Myelodysplastic Syndromes (MDS)

While myelodysplastic syndromes (MDS) can have a detrimental impact on a person's life, there is also a lot of hope, courage, and tenacity that is required in the face of this challenging diagnosis. The goal of this article is to help and encourage patients and their loved ones by shining light on the numerous elements of this condition.

Introduction

While myelodysplastic syndromes (MDS) can have a detrimental impact on a person's life, there is also a lot of hope, courage, and tenacity that is required in the face of this challenging diagnosis. The goal of this article is to help and encourage patients and their loved ones by shining light on the numerous elements of this condition.

Myelodysplastic disorders are characterized by defective bone marrow cells that produce blood. It is critical to understand that being informed about MDS is the first step in managing and coping with the condition, even if the diagnosis may cause concerns. The path is obviously difficult, but it also provides an opportunity to reinvent and construct a future outside the constraints imposed by MDS.

One ray of hope comes from the ongoing advances in medical research and therapy choices for Myelodysplastic Syndromes. Reducing symptoms, preventing complications, and slowing down MDS are the goals of management. Treatments include medications that increase the formation of red blood cells and blood transfusions. Occasionally, a bone marrow transplant—also known as a stem cell transplant—may be recommended, which involves replacing your unhealthy bone marrow with healthy bone marrow from a donor.

Cytopenias, or decreased blood cell counts, are a crucial aspect of MDS. For those with MDS, this results in symptoms like infections, anemia, bleeding, and easy bruising.

  • Anemia (low red blood cell counts), neutropenia (low white blood cell counts), and thrombocytopenia (low platelet counts) are the three major types of blood cell cytopenias. 

  • Dysplasia may cause mature blood cells circulating in the blood to malfunction, in addition to a decrease in the amount of blood cells. Dysplasia is characterized as a cell's abnormal shape and appearance, or morphology. 

  • The word myelo-, which means "marrow" in Greek, defines myelodysplasia. It refers to the abnormal shape and appearance of adult blood cells, often known as morphology. 

Because the bone marrow struggles to produce healthy cells slowly, MDS does not necessarily become fatal immediately. Sadly, some people pass away from the disease's direct effects, such as low platelet and blood cell counts, which make it difficult for the body to fight infections and stop bleeding. Furthermore, acute myeloid leukemia (AML) can develop in about 30% of MDS cases.

Living with MDS can be emotionally draining, but the strength of support networks can make a big impact. A robust support network is essential for managing the challenges of Myelodysplastic Syndromes, whether it be the unshakable support of family, the empathy of friends, or the advice of healthcare professionals. Sharing stories and connecting with people on similar paths can provide a sense of support and comfort.

Bone Marrow and Blood

Let's start with the fundamentals of bone marrow and blood to better understand MDS. Imagine bone marrow as the gelatinous, mushy material found inside our bones. There are two varieties: yellow, which resembles fatty tissue more, and red, which resembles myeloid tissue. About 220 billion new red, white, and platelet blood cells are produced daily by this incredible bone marrow factory.

Hematopoietic and mesenchymal stem cells are now seen in the bone marrow. The main players in the production of red blood cells are hematopoietic stem cells, which reside in the red bone marrow. Conversely, mesenchymal stem cells, which are present in yellow bone marrow, contribute to the formation of fat, cartilage, and bone. These stem cells can differentiate into several types of cells, making them the adaptable rookies of the body.

There is a problem with MDS: these stem cells may not mature appropriately and may accumulate in the bone marrow. Alternatively, they may live shorter lives than usual, which would result in a lower concentration of mature blood cells in the blood. It appears as though a hitch in the normally busy production line causes some disturbances in the blood cell division.

Myeloid and lymphoid lineages are the two primary cell types that can be produced by hematopoietic stem cells in the bone marrow. These comprise T cells, B cells, and natural killer (NK) cells in addition to monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, dendritic cells, and megakaryocytes, or platelets.

Causes and Risk Factors of MDS

Our bodies are made up of cells, each with its own set of instructions stored in structures known as chromosomes. Now, chromosomes are like our body's instruction manual, with numerous smaller portions known as genes. Each gene is responsible for a specific aspect of our body's function. Different genetic and chromosomal changes, which can arise spontaneously or be associated with specific causes, can impact the development of MDS. Patients exhibit abnormalities such as translocations or aneuploidy (changes in the number of chromosomes) in more than 80% of instances. 

5q Depletion

The most common abnormal karyotype is 5q deletion, which is divided into two types: 

  • Treatment-related MDS with 5q deletion (mostly induced by alkylating medications): Cancer treatments, such as chemotherapy or radiation, while necessary in the battle against cancer cells, can occasionally disrupt the natural function of bone marrow cells. In some circumstances, this disturbance may result in the formation of Myelodysplastic Syndromes, particularly those with the unique 5q deletion. 

  • De novo isolated 5q deletion: The Latin term "de novo" means "anew" or "from the beginning." In genetics, it refers to a novel genetic variation or mutation that was not acquired from either parent. It's similar to a genetic surprise that occurs during the creation of reproductive cells or the early stages of fetal development. In this usage, "isolated" refers to a deletion that arises independently of a wider genetic syndrome or disorder. It is a single event that occurs in a specific chromosomal region. "5q" refers to the long arm of chromosome 5, and "deletion" indicates that a portion of that area is missing or deleted. 

Patients who have had a 5q deletion induced by previous chemotherapy treatments generally have other cytogenetic abnormalities and/or TP53 mutations. A 5q deletion that is not associated with other cytogenetic abnormalities has a far better prognosis. Normal karyotype, deletion 7q (-7), trisomy 8, and -Y are also common cytogenetic abnormalities.

Let us take a look at the implications of these deletions in detail.

5q deletion-

In the case of 5q deletion, "5q" refers to the fifth chromosome, and "deletion" indicates that a portion of that chromosome is missing. Consider your instruction manual missing a page; it could influence how things work.

There are several reasons why a portion of the fifth chromosome may be deleted. It might occur at random or be linked to specific health concerns. 5q deletion is frequently related with myelodysplastic syndromes (MDS). 5q deletions are uncommon. Any of the following reasons couldbe result of 5q deletion:

  • Refractory anemia is the outcome of a drop in the amount of red blood cells in the blood. 

  • Refractory neutropenia is characterized by a decline in neutrophils, a subset of white blood cells. In the event that they drop below typical ranges, the patient is deemed "neutropenic" and vulnerable to infections.

  • Refractory thrombocytopenia, which is rare in Del 5(q) MDS patients, is a reduction in platelet quantity and quality.

Since deletion 5q rarely develops into acute leukemia, it is categorized as a low risk MDS. Chromosome 5 loss may also be seen in other MDS subgroups. Nevertheless, although they have a higher IPSS score, they are not regarded as belonging to this category because of several complex chromosomal abnormalities. Bruising, bleeding (from the gums or nose), recurrent infections, and an increased need for blood transfusions are all indicators that the illness may be worsening.

7q deletion

A chromosome anomaly known as a chromosome 7q deletion happens when a copy of the genetic material on chromosome 7's long arm (q) is absent. The size, location, and genes involved in the deletion determine the severity of the illness as well as its signs and symptoms. Individuals with chromosome 7q deletion frequently have behavioral issues, intellectual disabilities, developmental delays, and unique facial traits. Although most cases are not inherited, individuals can give their offspring the deletion. Each person's signs and symptoms determine how they should be treated.

Trisomy 8

Patients with illnesses affecting their myeloid lineage white blood cells often have bone marrow-derived cells that have trisomy 8 (gain of an additional 8 chromosome). Trisomy 8 MDS patients appear to upregulate WT1, an oncogene that can mutation in AML (but very rarely in MDS), however its role in pathogenesis is unclear. Some patients with isolated trisomy 8 may respond significantly to immunological suppression with antithymocyte globulin (ATG) since ATG can treat hematopoiesis dysfunction in MDS patients with trisomy 8. 

Loss of Y chromosome (LOY, -Y)

LOY is one of the most prevalent recurrent cytogenetic abnormalities in MDS, occurring in up to 30% of males. In some circumstances, it can be difficult to determine if LOY in MDS is a disease-associated alteration or simply incidental aging-associated somatic mosaicism. In contrast to other more strongly MDS-associated cytogenetic abnormalities such as del(5q) or del(7q), LOY cannot be utilized to define MDS under current World Health Organization (WHO) diagnostic criteria. While LOY in a higher proportion of metaphases has been consistently linked to MDS, it is unclear whether there is a corresponding increase in pathogenic MDS-associated mutations, which typically affect genes involved in DNA methylation, DNA damage repair, chromatin modification, RNA processing, transcription, and signal transduction.

MDS is associated with about 100 somatic point mutations, and there is some overlap with AML. The most prevalent somatic alterations include mutations in:

  • TET2- The TET2 enzyme regulates DNA methylation, which is vital for cellular proliferation and development. So to speak, TET2 acts as the master architect, creating the genetic landscape while skillfully balancing the preservation of key traits and the need for adaptation. DNA hypermethylation caused by TET2 mutation is related with a higher likelihood of MDS development, and a poor prognosis in AML.

  • SF3B1- SF3B1 (splicing factor 3b subunit 1) is required for branch site recognition and the initial phases of spliceosome assembly. It is an essential component in the symphony of cellular processes. It is involved in the splicing process, which is an important stage in the synthesis of proteins. Splicing is similar to constructing a musical piece in that multiple portions of the genome (musical notes) combine to generate a complete and functional mRNA transcript (sheet music). SF3B1 mutations in MDS cause several splicing abnormalities that define a disease defined by RS, inefficient erythropoiesis, a low probability of progression to leukemia, and a high overall survival (OS), implying a mode of action in MDS. 

  • ASXL1- The ASXL1 protein affects the expression of several genes, including the HOX genes, which are critical in prenatal development. Just like an effective city planner ensures that every building fulfills its purpose, ASXL1 regulates the activity of other genes to ensure that they function properly. ASXL1 can be compared to a diligent city planner, responsible for keeping order and guaranteeing the proper operation of the complex genetic metropolis within your cells. ASXL1 mutations in MDS can cause the expression of a C-terminal truncation mutant ASXL1 protein, resulting in decreased hematopoiesis.

  • DNMT3A- DNA (cytosine-5)-methyltransferase 3A (DNMT3A) may be a preventive factor against all types of cancers. DNMT3A, or DNA methyltransferase 3A, operates like a meticulous librarian, inserting tiny, important bookmarks into the pages of DNA books. These bookmarks, which take the form of methyl groups, help determine which chapters are "read" and used by the cell machinery, and which are left unopened. CDKN2B regulates hematopoietic progenitor cells, and DNMT3A mutations are related with CDKN2B promoter methylation.

  • SRSF2- SRSF2 has been shown to regulate gene transcription, pre-mRNA splicing, mRNA transport, and mRNA stability. It has been found to play a multifaceted role in MDS oncogenesis by affecting transcription, splicing, translation, and genomic stability. SRSF2 orchestrates the movement of genetic information within the nucleus, much like a traffic conductor oversees the flow of automobiles at a busy intersection. Its principal role is to regulate a process known as RNA splicing, which is similar to determining which routes cars (RNA molecules) travel to reach their destinations (final protein products).

  • RUNX1- RUNX1 is an important hematopoietic stem cell regulator. RUNX1 interprets the genetic code and controls the expression of genes involved in critical processes such as blood cell formation, immunological response, and tissue repair. It ensures that these processes occur in a coordinated and synchronized manner, allowing the body to work in complete harmony.

  • TP53- If our cells were a cite, where operations are meticulously organized, TP53 stands tall as the attentive guardian. When TP53 detects a threat, it goes into action. TP53 assesses the damage, halts cell division as needed, and initiates repair processes to repair the genetic material. In extreme instances where repair is not possible, TP53 may make the difficult decision to cause programmed cell death (apoptosis) in order to prevent injured cells from inflicting harm. The TP53 gene encodes a protein known as tumor protein p53 (p53). This protein functions as a tumor suppressor, which means it controls cell division by preventing cells from growing and dividing (proliferating) too quickly or in an uncontrolled manner. 

  • U2AF1- U2AF1 interprets the genetic information encoded in pre-mRNA in the same way as a conductor reads a musical score to bring out the nuances. It simplifies the splicing process, which is similar to selecting and mixing musical notes to create a coherent tune. The conductor ensures that the appropriate instruments (exons) are flawlessly combined while removing extraneous sections (introns), resulting in a refined and precise representation of the genetic composition. U2af1 is a gene that is essential for hematopoietic cancer cell survival. Mutant U2AF1-induced differential alternative splicing leads to oxidative stress in bone marrow stromal cells. U2af1 is necessary for hematopoietic stem and progenitor cell survival and function. 

  • EZH2- EZH2 is an important gene regulator. It attaches chemical markers, specifically methyl groups, to histone proteins that are connected with genes. These markings function as editorial comments, guiding the cellular machinery on how to read and react to the genetic information recorded in the DNA. EZH2 controls human erythropoiesis by causing histone and non-histone methylation. Mutations in the EZH2 gene have been linked to cancer development and progression. 

These mutations have been connected to a wide range of traits. TP53 gene mutations are associated with complex cytogenetics and poor overall survival. RUNX1 and TP53 are linked to worse thrombocytopenia. Hypomethylating medicines work effectively on TET2 mutations.

Metabolic theory and MDS

Metabolism is the process by which the body converts food into energy. Changes in cellular metabolic processes, according to metabolic theory, may contribute to the development and progression of illnesses like MDS.

In the context of MDS, metabolic theory suggests that changes in energy-producing processes within bone marrow cells may contribute to the abnormalities seen in MDS patients. This altered metabolism may have an impact on the balance of cell growth and death, affecting the formation of healthy blood cells. Consider it a delicate dance: when energy generation in cells is disrupted, the rhythm of cell development and function can go out of sync, resulting in the abnormal blood cell formation seen in MDS.

Let us understand how the metabolic theory plays a role in MDS. Hematopoietic stem cells, or HSCs, are potent bone marrow-derived cells are remarkably capable of differentiating into red blood cells, white blood cells, and platelets. Think of your body's hematopoietic stem cells as the ultimate multitaskers. Their capacity for self-renewal allows them to divide and produce similar stem cells, guaranteeing a steady supply. They have the ability to develop into specialized cells, each of which has a distinct purpose.

HSCs possess two incredible traits: pluripotency, which allows them to change into many types of blood cells, and self-renewal, which allows them to multiply without adhering to a particular kind. They thereby occupy the highest position in the hierarchy of blood cell formation. Throughout one’s life, these supercells are in charge of maintaining a silent reserve of themselves and, when necessary, replenishing your blood system with completely mature blood cells. HSCs awaken from their quiescent condition in response to an emergency, such as an infection or blood loss, and begin to proliferate, transforming into the precise blood cells your body requires at that precise moment. Different metabolic needs and metabolic states are linked to the various HSC and HSPC functioning states.

Inside your body, hematopoietic stem and progenitor cells, or HSPCs, are like intelligent energy managers. Their primary energy sources are glycolysis, which is similar to fast but inefficient batteries, and mitochondrial OXPHOS, which is similar to a powerful but slower generator. HSCs prefer to use the fast but weak batteries (glycolysis) when conditions are quiet and they don't require a lot of energy. But HSCs switch to the more potent but slower generator (mitochondrial metabolism) when there's a lot of work to be done, such as when the body wants to repair a wound or fight off an infection. 

It's interesting to note that these cells may alter the number and activity of their mitochondria, based on what the body needs. Glycolysis is the main energy source used by HSCs during the quiescent phase. It's their way of being ready to go when things are calm without using up too much energy. On the other hand, when it is time for them to mobilize, these cells use one of the most powerful energy producers—mitochondrial metabolism. 

When dealing with cancer, the body's metabolic pathways get distorted, allowing cancer cells to survive and grow. The bone marrow environment adapts to accommodate these cancer cells, resulting in a pro-inflammatory state. The regulators of these metabolic pathways are prospective targets for treating a variety of tumors, including myeloid malignancies.

Iron chelation is one example of a metabolism-focused therapy for myelodysplastic syndromes (MDS). Originally assumed to alleviate iron excess and its negative effects, it turns out to immediately address metabolic disorders such as insulin resistance. According to research, iron chelation appears to delay heart-related complications in MDS patients. Iron chelators, on a molecular level, help restore mitochondrial function and reduce toxic chemicals. So, adjusting iron metabolism can benefit MDS patients. 

Accepting dietary changes, such as calorie restriction, is becoming more acceptable as a supplementary treatment for a number of cancers. These treatments appear to be useful because they increase insulin sensitivity and important predictors like phosphatidylcholine and proline levels. Fasting-induced hormonal changes, particularly in adiponectin, leptin, insulin, and IGF1 levels, are relevant to illnesses like MDS because they influence HSC mortality in the bone marrow

Caloric restriction not only has anti-inflammatory properties, but it also helps the gut flora, making it an intriguing study in MDS. Beyond therapies, research into the MDS metabolome has shown a gene panel that exceeds conventional prognostic methods. However, the use of this gene-based method in clinical practice has yet to be established.

Recognizing MDS as a disorder characterized by unique metabolic alterations sheds light not just on present medication mechanisms, but also on potential new targets. Understanding the involvement of the bone marrow microenvironment in metabolic control will lead to novel treatment methods for this cancer.

Types of Myelodysplastic Syndromes

Myelodysplastic syndromes are classified according to abnormalities in blood cells and bone marrow. According to WHO (2016), MDS is classified as follows:

  • MDS with single lineage dysplasia (MDS-SLD) 

  • MDS with ring sideroblasts (MDS-RS)

  • MDS associated with multi-lineage dysplasia (MDS-MLD)

  • MDS with excess blasts (MDS-EB-1) with 5% to 9% blasts in the bone marrow and MDS with excess blasts (MDS-EB-2) with 10% to 19% blasts in the bone marrow 

  • MDS with isolated del(5q)

  • Unclassifiable (MDS-U) MDS 

In addition, the WHO 2016 classification includes overlap syndromes such as:

  • Myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T)

  • Myelodysplastic/myeloproliferative neoplasm, unclassifiable

Refractory anemia (RA)

RA is characterized by anemia, dyserythropoiesis in more than 10% of erythroid precursors, and a low number of blasts in bone marrow (<5%) and peripheral blood (≤1%). Refractory anemia is a feature of all myelodysplastic syndromes (MDSs). MDS bone marrow is typically hypercellular due to inefficient hematopoiesis, as opposed to aplastic anemia bone marrow, which is hypocellular. Refractory anemia develops when the patient's blood has inadequate red blood cells. The platelet and white blood cell levels are normal.

Refractory anemia with ring sideroblasts: 

Refractory anemia with ring sideroblasts (RARS) is a kind of myelodysplastic syndrome (MDS) marked by anemia and at least 15% ring sideroblasts in the marrow. Typically, patients will have normochromic, normocytic anemia, and erythroid hyperplasia.

RARS is associated with an increased chance of progression to overt acute myeloid leukemia (AML), as well as a shorter overall survival time, albeit these risks are not as substantial as those associated with higher-grade MDS. The existence of ring sideroblasts alone is insufficient to diagnose RARS; a thorough examination of all nonerythroid lineages is required. RARS does not exhibit dysplasia in the granulocytic or megakaryocytic lineages. Myeloblasts make up less than 5% of nucleated bone marrow cells, and Auer rods are missing.

Refractory anemia with excess blasts: 

The patient suffers from anemia while blasts make up 5% to 19% of bone marrow cells. White blood cell and platelet changes are also noted. Excess blasts in patients with refractory anemia may result in acute myeloid leukemia (AML). 

People with RAEB may experience one, two, or all three of the following difficulties with blood cell production:

  • Refractory anemia: a decrease in the amount of circulating red cells, resulting in anemic symptoms.

  • Refractory neutropenia refers to a decrease in neutrophils, a type of white blood cell. When these levels go below normal, the patient is labeled 'neutropenic' and prone to infections.

  • Refractory thrombocytopenia: a drop in platelet count, making the person prone to severe bruising and bleeding. Platelet quality may also be altered, increasing the risk of bleeding even if platelet numbers are relatively normal.

Refractory cytopenia in multilineage dysplasia

The World Health Organization (WHO) Classification of Tumors of Hematopoietic and Lymphoid Tissues defines MDS-MLD as a form of MDS characterized by one or more cytopenias and dysplastic alterations in two or more myeloid lineages (erythroid, granulocytic, or megakaryocytic). 

Cytopenia is defined as a hemoglobin level below 10 g/dL, an absolute neutrophil count of less than 1,800/μL, and a platelet count below 100,000/μL. A diagnosis of MDS-MLD requires the presence of dysplastic characteristics in more than 10% of cells from two or more lineages. There are less than 1% blasts in peripheral blood and less than 5% blasts in bone marrow. Auer rods are absent.

Unclassifiable Myelodysplastic syndrome

The WHO improved the MDS-U category in the 2008 classification to better characterize individuals who don't have enough data to be placed in an other, more "specific" MDS group. There are three circumstances include:

  • Patients with refractory cytopenia, either with unilineage or multilineage dysplasia, and 1% circulating blasts (MDS-U BL)

  • Patients with pancytopenia and unilineage dysplasia (MDS-U Pan)

  • Patients with less than 5% bone marrow (BM) blasts, less than 1% peripheral blood (PB) blasts, less than 10% dysplastic cells in the BM, and the presence of MDS-defining cytogenetic abnormalities (MDS-U CG) 

The World Health Organization's 2016 Classification of Myeloid Neoplasms defines various subtypes of myelodysplastic neoplasms, as well as overlap syndromes with both myeloproliferative and myelodysplastic characteristics. This classification is based on morphology, dysplasia, and karyotype abnormalities, particularly 5q deletion.

There are also a number of other overlap syndromes with myeloproliferative and myelodysplastic features such as chronic myelomonocytic leukemia (CMML), atypical chronic myeloid leukemia (CML), and juvenile myelomonocytic leukemia (JMML).

Symptoms

Myelodysplastic disorders do not usually have early warning signs and symptoms. They could be identified by a routine blood test. Signs and symptoms can be caused by myelodysplastic syndromes or other illnesses. If you experience any of the following symptoms, see your doctor:

  • Shortness of breath.

  • Weakness or feeling tired.

  • Having skin that is paler than usual.

  • Easy bruising or bleeding.

  • Petechiae (flat, pinpoint spots under the skin caused by bleeding).

Low red cell count (anemia)

When persons with MDS are first identified, the majority of them are anemic. Anemia is defined by a persistently low hematocrit or hemoglobin (the blood protein that carries oxygen to the body's tissues). Anemic individuals frequently experience tiredness and describe feeling tired all of the time. 

  • Patients with moderate anemia may feel OK or only slightly tired. Almost all moderate anemia patients complain of weariness, which may be accompanied by heart palpitations, shortness of breath, and a pale complexion. 

  • Almost all patients with severe anemia seem pale and complain of persistent overwhelming weariness and shortness of breath. Because severe anemia lowers blood supply to the heart, elderly people may be more prone to cardiovascular symptoms such as chest pain. 

  • Although chronic anemia is rarely fatal, it can have a significant impact on a patient's quality of life.

Low white cell count (neutropenia)

A decrease in the number of white blood cells decreases the body's resistance against bacterial infection. Skin infections, sinus infections (which cause nasal congestion), lung infections (which cause coughing and shortness of breath), and urinary tract infections (which cause painful and frequent urination) are more common in neutropenic patients. Fever may accompany many disorders.

Low platelet count (thrombocytopenia)

Thrombocytopenia patients are more likely to bruise and bleed after minor bumps and scratches. Patients regularly have nosebleeds and gum bleeding, particularly after dental operations. Before dental treatment, consult with your hematologist, who may prescribe prophylactic antibiotics. Infection and bleeding are hazardous for most MDS patients.

Diagnosis

Your doctor may do the following tests and procedures in addition to asking about your personal and family health history and performing a physical exam:

Full blood count (CBC) with differential

This set of tests screens for various disorders in your blood. These include anemia, infections, and leukemia. It can assist you in determining how your overall health is. The test collects a large amount of information from your blood sample:

  • The quantity and composition of white blood cells (WBCs)
    Your body contains five types of white blood cells. Everyone contributes to the fight against illnesses. A high count of WBCs, or a specific type of WBC, may indicate an infection or inflammation somewhere in your body. Low WBC counts may indicate that you are at risk for infection.

  • The count of red blood cells
    RBCs transport oxygen throughout the body and eliminate excess CO2. Too few RBCs may be a symptom of anemia.

  • The size of your red blood cells
    This test is referred to as red cell distribution width (RDW, RDW-CV, or RDW-SD). If you have anemia, you may notice larger disparities in red blood cell size.

  • Hematocrit (HCT)
    This refers to the proportion of red blood cells in a specific amount of whole blood. A low hematocrit may indicate excessive bleeding. It could also suggest that you have an iron deficit or another illness. Dehydration and other conditions can produce an elevated hematocrit.

  • Hemoglobin (Hgb or Hb)
    Hemoglobin is a protein found in red blood cells. It transports oxygen from the lungs to the rest of the body. Abnormalities can indicate a variety of conditions, including anemia and lung disease.

  • The average diameter of your red blood cells
    This test is referred to as mean corpuscular volume (MCV). MCV increases when your red blood cells are larger than normal. This occurs when you have anemia due to a lack of vitamin B-12 or folate. If your red blood cells are smaller, this may indicate another type of anemia, such as iron deficiency anemia.

  • A platelet count
    Platelets are cell fragments that aid in blood coagulation. A low platelet count may indicate a higher risk of bleeding. Too many may indicate a variety of conditions.

  • Mean corpuscular hemoglobin
    This test determines how much hemoglobin your red blood cells contain.

Peripheral blood smear (PBS)

A peripheral blood smear test is a technique used by healthcare experts to evaluate your red and white blood cells, as well as your platelets. Unlike certain blood tests, which are done by machines, healthcare personnel examine blood cells under a microscope.

Your healthcare provider may prescribe a peripheral blood smear in conjunction with a complete blood count (CBC), or if your CBC results suggest aberrant blood cell activity, they may order a PBS. For example, your CBC results may show that your white blood cells, red blood cells, and/or platelets are abnormal, or that you have an abnormal amount of any specific type of cell. A microscopic examination of your cells may assist your healthcare professional in determining how and/or why your blood cells appear abnormal, or whether you have an abnormal quantity of cells.

A peripheral blood smear test alone does not provide a diagnosis. Healthcare providers determine diagnoses based on your medical history, physical examination, and the results of laboratory tests such as PBS.

Cytogenetic analysis

A laboratory test that counts the chromosomes of cells in bone marrow or blood and looks for anomalies such as damaged, missing, altered, or extra chromosomes. Changes in specific chromosomes could indicate cancer. Cytogenetic analysis is used to help in cancer diagnosis, therapy planning, and assessing how well a treatment is working. 

Cytogenetic testing reveals information about a cell's chromosomes. These tests can be used to identify certain genetic illnesses or forms of cancer. Doctors utilize cytogenetic tests to determine chromosomal alterations. These tests are sometimes performed on a small sample of blood obtained during a routine blood draw. Your doctor may also utilize a bone marrow sample for cytogenetic analysis. Two approaches are commonly used to get a bone marrow sample:

  • A bone marrow aspiration- the removal of fluid from the bone marrow.

  • A bone marrow biopsy- the removal of cells from the bone marrow.

These tests may then be forwarded to a laboratory, where a pathologist will examine the cells and help make a diagnosis. When you get cytogenetic tests, the laboratory exclusively looks for particular abnormalities that are linked to cancer. These tests do not reveal your unique genetic information. If you take these tests, neither the laboratory nor your doctor's office will know what your typical genes are.

Blood chemistry studies

A method in which a blood sample is examined to determine the levels of various compounds produced in the blood by organs and tissues, such as vitamin B12 and folate. A compound in an unusual (higher or lower than normal) concentration can be a symptom of illness.

These tests may then be forwarded to a laboratory, where a pathologist will examine the cells and help make a diagnosis. When you get cytogenetic tests, the laboratory exclusively looks for particular abnormalities that are linked to cancer. These tests do not reveal your unique genetic information. If you take these tests, neither the laboratory nor your doctor's office will know what your typical genes are.

Bone marrow aspiration and biopsy

In this, a hollow needle is inserted into the hipbone or breastbone in order to extract bone marrow, blood, and a small sample of bone. A pathologist examines bone marrow, blood, and bone with a microscope for aberrant cells.

The removed tissue sample might be subjected to the following tests:

  • Immunocytochemistry: A laboratory test that looks for specific antigens (markers) in a patient's bone marrow sample using antibodies. Typically, antibodies are linked to an enzyme or a fluorescent dye. The enzyme or dye is activated once the antibodies bind to the antigen in the patient's cell sample, and the antigen is visible under a microscope. This type of test is used to aid in cancer detection and differentiate between myelodysplastic syndromes, leukemia, and other illnesses.

  • Immunophenotyping is a laboratory technique that employs antibodies to identify cancer cells based on antigens or markers on their surfaces. This test is used to help diagnose some types of leukemia and other blood disorders.

  • Flow cytometry is a laboratory test used to detect the number of cells in a sample, the percentage of living cells in the sample, and specific cell characteristics such as size, shape, and the presence of tumor (or other) markers on the cell surface. A sample of a patient's blood, bone marrow, or other tissue is stained with a fluorescent dye, placed in a fluid, and then passed one at a time through a light beam. The fluorescent dye-stained cells react to the light beam, which determines the test findings. This test is used to help in the diagnosis and treatment of some cancers, including leukemia and lymphoma.

FISH (fluorescence in situ hybridization) is a technique used in laboratories to examine and count genes or chromosomes in cells and tissues. Fluorescent dye-containing DNA fragments are generated in the lab and added to a sample of a patient's cells or tissues. When these dyed DNA fragments bind to specific genes or chromosomal areas in the sample, they glow under a fluorescent microscope. The FISH test aids in cancer diagnosis and treatment planning.

Treatment Options

Supportive Care

Supportive care is provided to alleviate the effects of the disease or its treatment. The following are examples of supportive care:

Transfusion therapy

Transfusion therapy (blood transfusion) is a method of replacing damaged blood cells with red blood cells, white blood cells, or platelets. A red blood cell transfusion is delivered when the red blood cell count is low and signs or symptoms of anemia, such as shortness of breath or feeling exceedingly weary, arise. When a patient is bleeding, undergoing surgery that may result in bleeding, or has a very low platelet count, a platelet transfusion is usually delivered.

Patients who get several blood cell transfusions may suffer tissue and organ damage due to excessive iron buildup. In these patients, iron chelation therapy may be performed to remove excess iron from the blood.

Erythropoiesis-stimulating agents

Erythropoietin (EPO) is a glycoprotein hormone that is spontaneously produced by the kidney's peritubular cells and stimulates red blood cell formation. The majority of EPO produced in the human body comes from peritubular cells in the renal cortex. pO2 (partial pressure of oxygen), which reflects the amount of oxygen gas dissolved in the blood, directly controls EPO production. The lower the pO2, the higher the production of EPO. Erythropoietin stimulating agents (ESAs) are pharmaceutically generated recombinant EPOs. ESAs include epoetin, darbepoetin, and methoxypolyethylene glycol-epoetin beta. ESAs are typically used in cases of poor red blood cell production. 

The most serious side effects of ESA are linked to an increased risk of thrombotic events, particularly in surgical patients. The use of erythropoietin-stimulating drugs increases blood viscosity due to increased erythrocyte synthesis. This, together with the diminished vasodilatory impact caused by a low baseline pO2, increases the risk of ischemic stroke and myocardial infarctions. There is also an elevated risk of venous thromboembolism, and some have suggested using antithrombotic prophylaxis in individuals taking ESA medication.

Medication Administration

Lenalidomide

Patients with myelodysplastic syndrome who have an isolated del(5q) chromosomal abnormality and require regular red blood cell transfusions may be treated with lenalidomide. Lenalidomide is a medicine used to decrease the need for red blood cell transfusions. In patients with IPSS low- or intermediate-1-risk (Int-1-risk) MDS with del(5q), either alone or in conjunction with additional cytogenetic abnormalities (United States); in patients with isolated del(5q), lenalidomide is approved for the treatment of transfusion-dependent (TD) anemia when other therapeutic options are insufficient or inadequate (European Union).

Evidence underlines that lenalidomide is beneficial not only in lowering the need for red blood cell transfusions but also in changing the course of the disease by suppressing the del(5q) clone, which causes cytogenetic responses (CyRs) and, in turn, lowers the risk of AML progression and lengthens survival.

Immunosuppressive therapy (IST)

Studies demonstrating that up to 48% of MDS patients had indications of an autoimmune illness provide justification for the use of IST in this patient population; nevertheless, the significance of this result for prognosis remains debatable. Furthermore, it has been observed that individuals with both lower-risk MDS and aplastic anemia have decreased hematopoiesis due to dysregulation of T-cell function, which may be rectified with IST. 

With varied degrees of efficacy, IST has been tested in the treatment of MDS in several forms. Transfusion independence and sustained objective responses have been shown to reach up to 27% and 55%, respectively, in earlier research. Consensus guidelines advise non-del(5q-) MDS patients with low or intermediate-1 risk to take into consider undergoing IST. The most widely used of these are monoclonal antibodies (etanercept, alemtuzumab), cyclosporine A (CsA), and anti-thymocyte globulin (ATG), which can be administered alone or in combination.

Azacitidine and decitabine

The Food and Drug Administration (FDA) has authorized the use of lower-intensity chemotherapeutic drugs, such as decitabine (DAC) and azitidine (AZA), to treat MDS. Azacitidine and decitabine are medications that kill rapidly proliferating cells in order to treat myelodysplastic syndromes. They also help genes involved in cell proliferation function properly. Treatment with azacitidine and decitabine may slow the progression of myelodysplastic syndromes to acute myeloid leukemia.

The structures of these two drugs differ slightly: DAC is a deoxyribonucleoside, whereas AZA is a ribonucleoside. Their mechanisms of action also have slight differences-  DNA methyltransferases are depleted by the actions of both AZA and DAC. These two agents' modes of action, however, differ: 10-20% percent of AZA can also be transformed into 5-aza-2'-deoxycytidine, which can result in the reexpression of tumor suppressor genes. 80-90% of AZA gets incorporated into RNA, eventually blocking tumor-related protein production. Low-dose DAC reactivates dormant tumor suppressor genes, while high-dose DAC cytotoxically prevents DNA cross-linking and synthesis. Clinical results suggest that AZA is more successful than DAC, despite preclinical studies showing that DAC is more effective than AZA in vivo. 

Stem Cell Transplant

For myelodysplastic syndrome (MDS), allogeneic hematopoietic stem cell transplantation (HCT) is the sole treatment that can be effective. To destroy all of the cells in the bone marrow, including the aberrant bone marrow cells, the patient undergoing this treatment will either get high-dose chemotherapy or total body radiation. The patient then receives fresh stem cells that can produce blood. 

Two primary categories of SCT are:

  • Allogeneic stem cell transplant
    After the bone marrow is destroyed, the patient undergoing an allogeneic stem cell transplant receives blood-forming stem cells from a different person—the donor. For MDS, this kind of transplant is usually utilized. When the donor's cell type closely matches that of the patient and the donor is a close relative of the patient, such as a brother or sister, the treatment's outcomes typically work well. Rarely, the patient and donor match, but they are unrelated.

  • Autologous stem cell transplant
    The recipient of an autologous stem cell transplant receives their own stem cells back. Patients with MDS usually do not receive this kind of transplant because their bone marrow contains aberrant stem cells.

The early side effects of a stem cell transplant are comparable, albeit more severe, to those associated with radiation and chemotherapy. Low blood counts are among the most dangerous side effects, as they increase the risk of severe infections and bleeding.

Graft-versus-host disease (GVHD) is another potentially fatal allogeneic transplant adverse effect. This happens when the patient's tissues are attacked by the donor's new immune cells, who mistake them for alien objects. Any area of the body may be affected by GVHD, which carries a risk to life.

Not every MDS patient who receives a transplant is cured, despite the fact that allogeneic stem cell therapy is now the sole treatment that can cure some patients. Additionally, this treatment's side effects may cause some patients to pass away. Medical professionals frequently advise against seeking a stem cell transplant until the MDS reaches a more severe stage.

Prognosis

MDS patients' prognoses vary widely depending on various characteristics, including cytogenetics and the severity of cytopenias. To guide treatment and the likely clinical course, clinicians use the International Prognostic Scoring System (IPSS) and revised IPSS (R-IPSS) risk stratification systems. The R-IPSS risk stratifies patients based on cytogenetics, blast percentage, and distinct scores for absolute neutrophil count, hemoglobin value, and platelet value, and it has been shown to predict prognosis better than the previous IPSS. Patients in the very high-risk category for R-IPSS, for example, had a median overall survival of 0.8 years, compared to 8.8 years for patients in the very low-risk category.

In addition to a clinical assessment of the patient's age and co-morbidities, these systems can be used to determine the best therapeutic options. The IPSS considers the number of blasts in the bone marrow, the karyotype, and the number of cell lineages with cytopenias. Normal karyotypes, -Y, deletion 5q, and deletion 20q have an excellent prognosis. High-risk karyotypes include complex cytogenetics (more than three abnormalities) and chromosome 7 abnormalities. All other karyotypes are regarded as intermediate risk. A risk score is computed based on these variables to determine if the risk is low, intermediate-1 or intermediate-2, or high.

Patients classified as high-risk or with a poor prognosis will almost likely require treatment and may be candidates for allogeneic stem cell transplant to maintain their remission. Patients with isolated 5q deletion have better prognosis than those with other types of MDS, with one study indicating a 5-year survival rate of 40% if no treatment was given and 54% if treatment was given.

Care

While MDS treatment shows great potential, there are several precautions that can help you relieve symptoms. People with myelodysplastic syndromes have low white blood cell counts, which makes them vulnerable to recurring, and frequently serious, infections. To lower your chance of infection, here are a few precautions one can take:

  • Please wash your hands. Hands should be washed regularly and thoroughly with warm, soapy water, particularly before eating or preparing food. When water isn't accessible, keep an alcohol-based hand sanitizer on hand.

  • Food intake should be handled with caution. Cook all meat and fish thoroughly. Avoid fruits and vegetables that cannot be peeled, particularly lettuce, and wash all food before peeling it. To increase safety, you should avoid any raw foods.

  • Avoid close contact with sick people, especially family members and coworkers.

Conclusion

Myelodysplastic syndrome, or MDS, primarily affects the elderly and is frequently detected inadvertently in patients who do not exhibit symptoms. Some patients may exhibit symptoms such as decreased platelets, white blood cells, and red blood cells. Blood and bone marrow are examined for abnormalities during the diagnostic process. Treatments centered on assistance, such as drugs and transfusions. In some situations, chemotherapy such as lenalidomide, azacitidine, or decitabine may be required. Allogeneic hematopoietic stem cell transplantation is the only possible treatment along with metabolic interventions. 


https://academic.oup.com/ajcp/article/148/1/49/3867561?login=false https://www.ncbi.nlm.nih.gov/books/NBK534126/ https://www.annalsofoncology.org/article/S0923-7534(19)35341-4/fulltext https://acsjournals.onlinelibrary.wiley.com/doi/10.1002/cncr.30585 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6939518/ https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8416074/ https://www.cancer.gov/types/myeloproliferative/patient/myelodysplastic-treatment-pdq# https://www.mayoclinic.org/diseases-conditions/myelodysplastic-syndrome/diagnosis-treatment/drc-20366980 https://emedicine.medscape.com/article/207347-overview#a4 https://www.cancer.org/cancer/types/myelodysplastic-syndrome/about/what-is-mds.html https://my.clevelandclinic.org/health/diseases/6192-myelodysplastic-syndrome-myelodysplasia https://www.mds-foundation.org/what-is-mds/