Multiple Myeloma

Back

Practice Essentials

Multiple myeloma (MM) is a plasma cell malignancy in which monoclonal plasma cells proliferate in bone marrow, resulting in an overabundance of monoclonal paraprotein (M protein), destruction of bone, and displacement of other hematopoietic cell lines. First described in 1848, MM is part of a spectrum of diseases ranging from monoclonal gammopathy of unknown significance (MGUS) to plasma cell leukemia. See the image below.



View Image

Bone marrow aspirate demonstrating plasma cells of multiple myeloma. Note the blue cytoplasm, eccentric nucleus, and perinuclear pale zone (or halo). ....

Signs and symptoms

MM can range from asymptomatic to severely symptomatic with complications requiring emergent treatment. Presenting signs and symptoms of MM include the following:

See Presentation for more detail.

Diagnosis

MM is often discovered through routine blood screening when patients are being evaluated for unrelated problems. In one third of patients, the condition is diagnosed after a pathologic fracture occurs, usually involving the axial skeleton.

Examination for MM may reveal the following:

In patients with MM and amyloidosis, the characteristic examination findings include the following:

Testing

The International Myeloma Workshop guidelines for standard investigative workup in patients with suspected MM include the following[1] :

Routine laboratory tests include the following:

National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines also recommend the use of serum free light chain assay and plasma cell fluorescence in situ hybridization (FISH) on bone marrow: del 13, del 17p13, t(4;14), t(11;14), t(14;16), t(14;20), 1q21 amplification, 1p deletion as part of the initial diagnostic workup.[2]

Imaging studies

See Workup for more detail.

Management

There is currently no cure for MM. However, advances in therapy, such as autologous stem cell transplantation, radiation, and surgical care in certain cases, have helped to lessen the occurrence and severity of adverse effects of this disease and to manage associated complications.[3, 4, 5]

Chemotherapy and immunosuppression

Several drug therapies are valuable in the treatment of symptomatic MM. Clinicians treat many patients with high-dose therapy and peripheral blood or bone marrow stem cell transplantation.

Chemotherapy regimens used in patients with MM include the following:

For primary induction therapy in patients with MM who are candidates for transplantation, NCCN guidelines recommend the following combinations as preferred regimens[2] :

Other recommended regimens, according to the NCCN, are as follows:

The NCCN considers the following regimens useful in certain circumstances (although triplet regimens should be used as the standard, patients not considered candidates for a 3-drug regimen can be started on a 2-drug regimen, with the third drug added once performance status improves):

In September 2019 the US Food and Drug Administration approved the combination of bortezomib/thalidomide/dexamethasone with daratumumab for newly diagnosed young patients with MM who are eligible for transplantation.

For primary induction therapy in patients who are not transplant candidates, the NCCN guidelines list the following as preferred regimens[2] :

For maintenance therapy, the NCCN recommends lenalidomide (category 1); bortezomib may also be used.

For MM that relapses after more than 6 months, the regimen used for primary induction therapy can be repeated. For relapses that occur sooner,  regimens that are NCCN category 1 preferred include the following[2] :

Isatuximab–pomalidomide–low-dose dexamethasone is approved for relapsed and refractory MM in patients who have received at least 2 prior therapies including lenalidomide and a proteasome inhibitor.[6, 7]

See Treatment and Medication for more detail.

Pathophysiology

The development of MM is commonly preceded by monoclonal gammopathy of undetermined significance (MGUS), a premalignant condition that results when plasma cells undergo mutations that restore their capacity for proliferation. In MGUS, these clonal plasma cells take up less than 10% of bone marrow. The serum protein value is less than 3 g/dL and myeloma-related end-organ damage is absent. An intermediate disease stage between MGUS and MM, termed smoldering MM, is characterized by an M protein level of  3 g/dL or more and over 10% clonal plasma cells in bone marrow, but no symptoms of myeloma-related end-organ damage.[8]   .

A variety of cytogenetic abnormalities are found in MGUS and MM. Approximately half of cases are hyperdiploid, usually with extra copies of the odd-numbered chromosomes. Most of the remainder are nonhyperdiploid and are characterized by a primary translocation involving the Ig heavy-chain gene at 14q32.[8] In addition, virtually all cases involve dysregulation of the cyclin D/retinoblastoma (cyclin D/RB) pathway.  This genetic heterogeneity contributes to the rapid emergence of drug resistance in MM.[9]  

Increasing evidence suggests that the bone marrow microenvironment of tumor cells plays a pivotal role in the pathogenesis of myelomas.[10] This discovery has resulted in the expansion of treatment options.

The role of cytokines in the pathogenesis of MM is an important area of research. Interleukin (IL)-6 is also an important factor promoting the in vitro growth of myeloma cells. Other cytokines are tumor necrosis factor and IL-1b.

The pathophysiologic basis for the clinical sequelae of MM involves the skeletal, hematologic, renal, and nervous systems, as well as general processes (see below).

Skeletal processes

Plasma-cell proliferation causes extensive skeletal destruction with osteolytic lesions, anemia, and hypercalcemia. Mechanisms for hypercalcemia include bony involvement and, possibly, humoral mechanisms. Isolated plasmacytomas (which affect 2-10% of patients) lead to hypercalcemia through production of the osteoclast-activating factor.

Destruction of bone and its replacement by tumor may lead to pain, spinal cord compression, and pathologic fracture. The mechanism of spinal cord compression symptoms may be the development of an epidural mass with compression, a compression fracture of a vertebral body destroyed by multiple myeloma, or, rarely, an extradural mass. With pathologic fracture, bony involvement is typically lytic in nature.

Hematologic processes

Bone marrow infiltration by plasma cells results in neutropenia, anemia, and thrombocytopenia. M components may interact specifically with clotting factors, leading to defective aggregation.

Renal processes

The most common mechanisms of renal injury in MM are direct tubular injury, amyloidosis, or involvement by plasmacytoma.[11, 12] Renal conditions that may be observed include hypercalcemic nephropathy, hyperuricemia due to renal infiltration of plasma cells resulting in myeloma, light-chain nephropathy, amyloidosis, and glomerulosclerosis.

Neurologic processes

The nervous system may be involved as a result of radiculopathy and/or cord compression due to nerve compression and skeletal destruction (amyloid infiltration of nerves).

General processes

General pathophysiologic processes include hyperviscosity syndrome. This syndrome is infrequent in MM and occurs with overproduction of IgG1, IgG3, or IgA. Sludging in the capillaries can result in purpura, retinal hemorrhage, papilledema, coronary ischemia, or central nervous system (CNS) symptoms (eg, confusion, vertigo, seizure). Cryoglobulinemia causes Raynaud phenomenon, thrombosis, and gangrene in the extremities.

Etiology

The precise etiology of MM has not yet been established. Roles have been suggested for a variety of factors, including genetic causes, environmental or occupational causes, MGUS, radiation, chronic inflammation, and infection.

Genetic causes

MM has been reported in two or more first-degree relatives and in identical twins, although no evidence suggests a hereditary basis for the disease. A study by the Mayo clinic found MM in eight siblings from a group of 440 patients; these eight siblings had different heavy chains but the same light chains.

Some studies have shown that abnormalities of certain oncogenes, such as c-myc, are associated with development early in the course of plasma cell tumors and that abnormalities of oncogenes such as N-ras and K-ras are associated with development after bone marrow relapse. Abnormalities of tumor suppressor genes, such as TP53, have been shown to be associated with spread to other organs.[13]

Ongoing research is investigating whether human leukocyte antigen (HLA)-Cw5 or HLA-Cw2 may play a role in the pathogenesis of multiple myeloma.

Environmental or occupational causes

Case-controlled studies have suggested a significant risk of developing MM in individuals with significant occupational exposures in the agriculture, food, and petrochemical industries. An increased risk has been reported in farmers, especially in those who use herbicides and insecticides (eg, chlordane), and in people exposed to benzene and other organic solvents. There is conflicting evidence regarding long-term (>20 y) exposure to hair dyes and possible increased risk of developing MM.[14]

MGUS/Smoldering Multiple Myeloma (SMM)

Monoclonal gammopathy of undetermined significance (MGUS) is defined by the presence of three criteria:

MGUS is seen in 2-3% of the elderly Caucasian population. It is divided into the following three subtypes:

Patients with non-IgM MGUS have a risk of progression to MM at rate of 1% per year. For these patients, risk factors for progression to MM are as follows:

Patients with IgM MGUS have a risk of progression to Waldenstrom macroglobulinemia and less frequently lymphoma or amyloid light chain (AL) amyloidosis. IgM MGUS rarely progresses into MM. Light chain MGUS has a tendency to progress to light chain MM, AL amyloidosis, or light chain deposition disease.

A study by Wadhera et al examined secondary MGUS that developed in patients with MM. Of 1942 patients with MM, 128 (6.6%) developed a secondary MGUS at a median of 12 months from the diagnosis of MM. Overall survival was superior in patients with MM who developed secondary MGUS compared with the rest of the cohort.[15]

Smoldering MM is present when the serum M protein concentration is > 3 g/dL or the bone marrow plasma cell concentration is > 10% but there is no evidence of end-organ damage. Risk factors for progression of SMM to MM include any of the following:

The time to progression decreases with increasing numbers of risk factors, as follows:

Radiation

Radiation may play a role in some patients. An increased risk has been reported in atomic-bomb survivors exposed to more than 50 Gy: In 109,000 survivors of the atomic bombing of Nagasaki during World War II, 29 died from multiple myeloma between 1950 and 1976. Some more recent studies, however, do not confirm that these survivors have an increased risk of developing multiple myeloma.

A study of workers at the Oak Ridge Diffusion Plant in eastern Tennessee showed only a weak correlation of risk of multiple myeloma to uranium exposure.[16]

Chronic inflammation

A relationship between MM and preexisting chronic inflammatory diseases has been suggested. However, a case-control study provides no support for the role of chronic antigenic stimulation.

Infection

Human herpesvirus 8 (HH8) infection of bone marrow dendritic cells has been found in patients with multiple myeloma and in some patients with MGUS.

Epidemiology

MM accounts for 10% of all hematologic cancers.[17, 18]  The American Cancer Society estimates that in the United States, approximately 32,270 new cases of MM (17,530 in men and 14,740 in women) will be diagnosed in 2020.[13] The lifetime risk of getting MM is approximately one in 125 (0.8%).[19] Approximately 12,830 deaths from MM (7,190 in men and 5,640 in women) are expected to occur in 2020.[13]  Rates for new MM cases have not changed significantly over the last decade, while death rates fell from 3.49 to 3.24 per 100,000 from 2006 to 2016.[19]

In the US, the annual incidence of MM per 100,000 persons is 8.1 cases in white men, 4.9 cases in white women, 16.3 cases in black men, and 11.9 cases in black women. For Hispanics, the rates are 8.2 in men and 5.5 in women. Rates are lowest for Asians/Pacific Islanders, at 4.9 in men and 3.0 in women.[19]  According to a study of the ethnic disparities among patients with MM, Hispanics had the youngest median age at diagnosis (65 years) and whites had the oldest (71 years). Asians had the best overall survival rates, while Hispanics had the worst.[20]

The median age of patients with MM is 68 years for men and 70 years for women. Only 18% of patients are younger than 50 years, and only 3% of patients are younger than 40 years. The male-to-female ratio in MM is approximately 3:2.

Prognosis

MM is a heterogeneous disease, with survival ranging from 1 year to more than 10 years. Median survival in unselected patients with MM is 3 years. The 5-year relative survival rate is 46.6%.[19] Survival is higher in younger people and lower in the elderly.[13, 21]

The tumor burden and the proliferation rate are the two key indicators for the prognosis in patients with MM. Many schemas have been published to aid in determining the prognosis. One schema uses C-reactive protein (CRP) and beta-2 microglobulin (which is an expression of tumor burden) to predict survival, as follows[22] :

Poor prognostic factors include the following:

The prognosis by treatment is as follows:

Infections are an important cause of early death in MM. In a United Kingdom study, 10% of patients died within 60 days after diagnosis of MM, and 45% of those deaths were due to infection.[23] In a Swedish study, 22% of patients died of infection within the first year after diagnosis. The risk of both bacterial infections (eg, meningitis, septicemia, pneumonia) and viral infections (eg, herpes zoster, influenza) was seven times higher in patients with MM than in matched controls. The Swedish investigators also found that the risk of infections has increased in recent decades, and they argue that the use of more intensive treatment measures for MM (ie, newer drugs and high-dose chemotherapy with transplantation) has contributed to the increased risk.[24]

Patient Education

Patient education is very important in the management of MM. The International Myeloma Foundation (IMF) offers educational resources, a quarterly newsletter, and conferences. Patients or physicians can contact the IMF by phone at (800) 452-CURE (800-452-2873) in the United States and Canada or on the Web at International Myeloma Foundation.

Patient education should address, at a minimum, the following questions:

For patient education information, see Blood and Lymphatic System Center, as well as Myeloma.

History

Presenting signs and symptoms of multiple myeloma (MM) include bone pain, pathologic fractures, weakness, anemia, infection (often pneumococcal), hypercalcemia, spinal cord compression, and renal failure. In approximately 30% of cases, MM is discovered through routine blood screening when patients are being evaluated for unrelated problems. Typically, a large gap between the total protein and the albumin levels observed on an automated chemistry panel suggests a problem (ie, protein minus albumin equals globulin).

In one third of patients, MM is diagnosed after a pathologic fracture occurs; such fractures commonly involve the axial skeleton. Two thirds of patients complain of bone pain, commonly with lower back pain. This bone pain is frequently located in the back, long bones, skull, and/or pelvis.

Patients may also complain of nonspecific constitutional symptoms related to hyperviscosity and hypercalcemia.

Bone pain

Bone pain is the most common presenting symptom in MM. Most case series report that 70% of patients have bone pain at presentation. The lumbar spine is one of the most common sites of pain.

Pathologic fractures and bone lesions

Pathologic fractures are very common in MM; 93% of patients have more than one site of bony involvement. A severe bony event is a common presenting issue.

Spinal cord compression

The symptoms that should alert physicians to consider spinal cord compression are back pain, weakness, numbness, or dysesthesias in the extremities. Because spinal cord compressions in MM occur at multiple levels, comprehensive evaluation of the spine is warranted. Patients who are ambulatory at the start of therapy have the best likelihood of preserving function and avoiding paralysis.

Bleeding

Occasionally, a patient may come to medical attention for bleeding resulting from thrombocytopenia. Rarely, monoclonal protein may absorb clotting factors and lead to bleeding.

Hypercalcemia

Confusion, somnolence, bone pain, constipation, nausea, and thirst are the presenting symptoms of hypercalcemia. This complication may be present in as many as 30% of patients with MM at presentation. In most solid malignancies, hypercalcemia carries an ominous prognosis, but in MM, its occurrence does not adversely affect survival.

Infection

Abnormal humoral immunity and leukopenia may lead to infection. Pneumococcal organisms are commonly involved, but shingles (ie, herpes zoster) and Haemophilus infections are also more common in patients with MM.

Hyperviscosity

Hyperviscosity may be associated with a number of symptoms, including generalized malaise, infection, fever, paresthesia, sluggish mentation, and sensory loss. Patients may report headaches and somnolence, and they may bruise easily and have hazy vision. Patients with MM typically experience these symptoms when their serum viscosity is greater than 4 times that of normal serum.

Epistaxis may be a presenting symptom of MM with a high tumor volume. Occasionally, patients may have such a high volume of monoclonal protein that their blood viscosity increases, resulting in complications such as stroke, myocardial ischemia, or infarction.

Neurologic symptoms

Carpal tunnel syndrome is a common complication of myeloma. Meningitis (especially that resulting from pneumococcal or meningococcal infection) is more common in patients with MM. Some peripheral neuropathies have been attributed to MM. Long-term neurologic function is directly related to the rapidity of the diagnosis and the institution of appropriate therapy for MM.

Anemia

Anemia, which may be quite severe, is the most common cause of weakness in patients with MM.

Physical Examination

On head, ears, eyes, nose, and throat (HEENT) examination, the eyes may show exudative macular detachment, retinal hemorrhage, or cotton-wool spots. Pallor from anemia may be present. Ecchymoses or purpura from thrombocytopenia may be evident.

Bony tenderness is not uncommon in MM, resulting from focal lytic destructive bone lesions or pathologic fracture. Pain without tenderness is typical. Pathologic fractures may be observed. In general, painful lesions that involve at least 50% of the cortical diameter of a long bone or lesions that involve the femoral neck or calcar femorale are at high (50%) risk for a pathologic fracture. The risk of fracture is lower in upper-extremity lesions than in lower-extremity lesions. Even a small cortical defect can decrease torsional strength by as much as 60% (stress riser effect).

Neurologic findings may include a sensory level change (ie, loss of sensation below a dermatome corresponding to a spinal cord compression), neuropathy, myopathy, a Tinel sign, or a Phalen sign due to carpel tunnel compression secondary to amyloid deposition.

Extramedullary plasmacytomas, which consist of soft-tissue masses of plasma cells, are not uncommon. Plasmacytomas have been described in almost every site in the body. Although the aerodigestive tract is the most common location, reports also describe orbital, ear canal, cutaneous, gastric, rectal, prostatic, and retroperitoneal lesions.

On evaluation of the abdomen, hepatosplenomegaly may be discovered. Cardiovascular system examination may reveal cardiomegaly secondary to immunoglobulin deposition.

Amyloidosis may develop in some patients with MM. The characteristic physical examination findings that suggest amyloidosis include the following:

The shoulder pad sign is defined by bilateral swelling of the shoulder joints secondary to amyloid deposition. Physicians describe the swelling as hard and rubbery. Amyloidosis may also be associated with carpal tunnel syndrome and subcutaneous nodules.

Macroglossia may occur secondary to amyloid deposition in the tongue and is a common finding in patients with amyloidosis (see the image below).



View Image

Amyloidosis infiltrating the tongue in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Skin lesions that have been described as waxy papules or nodules may occur on the torso, ears, or lips.

Post-proctoscopic peripalpebral purpura strongly suggests amyloidosis. The term originated in the time when rectal biopsy was the initial procedure of choice for diagnosing amyloidosis, and the hemodynamic effect of the procedure—comparable to a prolonged Valsalva maneuver—would lead to burst capillaries in patients with amyloid infiltration of the vessels around the eyes, Patients also may develop these raccoonlike dark circles around their eyes as a result of coughing, vomiting, or forced expiration during spirometric testing).

Complications

Renal failure and insufficiency are seen in 25% of patients with MM,[25]  and may reflect any of the following:

Anemia, neutropenia, or thrombocytopenia is due to bone marrow infiltration of plasma cells. Thrombosis and Raynaud phenomenon due to cryoglobulinemia may be present.

Bone disease may result in the following:

Radiculopathy and/or cord compression may occur because of skeletal destruction and nerve compression.

Bacterial infection may develop; it is the leading cause of death in patients with myeloma. The highest risk is in the first 2-3 months of chemotherapy.

Purpura, retinal hemorrhage, papilledema, coronary ischemia, seizures, and confusion may occur as a result of hyperviscosity syndrome.

Hypercalcemia may cause polyuria and polydipsia, muscle cramps, constipation, and a change in the patient’s mental status.

Approach Considerations

The International Myeloma Workshop developed guidelines for standard investigative workup in patients suspected to have multiple myeloma. These guidelines include the following:[1]

Consider the risk of acute kidney injury, especially in the setting of contrast medium injection for imaging studies. Take care to limit patients’ exposure and maintain hydration.

Blood Studies

Perform a complete blood count (CBC) to determine if the patient has anemia, thrombocytopenia, or leukopenia. The CBC and differential may show pancytopenia. The reticulocyte count is typically low. Peripheral blood smears may show rouleau formation.

The erythrocyte sedimentation rate (ESR) is typically increased. Coagulation studies may yield abnormal results.

Obtain a comprehensive metabolic panel to assess levels of the following:

Urine Collection

Obtain a 24-hour urine collection for quantification of the Bence Jones protein (ie, lambda light chains), protein, and creatinine clearance. Quantification of proteinuria is useful for the diagnosis of MM (>1 g of protein in 24 h is a major criterion) and for monitoring the response to therapy. Creatinine clearance can be useful for defining the severity of the patient’s renal impairment.

Electrophoresis and Immunofixation

Serum protein electrophoresis (SPEP) is used to determine the type of each protein present and may indicate a characteristic curve (ie, where the spike is observed). Urine protein electrophoresis (UPEP) is used to identify the presence of the Bence Jones protein in urine. Immunofixation is used to identify the subtype of protein (ie, IgA lambda).

National Comprehensive Cancer Network (NCCN) guidelines also recommend the use of serum free light chain assay and plasma cell fluorescence in situ hybridization (FISH) for del 13, del 17p13, t(4;14), t(11;14), 1q21 amplification as part of the initial diagnostic workup.[2]

Chemical screening, including calcium and creatinine SPEP, immunofixation, and immunoglobulin quantitation, may show azotemia, hypercalcemia, an elevated alkaline phosphatase level, and hypoalbuminemia. A high lactate dehydrogenase (LDH) level is predictive of an aggressive lymphomalike course.

SPEP is a useful screening test for detecting M proteins. An M component is usually detected by means of high-resolution SPEP. The kappa-to-lambda ratio has been recommended as a screening tool for detecting M-component abnormalities. An M-component serum concentration of 30 g/L is a minimal diagnostic criterion for MM. In about 25% of patients, M protein cannot be detected by using SPEP.

Routine urinalysis may not indicate the presence of Bence Jones proteinuria. Therefore, a 24-hour urinalysis by means of UPEP or immunoelectrophoresis may be required. UPEP or immunoelectrophoresis can also be used to detect an M component and kappa or lambda light chains. The most important means of detecting MM is electrophoretic measurement of immunoglobulins in both serum and urine.

Quantitative Immunoglobulin Levels (IgG, IgA, IgM)

Suppression of nonmyelomatous immunoglobulin is a minor diagnostic criterion for MM. The level of MM protein (ie, M protein level), as documented by the immunoglobulin level, can be useful as a marker to assess the response to therapy.

Beta-2 Microglobulin and C-Reactive Protein

Beta-2 microglobulin is a surrogate marker for the overall body tumor burden. The level of beta-2 microglobulin is increased in patients with renal insufficiency without MM, which is one reason that it is a useful prognosticator in MM.[22] (See Prognosis.) Patients with MM and impaired renal function have a worse prognosis.

C-reactive protein (CRP) is a surrogate marker of interleukin (IL)-6 activity. IL-6 is often referred to as the plasma cell growth factor. Like beta-2 microglobulin, CRP is useful for prognostication.[22] (See Prognosis.)

Serum Viscosity

Check the serum viscosity in patients with central nervous system (CNS) symptoms, nosebleeds, or very high M protein levels. These findings may indicate hyperviscosity syndrome.

Radiography

Simple radiography is indicated for the evaluation of skeleton lesions, and a skeletal survey is performed when myeloma is in the differential diagnosis. Plain radiography remains the gold standard imaging procedure for staging newly diagnosed and relapsed myeloma, according to an International Myeloma Working Group consensus statement.[26]

Perform a complete skeletal series at diagnosis of MM, including the skull (a very common site of bone lesions in persons with MM; see the image below), the long bones (to look for impending fractures), and the spine.



View Image

Radiograph of the skull demonstrating a typical lytic lesion in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematolo....

Conventional plain radiography can usually depict lytic lesions. Such lesions appear as multiple, rounded, punched-out areas, most often in the skull, vertebral column, ribs, and/or pelvis. Less common but not rare sites of involvement include the long bones. Plain radiographs can be supplemented by computed tomography (CT) scanning to assess cortical involvement and risk of fracture. Diffuse osteopenia may suggest myelomatous involvement before discrete lytic lesions are apparent.

Findings from this evaluation may be used to identify impending pathologic fractures, allowing physicians the opportunity to repair debilities and prevent further morbidity.

Also see the topic Imaging Multiple Myeloma.

Magnetic Resonance Imaging

Magnetic resonance imaging (MRI) is useful in detecting thoracic and lumbar spine lesions, paraspinal involvement, and early cord compression. Findings from MRI of the vertebrae are often positive when plain radiographs are not. MRI can depict as many as 40% of spinal abnormalities in patients with asymptomatic gammopathies in whom radiographic studies are normal. For this reason, evaluate symptomatic patients with MRI to obtain a clear view of the spinal column and to assess the integrity of the spinal cord.

Also see the topic Imaging Multiple Myeloma.

Positron Emission Tomography

Comparative studies have suggested the possible utility of positron emission tomography (PET) scanning in the evaluation of MM.[27, 28] For example, a comparison study of PET scanning and whole-body MRI in patients with bone marrow biopsy-proven multiple myeloma found that although MRI had higher sensitivity and specificity than PET in the assessment of disease activity, when used in combination and with concordant findings, the 2 modalities had a specificity and positive predictive value of 100%.

These researchers suggest that the combination of modalities may be valuable for assessing the effectiveness of treatment, when aggressive and expensive regimens are used.[28] However, PET scanning has not yet been integrated into standard practice. The International Myeloma Working Group notes the potential usefulness of PET scanning in selected patients but suggests that such studies ideally should be performed in the context of a clinical trial.[26]

A study by Zamagni et al found that 18-F fluorodeoxyglucose (FDG) PET/CT scan findings were reliable predictors of prognosis among patients with multiple myeloma who had undergone thalidomide-dexamethasone induction therapy and double autotransplantation.[29]

Also see the topic Imaging Multiple Myeloma.

Bone Scan

Do not use bone scans to evaluate MM. Cytokines secreted by MM cells suppress osteoblast activity; therefore, typically, no increased uptake is observed. On technetium bone scanning, more than 50% of lesions can be missed.

Aspiration and Biopsy

MM is characterized by an increased number of bone marrow plasma cells. Plasma cells show low proliferative activity, as measured by using the labeling index. This index is a reliable parameter for the diagnosis of MM. High values are strongly correlated with progression of the disease.

Obtain bone marrow aspirate and biopsy samples from patients with MM to calculate the percentage of plasma cells in the aspirate (reference range, up to 3%) and to look for sheets or clusters of plasma cells in the biopsy specimen. Bone marrow biopsy enables a more accurate evaluation of malignancies than does bone marrow aspiration.

Histologic Findings

Plasma cells are 2-3 times larger than typical lymphocytes; they have eccentric nuclei that are smooth (round or oval) in contour with clumped chromatin and have a perinuclear halo or pale zone (see the image below). The cytoplasm is basophilic.



View Image

Bone marrow aspirate demonstrating plasma cells of multiple myeloma. Note the blue cytoplasm, eccentric nucleus, and perinuclear pale zone (or halo). ....

Many MM cells have characteristic, but not diagnostic, cytoplasmic inclusions, usually containing immunoglobulin. The variants include Mott cells, Russell bodies, grape cells, and morula cells. Bone marrow examination reveals plasma cell infiltration, often in sheets or clumps (see the image below). This infiltration is different from the lymphoplasmacytic infiltration observed in patients with Waldenstrom macroglobulinemia.



View Image

Bone marrow biopsy demonstrating sheets of malignant plasma cells in multiple myeloma. All images and text are (c) 2002 by the American Society of Hem....

Analysis of bone biopsy specimens may reveal plasmacytic, mixed cellular, or plasmablastic histologic findings. Approximate median survival by histologic type is as follows:

Cytogenetic Analysis

Cytogenetic analysis of the bone marrow may contribute significant prognostic information in multiple myeloma. The most significant cytogenetic abnormality appears to be deletion of 17p13. This abnormality is associated with shorter survival, more extramedullary disease, and hypercalcemia. This locus is the site of the TP53 tumor suppressor gene. Chromosome 1 abnormalities and c-myc defects are also significant prognostic factors in multiple myeloma.

Although not as well defined as in other hematologic malignancies, such as acute leukemia, risk-adapted therapy based on cytogenetic abnormalities is at the forefront of myeloma research.

Staging

Staging is a cumulative evaluation of all of the diagnostic information garnered and is a useful tool for stratifying the severity of patients’ disease. Currently, two staging systems for multiple myeloma are in use: the Salmon-Durie system, which has been widely used since 1975; and the International Staging System, developed by the International Myeloma Working Group and introduced in 2005.[30, 31]  A revision of the International Staging System, published in 2015, added genetic information to the standard laboratory tests.[32]  See also Multiple Myeloma Staging.

Salmon-Durie staging system

The Salmon-Durie classification of MM is based on three stages and additional subclassifications.

In stage I, the MM cell mass is less than 0.6 × 1012 cells/m2, and all of the following are present:

In stage II, the MM cell mass is 0.6-1.2 × 1012 cells/m2 or more. The other values fit neither those of stage I nor those of stage III.

In stage III, the MM cell mass is >1.2 × 1012 cells/m2, and all of the following are present:

Subclassification A includes relatively normal renal function (serum creatinine value < 2 mg/dL), whereas subclassification B includes abnormal renal function (serum creatinine value > 2 mg/dL)

Median survival is as follows:

Disease in subclassification B has a significantly worse outcome (eg, 2-12 mo survival in 4 separate series).

International Staging System

The International Staging System of the International Myeloma Working Group is also based on three stages.

Stage I consists of the following:

Stage II consists of the following:

Stage III consists of the following:

Median survival is as follows:

Revised International Staging System

In the 2015 revision of the International Staging System (ISS) , stage I comprises all of the following:

Stage II consists of all other possible combinations of ISS criteria, chromosomal abnormalities, and LDH other than those of stage I or III.

Stage III consists of the following:

Approach Considerations

Physicians must understand both the natural history of multiple myeloma (MM) and the limitations of current therapy in the treatment of the disease. The objective in therapy is to obtain the deepest response in the first round by choosing the appropriate regimen; this should lead to better overall survival in both transplant and non-transplant patients. In situations with no definite data on therapeutic choices, participation in clinical trials should be encouraged. For a summary of treatment approaches to MM, see Multiple Myeloma Treatment Protocols.

Progression of disease and timing of treatment

An important study by Dimopoulos and associates evaluated the risk of disease progression in asymptomatic subjects with MM.[33] This study evaluated 638 consecutive untreated subjects with MM. Of these subjects, 95 were asymptomatic and were not treated until their M protein value rose to greater than 5 g/dL. These subjects developed increased bone disease or symptoms of bone disease.

The individuals in this group were designated as either low risk (ie, no bone disease, M protein level < 3 g/dL, or Bence Jones protein level < 5 g/24 h) or high risk (ie, lytic bone disease and serum M protein level >3 g/dL or Bence Jones protein level >5 g/24 h). Intermediate-risk subjects did not have bone disease or an M protein level greater than 3 g/dL or a Bence Jones protein level greater than 5 g/24 h. The patients were evaluated every 2 months.

The median time for disease progression was 10 months in the high-risk group, 25 months in the intermediate-risk group, and 61 months in the low-risk group.[33] At the time of progression, subjects were treated with standard chemotherapy. Their response rates did not significantly differ from those of unselected populations. Median survival time from the institution of chemotherapy did not differ among the groups. Thus, asymptomatic subjects did not benefit from early treatment, and delayed treatment did not affect treatment efficacy (ie, survival).

A systematic review by He et al demonstrated a reduction in vertebral compressions and time to progression with early systemic treatment for asymptomatic patients, but this study also revealed an increase in acute leukemia in the early treatment group.[34] The failure to demonstrate improved survival may be due to the small number of patients studied.

The 2009 International Myeloma Workshop concluded that detection of any cytogenic abnormality suggests higher-risk disease, including chromosomal 13 or 13q deletion, t(4;14), and del17p and fluorescence in situ hybridization detection of t(4;14), t(14;16), and del17p.[35] Fluorescence in situ hybridization detection of 13q deletion alone is not considered a high-risk feature. International Staging System stages II and II and high serum beta(2)-microglobulin levels are suggestive of higher risk disease.

A study by Klein et al determined that the prognostic significance of t(4;14) may be eliminated or lessened among patients who receive lenalidomide and dexamethasone; however, del(17p13) and +1q21 are still associated with a dismal overall survival.[36] A study by Neben et al concludes that long-term administration of bortezomib in patients with del(17p13) may result in better overall and progression-free survival.[37]

Current therapeutic approaches

Overall, the care of patients with MM is complex and should focus on treatment of the disease process and any associated complications.[3, 4, 5] Although MM remains incurable, several drug therapies are valuable in the treatment of patients with MM, as are autologous stem cell transplantation, radiation, and surgical care in certain cases.

Several studies are evaluating the role of treatment in patients with high-risk smoldering multiple myeloma (SMM). Previous smaller studies evaluating thalidomide did not show a clear evidence of benefit with treatment in patients with SMM; however, these included patients with all risk levels of SMM.

In a phase III trial that was restricted to patients with high-risk SMM, the PETHEMA group found evidence of benefit from treatment with lenalidomide versus observation. After a median follow-up of 40 months, study patients who were randomized to lenalidomide and dexamethasone induction followed by lenalidomide maintenance demonstrated significantly prolonged median time to progression (median not reached vs 21 months) and higher 3-year survival rate (94% vs. 80%).[38]

Lenalidomide as single-agent therapy (without dexamethasone induction) may also slow progression of SMM to MM. A phase III trial in 182 patients found that after 3 years, SMM had not progressed to MM in 91% of patients receiving lenalidomide, compared with 66% of those who underwent observation only.  Many patients stopped taking lenalidomide early due to side effects (eg, fatigue, neutropenia); however, preliminary results suggest that even a short course of treatment may be beneficial.[39]

In addition, the success of three-drug combinations for MM has led to trials of their use in SMM. Triplets currently under study include carfilzomib, lenalidomide, and dexamethasone and daratumumab, lenalidomide, and dexamethasone.

Longer follow-up will be necessary, however. Concern for second primary malignancies (SPMs) with the use of lenalidomide is also a significant issue. Consequently, watchful observation and frequent monitoring remains the standard of care for patients with SMM. 

Patients with MM for whom therapy is indicated typically receive chemotherapy. Greater understanding of the cell biology of MM and the ability to identify prognostic factors has led to the increasing individualization of treatment for affected patients. Physicians treat many patients with high-dose therapy and peripheral blood or bone marrow stem cell transplantation.

For primary induction therapy in patients with MM who are candidates for transplantation, National Comprehensive Cancer Network (NCCN) guidelines recommend the following combinations as preferred regimens[2] :

Other recommended regimens, according to the NCCN, are as follows:

The NCCN considers the following regimens useful in certain circumstances (although triplet regimens should be used as the standard, patients not considered candidates for a 3-drug regimen can be started on a 2-drug regimen, with the third drug added once performance status improves):

Patients should be assessed for response after two cycles of one of the above regimens.

Patients with MM who are treated with lenalidomide or thalidomide are at significantly increased risk for thrombotic events, and many physicians incorporate anticoagulation strategies in their management. A study by Palumbo et al determined that aspirin and low-dose warfarin had similar efficacy in reducing serious thromboembolic events, acute cardiovascular events, and sudden deaths in patients with myeloma receiving thalidomide-based regimens compared with low-molecular weight heparin, except in elderly patients.37 In addition, the NCCN recommends that clinicians consider harvesting peripheral blood stem cells before patients have prolonged exposure to lenalidomide.[2]

As monotherapy or in combination, interferon alfa-2b and prednisone modestly prolong the disease-free interval.

A study by the Southwest Oncology Group compared lenalidomide plus dexamethasone to placebo plus dexamethasone in patients with newly diagnosed myeloma.[40] The study determined that lenalidomide plus dexamethasone had superior 1-year progression-free survival, overall response rate, and very good partial response rate, suggesting that it is safe and effective as initial therapy for patients with newly diagnosed myeloma. In February 2015, the US Food and Drug Administration (FDA) expanded the approval of lenalidomide, in combination with dexamethasone, to include newly diagnosed MM. The original indication was for patients who had received at least 1 prior therapy.

A phase III randomized, open-label trial of 119 patients with high-risk smoldering MM found that early treatment with lenalidomide plus dexamethasone, followed by maintenance therapy with lenalidomide, delayed progression to symptomatic disease and increased overall survival.[41, 42]

Adjunctive therapy for MM includes radiation therapy to target areas of pain, impending pathologic fracture, or existing pathologic fracture. Bisphosphonate therapy serves as prophylaxis (ie, primary, secondary) against skeletal events (eg, hypercalcemia, spinal cord compression, pathologic fracture, need for surgery, need for radiation). Evidence suggests that it may be effective in treating bone pain and in decreasing the likelihood of lesion recurrence.[43, 44, 45]

Adjunctive therapy may also include any of the following, as appropriate:

Bone disease guidelines

Bisphosphonates (eg, zoledronic acid, pamidronate) or denosumab for prevention of skeletal related events (SREs) should be considered for all patients with MM receiving first-line antimyeloma therapy, regardless of presence of osteolytic bone lesions.[45, 46]  See Guidelines/Management of Multiple Myeloma–related Bone Disease

Chemotherapy and Immunotherapy

In patients with symptomatic MM, chemotherapy is required. In asymptomatic patients with MM, treatment is delayed until disease clinically progresses or until serum or urine levels of M protein substantially increase.

The M-component level in serum and/or urine is an indicator of the tumor burden; its reduction after chemotherapy is used as a sign of response. A 50% reduction in M-component is considered a good clinical response (according to the Chronic Leukemia-Myeloma Task Force). The historical standard regimen of melphalan plus prednisone induces a response in 50-60% of patients with MM. Disappearance of the M component on electrophoresis occurs in only 3% of patients, and cure is extraordinarily rare.

The first step before starting therapy in MM is to determine whether a patient is a candidate for an autologous stem cell transplant. Eligibility depends primarily on the patient’s age and comorbidities. Typically an age of 65 years is used as a cut-off point for transplant eligibility. Thus, treatment for MM is best looked at in terms of the following three categories of patients:

Young, newly diagnosed patients who are potential transplant candidates

Conventionally, VAD (vincristine, doxorubicin [Adriamycin], and dexamethasone) chemotherapy has been used to decrease the tumor burden in MM as preparation for transplantation. VAD is administered as a 4-day continuous intravenous infusion of vincristine and doxorubicin, with 4 daily oral doses of dexamethasone. Patients require a central venous catheter for delivery of the infusion. In selected patients, this therapy can be performed in an outpatient setting.

Many researchers feel that the high-dose steroid component of VAD accounts for much of its efficacy. In some patients, high-dose dexamethasone alone may produce significant clinical responses.

Significant concerns with the use of infusion therapy include the risk of soft-tissue injury if the chemotherapy agent infiltrates, the risk of cardiac injury from the doxorubicin, and the risk of infection or hyperglycemia from the high-dose steroids. Some patients also experience adverse central nervous system (CNS) effects from the high-dose steroids. Given these risks, and the higher response rates of new agents (thalidomide, lenalidomide, and bortezomib), VAD is now considered suboptimal treatment. Overall, data on these novel agents are very encouraging and promising. Nevertheless, oncologists will need further studies to help define the exact timing and role of novel agents in the treatment of MM.

Regimens with thalidomide 

Thalidomide has proved effective against MM. The superiority of induction regimens containing thalidomide was demonstrated in randomized trials that compared VAD with thalidomide plus dexamethasone[47] ; thalidomide and doxorubicin plus dexamethasone[48] ; and thalidomide plus VAD.[49]

Thalidomide has a well-established role as first-line therapy, either as a single agent or in combination with steroids in patients with MM. The toxicity of this drug is predominantly sensory neuropathy, and because of the drug’s teratogenicity, close monitoring is required to avoid inadvertent administration during pregnancy.

Regimens with lenalidomide

An analogue of thalidomide, lenalidomide (Revlimid) is now a standard component of MM therapy. In July 2013, Celgene Corp announced that a phase III trial of lenalidomide (Revlimid) met the main goal of improving progression-free survival (PFS) in patients with newly diagnosed MM.[50] The drug was already approved for use in previously-treated MM, mantle cell lymphoma, and transfusion-dependent anemia caused by myelodysplastic syndromes.

In the late-stage study, treatment with lenalidomide combined with dexamethasone in patients with newly diagnosed MM resulted in significantly longer survival without the cancer worsening, than did treatment with a regimen consisting of melphalan, prednisone, and thalidomide (MPT).[50]

In a randomized, double-blind, placebo-controlled trial, lenalidomide plus high-dose dexamethasone proved superior to high-dose dexamethasone alone as treatment for newly diagnosed MM.[47] The overall response rate was 84% in the lenalidomide plus high-dose dexamethasone group versus 53% in the high-dose dexamethasone group, with 22% of patients achieving complete remission in the lenalidomide plus high-dose dexamethasone arm.

PFS and overall survival favored lenalidomide plus high-dose dexamethasone, but 12-month survival for both arms was > 90%. A very important observation, however, was the high incidence of deep venous thrombosis in the lenalidomide plus high-dose dexamethasone arm.[47]

In another randomized trial of lenalidomide plus high-dose dexamethasone (LD) versus lenalidomide plus low-dose dexamethasone (Ld) in newly diagnosed MM, Rajkumar found that although the overall response rate within the first 4 months favored LD, analysis at 1 year, overall survival was 96% in the Ld arm compared with 87% in the LD arm (p=0.0002). As a result, the trial was stopped, and patients on high-dose therapy were crossed over to low-dose therapy.[51]

Another trial assessed the safety and efficacy of the combination regimen clarithromycin (Biaxin), lenalidomide (Revlimid), and dexamethasone (BiRD) as first-line therapy for MM.[52] Of the 72 patients enrolled, 65 had an objective response (90.3%). A combined stringent and conventional complete response rate of 38.9% was achieved, and 73.6% of the patients achieved at least a 90% decrease in M-protein levels. BiRD was found to be an effective regimen with manageable side effects in the treatment of symptomatic, newly diagnosed MM.

Patients tolerate lenalidomide therapy well, and nausea is usually minimal. Patients typically experience total alopecia, but other adverse effects (eg, peripheral neurotoxicity, constipation) are usually mild. Pancytopenia is expected, but is not severe enough to require hospitalization for infection or transfusion unless the patient also has some other cause of bone marrow suppression.

Regimens with bortezomib

Bortezomib (Velcade), a proteosome inhibitor, has shown striking activity against MM. Objective responses as high as 27.7% in patients with relapsed and heavily pretreated MM[53] led to its approval by the FDA in 2003. Subsequent studies reported response rates as high as 80% when bortezomib is combined with melphalan.

A randomized trial that compared bortezomib plus dexamethasone with VAD for induction showed response rates of 80% for the bortezomib plus dexamethasone arm versus 62.8% for the VAD arm.[54] This regimen has been shown to be active not only before but also after transplantation. Following high-dose therapy and autologous transplantation, the rate of very good partial response or better continued to favor bortezomib plus dexamethasone. This benefit was observed independent of beta-2 microglobulin or adverse cytogenetic risk groups.

Similarly, a superior response rate was seen when the combination of bortezomib, thalidomide, and dexamethasone (VTd) was compared with thalidomide plus dexamethasone in a large phase III study: 93% in the VTd arm versus 80% in the thalidomide-dexamethasone arm, in which patients went on to receive tandem autologous stem cell transplantation.[55] As in other studies, response was independent of adverse prognostic risk factors.

The phase III Velcade as Initial Standard Therapy in MM (VISTA) trial found that the combined treatment of bortezomib, melphalan, and prednisone (VMP) significantly prolongs overall survival compared with melphalan and prednisone (MP) after lengthy follow-up and extensive subsequent antimyeloma therapy. A notable outcome of this study was that first-line bortezomib use did not induce more resistant relapse. VMP used upfront appears more beneficial than first treating with conventional agents and saving bortezomib-based and other novel-agent–based treatment until relapse.[56]

A study by Harousseau et al confirms the role of bortezomib in the initial nonintensive management of MM.[57]

A study by Sher et al found that a combination of bortezomib, pegylated liposomal doxorubicin, and thalidomide, known as the VDT regimen, had overall response rate and complete plus near-complete response rates of 78% and 35%, respectively.[58] The study concluded that VDT was a tolerable and effective regimen that may induce high response rates in patients considered to be poor candidates for steroid-based treatments.

Bortezomib appears to be of especial benefit in patients with plasma cell leukemia and renal failure. The predominant adverse effects were neuropathy, hypotension, and thrombocytopenia. Despite these results, the exact timing of bortezomib administration in the treatment plan of patients with newly diagnosed MM is still evolving through ongoing research.

The FDA approved administration of bortezomib by the subcutaneous (SC) route in January 2012. A study by Moreau et al found that the efficacy of SC bortezomib is not inferior to that of standard IV administration. Moreau also observed a better safety profile with SC administration: in particular, the incidence of grade 2 or greater peripheral neuropathy was 24% for SC compared with 41% for IV; grade 3 or higher peripheral neuropathy occurred in 6% of patients with SC administration vs 16% for IV administration.[59] Starting therapy with SC administration may be considered for patients with pre-existing peripheral neuropathy and those at high risk for it.

A study by Mateos et al found that patients with cytogenetic abnormalities had similar response to bortezomib therapy but shorter survival. The authors concluded that the present treatment schema does not overcome the negative prognosis associated with high-risk cytogenetic abnormalities.[60]

Daratumumab

In September 2019, the FDA approved daratumumab with VTd for newly diagnosed patients with MM who are eligible for autologous stem cell transplantation (ASCT). Approval was based on results from the phase III CASSIOPEIA study (n=1085) that evaluated whether adding daratumumab to VTd before and after ASCT would improve the complete response rates in newly diagnosed patients. Results from the first part of the trial showed complete response rate post-consolidation was significantly higher with VTd plus daratumumab compared with VTd alone (29% vs 20%). At a median follow-up of 18.8 months, the addition of daratumumab resulted in a 53% reduction in the risk of disease progression or death compared with VTd alone.[61]

High-risk patients who are potential transplant candidates

Patients are considered at high risk if their MM meets any of the following criteria:

This group represents about 25% of those with newly diagnosed MM, with an expected median survival of 2 years or less. Although they respond to traditional therapies for induction, these individuals tend to relapse rapidly. Therefore, novel agents should be considered up front for these patients.

The advent of thalidomide, lenalidomide, and bortezomib has substantially improved outcomes in these high-risk groups. In fact, these novel agents appear to overcome the influence contributed by high-risk cytogenetics.[62, 63] Once a response has been achieved, then patients can be brought to autologous stem cell transplantation.

Newly diagnosed elderly patients who are not transplant candidates

All of the above regimens may be used in patients who are not being considered for transplantation. The following, however, can only be used in patients who will not be undergoing transplantation, as they impair stem cell reserve.

The gold standard has been the MP regimen since as far back as the 1950s. This regimen typically consists of melphalan 9 mg/m2 and prednisone 100 mg given on days 1-4, with courses repeated at 4- to 6-week intervals for at least 1 year. A meta-analysis of 4930 patients from 20 randomized trials compared MP with other drug combinations and showed a significantly higher response rate (60%) with this combination, with a response duration of 18 months and overall survival of 24 to 36 months.[64]

A three-arm study looked at MP plus thalidomide versus MP versus VAD induction, followed by high-dose melphalan and autologous stem cell transplantation in 447 patients between ages 65 and 75 years.[65]  Patients were randomized, with overall survival as the primary endpoint. The response rates in the MP plus thalidomide arm and transplantation arm were similar; the complete response rate was significantly better in the MP plus thalidomide and the transplantation arms than in the MP arm.[65]

MP plus thalidomide is now recommended as first-line treatment. MP plus lenalidomide has also shown promise.[66]

Hulin et al conducted a randomized, placebo-controlled, phase III trial to investigate the efficacy of adding thalidomide to MP in 229 elderly patients (>75 y) newly diagnosed with MM.[67] During each 6-week cycle, melphalan 0.2 mg/kg/d plus prednisone 2 mg/kg/d was given to all patients on days 1-4 for 12 cycles. In addition, patients were randomly assigned to receive thalidomide 100 mg/d PO (n = 113) or placebo (n = 116), continuously for 72 weeks.

Overall survival was significantly longer in the group that received thalidomide (median, 44 mo) compared with placebo (median, 29.1 mo).[67] Progression-free survival was also significantly prolonged in the thalidomide group (median, 24.1 mo) relative to the placebo group (median, 18.5 mo). However, the investigators noted peripheral neuropathy and neutropenia were significantly increased in the thalidomide group.[67]

A randomized, controlled trial evaluated the addition of thalidomide to standard MP chemotherapy in elderly patients with previously untreated MM. Although no impact on survival was observed, more patients in the thalidomide group achieved an objective response. Of note, thromboembolic events did not increase in the thalidomide group.[68]

A separate study by Fayers et al concluded that thalidomide added to MP therapy improved overall survival and progression-free survival in previously untreated elderly patients with multiple myeloma, extending the mean survival time by an average of 20%.[69]

A study by Gay et al assessed the addition of thalidomide and/or bortezomib to standard oral MP treatment in 1175 elderly patients with newly diagnosed myeloma.[70] The study found that these novel agents helped achieve maximal response in these patients.

A study by Morgan et al found that cyclophosphamide, thalidomide, and dexamethasone (CTD) produced higher response rates than melphalan and prednisolone among newly diagnosed elderly patients with multiple myeloma; however, CTD was not associated with improved survival outcomes.[71]

A phase III study by the ALCYONE Trial Investigators found that the addition of daratumumab to the combination of bortezomib, melphalan, and prednisone in patients with newly diagnosed multiple myeloma who are ineligible for autologous stem-cell transplantation resulted in a lower risk of disease progression or death.[72] At a median follow-up of 16.5 months, results of treatment with and without daratumumab were as follows:

   However, the rate of grade 3 or 4 infections was 23.1% in the daratumumab group and 14.7% in the control group.

The MAIA trial was an open-label, randomized, phase 3 study comparing lenalidomide with low-dose dexamethasone with or without daratumumab in patients with newly diagnosed multiple myeloma ineligible for ASCT. The study demonstrated an improvement in progression-free survival in the daratumumab combination arm compared to the control arm. Patients with a complete response or better was 47.6% in the daratumumab group and 24.9% in the control group. A total of 24.2% of the patients in the daratumumab group, as compared with 7.3% of the patients in the control group, had results below the threshold for minimal residual disease (1 tumor cell per 105 white cells).[73]

Maintenance therapy

In spite of advances in treatment, multiple myeloma remains an incurable disease. To improve overall survival (OS) in these patients, a number of trials have evaluated the role of maintenance therapy in both transplant-eligible and transplant- ineligible patients.

Five large phase III studies have looked at role of thalidomide maintenance after autologous stem cell transplant (ASCT). Three initial studies showed an improvement in both progression-free survival (PFS) and OS.[74, 75, 76] However, two subsequent studies—including one large study with 1970 patients—did not show an improvement in OS with thalidomide maintenance.[77, 78] Long-term use of thalidomide is also associated with significant neuropathy, thus limiting its use in maintenance therapy.

Given its favorable toxicity profile and efficacy at low doses, lenalidomide has also been studied for maintenance therapy. Two large trials, CALGB 100104 and IFM 05-02, have evaluated the role of lenalidomide in maintenance therapy, using slightly different protocols and having somewhat different outcomes.[79, 80] Patients in both studies received induction treatment followed by ASCT. In the IFM 05-02 study, however, all patients received 2 months of consolidation treatment with lenalidomide before being randomized to lenalidomide or placebo.

Both studies showed a significant improvement in time to progression (46 vs 27 months in CALGB study and 41 vs 23 months in IFM study). However, CALGB 100104 study showed significant improvement in OS (85 % vs 77 %), whereas IFM 05-02 did not show an improvement in OS. Both studies showed an increased incidence of hematologic toxicity and second primary malignancies (SPMs), particularly acute myelogenous leukemia/myelodysplastic syndrome in the lenalidomide arm.

The reason for the difference in the two studies in terms of OS benefit is not very clear. Since all the patients in the IFM trial received 2 months of consolidation treatment with lenalidomide following ASCT, it is possible that only short period of maintenance therapy, rather than continuous maintenance therapy, is required to achieve all the OS benefit seen in the CALGB trial.

A meta-analysis shows the benefit of maintenance lenalidomide, with a 51% reduction in the risk of recurrence.[81] This benefit outweighs the risk of SPM seen in the trials of lenalidomide maintenance.

Bortezomib has also been shown to be effective for maintenance therapy in the  HOVON-65/GMMG-HD4 trial.[82] In this trial, patients were randomized to either PAD (bortezomib, doxorubicin [Adriamycin], and dexamethasone) induction followed by bortezomib maintenance or to VAD induction followed by thalidomide maintenance. PFS in the PAD arm was significantly better than in the VAD arm (35 vs 28 months). Patients with high-risk cytogenetics, especially del(17p13) and t(4;14) abnormalities, seemed to benefit more with bortezomib maintenance.

Although several trials have shown the benefit of maintenance therapy after ASCT, the risk of SPM and the need for continuous treatment should be kept in mind. Individual patient characteristics should be taken in consideration before recommending maintenance therapy.

Maintenance therapy has also been evaluated in non–transplant eligible patients. Thalidomide has been studied as maintenance in a number of trials; most of the trials have shown only advantage in PFS, with no advantage in OS. The main problem with thalidomide has been the high incidence of neuropathy in these patients.

A trial of lenalidomide as maintenance therapy after induction with melphalan, prednisone, and lenalidomide showed a significant improvement in PFS (26 vs 7 months) but similar 4-year OS. Patients in the lenalidomide arm had more hematologic toxicity, including neutropenia, thrombocytopenia, and higher risk of second primary malignancy. However, given its overall tolerability, lenalidomide is a good option for induction and maintenance therapy in transplant-ineligible patients.[83]

A number of trials have also evaluated bortezomib in maintenance therapy. All of them have showed benefit in PFS but no clear OS benefit. Bortezomib given once a week in maintenance seems to be better tolerated and associated with lesser neuropathy.[84]

Patients with refractory disease or relapse

Patients who have a relapse after initial disease control may be treated with any of the agents not already utilized. If the relapse occurs longer than 6 months after the initial therapy, then the initial regimen can be used again.

Among the other choices for salvage therapy are the following preferred regimens[2] :

Bortezomib has a well-established role as salvage therapy, based on a phase III randomized trial showing a response rate of 38% versus 18% in patients receiving dexamethasone only.[53] Median progression-free survival was 6.22 months in the bortezomib arm versus 3.49 months in the dexamethasone-only group.

In 2012, the FDA approved carfilzomib (Kyprolis) for the treatment of patients with MM who have received at least two prior therapies including bortezomib and an immunomodulatory agent, and have demonstrated disease progression on or within 60 days of therapy completion. The approval was based on a phase 2b, single-arm, multicenter clinical study of 266 patients with relapsed multiple myeloma with other therapies. The study assessed for overall response rate (ORR), which was 22.9% over a median duration of 7.8 months.[85]

Panobinostat (Farydak) is a histone deacetylase (HDAc) inhibitor approved in February 2015. It is indicated in combination with bortezomib and dexamethasone for treatment of MM in patients who have received at least two prior regimens, including bortezomib and an immunomodulatory agent. The FDA approval was based on efficacy and safety data in a prespecified subgroup analysis of the phase III PANORAMA-1 (PANobinostat ORAl in Multiple MyelomA) trial, in which patients treated with panobinostat (n = 94) had a median progression-free survival of 10.6 months, compared with 5.8 months for patients in the placebo arm (n= 99) (hazard ratio= 0.52 [95% confidence interval: 0.36, 0.76]).[86]

Also in 2015, the FDA expanded carfilzomib’s indication for multiple myeloma based on data from the ASPIRE study. In this study, carfilzomib was combined with lenalidomide and dexamethasone (KRd) for patients with relapsed multiple myeloma who had received 1-3 prior lines of therapy. The study showed a significant improvement in progression-free survival (PFS) for patients treated in the KRd arm compared with those treated with lenalidomide and low-dose dexamethasone (Rd) alone. The median PFS was 26.3 months in the KRd arm compared to 17.6 months in the Rd arm.[87]

The following three new drugs were approved in November 2015:

Daratumumab gained approval from the FDA for patients with MM who had received at least three prior treatments, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), or whose disease is refractory to both a PI and an IMiD. The approval was based on the phase II MMY2002 (SIRIUS) study that showed treatment with single-agent daratumumab resulted in an ORR of 29.2% in patients who received a median of five prior lines of therapy, including a PI and an IMiD.[88]

Stringent complete response (sCR) was reported in 2.8%, very good partial response (VGPR) was reported in 9.4%, and partial response (PR) was reported in 17% of patients. For responders, the median duration of response was 7.4 months. At baseline, 97% of patients were refractory to their last line of therapy, 95% were refractory to both a PI and an IMiD, and 77% were refractory to alkylating agents.[88] These data are supported by similar results from a phase I/II trial.[89]

Ixazomib is a reversible proteasome inhibitor. It preferentially binds and inhibits the chymotrypsinlike activity of the beta 5 subunit of the 20S proteasome. Ixazomib is indicated in combination with lenalidomide and dexamethasone for patients with multiple myeloma who have received at least 1 prior therapy. Approval was based on data from the phase 3 TOURMALINE-MM1 trial, an international, randomized, double-blind clinical trial of 722 patients with treatment-refractory or recurrent multiple myeloma. It compared ixazomib with placebo the patients who also received lenalidomide and dexamethasone. Median progression-free survival was improved by 35% with ixazomib compared with placebo (20.6 vs 14.7 months; P = 0.012).[90]

Elotuzumab is a humanized IgG1 monoclonal antibody that specifically targets the SLAMF7 (signaling lymphocytic activation molecule family member 7) protein. SLAMF7 is expressed on myeloma cells and natural killer cells and plasma cells. Elotuzumab facilitates the interaction with natural killer cells to mediate the killing of myeloma cells through antibody-dependent cellular cytotoxicity. It is indicated for use in combination with lenalidomide and dexamethasone for MM in patients who have received 1-3 prior therapies.

Approval was based on the ELOQUENT-2 trial, a randomized, open-label clinical study that included 646 participants with multiple myeloma who had experienced relapse or who had not responded to previous treatment. The addition of elotuzumab to the combination of lenalidomide and dexamethasone extended progression- free survival to 19.4 months, as compared with 14.9 months seen in patients treated with lenalidomide and dexamethasone (P< 0.001). Additionally, the overall response rate (including complete and partial responses) was 78.5%, compared with 60.1% in patients receiving lenalidomide and dexamethasone (P< 0.001).[91]

The ELOQUENT-3 trial studies 117 patients with multiple myeloma that was refractory or relapsed and refractory to lenalidomide and a proteasome inhibitor. Patients received elotuzumab plus pomalidomide and dexamethasone or pomalidomide and dexamethasone alone (control group). Median progression-free survival was 10.3 months in the elotuzumab group and 4.7 months in the control group. The overall response rate was 53% in the elotuzumab group compared with 26% in the control group.[92]

The FDA approved the anti-CD38 monoclonal antibody isatuximab (Sarclisa), in March 2020. It is indicated for relapsed or refractory MM in combination with pomalidomide and dexamethasone in patients who have received at least 2 prior therapies including lenalidomide and a proteasome inhibitor. Approval was based on the ICARIA-MM clinical trial (n = 307). The median PFS was 11.5 months in the isatuximab-pomalidomide-dexamethasone group compared with 6.5 months in the pomalidomide-dexamethasone group (p = 0.001).[6]

Thalidomide is useful in the treatment of patients with relapsing and refractory MM. Its antiangiogenic properties have become increasingly apparent as a critical step in the proliferation and spread of malignant neoplasm.[93, 94] In a Mayo Clinic study, nearly one third of patients with advanced MM in whom current standard chemotherapy or stem cell transplantation failed were shown to respond to thalidomide for a median duration of nearly 1 year.[95]

An important prospective placebo-controlled trial of the addition of lenalidomide to dexamethasone in relapsed cases of MM demonstrated spectacular results.[96] The major response rate with lenalidomide was 61% compared with 19.9% in the placebo arm. There was a significant improvement in time to progression (11.1 in the lenalidomide plus dexamethasone group vs 4.7% in the placebo group). Overall survival was significantly improved.[96]

A study by Lacy et al found that pomalidomide overcame resistance in MM that was refractory to both lenalidomide and bortezomib.[97] In February 2013, pomalidomide was approved by the FDA for use in patients with MM who have received at least two previous therapies (including lenalidomide and bortezomib) and have disease progression on or within 60 days of completion of the last therapy.[98, 99]

This approval was supported by a phase II study comparing pomalidomide plus low-dose dexamethasone with pomalidomide alone in patients with relapsed MM refractory to their last therapy who had received lenalidomide and bortezomib. Of the 221 patients who were evaluable for response, 29.2% in the pomalidomide plus low-dose dexamethasone arm achieved a partial response or better, compared with 7.4% in the pomalidomide-alone arm.[98] The median duration of response for the former was 7.4 months; the median had not been reached for the latter.

In another study, Miguel et al found that the combination of pomalidomide with low-dose dexamethasone yielded a longer median progression-free survival (PFS) in 455 patients with refractory or relapsed and refractory MM than high-dose dexamethasone alone.[100] In the open-label, randomized study patients received 28-day cycles of either pomalidomide (4 mg/day on days 1-21) plus low-dose dexamethasone (40 mg/day on days 1, 8, 15, and 22) or only high-dose dexamethasone (40 mg/day on days 1-4, 9-12, and 17-20). At follow-up (median, 10 months), median PFS was 4.0 months for the combination therapy group, compared with 1.9 months for the monotherapy group, for a hazard ratio of 0.48. Rates of most adverse events were similar in the two groups.[100]

In January 2016, the FDA approved carfilzomib in combination with dexamethasone for relapsed or refractory multiple myeloma in patients who have received 1-3 prior lines of therapy. Approval was based on the ENDEAVOR study (n=929) where a statistically significant improvement in median progression-free survival was observed with carfilzomib plus dexamethasone compared with bortezomib plus dexamethasone in patients with relapsed multiple myeloma (26.3 mo vs 17.6 mo; p=0.0001). Overall survival data are not yet available.[101]

The first selective inhibitor of nuclear export (SINE), selinexor, was approved by the FDA in July 2019. Selinexor acts on tumor suppressor proteins (TSPs), growth regulators, and mRNAs of oncogenic proteins by blocking exportin 1 (XPO1). Inhibition of XPO1 leads to accumulation of TSPs in the nucleus, reductions in several oncoproteins (eg, c‐myc, cyclin D1), cell cycle arrest, and apoptosis of cancer cells. It is indicated in combination with dexamethasone for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least 4 prior therapies and whose disease is refractory to at least 2 proteasome inhibitors, at least 2 immunomodulatory agents, and an anti-CD38 monoclonal antibody.

The multicenter, single-arm, open-label STORM trial analyzed selinexor plus dexamethasone. STORM part 2 included 122 patients with relapsed/refractory disease who previously had 3 or more treatments including: an alkylating agent, glucocorticoids, bortezomib, carfilzomib, lenalidomide, pomalidomide, and an anti-CD38 monoclonal antibody. Trial participants also had myeloma that was refractory to glucocorticoids, a proteasome inhibitor, an immunomodulatory agent, an anti-CD38 monoclonal antibody, and to the last line of therapy that they had. FDA approval was based on results from the 83 patients from the STORM trial who were refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide, and daratumumab. This group had a 25.4% overall response rate, 1% stringent complete response rate, 5% very good partial response, and 19% partial response rate.[102, 103]  

Transplantation

Using the patient’s own (ie, autologous) bone marrow or peripheral blood stem cells facilitates more intense therapy for MM. After harvesting the stem cells from the patient, physicians can use otherwise lethal doses of total body irradiation and chemotherapy and then “rescue” the patient by reinfusing the harvested cells. This process of myeloablative therapy, followed by the reinfusion of stem cells, is termed autologous stem cell transplantation.

This sequence of therapy allows physicians to use melphalan at an approximately 10-20 times higher dose than is used in standard therapy.[48] In autologous transplantation, the reinfused stem cells or bone marrow act as a support to the patient but do not offer additional anticancer effects.

Tandem autologous transplantation has been proposed as a way of overcoming the incomplete response to a single transplant. A 2-arm trial of single versus tandem transplantation revealed no difference in overall survival at 54 months.[104]

Another two-arm study that compared single versus tandem transplants for newly diagnosed MM showed that whereas double autologous stem cell transplantation effected superior complete or near-complete response rates, relapse-free survival, and event-free survival (EFS), it failed to significantly prolong overall survival.[105] Benefits offered by double autologous stem cell transplantation were particularly evident among patients who failed to achieve at least a near-complete response after one autotransplantation.

A review of long-term outcomes of several autotransplantation trials for MM found that tandem transplantations were superior to both single transplantations and standard therapies and that tandem transplantations with thalidomide were superior to trials without thalidomide.[106] However, postrelapse survival (PRS) was superior when initial EFS exceeded 1280 days and when tandem transplantations had been administered, whereas PRS was shorter when EFS lasted 803 days or less and when trials had included thalidomide and bortezomib.[106]

Two randomized prospective studies compared standard chemotherapy with high-dose autologous transplantation. In the first study of 200 subjects, researchers observed better response rates (ie, 81% for the transplantation group vs 57% for the conventionally treated group) and better 5-year event-free survival rates (ie, 28% vs 10%).[107] The second study also showed a significant improvement in event-free survival rates and superior quality of life for subjects treated with the high-dose approach.

In highly selected patients with MM, physicians may use allogeneic (ie, from someone else) transplantation. In this approach, physicians administer myeloablative therapy and infuse stem cells (ie, peripheral blood or bone marrow) obtained from a donor, preferably a human leukocyte antigen (HLA)-identical sibling.

The advantage of this approach over autologous transplantation is that the patient is not at risk of being reinfused with MM cells. In addition, the donor’s immune system may fight the recipient’s cancer (ie, graft vs myeloma effect). Unfortunately, the donor’s immune system may also attack the recipient’s body (ie, graft vs host effect).

Physicians use allogeneic transplantation less often than autologous transplantation in MM patients, for several reasons. First, the risks of complications and death from allogeneic transplantation increase with age, and most patients with MM are older than the ideal age for allogeneic transplantation. 

Second, the transplantation-related mortality rate is quite high in patients with MM who undergo allogeneic transplantation. The death rate within 100 days of transplantation ranges from 10% to 56% in different case series.

Third, although some survivors experience long-term disease-free results after allogeneic transplantation, a retrospective case-matched analysis of allogeneic versus autologous transplantation showed a median survival of 34 months for the autologous transplantation group and 18 months for the allogeneic group.

The exception to this rule is the rare patient with a twin donor. In a limited study of 25 transplantations involving twins, outcomes with syngeneic transplantations were superior, with reduced transplantation-related mortality.

The development of a nonmyeloablative preparative regimen for MM allogeneic transplantation is changing the equation. A republished report of 52 high-risk patients who underwent nonmyeloablative transplants described a 17% mortality rate.[108] Progression-free survival at 18 months was roughly 30%.

A phase II trial of autologous stem cell transplantation followed by a nonmyeloablative matched sibling related donor transplant demonstrated this approach to be feasible, with low treatment-related mortality.[109] Further studies are needed to evaluate relative efficacy.

Allotransplants have markedly reduced activity; therefore, the use of nonmyeloablative regimens (mini-allotransplantation) may hold promise for more widely exploiting this feature.[110, 111]

A study by Moreau et al determined that achievement of very good partial response (VGPR) after induction therapy is an important prognostic factor in patients undergoing autologous stem cell transplantation.[112] VGPR was significantly improved with bortezomib-dexamethasone induction therapy.

A study by Harousseau et al also concluded that this combination significantly improved postinduction and posttransplantation complete response/near response rate at at least VGPR rates compared with VAD.[113] Cavo et al also concluded that this combination represents a new standard of care for patients with multiple myeloma who are eligible for transplant.[114]

In MM patients with progressive or relapsing disease following autologous stem-cell transplantation, treatment with the combination of bortezomib, thalidomide and dexamethasone is more effective than treatment with thalidomide and dexamethasone alone, although triple therapy is associated with a greater risk of grade 3 neurotoxicity.[115]

Interferon Alfa Therapy

Intense research has focused on the use of interferon alfa to treat MM. This drug does not appear to be effective for inducing remission, and a randomized controlled trial showed that patients do not benefit from the addition of interferon to melphalan and prednisone.[116] Interferon alfa does appear to prolong remission in selected patients with MM. For this use, it may be administered after conventional chemotherapy or bone marrow (ie, stem cell) transplantation has been completed.

The toxicity of interferon and the availability of alternate interventions have significantly limited the role of interferon alfa.

Radiation Therapy

MM is extremely sensitive to radiation. Physicians use radiation to treat symptomatic lesions and to stabilize bones at risk for fracture. Physicians also use radiation to treat spinal cord compression. Low-dose, double-hemibody irradiation has been studied as systemic therapy for refractory or relapsed MM, but without dramatic success.

If the pain is mild and if less than 50% of the bone is involved, a course of irradiation can be initiated. Radiation treatment can result in additional early bone loss due to inflammation, and weight bearing should be limited for the first 4-6 weeks.

Therapy for Skeletal-Related Events

Bisphosphonates

Bisphosphonates are specific inhibitors of osteoclastic activity and are used to treat bone resorption. They also have a role in the secondary prevention of bony complications in MM, including hypercalcemia, pathologic fracture, and spinal cord compression. Intravenous (IV) pamidronate (Aredia) has been shown to be effective in preventing skeletal complications; zoledronic acid (Zometa) may be significantly more potent than pamidronate. A study by Morgan et al found that the early use of zoledronic acid was superior to clodronic acid in preventing skeletal-related events among patients with newly diagnosed MM, irrespective of bone disease status at baseline.[43]

A randomized placebo-controlled trial of pamidronate in subjects with MM who had experienced one skeletal event demonstrated that the medication reduced the likelihood of a second skeletal event from 41% to 24% after 9 months of therapy.[44] The investigators also noted improvements in pain, narcotic usage, and quality of life scores.

A 2007 systematic review of the use of bisphosphonates in MM confirmed a number-needed-to-treat (NNT) of 10 for the prevention of vertebral fractures, although no impact on mortality was seen.[45]

The American Society of Clinical Oncology (ASCO) issued a clinical practice guideline governing bisphosphonate therapy for MM patients who have lytic destruction of bone or compression fracture of the spine from osteopenia.[45] ASCO recommends IV pamidronate, 90 mg delivered over at least 2 hours, or zoledronic acid, 4 mg delivered over at least 15 minutes every 3-4 weeks. Because the risk for osteonecrosis of the jaw is 9.5-fold greater with zoledronic acid than with pamidronate, patients may prefer pamidronate.[45]

Zoledronic acid doses should be reduced in patients with preexisting mild to moderate renal impairment (estimated creatinine clearance, 30-60 mL/min); the drug is not recommended for use in patients with severe renal impairment.[45] All patients receiving pamidronate or zoledronic acid therapy should be screened every 3-6 months for albuminuria. If unexplained albuminuria (>500 mg/24 hours) is found, ASCO recommends discontinuation of the drug until the renal problems resolve.[45]

A study by Morgan et al revealed the anticancer properties of zoledronic acid in addition to its ability to reduce skeletal-related events in MM.[117]

Denosumab

In January 2018, denosumab was approved by the FDA for prevention of skeletal-related events (SREs) in patients with MM. It was originally indicated for SREs in patients with solid tumors. Denosumab is a human monoclonal antibody targeting and binding to receptor activator of nuclear factor kappa-Β ligand (RANKL). Osteoclast-activating factors, such as RANKL, are implicated in an increased risk for SREs with MM.

In a phase III trial of denosumab compared with zoledronic acid in patients (n=1718) with bone metastases, denosumab was noninferior and showed an advantage in significantly reducing the risk for renal adverse events. A post hoc analysis at 15 months was also conducted, since many of the skeletal-related events (60%) occurred early, within 3 months, which led the authors to speculate that the data reflected events occurring before the treatment had enough time to take effect. Results did show superiority of denosumab (n = 450) over zoledronic acid (n = 459) in terms of the endpoint of time to the first SRE (hazard ratio [HR], 0.66; P = 0.039). Median progression-free survival showed difference of more than 10 months was observed between the denosumab (46.09 months) and zoledronic acid (35.38 months) groups (HR, 0.82; P = 0.036). No difference in overall survival was noted between the treatment groups.[118]

Osteonecrosis of the jaw

Osteonecrosis of the jaw is a rare but severe adverse effect of bisphosphonate therapy. Level 1 evidence (ie, systematic reviews or randomized controlled trials) indicate that approximately 1% of cancer patients exposed to zolendronic acid develop osteonecrosis of the jaw.[119] Dental extractions appear to be a risk factor, and guidelines recommend avoiding this where possible.

A position paper by the American Association of Oral and Maxillofacial Surgeons describes the differential diagnosis, prevention, and treatment of medication-related osteonecrosis of the jaw. Consultation with an appropriate dental professional is advised before prescribing a bisphosphonate.[119]

Adjunctive Therapy for Complications

Potential complications of MM include the following:

Treatment for myeloma-induced hypercalcemia is the same as that for other malignancy-associated hypercalcemia; however, the dismal outcome observed with hypercalcemia in solid tumors is not observed in MM.

To treat pathologic fractures, physicians should orthopedically stabilize (ie, typically pin) and irradiate these lesions. Careful attention to a patient’s bony symptoms, intermittent radiographic surveys, and the use of bisphosphonates may be useful to prevent fractures.[45, 120, 121] (See Surgical Care and Bisphosphonate Therapy.)

Spinal cord compression is one of the most severe adverse effects of MM. The dysfunction may be reversible, depending on the duration of the cord compression; however, once established, the dysfunction is only rarely fully reversed. Patients who may have spinal cord compression need a rapid evaluation, which may necessitate urgent transfer to a center equipped with MRI for diagnosis or a center with a radiation oncologist for urgent therapy.

Patients with spinal cord compression due to MM should begin corticosteroid therapy immediately to reduce swelling. Urgent arrangements must be made for radiation therapy in order to restore or stabilize neurologic function. Surgery may be indicated. (See Surgical Care.)

Erythropoietin may ameliorate anemia resulting from either MM alone or from chemotherapy and has been shown to improve quality of life.[122] A systematic review failed to demonstrate a survival advantage for the use of erythropoietin agents in the treatment of patients with cancer-related anemia.[123]

Acute renal impairment related to MM is typically managed with plasmapheresis to rapidly lower circulating abnormal proteins. Data about this approach are limited, but a small randomized study showed a survival advantage with the use of apheresis.[12] Hydration (to maintain a urine output of >3 L/d), management of hypercalcemia, and avoidance of nephrotoxins (eg, intravenous contrast media, antibiotics) are also key factors. Conventional therapy may take weeks to months to show a benefit.

Renal impairment resulting from MM is associated with a very poor prognosis. A case series demonstrated that patients with renal failure from myeloma may benefit from autologous stem cell transplants, and as many as one third may demonstrate improvement in their renal function with this approach.[124] A report by Ludwig et suggests that bortezomib-based therapy may restore renal function in MM patients with renal failure.[11]

Guidelines on the management of multiple myeloma complications by the European Myeloma Network include the following recommendations[125] :

Surgical Care

Surgical therapy for MM is limited to adjunctive therapy. It consists of prophylactic fixation of pending fractures, decompression of the spinal cord when indicated, and treatment of pathologic fractures.

Prophylactic treatment of impending fractures and the treatment of pathologic fractures may involve bracing. In general, bracing is not effective for the long bones, though it may be effective for treating spinal involvement without neurologic compromise.

Intramedullary fixation is the procedure of choice when surgery is necessary. If the metaphysis or joint surface is involved, resection of the diseased bone and reconstruction with a total joint or, more typically, a hemiarthroplasty is indicated. Modular implants may be required. Severe destruction of the diaphysis may require reconstruction with combinations of methylmethacrylate, intramedullary nails, or resection and prosthetic replacement.

Although surgical decompression of the spinal cord is sometimes appropriate, posterior laminectomy in this population has been reported to have a mortality rate of 6-10% and to not be superior to radiation. This surgical approach is probably best reserved for cases of MM in which radiation fails. Newer surgical interventions, such as kyphoplasty, in which cement is injected into compressed vertebrae, have been shown to improve function with few complications, although the studies reported have been small.

Dietary Measures

Patients with MM who are receiving bisphosphonate therapy should include adequate calcium in their diet.

The dietary supplement curcumin may slow the progression of smoldering multiple myeloma.[126]

Physical Activity

Patients with MM should be encouraged to be physically active to the extent appropriate for their individual bone status. Physical activity may help maintain bone strength.

In general, patients with activity-related pain in either the femur or the tibia should be given a walker or crutches until a radiographic workup has been completed. Radiation therapy elicits an inflammatory response, and for the first 6 weeks or so, bony resorption may actually weaken the target bone. Given that prophylactic treatment of an impending fracture is usually easier than reconstruction of a pathologic fracture, one should have a low threshold for initiating protected weight bearing.

Prevention of Multiple Myeloma

No preventive measures for MM are known. A study by Chang et al found that routine residential ultraviolet radiation exposure may have a protective effect against lymphomagenesis through mechanisms that may be independent of vitamin D.[127]

Consultations

Patients with MM often benefit from the expertise of an orthopedic surgeon who is versed in oncologic management because prophylactic fixation of impending pathologic fractures is occasionally warranted.

From the orthopedic perspective, because patients with MM have significant systemic comorbidities—including potentially severe hematologic, infectious, and metabolic diseases—the orthopedic surgeon treating the skeletal disease in a patient with myeloma should work in conjunction with the radiation oncologists and the medical oncologists.

Long-Term Monitoring

Patients with MM may require hospitalization for the treatment of pain or bony pathology.

Patients with MM are at high risk of infection, especially from encapsulated organisms. Vaccinations against pneumococcal organisms and influenza are recommended. Consider vaccinating patients against Haemophilus influenzae type b. Use of the herpes zoster vaccine should be considered.

The following laboratory results are helpful in the follow-up care of patients with MM:

Guidelines Summary

Diagnosis

Similar recommendations for the standard investigational workup for suspected myeloma have been issued by the following organizations:

The International Myeloma Working Group guidelines recommend the following diagnostic studies[128] :

The NCCN guidelines recommend the following diagnostic studies[2] :

The ESMO guidelines recommend basing the diagnosis of multiple myeloma on the following[129]

Diagnostic criteria

Multiple myeloma is defined as smoldering (asymptomatic) or active (symptomatic). The NCCN criteria for smoldering multiple myeloma are as follows[2] :

The NCCN also recommends that a patient whose bone survey is negative be assessed for bone disease with whole-body skeletal MRI, FDG PET/CT, of low-dose CT to differentiate active from smoldering MM.[2]

In the NCCN guidelines, active multiple myeloma is no longer diagnosed using the CRAB criteria (hyperCalcemia, Renal failure, Anemia, Bone lesions) for end-organ damage. The current diagnostic criteria for symptomatic multiple myeloma are as follows[2] :

Myeloma-defining events include the following [2] :

In November 2014, the International Myeloma Working Group added the following criteria to the CRAB criteria for multiple myeloma[1] :

The International Working Group noted that these findings have been “associated with near inevitable development of CRAB features in patients who would otherwise be regarded as having smouldering multiple myeloma.”[128]  The presence of any of the CRAB criteria or any of these three additional criteria justifies therapy.

Staging

In 2015, the  International Myeloma Working Group published the Revised International Staging System for Multiple Myeloma.[130] The revised system was subsequently recommended by the NCCN.[2]  The Revised International Staging System (R-ISS) was created by combining the International Staging System (ISS) with chromosomal abnormalities (CA) detected by interphase FISH (iFISH) after CD138 plasma cell purification, plus serum LDH assay results .[130]

Like the ISS, the R-ISS is based on three stages. Stage I criteria are as follows:

Stage II comprises patients who do not meet criteria for stage I or stage III

Stage III consists of the following:

Median progression-free survival is as follows:

Treatment

National Comprehensive Cancer Network (NCCN) general treatment recommendations for multiple myeloma include the following[2] :

For primary induction therapy in patients with MM who are candidates for transplantation, NCCN guidelines recommend the following combinations as preferred regimens[2] :

Other recommended regimens, according to the NCCN, are as follows:

The NCCN considers the following regimens useful in certain circumstances (although triplet regimens should be used as the standard, patients not considered candidates for a 3-drug regimen can be started on a 2-drug regimen, with the third drug added once performance status improves):

For primary induction therapy in patients who are not transplant candidates, the NCCN guidelines list the following as preferred regimens[2] :

Other NCCN-recommended regimens for these cases include the following:

For maintenance therapy, the NCCN recommends lenalidomide. Although lenalidomide is a category 1 recommendation, the NCCN notes that lenalidomide maintenance appears to be associated with increased risk of secondary cancers, especially after transplant; this should be discussed with patients. Other recommended agents are ixazomib (category 1) and bortezomib; bortezomib/lenalidomide is useful in certain circumstances.

For salvage therapy, the regimen used for primary induction can be repeated if relapse occurs after more than 6 months. Otherwise, category 1 preferred regimens include the following[2] :

Management of Multiple Myeloma–related Bone Disease

In May 2013, the International Myeloma Working Group (IMWG) released practice guidelines for the management of multiple myeloma–related bone disease.[46]  The recommendations, which were based on a review of the literature through August 2012 and a consensus of an interdisciplinary panel of experts, include the following:

Bisphosphonate Therapy

In 2007, the American Society of Clinical Oncology (ASCO) issued an update to their clinical practice guideline governing bisphosphonate therapy for multiple myeloma patients who have lytic destruction of bone or compression fracture of the spine from osteopenia. ASCO recommends IV pamidronate, 90 mg delivered over at least 2 hours, or zoledronic acid, 4 mg delivered over at least 15 minutes every 3-4 weeks. Because the risk for osteonecrosis of the jaw is 9.5-fold greater with zoledronic acid than with pamidronate, patients may prefer pamidronate.[45]

Zoledronic acid doses should be reduced in patients with preexisting mild to moderate renal impairment (estimated creatinine clearance, 30-60 mL/min); the drug is not recommended for use in patients with severe renal impairment. All patients receiving pamidronate or zoledronic acid therapy should be screened every 3-6 months for albuminuria. If unexplained albuminuria (>500 mg/24 hours) is found, ASCO recommends discontinuation of the drug until the renal problems resolve.[45]

Management of Complications

According to National Comprehensive Cancer Network (NCCN) guidelines, novel drugs such as bortezomib can be used with dexamethasone as primary treatment for multiple myeloma–associated amyloidosis. The combination of cyclophosphamide, thalidomide, and dexamethasone is also recommended for the primary treatment of amyloidosis.[2]

The NCCN, American Society of Clinical Oncology (ASCO), and International Myeloma Workshop clinical guidelines for prevention of venous thromboembolism agree that patients with multiple myeloma who are receiving thalidomide- or lenalidomide-based regimens with chemotherapy and/or dexamethasone should receive prophylactic anticoagulation therapy with either aspirin or low molecular weight heparin (LMWH) for lower-risk patients and LMWH for higher-risk patients.[131, 132, 133]

A joint American Society of Hematology (ASH) and ASCO clinical practice guideline on the use of erythropoiesis-stimulating agents (ESAs) in cancer was updated in 2010. The specific recommendations for patients with multiple myeloma receiving concurrent chemotherapy include the following[131] :

Guidelines on the management of multiple myeloma complications by the European Myeloma Network include the following recommendations[125] :

Medication Summary

Multiple myeloma (MM) is treated with several categories of medications. Chemotherapeutic agents are used to reduce the disease burden, and bisphosphonates are used to promote bone healing and to provide secondary prophylaxis against skeletal-related events (eg, hypercalcemia, bone fracture, spinal cord compression, need for radiation, and need for surgery). In addition, erythropoietin is used to treat anemia, either alone or in conjunction with chemotherapy.

Cyclophosphamide (Cytoxan, Neosar)

Clinical Context:  Cyclophosphamide is chemically related to nitrogen mustards. It is an alkylating agent, and its mechanism of action of active metabolites may involve cross-linking of DNA, which may interfere with growth of normal and neoplastic cells.

Melphalan (Alkeran)

Clinical Context:  The most widely used regimen is MP. Melphalan is an alkylating agent and a derivative of mechlorethamine that inhibits mitosis by cross-linking DNA strands. It is indicated for the palliative treatment of multiple myeloma.

Doxorubicin (Adriamycin, Rubex)

Clinical Context:  Doxorubicin is part of VAD therapy. It inhibits topoisomerase II and produces free radicals, which may cause destruction of DNA; these 2 events, in turn, can inhibit growth of neoplastic cells.

Doxorubicin liposomal (Doxil)

Clinical Context:  Doxorubicin liposomal is a pegylated formulation that protects the liposomes and, thereby, increases blood circulation time. The drug inhibits topoisomerase II and produces free radicals, which may cause destruction of DNA; these 2 events can, in turn, inhibit growth of neoplastic cells.

Vincristine (Oncovin)

Clinical Context:  Vincristine inhibits cellular mitosis by inhibition of intracellular tubulin function, binding to microtubules, and synthesis of spindle proteins in the S phase. Vincristine is part of VAD therapy. Its mechanism of action is complex and includes depolymerization of microtubules.

Bortezomib (Velcade)

Clinical Context:  Bortezomib is the first drug approved in the group of anticancer agents known as proteasome inhibitors. The proteasome pathway is an enzyme complex existing in all cells, which degrades ubiquitinated proteins that control the cell cycle and cellular processes and maintains cellular homeostasis. Reversible proteasome inhibition disrupts pathways supporting cell growth, thus decreasing cancer cell survival. Bortezomib is indicated for patients with multiple myeloma. Development of peripheral neuropathy is a limiting factor. A decreased incidence of peripheral neuropathy has been observed with SC administration compared with the IV route.

Carfilzomib (Kyprolis)

Clinical Context:  Proteasome inhibitor; elicits antiproliferative and proapoptotic activities in vitro in solid and hematologic tumor cells. It is indicated as monotherapy, in combination with dexamethasone, or in combination with lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma in patients who have received at least 1 prior line of therapy.  

Ixazomib (Ninlaro)

Clinical Context:  Reversible proteasome inhibitor. It preferentially binds and inhibits the chymotrypsinlike activity of the beta 5 subunit of the 20S proteasome. It is indicated in combination with lenalidomide and dexamethasone for patients with multiple myeloma who have received at least 1 prior therapy.

Panobinostat (Farydak)

Clinical Context:  Panobinostat is a histone deacetylase (HDAc) inhibitor. HDAc catalyzes the removal of acetyl groups from the lysine residues of histones and some nonhistone proteins. Inhibition of HDAc activity results in increased acetylation of histone proteins and an epigenetic alteration that results in a relaxing of chromatin, leading to transcriptional activation. It is indicated in combination with bortezomib and dexamethasone for treatment of multiple myeloma in patients who have received at least 2 prior regimens, including bortezomib and an immunomodulatory agent.

Class Summary

The choice of chemotherapy depends on several factors, including the patient’s performance status, age, renal function, desire for inpatient or outpatient therapy, and likelihood of receiving future autologous stem cell transplantation.

In patients with renal failure or highly aggressive disease, therapy with vincristine, Adriamycin (doxorubicin), and dexamethasone (VAD) may be preferred. In elderly patients or patients in whom autologous transplantation is not possible in the future, melphalan and prednisone (MP) therapy is preferred because of its ease of administration and low toxicity. 

Prednisone (Deltasone, Orasone, Meticorten)

Clinical Context:  The most widely used regimen is MP. Prednisone stabilizes lysosomal membranes and suppresses lymphocyte and antibody production.

Dexamethasone (Decadron)

Clinical Context:  Dexamethasone is part of many treatment regimens for multiple myeloma. Dexamethasone stabilizes lysosomal membranes and suppresses lymphocyte and antibody production.

Class Summary

Corticosteroids have anti-inflammatory properties and cause profound and varied metabolic effects. They modify the body’s immune response to diverse stimuli. 

Denosumab (Xgeva)

Clinical Context:  Monoclonal antibody that specifically targets RANKL. It binds to RANKL and inhibits its binding to RANK receptor, thereby preventing osteoclast formation. This results in decreased bone resorption and increases bone mass in osteoporosis. RANKL inhibition decreases tumor-induced bone destruction and SREs. Denosumab is indicated for prevention of SREs in patients with multiple myeloma.

Daratumumab (Darzalex)

Clinical Context:  Monoclonal antibody that binds with high affinity to the CD38 molecule, which is highly expressed on the surface of multiple myeloma cells. It is indicated for patients with multiple myeloma who have received at least 3 prior treatments, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), or who are double-refractory to a PI and IMiD. Other regimens for relapsed/refractory MM are approved for daratumumab in combination with dexamethasone plus bortezomib or lenalidomide or pomalidomide. It is also indicated for newly diagnosed MM in patients ineligible for ASCT as part of various combination regimens. May also be consider in newly diagnosed MM in patients who are eligible for ASCT in combination with bortezomib, thalidomide, and dexamethasone.

Elotuzumab (Empliciti)

Clinical Context:  Humanized IgG1 monoclonal antibody that specifically targets the SLAMF7 (signaling lymphocytic activation molecule family member 7) protein. SLAMF7 is expressed on myeloma cells and natural killer cells and plasma cells. Facilitates the interaction with natural killer cells to mediate the killing of myeloma cells through antibody-dependent cellular cytotoxicity. It is indicated in combination with lenalidomide and dexamethasone for multiple myeloma in patients who have received 1-3 prior therapies. Elotuzumab is also indicated in combination with pomalidomide and dexamethasone for patients with multiple myeloma who have received 2 or more prior therapies including lenalidomide and a proteasome inhibitor.

Isatuximab (Sarclisa, isatuximab-irfc)

Clinical Context:  Anti-CD38 monoclonal antibody indicated for relapsed or resistant multiple myeloma in combination with pomalidomide and dexamethasone in patients who have received at least 2 prior therapies, including lenalidomide and a proteasome inhibitor.

Class Summary

Monoclonal antibodies that target proteins expressed on multiple myeloma cells (eg, CD38, SLAMF7) have been approved.

Interferon alfa-2B (Intron A)

Clinical Context:  Interferon alfa-2B is a protein product manufactured by recombinant DNA technology. The mechanism of antitumor activity is not clearly understood; however, direct antiproliferative effects against malignant cells and modulation of host immune response may play important roles.

Class Summary

Interferons are naturally produced proteins with antiviral, antitumor, and immunomodulatory actions. Alfa-, beta-, and gamma-interferons may be administered topically, systemically, and intralesionally. 

Thalidomide (Thalomid)

Clinical Context:  Thalidomide, when used in combination with dexamethasone, is indicated for the treatment of patients with newly diagnosed multiple myeloma. Thalidomide is an immunomodulatory agent that may suppress excessive production of tumor necrosis factor (TNF)-alpha and may down-regulate selected cell-surface adhesion molecules involved in leukocyte migration. Because of concerns regarding teratogenicity, thalidomide can only be prescribed by registered physicians and is dispensed by registered pharmacists. Patients must participate in ongoing surveys to receive therapy, and only a 28-d supply can be prescribed at a time.

Lenalidomide (Revlimid)

Clinical Context:  Lenalidomide is indicated in combination with dexamethasone for multiple myeloma. It is structurally similar to thalidomide. Lenalidomide elicits immunomodulatory and antiangiogenic properties. It inhibits proinflammatory cytokine secretion and increases anti-inflammatory cytokines from peripheral blood mononuclear cells.

Pomalidomide (Pomalyst)

Clinical Context:  Thalidomide analogue indicated in combination with dexamethasone for patients with multiple myeloma who have received at least 2 prior therapies including lenalidomide and a proteasome inhibitor. Also used in combination with elotuzumab and dexamethasone.

Class Summary

Immunosuppressant agents inhibit key factors in the immune system that are responsible for immune reactions.

Selinexor (Xpovio)

Clinical Context:  Indicated in combination with dexamethasone for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least 4 prior therapies and whose disease is refractory to at least 2 proteasome inhibitors, at least 2 immunomodulatory agents, and an anti-CD38 monoclonal antibody.

Class Summary

The first selective inhibitor of nuclear export (SINE) was approved by the FDA in July 2019. These agents act on tumor suppressor proteins (TSPs), growth regulators, and mRNAs of oncogenic proteins by blocking exportin 1 (XPO1). Inhibition of XPO1 leads to accumulation of TSPs in the nucleus, reductions in several oncoproteins (eg, c‐myc, cyclin D1), cell cycle arrest, and apoptosis of cancer cells.

Pamidronate (Aredia)

Clinical Context:  Pamidronate inhibits normal and abnormal bone resorption. It appears to inhibit bone resorption without inhibiting bone formation and mineralization. The optimal timing and duration of therapy are being studied. Pamidronate is administered intravenously (IV) over 2 hours. Newer drugs similar in structure and function are being studied and may have improved efficacy and greater convenience.

Zoledronic acid (Zometa)

Clinical Context:  Zoledronic acid inhibits bone resorption, possibly by acting on osteoclasts or osteoclast precursors. It is effective in treating the hypercalcemia of malignancy.

Class Summary

Bisphosphonates inhibit bone resorption via action on osteoclasts or osteoclast precursors.

Epoetin alfa, erythropoietin (Epogen, Procrit)

Clinical Context:  Erythropoietin stimulates the division and differentiation of committed erythroid progenitor cells and induces the release of reticulocytes from bone marrow into the blood stream.

Erythropoietin is a naturally occurring hormone produced by the kidneys to stimulate bone marrow production of red blood cells. In patients with MM, administration of exogenous erythropoietin may correct anemia, leading to a significant improvement in performance status and quality of life.

Class Summary

Colony-stimulating factors induce erythropoiesis.

What is multiple myeloma (MM)?What are the signs and symptoms of multiple myeloma (MM)?How is multiple myeloma (MM) often discovered?Which physical findings are characteristic of multiple myeloma (MM)?What are the physical findings of multiple myeloma (MM) with amyloidosis?What are the International Myeloma Workshop guidelines for the workup of suspected multiple myeloma (MM)?Which routine lab tests are performed in the workup of multiple myeloma (MM)?Which imaging studies are performed in the workup of multiple myeloma (MM)?What are the treatment options for multiple myeloma (MM)?Which chemotherapy regimens are used in the treatment of multiple myeloma (MM)?What is the NCCN recommended primary induction therapy for patients with multiple myeloma (MM) who are candidates for transplantation?What are the NCCN recommended chemotherapy regimens for the treatment of multiple myeloma (MM)?What are the NCCN preferred regimens for treatment of relapsed multiple myeloma (MM)?What is the pathophysiology of multiple myeloma (MM)?What is the role of skeletal processes in the pathophysiology of multiple myeloma (MM)?What is the role of hematologic processes in the pathophysiology of multiple myeloma (MM)?What is the role of renal processes in the pathophysiology of multiple myeloma (MM)?What is the role of neurologic processes in the pathophysiology of multiple myeloma (MM)?What is the role of hyperviscosity syndrome in the pathophysiology of multiple myeloma (MM)?What causes multiple myeloma (MM)?What is the role of genetics in the etiology of multiple myeloma (MM)?What are environmental or occupational risk factors for multiple myeloma (MM)?How is monoclonal gammopathy of undetermined significance (MGUS) defined?What are the types of monoclonal gammopathy of undetermined significance (MGUS)?What are risk factors for progression of monoclonal gammopathy of undetermined significance (MGUS)?What is smoldering multiple myeloma (MM)?What is the role of radiation in the etiology of multiple myeloma (MM)?What is the role of chronic inflammation in the etiology of multiple myeloma (MM)?What is the role of infection in the etiology of multiple myeloma (MM)?What is the prevalence of multiple myeloma (MM)?What is the prognosis of multiple myeloma (MM)?What are poor prognostic factors for multiple myeloma (MM)?What are the survival rates for multiple myeloma (MM)?How do infections affect the prognosis of multiple myeloma (MM)?What should the patient education for multiple myeloma (MM) address?What are the signs and symptoms of multiple myeloma (MM)?How prevalent is bone pain in multiple myeloma (MM)?What is the prevalence of pathologic fractures in multiple myeloma (MM)?What are the symptoms of spinal cord compression in multiple myeloma (MM)?What causes bleeding in multiple myeloma (MM)?What are the symptoms of hypercalcemia in multiple myeloma (MM)?Which infections are common in patients with multiple myeloma (MM)?What are the symptoms of hyperviscosity multiple myeloma (MM)?What are neurologic symptoms in multiple myeloma (MM)?What causes weakness in multiple myeloma (MM)?Which physical findings are characteristic of multiple myeloma (MM)?Which physical findings suggest amyloidosis in multiple myeloma (MM)?What is the shoulder pad sign in multiple myeloma (MM)?What does a finding of macroglossia suggest in the evaluation of multiple myeloma (MM)?How are skin lesions characterized in multiple myeloma (MM)?What does a finding of post-proctoscopic peripalpebral purpura suggest in the evaluation for multiple myeloma (MM)?What is the prevalence of renal failure in multiple myeloma (MM)?What are possible hematologic complications of multiple myeloma (MM)?What are the skeletal complications of multiple myeloma (MM)?What are potential complications in multiple myeloma (MM)?How is multiple myeloma (MM) diagnosed?What are the diagnostic criteria for active (symptomatic) multiple myeloma (MM)?What are-defining events in multiple myeloma?How is active multiple myeloma (MM) differentiated from smoldering multiple myeloma (MM)?What are the differential diagnoses for Multiple Myeloma?What are the guidelines for the standard workup for suspected multiple myeloma (MM)?Which blood studies are performed in the workup of multiple myeloma (MM)?What is the role of urine testing in the workup of multiple myeloma (MM)?What is the role of electrophoresis and immunofixation in the workup of multiple myeloma (MM)?What is the role of quantitative immunoglobulin (IgG, IgA, IgM) testing in the workup of multiple myeloma (MM)?What is the role of beta-2 microglobulin and C-reactive protein (CRP) testing in the workup of multiple myeloma (MM)?What is the role of serum viscosity testing in the workup of multiple myeloma (MM)?What is the role of radiography in the workup of multiple myeloma (MM)?What is the role of MRI in the workup of multiple myeloma (MM)?What is the role of positron emission tomography (PET) scanning in the workup of multiple myeloma (MM)?What is the role of bone scanning in the workup of multiple myeloma (MM)?What is the role of bone marrow aspiration and biopsy in the workup of multiple myeloma (MM)?Which histologic findings are characteristic of multiple myeloma (MM)?What is the role of cytogenetic analysis of the bone marrow in the evaluation of multiple myeloma (MM)?Which systems are used to stage multiple myeloma (MM)?What is the Salmon-Durie classification of multiple myeloma (MM)?What is the international staging system for multiple myeloma (MM)?What is the revised international staging system for multiple myeloma (MM)?What is the approach to treatment of multiple myeloma (MM)?When should treatment for multiple myeloma (MM) be initiated?Which genetic mutations are indicators of high risk multiple myeloma (MM)?What are treatment options for multiple myeloma (MM)?What are the NCCN preferred regimens for primary induction therapy in patients with multiple myeloma (MM)?Which regimens does NCCN consider useful in select circumstances of multiple myeloma (MM)?What is the role of lenalidomide in the treatment of multiple myeloma (MM)?What is included in adjunctive therapy for multiple myeloma (MM)?How are skeletal related events (SREs) prevented in patients with multiple myeloma (MM) receiving first-line therapy?When are chemotherapy and immunotherapy indicated in the treatment of multiple myeloma (MM)?Which patient groups are candidates for autologous stem cell transplant to treat multiple myeloma (MM)?What is the role of VAD (vincristine, doxorubicin [Adriamycin], and dexamethasone) in the treatment of multiple myeloma (MM)?What is the role of thalidomide in the treatment of multiple myeloma (MM)?What is the role of combined thalidomide and lenalidomide (Revlimid) in the treatment of multiple myeloma (MM)?What is the efficacy of lenalidomide for the treatment of multiple myeloma (MM)?What are risks associated with lenalidomide for the treatment of multiple myeloma (MM)?What is the role of bortezomib for the treatment of multiple myeloma (MM)?What is the efficacy of combination therapy with bortezomib for the treatment of multiple myeloma (MM)?What is the efficacy of bortezomib for the treatment of multiple myeloma (MM)?What is the role of daratumumab in the treatment of multiple myeloma (MM)?Which high-risk patients with multiple myeloma (MM) are potential transplant candidates?What is the standard treatment for newly diagnosed patients with multiple myeloma (MM) who are not transplant candidates?What is the efficacy of melphalan and prednisone (MP) therapies for the treatment of multiple myeloma (MM)?What is the role of maintenance therapy in the treatment of multiple myeloma (MM)?What is the efficacy of maintenance therapy in the treatment of multiple myeloma (MM)?What are preferred regimens for treatment of multiple myeloma (MM) relapse?What is the efficacy of bortezomib in the treatment of refractory multiple myeloma (MM)?Which medications are FDA approved for treatment of refractory or relapsed multiple myeloma (MM)?What is the role of Daratumumab (Darzalex), Ixazomib (Ninlaro), and Elotuzumab (Empliciti) in the treatment of multiple myeloma (MM)?What is the efficacy of second-line therapies for multiple myeloma (MM)?What is the role of autologous stem cell transplantation in the treatment of multiple myeloma (MM)?What is the efficacy of tandem autologous stem cell transplantation in the treatment of multiple myeloma (MM)?What is the role of allogeneic stem cell transplantation in the treatment of multiple myeloma (MM)?What are some limitations of allogeneic stem cell transplantation less for the treatment of multiple myeloma (MM)?What is the role of nonmyeloablative transplantation regimens in the treatment of multiple myeloma (MM)?What is the role of bortezomib-dexamethasone induction therapy in the treatment of multiple myeloma (MM)?What are the treatment options for multiple myeloma (MM) in patients with progressive or relapsing disease following autologous stem-cell transplantation?What is the role of interferon alfa in the treatment of multiple myeloma (MM)?What is the role of radiation therapy in the treatment of multiple myeloma (MM)?What is the role of bisphosphonates in the treatment of multiple myeloma (MM)?What medication is FDA approved for prevention of skeletal-related events (SREs) in multiple myeloma (MM)?What is a possible severe adverse effect of bisphosphonate therapy for multiple myeloma (MM)?What are possible complications of multiple myeloma (MM)?How is hypercalcemia treated in patients with multiple myeloma (MM)?What are the guidelines on the European Myeloma Network management of multiple myeloma (MM) complications?What is the role of surgery in the treatment of multiple myeloma (MM)?What dietary modifications are used in the treatment of multiple myeloma (MM)?What physical activity modifications are needed in the treatment of multiple myeloma (MM)?How is multiple myeloma (MM) prevented?Which specialist consultations are needed for the treatment of multiple myeloma (MM)?What is included in the long-term monitoring of multiple myeloma (MM)?Which organizations have issued guidelines for the workup of multiple myeloma (MM)?What are the International Myeloma Working Group (IMWG) guidelines for the workup of multiple myeloma (MM)?What are the National Comprehensive Cancer Network (NCCN) guidelines for the workup of multiple myeloma (MM)?What are the European Society for Medical Oncology (ESMO) guidelines for the diagnosis evaluation of multiple myeloma (MM)?What are the National Comprehensive Cancer Network (NCCN) criteria for a diagnosis of smoldering multiple myeloma (MM)?What are the National Comprehensive Cancer Network (NCCN) criteria for a diagnosis of active multiple myeloma (MM)?What are considered myeloma defining events in the diagnosis of multiple myeloma (MM)?What is the Revised International Staging System (R-ISS) and how is it used in the diagnosis of multiple myeloma (MM)?What are the National Comprehensive Cancer Network (NCCN) general treatment recommendations for multiple myeloma (MM)?What are the NCCN guidelines for primary induction therapy in patients with multiple myeloma (MM) who are candidates for transplantation?Which regimens does the National Comprehensive Cancer Network (NCCN) consider useful in elderly or frail patients with multiple myeloma (MM)?What are the NCCN guidelines for primary induction therapy in patients with multiple myeloma (MM) who are not candidates for transplantation?What does the NCCN recommend for maintenance therapy in patients with multiple myeloma (MM)?What does the NCCN recommend for salvage therapy in patients with multiple myeloma (MM)?What are the International Myeloma Working Group (IMWG) practice treatment guidelines for bone disease associated with multiple myeloma (MM)?What are the American Society of Clinical Oncology (ASCO) clinical practice guidelines governing bisphosphonate therapy for multiple myeloma (MM) patients?Which novel drugs can be used as primary treatment for amyloidosis associated with multiple myeloma (MM)?What are the American Society of Hematology (ASH) and ASCO recommendations for use of erythropoiesis-stimulating agents (ESAs) in multiple myeloma (MM)?What are the European Myeloma Network treatment guidelines for multiple myeloma (MM) complications?What type of medications are used in the treatment of multiple myeloma (MM)?Which medications in the drug class Bisphosphonates are used in the treatment of Multiple Myeloma?Which medications in the drug class Selective Inhibitor of Nuclear Export are used in the treatment of Multiple Myeloma?Which medications in the drug class Immunosuppressant Agents are used in the treatment of Multiple Myeloma?Which medications in the drug class Interferons are used in the treatment of Multiple Myeloma?Which medications in the drug class Monoclonal Antibodies are used in the treatment of Multiple Myeloma?Which medications in the drug class Corticosteroids are used in the treatment of Multiple Myeloma?Which medications in the drug class Chemotherapeutic Agents are used in the treatment of Multiple Myeloma?Which medications in the drug class Colony-stimulating Factors are used in the treatment of Multiple Myeloma?

Author

Dhaval Shah, MD, Helen F Graham Cancer Center and Research Institute

Disclosure: Nothing to disclose.

Coauthor(s)

Karen Seiter, MD, Professor, Department of Internal Medicine, Division of Oncology/Hematology, New York Medical College

Disclosure: Received honoraria from Novartis for speaking and teaching; Received consulting fee from Novartis for speaking and teaching; Received honoraria from Celgene for speaking and teaching.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Emmanuel C Besa, MD, Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Disclosure: Nothing to disclose.

References

  1. Dimopoulos M, Kyle R, Fermand JP, Rajkumar SV, San Miguel J, et al. Consensus recommendations for standard investigative workup: report of the International Myeloma Workshop Consensus Panel 3. Blood. 2011 May 5. 117 (18):4701-5. [View Abstract]
  2. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology, Multiple Myeloma. Available at http://www.nccn.org/professionals/physician_gls/pdf/myeloma.pdf. Version 2.2020 — October 9, 2019; Accessed: March 4, 2020.
  3. Rajkumar SV. Myeloma today: Disease definitions and treatment advances. Am J Hematol. 2016 Jan. 91 (1):90-100. [View Abstract]
  4. Palumbo A, Rajkumar SV. Treatment of newly diagnosed myeloma. Leukemia. 2009 Mar. 23(3):449-56. [View Abstract]
  5. Bensinger WI. Role of autologous and allogeneic stem cell transplantation in myeloma. Leukemia. 2009 Mar. 23(3):442-8. [View Abstract]
  6. Attal M, Richardson PG, Rajkumar SV, San-Miguel J, Beksac M, Spicka I, et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet. 2019 Dec 7. 394 (10214):2096-2107. [View Abstract]
  7. Adding Isatuximab to Pomalidomide and Low-Dose Dexamethasone Improves PFS in Relapsed and Refractory Multiple Myeloma. ESMO. Available at https://www.esmo.org/oncology-news/adding-isatuximab-to-pomalidomide-and-low-dose-dexamethasone-improves-pfs-in-relapsed-and-refractory-multiple-myeloma. November 20, 2019; Accessed: March 5, 2020.
  8. van Nieuwenhuijzen N, Spaan I, Raymakers R, Peperzak V. From MGUS to Multiple Myeloma, a Paradigm for Clonal Evolution of Premalignant Cells. Cancer Res. 2018 May 15. 78 (10):2449-2456. [View Abstract]
  9. Kuehl WM, Bergsagel PL. Molecular pathogenesis of multiple myeloma and its premalignant precursor. J Clin Invest. 2012 Oct. 122 (10):3456-63. [View Abstract]
  10. Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma. Lancet. 2009 Jul 25. 374(9686):324-39. [View Abstract]
  11. Ludwig H, Drach J, Graf H, Lang A, Meran JG. Reversal of acute renal failure by bortezomib-based chemotherapy in patients with multiple myeloma. Haematologica. 2007 Oct. 92(10):1411-4. [View Abstract]
  12. Zucchelli P, Pasquali S, Cagnoli L, Ferrari G. Controlled plasma exchange trial in acute renal failure due to multiple myeloma. Kidney Int. 1988 Jun. 33(6):1175-80. [View Abstract]
  13. Key Statistics About Multiple Myeloma. American Cancer Society. Available at http://www.cancer.org/cancer/multiplemyeloma/detailedguide/multiple-myeloma-key-statistics. January 8, 2020; Accessed: March 4, 2020.
  14. Koutros S, Baris D, Bell E, Zheng T, Zhang Y, Holford TR, et al. Use of hair colouring products and risk of multiple myeloma among US women. Occup Environ Med. 2009 Jan. 66 (1):68-70. [View Abstract]
  15. Wadhera RK, Kyle RA, Larson DR, et al. Incidence, clinical course, and prognosis of secondary monoclonal gammopathy of undetermined significance in patients with multiple myeloma. Blood. 2011 Sep 15. 118(11):2985-7. [View Abstract]
  16. Yiin JH, Anderson JL, Daniels RD, Seel EA, Fleming DA, Waters KM, et al. A nested case-control study of multiple myeloma risk and uranium exposure among workers at the Oak Ridge Gaseous Diffusion Plant. Radiat Res. 2009 Jun. 171(6):637-45. [View Abstract]
  17. Caers J, Vande broek I, De Raeve H, Michaux L, Trullemans F, Schots R, et al. Multiple myeloma--an update on diagnosis and treatment. Eur J Haematol. 2008 Nov. 81(5):329-43. [View Abstract]
  18. Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia. 2009 Jan. 23(1):3-9. [View Abstract]
  19. Surveillance, Epidemiology, and End Results Program. SEER Stat Fact Sheets: Myeloma. National Cancer Institute. Available at http://seer.cancer.gov/statfacts/html/mulmy.html. Accessed: July 12, 2019.
  20. Ailawadhi S, Aldoss IT, Yang D, Razavi P, Cozen W, Sher T, et al. Outcome disparities in multiple myeloma: a SEER-based comparative analysis of ethnic subgroups. Br J Haematol. 2012 Apr 26. [View Abstract]
  21. Ludwig H, Durie BG, Bolejack V, Turesson I, Kyle RA, Blade J, et al. Myeloma in patients younger than age 50 years presents with more favorable features and shows better survival: an analysis of 10 549 patients from the International Myeloma Working Group. Blood. 2008 Apr 15. 111(8):4039-47. [View Abstract]
  22. Bataille R, Boccadoro M, Klein B, Durie B, Pileri A. C-reactive protein and beta-2 microglobulin produce a simple and powerful myeloma staging system. Blood. 1992 Aug 1. 80(3):733-7. [View Abstract]
  23. Augustson BM, Begum G, Dunn JA, Barth NJ, Davies F, Morgan G, et al. Early mortality after diagnosis of multiple myeloma: analysis of patients entered onto the United kingdom Medical Research Council trials between 1980 and 2002--Medical Research Council Adult Leukaemia Working Party. J Clin Oncol. 2005 Dec 20. 23(36):9219-26. [View Abstract]
  24. Blimark C, Holmberg E, Mellqvist UH, Landgren O, Björkholm M, Hultcrantz M, et al. Multiple myeloma and infections: a population-based study on 9253 multiple myeloma patients. Haematologica. 2015 Jan. 100(1):107-13. [View Abstract]
  25. Bladé J, Fernández-Llama P, Bosch F, Montolíu J, Lens XM, Montoto S, et al. Renal failure in multiple myeloma: presenting features and predictors of outcome in 94 patients from a single institution. Arch Intern Med. 1998 Sep 28. 158(17):1889-93. [View Abstract]
  26. Dimopoulos M, Terpos E, Comenzo RL, Tosi P, Beksac M, Sezer O, et al. International myeloma working group consensus statement and guidelines regarding the current role of imaging techniques in the diagnosis and monitoring of multiple Myeloma. Leukemia. 2009 Sep. 23(9):1545-56. [View Abstract]
  27. Hung GU, Tsai CC, Tsai SC, Lin WY. Comparison of Tc-99m sestamibi and F-18 FDG-PET in the assessment of multiple myeloma. Anticancer Res. 2005 Nov-Dec. 25(6C):4737-41. [View Abstract]
  28. Shortt CP, Gleeson TG, Breen KA, McHugh J, O'Connell MJ, O'Gorman PJ, et al. Whole-Body MRI versus PET in assessment of multiple myeloma disease activity. AJR Am J Roentgenol. 2009 Apr. 192(4):980-6. [View Abstract]
  29. Zamagni E, Patriarca F, Nanni C, et al. Prognostic relevance of 18-F FDG PET/CT in newly diagnosed multiple myeloma patients treated with up-front autologous transplantation. Blood. 2011 Dec 1. 118(23):5989-95. [View Abstract]
  30. Durie BG, Salmon SE. A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer. 1975 Sep. 36(3):842-54. [View Abstract]
  31. Greipp PR, San Miguel J, Durie BG, Crowley JJ, Barlogie B, Bladé J, et al. International staging system for multiple myeloma. J Clin Oncol. 2005 May 20. 23(15):3412-20. [View Abstract]
  32. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J Clin Oncol. 2015 Sep 10. 33 (26):2863-9. [View Abstract]
  33. Dimopoulos MA, Moulopoulos A, Smith T, Delasalle KB, Alexanian R. Risk of disease progression in asymptomatic multiple myeloma. Am J Med. 1993 Jan. 94(1):57-61. [View Abstract]
  34. He Y, Wheatley K, Clark O, Glasmacher A, Ross H, Djulbegovic B. Early versus deferred treatment for early stage multiple myeloma. Cochrane Database Syst Rev. 2003. CD004023. [View Abstract]
  35. Munshi NC, Anderson KC, Bergsagel PL, et al. Consensus recommendations for risk stratification in multiple myeloma: report of the International Myeloma Workshop Consensus Panel 2. Blood. 2011 May 5. 117(18):4696-700. [View Abstract]
  36. Klein U, Jauch A, Hielscher T, et al. Chromosomal aberrations +1q21 and del(17p13) predict survival in patients with recurrent multiple myeloma treated with lenalidomide and dexamethasone. Cancer. 2011 May 15. 117(10):2136-44. [View Abstract]
  37. Neben K, Lokhorst HM, Jauch A, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 2012 Jan 26. 119(4):940-8. [View Abstract]
  38. Mateos MV, Hernández MT, Giraldo P, et al. Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. N Engl J Med. 2013 Aug 1. 369(5):438-47. [View Abstract]
  39. Drug Shows Promise in Slowing Progression of Smoldering Myeloma to Cancer. National Cancer Institute. Available at https://www.cancer.gov/news-events/cancer-currents-blog/2019/lenalidomide-slows-progression-smoldering-myeloma. June 7, 2019; Accessed: March 5, 2020.
  40. Zonder JA, Crowley J, Hussein MA, Bolejack V, Moore DF Sr, Whittenberger BF, et al. Lenalidomide and high-dose dexamethasone compared with dexamethasone as initial therapy for multiple myeloma: a randomized Southwest Oncology Group trial (S0232). Blood. 2010 Dec 23. 116(26):5838-41. [View Abstract]
  41. Nelson R. High-risk smoldering myeloma benefits from early treatment. Medscape Medical News. August 1, 2013.
  42. Mateos MV, Hernandez MT, Giraldo P, et al. Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. N Engl J Med. 2013 Aug 1. 369(5):438-47. [View Abstract]
  43. Morgan GJ, Child JA, Gregory WM, et al. Effects of zoledronic acid versus clodronic acid on skeletal morbidity in patients with newly diagnosed multiple myeloma (MRC Myeloma IX): secondary outcomes from a randomised controlled trial. Lancet Oncol. 2011 Aug. 12(8):743-52. [View Abstract]
  44. Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA, Bordoni R, George S, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. Myeloma Aredia Study Group. N Engl J Med. 1996 Feb 22. 334(8):488-93. [View Abstract]
  45. [Guideline] Kyle RA, Yee GC, Somerfield MR, Flynn PJ, Halabi S, Jagannath S, et al. American Society of Clinical Oncology 2007 clinical practice guideline update on the role of bisphosphonates in multiple myeloma. J Clin Oncol. 2007 Jun 10. 25 (17):2464-72. [View Abstract]
  46. [Guideline] Terpos E, Morgan G, Dimopoulos MA, Drake MT, Lentzsch S, Raje N, et al. International Myeloma Working Group Recommendations for the Treatment of Multiple Myeloma-Related Bone Disease. J Clin Oncol. 2013 May 20. [View Abstract]
  47. Zonder JA, Crowley J, Hussein MA, et al. Superiority of lenalidomide (Len) plus high-dose dexamethasone (HD) compared to HD alone as treatment of newly-diagnosed multiple myeloma (NDMM): results of the randomized, double-blinded, placebo-controlled SWOG trial S0232 [abstract 77]. Blood. 2007. 110:32a.
  48. Lokhorst HM, Sonneveld P, Cornelissen JJ, Joosten P, van Marwijk Kooy M, Meinema J, et al. Induction therapy with vincristine, adriamycin, dexamethasone (VAD) and intermediate-dose melphalan (IDM) followed by autologous or allogeneic stem cell transplantation in newly diagnosed multiple myeloma. Bone Marrow Transplant. 1999 Feb. 23(4):317-22. [View Abstract]
  49. Zervas K, Mihou D, Katodritou E, Pouli A, Mitsouli CH, Anagnostopoulos A, et al. VAD-doxil versus VAD-doxil plus thalidomide as initial treatment for multiple myeloma: results of a multicenter randomized trial of the Greek Myeloma Study Group. Ann Oncol. 2007 Aug. 18(8):1369-75. [View Abstract]
  50. Lenalidomide Improves Survival in Newly-Diagnosed Multiple Myeloma. Medscape Medical News. Available at http://www.medscape.com/viewarticle/807706. Accessed: July 23, 2013.
  51. Rajkumar SV, Jacobus S, Callander NS, Fonseca R, Vesole DH, Williams ME, et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol. 2010 Jan. 11(1):29-37. [View Abstract]
  52. Niesvizky R, Jayabalan DS, Christos PJ, Furst JR, Naib T, Ely S, et al. BiRD (Biaxin [clarithromycin]/Revlimid [lenalidomide]/dexamethasone) combination therapy results in high complete- and overall-response rates in treatment-naive symptomatic multiple myeloma. Blood. 2008 Feb 1. 111(3):1101-9. [View Abstract]
  53. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med. 2003 Jun 26. 348(26):2609-17. [View Abstract]
  54. Harousseau JL, Mathiot C, Attal M, et al. VELCADE/dexamethasone (Vel/D) versus VAD as induction treatment prior to autologous stem cell transplantation (ASCT) in newly diagnosed multiple myeloma (MM): updated results of the IFM 2005/01 trial [abstract 450]. Blood. 2007. 110:139a.
  55. Cavo M, Patriarca F, Tacchetti P, et al. . Bortezomib (Velcade[R])-thalidomide-dexamethasone (VTD) vs thalidomide-dexamethasone (TD) in preparation for autologous stem-cell (SC) transplantation (ASCT) in newly diagnosed multiple myeloma (MM) [abstract 73]. Blood. 2007. 110:30a.
  56. Mateos MV, Richardson PG, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, et al. Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow-up and impact of subsequent therapy in the phase III VISTA trial. J Clin Oncol. 2010 May 1. 28(13):2259-66. [View Abstract]
  57. Harousseau JL, Palumbo A, Richardson PG, Schlag R, Dimopoulos MA, Shpilberg O, et al. Superior outcomes associated with complete response in newly diagnosed multiple myeloma patients treated with nonintensive therapy: analysis of the phase 3 VISTA study of bortezomib plus melphalan-prednisone versus melphalan-prednisone. Blood. 2010 Nov 11. 116(19):3743-50. [View Abstract]
  58. Sher T, Ailawadhi S, Miller KC, et al. A steroid-independent regimen of bortezomib, liposomal doxorubicin and thalidomide demonstrate high response rates in newly diagnosed multiple myeloma patients. Br J Haematol. 2011 Jul. 154(1):104-10. [View Abstract]
  59. Moreau P, Pylypenko H, Grosicki S, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. Lancet Oncol. 2011 May. 12(5):431-40. [View Abstract]
  60. Mateos MV, Gutierrez NC, Martin-Ramos ML, et al. Outcome according to cytogenetic abnormalities and DNA ploidy in myeloma patients receiving short induction with weekly bortezomib followed by maintenance. Blood. 2011 Oct 27. 118(17):4547-53. [View Abstract]
  61. Moreau, Philippe, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet. 2019 Jul 6. 394 (10192):29-38. [View Abstract]
  62. Jagannath S, Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, et al. Bortezomib appears to overcome the poor prognosis conferred by chromosome 13 deletion in phase 2 and 3 trials. Leukemia. 2007 Jan. 21(1):151-7. [View Abstract]
  63. Sagaster V, Ludwig H, Kaufmann H, Odelga V, Zojer N, Ackermann J, et al. Bortezomib in relapsed multiple myeloma: response rates and duration of response are independent of a chromosome 13q-deletion. Leukemia. 2007 Jan. 21(1):164-8. [View Abstract]
  64. Myeloma Trialists' Collaborative Group. Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma: an overview of 6,633 patients from 27 randomized trials. J Clin Oncol. 1998 Dec. 16(12):3832-42. [View Abstract]
  65. Facon T, Mary JY, Hulin C, Benboubker L, Attal M, Pegourie B, et al. Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial. Lancet. 2007 Oct 6. 370(9594):1209-18. [View Abstract]
  66. Palumbo A, Falco P, Falcone A, Benevolo G, Canepa L, Gay F, et al. Melphalan, prednisone, and lenalidomide for newly diagnosed myeloma: kinetics of neutropenia and thrombocytopenia and time-to-event results. Clin Lymphoma Myeloma. 2009 Apr. 9(2):145-50. [View Abstract]
  67. Hulin C, Facon T, Rodon P, Pegourie B, Benboubker L, Doyen C, et al. Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial. J Clin Oncol. 2009 Aug 1. 27(22):3664-70. [View Abstract]
  68. Waage A, Gimsing P, Fayers P, Abildgaard N, Ahlberg L, Björkstrand B, et al. Melphalan and prednisone plus thalidomide or placebo in elderly patients with multiple myeloma. Blood. 2010 Sep 2. 116(9):1405-12. [View Abstract]
  69. Fayers PM, Palumbo A, Hulin C, et al. Thalidomide for previously untreated elderly patients with multiple myeloma: meta-analysis of 1685 individual patient data from 6 randomized clinical trials. Blood. 2011 Aug 4. 118(5):1239-47. [View Abstract]
  70. Gay F, Larocca A, Wijermans P, et al. Complete response correlates with long-term progression-free and overall survival in elderly myeloma treated with novel agents: analysis of 1175 patients. Blood. 2011 Mar 17. 117(11):3025-31. [View Abstract]
  71. Morgan GJ, Davies FE, Gregory WM, et al. Cyclophosphamide, thalidomide, and dexamethasone (CTD) as initial therapy for patients with multiple myeloma unsuitable for autologous transplantation. Blood. 2011 Aug 4. 118(5):1231-8. [View Abstract]
  72. Mateos MV, et al, for the ALCYONE Trial Investigators. Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma. N Engl J Med. 2017 Dec 12. [View Abstract]
  73. Facon T, et al. Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. N Engl J Med. 2019 May 30. 380 (22):2104-2115. [View Abstract]
  74. Attal M, Harousseau JL, Leyvraz S, et al. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood. 2006 Nov 15. 108(10):3289-94. [View Abstract]
  75. Barlogie B, Pineda-Roman M, van Rhee F, et al. Thalidomide arm of Total Therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities. Blood. 2008 Oct 15. 112(8):3115-21. [View Abstract]
  76. Spencer A, Prince HM, Roberts AW, et al. Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure. J Clin Oncol. 2009 Apr 10. 27(11):1788-93. [View Abstract]
  77. Lokhorst HM, van der Holt B, Zweegman S, et al. A randomized phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma. Blood. 2010 Feb 11. 115(6):1113-20. [View Abstract]
  78. Morgan GJ, Gregory WM, Davies FE, et al. The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis. Blood. 2012 Jan 5. 119(1):7-15. [View Abstract]
  79. McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012 May 10. 366(19):1770-81. [View Abstract]
  80. Attal M, Lauwers-Cances V, Marit G, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012 May 10. 366(19):1782-91. [View Abstract]
  81. Yang B, Yu RL, Chi XH, Lu XC. Lenalidomide treatment for multiple myeloma: systematic review and meta-analysis of randomized controlled trials. PLoS One. 2013. 8 (5):e64354. [View Abstract]
  82. Sonneveld P, Schmidt-Wolf IG, van der Holt B, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/ GMMG-HD4 trial. J Clin Oncol. 2012 Aug 20. 30(24):2946-55. [View Abstract]
  83. Palumbo A, Hajek R, Delforge M, et al. Continuous lenalidomide treatment for newly diagnosed multiple myeloma. N Engl J Med. 2012 May 10. 366(19):1759-69. [View Abstract]
  84. Palumbo A, Mina R. Part II: role of maintenance therapy in transplant-ineligible patients. J Natl Compr Canc Netw. 2013 Jan 1. 11(1):43-9. [View Abstract]
  85. Kyprolis (carfilzomib) [package insert]. South San Francisco, CA: Onyx Pharmaceuticals. July 2012. Available at
  86. San-Miguel JF, Hungria VT, Yoon SS, Beksac M, Dimopoulos MA, Elghandour A, et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 2014 Oct. 15(11):1195-206. [View Abstract]
  87. Stewart AK, Rajkumar SV, Dimopoulos MA, Masszi T, Špička I, Oriol A, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med. 2015 Jan 8. 372 (2):142-52. [View Abstract]
  88. Lonial S, Weiss BM, Usmani SZ, Singhal S, Chari A, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet. 2016 Apr 9. 387 (10027):1551-60. [View Abstract]
  89. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N Engl J Med. 2015 Sep 24. 373 (13):1207-19. [View Abstract]
  90. Moreau P, et al, for the TOURMALINE-MM1 Study Group. Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med. 2016 Apr 28. 374 (17):1621-34. [View Abstract]
  91. Lonial S, Dimopoulos M, Palumbo A, White D, Grosicki S, Spicka I, et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N Engl J Med. 2015 Aug 13. 373 (7):621-31. [View Abstract]
  92. Dimopoulos MA, Dytfeld D, Grosicki S, Moreau P, Takezako N, Hori M, et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N Engl J Med. 2018 Nov 8. 379 (19):1811-1822. [View Abstract]
  93. Ludwig H, Hajek R, Tóthová E, Drach J, Adam Z, Labar B, et al. Thalidomide-dexamethasone compared with melphalan-prednisolone in elderly patients with multiple myeloma. Blood. 2009 Apr 9. 113(15):3435-42. [View Abstract]
  94. van Rhee F, Dhodapkar M, Shaughnessy JD Jr, Anaissie E, Siegel D, Hoering A, et al. First thalidomide clinical trial in multiple myeloma: a decade. Blood. 2008 Aug 15. 112(4):1035-8. [View Abstract]
  95. Kumar S, Witzig TE, Rajkumar SV. Thalidomid: current role in the treatment of non-plasma cell malignancies. J Clin Oncol. 2004 Jun 15. 22(12):2477-88. [View Abstract]
  96. Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007 Nov 22. 357(21):2133-42. [View Abstract]
  97. Lacy MQ, Allred JB, Gertz MA, et al. Pomalidomide plus low-dose dexamethasone in myeloma refractory to both bortezomib and lenalidomide: comparison of 2 dosing strategies in dual-refractory disease. Blood. 2011 Sep 15. 118(11):2970-5. [View Abstract]
  98. Leleu X, Attal M, Arnulf B, Moreau P, Traulle C, Marit G, et al. Pomalidomide plus low dose dexamethasone is active and well tolerated in bortezomib and lenalidomide refractory multiple myeloma: IFM 2009-02. Blood. 2013 Jan 14. [View Abstract]
  99. Chustecka Z, Mulcahy N. FDA Approves Pomalidomide for Multiple Myeloma. Medscape Medical News. Available at http://www.medscape.com/viewarticle/779048. February 8, 2013; Accessed: January 11, 2018.
  100. Miguel JS, Weisel K, Moreau P, Lacy M, Song K, Delforge M, et al. Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013 Sep 2. [View Abstract]
  101. Dimopoulos MA, Moreau P, Palumbo A, Joshua D, Pour L, Hájek R, et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol. 2016 Jan. 17 (1):27-38. [View Abstract]
  102. Ajai C, Vogl DT, et al. Results of the Pivotal STORM Study (Part 2) in Penta-Refractory Multiple Myeloma (MM): Deep and Durable Responses with Oral Selinexor Plus Low Dose Dexamethasone in Patients with Penta-Refractory MM. Blood 2018 132:598.
  103. Xpovio (selinexor) [package insert]. Newton, MA: Karyopharm Therapeutics, Inc. July 2019. Available at
  104. Moreau P, Hullin C, Garban F, Yakoub-Agha I, Benboubker L, Attal M, et al. Tandem autologous stem cell transplantation in high-risk de novo multiple myeloma: final results of the prospective and randomized IFM 99-04 protocol. Blood. 2006 Jan 1. 107(1):397-403. [View Abstract]
  105. Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. J Clin Oncol. 2007 Jun 10. 25(17):2434-41. [View Abstract]
  106. Barlogie B, Attal M, Crowley J, van Rhee F, Szymonifka J, Moreau P, et al. Long-term follow-up of autotransplantation trials for multiple myeloma: update of protocols conducted by the intergroupe francophone du myelome, southwest oncology group, and university of arkansas for medical sciences. J Clin Oncol. 2010 Mar 1. 28(7):1209-14. [View Abstract]
  107. Attal M, Harousseau JL, Stoppa AM, Sotto JJ, Fuzibet JG, Rossi JF, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Français du Myélome. N Engl J Med. 1996 Jul 11. 335(2):91-7. [View Abstract]
  108. Gerull S, Goerner M, Benner A, Hegenbart U, Klein U, Schaefer H, et al. Long-term outcome of nonmyeloablative allogeneic transplantation in patients with high-risk multiple myeloma. Bone Marrow Transplant. 2005 Dec. 36(11):963-9. [View Abstract]
  109. Vesole DH, Zhang L, Flomenberg N, Greipp PR, Lazarus HM, Huff CA. A Phase II trial of autologous stem cell transplantation followed by mini-allogeneic stem cell transplantation for the treatment of multiple myeloma: an analysis of Eastern Cooperative Oncology Group ECOG E4A98 and E1A97. Biol Blood Marrow Transplant. 2009 Jan. 15(1):83-91. [View Abstract]
  110. Bruno B, Rotta M, Patriarca F, Mattei D, Allione B, Carnevale-Schianca F, et al. Nonmyeloablative allografting for newly diagnosed multiple myeloma: the experience of the Gruppo Italiano Trapianti di Midollo. Blood. 2009 Apr 2. 113(14):3375-82. [View Abstract]
  111. Rotta M, Storer BE, Sahebi F, Shizuru JA, Bruno B, Lange T, et al. Long-term outcome of patients with multiple myeloma after autologous hematopoietic cell transplantation and nonmyeloablative allografting. Blood. 2009 Apr 2. 113(14):3383-91. [View Abstract]
  112. Moreau P, Attal M, Pégourié B, et al. Achievement of VGPR to induction therapy is an important prognostic factor for longer PFS in the IFM 2005-01 trial. Blood. 2011 Mar 17. 117(11):3041-3044. [View Abstract]
  113. Harousseau JL, Attal M, Avet-Loiseau H, et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial. J Clin Oncol. 2010 Oct 20. 28(30):4621-9. [View Abstract]
  114. Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet. 2010 Dec 18. 376(9758):2075-85. [View Abstract]
  115. Garderet L, Iacobelli S, Moreau P, Dib M, Lafon I, Niederwieser D, et al. Superiority of the Triple Combination of Bortezomib-Thalidomide-Dexamethasone Over the Dual Combination of Thalidomide-Dexamethasone in Patients With Multiple Myeloma Progressing or Relapsing After Autologous Transplantation: The MMVAR/IFM 2005-04 Randomized Phase III Trial From the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. J Clin Oncol. 2012 May 14. [View Abstract]
  116. Cooper MR, Dear K, McIntyre OR, Ozer H, Ellerton J, Canellos G, et al. A randomized clinical trial comparing melphalan/prednisone with or without interferon alfa-2b in newly diagnosed patients with multiple myeloma: a Cancer and Leukemia Group B study. J Clin Oncol. 1993 Jan. 11(1):155-60. [View Abstract]
  117. Morgan GJ, Davies FE, Gregory WM, Cocks K, Bell SE, Szubert AJ, et al. First-line treatment with zoledronic acid as compared with clodronic acid in multiple myeloma (MRC Myeloma IX): a randomised controlled trial. Lancet. 2010 Dec 11. 376(9757):1989-99. [View Abstract]
  118. Terpos E, Roodman GD, Willenbacher W, Shimizu K, Garcia-Sanz R, Durie B, et al. Comparison of denosumab (DMB) with zoledronic acid (ZA) for the treatment of bone disease in patients with newly diagnosed multiple myeloma; an international, randomized, double blind trial (oral presentation S782). Presented at the 22nd Annual Congress of the European Hematology Association, Madrid Spain. 2017 June 25.
  119. [Guideline] Ruggiero SL, Dodson TB, Fantasia J, Goodday R, Aghaloo T, Mehrotra B, et al. American Association of Oral and Maxillofacial Surgeons position paper on medication-related osteonecrosis of the jaw--2014 update. J Oral Maxillofac Surg. 2014 Oct. 72(10):1938-56. [View Abstract]
  120. Djulbegovic B, Wheatley K, Ross J, Clark O, Bos G, Goldschmidt H, et al. Bisphosphonates in multiple myeloma. Cochrane Database Syst Rev. 2001. CD003188. [View Abstract]
  121. Bloomfield DJ. Should bisphosphonates be part of the standard therapy of patients with multiple myeloma or bone metastases from other cancers? An evidence-based review. J Clin Oncol. 1998 Mar. 16(3):1218-25. [View Abstract]
  122. Ludwig H, Fritz E, Kotzmann H, Höcker P, Gisslinger H, Barnas U. Erythropoietin treatment of anemia associated with multiple myeloma. N Engl J Med. 1990 Jun 14. 322(24):1693-9. [View Abstract]
  123. Wilson J, Yao GL, Raftery J, Bohlius J, Brunskill S, Sandercock J, et al. A systematic review and economic evaluation of epoetin alpha, epoetin beta and darbepoetin alpha in anaemia associated with cancer, especially that attributable to cancer treatment. Health Technol Assess. 2007 Apr. 11(13):1-202, iii-iv. [View Abstract]
  124. Parikh GC, Amjad AI, Saliba RM, Kazmi SM, Khan ZU, Lahoti A, et al. Autologous hematopoietic stem cell transplantation may reverse renal failure in patients with multiple myeloma. Biol Blood Marrow Transplant. 2009 Jul. 15(7):812-6. [View Abstract]
  125. [Guideline] Terpos E, Kleber M, Engelhardt M, et al; European Myeloma Network. European Myeloma Network Guidelines for the Management of Multiple Myeloma-related Complications. Haematologica. 2015 Oct. 100 (10):1254-66. [View Abstract]
  126. Golombick T, Diamond TH, Manoharan A, Ramakrishna R. Monoclonal gammopathy of undetermined significance, smoldering multiple myeloma, and curcumin: a randomized, double-blind placebo-controlled cross-over 4g study and an open-label 8g extension study. Am J Hematol. 2012 May. 87(5):455-60. [View Abstract]
  127. Chang ET, Canchola AJ, Cockburn M, et al. Adulthood residential ultraviolet radiation, sun sensitivity, dietary vitamin D, and risk of lymphoid malignancies in the California Teachers Study. Blood. 2011 Aug 11. 118(6):1591-9. [View Abstract]
  128. [Guideline] Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014 Nov. 15 (12):e538-48. [View Abstract]
  129. [Guideline] Moreau P, San Miguel J, Sonneveld P, Mateos MV, Zamagni E, Avet-Loiseau H, et al. Multiple myeloma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2017 Apr 27. [View Abstract]
  130. [Guideline] Palumbo A, Avet-Loiseau H, Oliva S,et al. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J Clin Oncol. 2015 Sep 10. 33 (26):2863-9. [View Abstract]
  131. [Guideline] Rizzo JD, Brouwers M, Hurley P, Seidenfeld J, Arcasoy MO, Spivak JL, et al. American Society of Clinical Oncology/American Society of Hematology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. J Clin Oncol. 2010 Nov 20. 28 (33):4996-5010. [View Abstract]
  132. [Guideline] National Comprehensive Cancer Network. Cancer-Associated Venous Thromboembolic Disease. V1.2016. NCCN. Available at https://www.nccn.org/professionals/physician_gls/pdf/vte.pdf. July 22, 2016; Accessed: May 12, 2017.
  133. Palumbo A, Rajkumar SV, Dimopoulos MA, et al. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia. 2008 Feb. 22 (2):414-23. [View Abstract]
  134. Vickrey E, Allen S, Mehta J, Singhal S. Acyclovir to prevent reactivation of varicella zoster virus (herpes zoster) in multiple myeloma patients receiving bortezomib therapy. Cancer. 2009 Jan 1. 115(1):229-32. [View Abstract]
  135. Petrucci MT, Giraldo P, Corradini P, et al. A prospective, international phase 2 study of bortezomib retreatment in patients with relapsed multiple myeloma. Br J Haematol. 2013 Jan 7. [View Abstract]
  136. Rodon P. Management and treatment of multiple myeloma in elderly patients. Ann Long-Term Care. 2002. 10:20-7.
  137. Palumbo A, Cavo M, Bringhen S, et al. Aspirin, warfarin, or enoxaparin thromboprophylaxis in patients with multiple myeloma treated with thalidomide: a phase III, open-label, randomized trial. J Clin Oncol. 2011 Mar 10. 29(8):986-93. [View Abstract]
  138. Jackson GH, Morgan GJ, Davies FE, Wu P, Gregory WM, Bell SE, et al. Osteonecrosis of the jaw and renal safety in patients with newly diagnosed multiple myeloma: Medical Research Council Myeloma IX Study results. Br J Haematol. 2014 Jul. 166(1):109-17. [View Abstract]

Bone marrow aspirate demonstrating plasma cells of multiple myeloma. Note the blue cytoplasm, eccentric nucleus, and perinuclear pale zone (or halo). All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Amyloidosis infiltrating the tongue in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Radiograph of the skull demonstrating a typical lytic lesion in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Bone marrow aspirate demonstrating plasma cells of multiple myeloma. Note the blue cytoplasm, eccentric nucleus, and perinuclear pale zone (or halo). All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Bone marrow biopsy demonstrating sheets of malignant plasma cells in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Amyloidosis infiltrating the tongue in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Radiograph of the skull demonstrating a typical lytic lesion in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Bone marrow aspirate demonstrating plasma cells of multiple myeloma. Note the blue cytoplasm, eccentric nucleus, and perinuclear pale zone (or halo). All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.

Bone marrow biopsy demonstrating sheets of malignant plasma cells in multiple myeloma. All images and text are (c) 2002 by the American Society of Hematology. All rights reserved.