Hematologic Manifestations of Solid Tumors


Hematologic abnormalities are commonly seen among patients with malignancy. These derangements range from the incidental to the life-threatening and may complicate management or prompt the initiation of additional or alternative therapies. Hematologic abnormalities can be seen as the initial manifestation of cancer, providing a crucial diagnostic clue. In addition, the hematologic aspects of cancer, and the therapies that we use to treat these irregularities, can provide insight into the biology of tumorigenesis. This chapter reviews some of the impact that malignancy can have on erythrocytes, leukocytes, platelets, and hemostasis.

Erythrocytes

Anemia

The most common hematologic manifestation of cancer is anemia; anywhere from 30% to 90% of patients with cancer have documented anemia before the initiation of any treatment. Anemia may be the first presentation of malignancy; for instance, anemia is frequently noted in patients with colon cancer and more frequent in advanced stage disease. The pathogenesis of cancer-related anemia is multifactorial, and can arise from direct cancer tissue invasion with resultant blood loss, tumor involvement of the bone marrow, or hemolysis; it can be a direct effect of chemotherapy or radiation therapy, be associated with chronic renal dysfunction secondary to the malignancy or treatment, among other causes. These causes can be generally divided according to the pathogenesis of the anemia: that resulting from direct blood loss, anemia caused by erythrocyte destruction, and anemia caused by a hypoproliferative marrow state.

A number of cancer-specific mechanisms can be responsible for a hypoproliferative state, including nutritional deficiencies, decreased erythropoietin (EPO) production because of acute or chronic kidney injury, and injury to the bone marrow itself. The most common nutritional abnormality in cancer-related anemia is iron deficiency; roughly 40% of cancer patients may have iron-deficiency, which can be secondary to poor oral intake, as well as from blood loss, commonly seen in gastrointestinal and gynecologic malignancies. Compounding any deficiency in total body iron stores are the myelosuppressive effects on erythropoiesis and iron homeostasis that may result from cancer-associated inflammation and cytokine activation. Many cancers are associated with a systemic inflammatory state, manifested by elevated levels of inflammatory cytokines such as IL-2 and IL-6, which can directly inhibit erythropoiesis and also suppress the amount of iron available for erythropoiesis.

Various systemic mediators of anemia are disrupted in malignancy. When serum EPO concentrations are measured among cancer patients, these levels are inappropriately low in comparison to patients without malignancy but who have the same degree of anemia caused by iron deficiency. Cancer patients also often demonstrate increased levels of hepcidin, a protein critical to iron homeostasis, which acts by decreasing the binding to and breaking down of the iron transporter ferroportin. This subsequently results in increased storage of iron in the form of ferritin, and at the same time decreases free iron that would be used in erythropoiesis. In contrast to iron deficiency, vitamin B 12 deficiency is less frequently observed in patients with anemia and cancer, seen in only 5% to 10% of patients.

Many chemotherapeutic agents have hematologic sequelae; patients undergoing treatment may experience varying degrees of anemia resulting from chemotherapy-induced bone marrow suppression. The European Cancer Anaemia Survey prospectively studied over 15,000 patients with a variety of solid tumors undergoing treatment. Before therapy, 10% of patients had hemoglobin levels below 10 g/dL, and during therapy this increased to almost 40% of patients and was correlated with a worsening performance status. In the same study, radiation therapy alone had less impact on the incidence of anemia, but the combination of chemotherapy and radiation led to a greater degree of anemia than chemotherapy alone. Anemia caused by chemotherapy is related to the cumulative dose, combination of chemotherapeutic agents, and dose schedule. For example, in the treatment of lung cancer with cisplatin and paclitaxel, anemia worsens with an increasing total dose of cisplatin, or if paclitaxel is given over a 24-hour period rather than shorter courses.

Anemia in cancer patients can also occur in the setting of red cell destruction, such as is seen in autoimmune hemolytic anemia, or in the setting of microangiopathic hemolytic anemia (MAHA), which is associated with concurrent thrombocytopenia. Although the true incidence of MAHA among cancer patients is difficult to determine, it is a well-defined entity that has been described in a small (perhaps <10%) subset of cancers, particularly mucin-producing disseminated adenocarcinomas. It is important to distinguish secondary thrombotic microangiopathies (TMAs), which can occur in patients with malignancy, from thrombotic thrombocytopenic purpura (TTP), given the marked differences in underlying pathophysiology. TTP is the result of congenital or acquired absence or inhibition of the von Willebrand cleaving enzyme called “A Disintegrin And Metalloproteinase with Thrombospondin Motifs-13” (ADAMTS13). In contrast, secondary TMAs may have a slight decrease in ADAMTS13 activity, but do not have the severe deficiency of ADAMTS13 activity, typically characterized by a level of less than 10%, that is seen in TTP. Accordingly, patients with secondary TMAs typically have a poor response to therapeutic plasma exchange. Cancer-associated TMAs may arise from direct endothelial damage rather than diminished ADAMTS13 activity. Cancer patients may also develop a MAHA related to the specific medications used during treatment, including cyclosporine, mitomycin, and gemcitabine, and agents inhibiting vascular endothelial growth factor (anti-VEGF) signaling. (See box on Management of Cancer-Related Anemia .)

Management of Cancer-Related Anemia

Hemoglobin levels typically decrease early in the course of chemotherapy treatment; more than half of patients experience a greater than 1 g/dL drop over the course of the first 9 weeks of therapy. The treatment of anemia related to malignancy depends upon correct identification of the underlying etiology. As noted previously, iron-deficiency anemia is common in patients with malignancy. Among those patients with cancer who have an absolute iron deficiency (transferrin saturation <20%, ferritin <30 ng/mL), there is evidence that they may benefit from a short course of either oral or low-dose intravenous iron. In this setting, the addition of erythropoiesis-stimulating agents (ESAs) is not necessary.

For many patients, transfusion of blood products is an effective therapeutic intervention. Red cell transfusion provides rapid symptomatic relief and is also a source of iron; one unit of packed red blood cells (RBCs) contains roughly 200 mg of iron. Logistic limitations with RBC transfusion, and transfusion-related morbidities, particularly with a high cumulative transfusion burden, have spurred the incorporation of ESAs as alternative or adjunct agents for treating anemia in cancer patients. The use of ESAs during myelosuppressive treatment may increase the hemoglobin level and decrease transfusion requirements by up to 50%. However, ESA use is associated with an increased rate of cardiovascular and thrombotic events. This relationship between ESA use and thrombosis may be related to the target hemoglobin concentration during therapy, since higher hemoglobin targets are associated with increased rates of thrombotic events in cancer patients.

In addition to thrombotic events, ESA use has been associated with poorer overall survival or time to cancer progression in some settings. Data regarding ESAs and progression of disease are conflicting; some studies in patients with breast cancer and patients with head and neck cancer suggested worsening progression-free survival or worsened local control of disease with ESA use. A number of mechanisms have been proposed, including decreased chemosensitivity in the setting of ESA use, or alterations in tumor vascularity and oxygen supply. Aberrant expression of the EPO receptor has been identified on the cell surface of certain tumor cells, while other putative mechanisms include augmentation of red cell mass and effects on tumor oxygenation. Nonetheless, larger studies on the impact of ESA on progression suggest any effect may be small overall.

Related in part to the above concerns, ESA use is generally restricted to specific limited indications in patients with cancer; notably, such practice recommendations are generally limited to solid tumors with chemotherapy-induced anemia. In specific hematologic malignancies, such as myelodysplastic syndromes (MDS), ESA therapies remain a backbone of the treatment of anemia. For the treatment of anemia in patients with solid tumors, transfusion of blood products and, if indicated, iron therapy, remain the standard of care. There are other scenarios where ESAs may be useful, particularly among patients with moderate or severe chronic kidney disease, or in palliative settings, as well as for specific patient scenarios such as the treatment of patients who identify as Jehovah’s Witnesses. In such situations, reversible causes of anemia should be ruled out before ESA use, and the minimal amount of ESA be used to avoid RBC transfusion.

Erythrocytosis

Outside of patients with myeloproliferative neoplasms, and specifically polycythemia vera, erythrocytosis is an uncommon manifestation of cancer. Polycythemia vera, and other myeloproliferative neoplasms, are typified by acquisition of somatic mutations that upregulate the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, most commonly via the JAK2 V617F mutation, and phenotypically present as an expansion of myeloid-origin cells, with an elevated hemoglobin and suppressed EPO levels. In these cancers, erythrocytosis is caused by primary expansion of the malignant clone. For such patients, various therapeutic interventions may be considered, including therapeutic phlebotomy or hydroxyurea (see Chapter 70 , The Polycythemias). In solid tumors, erythrocytosis is an uncommon paraneoplastic phenomenon. Paraneoplastic erythropoiesis has been described in several tumors, including renal cell carcinoma. In some instances, there may be association with a common predisposing condition; for instance, patients with von Hippel-Lindau disease are at risk for renal and central nervous system tumors, and the mechanism of erythrocytosis is typically through paraneoplastic EPO expression. Nonetheless, elevated RBC counts are rare in patients with malignancy, particularly those receiving chemotherapy, and usually do not require intervention.

Management of Cancer-Related Thrombocytopenia

Treatment of thrombocytopenia in cancer patients is generally supportive. For patients with active bleeding, platelet transfusion and correction of other coagulopathies are the mainstays of therapy. The use of agents such as tranexamic acid in these patients may also be considered. In the absence of active bleeding, a threshold of <10 × 10 9 /L for prophylactic platelet transfusion is generally recommended. This is based on a study that randomized 600 patients with hematologic malignancies to prophylaxis or no prophylactic transfusions. Both were treated for bleeding events. Rates of World Health Organization grade 2, 3, or 4 bleeding were high in both groups, but higher in the no-prophylaxis group (50%), compared with the prophylaxis group (43%). Prophylactic platelet transfusion has been used in patients with solid tumors, but without as much evidence-based support.

Other therapies, with or without platelet transfusion, may also have a role in chemotherapy-related thrombocytopenia. Thrombopoietin (TPO) receptor agonists are an area of increasing interest in the management of thrombocytopenia during chemotherapy, particularly as a means to maintain treatment schedules, which may be delayed by thrombocytopenia. For instance, a randomized phase II trial compared placebo to eltrombopag in patients receiving gemcitabine-based chemotherapy and found that fewer patients receiving eltrombopag required dose delays and/or reductions in chemotherapy compared with those receiving placebo. Nonetheless, specific interactions with other chemotherapeutic agents will likely be important to understand; for instance, in a trial of patients with MDS randomized to receive azacitidine alone or in combination with eltrombopag, the patients receiving eltrombopag actually had worsened platelet transfusion requirements.

Platelets

Thrombocytopenia

Chemotherapy and immunosuppressive agents are the most common causes of thrombocytopenia in the cancer patient. For instance, patients receiving cisplatin/gemcitabine for bladder cancer or carboplatin/gemcitabine for lung cancer have rates of clinically significant thrombocytopenia of 30% to 60%. Among patients with hematologic malignancy, thrombocytopenia is even more common, particularly with induction or conditioning chemotherapy regimens. The typical mechanism by which chemotherapy causes thrombocytopenia is through marrow suppression; although, some agents are also associated with immune-mediated thrombocytopenia, and thrombocytopenia may also occur as a sequelae of other processes such as MAHA or TMA (above). Immune thrombocytopenia is typically characterized by a sudden and isolated drop in platelet count shortly after drug administration. Several chemotherapeutics, including gemtuzumab, trastuzumab, and oxaliplatin, have been associated with immune-mediated thrombocytopenia, potentially related to the presence of drug-dependent antibodies.

Thrombocytopenia may also be a result of bone marrow infiltration by tumor cells, thrombotic microangiopathy, consumptive coagulopathy, or as an autoimmune manifestation of the malignancy itself. Bone marrow involvement, often occult, is more common in prostate, lung, and breast cancer; such patients typically have multiple cell lines involved, and may display a leukoerythroblastic appearance on the peripheral smear. Associated laboratory features of disseminated intravascular coagulation (DIC), including an elevated D-dimer and fibrinogen degradation products, can be seen in cancer patients, and are more common with advanced disease. (See box on Management of Cancer-Related Thrombocytopenia .)

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here