Introduction to Leukocyte Neoplasms

Published on 04/03/2015 by admin

Filed under Hematology, Oncology and Palliative Medicine

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 3435 times

Introduction to Leukocyte Neoplasms

Peter D. Emanuel

Etiology of Leukocyte Neoplasms

For most leukocyte neoplasms, causes directly related to the development of the malignancy are unknown. There are, however, a few exceptions. Environmental toxins can induce genetic changes, as discussed later in this chapter, leading to a malignancy phenotype. Environmental exposures known to lead to hematopoietic malignancies include radiation exposure, as experienced by survivors of atomic explosions, and exposure to organic solvents, such as benzene. There are two types of lymphoid malignancies in which viruses may play a pathogenetic role. The Epstein-Barr virus has been implicated as an etiologic factor in the development of Burkitt non-Hodgkin lymphoma. Similarly, human T-cell lymphotropic virus type 1 (HTLV-1) is the likely cause of adult T-cell leukemia/lymphoma. As discussed further in this chapter, there are some known familial cancer predisposition syndromes. In addition, as more cancer survivors live longer, it is clear that some alkylating agents and other forms of chemotherapy used to treat various forms of cancer can induce deoxyribonucleic acid (DNA) damage in hematopoietic cells, leading to hematologic malignancies.

Classification Schemes for Leukocyte Neoplasms

The French-American-British (FAB) classification of the acute leukemias was devised in the 1970s and 1980s. The FAB schemas were based largely on morphologic characteristics and relied heavily on examination of routine histologic stain preparations to distinguish lymphoid neoplasms from myeloid neoplasms (Figure 29-1). Although these types of diagnostic criteria have not been abandoned, pathologists are now moving toward more precise classification of many of the leukocyte neoplasms based on recurring chromosomal and genetic lesions found in many patients. These lesions are related to disruptions of oncogenes, tumor suppressor genes, and other regulatory elements that control proliferation, maturation, apoptosis, and other vital cell functions. In 2001 the World Health Organization (WHO) published new classification schemes for nearly all of the tumors of hematopoietic and lymphoid tissues.1 In some cases WHO melded the older morphologic schemes with the newer schemes. For instance, in the WHO classification scheme for acute myeloid leukemias (AMLs) there are some remnants of the old FAB classification, but new classifications were introduced for leukemias associated with consistently recurring chromosomal translocations. This 2001 WHO classification of hematologic malignancies has undergone a recent revision.2

Oncogenes

Oncogenes originally were identified as genes that carried rapidly transforming retroviruses derived from normal cellular homologues, proto-oncogenes. The oncogene definition has evolved to specify genes that cause dominant-acting cancer mutations, regardless of whether they are derived from a retrovirus. The typical proto-oncogene codes for a protein involved in normal cell cycle regulation. Regulation proteins provide the signal transduction that carries messages about cell division and maturation (differentiation) from outside the cell to the nucleus. Mutations of these genes may form oncogenes that disrupt normal cell cycle processes.

Most chromosomal translocations in leukemias involve oncogenes. The “dominant” transforming oncogene is able to alter the gene product and transform the cell into a malignant phenotype, even in the presence of a residual normal allele. Even the afore-mentioned two examples, CML and Burkitt lymphoma, t(9;22) and t(8;14), involve oncogenes that are activated when brought into proximity with their new partners on fusion genes. In the case of CML, the ABL proto-oncogene on chromosome 9 is activated when fused to the BCR component of chromosome 22. In the case of Burkitt lymphoma, the MYC proto-oncogene on chromosome 8 is fused with the immunoglobulin heavy chain locus on chromosome 14. Although oncogenic transformation was first identified by karyotypic analyses, molecular biologic techniques have evolved rapidly over the last three decades, so that more genetic translocations are being identified that create novel fusion genes invisible at the cytogenetic level.

Tumor Suppressor Genes

Tumor suppressor genes are so named because they code for proteins that resist malignant transformation. These genes do not act in a dominant fashion as in the case of oncogenes; rather, cells are transformed into a malignant phenotype only after both alleles of these genes have been lost or otherwise inactivated, the so-called two-hit mechanism proposed by Knudson.5 Although tumor suppressor genes are harder to isolate and identify, numerous such genes have now been identified, and many have been found to be associated with autosomal dominant familial cancer predisposition syndromes. Some well-known examples include the RB1 tumor suppressor gene involved in familial retinoblastoma, the TP53 gene in Li-Fraumeni syndrome, the WT1 gene in Wilms tumor, and the NF1 gene in familial neurofibromatosis type 1. Perhaps more importantly, many of these tumor suppressor genes and their protein products are altered in many sporadic cancers, including hematologic cancers.

Molecular Pathways Perturbed by Cellular Transformation

Regardless of the type of chromosomal or genetic abnormality, clinicians understand the molecular consequences that the formation of oncogenes or loss of tumor suppressor genes has on the proliferation and differentiation of hematologic tissues. Specialists recognize several mechanisms, including blocked differentiation, transcriptional repression, disruptions of cell signaling, progression, and apoptosis. The t(15;17) translocation found in acute promyelocytic leukemia (APL), which fuses the PML gene to the RARA (retinoic acid receptor alpha) gene, clearly results in a state of arrested differentiation, because RARA-induced differentiation is inhibited. Treating patients with APL with pharmacologically high doses of all-trans retinoic acid can overcome this block and permit APL cells to differentiate into normal neutrophils. In so doing, the APL cells lose their leukemic potential.

Other chromosomal abnormalities involve transcriptional repression of DNA and condensation abnormalities of chromatin, such as those involved in the core-binding factor leukemic subtypes of AML. A similar example on the lymphoid side of hematopoietic neoplasms are the chromosomal translocations involving the BCL6 gene. Normal BCL6 encodes for a transcriptional repressor responsible for recruiting the histone deacetylase complex, which regulates germinal center formation in lymph nodes. The mutation of BCL6 leads to overexpression of this normal protein so that DNA is excessively repressed, which in this case prevents lymphocytes from progressing beyond the germinal center stage of development.

Since the initial identification of the BCR/ABL fusion gene in CML, there are now many other examples of genetic abnormalities in myeloid malignancies in which the abnormality leads to disruption of cell signaling, often by way of activation of kinase cascades. FLT3 codes for a tyrosine kinase receptor preferentially expressed on hematopoietic stem cells that mediates proliferation and differentiation. A unique mutation resulting in an internal tandem duplication leads to constitutive activation of this pathway (i.e., always turned on) in many forms of AML and other hematopoietic malignancies. Other examples of gene mutations that alter kinase cascades in myeloid or lymphoid cells are c-KIT, NOTCH, JAK2, and RAS.

Many cyclin-dependent kinases are altered in lymphoid malignancies. The cyclin-dependent kinases tightly regulate cell cycle progression through the synthesis, proteolysis, and phosphorylation of cyclins.

Finally, another important molecular signaling pathway in cells involves programmed cell death, or apoptosis. This vital process allows organisms to eliminate redundant, damaged, aged, or infected cells. In the hematopoietic environment, apoptosis is essential to contain and control the massive expansion that the hematopoietic system is capable of generating at times of stress, infection, or hemorrhage. Caspases are a family of proteases that participate in the apoptotic cascade triggered in response to proapoptotic signals. The culmination of this apoptotic cascade is cellular disassembly. The BCL family contains many genes, some of which are proapoptotic and some of which are antiapoptotic. Many of the various forms of non-Hodgkin lymphoma seem to involve disruptions of BCL2, BCL6, BCL10, or other members of the caspase and BCL family of genes comprising the apoptotic cascade.

The list of chromosomal and molecular aberrations known to occur in the various leukocyte neoplasms continues to grow on an almost daily basis. Indexing this list is far beyond the scope of this chapter, but some condensed lists are provided in Chapter 31. More complete indices can be found in other publications such as the WHO reclassification scheme,1 its 2008 revision, or other hematology textbooks.610

Therapy for Leukocyte Neoplasms

The various forms of therapy available today for leukocyte neoplasms can be roughly divided into the following categories: chemotherapy, radiation therapy, supportive therapy, targeted therapy, and stem cell transplantation. In contrast to many solid tumors, numerous hematologic malignancies now have cure rates that are substantially higher than they were two or three decades ago. Many new and exciting therapies that are less toxic are now under development or are already employed in patient settings. These therapies are bringing more optimism to the care of patients with leukocyte neoplasms than ever before. Selection of the best therapy must start, however, with an accurate diagnosis. Even the most effective therapies do not work if they are applied in the wrong circumstances.

Curative treatment strategies are a realistic goal for patients with Hodgkin lymphoma, CML, hairy cell leukemia, and some forms of non-Hodgkin lymphoma, and for children with acute lymphoblastic leukemia. Cure may be attainable in other patients with acute lymphoblastic or myeloid leukemia, and long-term remissions may be achievable in adults with multiple myeloma. For patients with other leukocyte neoplasms such as mantle cell lymphoma, chronic lymphocytic leukemia, or a therapy-related leukemia, cure remains elusive, and therapy must be directed more toward attaining remissions or providing supportive care.

Chemotherapy

Chemotherapy is oral or parenteral cancer treatment with compounds that possess antitumor properties. The methods of action of the chemotherapy drugs vary considerably. Chemotherapy agents can be classified in two ways: by their effects on the cell cycle and by their biochemical mechanism of action. Some chemotherapy drugs can affect cells only in specific phases of the cell cycle (phase specific), whereas other drugs act without regard to the cell cycle (phase nonspecific) and affect cells during any phase of the cell cycle. Agents in this latter category usually have a linear dose-response curve (i.e., the higher the dose, the more cells are killed). There are two subgroups:

Phase-specific agents are effective only if present during a certain phase in the cell cycle (see Chapters 31 and 32). Within a certain dosage range, agents in this category show no increase in killing of cells with a further increase in dosage. Examples are l-asparagine amidohydrolase (G1 phase), antimetabolites such as methotrexate (S phase), and vinca alkaloids (M phase).

Chemotherapeutic agents affect both neoplastic and normal cells. The effect is most pronounced on rapidly dividing cells, such as those of the mucosa of the gastrointestinal tract and the bone marrow. This limits the dosage and usually determines the maximum tolerated dose for a patient. Chemotherapy agents are categorized in Table 29-1.

TABLE 29-1

Chemotherapy Agents

Agent Other Names Uses Toxic Effects
Alkylating Agents
Busulfan Myleran CML, pretransplantation Myelosuppression, infertility
Cyclophosphamide Cytoxan, Neosar Lymphoma, MM, ALL, pretransplantation Marrow suppression, N&V, cystitis
Nitrogen mustard Mechlorethamine Hodgkin lymphoma, NHL Myelosuppression, N&V, infertility
Chlorambucil Leukeran CLL, Waldenström macroglobulinemia, NHL, Hodgkin disease Myelosuppression, hair loss
Melphalan Alkeran MM Myelosuppression
Carmustine BCNU Hodgkin disease, NHL, MM Myelosuppression
Dacarbazine DTIC Hodgkin disease Myelosuppression, N&V
Plant Alkaloids
Vincristine Oncovin ALL, NHL, Hodgkin disease, CLL, MM Neurotoxicity, hair loss
Vinblastine Velban ALL, NHL, Hodgkin disease, CLL, MM Myelosuppression
Etoposide VP-16 NHL, pretransplantation Myelosuppression, hair loss
Antitumor Antibiotics
Daunorubicin Daunomycin AML, ALL Myelosuppression, cardiotoxicity, N&V, hair loss
Doxorubicin Adriamycin ALL, AML, Hodgkin disease, NHL, CLL, MM Myelosuppression, cardiotoxicity, N&V, hair loss
Bleomycin Bleo Hodgkin lymphoma, NHL Lung toxicity, gastrointestinal toxicity
Idarubicin Idamycin AML Myelosuppression
Antimetabolites
Methotrexate Amethopterin, MTX ALL, NHL Myelosuppression, gastrointestinal toxicity
Ara-C Cytosine arabinoside, cytarabine AML, NHL, Hodgkin disease Myelosuppression, gastrointestinal toxicity, hair loss
Mercaptopurine 6-MP ALL, CML Hepatotoxicity, myelosuppression
Thioguanine 6-TG AML Myelosuppression
Pentostatin 2′-Deoxycoformycin Hairy cell leukemia, CLL, lymphomas Neurotoxicity, myelosuppression
Fludarabine CLL, lymphomas, Waldenström macroglobulinemia Neurotoxicity, myelosuppression
2-CDA CDA, 2-chlorodeoxyadenosine, 2-cladribine Hairy cell leukemia, CLL, lymphomas, AML, Waldenström macroglobulinemia Neurotoxicity, myelosuppression
Glucocorticoids
Prednisone ALL, CLL Fluid retention, muscle weakness
Methylprednisolone Hodgkin disease, NHL, AMM Fluid retention, muscle weakness
Hydrocortisone Hodgkin disease, NHL, AMM Fluid retention, muscle weakness
Dexamethasone Decadron MM Fluid retention, muscle weakness
Others
Hydroxyurea Hydrea CML, PV, AMM Leukopenia, N&V
Asparaginase l-Asparagine amidohydrolase ALL, refractory NHL Nephrotoxicity, N&V
Cisplatin Cis-platinum Solid tumors, Hodgkin disease, NHL Nephrotoxicity, ototoxicity
Procarbazine Hodgkin disease, NHL Myelosuppression

image

ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; AMM, agnogenic myeloid metaplasia; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; N&V, nausea and vomiting; PV, polycythemia vera.

Radiation Therapy

The effectiveness of x-rays against Hodgkin and non-Hodgkin lymphomas was described shortly after radiation’s discovery. Radiation kills cells by producing unstable ions that damage DNA and may cause instant or delayed death of the cell. The toxic effects of radiotherapy can occur during therapy or much later. Complications can be reduced through the use of combined anterior and posterior treatment ports and the application of maximal shielding techniques to prevent damage to normal tissues. The hematopoietic system, the gastrointestinal tract, and the skin are most often affected during radiotherapy. The toxic effects are usually reversible when radiation is stopped. The epithelium of the entire gastrointestinal tract is a rapidly dividing cellular system that is very sensitive to radiation. There may be drying up of saliva and loss of taste. If the stomach is irradiated, anorexia, nausea, and vomiting may occur. Intestinal irradiation may result in malabsorption and diarrhea. Irradiated skin becomes erythematous and tender. Permanent loss of body hair and hyperpigmentation also may occur in irradiated areas. Spinal and pelvic irradiation can cause marrow suppression, sometimes lowering blood counts to the life-threatening range.

Supportive Therapy

Numerous substances that are naturally produced in the human body have now been created in the laboratory using cloning techniques. Several are commercially produced and have been cleared by the Food and Drug Administration (FDA) for general use in the supportive care of cancer patients, particularly patients with hematologic malignancies. Colony-stimulating factors (CSFs), a class of cytokines, normally act in the bone marrow microenvironment to stimulate blood cell formation. Erythropoietin is the main stimulatory CSF responsible for red blood cell formation. Normal erythropoietin and a long-acting formulation of this molecule are available to aid in the care of cancer patients with anemia induced by chemotherapy. Similarly, granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) are used to expand rapidly the number of mature neutrophils capable of fighting infection within the body. Recombinant forms of G-CSF and GM-CSF and a long-acting form of G-CSF are now FDA cleared and are commonly employed to support cancer patients with chemotherapy-induced neutropenia. CSFs not only have enabled physicians to improve patients’ quality of life, but also have allowed more efficient and effective delivery of chemotherapy regimens by preventing delays of or dosage reductions in chemotherapy courses owing to low blood counts. On the other hand, use of erythropoietin support has recently become restricted because of reports of possible higher tumor relapse rates and shortened survival.

Targeted Therapy

As more has been learned about the specific genetic lesions that cause cancers, researchers have worked to develop targeted therapies that act specifically on the tumor cell and leave normal cells untouched.11 As a result of these advances in translational research, cancer therapy is realizing the dream of targeted therapeutics and is moving away from nonspecific therapies such as chemotherapy and radiation. The Philadelphia chromosomal abnormality, t(9;22) associated with CML was first reported in 1960. The Philadelphia chromosome results from the juxtaposition of two genes, BCR and ABLl, and the resulting fusion protein has elevated tyrosine kinase activity. It took until almost the year 2000 to develop an agent that specifically targets the product of the Philadelphia chromosome. Imatinib mesylate (Gleevec) is a tyrosine kinase inhibitor that inhibits the kinase activity of all proteins that contain ABL. Imatinib is not specific solely for the BCR/ABLl tyrosine kinase; it also inhibits the tyrosine kinase activity of the c-KIT and the platelet-derived growth factor receptor. It is through its activity on these other receptors that imatinib has found usefulness against other tumors as a targeted therapeutic. In CML, however, imatinib has quickly become the standard treatment for patients newly diagnosed with the disease who are not candidates for immediate transplant (see Chapter 34). Resistance to imatinib has developed in some CML cells, and second-generation, more powerful kinase inhibitors, such as dasatinib and nilotinib, have now been cleared by the FDA. Small-molecule inhibitors such as imatinib are typically available in oral form and have relatively few side effects.

Rather than utilizing small-molecule inhibitors, treatment of lymphoid neoplasms has relied more on monoclonal antibodies for targeted therapeutic strategies. Rituximab specifically targets the CD20 antigen present on many non-Hodgkin lymphoma cells. When the antibody binds the lymphoma cell, complement is activated, and the cell lyses. Other possible scenarios leading to cell death with the use of monoclonal antibodies include antibody-mediated cellular cytotoxicity and stimulation of apoptosis.10 In contrast to small-molecule inhibitors, monoclonal antibodies generally must be delivered intravenously or subcutaneously. Monoclonal antibodies such as rituximab have evolved from the original monoclonal antibodies that were derived from mice. Rituximab represents a humanized form of an antibody designed or modified to be “more human” so that the patient’s immune system will not raise an antibody against the monoclonal antibody. Additional modifications are now being developed, such as radioactively labeled monoclonal antibodies that can carry a killing dose of radioactivity directly to the mutated cell.

Stem Cell Transplantation

As more has been learned about hematopoietic stem cells, the therapeutic method of bone marrow transplantation has evolved to be more aptly termed stem cell transplantation, because a variety of different sources in addition to bone marrow can be used to obtain hematopoietic stem cells. Along with bone marrow, peripheral blood and umbilical cord blood are rich sources. Regardless of the source, hematopoietic stem cells are considered adult stem cells, even when they come from umbilical cord blood, as opposed to embryonic stem cells, which are the subject of considerable ethical debate. Stem cell transplantation still remains an expensive and rigorous treatment alternative.

When the decision to transplant has been made and a donor has been found, an extensive hospital stay is usually required. The pretransplantation conditioning regimen uses high-dose therapy to kill the patient’s cancer cells and bone marrow cells. This regimen reduces the body’s immunity to dangerously low levels and necessitates special protective isolation. Granulocyte counts approaching 0 are commonly seen immediately before and after transplantation. After the infusion of donor stem cells, the recipient remains in a severely immunosuppressed condition for 2 weeks or longer. Strict isolation at this point is crucial. Prophylactic antibiotics and intravenous nutrition are also essential to keep the patient alive until the marrow engrafts. The return of granulocytes, reticulocytes, and platelets to normal levels is monitored closely in the peripheral blood. Hematology laboratory evaluation and management of red blood cell and platelet transfusions are crucial components of stem cell transplantation. After the patient’s release from the hospital, the blood counts, along with bone marrow aspirate and core biopsy specimens, continue to be monitored to measure the progress of engraftment of the donor stem cells.

Stem cell transplants for treatment of malignant disease have come from donors of three general types: (1) an identical twin donor (syngeneic transplant), (2) a donor genetically different from the recipient (allogeneic transplant), or (3) the patient’s own marrow or peripheral blood stem cells (autologous transplant) (Figure 29-2). Syngeneic transplants are most desirable because of the perfect match of cells. For obvious reasons, however, they are rare, and they are not discussed further in this chapter.

Allogeneic Transplantation

Most stem cell donors are genetically different from the recipient. The intent is to match as many of the human leukocyte antigens (HLAs) as possible. Within any given family, there can be only four HLA haplotypes (two from the mother and two from the father), and there is one chance in four that a sibling of the patient will be HLA identical. In addition to HLA-identical grafts, grafts from HLA-mismatched donors within families have been used.

A major complication of an allogeneic marrow graft is the immunologic reaction of donor T cells against the tissues of the recipient, which results in graft-versus-host disease (GVHD). Two forms of GVHD are recognized: acute and chronic. Acute GVHD develops in the immediate posttransplantation period or shortly thereafter. It is characterized by a skin rash, liver dysfunction, and diarrhea. Chronic GVHD, by definition, develops more than 100 days after transplantation. It is frequently generalized in the form of a multisystem autoimmune disease. Skin lesions, joint contractures, chronic hepatitis, malabsorption, and chronic obstructive pulmonary disease are frequent features of the chronic GVHD syndrome. Clinically significant GVHD is associated with a fatality risk 25 times higher than that in patients without GVHD.

T-cell depletion of donor bone marrow is the most effective means of preventing acute and chronic GVHD, but this benefit has been offset by the substantial increase in the risk of leukemic relapse and infections. There is considerable clinical evidence that allogeneic grafts lower the risk of leukemic relapse. This antileukemia effect is most pronounced in the presence of chronic GVHD.

Autologous Transplantation

In autologous stem cell transplantation, cells are harvested from the patient and, after conditioning, transplanted back. Stem cells harvested during remission, presumably contaminated with malignant cells, are purged in vitro through the use of antileukemic monoclonal antibodies or cytotoxic drugs. After conditioning of the patient with cyclophosphamide, total body irradiation, or other techniques to eradicate remaining malignant cells, the purged autologous stem cells are reinfused. Requirements for success in autologous transplantation are the presence of normal pluripotent stem cells and reduction in the number of malignant cells to a level insufficient to cause recurrence from reinfused marrow.

A comparison of autologous transplantation with matched allogeneic transplantation shows that (1) although allogeneic transplantation is not available to every patient, almost every patient is eligible for autologous transplantation; (2) among autologous transplant recipients, posttransplantation morbidity and mortality are lower and hospital stays are shorter; and (3) the relapse rate is higher among recipients of autologous transplants than among those receiving allogeneic transplants.

Even with the continued improvement in technique and supportive care, stem cell transplantation carries many risks. Death after transplantation is most likely caused by the following:

Summary

• Most malignancies of the hematopoietic system are acquired genetic diseases.

• Most leukocyte malignancies are not localized, but rather are systemic at the initiation of the malignant process.

• For most leukocyte neoplasms, causes directly related to the development of the malignancy are unknown, but a few exceptions exist. Some known causes include environmental toxins, certain viruses, previous chemotherapy, and familial predisposition.

• There are several classification schemes for leukocyte neoplasia, including the FAB system, based primarily on morphology and cytochemical staining, and the WHO system, which retains some elements of the FAB scheme but emphasizes molecular and cytogenetic changes.

• Chromosomal translocations in hematologic malignancies illustrate that a single mutation, or a series of mutations in stepwise fashion, can lead to malignant transformation by disrupting the molecular machinery of the cell.

• Most chromosomal translocations in leukemias involve oncogenes. The “dominant” transforming oncogene is able to alter the gene product and transform the cell into a malignant phenotype, even in the presence of a residual normal allele.

• In contrast to oncogenes, tumor suppressor genes contribute to the malignant process only if both alleles have been lost or otherwise inactivated.

• The formation of oncogenes or the loss of tumor suppressor genes has molecular effects on the proliferation and differentiation of hematologic tissues.

• Current treatments for leukocyte neoplasms can be roughly divided into the following categories: chemotherapy, radiation therapy, supportive therapy, targeted therapy, and stem cell transplantation.

Review Questions

1. Which of the following viruses is known to cause lymphoid malignancies in humans?

2. Tumor suppressor genes cause cancers such as leukemia when mutations result in:

3. Oncogenes are said to act in a dominant fashion because:

4. All of the following are among the cellular abnormalities produced by oncogenes except:

5. Chemotherapeutic agents are divided into which two major subgroups?

6. G-CSF is provided as supportive treatment during leukemia treatment regimens to:

7. Imatinib is an example of what type of leukemia treatment?

8. Monoclonal antibodies may kill cancer cells by all of the following mechanisms except:

9. Hematopoietic stem cells for transplantation are harvested from all of the following tissues except:

10. Compared with autologous bone marrow transplantation, allogeneic transplantation has: