production of blood cells. Risk of developing another blood disorder called acute myeloid leukemia (AML) increases with MDS, so early diagnosis and treatment is very important.
  • This protocol reviews how MDS develops, types of MDS, and how these conditions are typically diagnosed and treated. Natural therapies that may complement conventional MDS treatments will be summarized as well.
  • Supplementation with several natural compounds, including vitamin K2, maitake-derived beta-glucan, and green tea extracts may help keep blood cells healthy and complement conventional treatment for people with MDS.

Myelodysplastic syndromes (MDS) are a group of cancers that prevent your bone marrow from producing enough healthy blood cells. MDS are not necessarily terminal diseases, but they are not easily curable either. Depending on how progressive your MDS is, you may not immediately need aggressive treatment. In some cases, MDS progress into a form of leukemia. A bone marrow transplant is the only treatment that has potential to cure MDS, though it is a risky procedure and not appropriate for everyone.1,2

MDS encompass a group of varied but related diseases. The kind you develop depends on both genetic as well as epigenetic phenomena that remain poorly characterized to date. Depending on the type of MDS you have, you may develop deficiencies in red blood cells, white blood cells, and/or platelets. Your symptoms will depend on which type of blood cell(s) your bone marrow is not producing correctly. Extreme tiredness, shortness of breath, easy bleeding, or frequent infections are common, though it is also not unusual for some patients not to experience symptoms, especially in the early stages of MDS.2

There are as many as 170,000 people living with MDS in the United States, where an average of 33‒55 people are diagnosed every day. While MDS can affect people of all ages, 75% of patients with MDS are older than 60 years. One in three patients with MDS eventually develop another blood disorder called acute myeloid leukemia (AML). Regrettably, the development of AML in the setting of pre-existing MDS is difficult to treat. The average survival is approximately five months for high-risk MDS patients and up to six years for lower-risk patients who do not receive a bone marrow transplant.3,4 Allogeneic bone marrow transplant has the potential to significantly prolong overall survival in select patients, but quality of life is frequently compromised.

In this protocol, you will learn how MDS develop and which factors increase risk. You will also learn how MDS are typically diagnosed and treated. You will discover several emerging treatment strategies currently being pursued in research settings and learn how to find ongoing clinical trials in which you may be able to participate. In addition, you will learn about various dietary and lifestyle considerations, as well as natural interventions, that may be supportive in cases of MDS.

2 Historical Background

In 1900, physicians first began describing patients with what are now regarded as telltale signs of MDS: severe shortages in different types of blood cells, cells that do not fully mature or that develop abnormally, and bone marrow that is overcrowded with cells that never leave to enter the bloodstream.5 MDS were first noted in the medical literature in the 1930s, where they were described as pre-leukemic conditions. Historically, these disorders have been referred to as refractory anemia, oligoblastic anemia, myelodysplastic anemia, preleukemia, and smoldering leukemia. MDS were not regarded as separate, distinct disorders until 1976.6,7

MDS were first classified in a 1982 publication by the French-American-British group.5,7 This system developed subgroups based on the condition of blood and bone marrow, as well as the presence of specific abnormalities. During the 1990s, the importance of other clinical variables was recognized. In particular, chromosome analysis largely contributed to improved clinical outcomes.5

In 1997, patient cohorts were gathered from the United States, Europe, and Japan and reviewed thoroughly to create the database for the International Prognostic Scoring System (IPSS).5,7 This was the first widely used classification system for MDS. Based on the blood cell counts, percentages of bone marrow cells, and genetic risk profile, the IPSS was used to divide patients into risk categories. This helped identify probabilities of survival and risk of disease progression.5

3 Biology and Development of MDS

Blood Cells and Genes

Blood consists of fluid, which is mainly water, and a combination of three main cell types: platelets that clot to control bleeding, red blood cells that deliver oxygen throughout the body, and white blood cells, which are part of the immune system.2 Blood cells grow and develop within bone marrow, the spongey tissue inside the center of most bones. Immature cells with the ability to turn into mature, functional cells are called stem cells. Immature blood cells are referred to as hematopoietic stem cells, which develop into young blood cells called blasts. Blasts mature into platelets, red blood cells, and white blood cells before leaving the bone marrow and entering the bloodstream.

All cells in the body contain genes, which are instructions for how cells function and build new cells.2 Genes are stored in the form of deoxyribonucleic acid (DNA) and organized into long strands called chromosomes. Changes in genes, which are referred to as mutations, have the possibility of turning normal cells into cancer cells.

4 MDS Basics

Blood stem cells are direct precursors for the maturation and function of mature blood cells.2,8 With MDS, blood stem cells do not generate enough new blood cells and make blood cells that are defective. These cells have abnormal size, shape, and appearance, which is called dysplasia. They do not mature into normal, healthy blood cells; often do not leave the bone marrow; and may die early either in the bone marrow or shortly after entering the bloodstream. This results in having low numbers of one or more types of blood cells in the bloodstream, referred to as cytopenia.

5 Symptoms of MDS

Most symptoms of MDS are due to a lack of healthy platelets, red blood cells, or white blood cells.2,8 Anemia occurs due to a shortage of healthy red blood cells and can cause fatigue, weakness, dizziness, shortness of breath, and pale skin. With low amounts of white blood cells, referred to as neutropenia, patients may experience frequent or severe infections. Having low numbers of platelets, which is called thrombocytopenia, can cause bruising, bleeding, and make it more difficult for wounds to heal.

In some patients, MDS are slow-developing conditions and some patients do not experience symptoms early in the course of the disease.2,8 In other patients, defective blood stem cells and blasts can accumulate and overcrowd the bone marrow. In certain patients, MDS may progress into a fast-growing cancer called acute myeloid leukemia (AML).2,8 In the past, MDS were considered “pre-leukemic” conditions, but evidence clearly indicates that not all patients with MDS necessarily develop AML, which occurs roughly about one-third of the time.

6 Causes and Risk Factors

Causes for MDS are generally unknown, and little is known about what contributing factors may cause the gene mutations that lead to disease. However, although research is ongoing, evidence suggests several potential risk factors.

Environmental and Therapeutic Exposure

Studies have shown that chemotherapies, particularly a class called alkylating agents, and radiation therapy used to treat previous cancers could possibly lead to MDS.9-12 Occupational exposure to certain other chemicals, like benzene13,14 or pesticides,15-17 have also been associated with higher risk for MDS.

Modifiable Lifestyle Considerations

In a review18 of available small studies, smoking13,19-22 and alcohol21,23 use were each evaluated as potential risk factors for MDS in four studies. Body mass index (BMI)21,24 and anemia were each evaluated in two studies. A BMI of 30 kg/m2 or greater more than doubled risk of MDS in a study of both men and women in the United States.21 History of autoimmune diseases, certain infectious diseases, anemia, and use of anti-tuberculosis drugs were associated with increased risk of MDS as well. Conversely, vigorous physical activity and tea and dietary isoflavone intake were associated with lower MDS risk.18

7 Types of MDS

MDS are commonly organized into subtypes using a classification system developed by the World Health Organization (WHO) and National Comprehensive Cancer Network (NCCN) guidelines.8,25 The 2017 WHO Revision contains additional subtypes in a category for myelodysplastic syndromes/myeloproliferative neoplasms (MDS/MPN).8,26 Factors used to determine MDS and MDS/MPN subtypes include the types of cytopenias and/or dysplasias, amount of blasts in the blood and bone marrow, type of gene mutations found in the bone marrow, and presence of defective red blood cells that contain too much iron, called sideroblasts.

Definitions of MDS Subtypes According to WHO/NCCN*

Subtype

Blood

Bone Marrow

MDS with single lineage dysplasia (MDS-SLD)

Cytopenia in 1‒2 types of blood cells

Dysplasia in 1‒3 types of blood cells; under 5% blasts

MDS with multilineage dysplasia (MDS-MLD)

Cytopenia in 1‒3 types of blood cells

Dysplasia in 2‒3 types of blood cells; under 15% sideroblasts; under 5% blasts

MDS with ring sideroblasts (MDS-RS)

Anemia; no blasts

15% or more sideroblasts

MDS with excess blasts 1 (MDS-EB1)

Cytopenia in 1‒3 types of blood cells; blasts 4% or less; low monocytesα

Dysplasia in 1‒3 types of blood cells; 5‒9% blasts; no Auer rodsß

MDS with excess blasts 2 (MDS-EB2)

Cytopenia in 1‒3 types of blood cells; 5‒19% blasts; low monocytesα

Dysplasia in 1‒3 types of blood cells; 10‒19% blasts; ± Auer rodsß

MDS, unclassifiable (MDS-U)

Cytopenia in 2‒3 types of blood cells; ± 1% blasts

Dysplasia in 0‒1 type of blood cell; MDS gene mutations present; under 5% blasts

MDS with isolated del(5q)d

Anemia; platelets normal or increased

Dysplasia in 1 type of blood cell; isolated del(5q)d; under 5% blasts; ± one other mutation except -7d or del(7q)d

Refractory cytopenia of childhood (Provisional WHO category)

Cytopenia in 2‒3 types of blood cells; under 2% blasts

Dysplasia in 1‒3 types of blood cells; under 5% blasts

Definitions of MDS/MPN Subtypes According to WHO/NCCN*

Subtype

Blood

Bone Marrow

Chronic myelomonocytic leukemia (CMML)-0

Elevated monocytesα; under 2% blasts

Dysplasia in 1‒3 types of blood cells; under 5% blasts

CMML-1

Elevated monocytesα; 2‒4% blasts

Dysplasia in 1‒3 types of blood cells; 5‒9% blasts

CMML-2

Elevated monocytesα; 5‒19% blasts or Auer rodsß

Dysplasia in 1‒3 types of blood cells; 10‒19% blasts or Auer rodsß

Atypical chronic myeloid leukemia (aCML), BCR-ABL negative

Elevated white blood cells; neutrophilsα over 10%; under 20% blasts; improper granulocyteα development

Excessive accumulation and overcrowding of cells; under 20% blasts

Juvenile myelomonocytic leukemia (JMML)

Elevated monocytesα; under 20% blasts; increased fetal hemoglobin

Elevated monocytesα; under 20% blasts; no Philadelphia chromosomed; GM-CSF hypersensitivity

MDS/MPN, unclassifiable (“Overlap Syndrome”)

Dysplasia + myeloproliferative features and no prior MDS or MPN

Dysplasia + myeloproliferative features

MDS/MPN with ring sideroblasts and thrombocytosis
(MDS-MPN-RS-T)

Dysplasia + myeloproliferative features; elevated platelets; 15% or more sideroblasts

Dysplasia + myeloproliferative features

*To be interpreted with the help of a clinician skilled in the management of MDS. αA type of white blood cell. ßType of abnormal presence found in defective blasts. dType of gene mutation. Over 5% for patients with SF3B1 gene mutations. 5% or more for patients with SF3B1 gene mutations.
GM-CSF, granulocyte-macrophage colony-stimulating factor; MDS, myelodysplastic syndromes; MPN, myeloproliferative overlap neoplasms; NCCN, the National Comprehensive Cancer Network; WHO, World Health Organization

8 Testing for MDS

Tests for MDS involve a complete medical history and physical exam to review symptoms and when they began. Sometimes MDS tests are completed to review unsuspected results from blood tests done for other reasons.2,8 If a person is suspected to have a MDS, tests should be conducted to examine his or her blood and bone marrow for evidence of MDS or a different health condition.2,8,27

Complete Blood Count and Differential

A complete blood count (CBC) measures the number of white blood cells, red blood cells, and platelets. A CBC for MDS includes a differential to measure specific types of white blood cells, like neutrophils, monocytes, and lymphocytes.2,8,27

Reticulocyte Count

Reticulocytes are immature red blood cells. Measuring the number of reticulocytes in the bloodstream reflects on how quickly reticulocytes are being made and released from healthy bone marrow and can also help determine the cause of anemia.2,8,27

Blood Smear

By examining a drop of blood on a microscope slide, blood cells are assessed for size, shape, and maturity to look for abnormalities, or dysplasias. This process also can help count different types of blood cells and check for blast cells in the bloodstream, which are normally only in the bone marrow.2,8,27

Serum EPO

Erythropoietin (EPO) is a hormone made by the kidneys that stimulates bone marrow to make more red blood cells when blood oxygen levels are low. Measuring EPO can help determine the cause of anemia. A low EPO level can itself cause anemia and may be a sign of a different health condition. In patients with known MDS, low EPO levels can worsen anemia.2,28

Iron, Ferritin, Folate, and Vitamin B12

Checking for shortages of certain substances can help determine the cause of anemia. Iron is needed to make hemoglobin, the protein in red blood cells that carries oxygen. Ferritin represents the amount of iron stored in the body. Folate and vitamin B12 are nutrients needed to make red blood cells. Low levels of these can cause anemia and shortages in folate or vitamin B12 can also cause red blood cell dysplasia.8

Thyroid Hormones

Hormones made by the thyroid help control energy usage and can also affect other body functions. High thyroid-stimulating hormone (TSH) levels may indicate an underactive thyroid, which can cause anemia.29

Copper

Copper is a mineral that supports many bodily processes, and low levels can cause low numbers of red blood cells, white blood cells, and have been associated with dysplasia. Copper deficiency could potentially be confused for MDS. Because copper plays a role in mobilizing iron from the liver to tissues throughout the body, copper deficiency can cause iron to accumulate in the liver and not reach other tissues. This can lead to problems with blood cell formation in the bone marrow secondary to lack of iron availability.30 The abnormalities in blood cell production may be mistaken for MDS. Most commonly, copper deficiency manifests as low red blood cell and white blood cell counts. Low copper levels can be detected via blood testing.30 Measuring copper levels is not a standard test for MDS, but it is sometimes needed to rule out other causes of dysplasia.31-35

HIV Testing

Human immunodeficiency virus (HIV), which can cause cytopenia and dysplasia, may need to be ruled out in certain cases.2,27

Flow Cytometry

Flow cytometry is a technique that can be used to measure different characteristics of cells, including their size, count, and type. In some cases of MDS, flow cytometry is used to identify specific types of cells in blood or bone marrow by analyzing proteins on the surface of the cells.36-41

Bone Marrow Biopsy and Aspiration

To confirm a diagnosis of MDS, a small piece of solid bone and a bone marrow sample are biopsied for testing, typically from the hip bone. At the same time, a bone marrow aspiration collects bone marrow from inside the bone. These tests provide specific information on cytopenias, dysplasias, and blast cells from the bone marrow. Whereas in healthy bone marrow less than 5% of the cells are blasts, samples from patients with MDS can show blasts in up to 19% of cells. With AML, more than 20% blasts can be measured in the bone marrow. The samples are also dyed to test for ring sideroblasts, dysfunctional red blood cells that have too much iron.42-46

Genetic Tests

Bone marrow and sometimes blood are examined by cytogenetic testing under a microscope to see the chromosomes inside cells. The chromosomes are detailed into an outline, called a karyotype. Sometimes fluorescence in situ hybridization (FISH)47,48 is also used to study chromosomes. Chromosomes with missing or misplaced parts may provide valuable disease state information. In MDS, it is common to have abnormal chromosomes, though not all patients do.41,49-57

Molecular Testing

More sensitive than karyotyping or FISH, molecular testing in blood and bone marrow detects small defects in genes, known as mutations. Often, a patient with a normal karyotype can have detectable mutations at the molecular level.58-62 Recurrent gene mutations that occur commonly in MDS can be looked for specifically using DNA sequencing.61,63

PDGFRβ Gene Mutation

Changes to the platelet-derived growth factor receptor beta (PDGFRβ) gene occur in a subtype of MDS called chronic myelomonocytic leukemia (CMML). Knowing if cells have a PDGFRβ gene mutation helps guide treatment decisions for some patients.53,57,64-66

9 Prognostic Scoring and Risk Groups

Prognostic Factors

Prognostics predict patterns and outcomes of disease. Certain factors related to blood counts, bone marrow assessment, and karyotype/molecular profile affect the predicted outcome of MDS and help classify the severity of the diseases. An important aspect that affects the prognosis of MDS is the risk of progressing to AML. Other prognostic factors include MDS subtype, number and severity of cytopenias, amount of blasts in the bone marrow, and type and number of chromosome changes.8,58,67,68

Prognostic Scoring and Grouping

Prognostic scoring uses points to rate the severity of MDS in individual patients based on their prognostic factors. Two common scoring systems are the Revised International Prognostic Scoring System (IPSS-R) and the WHO classification-based Prognostic Scoring System (WPSS).69-71

Prognostic factors have different scores based on their severity. When the scores for individual prognostic factors are added together, they provide an overall risk score. A lower total score means a better outlook. The risk score describes how slow or fast MDS will likely grow and progress to AML if not treated. Risk scores are used to assign risk groups that guide decisions on treatment.8,69-72

Prognostic Scoring Systems and Risk Groups*

System

Prognostic Factors Scored

Total Risk Score

Risk Group

IPSS-R

  • Percent of blasts in the bone marrow
    • Score of 0, 1, 2, or 3 points
      • ≤2%: 0 points
      • >2% to <5%: 1 point
      •  5% to 10%: 2 points
      • >10%: 3 points
  • Chromosome changes
    • Score of 0, 1, 2, 3, or 4 points
      • Scores assigned based upon number and type of genetic changes observed through genetic testing
  • Hemoglobin level
    • Score of 0, 1, or 1.5 points
      • ≥10: 0 points
      • 8 to <10: 1 point
      • <8: 1.5 points
  • Platelet count
    • Score of 0, 0.5, or 1 point
      • ≥100: 0 points
      • 50 to <100: 0.5 points
      • <50: 1 point
  • Neutrophil count
    • Score of 0 or 0.5 points
      • ≥0.8: 0 points
      • <0.8: 0.5 points

1.5 points or lower

Very Low

2‒3 points

Low

3.5‒4.5 points

Intermediate

5‒6 points

High

6.5 points or higher

Very High

WPSS

  • MDS subtype
    • Score of 0, 1, 2, or 3 points
      • Determined by clinician based upon WHO classification
  • Chromosome changes
    • Score of 0, 1, or 2 points
      • Scores assigned based upon number and type of genetic changes observed through genetic testing
  • Presence of severe anemia
    • Score of 0 or 1 point
      • Whether or not patient needs regular blood transfusion

0

Very Low

1

Low

2

Intermediate

3‒4

High

5‒6

Very High

*To be interpreted with the help of a clinician skilled in the management of MDS.
IPSS-R, Revised International Prognostic Score System; MDS, myelodysplastic syndromes; WPSS, World Health Organization classification-based Prognostic Scoring System

Determining Low Risk vs. High Risk

It is important to note that risk groups are based on averages from large groups of patients, so some cases of MDS fare better or worse than predicted. For example, factors like age, general health, ability to do daily activities, and the presence of other health conditions can affect risk, although these are not currently accounted for in available scoring systems. Also, risk groups can only help predict the likely course of untreated MDS over time, but cannot provide an estimate as to how patients will respond to treatment or how long an individual will live.8,69-72

Risk groups measured with each scoring system are often separated into two categories: lower-risk and higher-risk MDS. Generally, lower-risk MDS are likely to progress more slowly over time, not cause many or severe symptoms, and involve less intensive treatment. Higher-risk MDS are more likely to progress faster or develop into AML, cause more symptoms, and involve more intensive treatment.8,69-72

Risk Groups: Lower-risk versus Higher-risk*

Prognostic Scoring System

Lower-risk MDS

Higher-risk MDS

IPSS-R

Very Low, Low, Intermediate

Intermediate, High, Very High

WPSS

Very Low, Low, Intermediate

High, Very High

*To be interpreted with the help of a clinician skilled in the management of MDS.
IPSS-R, Revised International Prognostic Score System; MDS, myelodysplastic syndromes; WPSS, World Health Organization classification-based Prognostic Scoring System

10 Conventional Treatment

MDS treatments may include supportive care, drug therapy, and stem cell transplantation. Supportive care might involve blood transfusions, medications to increase the production of red blood cells, and antibiotics.

Chemotherapy

Chemotherapy uses different intravenous and oral drugs to kill abnormal cells or stop new ones from being made. The drugs used for MDS work in various ways and two or more agents may be used in a combination regimen. These medications can also affect normal cells, so they are given in varying cycles of treatment days followed by rest.

Low-intensity chemotherapy describes agents that are less likely to cause severe side effects. High-intensity chemotherapy is typically reserved to treat AML or before a hematopoietic cell transplant (HCT) for MDS. Two low-intensity chemotherapies are approved to treat MDS: azacitidine73-76 and decitabine.77-81 These drugs are in a class known as hypomethylating agents. They modify gene expression in abnormal cells and stunt their growth, which in turn activates genes for growth of normal, healthy cells in the bone marrow.82-85 Common side effects of hypomethylating agents are low white blood cell, platelet, or red blood cell counts and fever.

Immunosuppressive Therapy

With some types of MDS, the immune system attacks bone marrow tissue. Anti-thymocyte globulin (ATG) [equine] and cyclosporine are immunosuppressive drugs used for MDS. They suppress parts of the immune system to prevent damage to bone marrow.86-89

Cyclosporine inhibits an enzyme called calcineurin, preventing it from activating T cells and thereby restraining the immune system. Side effects might include increased blood pressure and elevated potassium levels. Vaccinations may be less effective in people taking cyclosporine, and it is important to avoid live attenuated vaccines while taking cyclosporine.

ATG is an antibody that stifles the immune system by eliminating certain types of white blood cells from the bloodstream. It may cause fever, chills, rash, low platelet or white blood cell counts, and joint pain. In certain situations, ATG and cyclosporine are used in combination.8

Immunomodulator

Lenalidomide is approved to treat MDS with cells that are missing part of chromosome 5, or del(5q). It supports the immune system by preventing the release of proteins that signal for inflammation and promoting development of T cells. This in turn helps bone marrow make normal cells and kill abnormal cells.90-100 Lenalidomide may be useful in some patients without del(5q) as well.101 In treating MDS, some of the common side effects of lenalidomide are low platelet and white blood cell counts, itching or rash, joint pain, and fever.

Targeted Therapy

Targeted therapies, like imatinib mesylate, are drugs that target unique features of cancer cells, and thus may be less likely to affect normal cells. A tyrosine kinase inhibitor (TKI), imatinib targets proteins that help cancer cells grow and is used especially in CMML expressing PDGFRβ gene mutations.8,64-66 Examples of common side effects include swelling, nausea and vomiting, muscle cramps, and musculoskeletal pain.

Allogeneic Hematopoietic Cell Transplant

An allogeneic hematopoietic cell transplant (HCT) destroys bone marrow cells with chemotherapy and replaces them with new blood stem cells, or hematopoietic stem cells, from a donor. The goal of HCT is to cure the cancer by allowing healthy blood stem cells to grow and form new bone marrow and blood cells. This process, called engraftment, takes about 2 to 4 weeks.102-106 The transplanted cells also give rise to new healthy T cells that can recognize and attack remaining malignant cells. This is known as the graft-versus-leukemia (GVL) or graft-versus-tumor (GVT) effect.107 A donor lymphocyte infusion may be used to donate lymphocytes, a type of white blood cell, to stimulate the GVL effect to treat MDS that reoccur or progress after an HCT.108

Drug Treatments for MDS*

Type of Drug

Generic (Brand)

Typical Treatment Cycles

Hypomethylating agents (low-intensity chemotherapy)

Azacitidine (Vidaza)

75 mg/m2 by subcutaneous or IV injection once daily for 7 days, then 21 days of rest for 4‒6 cycles

 

Decitabine (Dacogen)

20 mg/m2 by IV infusion over 1 hour once daily for 5 days, then 23 days of rest for 4‒6 cycles

Immunosuppressive therapy

ATG (Atgam)

10‒20 mg/kg by IV infusion over at least 4 hours once daily for 4‒5 days; cycles vary

 

Cyclosporine (Neoral, Sandimmune)

5 mg/kg by mouth 1‒2 times daily for 6 months

Immunomodulator

Lenalidomide (Revlimid)

10 mg by mouth once daily for 21 days, then 7 days rest; or 10 mg by mouth daily for 28 days

Targeted therapy

Imatinib mesylate (Gleevec)

400 mg by mouth once daily; cycles vary

*To be interpreted with the help of a clinician skilled in the management of MDS.
ATG, Anti-thymocyte globulin, equine; MDS, myelodysplastic syndromes. IV, intravenous

Treatment Approaches

Treatment options are guided by many factors, including MDS risk groups, patient symptoms, presence of chromosome abnormalities, serum EPO levels, and features that might indicate likelihood of response to certain medications.8

Initial Treatments for Lower-risk MDS with Anemia*

Test Results and Assessments

Treatment Options

Del(5q) and/or another chromosome change

  • Lenalidomide

No del(5q) and/or other chromosome changes, and serum EPO <500 mU/mL

  • Epoetin alfa ± G-CSF, or
  • Darbepoetin alfa ± G-CSF

No del(5q) and/or other chromosome changes, and serum EPO >500 mU/mL

If likely to respond to immunosuppressive therapy:

  • ATG + cyclosporine

If not likely to respond to immunosuppressive therapy:

  • Azacitidine
  • Decitabine
  • Consider lenalidomide
  • Clinical trial

Treatments for Loer-riswk MDS without Anemia*

Treatment Options

  • Azacitidine or decitabine
  • Immunosuppressive therapy for certain patients
  • Clinical trial

Initial Treatment for Higher-risk MDS*

Test Results and Assessments

Treatment Options

Allogenic HCT is a viable option, if a well-matched donor is available and the patient is healthy enough to tolerate the grueling procedure

  • Allogenic HCT
  • Azacitidine or decitabine followed by allogenic HCT
  • High-intensity chemotherapy followed by allogenic HCT

Allogenic HCT may be a viable option, but a well-matched donor is not available

  • Azacitidine
  • Decitabine
  • Clinical trial

Allogenic HCT is not a viable option given the patient’s advanced age and/or underlying health status, or a well-matched donor is not available

  • Azacitidine
  • Decitabine
  • Clinical trial

*To be interpreted with the help of a clinician skilled in the management of MDS.
ATG, Anti-thymocyte globulin, equine; EPO, erythropoietin; G-CSF, granulocyte colony stimulating factor; HCT, hematopoietic cell transplant; MDS, myelodysplastic syndromes.

Lower-risk MDS with anemia. Initial treatment for lower-risk MDS with anemia should consider the level of serum EPO in the blood and chromosome changes in the MDS cells, like del(5q).2,28 If MDS cells have a del(5q) chromosome change, with or without other chromosome abnormalities, lenalidomide treatment may be recommended. However, lenalidomide should not be given if patients have an abnormal chromosome 7, complex karyotype, or very low number of neutrophils or platelets.8,90-100

For patients with MDS and anemia, but without del(5q), serum EPO levels guide treatment. Epoetin alfa and darbepoetin alfa are erythropoietin-stimulating agents (ESAs) given for serum EPO levels ≤ 500 mU/mL. ESAs are sometimes given with G-CSF in order to stimulate growth of white blood cells.2,28 If tests show serum EPO > 500 mU/mL and factors indicate a high chance that MDS will respond to immunosuppressive therapy (IST), then ATG with cyclosporine may be helpful.86-89 If MDS are not likely to respond to IST, then patients usually receive azacitidine or decitabine.8

Lower-risk MDS without anemia. Azacitidine73-76 or decitabine82-85 are both low-intensity chemotherapy drugs used for lower-risk MDS without anemia. For some patients, if features of the MDS cells and other factors indicate that they are likely to respond to IST, treatment using ATG with or without cyclosporine is available.86-89

Higher-risk MDS. Patients with higher-risk MDS should be evaluated for treatment with an allogeneic HCT and high-intensity chemotherapy. While high-intensity treatments can improve survival and slow or stop MDS progression to AML, side effects can be severe and not all patients can tolerate therapy. In some instances, an HCT is performed right away. In other instances, patients receive azacitidine, decitabine, or high-intensity chemotherapy first.102-106

If an allogeneic HCT is not a good option, azacitidine may help improve blood cell counts, lower risk for progression to AML, and can increase survival time. Similarly, decitabine can lower the chance that MDS will progress to AML.8

Treatment of anemia. With MDS, anemia will be treated if it is causing symptoms. While ruling out and/or treating other possible causes of anemia, patients may receive iron, folate, or vitamin B12, red blood cell transfusions, and other supportive care. If a del(5q) mutation is present without an abnormal chromosome 7 or very low neutrophil or platelet counts, lenalidomide serves as an option.2,8

If no relevant chromosome abnormalities are present, patients should receive epoetin alfa or darbepoetin alfa for serum EPO levels ≤ 500 mU/mL. If 15% or more of the bone marrow is ring sideroblast, then G-CSF is typically given alongside the ESA.109-116

Initial Treatments for MDS-related Anemia*

Test Results

Treatment Options

del(5q) and/or one other chromosome change

  • Lenalidomide

Serum EPO <500 mU/mL and <15% ring sideroblasts

  • Epoetin alfa
  • Darbepoetin alfa

Serum EPO <500 mU/mL and ³15% ring sideroblasts

  • Epoetin alfa + G-CSF
  • Darbepoetin alfa + G-CSF

Serum EPO >500 mU/mL

  • See options for initial treatment of lower-risk MDS with anemia

*To be interpreted with the help of a clinician skilled in the management of MDS.
EPO, erythropoietin; G-CSF, granulocyte colony stimulating factor; MDS, myelodysplastic syndromes

11 Novel and Emerging Therapies

Following are several clinical developments in the MDS “pipeline.” None of the following products are currently approved for MDS. While this is not an exhausted list, these represent some important advancements in MDS research.

Clinical Trials

New treatment approaches for MDS are currently being developed and researched. Especially for certain patients who do not meet the criteria for some treatments or do not see benefit when using current therapies, clinical trials may provide suitable options and should be considered when available. The Leukemia & Lymphoma Society (LLS) provides assistance for patients through the Clinical Trial Support Center (https://www.lls.org/treatment/types-of-treatment/clinical-trials).

Venetoclax

Venetoclax (Venclexta) is a highly selective, oral inhibitor of the B-cell lymphoma 2 (Bcl-2) protein which interferes with cell death. Venetoclax helps restore the process of cancer cell death. This represents a newly available approach for the treatment of chronic lymphocytic leukemia (CLL) or small lymphocytic leukemia (SLL). It can also be used in combination with the hypomethylating agents, azacitidine or decitabine, or with cytarabine for patients 75 years or older with AML or those who have comorbidities that prevent the use of intensive induction chemotherapy.117

Guadecitabine

Guadecitabine is a next-generation hypomethylating agent (HMA) that is resistant to the enzyme that breaks down typical HMAs. It is administered under the skin and thought to release more gradually to provide longer exposure to the drug. Long-term phase II studies for MDS have been completed and an ongoing phase III study is currently underway.118,119

Immune Checkpoint Inhibition

A form of cancer immunotherapy, checkpoint inhibitors (CPIs) are agents that target and deactivate key controllers of the immune system called “checkpoints.” Checkpoint controllers weaken the immune system’s response to various cancers, so suppressing checkpoints allows the body’s immune system to mount a better response to cancer.118,120,121 More so, evidence suggests previous exposure to common MDS HMAs, like azacitidine, can induce certain immune checkpoints.120

Targeted Therapies

Many efforts are focusing on drug therapies that target genes common to MDS. TP53118,122,123 and isocitrate dehydrogenase (IDH) mutations,118,124 for instance, are just two examples of many aberrations associated with aggressive MDS and AML diseases. Mutations like these can result in poor clinical course and poor response to medications, so developing agents to target them is key. The IDH2 inhibitor enasidenib (Idhifa) was approved for treating AML after a phase I/II trial that included 30 patients with previous MDS.118,125

Targeting Innate Immunity

Toll-like receptors (TLRs) are a family of receptors involved in immune responses. With MDS, high levels of TLR2 are often expressed in the bone marrow, especially after treatment with HMAs, and disrupt the development of blood cells. When the experimental anti-TLR2 drug, tomaralimab, was given to 22 low-risk MDS patients in a phase I/II trial, it produced either complete or partial reduction of MDS in 50% of patients and little dose-limiting toxicity was observed.118,126

Amifostine

Used as a protectant, amifostine (Ethyol) binds excess chemotherapies in the body to protect patients from their harmful side effects.127,128 Due to its ability to stimulate growth of blood stem cells from healthy bone marrow and ameliorate cytopenias, amifostine is sometimes used as a single agent in the treatment of MDS.129-131

Sucrosomial Iron

MDS often cause certain types of red blood cell shortages that require supportive care with therapeutic iron administered through IV lines. IV iron, however, can cause gastrointestinal upset in up to 50% of patients and requires accessing a vein for administration by a trained clinician.132 Sucrosomial iron is a form of iron supplementation developed in Italy that is taken orally. Absorbed differently than other oral iron supplements, it has shown efficacy in MDS with less gastrointestinal side effects.132,133 When 92 patients with MDS and refractory anemia were followed for a median of 22 months while taking 28 mg sucrosomial iron daily, patients experienced high rates of incremental improvements. The regimens also included 400 mg vitamin B12 and 7.5 mg calcium levofolinate by mouth daily.132,134 Preliminary data suggest that oral sucrosomial iron may be as effective as IV iron in patients with MDS.132

12 Lifestyle Considerations

Smoking

Smoking is a modifiable risk factor associated with MDS,13,19-22 as well as other types of cancer. Specifically for MDS, evidence shows that current tobacco smokers have a higher risk than former smokers, and heavy smokers have a higher risk than lighter smokers.21 For patients with MDS who smoke, it is imperative to stop. Various tobacco cessation programs are available through nonprofit organizations that offer helpful tips and resources.135,136 The Centers for Disease Control and Prevention (CDC) also provides resources to aid in quitting tobacco smoking.

Exercise and Obesity

Obesity is associated with MDS.21,24 Trends in data also show a relationship between body mass index (BMI) and MDS. Defined as a BMI of 30 kg/m2 or more, obesity increases the risk of developing MDS by more than two-fold.21 For patients living with or at risk for MDS, exercise could be a way to manage this risk factor and can help mitigate side effects of disease or treatments.21,24,137

Body Mass Index (BMI) Calculations*

Measurement Units

Formula and Calculation

Example

Kilograms and meters

weight (kg) = BMI kg/m2
[height (m)]2

The formula for BMI is weight in kilograms divided by height in meters squared. If height has been measured in centimeters, divide by 100 to convert this to meters.

 

81 kg = 25 kg/m2
1.8 m2

Pounds and inches

703 x weight (lbs) = BMI kg/m2
[height (in)]2

When using American measurements, pounds should be divided by inches squared. This should then be multiplied by 703 to convert from lbs/inches2 to kg/m2.

 

703 x 180 lbs = 25 kg/m2
71 in2

*To be interpreted with the help of a skilled clinician.

13 Nutrients

The integrative interventions below may complement conventional MDS treatments for some people. Patients should always consult clinicians skilled in the management of MDS before starting a new regimen with any agent.

Vitamin K

Evidence suggests vitamin K may selectively clear abnormal blast cells in MDS.138,139 Menatetrenone (MK-4), a vitamin K compound, was given at 15 mg three times daily by mouth to nine patients with MDS for 16 weeks. Five patients in the vitamin K group experienced resolution of cytopenias, compared to one patient in the no menatetrenone group. Three major responses were observed in white blood cells, while increases in red blood cells and platelet count occurred in two patients each.140

A different multicenter phase II trial was conducted to investigate use of MK-4 alone versus MK-4 plus an active vitamin D3 metabolite for low-risk MDS with anemia, cytopenia, and multiple dysplasias. Out of 38 patients who received MK-4 (45 mg by mouth daily for 16 weeks), four saw improvements in anemia and one in thrombocytopenia. When 20 of the patients who did not respond to vitamin K alone took this in combination with 0.75 mcg of the vitamin D3 metabolite daily, six had improvements in cytopenias indicating that combination therapy may be helpful.141

Japanese researchers reported a case in 1999 in which an 80-year-old woman with MDS and refractory anemia was successfully treated with 45 mg daily of MK-4. Before treatment, the woman was heavily dependent on blood transfusions. Her blood cell counts gradually improved during 14 months of treatment with MK-4, to the extent that she no longer needed transfusions. Remarkably, her blood cell counts deteriorated again when she discontinued the MK-4 supplementation but recovered once more when she reinstated MK-4 supplementation. The researchers suggested that MK-4 may help restore hematopoiesis in certain MDS cases.

In preclinical research published in 2019, a team of researchers out of Germany and France found that drugs that block the effects of vitamin K, like the anticoagulant drug warfarin (Coumadin), altered the physiology of the bone marrow microenvironment and reduced functional hematopoietic stem cells 8-fold. Vitamin K-antagonist drugs were not found to be directly toxic to hematopoietic stem cells, but they modified the function of some key proteins in the bone marrow microenvironment—an effect to which the scientists attributed the decrease in functional hematopoietic stem cells. In an accompanying analysis of patient records, the researchers also found that use of vitamin K antagonists was more common in people who had been diagnosed with a MDS than in healthy people. However, the researchers stressed that their findings did not establish a causal link between vitamin K antagonists and MDS in humans, and that further research should be done to explore this connection.142

Importantly, people taking warfarin to prevent blood clots should not initiate vitamin K supplementation without first talking with their physician. Vitamin K, especially in high doses, may interfere with the efficacy of warfarin.

Vitamin D

Vitamin D can modulate different types of immune responses.143 Low levels of vitamin D3 have been associated with shorter survival after azacitidine treatment in patients with MDS. A study at a hospital in Spain analyzed vitamin D3 levels in 58 patients with MDS between 2006 and 2014. The estimated probability of surviving 2 years was 40% in patients with vitamin D3 levels over 13 ng/mL compared with 14% in those with lower levels.144 Unfortunately, participants’ vitamin D levels in this study were all below 50 ng/mL, which many experts consider a lower bound for the target healthy vitamin D range (50–80 ng/mL). More studies are needed to prospectively evaluate higher vitamin D levels and/or vitamin D supplementation in the context of MDS outcomes.

One study on the effects of vitamin D3 metabolites for MDS followed 19 patients for an average of more than 26 months. Five patients received 266 mcg calcifediol (25-hydroxyvitamin D3) three times weekly by mouth and 14 patients received calcitriol 0.25 to 0.75 mcg once daily by mouth. Eleven patients saw improvements in transfusion requirements and in counts for platelets, white blood cells, and red blood cells. Two patients experienced complete normalization of all blood cells.145

Maitake Mushroom

Preclinical research has shown that Maitake-derived beta-glucan may stimulate the growth of blood cells in bone marrow, increase the number of white blood cells in the bloodstream, and protect cells from potentially harmful molecules called reactive oxygen species (ROS).146-148 In a phase II clinical trial, 18 patients with MDS were given 3 mg/kg Maitake by mouth twice daily for 12 weeks. The treatment was well tolerated and increased the function of endogenous neutrophils and monocytes, which are types of white blood cells. These results suggest Maitake may have immunomodulatory potential and may enhance immune responses against bacterial infections for patients with MDS. However, there were small decreases in neutrophil count and hemoglobin levels among people taking the Maitake extract, although the changes were so small as to be deemed not clinically meaningful.148

Green Tea

The polyphenols epigallocatechin gallate (EGCG), epicatechin gallate, epigallocatechin, and epicatechin are well-known active plant compounds in green tea.149 Some epidemiology data suggest consuming green tea may protect against MDS. One study matched 208 patients with MDS to 208 control patients at a hospital based in China, accounting for gender, age, and residential locality. Compared with non-tea drinkers, regular tea consumption was associated with a decreased risk of developing MDS. This held true for those who consumed tea >20 years, ≥2 cups daily, and dried tealeaves ≥750 grams per annum and was associated across gender, good and intermediate/poor genetic risk groups, and lower versus higher risk groups.150 However, a large Japanese cohort study of 45,937 men and 49,870 women age 40‒69 years failed to find an association between tea consumption and risk of MDS.151 Further study is necessary to elucidate the role of tea consumption on MDS.

Ginger

6-shogaol is derived from the main active compound in ginger root, 6-gingerol, which also gives ginger its distinctive flavor. Shogaols from ginger display several liver-protective effects and can induce Nrf2, which controls genes for several liver-protective enzymes.152 Serum ferritin is often elevated in patients with MDS, and some research suggests ferritin levels are a potential prognostic factor. In a 6-month study measuring serum ferritin, six patients with low-risk MDS were given 1 gel capsule daily of 20 mg ginger extract, standardized at 20% 6-shogaol. Three patients experienced decreases in serum ferritin, despite having increased baseline levels, and one also had improved liver markers. Due to decreases in serum ferritin levels during the initial study, two of the three patients repeated the study for another six months. No ill effects from 6-shogaol were reported by patients.153

Coenzyme Q10

Coenzyme Q10 (CoQ10) occurs naturally in the body, mostly in the heart, liver, kidneys, and pancreas, and is used to support healthy cellular membranes and serum. Lower levels of CoQ10 reportedly occur with age, use of certain drugs, and cancer. It is thought to restore mitochondrial energy production that is impaired in patients with MDS. In 29 patients with MDS, 1,200 mg CoQ10 was given daily by mouth for a minimum of 16 weeks. Of the seven patients who responded to treatment, three had major responses in all three types of blood cells, including one patient who continued CoQ10 for 17 months before having a delayed response. Another patient’s red blood cell and platelet counts normalized, amongst other patients corrected cytopenias and improvements in quality of life. Two patients even experienced cellular genetic responses.154,155

Curcumin

Curcumin is the active ingredient in turmeric. By influencing various pathways, for example those regulated by growth factors or Nrf2, curcumin has a wide range of anti-inflammatory, antioxidant, and chemopreventive benefits.156,157 Supplemental curcumin has been shown in preclinical studies to sensitize MDS tissue samples to treatment with arsenic trioxide and has displayed synergy when used in combination with azacitidine.158,159

Luteolin

Nuclear factor erythroid 2-related factor 2 (NRF2) can play a pivotal role in protecting some types of cancer cells from certain chemotherapies. Luteolin, a common dietary flavonoid, inactivates NRF2.160,161 A study of 137 tissue samples from patients with MDS showed that patients with high-risk MDS often have greater levels of NRF2 in bone marrow than those with low-risk MDS. Furthermore, it showed that when 2 μM of luteolin was applied to the samples it enhanced the efficacy of a chemotherapy called cytarabine when given together. This suggests luteolin combined with conventional chemotherapy to target NRF2 could provide new therapy options in high-risk MDS.160 In another study of tissue samples from four patients, luteolin appeared to directly clear MDS-derived cells.161

Withaferin A

Withaferin A is an active substance from the Indian herb, Withania somnifera, commonly known as Indian winter cherry. Widely distributed across South Asian field, it is a traditional medicine used to treat inflammatory diseases, autoimmune diseases, and malignant tumors.162 Withaferin A showed that it might also suppress the growth of MDS cells in one study, when it was applied to tissue samples obtained from a patient with MDS.162 Another study used animal and human tissue samples to investigate how Withaferin A is able to do this. It found that Withaferin A had potential direct toxicity to MDS cells, but did not significantly affect normal human bone marrow cells.163

Copper

Copper is an essential element for all living organisms, but the daily copper requirement in humans is very low so deficiencies are not common. Nevertheless, copper shortages can cause blood abnormalities that can mimic MDS.30,164

Copper deficiencies can result from tube feeding, gastric bypass surgery, celiac disease, intestinal surgery, and other malabsorptive conditions. Copper deficiency can also arise as a consequence of excessive zinc intake, use of the chelating drug penicillamine, or from chronic use of prescription and over-the-counter proton pump inhibitors for heartburn or gastric reflux.30,165

2019

  • Nov: Comprehensive update & review

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. NCCN Guidelines for Patients: Myelodysplastic Syndromes. https://www.nccn.org/patients/guidelines/mds/index.html. Accessed October 1, 2019.
  2. Greenberg P. The myelodysplastic syndromes. In: Hoffman R BE, Shattil S, ed. Hematology: Basic Principles and Practice. 3rd ed. New York: Churchill Livingstone; 2000:1106-1129.
  3. MDS Foundation. Myelodysplastic Syndromes Foundation: What is MDS? https://www.mds-foundation.org/what-is-mds/. Accessed October 1, 2019.
  4. Zeidan AM, Shallis RM, Wang R, Davidoff A, Ma X. Epidemiology of myelodysplastic syndromes: Why characterizing the beast is a prerequisite to taming it. Blood reviews. 2019;34:1-15.
  5. Hellstrom-Lindberg E. Myelodysplastic syndromes: an historical perspective. Hematology Am Soc Hematol Educ Program. 2008:42.
  6. National Organization for Rare Disease (NORD): Myelodysplastic Syndromes. https://rarediseases.org/rare-diseases/myelodysplastic-syndromes/. Published 2017. Accessed October 1, 2019.
  7. Steensma DP. Historical perspectives on myelodysplastic syndromes. Leuk Res. 2012;36(12):1441-1452.
  8. National Comprehensive Cancer Netowork. Myelodysplastic Syndromes (Version 1.2020) Web site. https://www.nccn.org/professionals/physician_gls/pdf/mds.pdf. Published 2019. Accessed October 1, 2019.
  9. McLaughlin P, Estey E, Glassman A, et al. Myelodysplasia and acute myeloid leukemia following therapy for indolent lymphoma with fludarabine, mitoxantrone, and dexamethasone (FND) plus rituximab and interferon alpha. Blood. 2005;105(12):4573-4575.
  10. Pedersen-Bjergaard J, Andersen MK, Christiansen DH. Therapy-related acute myeloid leukemia and myelodysplasia after high-dose chemotherapy and autologous stem cell transplantation. Blood. 2000;95(11):3273-3279.
  11. Pedersen-Bjergaard J, Daugaard G, Hansen SW, Philip P, Larsen SO, Rorth M. Increased risk of myelodysplasia and leukaemia after etoposide, cisplatin, and bleomycin for germ-cell tumours. Lancet. 1991;338(8763):359-363.
  12. West RR, Stafford DA, Farrow A, Jacobs A. Occupational and environmental exposures and myelodysplasia: a case-control study. Leuk Res. 1995;19(2):127-139.
  13. Lv L, Lin G, Gao X, et al. Case-control study of risk factors of myelodysplastic syndromes according to World Health Organization classification in a Chinese population. Am J Hematol. 2011;86(2):163-169.
  14. 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. 1998;103(1):189-197.
  15. Jin J, Yu M, Hu C, et al. Pesticide exposure as a risk factor for myelodysplastic syndromes: a meta-analysis based on 1,942 cases and 5,359 controls. PLoS One. 2014;9(10):e110850.
  16. Pekmezovic T, Suvajdzic Vukovic N, Kisic D, et al. A case-control study of myelodysplastic syndromes in Belgrade (Serbia Montenegro). Annals of hematology. 2006;85(8):514-519.
  17. Strom SS, Gu Y, Gruschkus SK, Pierce SA, Estey EH. Risk factors of myelodysplastic syndromes: a case-control study. Leukemia. 2005;19(11):1912-1918.
  18. Sweeney MR, Applebaum KM, Arem H, Braffett BH, Poynter JN, Robien K. Medical Conditions and Modifiable Risk Factors for Myelodysplastic Syndrome: A Systematic Review. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2019;28(9):1502-1517.
  19. Du Y, Fryzek J, Sekeres MA, Taioli E. Smoking and alcohol intake as risk factors for myelodysplastic syndromes (MDS). Leuk Res. 2010;34(1):1-5.
  20. Bjork J, Johansson B, Broberg K, Albin M. Smoking as a risk factor for myelodysplastic syndromes and acute myeloid leukemia and its relation to cytogenetic findings: a case-control study. Leuk Res. 2009;33(6):788-791.
  21. Ma X, Lim U, Park Y, et al. Obesity, lifestyle factors, and risk of myelodysplastic syndromes in a large US cohort. American journal of epidemiology. 2009;169(12):1492-1499.
  22. Ugai T, Matsuo K, Sawada N, et al. Smoking and alcohol and subsequent risk of myelodysplastic syndromes in Japan: the Japan Public Health Centre-based Prospective Study. Br J Haematol. 2017;178(5):747-755.
  23. Liu P, Holman CD, Jin J, Zhang M. Alcohol consumption and risk of myelodysplastic syndromes: a case-control study. Cancer Causes Control. 2016;27(2):209-216.
  24. Poynter JN, Richardson M, Blair CK, et al. Obesity over the life course and risk of acute myeloid leukemia and myelodysplastic syndromes. Cancer epidemiology. 2016;40:134-140.
  25. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391-2405.
  26. Orazi Aea. Myelodysplastic Syndromes/Myeloproliferative Neoplasms, Chapter 5. In: Swerdlow S CE, Harris NL, et al, ed. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues. Vol 2. Revised 4th ed.: IARC Press, Lyon; 2017:82-96.
  27. Kaloutsi V, Kohlmeyer U, Maschek H, et al. Comparison of bone marrow and hematologic findings in patients with human immunodeficiency virus infection and those with myelodysplastic syndromes and infectious diseases. American journal of clinical pathology. 1994;101(2):123-129.
  28. Jacobs A, Janowska-Wieczorek A, Caro J, Bowen DT, Lewis T. Circulating erythropoietin in patients with myelodysplastic syndromes. Br J Haematol. 1989;73(1):36-39.
  29. Jafarzadeh A, Poorgholami M, Izadi N, Nemati M, Rezayati M. Immunological and hematological changes in patients with hyperthyroidism or hypothyroidism. Clinical and investigative medicine Medecine clinique et experimentale. 2010;33(5):E271-279.
  30. D'Angelo G. Copper deficiency mimicking myelodysplastic syndrome. Blood Res. 2016;51(4):217-219.
  31. Angotti LB, Post GR, Robinson NS, Lewis JA, Hudspeth MP, Lazarchick J. Pancytopenia with myelodysplasia due to copper deficiency. Pediatr Blood Cancer. 2008;51(5):693-695.
  32. Fong T, Vij R, Vijayan A, DiPersio J, Blinder M. Copper deficiency: an important consideration in the differential diagnosis of myelodysplastic syndrome. Haematologica. 2007;92(10):1429-1430.
  33. Gregg XT, Reddy V, Prchal JT. Copper deficiency masquerading as myelodysplastic syndrome. Blood. 2002;100(4):1493-1495.
  34. Haddad AS, Subbiah V, Lichtin AE, Theil KS, Maciejewski JP. Hypocupremia and bone marrow failure. Haematologica. 2008;93(1):e1-5.
  35. Koca E, Buyukasik Y, Cetiner D, et al. Copper deficiency with increased hematogones mimicking refractory anemia with excess blasts. Leuk Res. 2008;32(3):495-499.
  36. Bellos F, Kern W. Flow cytometry in the diagnosis of myelodysplastic syndromes and the value of myeloid nuclear differentiation antigen. Cytometry B Clin Cytom. 2017;92(3):200-206.
  37. Cremers EMP, Westers TM, Alhan C, et al. Multiparameter flow cytometry is instrumental to distinguish myelodysplastic syndromes from non-neoplastic cytopenias. European journal of cancer (Oxford, England : 1990). 2016;54:49-56.
  38. Della Porta MG, Picone C. Diagnostic Utility of Flow Cytometry in Myelodysplastic Syndromes. Mediterr J Hematol Infect Dis. 2017;9(1):e2017017.
  39. Porwit A, van de Loosdrecht AA, Bettelheim P, et al. Revisiting guidelines for integration of flow cytometry results in the WHO classification of myelodysplastic syndromes-proposal from the International/European LeukemiaNet Working Group for Flow Cytometry in MDS. Leukemia. 2014;28(9):1793-1798.
  40. Selimoglu-Buet D, Wagner-Ballon O, Saada V, et al. Characteristic repartition of monocyte subsets as a diagnostic signature of chronic myelomonocytic leukemia. Blood. 2015;125(23):3618-3626.
  41. Westers TM, Ireland R, Kern W, et al. Standardization of flow cytometry in myelodysplastic syndromes: a report from an international consortium and the European LeukemiaNet Working Group. Leukemia. 2012;26(7):1730-1741.
  42. Lambertenghi-Deliliers G, Orazi A, Luksch R, Annaloro C, Soligo D. Myelodysplastic syndrome with increased marrow fibrosis: a distinct clinico-pathological entity. Br J Haematol. 1991;78(2):161-166.
  43. Manoharan A, Horsley R, Pitney WR. The reticulin content of bone marrow in acute leukaemia in adults. Br J Haematol. 1979;43(2):185-190.
  44. Maschek H, Georgii A, Kaloutsi V, et al. Myelofibrosis in primary myelodysplastic syndromes: a retrospective study of 352 patients. Eur J Haematol. 1992;48(4):208-214.
  45. Pagliuca A, Layton DM, Manoharan A, Gordon S, Green PJ, Mufti GJ. Myelofibrosis in primary myelodysplastic syndromes: a clinico-morphological study of 10 cases. Br J Haematol. 1989;71(4):499-504.
  46. Steensma DP, Hanson CA, Letendre L, Tefferi A. Myelodysplasia with fibrosis: a distinct entity? Leuk Res. 2001;25(10):829-838.
  47. Alter BP, Baerlocher GM, Savage SA, et al. Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita. Blood. 2007;110(5):1439-1447.
  48. Du HY, Pumbo E, Ivanovich J, et al. TERC and TERT gene mutations in patients with bone marrow failure and the significance of telomere length measurements. Blood. 2009;113(2):309-316.
  49. Kussick SJ, Fromm JR, Rossini A, et al. Four-color flow cytometry shows strong concordance with bone marrow morphology and cytogenetics in the evaluation for myelodysplasia. American journal of clinical pathology. 2005;124(2):170-181.
  50. van de Loosdrecht AA, Alhan C, Bene MC, et al. Standardization of flow cytometry in myelodysplastic syndromes: report from the first European LeukemiaNet working conference on flow cytometry in myelodysplastic syndromes. Haematologica. 2009;94(8):1124-1134.
  51. Wood BL. Myeloid malignancies: myelodysplastic syndromes, myeloproliferative disorders, and acute myeloid leukemia. Clinics in laboratory medicine. 2007;27(3):551-575, vii.
  52. Wood BL, Arroz M, Barnett D, et al. 2006 Bethesda International Consensus recommendations on the immunophenotypic analysis of hematolymphoid neoplasia by flow cytometry: optimal reagents and reporting for the flow cytometric diagnosis of hematopoietic neoplasia. Cytometry B Clin Cytom. 2007;72 Suppl 1:S14-22.
  53. Baxter EJ, Kulkarni S, Vizmanos JL, et al. Novel translocations that disrupt the platelet-derived growth factor receptor beta (PDGFRB) gene in BCR-ABL-negative chronic myeloproliferative disorders. Br J Haematol. 2003;120(2):251-256.
  54. Chan WC FK, Morice WG, Catovsky D. T-cell large granular lymphocytic leukaemia. In: Swerdlow S, Campo, E, Harris, NL, et al, ed. WHO classification of tumours of haematopoietic and lymphoid tissues. 4th ed.: Lyon: IARC; 2008:272-273.
  55. Della Porta MG, Picone C, Pascutto C, et al. Multicenter validation of a reproducible flow cytometric score for the diagnosis of low-grade myelodysplastic syndromes: results of a European LeukemiaNET study. Haematologica. 2012;97(8):1209-1217.
  56. Morgan EA, Lee MN, DeAngelo DJ, et al. Systematic STAT3 sequencing in patients with unexplained cytopenias identifies unsuspected large granular lymphocytic leukemia. Blood Adv. 2017;1(21):1786-1789.
  57. Steer EJ, Cross NC. Myeloproliferative disorders with translocations of chromosome 5q31-35: role of the platelet-derived growth factor receptor Beta. Acta haematologica. 2002;107(2):113-122.
  58. Bejar R. Prognostic models in myelodysplastic syndromes. Hematology Am Soc Hematol Educ Program. 2013;2013:504-510.
  59. Cazzola M, Della Porta MG, Malcovati L. The genetic basis of myelodysplasia and its clinical relevance. Blood. 2013;122(25):4021-4034.
  60. Greenberg PL. The multifaceted nature of myelodysplastic syndromes: clinical, molecular, and biological prognostic features. Journal of the National Comprehensive Cancer Network : JNCCN. 2013;11(7):877-884; quiz 885.
  61. Kohlmann A, Bacher U, Schnittger S, Haferlach T. Perspective on how to approach molecular diagnostics in acute myeloid leukemia and myelodysplastic syndromes in the era of next-generation sequencing. Leuk Lymphoma. 2014;55(8):1725-1734.
  62. Tothova Z, Steensma DP, Ebert BL. New strategies in myelodysplastic syndromes: application of molecular diagnostics to clinical practice. Clin Cancer Res. 2013;19(7):1637-1643.
  63. Sakaguchi H, Okuno Y, Muramatsu H, et al. Exome sequencing identifies secondary mutations of SETBP1 and JAK3 in juvenile myelomonocytic leukemia. Nat Genet. 2013;45(8):937-941.
  64. Apperley JF, Gardembas M, Melo JV, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. The New England journal of medicine. 2002;347(7):481-487.
  65. David M, Cross NC, Burgstaller S, et al. Durable responses to imatinib in patients with PDGFRB fusion gene-positive and BCR-ABL-negative chronic myeloproliferative disorders. Blood. 2007;109(1):61-64.
  66. Magnusson MK, Meade KE, Nakamura R, Barrett J, Dunbar CE. Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor beta receptor fusion oncogene. Blood. 2002;100(3):1088-1091.
  67. Padron E, Garcia-Manero G, Patnaik MM, et al. An international data set for CMML validates prognostic scoring systems and demonstrates a need for novel prognostication strategies. Blood cancer journal. 2015;5:e333.
  68. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469-2475.
  69. Della Porta MG, Tuechler H, Malcovati L, et al. Validation of WHO classification-based Prognostic Scoring System (WPSS) for myelodysplastic syndromes and comparison with the revised International Prognostic Scoring System (IPSS-R). A study of the International Working Group for Prognosis in Myelodysplasia (IWG-PM). Leukemia. 2015;29(7):1502-1513.
  70. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454-2465.
  71. Malcovati L, Della Porta MG, Strupp C, et al. Impact of the degree of anemia on the outcome of patients with myelodysplastic syndrome and its integration into the WHO classification-based Prognostic Scoring System (WPSS). Haematologica. 2011;96(10):1433-1440.
  72. Bouayed J, Rammal H, Soulimani R. Oxidative stress and anxiety: relationship and cellular pathways. Oxid Med Cell Longev. 2009;2(2):63-67.
  73. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic response to three alternative dosing schedules of azacitidine in patients with myelodysplastic syndromes. J Clin Oncol. 2009;27(11):1850-1856.
  74. Martin MG, Walgren RA, Procknow E, et al. A phase II study of 5-day intravenous azacitidine in patients with myelodysplastic syndromes. Am J Hematol. 2009;84(9):560-564.
  75. Silverman LR, Fenaux P, Mufti GJ, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117(12):2697-2702.
  76. 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. 2006;24(24):3895-3903.
  77. Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007;109(1):52-57.
  78. Kantarjian HM, O'Brien S, Shan J, et al. Update of the decitabine experience in higher risk myelodysplastic syndrome and analysis of prognostic factors associated with outcome. Cancer. 2007;109(2):265-273.
  79. Lubbert M, Wijermans P, Kunzmann R, et al. Cytogenetic responses in high-risk myelodysplastic syndrome following low-dose treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Br J Haematol. 2001;114(2):349-357.
  80. van den Bosch J, Lubbert M, Verhoef G, Wijermans PW. The effects of 5-aza-2'-deoxycytidine (Decitabine) on the platelet count in patients with intermediate and high-risk myelodysplastic syndromes. Leuk Res. 2004;28(8):785-790.
  81. Wijermans P, Lubbert M, Verhoef G, et al. Low-dose 5-aza-2'-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol. 2000;18(5):956-962.
  82. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. The Lancet Oncology. 2009;10(3):223-232.
  83. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106(8):1794-1803.
  84. Lubbert M, Suciu S, Baila L, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J Clin Oncol. 2011;29(15):1987-1996.
  85. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20(10):2429-2440.
  86. Sloand EM, Wu CO, Greenberg P, Young N, Barrett J. Factors affecting response and survival in patients with myelodysplasia treated with immunosuppressive therapy. J Clin Oncol. 2008;26(15):2505-2511.
  87. Garg R, Faderl S, Garcia-Manero G, et al. Phase II study of rabbit anti-thymocyte globulin, cyclosporine and granulocyte colony-stimulating factor in patients with aplastic anemia and myelodysplastic syndrome. Leukemia. 2009;23(7):1297-1302.
  88. Molldrem JJ, Caples M, Mavroudis D, Plante M, Young NS, Barrett AJ. Antithymocyte globulin for patients with myelodysplastic syndrome. Br J Haematol. 1997;99(3):699-705.
  89. Passweg JR, Giagounidis AA, Simcock M, et al. Immunosuppressive therapy for patients with myelodysplastic syndrome: a prospective randomized multicenter phase III trial comparing antithymocyte globulin plus cyclosporine with best supportive care--SAKK 33/99. J Clin Oncol. 2011;29(3):303-309.
  90. Jadersten M, Saft L, Pellagatti A, et al. Clonal heterogeneity in the 5q- syndrome: p53 expressing progenitors prevail during lenalidomide treatment and expand at disease progression. Haematologica. 2009;94(12):1762-1766.
  91. Jadersten M, Saft L, Smith A, et al. TP53 mutations in low-risk myelodysplastic syndromes with del(5q) predict disease progression. J Clin Oncol. 2011;29(15):1971-1979.
  92. List A, Dewald G, Bennett J, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. The New England journal of medicine. 2006;355(14):1456-1465.
  93. Mallo M, Del Rey M, Ibanez M, et al. Response to lenalidomide in myelodysplastic syndromes with del(5q): influence of cytogenetics and mutations. Br J Haematol. 2013;162(1):74-86.
  94. Sebaa A, Ades L, Baran-Marzack F, et al. Incidence of 17p deletions and TP53 mutation in myelodysplastic syndrome and acute myeloid leukemia with 5q deletion. Genes, chromosomes & cancer. 2012;51(12):1086-1092.
  95. Giagounidis A, Mufti GJ, Mittelman M, et al. Outcomes in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with isolated deletion 5q treated with lenalidomide: a subset analysis from the MDS-004 study. Eur J Haematol. 2014;93(5):429-438.
  96. Kuendgen A, Lauseker M, List AF, et al. Lenalidomide does not increase AML progression risk in RBC transfusion-dependent patients with Low- or Intermediate-1-risk MDS with del(5q): a comparative analysis. Leukemia. 2013;27(5):1072-1079.
  97. List A, Kurtin S, Roe DJ, et al. Efficacy of lenalidomide in myelodysplastic syndromes. The New England journal of medicine. 2005;352(6):549-557.
  98. Nimer SD. Clinical management of myelodysplastic syndromes with interstitial deletion of chromosome 5q. J Clin Oncol. 2006;24(16):2576-2582.
  99. Raza A, Reeves JA, Feldman EJ, et al. Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate-1 risk myelodysplastic syndromes with karyotypes other than deletion 5q. Blood. 2008;111(1):86-93.
  100. Toma A, Kosmider O, Chevret S, et al. Lenalidomide with or without erythropoietin in transfusion-dependent erythropoiesis-stimulating agent-refractory lower-risk MDS without 5q deletion. Leukemia. 2016;30(4):897-905.
  101. Santini V, Almeida A, Giagounidis A, et al. Randomized Phase III Study of Lenalidomide Versus Placebo in RBC Transfusion-Dependent Patients With Lower-Risk Non-del(5q) Myelodysplastic Syndromes and Ineligible for or Refractory to Erythropoiesis-Stimulating Agents. J Clin Oncol. 2016;34(25):2988-2996.
  102. Bierings M, Nachman JB, Zwaan CM. Stem cell transplantation in pediatric leukemia and myelodysplasia: state of the art and current challenges. Curr Stem Cell Res Ther. 2007;2(1):53-63.
  103. Eissa H, Gooley TA, Sorror ML, et al. Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia: relapse-free survival is determined by karyotype and comorbidities. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2011;17(6):908-915.
  104. Festuccia M, Deeg HJ, Gooley TA, et al. Minimal Identifiable Disease and the Role of Conditioning Intensity in Hematopoietic Cell Transplantation for Myelodysplastic Syndrome and Acute Myelogenous Leukemia Evolving from Myelodysplastic Syndrome. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2016;22(7):1227-1233.
  105. Shaw PJ, Kan F, Woo Ahn K, et al. Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors. Blood. 2010;116(19):4007-4015.
  106. Strahm B, Nollke P, Zecca M, et al. Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG-MDS 98 study. Leukemia. 2011;25(3):455-462.
  107. Dickinson AM, Norden J, Li S, et al. Graft-versus-Leukemia Effect Following Hematopoietic Stem Cell Transplantation for Leukemia. Front Immunol. 2017;8:496.
  108. Fukumoto JS, Greenberg PL. Management of patients with higher risk myelodysplastic syndromes. Critical reviews in oncology/hematology. 2005;56(2):179-192.
  109. Stasi R, Abruzzese E, Lanzetta G, Terzoli E, Amadori S. Darbepoetin alfa for the treatment of anemic patients with low- and intermediate-1-risk myelodysplastic syndromes. Ann Oncol. 2005;16(12):1921-1927.
  110. Park S, Grabar S, Kelaidi C, et al. Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF: the GFM experience. Blood. 2008;111(2):574-582.
  111. Musto P, Lanza F, Balleari E, et al. Darbepoetin alpha for the treatment of anaemia in low-intermediate risk myelodysplastic syndromes. Br J Haematol. 2005;128(2):204-209.
  112. Mannone L, Gardin C, Quarre MC, et al. High-dose darbepoetin alpha in the treatment of anaemia of lower risk myelodysplastic syndrome results of a phase II study. Br J Haematol. 2006;133(5):513-519.
  113. Kelaidi C, Beyne-Rauzy O, Braun T, et al. High response rate and improved exercise capacity and quality of life with a new regimen of darbepoetin alfa with or without filgrastim in lower-risk myelodysplastic syndromes: a phase II study by the GFM. Annals of hematology. 2013;92(5):621-631.
  114. Jadersten M, Malcovati L, Dybedal I, et al. Erythropoietin and granulocyte-colony stimulating factor treatment associated with improved survival in myelodysplastic syndrome. J Clin Oncol. 2008;26(21):3607-3613.
  115. Hellstrom-Lindberg E, Ahlgren T, Beguin Y, et al. Treatment of anemia in myelodysplastic syndromes with granulocyte colony-stimulating factor plus erythropoietin: results from a randomized phase II study and long-term follow-up of 71 patients. Blood. 1998;92(1):68-75.
  116. Giraldo P, Nomdedeu B, Loscertales J, et al. Darbepoetin alpha for the treatment of anemia in patients with myelodysplastic syndromes. Cancer. 2006;107(12):2807-2816.
  117. Kucukyurt S, Eskazan AE. New drugs approved for acute myeloid leukemia (AML) in 2018. Br J Clin Pharmacol. 2019.
  118. Gil-Perez A, Montalban-Bravo G. Management of myelodysplastic syndromes after failure of response to hypomethylating agents. Therapeutic advances in hematology. 2019;10:2040620719847059.
  119. Sebert M, Renneville A, Bally C, et al. A phase II study of guadecitabine in higher-risk myelodysplastic syndrome and low blast count acute myeloid leukemia after azacitidine failure. Haematologica. 2019;104(8):1565-1571.
  120. Boddu P, Kantarjian H, Garcia-Manero G, Allison J, Sharma P, Daver N. The emerging role of immune checkpoint based approaches in AML and MDS. Leuk Lymphoma. 2018;59(4):790-802.
  121. Zeidan AM, Knaus HA, Robinson TM, et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin Cancer Res. 2018;24(15):3519-3527.
  122. Welch JS, Petti AA, Miller CA, et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. The New England journal of medicine. 2016;375(21):2023-2036.
  123. Zhang L, McGraw KL, Sallman DA, List AF. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications. Leuk Lymphoma. 2017;58(8):1777-1790.
  124. DiNardo CD, Jabbour E, Ravandi F, et al. IDH1 and IDH2 mutations in myelodysplastic syndromes and role in disease progression. Leukemia. 2016;30(4):980-984.
  125. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722-731.
  126. Wei Y, Dimicoli S, Bueso-Ramos C, et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia. 2013;27(9):1832-1840.
  127. Arboscello E, Botta M, Lerza R, et al. Amifostine promotes hemopoietic progenitor cell mobilization in patients with myelodysplastic syndrome. Anticancer research. 2002;22(3):1819-1824.
  128. Schanz J, Jung H, Wormann B, et al. Amifostine has the potential to induce haematologic responses and decelerate disease progression in individual patients with low- and intermediate-1-risk myelodysplastic syndromes. Leuk Res. 2009;33(9):1183-1188.
  129. Galanopoulos A, Kritikou-Griva E, Gligori J, et al. Treatment of patients with myelodysplastic syndrome with amifostine. Leuk Res. 2001;25(8):665-671.
  130. Viniou N, Terpos E, Galanopoulos A, et al. Treatment of anemia in low-risk myelodysplastic syndromes with amifostine. In vitro testing of response. Annals of hematology. 2002;81(4):182-186.
  131. Grossi A, Fabbri A, Santini V, et al. Amifostine in the treatment of low-risk myelodysplastic syndromes. Haematologica. 2000;85(4):367-371.
  132. Gomez-Ramirez S, Brilli E, Tarantino G, Munoz M. Sucrosomial((R)) Iron: A New Generation Iron for Improving Oral Supplementation. Pharmaceuticals (Basel). 2018;11(4).
  133. Giordano G, Cutuli MA, Lucchesi A, et al. Iron Support in Erythropoietin Treatment in Myelodysplastic Syndrome Patients Affected by Low-Risk Refractory Anaemia: Real-Life Evidence from an Italian Setting. Acta haematologica. 2019:1-8.
  134. Giordano G, Mondello P, Tambaro R, et al. Biosimilar epoetin alpha is as effective as originator epoetin-alpha plus liposomal iron (Sideral(R)), vitamin B12 and folates in patients with refractory anemia: A retrospective real-life approach. Molecular and clinical oncology. 2015;3(4):781-784.
  135. Substance Abuse and Mental Health Services Administration-Health Resources and Services Administration (SAMHSA-HRSA): Tobacco Cessation. https://www.integration.samhsa.gov/health-wellness/wellness-strategies/tobacco-cessation-2. Accessed October 1, 2019.
  136. Center for Disease Control and Prevention. Fast Facts and Fact Sheets. Quitting Smoking. . https://www.cdc.gov/tobacco/data_statistics/fact_sheets/cessation/quitting/index.htm. Accessed Octover 1, 2019.
  137. Schuler MK, Hentschel L, Gobel J, et al. Effects of a home-based exercise program on physical capacity and fatigue in patients with low to intermediate risk myelodysplastic syndrome-a pilot study. Leuk Res. 2016;47:128-135.
  138. Abe Y, Muta K, Hirase N, et al. [Vitamin K2 therapy for myelodysplastic syndrome]. Rinsho Ketsueki. 2002;43(2):117-121.
  139. Yaguchi M, Miyazawa K, Otawa M, et al. Vitamin K2 selectively induces apoptosis of blastic cells in myelodysplastic syndrome: flow cytometric detection of apoptotic cells using APO2.7 monoclonal antibody. Leukemia. 1998;12(9):1392-1397.
  140. Takami A, Asakura H, Nakao S. Menatetrenone, a vitamin K2 analog, ameliorates cytopenia in patients with refractory anemia of myelodysplastic syndrome. Annals of hematology. 2002;81(1):16-19.
  141. Akiyama N, Miyazawa K, Kanda Y, et al. Multicenter phase II trial of vitamin K(2) monotherapy and vitamin K(2) plus 1alpha-hydroxyvitamin D(3) combination therapy for low-risk myelodysplastic syndromes. Leuk Res. 2010;34(9):1151-1157.
  142. Verma D, Kumar R, Pereira RS, et al. Vitamin K antagonism impairs the bone marrow microenvironment and hematopoiesis. Blood. 2019;134(3):227-238.
  143. Aranow C. Vitamin D and the immune system. Journal of investigative medicine : the official publication of the American Federation for Clinical Research. 2011;59(6):881-886.
  144. Radujkovic A, Schnitzler P, Ho AD, Dreger P, Luft T. Low serum vitamin D levels are associated with shorter survival after first-line azacitidine treatment in patients with myelodysplastic syndrome and secondary oligoblastic acute myeloid leukemia. Clin Nutr. 2017;36(2):542-551.
  145. Mellibovsky L, Diez A, Perez-Vila E, et al. Vitamin D treatment in myelodysplastic syndromes. Br J Haematol. 1998;100(3):516-520.
  146. Lin H, Cheung SW, Nesin M, Cassileth BR, Cunningham-Rundles S. Enhancement of umbilical cord blood cell hematopoiesis by maitake beta-glucan is mediated by granulocyte colony-stimulating factor production. Clinical and vaccine immunology : CVI. 2007;14(1):21-27.
  147. Lin H, de Stanchina E, Zhou XK, et al. Maitake beta-glucan promotes recovery of leukocytes and myeloid cell function in peripheral blood from paclitaxel hematotoxicity. Cancer immunology, immunotherapy : CII. 2010;59(6):885-897.
  148. Wesa KM, Cunningham-Rundles S, Klimek VM, et al. Maitake mushroom extract in myelodysplastic syndromes (MDS): a phase II study. Cancer immunology, immunotherapy : CII. 2015;64(2):237-247.
  149. Henning SM, Wang P, Carpenter CL, Heber D. Epigenetic effects of green tea polyphenols in cancer. Epigenomics. 2013;5(6):729-741.
  150. Liu P, Zhang M, Jin J, Holman CD. Tea consumption reduces the risk of de novo myelodysplastic syndromes. Leuk Res. 2015;39(2):164-169.
  151. Ugai T, Matsuo K, Sawada N, et al. Coffee and green tea consumption and subsequent risk of acute myeloid leukemia and myelodysplastic syndromes in Japan. International journal of cancer Journal international du cancer. 2018;142(6):1130-1138.
  152. Zhuang X, Deng ZB, Mu J, et al. Ginger-derived nanoparticles protect against alcohol-induced liver damage. J Extracell Vesicles. 2015;4:28713.
  153. Golombick T, Diamond TH, Manoharan A, Ramakrishna R, Badmaev V. Effect of the Ginger Derivative, 6-Shogaol, on Ferritin Levels in Patients With Low to Intermediate-1-Risk Myelodysplastic Syndrome-A Small, Investigative Study. Clin Med Insights Blood Disord. 2017;10:1179545X17738755.
  154. Kalman Filanovsky MH, Vita Mirkin, Andrei Braester, Olga Shvetz, Anfisa Stanevsky, Erica Sigler, Ekaterina Votinov, Yehudit Zaltsman-Amir, Atan Gross, Lev Shvidel. Clinical Benefit and Improvement of Mitochondrial Function in Low Risk Myelodysplastic Syndrome Treated By Combination Ultra Coenzyme Q10 and L-Carnitine. Blood. 2017;130:1704.
  155. Galili N, Sechman EV, Cerny J, et al. Clinical response of myelodysplastic syndromes patients to treatment with coenzyme Q10. Leuk Res. 2007;31(1):19-26.
  156. Hatcher H, Planalp R, Cho J, Torti FM, Torti SV. Curcumin: from ancient medicine to current clinical trials. Cellular and molecular life sciences : CMLS. 2008;65(11):1631-1652.
  157. Ma L, Zhang X, Wang Z, et al. Anti-cancer effects of curcumin on myelodysplastic syndrome through the inhibition of enhancer of zeste homolog-2 (EZH2). Curr Cancer Drug Targets. 2019.
  158. Martin I, Navarro B, Solano C, et al. Synergistic Antioncogenic Activity of Azacitidine and Curcumin in Myeloid Leukemia Cell Lines and Patient Samples. Anticancer research. 2019;39(9):4757-4766.
  159. Zeng Y, Weng G, Fan J, et al. Curcumin reduces the expression of survivin, leading to enhancement of arsenic trioxide-induced apoptosis in myelodysplastic syndrome and leukemia stem-like cells. Oncology reports. 2016;36(3):1233-1242.
  160. Lin P, Ren Y, Yan X, et al. The high NRF2 expression confers chemotherapy resistance partly through up-regulated DUSP1 in myelodysplastic syndromes. Haematologica. 2019;104(3):485-496.
  161. Dong W, Lin Y, Cao Y, Liu Y, Xie X, Gu W. Luteolin induces myelodysplastic syndromederived cell apoptosis via the p53dependent mitochondrial signaling pathway mediated by reactive oxygen species. Int J Mol Med. 2018;42(2):1106-1115.
  162. Okamoto S, Tsujioka T, Suemori S, et al. Withaferin A suppresses the growth of myelodysplasia and leukemia cell lines by inhibiting cell cycle progression. Cancer science. 2016;107(9):1302-1314.
  163. Oben KZ, Alhakeem SS, McKenna MK, et al. Oxidative stress-induced JNK/AP-1 signaling is a major pathway involved in selective apoptosis of myelodysplastic syndrome cells by Withaferin-A. Oncotarget. 2017;8(44):77436-77452.
  164. Dalal N, Hooberman A, Mariani R, Sirota R, Lestingi T. Copper deficiency mimicking myelodysplastic syndrome. Clin Case Rep. 2015;3(5):325-327.
  165. Thakral B, Saluja K, Eldibany M. Zinc-induced copper deficiency: a diagnostic pitfall of myelodysplastic syndrome. Pathology. 2014;46(3):246-248.