Multiple myeloma and cardiac amyloidosis


Unlike solid tumors, myeloma staging is a composite score based on the four tests: blood albumin, beta-2 microglobulin, lactate dehydrogenate (LDH), and bone marrow Fluorescent in situ hybridization (FISH). Stage I is albumin 3.5 g/dL or greater, beta-2 microglobulin is less than 3.5 mg/L, normal LDH, and no high-risk FISH. Stage III is beta-2 microglobulin 5.5 mg/L or greater and either high LDH or high-risk FISH. Stage II is neither stage I nor III. T two amyloid staging systems are commonly used. Both are from the Mayo Clinic. The first is purely based on troponin and NT-proBNP (above), and the second is the Mayo (2012) system, which also includes the immunoglobulin free light chain, a measure of tumor burden. The Mayo (2004) system uses the following cut-offs: Stage I, troponin T less than 0.035 mcg/L and NT-proBNP less than 332 ng/L; stage II, either troponin T 0.035 μg/L or greater or NT-proBNP greater than 332 ng/L; stage IIIa, both troponin T 0.035 μg/L or greater, and NT-proBNP greater than 332 ng/L, but NT-proBNP less than 8500 ng/L; stage IIIa, both troponin T 0.035 μg/L or greater and NT-proBNP greater than 8500 ng/L. See Table 51.2 for conversion factors when troponin I, high-sensitivity troponin T or BNP are the only available assays. A hard cut-off for ASCT is a troponin T greater than 0.06 μg/L ASCT, autologous stem cell transplant after high-dose melphalan; B, bor, or V, bortezomib (Velcade); C or Cy, cyclophosphamide; CHF, congestive heart failure; D or d, dexamethasone; Dara, daratumumab (Darzylex); HFpEF, heart failure with preserved ejection fraction; K, carfilzomib (Kyprolis); M, melphalan; P, prednisone; R, lenalidomide (Revlimid); R-ISS, revised international staging system; T, thalidomide.

KEY POINTS

  • In the United States the 2018 annual estimate for new cases of multiple myeloma (MM) is 30,770 and of deaths is 12,770.

  • The incidence of immunoglobulin light-chain (AL) amyloidosis is 12 cases per million per year. It is imperative that AL amyloidosis be distinguished from both the inherited and the acquired form of transthyretin (ATTR) amyloidosis.

  • Neither disease is thought to be curable, although young patients with Revised International Staging System (R-ISS) stage I myeloma are expected to have a median overall survival of 8 to 10 years. More than 20% of patients with AL amyloidosis are alive at 10 years.

  • For fit patients with AL amyloidosis, autologous stem cell transplant (ASCT) is considered to an important part of therapy, although there are differing interpretations of the data in terms of the relative importance of ASCT for this disease.

  • Treatment of amyloidosis is directed to the cause of fibril production.

  • Cardiovascular aspects with therapy pertain to the use of immune modulatory drugs in MM, which require consideration for thrombo prophylaxis, and proteasome inhibitor in both MM and AL amyloidosis, which can lead to heart failure presentations (esp., carfilzomib).

Introduction

Multiple myeloma (MM) and immunoglobulin light chain amyloidosis are plasma cell disorders. Both are characterized by increased clonal plasma cells in the bone marrow and detectable monoclonal proteins in the serum and/or urine. After lymphoma, myeloma is the second most common hematologic malignancy, comprising 1.8% of all malignancies. Immunoglobulin light chain (AL) amyloidosis is about one-fifth as common as MM, and approximately 10% to 15% of patients with myeloma will have AL amyloidosis. Both are diseases of the elderly, with median ages at diagnosis of approximately 70 years. , Men are 1.6 times more likely to develop MM, and African-Americans are twice as likely to develop MM than whites.

Diagnosis and differential diagnosis

As shown in the Central Illustration, clinical manifestations are dissimilar for the two diseases. In myeloma, the defining symptoms and signs are anemia, bone pain (or asymptomatic lesions), hypercalcemia, renal dysfunction, and high levels of serum monoclonal proteins (most commonly found on serum protein electrophoresis), and bone marrow plasma cells. When present, fatigue is a function of anemia, renal failure, or hypercalcemia. Occasionally, significant abnormalities are incidentally found even before patients are symptomatic. One must distinguish MM from its truly asymptomatic form (smoldering myeloma) and from its precancerous form (monoclonal gammopathy of undetermined significance).

The diagnosis of AL amyloidosis is often more challenging than that of MM. Fatigue and edema are the most common presenting symptoms. Fatigue often relates to heart failure with preserved ejection fraction (HFpEF) and edema to either cardiomyopathy and/or nephrotic syndrome. The cardiomyopathy of AL amyloidosis is frequently overlooked in part owing to the preserved ejection fraction, and there are significant delays before a diagnosis is made. Patients may have hepatomegaly with a cholestatic picture, a small fiber peripheral neuropathy, autonomic dysfunction, signs of microvascular involvement (periorbital purpura, jaw claudication), early satiety, or changes in their bowel patterns. The diagnosis is made by a tissue biopsy with (sub-) typing of the amyloid to prove that it is indeed AL amyloidosis rather than another form of inherited or acquired amyloidosis ( Table 51.1 ). Elevated serum immunoglobulin free light chains and/or the presence of a monoclonal protein by immunofixation are supportive of a diagnosis of AL amyloidosis, but not definitive. The differential diagnosis for AL amyloidosis includes monoclonal gammopathy of undetermined significance, MM, and other types of amyloidosis, especially wild type transthyretin (ATTR wt ) amyloidosis. The diagnosis of acquired form of transthyretin (ATTR) amyloidosis can be made with a positive 99m-technetium pyrophosphate (PYP) or 99m-technetium (99mTc) and 3,3-diphosphono-1,2-propanodicarboxylic acid (DPD) scan in the ABSENCE of a monoclonal protein by immunofixation or by serum immunoglobulin free light chain assay ( Fig. 51.1 ). Cardiac imaging with echocardiography and magnetic resonance imaging can help support a diagnosis of AL amyloidosis, but neither is diagnostic nor can either distinguish AL from ATTR. This holds true also for echo strain imaging, although apical sparring on the bull’s eye view of global longitudinal strain (“cherry on top” appearance) should prompt further investigation into amyloidosis (sensitivity 96%, specificity 88% in patients without coronary artery disease). Increased left ventricular wall thickness, septal hypertrophy with granular sparking myocardium, and pericardial and pleural effusions are other echocardiographic features of amyloidosis. Lack of R-wave progression in the precordial leads (pseudoinfarction pattern) and low voltage are the classic electrocardiographic features, but have low sensitivity. A QTc greater than 440 ms and a Sokolow-Lyon index equal to or less than 1.5 mV have a sensitivity of 85% and a specificity of 100% for cardiac amyloidosis, mainly the AL subtype. The diagnosis of AL always requires a Congo red positive tissue biopsy that has been typed to be composed of immunoglobulins.

TABLE 51.1
Nomenclature of Most Common Forms of Amyloidosis Affecting the Heart
AMYLOID TYPE PRECURSOR PROTEIN CLINICAL
AL or AH Immunoglobulin (light or heavy chain) Immunoglobulin amyloidosis (formerly known as primary)Systemic or localized amyloidosis
ATTR wt Transthyretin Age-related (formerly known as senile) amyloidosis; heart and ligaments
ATTR v Transthyretin, mutants Familial amyloidosis; most often heart and/or nerve
AApoAI Apolipoprotein AI, mutant Kidney, liver, rarely heart, larynx, skin
AApoAII Apolipoprotein AII, mutant Kidney, heart
AApoAIV Apolipoprotein AIV, no mutation recognized Heart, kidney
AA Serum amyloid A (SAA) Secondary or familial Mediterranean fever; familial periodic fever syndromes associated with mutated tumor necrosis factor receptor; most often kidney; rarely heart
AA, Secondary amyloid; AH, immunoglobulin heavy chain amyloid; AL, immunoglobulin light chain; AApo, apolipoprotein amyloid; ATTR, acquired form of transthyretin.

FIG. 51.1, Approach to a patient with suspected cardiac amyloidosis. ECV, Extracellular volume; FLC, free light chain ratio; GLS, global longitudinal strain; HF, heart failure; HC, immunohistochemistry; LGE, late gadolinium enhancement; LVH, left ventricular hypertrophy; SCT, stem cell transplantation; TTR, transthyretin.

Prognosis

For patients with MM, staging is not directly related to tumor burden, but rather the genetics of the tumor (i.e., plasma cells), which is most commonly determined by f luorescent in situ hybridization (FISH). Certain cytogenetics, such as deletion 17p, translocation t(4;14), and t(14;20), are considered high-risk features. These bone marrow plasma cell abnormalities, along with serum albumin, serum beta-2 microglobulin, and lactate dehydrogenase (LDH) are the components of the Revised International Staging System (R-ISS), and patients are placed into three groups with discrete outcomes ( Table 51.2 ). The relative risk of death between each of the stages is approximately 3, yielding approximate 5-year overall survival rates of 80%, 60%, and 40%, respectively.

TABLE 51.2
Prognostic Staging Systems in Multiple Myeloma
DURIE-SALMON INTERNATIONAL STAGING SYSTEM REVISED INTERNATIONAL STAGING SYSTEM
Stage I
  • Low cell mass: < 0.6 × 10 12 cells/m 2 PLUS all of the following:

  • Hgb > 10 g/dL

  • Serum IgG <5 g/dL

  • Serum IgA < 3 g/dL

  • Normal serum calcium

  • Urine monoclonal protein excretion < 4 g/day

  • No generalized lytic bone lesions

62 months β2M < 3.5 mg/L and serum albumin ≥3.5 g/dL 62 months β2M < 3.5 mg/L and serum albumin ≥ 3.5 g/dL
No high-risk cytogenetics (FISH)
Normal LDH
82 months
Stage II Neither stage I nor stage III 44 months Neither stage I nor stage III 44 months Neither stage I nor stage III 62 months
Stage III
  • High cell mass: > 1.2 × 10 12 cells/m 2 PLUS 6 1 of the following:

  • Hgb < 8.5 g/dL

  • Serum IgG >7 g/dL

  • Serum IgA > 5 g/dL

  • Serum calcium > 12 mg/dL (3 μmol/L)

  • Urine monoclonal protein excretion > 12 g/day

  • Advanced lytic bone lesions

29 months β2M ≥ 5.5 mg/L 29 months
  • β2M ≥ 5.5 mg/L

  • PLUS ≥ 1 of the following:

  • High-risk cytogenetics (FISH)T(4;14)

  • T(14;16)

  • Del(17p)

  • LDH > ULN

40 months
Subclass A: Serum creatinine < 2 mg/dL
B: Serum creatinine ≥ 2 mg/dL
FISH, f luorescent in situ h ybridization; LDH, lactate dehydrogenase; ULN, upper limit of normal.

The greatest risk of death for patients with AL amyloidosis is during the first 6 to 12 months after diagnosis. It is during this period that approximately 30% of patients die. After the first year, the slope of the survival curves, especially for those with stage III disease, becomes less steep. Overall, for patients with AL amyloidosis, 6-year survival rates are 85%, 45%, 30%, and less than 10% for patients with stage I, II, IIIa and IIIb, respectively. Among those patients who achieve a deep hematologic response (a very good partial response or better), more than 80% of them are alive at 6 years. Staging for patients with AL amyloidosis is done using serum troponin, N-terminal pro-brain natriuretic peptide (NT-proBNP), and immunoglobulin free light chain ( Tables 51.3 and 51.4 ).

TABLE 51.3
Prognostic Biomarker-Based Staging Systems in Amyloidosis
AL AMYLOIDOSIS MODEL MEDIAN OVERALL SURVIVAL
Revised Mayo Model a cTnT
≥ 0.025 ug/L
Stage I = all negative 94.1 months
NT-pro BNP ≥ 1800 ng/L Stage II = 1 positive 40.3 months
Difference between involved and uninvolved light chain ≥ 180 mg/L Stage III = 2 positive 14.0 months
Stage IV = all positive 5.8 months
ATTR Amyloidosis Median Overall Survival
National Amyloidosis Center b
NT-pro BNP > 3000 ng/L Stage I = all negative 62.9 months
eGFR < 45 mL/min/1.73 m 2 Stage II = 1 positive 46.7 months
Stage III = all positive 24.1 months
ATTR, Acquired form of transthyretin; NT -proBNP, N-terminal pro-brain natriuretic peptide.

a Kumar S, Dispenzieri A, Lacy MQ, et al. Revised prognostic staging system for light chain amyloidosis incorporating cardiac biomarkers and serum free light chain measurements. J Clin Oncol. 2012;30:989–995.

b Gillmore JD, Damy T, Fontana M, et al. A new staging system for cardiac transthyretin amyloidosis. Eur Heart J. 2018;39(30):2799–2806.

TABLE 51.4
AL Amyloidosis Staging Conversions
Modified from Muchtar E, Kumar SK, Gertz MA, et al. Staging systems use for risk stratification of systemic amyloidosis in the era of high-sensitivity troponin T assay. Blood. 2019;133:763–766.
Model cTnT, µg/L cTnI, µg/L Hs-cTnT, ng/L NT-proBNP, ng/L BNP, ng/L
Mayo 2004 model a ≥ 0.035 ≥ 0.1 ≥ 50 e ≥ 332 81 g
Euro 2015 modification of Mayo 2004 model b ≥ 0.035 ≥ 0.1 f ≥ 50 ng/L e , f
  • ≥ 332

  • ≥ 8500

Mayo 2012 model c ≥ 0.025 ND >40 f ≥ 1800 ≥ 400 h
Mayo ASCT troponin risk marker d ≥ 0.06 ND ≥75 f NA NA

ASCT, Autologous stem cell transplantation; NA, not applicable; ND, no data.

a Original three-stage model using cTnT and NT-proBNP cut-points as listed. cTnI also tested in same paper.

b Original three-stage model using cTnT and NT-proBNP cut-points as listed, but separate stage III into IIIa and IIIb based on whether or not NT-proBNP is higher than 8500 ng/L.

c A four-stage model using cTnT and NT-proBNP cut-points as listed along with difference of involved free light chain ≥ 18 mg/dL.

d Simple binary troponin T threshold predicting for transplant-related mortality 25% vs. 4%.

e In separate study, Hs-cTnT 54 found to be comparable to cTnT cut-point of 0.035, but reanalysis using quartic formula yielded 51 ng/L.

f Extrapolated numbers are based on quartic formula applied to a dataset of 224 newly diagnosed AL amyloidosis cases.

g Separate study demonstrated similarity between original troponin T cut-offs and troponin I and BNP cut-offs.

h In Mayo 2012 study, this BNP threshold was found to be comparable to NT-proBNP.

Treatment and side effects

Many of the drugs to treat these two conditions are the same as are the side effects but typically more severe in AL amyloidosis. Over the course of 15 years, nine drugs have been approved for MM, several of which are shown in Central Illustration. Other drugs used to treat myeloma not shown in the Central Illustration include pomalidomide (another immune modulatory drug [IMiD]), ixazomib (another proteasome inhibitor), daratumumab (an antibody directed at CD38), elotuzumab (an antibody directed at SLAMF7), and panobinostat (a histone deacetylase inhibitor). Additional promising drugs for the treatment of myeloma are in clinical trials (discussed below). Although there are no US Food and Drug Administration (FDA) approved drugs for the treatment of AL amyloidosis, many myeloma drugs are employed to treat this otherwise progressive and lethal disease. Such pharmacologic advances have resulted in approximately a doubling of overall survival for both MM and AL amyloidosis in the past two decades. , ,

Among patients with MM, treatment strategies are aligned with age (and functional status) and cytogenetic risk more so than tumor burden or R-ISS stage. The general MM treatment plan is induction (typically the first 4 months of therapy), followed by consolidation (ASCT in those eligible or continuation of induction for non-ASCT candidates), and maintenance (most often lenalidomide, but in certain cases bortezomib, ixazomib, or carfilzomib).

The general strategy to treat AL amyloidosis is based on the extent of cardiac involvement (dysfunction) followed in part by tumor burden as measured by serum free light chains. The distinction between amyloid with and without myeloma is in part semantics because so much of the prognosis is dependent on cardiac involvement. Early death relates predominantly to the severity of cardiac impairment (Central Illustration). That said, clonal burden has an impact on both presentation and long-term outcomes. A patient with AL amyloidosis who has fewer than 10% bone marrow plasma cells at diagnosis is (1) less likely to have cardiac involvement, (2) more likely to have a very deep response to chemotherapy, that is a complete response; and (3) very likely to expect a median overall survival of greater than 10 to 15 years assuming they survive the first year after diagnosis. In contrast, those patients with AL amyloidosis with more than 10% to 20% bone marrow plasma cells, whether or not they have classic myeloma bone lesions or classic myeloma kidney, are (1) more likely to have cardiac involvement, (2) less likely to have a deep response to chemotherapy, and (3) in general have shorter long- term survival, even if they survive past the first year. ,

Toxicity of therapy

Each of the different chemotherapeutic agents used to kill bone marrow plasma cells have their own cardiac toxicities, but a few additional general principles need to be considered as well. Virtually all chemotherapy is myelosuppressive, which increases a patient’s risk of sepsis syndrome and all the cytokine-mediated toxicities that accompany it. Practically every regimen incorporates corticosteroids into the treatment plan—most often as a potent killer of plasma cells, but also sometimes as a knee-jerk antiemetic. The fluid retention that accompanies high-doses of weekly—and even sometimes daily—corticosteroids can aggravate hypertension, diastolic dysfunction, and systolic dysfunction, resulting in congestive heart failure (CHF). In addition, chronic steroid use can result in reduced adrenal reserve or frank adrenal insufficiency, resulting in cardiovascular collapse when stressed. The use of beta-blockers in patients with myeloma has been associated with a better overall survival in a retrospective study. In contrast, beta-blockers and calcium channel blockers (anything that slows the heart rate) can precipitate worsening functional status and even CHF among patients with AL amyloidosis.

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