Nonmalignant Leukocyte Disorders PDF

Summary

This document covers nonmalignant leukocyte disorders, including their objectives, outline, and a case study. It discusses various conditions like primary immunodeficiency disorders, and lysosomal storage diseases. The keywords related to the document are leukocyte disorders, hematology, medical science, blood diseases.

Full Transcript

PART V Leukocyte Disorders 26 Nonmalignant Leukocyte Disorders Steven Marionneaux* OBJECTIVES Aer the completion of this chapter, the reader will be able to: 6. List acquired conditions associated with quantitative 1. Compare the genetic defects, pathologic mechanisms,...

PART V Leukocyte Disorders 26 Nonmalignant Leukocyte Disorders Steven Marionneaux* OBJECTIVES Aer the completion of this chapter, the reader will be able to: 6. List acquired conditions associated with quantitative 1. Compare the genetic defects, pathologic mechanisms, changes in leukocyte subtypes. and clinical and laboratory ndings in the primary 7. Compare invivo and invitro alterations in leukocyte immunodeciencies discussed in the chapter. morphology with normal cells. 2. Explain how the presence of Pelger-Huët cells during blood 8. Outline the pathogenesis and clinical and laboratory lm review could be misinterpreted as a neutrophilic le shi. features of infectious mononucleosis. 3. Describe peripheral blood lm abnormalities in Alder- 9. Given the patient history, clinical and laboratory ndings, Reilly anomaly and May-Hegglin anomaly and their and representative cells from a peripheral blood lm or similarity to other conditions. bone marrow smear, interpret results to determine the 4. Associate various lysosomal storage diseases with their type of nonmalignant leukocyte disorder, and recommend underlying pathologic conditions and clinical and additional tests to conrm a diagnosis. laboratory manifestations. 5. Dene neutrophilia, neutropenia, lymphocytosis, lymphocytopenia, monocytosis, monocytopenia, eosinophilia, and basophilia. OUTLINE Primary Immunodeciency Disorders/Inborn Errors of Neutrophil Hypersegmentation Immunity Alder-Reilly Anomaly Severe Combined Immune Deciency May-Hegglin Anomaly Wiskott-Aldrich Syndrome Quantitative Abnormalities of Leukocytes 22q11.2 Deletion Syndrome Quantitative Abnormalities of Neutrophils Bruton Tyrosine Kinase Deciency Quantitative Abnormalities of Eosinophils Chédiak-Higashi Syndrome Quantitative Abnormalities of Basophils Congenital Defects of Phagocyte Number and/or Function Quantitative Abnormalities of Monocytes Lysosomal Storage Disorders Quantitative Abnormalities of Lymphocytes Mucopolysaccharidoses Secondary Morphologic Changes in Leukocytes Sphingolipidoses Storage Artifacts in Leukocytes Morphologic Abnormalities of Leukocytes Without Secondary Changes in Neutrophils and Monocytes Associated Immune Deciency Eosinophils and Basophils Pelger-Huët Anomaly Lymphocytes Pseudo– or Acquired Pelger-Huët Anomaly Infectious Mononucleosis *e author extends appreciation to Scott Dunbar for his assistance with some of the gures in this chapter. 464 CHAPTER 26 Nonmalignant Leukocyte Disorders 465 CASE STUDY After studying the material in this chapter, the reader should be able to respond 3. Provide an approach to correcting the results of the WBC differential that to the following case study. Answers can be found in Appendix C. does not require repeating the test. A 25-year-old woman was seen by her primary care physician with a complaint 4. What are clinical implications of reporting the manual differential results of fatigue. Physical examination revealed pale conjunctiva. The remainder of above if a left shift does not actually exist? the examination was unremarkable. The patient indicated she was experiencing heavier than normal bleeding during menses. Complete blood count results were as follows. Reference intervals can be found after the Index at the end of the book. WBC 8.1 × 109/L MCHC 29 g/dL HGB 8.6 g/dL PLT 172 × 109/L MCV 65 fL Because of the low mean cell volume, a blood lm review was performed. Appar- ent immature granulocytes were present (Figure 26.1) and a manual differential was performed: Segmented neutrophils 9 Lymphocytes 30 Bands 21 Monocytes 7 Metamyelocytes 16 Eosinophils 2 Myelocytes 14 Basophils 1 1. Do you agree with the classication of the immature granulocytes shown? If Figure 26.1 Examples of Cells Classied as Immature Granulocytes in the Differential for the Patient in the Case. (Peripheral blood, Wright- not, provide an explanation in morphologic terms. Giemsa stain, Cellavision DC-1, ×1000.) (Courtesy Cellavision AB, Lund, 2. The patient was subsequently diagnosed with iron deciency anemia. Based Sweden.) on the cell images shown, what other hematologic diagnosis is suspected? is chapter focuses on leukocyte disorders not associated with Severe Combined Immune Deficiency clonal or neoplastic changes in hematopoietic precursor cells. Severe combined immune deciency (SCID) includes a group e cause of these disorders can be genetic or acquired, and of IEIs associated with defects in both cellular and humoral result in quantitative, functional, and/or morphologic changes immunity.1 Nearly all patients with SCID experience a marked in one or more lineages: neutrophils, lymphocytes, monocytes, decrease in circulating T cells, poorly functioning B cells, eosinophils, and/or basophils. Some of these disorders are asso- hypogammaglobulinemia, and profound clinical symptoms ciated with signicant clinical manifestations. that manifest in infancy. Le untreated, most patients with SCID die within the rst 2 years of life from overwhelming PRIMARY IMMUNODEFICIENCY DISORDERS/ bacterial, viral, and/or fungal infections. Potentially cura- tive options in SCID are allogeneic hematopoietic stem cell INBORN ERRORS OF IMMUNITY transplantation (HSCT) and gene therapy, in which the goal As of June 2022, nearly 500 (inherited) inborn errors of immu- is reconstitution of the failed immune system. e outcomes nity (IEI) or primary immunodeciency disorders have been in patients treated with HSCT have improved over the past identied, each originating from a single germline genetic decade. Most patients with SCID who undergo HSCT can abnormality aecting the innate or adaptive immune system.1 expect to survive well into adulthood.3 In the United States, Most individual IEIs are rare; however, as a group, the preva- gene therapy is not yet approved for SCID. However, gene ther- lence of diagnosed IEIs the United States is approximately 1 in apy is an option for patients treated in clinical trials in which 1200 individuals.2 there has been promising success. However, even with gene e mechanisms of disease, clinical manifestations, and therapy, there remains a signicant risk of leukemic transfor- disease severity among the IEIs can vary based on the nature mation in SCID patients.1 of the gene defect and its pattern of inheritance. In each IEI, a missense mutation, deletion, or frame shi mutation leads to a Common Gamma Chain Deficiency/X-Linked Severe reduced quantity and/or quality of the encoded protein, altering Combined Immune Deficiency normal functioning within the innate or adaptive immune sys- Common gamma chain deciency/X-linked SCID is classied tem. is can have a wide range of eects from life-threatening as T–B+ SCID1 and is the most common SCID. is disease manifestations that arise in infants to manageable disorders initiates from a mutation in IL2RG located at Xq13.1. IL2RG diagnosed in adults. normally encodes the gamma chain protein within receptor is section illustrates the heterogenous clinical presenta- complexes that bind interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15, tions associated with IEI by reviewing a select number of dis- and IL-21. ese interleukins provide growth, dierentiation, eases within the classication schema from the International and survival signals for T and natural killer (NK) lymphocytes. Union of Immunological Societies Expert Committee1 and will In common gamma chain deciency, a mutation in IL2RG mainly include diseases associated with peripheral blood mor- leads to truncated gamma chain proteins and faulty signal phologic abnormalities. transduction that impairs T and NK cell development. Infants 466 PART V Leukocyte Disorders with the disease have no thymus, tonsils, or lymph nodes and ere is, however, a signicant risk for the development of acute oen experience severe life-threatening infections within the leukemia. rst few months of life as protective maternal immunoglobu- lins are depleted. Prompt diagnosis of common gamma chain 22q11.2 Deletion Syndrome deciency is key in implementing preventive measures in antic- 22q11.2 deletion syndrome (22DS)/DiGeorge syndrome/velo- ipation of these infections. e white blood cell (WBC) count cardiofacial syndrome is classied as a combined immuno- in aected infants is decreased as a result of severely decreased deciency with associated syndromic features.1 e genetic T and NK lymphocytes. B cells are generally adequate in num- aberration in 22DS is a heterozygous microdeletion of 1.5 to ber but are dysfunctional without T cell signaling. In common 3 million base pairs on chromosome 22 at q11.2 resulting in a gamma chain deciency, there is failure to thrive and death usu- loss of over 100 genes and their encoded protein products.9,10 ally occurs before 2 years without HSCT. e deletion breakpoint and genes lost in the deletion can dier among patients, contributing to the variable clinical phenotype. Adenosine Deaminase Deficiency (ADA-SCID) Haploinsuciency caused by the loss of T-box transcription Adenosine deaminase deciency (ADA-SCID) is classied as factor 1 gene (TBX1), DiGeorge Syndrome Critical Region 8 T–B– SCID1 and represents 10% to 20% of SCID cases. It is an (DGCR8), and several microRNAs result in congenital malfor- autosomal recessive disorder caused by a mutation in the ADA mations and other clinical manifestations in 22DS.10 Variable gene located at chromosome 20q13.12. Adenosine deaminase degrees of thymic hypoplasia proportionally aect the severity is a key component in the metabolic breakdown of adenosine of immune dysfunction, autoimmunity, and T lymphopenia. triphosphate (ATP) and RNA in all cells. In ADA-SCID, there Abnormal development of the embryonic pharyngeal system is intracellular and extracellular accumulation of toxic levels of during gestation produces characteristic cardiac anomalies, thy- adenosine and deoxyadenosine in lymphocytes, causing pro- mus and parathyroid hypoplasia, and craniofacial deformations found decreases in T, B, and NK cells. Patients with ADA-SCID in 22DS patients. A wide array of additional clinical ndings experience severe recurring, life-threatening bacterial, viral, includes intrauterine growth restriction, major infections, fail- and fungal infections beginning early in life. Additional man- ure to thrive, short stature, skeletal defects, neuropsychiatric ifestations include skeletal abnormalities, neurologic decits, diseases, developmental delays, gastrointestinal diseases, oph- and skin rashes.1 thalmologic disorders, hypocalcemia, immune thrombocy- topenia, and autoimmune hemolytic anemia. Genetic testing Wiskott-Aldrich Syndrome conrms the 22DS diagnosis in patients with the characteristic Wiskott-Aldrich syndrome (WAS) is classied as a combined presentation. e survival of aected infants is oen less than immunodeciency with associated or syndromic features.1 1 year without thymic tissue transplantation or HLA-matched WAS is a rare X-linked disease caused by one of more than HSCT with memory T cells. Gra-versus-host disease is a 450 mutations in the WAS gene,4 which results in decreased common complication aer HSCT. In a survey of 17 athymic Wiskott-Aldrich syndrome protein (WASP) expression. patients with 22DS who underwent HSCT between 1995 and Disease severity and clinical manifestations in WAS can vary 2006, 41% were alive at a median followup of 5.8 years.11 based on the type of mutation present and its impact on WASP expression. WASP controls RNA polymerase II–depen- Bruton Tyrosine Kinase Deficiency dent transcription and has a critical role in actin cytoskeleton Classied as an antibody deciency, Bruton tyrosine kinase remodeling of hematopoietic cells. Low WASP levels have an (BTK) deciency/X-linked agammaglobulinemia is caused by impact on the stability of the immunologic synapse, the site a mutation in the gene encoding BTK (Xq21.3-Xq22), a signal where T-cells and antigen-presenting cells interact. Abnormal transduction protein that promotes B-cell lineage develop- reorganization of the cytoskeleton results in defective T cell ment.1 A mutation in BTK leads to marked reduction in B cells, function and impaired interaction with lymphocytes, lead- absent tonsils and adenoids, inability to produce plasma cells, ing to decreased B cells. NK cells are normal or increased in and therefore severe decreases in all immunoglobulin isotypes number but have diminished cytotoxic activity. Regulatory and grossly defective antibody responses. e lack of a humoral T cells involved in autoimmunity are dysfunctional.5 WASP immune response leaves the patient susceptible to infections deciency also diminishes neutrophil chemotaxis, phagocy- from encapsulated bacteria, bloodborne enteroviruses, and cer- tosis and other steps involved in the killing of microorgan- tain fungal and parasitic infections. isms, which increases susceptibility to infections. ere is a e disease manifests clinically aer maternal antibodies clear risk of bleeding because of thrombocytopenia, small abnormal from the BTK-decient infant’s circulation, between 3 and 18 platelets, and shortened platelet survival, which is the result of months. Recurrent otitis, conjunctivitis, diarrhea, and sinus and increased immune-related and nonimmune-related destruc- skin infections are common. e disease is oen rst suspected tion and ineective megakaryocytopoiesis. when the child experiences a severe life-threatening bacterial WAS diagnosis is typically conrmed through WAS gene infection. Molecular genetic testing is needed to conrm the diag- sequencing in patients with suspected disease. HSCT can be nosis. e management of BTK deciency consists primarily of curative in patients with human leukocyte antigen (HLA)- immunoglobulin replacement therapy. Most patients diagnosed matched donors.6,7 Gene therapy is a promising approach most today will live well into adulthood because of earlier recognition, appropriate for patients with WAS who lack a matched donor.8 advancements in immunoglobulin therapies, and improvements CHAPTER 26 Nonmalignant Leukocyte Disorders 467 A B C Figure 26.2 Chédiak-Higashi Syndrome. Fused cytoplasmic granules in (A), Neutrophil. (B), Basophil. (C), Eosinophil. (Peripheral blood, Wright-Giemsa stain, Cellavision DC-1, ×1000.) (Courtesy Cellavision AB, Lund, Sweden.) in antimicrobials. Recent studies of gene therapy to restore nor- phagocytic cells of the innate immune system, primarily neutro- mal BTK function in mice have shown promising ndings.12 phils, monocytes, and/or macrophages. As of 2022, there were 42 distinct disease entities, each originating from a unique genetic Chédiak-Higashi Syndrome abnormality.1 ese diseases are organized into four subgroups, Chédiak-Higashi syndrome (CHS) is a rare autosomal reces- (1) congenital neutropenias, (2) defects of motility, (3) defects sive disease of immune dysregulation classied as a familial of respiratory burst, and (4) other nonlymphoid defects. A com- hemophagocytic lymphohistiocytosis syndrome with hypopig- mon thread in all phagocyte diseases is an increased susceptibil- mentation.1 13 CHS is caused by a mutation in the lysosomal ity to bacterial and fungal infections. Dermatologic symptoms, tracking regulator (LYST) gene on chromosome 1q42.3. e delayed wound healing, growth failure, and inammation of the mutation leads to the formation of giant fused cytoplasmic gran- oral mucosa are also common. A wide array of additional clin- ules/inclusions in leukocytes, platelets, broblasts, melanocytes, ical symptoms are associated with the individual diseases. is and other granulated cells in nerve tissue, kidney, and gastric section highlights several phagocyte disorders that arise from mucosa. On peripheral blood lms, the inclusions can be seen dierent underlying mechanisms. in granulocytes (Figure 26.2), monocytes, large granular lym- phocytes (NK and cytotoxic T cells), and platelets. Patients with Congenital Neutropenia CHS oen have cytopenias, particularly neutropenia. Clinical Congenital neutropenia (absolute neutrophil count [ANC] less manifestations present in infancy as partial albinism, severe than 1.5 × 109/L) is associated with a variety of nonmalignant recurrent life-threatening bacterial infections, mild bleeding, conditions covered in this chapter in which the decrease in neu- easy bruising, and progressive neurologic impairment. In sus- trophils results from decreased production, increased destruc- pected patients, genetic testing for mutations in LYST is done tion, or apoptosis. e congenital neutropenias (CNs) are a to conrm the diagnosis. Patients with CHS are at high risk of group of inherited diseases in which the primary defect is in the developing an aggressive, life-threatening syndrome of exces- production of neutrophils.1 Common to all CNs is early clinical sive immune activation known as hemophagocytic lymphohis- onset, frequently in infancy, and an increased risk of primarily tiocytosis. Most children with CHS will succumb to the disease bacterial and fungal infections arising from neutropenia rather before 7 years unless treated with HSCT.14 HSCT using matched than some other immune deciency. In some CNs, there are donors has proven successful; however, it does not prevent neu- nonhematologic manifestations such as neurologic/cognitive rologic deterioration.15 defects, dysmorphic features, and organ dysfunction. Pseudo–Chédiak-Higashi (PCH) granules/inclusions resem- Severe congenital neutropenia. Severe congenital ble the fused lysosomal granules in CHS. PCH granules are neutropenia (SCN) is a subset of CN characterized by an ANC seen most oen in acute myeloid leukemia (AML). One study less than 0.5 × 109/L (oen less than 0.2 × 109/L) and recurring, reported PCH granules in the leukemic cells of 5 of 20 children severe, oen life-threatening bacterial and fungal infections with with AML.16 PCH granules are also associated with chronic or without nonhematologic manifestations and an increased myeloid leukemia, myelodysplastic neoplasms (MDSs), and risk of malignancy, frequently MDS or AML. Mutations in the rarely, in acute lymphoblastic leukemia.17 neutrophil elastase gene ELANE (causing SCN1) are responsible for 50% to 60% of all SCN cases.18 Ten subtypes of SCN are Congenital Defects of Phagocyte Number and/or listed in Table 26.1 1 19 20 Function Bone marrow examination in patients with SCN shows a e congenital defects of phagocytes include a heteroge- maturation arrest in the neutrophil series at the myelocyte/pro- neous group of qualitative and quantitative disorders aecting myelocyte stage and dysmorphic changes in neutrophilic cells. 468 PART V Leukocyte Disorders TABLE 26.1 Severe Congenital Neutropenias Subtype Gene Inheritance Location Disease Associated Features* SCN1 ELANE AD 19p13.3 Elastase deciency Mild anemia, thrombocytopenia, monocytosis, eosinophilia, osteopenia SCN2 GFI1 AD 1p22.1 GFI1 deciency Monocytosis, lymphopenia, IG SCN3 HAX1 AR 1q21.3 Kostmann disease Neurologic defects SCN4 G6PC3 AR 17q21.31 G6PC3 deciency BM dysplasia, anemia, thrombocytopenia, HSM; cardiac, urogenital, skeletal, endocrine, and vascular abnormalities, deafness SCN5 VPS45 AR 1q21.2 VPS45 deciency BM brosis, EMH, anemia, anisopoikilocytosis, thrombocytopenia, NRBC, enlarged kidneys, HSM, delayed development, blindness, no response to G-CSF in some SCN6 JAGN1 AR 3p25.3 JAGN1 deciency Short stature, facial dysmorphia, hypothyroidism, urogenital anomalies SCN7 CSFCR AR 1p34.3 GCSF receptor deciency No response to G-CSF SCN8 SRP54 AD 14q13.2 SRP54 deciency Groups A and B patients: short stature, pancreas dysfunction, poor response to G-CSF Group B also has developmental delay, autism SCN9 CLPB AD 11q13.4 3-Methylglutaconic deciency Neurocognitive defects, cataracts, cardiac defects, facial dysmorphia, hypothroidism SCNX WAS XL Xp11.23 X-linked neutropenia Low normal platelets, monocytopenia *SCN1–SCN9 and SCNX are all associated with severe neutropenia and recurring acute, severe, life-threatening infections. AD, Autosomal dominant; AR, autosomal recessive; BM, bone marrow; EMH, extramedullary hematopoiesis; HSM, hepatosplenomegaly; IG, immature granulocytes in blood; NRBC, nucleated red blood cells in peripheral blood; SCN, severe congenital neutropenia; XL, sex-linked. e mainstay of treatment in SCN is long-term granulo- chemotaxis. Patients are usually diagnosed in infancy with cyte colony-stimulating factor (G-CSF), which is eective in pancreatic insuciency and associated malabsorption and most patients at increasing the absolute neutrophil count and steatorrhea. ere are also skeletal abnormalities, severe decreasing the rate of infections and mortality. While the risk infections, and failure to thrive. Additional manifestations of malignancy in SCN is thought to be a complication of the of SDS oen include developmental delays and bone marrow disease, long-term G-CSF therapy increases this risk.21 Aer failure with dysplasia, cytopenias, and increased risk of 15 years on G-CSF, the cumulative risk of AML/MDS is 22%.22 AML or MDS, particularly in patients who develop multiple Patients who do not respond to G-CSF or require higher doses hematopoietic cell clones with TP53 mutations.23 e to maintain adequate neutrophil counts and infection prophy- diagnosis of SDS in suspected patients is achieved though laxis are oen considered for HSCT. molecular genetic testing, typically DNA sequencing. G-CSF Cyclic neutropenia. Cyclic neutropenia is a CN caused by is used in patients with neutropenia and severe infections. ELANE mutations but is a less severe disease compared to Allogeneic HSCT can potentially cure SDS patients with bone SCN1, which also stems from ELANE mutations. Patients with marrow failure and MDS or AML; however, there is no eect cyclic neutropenia experience characteristic episodes of severe on the nonhematologic manifestations. Outcome data in SDS neutropenia (less than 0.2 × 109/L) together with absolute patients treated with HSCT are limited. monocytosis in approximately 21-day cycles, each lasting 3 to 5 or more days. During the ANC nadir, patients oen experience Defects of Motility mouth ulcers, sore throat, gingivitis, rash, fatigue, fever, and/ As leukocytes (primarily neutrophils and monocytes) leave cir- or cervical lymphadenopathy. e risk of life-threatening culation to move to a site of infection, they adhere to the vascu- infections in cyclic neutropenia is much lower than in SCN1 lar endothelium and tissue matrix and roll along the vessel wall. and there is no increased risk of malignant transformation. Adhesion molecules such as selectins and integrins expressed on Shwachman-Diamond syndrome. Shwachman-Diamond endothelial cells and leukocytes mediate this process, interact- syndrome (SDS) is classied as a CN with associated neutrophil ing with ligands on the surface of leukocytes to slow the speed of functional defects.1 SDS is a rare disease but a common bone neutrophils in circulation. Ligand binding induces high-anity marrow failure syndrome. SDS is caused by a mutation in attachment of integrins with endothelial cell receptors, facilitat- SBDS which encodes SBDS, a key component in ribosome ing leukocyte transmigration between gaps in endothelial cells maturation, cell proliferation, and maintenance of the bone (diapedesis) (Chapter 10). marrow microenvironment. Biallelic mutations in SBDS Leukocyte adhesion disorders. Leukocyte adhesion occur in most patients, resulting in a truncated nonfunctional disorders (LADs) are rare autosomal recessive primary SBDS protein and both neutropenia and impaired neutrophil immunodeciency disorders characterized by the inability of CHAPTER 26 Nonmalignant Leukocyte Disorders 469 neutrophils and monocytes to move from peripheral circulation however, the integrins fail to respond to external signals, to sites of infection. Patients with LAD have recurring infections which limits neutrophil adhesion, NK cell activity, and platelet and impaired wound healing and, depending on the LAD function. Patients with LAD-III experience severe recurrent subtype, they may also have developmental abnormalities and/ infections, osteoporosis, and bleeding. Without treatment with or increased risk of bleeding. Absolute neutrophilia is present HSCT, the prognosis of infants with LAD-III is poor. in LAD, especially evident during infections. LAD subtype is determined through genetic testing, typically DNA sequencing. Defects of Respiratory Burst: Chronic Granulomatous Allogeneic HSCT has been shown to be a potentially curative Disease therapy in severe LAD-I and LAD-III. A 2021 review of Chronic granulomatous disease (CGD) is a rare condition transplant data in 84 patients from 33 sites found a 3-year caused by the decreased ability of neutrophils, monocytes, overall survival estimate of 83%.24 ere are three subtypes of macrophages, and eosinophils to kill phagocytized bacteria LAD. and yeast. This is due to a failure in the respiratory burst Leukocyte adhesion disorder type I. LAD-1 is the most and failed production of powerful antimicrobial superox- common type, caused by a mutation in ITGB2, which encodes ide anions and other reactive oxygen species, including the CD18 subunit of β2 integrins. e mutation leads to hydrogen peroxide, hypochlorous acid, and others. The decreased production or a truncated form of β2 integrin and failed respiratory burst in CGD is caused by a mutation diminished neutrophil adhesion to endothelial cells, recognition in one of six genes that encode protein components in the of bacteria, and outside-in signaling. In LAD-I, neutrophils reduced form of nicotinamide adenine dinucleotide phos- accumulate in peripheral circulation but cannot reach sites of phate (NADPH) oxidase, which is required in the respiratory infection in tissues because of the adhesion problem. T-cell burst. Approximately two-thirds of CGD cases are inherited function is also impaired in LAD-I. As of March 2023, 177 as X-linked recessive and one-third are autosomal recessive. dierent mutations had been described25 that can have diering X-linked recessive patients generally have a more severe dis- eects on the quantity and quality of the encoded β2 integrin. ease course with shortened survival compared with autoso- Shortly aer birth, LAD-I infants experience severe recurring mal recessive patients. In normal individuals, phagocytosis infections, most oen of the skin, oral mucosa, gastrointestinal of foreign organism leads to phosphorylation and binding tract, respiratory tract, and perirectal area. of cytosolic p47phox and p67phox. Primary granules con- In addition, LAD-I infants usually have lymphadenopathy, taining antibacterial neutrophil elastase and cathepsin G splenomegaly, neutrophilia, and lack of pus formation. e clin- and secondary granules containing the cytochrome com- ical severity of LAD-I is heterogeneous and largely related to the plex gp91phox and p22phox migrate to the phagolysosome. specic ITGB2 mutation and its impact on β2 integrins.25 NADPH oxidase forms when p47phox and p67phox, along Leukocyte adhesion disorder type II. LAD-II is caused with p40phox and RAC2, combine with the cytochrome by 1 of 16 mutations in SLC35C1, which codes for a fucose complex. The principal antimicrobial agent, superoxide, transporter in the Golgi system.25 e mutation leads to a lack is generated in the phagolysosome during the respiratory of or a dysfunctional transporter and diminished fucosylation burst, when an electron from NADPH is added to oxygen. In of cell surface glycoproteins that serve as ligands for L-selectins addition, NADPH has regulatory functions in the generation on leukocytes. e adhesion and rolling of leukocytes along of other antimicrobial agents. Most cases of CGD are due endothelial cells of the vessel wall toward the site of infection to mutations in the genes coding for gp91phox or p47phox. depends on the binding of L-selectins on leukocytes with E or The six genes implicated in CGD and their encoded protein P selectins on endothelial cells, with their matching ligands products are listed in Table 26.2 26 on opposite cells. erefore in LAD-II, a lack of fucosylated Patients with CGD experience life-threatening catalase- glycoprotein ligands for L-selectin decreases the adhesion positive bacterial and fungal infections, as well as inammatory of leukocytes to the vascular endothelium and impairs their complications such as colitis and granuloma formation in the transmigration to sites of infection. Fortunately, β2 integrins gastrointestinal tract, urinary tract, and other organs. Neutrophil are also activated through alternate pathways not aected by the SLC35C1 mutation. is provides partial functioning leukocyte adhesion and transmigration that is less impaired TABLE 26.2 Genes Involved in Chronic than that in LAD-I. Consequently, infections in LAD-II patients Granulomatous Disease tend to be fewer and less severe than in LAD-I. On the other Gene Location Inheritance Protein Product hand, the lack of fucosylation in LAD-II aects glycoproteins CYBB Xp21.1 XLR gp91phox other than selectins, leading to severe cognitive impairment, CYBA 16q24 AR p22phox skeletal abnormalities, growth retardation, and distinctive facial NCF1 7q11.23 AR p47phox features. NCF2 1q25 AR p67phox Leukocyte adhesion disorder type III. LAD-III is caused by 1 of 31 mutations in FERMT3 25 FERMT3 encodes kindlin-3 NCF4 22q13.1 AR p40phox (fermitin family homolog 3), which is required for normal CYBC1(EROS) 17q25.3 AR Essential for reactive inside-out signaling of hematopoietic cells and activation of β1, oxygen species (EROS) β2, and β3 integrins. In LAD-III, integrin expression is normal; AR, Autosomal recessive; XLR, sex-linked recessive. 470 PART V Leukocyte Disorders function tests such as dihydrorhodamine 123 or nitroblue tetra- (MPSs), lipoprotein storage disorders, and lysosomal transport zolium test can be used to screen suspected patients. Positive defects. e more common LSDs are presented in this chapter. screening tests are followed with DNA sequencing to conrm the diagnosis. e optimal management of mild to moderately Mucopolysaccharidoses severe CGD involves long-term antimicrobial prophylaxis plus e MPSs are a rare group of inherited disorders of mucopoly- quick recognition and aggressive treatment of acute infections saccharide or glycosaminoglycan degradation. e combined and inammatory complications. Over the past two decades, incidence in the United States for all subtypes is 1 per 100,000 live studies using HSCT in CGD have shown signicant improve- births.33 MPS types I, II, and III have the highest incidence. As of ments in outcomes and reductions in morbidity and mortality. 2021, only four cases of MPS type IX had been reported.33 e A 2018 review of data from nine studies reported an overall MPSs are caused by reduced activity in enzymes necessary for the event-free survival of more than 80% and overall survival of degradation of one or more substrates: dermatan sulfate, heparan approximately 90% at 3 years aer HSCT.27 sulfate, keratan sulfate, chondroitin sulfate, and hyaluronan. e partially degraded material builds up in lysosomes, causing serious Enzymatic White Blood Cell Defects: Myeloperoxidase clinical consequences and shortened survival. Table 26.3 provides Deficiency genetic, biological, and clinical information about MPS subtypes, Myeloperoxidase (MPO) deciency is the most common inher- along with the decient enzymes and accumulated substrates.32,34,35 ited disorder of phagocytes. MPO is an enzyme found in high e peripheral blood lms of patients with MPS may show concentration in primary azurophilic granules of neutrophils metachromatic Reilly bodies (acquired Alder-Reilly anomaly and in lower concentration in the lysosomal granules of mono- [ARA]) in neutrophils, monocytes, and lymphocytes. Bone cytes. In neutrophils, MPO catalyzes the conversion of H2O2 to marrow macrophages (histiocytes) also can demonstrate cyto- HOCL and other microbicidal reactive oxygen intermediates. plasmic metachromatic material. Diagnosis relies on assays that MPO plays an important role in the killing of phagocytized bac- measure the specic enzymes decient in each subtype. Genetic teria and yeast and participates in the formation of neutrophil testing to identify pathologic mutations can be performed for extracellular traps (NETs), another mechanism for eliminating diagnostic conrmation. MPS treatment includes supportive microorganisms.28 e MPO gene is located on chromosome care, enzyme replacement therapy, and/or HSCT. Gene therapy 17q23. Pathogenic mutations lead to decreased MPO pro- is a promising treatment approach and clinical studies are ongo- duction, limiting its participation in microbicidal activities. ing in patients with MPS types I, II, IIIA, and IV.36 Nevertheless, most MPO-decient individuals are asymptom- atic. is is because neutrophils employ additional pathways Sphingolipidoses for generating microbicidal agents. Although less potent, these e sphingolipidoses are a group of LSDs with functional pathways provide adequate protection against pathogens in the deciencies in enzymes needed for lysosomal breakdown of majority of individuals with MPO deciency. However, there sphingolipids, causing harmful accumulation of various phos- have been reports of increased susceptibility to fungal infections pholipids in tissues. e pathways involved in sphingolipid in MPO deciency when comorbid diseases are present.29,30 catabolism, specic enzyme deciencies, and corresponding Automated hematology analyzers that use MPO in the WBC storage diseases are outlined in Figure 26.3. Gaucher disease dierential method are limited in the analysis of MPO-decient and Niemann-Pick disease are examples of sphingolipidoses patients (Chapter 13). and are described in more detail in the following sections. Gaucher Disease LYSOSOMAL STORAGE DISORDERS Gaucher disease (GD) is the most common LSD. It is an auto- e inborn errors of metabolism consist of 1450 disorders somal recessive disorder triggered by a defect or deciency in divided into 24 categories and 124 groups.31 ese are mostly the catabolic enzyme β-glucocerebrosidase, which is necessary single gene defects aecting enzymes that metabolize substrates for glycolipid metabolism. As a result, unmetabolized substrate into various components. In most cases, the enzyme deciency sphingolipid glucocerebroside accumulates in the lysosomes leads to a buildup of toxic substances that are the source of clini- of macrophages throughout the body, including osteoclasts in cal manifestations. e inborn errors of metabolism include the bone and microglia in the brain.37 GD stems from a mutation in lysosomal storage disorders (LSDs) a group of over 70 inherited GBA located at 1q21-q22. More than 400 pathogenic mutations diseases. In the LSDs, single gene mutations negatively aect the have been reported and, although some correlations have been production, transport and secretion of lysosomal enzymes.32 found between specic mutations and patient outcomes, most Failed degradation of macromolecules and gradual buildup clinical phenotypes cannot be predicted by genotype.38 (“storage”) of undigested substrates within lysosomes result in ere are three types of GD, and type I is by far the most cell dysfunction, cell death, and a variety of clinical symptoms. common. Neurologic symptoms are key factors in dierenti- All cells containing lysosomes are potentially aected. LSDs are ating subtypes. e phenotype is quite heterogeneous. Some classied according to the undigested macromolecule(s) that patients are asymptomatic, whereas others experience a multi- accumulates in the cell. Subtypes of LSDs include sphingolipido- tude of clinical problems. Clinical information associated with ses, pseudobasophilia, mucolipidoses, mucopolysaccharidoses the three types of GD is provided in Table 26.4. CHAPTER 26 Nonmalignant Leukocyte Disorders 471 TABLE 26.3 Mucopolysaccharidoses Gene and Name Also Known As Enzyme Deficiency AS Location Clinical Features MPS I Hurler syndrome,* Alpha-l-iduronidase DS + HS IDUA Severe phenotype (Hurler): coarse facial features, Hurler-Scheie 4p16.3 skeletal abnormalities, severe intellectual disability, syndrome,† and hepatosplenomegaly, corneal clouding, umbilical Scheie syndrome† hernia, frequent upper respiratory tract infections MPS II Hunter syndrome Iduronate 2-sulfatase DS + HS IDS Severe phenotype: coarse facial features, Xq28 hepatosplenomegaly, short stature, skeletal deformities, cardiovascular disorders, deafness, neurocognitive issues MPS III A Sanlippo syndrome N-Sulfoglucosamine HS SGSH Hirsutism, coarse facial features, behavioral problems, sulfohydrolase 17q25.3 aggressive behavior, speech delay, hepatomegaly MPS III B Sanlippo syndrome Alpha-N- HS NAGLU Cardiomegaly, coarse facial features, progressive acetylglucosaminidase 17q21.2 dementia, convulsions, MPS III C Sanlippo syndrome Heparan-alpha- HS HGSNAT Delayed psychomotor development, behavioral problems, glucosaminide 8p11.21 sleep disorder, hearing loss, coarse facial features, N-acetyltransferase recurring infections, diarrhea, epilepsy, and retinitis pigmentosa MPS IV A Morquio syndrome N-Acetylgalactosamine-6- KS + CS GALNS Musculoskeletal abnormalities, short stature, pulmonary sulfatase 16q24.3 and cardiac dysfunction, hearing loss, corneal clouding MPS IV B Morquio syndrome Beta-galactosidase KS GLB1 Musculoskeletal abnormalities, short stature, pulmonary 3p22.3 and cardiac dysfunction, hearing loss, corneal clouding MPS VI Maroteaux-Lamy Arylsulfatase B DS + CS ARSB Severe form: coarse facial features, severe skeletal syndrome 5q14.1 abnormalities, joint stiffness, respiratory disease, cardiac disease, sleep apnea, pulmonary hypertension, corneal clouding, hepatosplenomegaly, macrocephaly, hearing loss MPS VII Sly disease Beta-glucuronidase HS + DS + GUSB Varies widely CS 7q11.21 Infantile: hydrops fetalis Severe: like MPS I (Hurler) Intermediate: skeletal abnormalities, coarse facial features, variable cognitive impairment Mild: skeletal abnormalities MPS IX Natowicz syndrome Hyaluronidase-1 HYAL HYAL1 Facial dysmorphism; hepatosplenomegaly; cardiac, 3p21.31 respiratory, and skeletal involvement; and neurologic, hematologic, and ocular symptoms *Most severe. †Attenuated phenotype. AS, Accumulated substrate; CS, chondroitin sulfate; DS, dermatan sulfate; HYAL, hyaluronan; HS, heparin sulfate; KS, keratan sulfate; MPS, mucopolysaccharidoses. In GD, macrophages containing excess glucocerebroside increased risk for developing hematopoietic neoplasms, most accumulate in the spleen, liver, and bone marrow and are pri- frequently plasma cell (multiple) myeloma.40 41 marily responsible for the disease manifestations. Gaucher Treatment of GD includes enzyme replacement therapy and cells are lysosome-engorged macrophages with eccentric substrate reduction therapy.42 HSCT oers the potential for nuclei and abundant brillar blue-gray cytoplasm with a stri- cure; however, it is not oen used because treatment-related ated or wrinkled appearance (sometimes described as onion mortality is high.43 Gene therapy, genome editing, and chaper- skin-like) (Figure 26.4). Gaucher cells stain positive with one therapy are additional modalities being pursued. trichrome, aldehyde fuchsin, periodic acid-Schi (PAS) and Pseudo-Gaucher cells. Psuedo-Gaucher cells are seen acid phosphatase.39 In untreated patients, Gaucher cells inl- in bone marrow specimens from patients with thalassemia, trate the hematopoietic space in bone resulting in anemia and myeloid neoplasms,44 45 acute lymphoblastic leukemia,46 non- thrombocytopenia. Hodgkin lymphoma,46 and plasma cell neoplasms.47 In these A commercially available test for β-glucosidase (glucocere- diseases, pseudo-Gaucher cells are thought to result from brosidase) is available to conrm the GD diagnosis. Genetic increased cell turnover overwhelming the glucocerebrosidase testing performed in suspected patients to screen for the most enzyme, causing accumulation of glucosylceramide in common mutations, including N370S, 84GG, IVS2 +1G>A, and macrophages. erefore pseudo-Gaucher cells result from a L444P.39 All three types of GD have been associated with an relative rather than true decrease in enzyme. Pseudo- and true 472 PART V Leukocyte Disorders Figure 26.3 Pathways and Diseases of Sphingolipid Metabolism. (From Orkin, S. H., Fisher, D. E., & Look, A. T., et al.. Nathan and Oski’s Hematology of Infancy and Childhood. [7th ed.]. Philadelphia: Saunders.) TABLE 26.4 Clinical Subtypes of Gaucher Disease* Type I: Nonneuronopathic Type II: Acute Neuronopathic Type III: Subacute Neuronopathic Age at presentation Childhood/adulthood Infancy Childhood/adulthood Hepatosplenomegaly + → +++ + + → +++ Skeletal abnormality – → +++ – ++ → +++ CNS disease – +++ + → +++ Life span 6–80+ years T) mutation in polycythemia vera (Chapter 32) and Numerous tumor suppressor genes have now been identi- internal tandem duplication such as the FLT3-ITD mutation in ed, and many have been found to be associated with autosomal acute myeloid leukemia (Chapter 31). JAK2 encodes a nonrecep- dominant familial cancer predisposition syndromes. Some well- tor tyrosine kinase associated with the intracellular domain of known examples of loss or inactivation of tumor suppressor growth factor receptors, such as the erythropoietin receptor, and genes include TP53 in Li-Fraumeni syndrome (mentioned in 494 PART V Leukocyte Disorders TABLE 27.3 Comparison of Oncogenes and Tumor Suppressor Genes Oncogenes Tumor Suppressor Genes Normal functions Promote cell proliferation and differentiation, signal Detect damaged DNA and delay cell cycle to allow for DNA repair or transduction, apoptosis apoptosis Effect of mutations Leukemogenic when inappropriately activated (gain-of-function Leukemogenic when inactivated or deleted (loss-of-function mutation); mutation); causes constitutive (continuous), dysregulated, causes inability to prevent cells with damaged DNA from progressing and/or overexpression of oncogene product through the cell cycle; helps sustain malignant phenotypes Genetics Dominant; only one mutant allele needed for leukemogenesis Recessive; deletion or inactivation of both alleles needed for leukemogenesis Examples of mutations Chromosomal rearrangement (translocation, inversion), point Deletion, inactivating mutation, epigenetic silencing mutation, internal tandem duplication, gene amplication Examples relevant to ABL1, JAK2, BCL2 TP53, RB1, WT1 hematologic neoplasms “Incidence, Prevalence, and Etiology”), RB1 involved in familial BOX 27.2 General Categories of Therapy retinoblastoma, and WT1 in Wilms tumor. More importantly, for Hematologic Neoplasms these tumor suppressor genes are deleted or inactivated in many sporadic (nonfamilial) cancers, including hematologic neo- Chemotherapy plasms, which leads to accumulation of additional mutations Cell cycle effects: phase-specic or phase-nonspecic agents and a more clinically aggressive state. An example is chronic Biochemical mode of action: alkylating agents, plant alkaloids, antimetabo- lites, antitumor antibiotics, glucocorticoids lymphocytic leukemia, which has a more aggressive course Radiation therapy when the TP53 gene is deleted or mutated, warranting a dier- Supportive therapy ent approach to therapy (Chapter 34).25 Growth factors and cytokines Table 27.3 compares characteristics of oncogenes and tumor Targeted therapy suppressor genes. Targeted molecular therapy Immunotherapy DNA Repair Genes Cellular therapy Mutations in DNA repair genes are also involved in hematologic Hematopoietic stem cell transplantation neoplasms. Mutations in these genes cause genetic instability Syngeneic and increased mutation rates, leading to an increased risk of Allogeneic malignant transformation. An example is the Fanconi anemia Autologous gene, FA, which is important for maintaining genomic stability in hematopoietic tissues (mentioned in “Incidence, Prevalence, treatment. In contrast to many solid tumors, numerous hema- and Etiology”). tologic neoplasms now have cure rates that are substantially Regardless of the type of chromosome or genetic abnormality, higher than they were two or three decades ago. Many exciting oncogene activation or the loss or inactivation of tumor suppres- new therapies that are less toxic are now under development sor or DNA repair genes has adverse molecular eects on prolif- or are already employed in patient settings. ese therapies are eration, dierentiation, maturation, survival, apoptosis, cell cycle bringing more optimism to the care of patients with hematologic control, and DNA repair mechanisms in hematopoietic cells and neoplasms. Selection of the best therapy must start, however, an increased risk of malignant transformation. e list of chro- with an accurate diagnosis. Even the most eective therapies do mosomal and molecular aberrations known to occur in various not work if they are applied in the wrong circumstances. hematologic neoplasms continues to grow on an almost daily Curative treatment strategies are now a realistic goal for patients basis. Indexing this list is far beyond the scope of this chapter, with Hodgkin lymphoma, CML, hairy cell leukemia, some forms of but specic examples will be discussed in the chapters that follow. non-Hodgkin lymphoma, and for children with acute lymphoblas- tic leukemia. Cure may be attainable in other patients with acute lymphoblastic or myeloid leukemia, and long-term remissions THERAPY may be achievable in adults with multiple myeloma. For patients Treatment for leukemia and lymphoma may involve chemo- with other hematologic neoplasms such as mantle cell lymphoma, therapy, radiation, supportive therapy, targeted therapies, and chronic lymphocytic leukemia, or a therapy-related leukemia, cure hematopoietic stem cell transplantation (HSCT) (Box 27.2). remains elusive, and therapy must be directed more toward attain- Research in leukemia-specic therapy using molecular targets ing remissions or providing supportive care. and immunotherapy and cellular therapy is ongoing and rap- idly advancing. New schemes for risk stratication and treat- Chemotherapy ment modalities have resulted in prolonged survival and cures Chemotherapy is oral or parenteral cancer treatment with com- for conditions that were uniformly fatal with conventional pounds that possess antitumor properties. Methods of action of CHAPTER 27 Introduction to Hematologic Neoplasms 495 chemotherapy drugs vary considerably. Chemotherapy agents and Drug Administration (FDA) for supportive care of patients can be classied in two ways: by their eects on the cell cycle with cancer, including patients with hematologic neoplasms. and by their biochemical mechanism of action. Some che- Growth factors and colony-stimulating factors (CSFs), a class of motherapy drugs can aect cells only in specic phases of the cytokines, normally act in the bone marrow microenvironment cell cycle (phase specic), whereas other drugs act during any to stimulate blood cell formation (Chapter 5). Erythropoietin phase of the cell cycle (phase nonspecic). A general biochem- promotes red blood cell formation, and recombinant forms are ical classication of chemotherapy agents includes alkylating administered to cancer patients with anemia induced by che- agents, plant alkaloids, antimetabolites, antitumor antibiotics, motherapy. Similarly, granulocyte colony-stimulating factor and glucocorticoids. Chemotherapy drugs are usually given in (G-CSF) and granulocyte-macrophage colony-stimulating fac- combination according to various published protocols or in the tor (GM-CSF) are used to rapidly expand the number of mature context of a clinical trial. neutrophils to ght or prevent infection. Recombinant forms of Chemotherapy is usually administered in three phases: G-CSF and GM-CSF are administered to cancer patients with induction, consolidation, and maintenance. e goal of induc- chemotherapy-induced neutropenia. ese agents not only tion is to rapidly decrease the tumor burden and achieve remis- have improved patients’ quality of life but also have allowed sion. ere are dierent types of remission depending on the more ecient and eective delivery of chemotherapy regimens leukemia and the sensitivity of methods used for detection of by preventing delays of or dosage reductions in chemotherapy malignant cells. For example, a hematologic remission in acute courses as a result of low blood cell counts. leukemia may include a normocellular bone marrow, recovery of peripheral blood cell counts, and no microscopic evidence of Targeted Therapy leukemia cells, whereas a cytogenetic remission is the absence As more has been learned about specic genetic lesions that of the cytogenetic defect determined by karyotyping methods cause hematologic neoplasms, researchers have worked to (Chapter 30). A molecular remission is the absence of leukemia develop targeted therapies that act specically on malignant cell nucleic acid sequences using highly sensitive molecular cells and leave normal cells untouched. As a result of these methods capable of detecting one leukemia cell among 105 or advances, cancer therapy is realizing the dream of targeted ther- 106 normal cells (Chapter 29). Aer hematologic remission is apeutics and is moving away from nonspecic therapies such achieved, millions of leukemia cells may still be undetected in as chemotherapy and radiation. In 2001 the FDA cleared ima- the body (called minimal residual disease), so a consolidation tinib mesylate for treatment of chronic-phase CML as the rst phase is done with dierent chemotherapy agents to further rationally designed molecular targeted therapy for a cancer.26 reduce the number of leukemia cells. A maintenance phase may e t(9;22) translocation in CML results in production of the be incorporated in the treatment plan, which is continued for a BCR::ABL1 gene fusion, creating a fusion protein with consti- longer period with less intensive agents to eradicate any remain- tutive and unregulated tyrosine kinase activity. Imatinib is an ing leukemia cells to prevent a relapse. orally administered, small tyrosine kinase inhibitor (TKI) mol- Chemotherapy agents aect both malignant and normal ecule that binds to the ABL1 domain of the fusion protein and cells. eir eects are most pronounced on rapidly dividing selectively blocks its tyrosine kinase activity.26,27 It reduces the cells, such as those in gastrointestinal mucosa and bone mar- massive cell proliferation and induces apoptosis of CML cells row. is limits dosage and usually determines the maximum and remission with very few side eects.27 Imatinib is now the tolerated dose for a patient. Neutrophil and platelet counts are rst-line treatment for chronic-phase CML and has resulted in routinely monitored because they are the rst cells to decrease long-term remissions of 10 years and longer (Chapter 32).26,27 aer chemotherapy because of their shorter life spans. Newer and more potent TKIs have also been developed (such as dasatinib and nilotinib) to treat imatinib resistance that devel- Radiation Therapy ops in some patients with CML; these second-generation drugs Radiation kills cells by producing unstable ions that damage have also been cleared by the FDA for rst-line treatment in DNA and may cause instant or delayed death of cells. Toxic CML.26–28 Imatinib is also used in combination with chemo- eects of radiotherapy can occur during therapy or much later. therapeutic agents to treat BCR::ABL1 positive acute lympho- Complications can be reduced through use of combined ante- blastic leukemia.28 Other TKIs are in development, and their rior and posterior treatment ports and application of maximal eectiveness and safety are being evaluated in other hemato- shielding techniques to prevent damage to normal tissues. e logic neoplasms with dysregulated kinases, such as JAK2 inhib- hematopoietic system, gastrointestinal tract, and skin are most itors in polycythemia vera and FLT3 inhibitors in acute myeloid oen aected during radiotherapy. Spinal and pelvic irradia- leukemia.29,30 tion can cause marrow suppression, sometimes lowering blood In APL, the t(15;17) translocation, results in fusion of the ret- counts to life-threatening levels. Toxic eects are usually revers- inoic acid receptor gene, RARA, with the PML gene. RARA pro- ible when radiation is stopped. tein is a ligand-dependent transcription factor that when bound to retinoic acid, upregulates expression of target genes required Supportive Therapy for myeloid dierentiation.31 However, RARA in the PML::RARA Numerous substances that are naturally produced in the human fusion does not respond to retinoic acid, resulting in silencing of body have now been developed using recombinant technologies. transcription of genes needed for dierentiation and a block in Some are commercially produced and cleared by the US Food maturation beyond the promyelocytic stage.31 Now the prognosis 496 PART V Leukocyte Disorders of patients with APL is dramatically improved with the availability of intense research, and their use will continue to improve of dierentiation therapy using all-trans retinoic acid (ATRA) as patient outcomes and survival as they more specically target rst-line treatment along with chemotherapeutic agents (Chapter malignant cells and not patients’ normal cells. 31). ATRA binds to RARA in the fusion protein and overcomes the block, allowing transcription and expression of genes needed Hematopoietic Stem Cell Transplantation for myeloid dierentiation and maturation.31 As more has been learned about hematopoietic stem cells In lymphoid neoplasms, treatment includes immunotherapy (HSCs), the therapeutic method of bone marrow transplanta- using monoclonal antibodies for targeted therapeutic strategies. tion has evolved to be more aptly termed hematopoietic stem cell Monoclonal antibodies generally must be delivered intrave- transplantation (HSCT) because dierent sources in addition nously or subcutaneously. An example is rituximab (anti-CD20), to bone marrow can be used to obtain HSCs. Along with bone which binds to the CD20 antigen present on malignant cells marrow, peripheral blood and umbilical cord blood (UCB) are in many B-lymphoid neoplasms and has shown ecacy in rich sources. Bone marrow is harvested through multiple nee- non-Hodgkin lymphoma and chronic lymphocytic leukemia.32 dle aspirations, typically from the posterior iliac crests, and it is e antibody targets CD20+ lymphocytes for destruction by done in a sterile surgical environment, usually under general or antibody-mediated and complement-mediated cytotoxicity.32 regional anesthesia.38 Collection of peripheral blood stem cells Additional modications of immunotherapy are now being is less invasive in that they are harvested by pheresis aer mobi- developed, such as the bispecic T cell engager (BiTE) monoclo- lization out of bone marrow by cytokines and chemokines such nal antibody, blinatumomab.33 It can bind both CD19 and CD3, as G-CSF and plerixafor.38 UCB is collected by inserting a sterile bringing CD3+ T-lymphoid cells closer to CD19+ B-lymphoid needle into the umbilical vein aer the infant is delivered and cells to promote their destruction by cytokines.33 It has shown the cord is clamped and cut, either before or aer delivery of the ecacy in relapsed or refractory non-Hodgkin lymphoma placenta.38 Regardless of the source, HSCs are considered adult and precursor acute lymphoblastic leukemia.33 Monoclonal stem cells, even when they come from umbilical cord blood, as antibodies can also be conjugated with toxins (called immu- opposed to embryonic stem cells, which are the subject of con- noconjugates) to kill target cells. An example is inotuzumab siderable ethical debate. HSCT remains an expensive and di- ozogamicin, an anti-CD22 monoclonal antibody conjugated cult treatment alternative. with calicheamicin, an antibiotic with cytotoxic activity.34 e When the decision to perform HSCT has been made and conjugated antibody binds to CD22+ B-lymphoid cells, bring- a donor has been found, an extensive hospital stay is usually ing the toxin close to the cell where it rapidly enters, binds to, required. Pretransplantation conditioning regimens use high- and damages DNA and induces apoptosis.34 It has been used dose therapy to kill the patient’s malignant cells and normal with encouraging results in relapsed or refractory acute lym- bone marrow cells. is regimen reduces the body’s immunity phoblastic leukemia.34 to dangerously low levels and necessitates special protective Cellular therapy is also being implemented, with high e- isolation. Granulocyte counts approaching zero are commonly cacy in patients with high-risk hematologic neoplasms. One seen immediately before and aer transplantation. Aer infu- such therapy uses CD19-specic chimeric antigen recep- sion of donor HSCs, the recipient remains in a severely immu- tor T (CAR-T) cells, in which patient T cells are collected by nosuppressed condition for 2 weeks or longer. Strict isolation pheresis and are genetically engineered exvivo using lentiviral at this point is crucial. Prophylactic antibiotics and intrave- or retroviral vectors to express protein complexes that recog- nous nutrition are also essential to keep the patient alive until nize only the patient’s leukemia cells.35 Engineered CAR-T cells marrow engrament. Recovery of granulocytes, reticulocytes, then specically bind to patient’s leukemia cells and target them and platelets to normal levels is monitored closely in periph- for destruction.35 CAR-T cells have been successful in achiev- eral blood. Evaluation and management of red blood cell and ing remission in patients with high-risk, refractory, or relapsed platelet transfusions are crucial components of HSCT. Aer acute lymphoblastic leukemia and non-Hodgkin lymphoma.35 discharge, peripheral blood cell counts and bone marrow con- Gene editing technologies, such as CRISPR/Cas9 targeted tinue to be monitored to measure the progress of engrament nuclease, are also being used to improve the potency and safety of donor HSCs. of CAR-T cells.35 HSCs for treatment of malignant disease come from donors Epigenetic therapies are also used in hematologic neo- of three general types: (1) syngeneic, or from an identical twin; plasms to reverse epigenetic silencing of gene transcription. (2) allogeneic, usually from an HLA-identical sibling or HLA- For example, histone deacetylase inhibitors are being used in matched unrelated donor; or (3) autologous, in which the cutaneous T-cell lymphoma to prevent deacetylation, allowing patient’s own marrow or peripheral blood stem cells are used chromatin to open so that repressed genes can be transcribed (Figure 27.2). Most HSC donors are allogeneic, but for optimal and expressed.22,36 Likewise, hypomethylating treatment, using outcomes, it is important to match as many HLA antigens as drugs that remove methyl groups from gene promoters allow- possible. Within any given family, there can be only four HLA ing expression of genes, is used in high-risk myelodysplastic haplotypes (two maternal and two paternal), and there is one neoplasms.22,37 chance in four that a sibling will be HLA identical. If an HLA- ese and other targeted therapies will be discussed in more identical sibling or fully HLA-matched unrelated donor is not detail in chapters covering specic hematologic neoplasms. available, HLA-partially matched, related, or unrelated donors Development and optimization of targeted therapies is an area have been used. CHAPTER 27 Introduction to Hematologic Neoplasms 497 ALLOGENEIC AUTOLOGOUS Donor (related Recipient or unrelated) Treatment (patient) 1 Peripheral blood 1 Peripheral blood stem cells stem cells collected collected or bone or bone marrow marrow harvested harvested Blood/marrow Blood/marrow processing and 2 processing; 2 storage; may may include include purging T cell depletion of malignant cells Blood/ 3 4 marrow 3 4 Blood/marrow infusion infusion Conditioning Conditioning regimen regimen Recipient Recipient Recipient Recipient (patient) (patient) Figure 27.2 Overview of Hematopoietic Stem Cell Transplantation in Allogeneic and Autologous Donors. Even with continued improvement in technique and sup- such as infections or bleeding from bone marrow suppression, portive care, HSCT carries many risks. Death aer transplan- gra-versus-host disease, regrowth of malignant cells, and fail- tation is caused by complications of the conditioning regimens, ure of donor HSCs to engra. SUMMARY   Most neoplasms of the hematopoietic system are acquired   For most hematologic neoplasms, causes directly related genetic diseases. ey include leukemias, lymphomas, and to development of the malignancy are unknown, but a few myeloid proliferations and neoplasms. exceptions exist. Some known causes include environmental   Leukemias originate in bone marrow, and leukemia cells toxins, certain viruses, previous chemotherapy, and familial readily pass into peripheral blood, but they can also inltrate predisposition. lymphoid tissues (spleen, liver, lymph nodes) as well as other   Classiĕcation schemes for hematologic neoplasms include organs and tissues of the body. Lymphomas are solid tumors the older French-American-British (FAB) system, based pri- of lymphoid cells that usually originate in the lymphatic marily on morphology and cytochemical staining, and the system and proliferate in lymph nodes and other lymphoid World Health Organization (WHO) system, which retains organs and tissues. some elements of the FAB scheme but emphasizes molecular   Leukemias are divided into lymphoid, myeloid, and and cytogenetic abnormalities. mixed-phenotype lineages, and further into acute (precur-   Types of mutations found in hematologic neoplasms include sor cell) and chronic (mature cell) categories. In acute leuke- chromosomal rearrangements (such as translocation or mias, onset is sudden, progression is rapid, and the outcome inversion), gains or losses of chromosomes (aneuploidy), is oen fatal in weeks or months if le untreated. In chronic total or partial gene deletions, point mutations, insertions, leukemias, onset is insidious, and progression is slower, with and gene duplication/amplication. a longer survival compared with acute leukemia. 498 PART V Leukocyte Disorders   Hematologic neoplasms illustrate that a single mutation, or   Most hematologic neoplasms are systemic (diČusely involv more commonly a series of mutations, can lead to malignant ing the peripheral blood and/or bone marrow) at initiation transformation by disrupting the molecular machinery of of the malignant process. erefore with rare exceptions, hematopoietic cells. most treatments for hematologic neoplasms given with cura-   Most chromosomal translocations in leukemias involve tive intent are not localized, such as radiation or surgery, but oncogenes. Activation of the dominant transforming onco- must by nature be systemic treatments. gene alters its gene product or its expression and transforms   Current treatments for hematologic neoplasms can be gen the cell into a malignant phenotype, even in the presence of erally divided into the following categories: chemotherapy, a residual normal allele. radiation therapy, supportive therapy, targeted therapy, and   In contrast to oncogenes, tumor suppressor genes contribute hematopoietic stem cell transplantation. to the malignant process only if both alleles have been lost or otherwise inactivated. Now that you have completed this chapter, go back and read again   Activation of oncogenes or the loss or inactivation of tumor the case study at the beginning and respond to the questions pre- suppressor or DNA repair genes has adverse molecular sented. Answers can be found in Appendix C. eects on proliferation, survival, dierentiation, and matu- ration of hematopoietic cells. REVIEW QUESTIONS Answers can be found in Appendix C. 6. Which one of the following is an example of a tumor sup- pressor gene? 1. Lymphomas dier from leukemias in that they are: a. ABL1 a. Solid tumors b. RARA b. Not considered systemic diseases c. TP53 c. Never found in peripheral blood d. JAK2 d. Do not originate from hematopoietic cells 7. G-CSF is provided as supportive treatment during leuke- 2. Which one of the following viruses is known to CAUSE mia treatment regimens to: lymphoid neoplasms in humans? a. Suppress GVHD a. HIV-1 b. Overcome anorexia b. HTLV-1 c. Prevent anemia c. Hepatitis B d. Reduce the risk of infection d. Parvovirus B 8. Imatinib is an example of what type of leukemia treatment? 3. Loss of function of tumor suppressor genes increase the a. Supportive care risk of hematologic neoplasms by: b. Chemotherapy a. Suppressing cell division c. Bone marrow conditioning agent b. Activating tyrosine kinases, which promote proliferation d. Targeted therapy c. Promoting excessive apoptosis of hematopoietic cells 9. Which one of the following statements is FALSE about epi- d. Allowing cells with damaged DNA to progress through genetic mechanisms? the cell cycle a. Epigenetic mechanisms control how genes are expressed 4. Oncogenes are said to act in a dominant fashion because: and silenced. a. Leukemia is a dominating disease that is systemic b. MicroRNAs can bind to specic mRNAs and block their b. e oncogene product is a gain-of-function mutation translation. c. A mutation in only one allele is sucient to promote a c. Hypermethylation of CpG islands in gene promoters malignant phenotype results in their overactivation. d. ey are inherited by autosomal dominant transmission d. Histone deacetylases keep chromatin of target genes in a 5. Which one of the following is NOT one of the cellular closed inactive state. abnormalities produced by oncogenes? 10. Which one of the following is NOT a source of hematopoi- a. Constitutive activation of a growth factor receptor etic stem cells for transplantation? b. Constitutive activation of a signaling protein a. Spleen c. Acceleration of DNA catabolism b. Bone marrow d. Dysregulation of apoptosis c. Peripheral blood d. Umbilical cord blood CHAPTER 27 Introduction to Hematologic Neoplasms 499 REFERENCES 19. Daley, G. Q., Van Etten, R. A., & Baltimore, D. (1990). Induction of chronic myelogenous leukemia in mice by the P210 bcr/abl 1. Nowell, P. C., & Hungerford, D. A. (1960). A minute chromosome gene of the Philadelphia chromosome. Science, 247, 824–830. in human granulocytic leukemia (Abstract). Science, 132, 1497. 20. Ko, T. K., Javed, A., Lee, K. L., etal. (2020). An integrative model 2. Rowley, J. D. (1973). A new consistent chromosomal abnormality of pathway convergence in genetically heterogeneous blast crisis in chronic myelogenous leukemia identied by quinacrine chronic myeloid leukemia. Blood, 135, 2337–2353. uorescence and Giemsa staining. Nature, 243, 290–293. 21. Lodish, H., Birk, A., Kaiser, C. A., etal. (2021). Cancer. In 3. Taub, R., Kirsch, I., Morton, C., etal. (1982). Translocation of the Molecular Cell Biology. (9th ed., pp.). New York: W.H. Freeman c-myc gene into the immunoglobulin heavy chain locus in human MacMillan. Burkitt lymphoma and murine plasmacytoma cells. Proc Natl 22. Yang, H., & Green, M. R. (2020). Harnessing lymphoma Acad Sci USA, 79, 7837–7841. epigenetics to improve therapies. Blood, 136, 2386–2391. 4. National Cancer Institute, Surveillance, Epidemiology, and End 23. Weiss, C. N., & Ito, K. (2017). A macro view of microRNAs: the Results Program (SEER). Cancer Stat Facts: Leukemia. https://seer. discovery of microRNAs and their role in hematopoiesis and cancer.gov/statfacts/html/leuks.html. Accessed December 28, 2022. hematologic disease. Int Rev Cell Mol Biol, 334, 99–175. 5. National Cancer Institute, Surveillance, Epidemiology, and 24. Knudson, A. G. (1985). Hereditary cancer, oncogenes, and End Results Program (SEER). Cancer Stat Facts: Non-Hodgkin antioncogenes. Cancer Res, 45, 1437–1443. Lymphoma. https://seer.cancer.gov/statfacts/html/nhl.html. 25. Zenz, T., Eichhorst, B., Busch, R., etal. (2010). TP53 mutation Accessed December 28, 2022. and survival in chronic lymphocytic leukemia. J Clin Oncol, 28, 6. National Cancer Institute, Surveillance, Epidemiology, and End 4473–4479. Results Program (SEER). Cancer Stat Facts: Hodgkin Lymphoma. 26. Sampaio, M. M., Santos, M. L. C., Marques, H. S., etal. (2021). https://seer.cancer.gov/statfacts/html/hodg.html. Accessed Chronic myeloid leukemia-from the Philadelphia chromosome December 28, 2022. to specic target drugs: a literature review. World J Clin Oncol, 12, 7. WHO Classication of Tumours Editorial Board. 69–94. Haematolymphoid tumours. Lyon (France): International Agency 27. Hochhaus, A., Larson, R. A., Guilhot, F., etal. (2017). Long-term for Research on Cancer; forthcoming. (WHO classication of outcomes of imatinib treatment for chronic myeloid leukemia. N tumours series, 5th ed.; vol. 11). https://publications.iarc.fr. Eng J Med, 376, 917–927. 8. Gaglia, M. M., & Munger, K. (2018). More than just oncogenes: 28. Haddad, F. G., & Short, N. J. (2022). Evidence-based minireview: mechanisms of tumorigenesis by human viruses. Curr Opin Virol, what is the optimal tyrosine kinase inhibitor for adults with 32, 48–59. newly diagnosed Philadelphia chromosome-positive acute 9. Fiore, D., Cappelli, L. V., Broccoli, A., etal. (2020). Peripheral T cell lymphoblastic leukemia? Hematology Am Soc Hematol Educ lymphomas: from the bench to the clinic. Nat Rev Cancer, 20, 323–342. Program, 2022, 213–217. 10. Yin, H., Qu, J., Peng, Q., et al. (2019). Molecular mechanisms 29. Springuel, L., Renauld, J. C., & Knoops, L. (2015). JAK kinase of EBV-driven cell cycle progression and oncogenesis. Med targeting in hematologic malignancies: a sinuous pathway from Microbiol Immunol, 208, 573–583. identication of genetic alterations towards clinical indications. 11. Baptista, M. J., Tapia, G., Muñoz-Marmol, A. M., etal. (2022). Hematologica, 100, 1240–1253. Genetic and phenotypic characterisation of HIV-associated 30. Kennedy, V. E., & Smith, C. C. (2020). FLT3 mutations in acute aggressive B-cell non-Hodgkin lymphomas, which do not myeloid leukemia: key concepts and emerging controversies. occur specically in this population: diagnostic and prognostic Front Oncol, 10, 612880. implications. Histopathology, 81, 826–840. 31. Liquori, A., Ibanez, M., Sargas, C., etal. (2020). Acute 12. Sweet-Cordero, E. A., & Biegel, J. A. (2019). e genomic promyelocytic leukemia: a constellation of molecular events landscape of pediatric cancers: implications for diagnosis and around a single PML::RARA fusion gene. Cancers, 12, 624. treatment. Science, 363, 1170–1175. 32. Feugier, P. (2015). A review of rituximab, the rst anti-CD20 13. Zhang, C., Liu, J., Xu, D., etal. (2020). Gain-of-function mutant p53 monoclonal antibody used in the treatment of B non-Hodgkin’s in cancer progression and therapy. J Mol Cell Biol, 12, 674–687. lymphomas. Future Oncol, 11, 1327–1342. 14. Huang, R. X., & Zhou, P. K. (2020). DNA damage response 33. Goebeler, M. E., & Bargou, R. (2016). Blinatumomab: a CD19/ signaling pathways and targets for radiotherapy sensitization in CD3 bispecic T cell engager (BiTE) with unique anti-tumor cancer. Signal Transduct Target er, 5, 1–27. ecacy. Leuk Lymphoma, 57, 1021–1032. 15. Labuhn, M., Perkins, K., Matzk, S., etal. (2019). Mechanisms 34. Kantarjian, H. M., DeAngelo, D. J., Stelljes, M., etal. (2016). of progression of myeloid preleukemia to transformed myeloid Inotuzumab ozogamicin versus standard care for acute leukemia in children with Down syndrome. Cancer Cell, 36, lymphoblastic leukemia. N Engl J Med, 375(8), 740–753. 123–138. 35. Perales, M. A., Kebriaei, P., Kean, L. S., etal. (2018). Building a 16. Campo, E., Jae, E. S., Cook, J. R., etal. (2022). e international safer and faster CAR: seatbelts, airbags, and CRISPR. Biol Blood consensus classication of mature lymphoid neoplasms: a report Marrow Transplant, 24, 27–31. from the clinical advisory committee. Blood, 140, 1229–1253. 36. Duvic, M. (2015). Histone deacetylase inhibitors for cutaneous 17. Arber, D. A., Orazi, A., Hasserjian, R. P., etal. (2022). T-cell lymphoma. Dermatol Clin, 33, 757–764. International consensus classication of myeloid neoplasms and 37. Voso, M. T., Lo-Coco, F., & Fianchi, L. (2015). Epigenetic therapy acute leukemias: integrating morphologic, clinical, and genomic of myelodysplastic syndromes and acute myeloid leukemia. Curr data. Blood, 140, 1200–1228. Opin Oncol, 27, 532–539. 18. Stam, K., Heisterkamp, N., & Grosveld, G. (1985). Evidence 38. Zubair, A. C., & Rowley, S. D. (2023). Practical aspects of hematologic of a new chimeric bcr/c-abl mRNA in patients with chronic stem cell harvesting and mobilization. In Homan, R., Benz, E. myelocytic leukemia and the Philadelphia chromosome. N Engl J J., Silberstein, L. E., etal. (Eds.), Hematology: Basic Principles and Med, 313, 1429–1433. Practice. (8th ed., pp. 1653–1666). Philadelphia: Elsevier.

Use Quizgecko on...
Browser
Browser