fbpx
Wikipedia

Plasma cell dyscrasias

Plasma cell dyscrasias (also termed plasma cell disorders and plasma cell proliferative diseases) are a spectrum of progressively more severe monoclonal gammopathies in which a clone or multiple clones of pre-malignant or malignant plasma cells (sometimes in association with lymphoplasmacytoid cells or B lymphocytes) over-produce and secrete into the blood stream a myeloma protein, i.e. an abnormal monoclonal antibody or portion thereof. The exception to this rule is the disorder termed non-secretory multiple myeloma; this disorder is a form of plasma cell dyscrasia in which no myeloma protein is detected in serum or urine (at least as determined by conventional laboratory methods) of individuals who have clear evidence of an increase in clonal bone marrow plasma cells and/or evidence of clonal plasma cell-mediated tissue injury (e.g. plasmacytoma tumors). Here, a clone of plasma cells refers to group of plasma cells that are abnormal in that they have an identical genetic identity and therefore are descendants of a single genetically distinct ancestor cell.

Plasma cell dyscrasia
Other namesPlasma cell proliferative diseases
SpecialtyHematology, oncology

At one end of this spectrum of hematological disorders, detection of one of these myeloma proteins in an individual's blood or urine is due to a common and clinically silent disorder termed MGUS, i.e. monoclonal gammopathy of undetermined significance. At the other end of this spectrum, detection of the myeloid protein is due to a hematological malignancy, i.e. multiple myeloma, Waldenström macroglobulinemia, or other B cell-associated neoplasm, that has developed, often in a stepwise manner, from their MGUS precursors.[1][2]

The clinical importance of understanding this spectrum of diseases is that it can be used to: a) advise individuals on the likelihood of their condition progressing to a malignant phase; b) monitor individuals for the many complications that may occur at any stage of the dyscrasias so that they can be treated to avoid or reduce their clinical impacts; and c) monitor patients for transitions to malignancy so that the malignancy can be treated at an early stage when treatment results are best.[3] Unless otherwise noted, the advice and monitoring given here are those recommended by the International Myeloma Working Group in 2014[4] and updated in 2016.[5]

Clonal plasma cells

Plasma cells are key effector elements of the adaptive immune system. They contribute to immunity by making antibodies that bind with and thereby initiate the process of neutralizing specific antigens that usually are found on the surface of invading pathogens and foreign substances. Plasma cells develop from B lymphocytes (i.e. B cells) which are stimulated to undergo this maturational development by T lymphocytes during the latter cells' processing of these antigens. As they are stimulated to become plasma cells, B cells refashion parts of their genome in efforts to create a new gene that encodes a functional antibody. In humans, antibodies are composed of two identical heavy chains which are of the gamma (γ), alpha (α), epsilon (ε), delta (δ), or mu (μ) subtypes and two identical light chains which are of the kappa (κ) or lambda (λ) subtypes. Antibodies are classified as IgG, IgA, IgE, IgD, and IgM based on their being made up of γ, α, ε, δ, or μ heavy chains, respectively. Formation of the genes that make these antibodies requires B cells and/or their descendent plasma cells to mutate, break, and recombine various genes at the immunoglobulin heavy chain antigen-binding locus on the long (i.e. "q") arm of human chromosome 14 at position 32.33 (notated as 14q32.33) and the immunoglobulin light chain antigen binding locus on the q arm of chromosome 22 at position 11.2 (i.e. 22 q11.2) by processes termed V(D)J recombination, somatic hypermutation, and immunoglobulin class switching. These genomic changes can go awry by placing a gene that controls cell growth an/or survival adjacent to a normally highly active antibody gene promoter and/or by causing the formation of extra chromosomes (see trisomy) or chromosomes with large deletions that result in the overexpression or under-expression, respectively, of genes that control cell growth and/or survival. In consequence of these "primary genomic changes", an expanding clone of cells develops; overproduces and secretes a monoclonal IgM, IgG, IgA, IgE, or IgD antibody, a κ or λ light chain, an α, γ, or μ heavy chain, or, very rarely, fragments of these proteins; and may accumulate "secondary genomic changes" that cause them to become malignant. The overproduced monoclonal proteins, termed myeloma proteins, commonly circulate in blood, may accumulate in urine, and are the hallmarks of plasma cell dyscrasias including their most malignant forms viz., multiple myeloma, light chain multiple myeloma, and plasma cell leukemia.[6][7][8] IgG-secretory, IgA-secretory, and light-chain secretory multiple myeloma represent 52%, 21%, and 16%, respectively, of all multiple myeloma cases; these myelomas are associated with various types of chromosomal aberrancies and mutations. IgD-secretory multiple myeloma occurs in only 1% to 2% of multiple myeloma cases and is commonly associated with somatic mutations in the gene encoding the gV (i.e. variable) region of the monoclonal antibody. IgE-secretory multiple myeloma has been reported in <50 cases as of 2013 and is characteristically associated with translocations between the q arms of chromosome 11 and 14, i.e. t(11;14)(q13;q32) translocations.[9]

In other cases, plasma cells and/or lymphoplasmacytoid cells (a type of B cell resembling, and possible precursor to, plasma cells) suffer other kinds of mutations that lead to the production of an IgM myeloma protein. Overproduction of this myeloma protein may progress to a different form of plasma cell/lymphoplasmacytoid cell malignancy, Waldenström macroglobulinaemia. Genetic mutations thought to be involved in the development and/or progression of the latter disease include the L265P mutation in the MYD88 gene found in >90% of Waldenström macroglobulinaemia patients as well as various mutations in the CXCR gene found in 27% to 40% of Waldenström macroglobulinaemia patients.[6][7][10][11]

The clonal plasma cells involved in plasma cell dyscrasias exhibit a high degree of genetic instability. For example, the clonal plasma cell population formed by initial genetic alterations that lead to multiple myeloma contains cells that develop further genetic changes that enhance their survival, proliferation, tissue-injuring, and metastatic capacities. This allows the new cell clones to crowd out older cell clones and thereby establish a more malignant disease. Repetition of such genetic changes underlie the evolution of a clinically silent plasma cell dyscrasia to an overt malignancy.[6][7][11] The progressive genetic changes in clonal plasma cells include accumulating numerous single nucleotide polymorphisms, increases and decreases in gene and chromosome copy numbers, and chromosomal translocations. Genes affected include those regulating genome stability itself (e.g. KIF2B[12]) as well as cellular activation, proliferation, and apoptosis (e.g. CIDEC,[13] TP52, ATM, KRAS, NRAS, Wnt, and NF-κB). In the most malignant form of plasma cell dyscrasias, primary plasma cell leukemia, the plasma cell population contains >1900 distinct DNA alterations in >600 genes.[14]

In general, the plasma cell dyscrasias are defined by 1) the presence of these genetically unstable clonal plasma cells, lymphoplasmacytoid cells, or B cells infiltrating the bone marrow or forming distinct masses in bone, and/or other tissues as defined by biopsy of involved tissues and 2) the presence of these cells' myeloma proteins (i.e. intact monoclonal antibody, free light chain, free heavy chain, shortened version of these proteins, or any combination of these proteins) in blood and/or urine as defined by various types of gel electrophoresis. Obviously, the latter criterion does not apply to the rare cases of true non-secretory myeloma.[6]

Myeloma protein toxicity

Myeloma proteins form as a result of gene mutations rather than physiological gene remodeling responses to an instigating foreign antigen: typically these proteins are non-functional. However, they sometimes cause serious tissue damage with the kidney being a particularly vulnerable target. The toxic effects of monoclonal proteins may occur at early stages in the plasma cell dyscrasia spectrum and require treatment independently of the mass or tissue-destructive effects of the myeloma protein-producing cells. Myeloma protein toxicities include:

  • Monoclonal free light chains, free heavy chains, or a mixture of these chains can deposit in the kidney and other organs to cause systemic monoclonal immunoglobulin deposition disease; free κ or λ light chains can deposit selectively in the kidney's proximal tubule to cause light chain proximal tubulopathy or in the kidneys distal tubule to cause light chain myeloma cast nephropathy; and various myeloma proteins can deposit selectively in the kidneys glomeruli to cause various forms of organized deposit and non-organized deposit glomerulonephritis diseases.[15]
  • Free κ or λ light chains can aggregate with each other to cause extracellular amyloid deposits and a disease termed amyloidosis in which the deposits injure and ultimately lead to the failure of such organs as the kidney, heart, liver, stomach, and intestines;[15] these deposits can also cause peripheral and autonomic neuropathies.[1]
  • IgM myeloma proteins or in rare cases other myeloma proteins such as IgA, free κ light chains, or free λ light chains may increase blood viscosity, deposit in peripheral blood vessels, and thereby cause vascular occlusion and gangrene of the extremities in the syndrome termed cryoglobulinemia.[7]
  • Monoclonal IgM myeloma proteins operating through their effects on increasing blood hyperviscosity can reduce blood flow to the central nervous system to cause blurred vision, headaches, vertigo, ataxia, and cold-induced hemolytic anemia.[8]
  • IgM, IgG, and to lesser extents κ and λ free light chain myeloma proteins can cause Immune thrombocytopenic purpura with extensive bleeding tendencies .[16]

Stages

MGUS stage

Monoclonal gammopathy of undetermined significance (MGUS), is defined as the presence in the blood or urine of a monoclonal antibody, antibody heavy chain, or antibody light chain in a person lacking symptoms or signs of a more serious plasma cell dyscrasia. The condition is typically discovered as an incidental finding when serum protein electrophoresis is done for various reasons unrelated to plasma cell dyscrasias. Protein electrophoresis generally detects one of the following patterns of monoclonal myeloma protein spikes representing: a) intact IgG, IgA, IgE, IgE, or IgM; b) intact IgG, IgA, IgE, IgD, or IgM plus high concentrations of a free (i.e. not bound to a heavy chain) κ or λ light chain; c) a free κ chain in great excess of a λ chain or a free λ chain in great excess of a κ chain; and d) free γ, δ, or μ heavy chains unbound to a light chain (free α and ε heavy chain myeloma protein spikes have not been reported). Among MGUS cases expressing an intact antibody, 70%, 15%, 12%, and 3% express either IgG, IgM, IgA, or two of these M proteins, respectively, with or without excessive levels of a light chain; these cases represent ~80% of all MGUS. About 20% of MGUS cases express either κ or λ light chains. As a group, these MGUS findings occur more commonly in men and are ~2-fold more common in individuals of African descent than Caucasians.[2][17] MGUS cases expressing free γ, δ, or μ heavy chains are extremely rare.[17][11] MGUS is categorized into the following sub-types based upon the identity and levels of the myeloma proteins detected as well as the prognoses for progressive disease indicated by these myeloma protein findings.[citation needed]

Non-IgM MGUS

Non-IgM MGUS, commonly termed MGUS, is diagnosed in individuals who exhibit a serum IgG, IgD, IgA, or IgE monoclonal protein with or without increased levels of blood and/or urine free κ or λ light chains. These patients typically also show small increases in bone marrow plasma cells. Further requirements for the diagnosis of non-IgM MGUS are: a) bone marrow clonal plasma cells <10% of total nucleated cells; b) absence of any of the four CRAB criteria (CRAB criteria are C = Calcium serum levels >1 milligram/deciliter above normal values and/or a serum level >11 milligram/deciliter; R = Renal insufficiency as defined by a glomerular filtration rate <40 milliliter/minute and/or a serum creatinine >2 gram/deciliter due to myeloma protein-induced kidney damage; A = Anemia, as defined by a blood hemoglobin level >2 gram/deciliter below normal and/or <10 gram/deciliter due to the plasma cell dyscrasia rather than e.g. iron deficiency or blood loss; B = Bone lesions, i.e. ≥1 lytic (i.e. bone re-adsorbing) bone lesion due to a plasmacytoma as detected by skeletal radiography, computed tomography, or positron emission tomography-computed tomography); c) no evidence of a plasmacytoma in bone or soft tissues, of amyloidosis, or of another plasma cell disorder; d) a ratio of free serum light chains (i.e. free κ/λ or λ/κ light chain ratio) less than 100, providing that the higher light chain concentration is >100 milligram/liter; and e) a circulating blood plasma cell absolute count of <2x109 and/or <20% of total circulating nucleated cells.[2][18][19] Presence of any one of the latter findings indicates that the plasma cell dyscrasia has progressed beyond the MGUS stage.[citation needed]

Non-IgM MGUS is a relatively stable condition afflicting 3% of people aged 50 and 5% of people aged 70; on average, it progresses to multiple myeloma at a rate of 0.5-1% cases per year, as defined in studies following patients over a 25-year period. A study conducted by the Mayo Clinic found that MGUS associated with non-IgG myeloma proteins or with serum myeloma protein levels greater than 15 gram/liter had a greater risk per year of progression to multiple myeloma. A Spanish study group found that MGUS patients demonstrating aneuploidy (i.e. abnormal number of chromosomes) in bone marrow cells or >95% of resident bone marrow plasma cells that are clonal in nature also have a greater risk per year of progression to myeloma.[6] In a more recent study, MGUS patients that had the presence of none, 1, 2, or 3 of the three following risk factors, serum M protein levels >15 gram/liter, a non-IgG isotype, and abnormal free light chain ratios, had 5, 32, 37, and 58% chances, respectively, of progressing to multiple myeloma within 20 years. In another study, MGUS patients with none, 1, or 2 of the following risk factors, >95% of bone marrow plasma cells that are clonal in nature and a 10% or greater rise in the levels of their monoclonal proteins within 3 years, had 2, 16, and 72% risks, respectively, of progression within 7 years.[3] However, estimates on the risks of progression for some of these parameters are tentative and subject to change. For example, the IgA form of MGUS, while once considered to have a poorer prognosis than IgG MGUS, was found to have a prognosis similar to IgG MGUS in a more recent study.[20]

IgM MGUS

While traditionally classified as such, it is not clear that IgM MGUS is a clonal plasma cell dyscrasia. IgM MGUS involves an increase in a B cell derivative with morphological features of both plasma cells and lymphocytes viz., lymphoplasmacytic cells. Studies indicate that both plasma cells and lymphoblastic cells infiltrate involved tissues and that one or perhaps both cell types harbor mutations in a) the MYD88 gene (~20% in IgM MGUS and >90% in IgM-related malignancies), almost all of which are L265P mutations (i.e. changing leucine to proline at the 265th amino acid position of the MYK88 protein thereby causing the protein to be continuously active in stimulating the same cell-activating pathways that Toll-like receptors activate intermittently and on a physiologically basis); b) the CXCR4 gene (8% in IgM MGUS, 25% in IgM-related malignancies); and c) increased gene copy number due to chromosomal rearrangements (36% in IgM MGUS, 82% in IgM-related malignancies).[17] It is clear that each cell type contributes to different features of IgM malignancies but not clear that clonal plasma cells are critical to the development or progression of IgM MGUS.[17] In all events, IgM MGUS is diagnosed in individuals who have serum IgM levels less than 30 gram/liter; have less than 10% of nucleated bone marrow cells with the lymphoplasmacytic morphology, and have no symptoms or findings of end organ dysfunction attributed to Waldenström macroglobulinemia such as anemia, decreases in any white blood cell count, cold agglutinin disease, hyperviscosity of blood, lymphadenopathy, hepatomegaly, splenomegaly, peripheral neuropathy, cryoglobulinemia, or constitutional symptoms.[2][8][17]

There may be a modest increase in the incidence of IgM MGUS in people of African descent. A study of 213 individuals diagnosed with IgM MGUS found that 10% at 5 years and 24% in 15 years progressed to more serious IgM-related diseases including non-Hodgkin lymphoma, Waldenstorm's macroglobulinemia, systemic amyloidosis, and chronic lymphocytic leukemia. A second long-term study of 116 individuals with IgM MGUS found a 15-fold increased risk of progressing to a lymphoid malignancy, mostly to Waldenstorm's macroglobulinemia. In general, progression to one of these malignant outcomes occurs at a rate of 2% to 3% per year. Individuals with higher serum IgM or lower serum albumin levels progress at faster rates than those with normal levels of these parameters.[17]

Light chain MGUS

Individuals diagnosed with light chain MGUS typically do not express detectable levels of an IgG, IgA, IgD, IgE, or IgM intact myeloma protein in their blood. Rather, they overexpress a monoclonal, aberrant free κ (i.e. kappa) or λ (i.e., lambda) immunoglobulin light chain. For diagnosis, the κ and λ free light chains are quantified by immunological methods and the ratio of κ to λ light chains is used to detect unbalanced light chain synthesis that is indicative of a monoclonal light chain plasma cell dyscrasia. Light chain MGUS is defined as a disorder in which a serum κ to λ free light chain ratio falls outside the normal range of 0.26–1.65 (mean =0.9) provided that it is not associated with: a) any of the CRAB criteria, b) a bone marrow plasma cell count of 10 or a higher percentage of nucleated cells, c) evidence of amyloid deposition (see Light chain deposition disease), and d) an accumulation of 0.5 or more grams of the monoclonal light chain in the urine over a 24-hour period. As so defined, light chain MGUS comprises ~19% of all MGUS cases, occurs in ~0.8% of the general population, and progresses to light chain multiple myeloma at the very slow rate of 0.3 cases per 100 years.[2][6][3]

Some early studies have reported that a very rapid rate of progression occurs in light chain MGUS patients who have free light chain κ/λ or λ/κ ratios equal to or greater than 100 (i.e. and κ/λ ratio outside of 0.02 to 100). About 80% of individuals bearing these light chain ratios were found to progress to light chain multiple myeloma within 2 years. In consequence, these individuals were recommended for being diagnosed and treated as having light chain multiple myeloma.[3] However, two more recent studies reported a 2-year progression rate for these patients of 64% and 30%. It is therefore suggested that the diagnosis of light chain multiple myeloma based solely on a free κ/λ light chain ratio of 0.02 to 100 may be premature.[20]

Monoclonal gammopathy of renal significance

Monoclonal gammopathy of renal significance or MGRS designates any MGUS disorder that has a clinically significant impact on renal function. MGRS can be caused by the deposition of a monoclonal immunoglobulin in, and consequent injury to, the kidneys. The diagnosis of this form of MGRS is made based on the presence of: 1) a disorder meeting the criteria for MGUS; b) decreased kidney function as evidence by, e.g. a Glomerular filtration rate of <40; and c) biopsy confirmed or suspicion of cast nephropathy, glomerulonephritis, of other morphological expressions of clonal immunoglobulin-induced kidney injury. Increased excretion of a urinary monoclonal light chain (typically >0.5 gram/day), which suggests the presence of a particularly severe form of kidney injury (myeloma cast nephropathy), supports but is not a requirement for the diagnosis of MGRS.[5][15][21] The disorder can also be caused by a monoclonal immunoglobulin that acts as an autoantibody that activates the blood complement system to cause complement-related kidney injury. This form of MGRS is usually associated with other syndromes like glomerulopathy associated with a monoclonal immunoglobulin or C4 dense deposit disease associated with a monoclonal immunoglobulin. Diagnosis depends or identifying these other syndromes and the identification of complement components on kidney biopsy.

Regardless of the exact pathophysiology causing monoclonal immunoglobulin-induced kidney injury, MGRS has a greater morbidity and mortality than other forms of MGUS. Since renal dysfunction usually improves with therapy directed at the underlying plasma cell dyscrasia, MGRS may warrant treatment even when other parameters of plasma cell dyscrasia severity (e.g. low levels of serum monoclonal immunoglobulin and bone marrow plasma cells) suggest the presence of minimal, non-malignant disease.[21]

Smoldering multiple myeloma stage

Smoldering multiple myeloma or SMM (also termed smoldering myeloma) is the next stage following MGUS in the spectrum of plasma cell dyscrasias. While still considered a pre-malignant condition, its chances of progressing to a malignant plasma cell dyscrasia are generally greater than that for MGUS.[20] SMM consists of the following subtypes which represent progression of their corresponding MGUS subtypes.[citation needed]

Non-IgM SMM

Non-IgM SMM (also termed IgG and IgA SMM because of the rarity of IgD and IgE SMM) is diagnosed in asymptomatic individuals based on criteria identical to those listed above for Non-IgM MGUS except that: their intact IgG or IgA myeloma protein levels are equal to or greater than 30 grams/liter rather than 15 grams/liter; their bone marrow shows plasma cells comprise between 10% and <60% rather than <10% of nucleated cells; and/or their 24-hour urine contains 0.5 gram or greater levels of Bence Jones, i.e. light chain myeloma, proteins. individuals must also lack evidence of more recently established multiple myeloma-defining criteria viz., CRAB features, amyloidosis, more than one solitary plasmacytoma, and/or serum or urine free light chain κ to λ or λ to κ ratios of 100 or greater.[2][20]

Overall, the risk of Non-IgM SMM progressing to multiple myeloma is 10% per year for the first 5 years but falls off sharply to 3% per year for the next 5 years and thereafter to 1% per year.[2]

Smoldering Waldenström macroglobulinemia

Smoldering Waldenström macroglobulinemia is diagnosed in asymptomatic individuals that have a serum IgM level 30 gram/liter and/or a bone marrow lymphoplasmacytoid cell infiltrate >10% of total nucleated cells. These cases should have no symptoms or findings of end organ dysfunction attributed to Waldenström macroglobulinemia such as anemia, decreases in any white blood cell count, cold agglutinin disease, hyperviscosity of blood, lymphadenopathy, hepatomegaly, splenomegaly, peripheral neuropathy, cryoglobulinemia, or constitutional symptoms.[17]

As determined by a Mayo Clinic study of 48 individuals, smoldering Waldenström macroglobulinemia's risk of progression to Waldenström macroglobulinemia is estimated to be ~12% per year and then falling of sharply for at least the next 5 years to 2% per year. In this study the only factor predictive of a more rapid progress was a finding of anemia (hemoglobin level <115 grams/liter). During a 15-year follow-up, the Clinic subsequently reported that patients progressed to Waldenström macroglobulinemia, amyloidosis, or a related IgM-associated neoplasm at a rate of 6%, 39%, 59%, and 68% after the first, third, fifth, and tenth year, respectively. However, the Southwest Oncology Group in a study on 231 individuals reported that the smoldering disease progressed to overt Waldenström macroglobulinemia over 9 years in only 26% of cases.[2][8][17]

Light chain SMM

Light chain smoldering multiple myeloma (light chain SMM) was previously termed idiopathic Bence Jones proteinuria. The condition is currently diagnosed in asymptomatic individuals who have a 24-hour urinary Bence Jones, i.e. light chain myeloma protein level, that is >0.5 grams and/or bone marrow plasma cells that are 10% to <60% of nucleated cells. These individuals must also; lack detectable IgG, IgA, IgD, IgE, or IgM myeloma proteins in sera; have a free κ/λ or λ/κ light chain ratio outside of 0.26 to 1.65 range but less than 100; and/or have no evidence for the presence of any one of the CRAB criteria, amyloidosis, or end organ damage attributable to the myeloma proteins or plasma cells.[2][3][6]

In a Mayo clinic study of 101 individuals with light chain SMM, the cumulative probability of progression to active multiple myeloma or light-chain amyloidosis in patients with light-chain SMM was 28%, 45%, and 56% after 5, 10, and 15 years, respectively. The major risk factors for progression were the level of urinary excretion of M protein, percentage of bone marrow plasma cells, and immunoparesis (i.e. reduced serum levels of intact immunoglobulins).[2][22]

Paraneoplastic complications

Serious and potentially life-threatening paraneoplastic complications can occur in plasma cell dyscrasias regardless of tumor cell burden, myeloma protein levels, or the presence of other criteria suggesting the dyscrasia has entered a malignant phase. Many of these complications are caused by the tissue-destructive effects of the myeloma proteins, are predictive of a rapidly progressive disease, and require chemotherapeutic or other treatments directed at lowering the burden of the myeloma protein-producing cells. The serious paraneoplasitic diseases that complicate the plasma cell dyscrasias and may require such treatments include the following.[citation needed]

Amyloidosis

Amyloidosis is a general term for a protein misfolding syndrome that involves the deposition of a low molecular weight beta-pleated sheet-containing protein in extracellular tissues. These proteins normally circulate in the blood but may undergo conformational changes that cause them to auto-aggregate along their beta-pleated sheets to become insoluble and form fibril deposits in and outside of the circulation. These deposits disrupt tissue architecture and, in the case of light chains, directly injure cells, thereby causing potentially cataclysmic organ failures. There are 31 types of circulating proteins that can become misfolded and lead to distinctly different types of amyloidosis; among these, myeloma proteins, particularly free light chains, are the predominant cause of the disease.[20][23] Increases in the levels of free κ or λ light chains are a common feature of plasma cell dyscrasias. These increases occur in: 40% of IgM MGUS, IgM SMM, and Waldenstroms macroglbulonemia cases; 60% to 70% of non-secretory multiple myelom cases; 90% to 95% of intact immunoglobulin multiple myeloma cases; and, by definition, 100% of light chain multiple myeloma cases.[2] There are two different types of plasma cell dyscrasia-associated amyloidosis syndromes: amyloid light chain amyloidosis (AL amyloidosis) in which amyloid deposits consist of free light chains and amyloid heavy chain amyloidosis (AH amyloidosis) in which amyloid deposits contain only free heavy chains. The deposits in a third type, AHL amyloidosis, consists of both free light chains and free heavy chains. AHL amyloidosis is here, as in some recent reports, grouped with AH amyloidosis.[24]

AL Amyloidosis

AL amyloidosis can occur at any stage in the plasma cell dyscrasia spectrum. Typically, patients developing this type of amyloidosis have had excess κ or λ free light chains in their urine for years before diagnosis. At diagnosis, however, they typically have a relatively small plasma cell burden (bone marrow plasma cells <5% to 7% of total nucleated cells) and in only <5% to 10% of cases do other findings indicate the presence of a malignant condition (i.e. definitive signs of multiple myeloma, Waldenström macroglobulinemia, or chronic lymphocytic leukemia associated with over-production of a clonal light chain). Nonetheless, these individuals often evidence serious involvement of the kidney (proteinuria, nephrotic syndrome) or heart (restrictive cardiomyopathy, arrhythmias) in 70% or 60% of cases, respectively, and of dysfunction in the peripheral nervous system (numbness, paresthesias) or autonomic nervous system (orthostatic hypotension) in 20% or 15% of cases, respectively. They may also exhibit evidence of liver involvement (liver failure, increases in circulating liver enzymes, bleeding due to factor X deficiency), gastrointestinal track deficiencies (malabsorption), and amyloid deposition in surface tissues (macroglossia, shoulder pad masses, cutaneous nodules). Arthritis in multiple joints, often manifested before diagnosis, is also a common feature of AL amyloidosis and has led to initial misdiagnoses of rheumatoid arthritis.[25] Diagnosis of the disease requires evidence of increased levels of a κ or λ myeloma protein in blood and/or blood, presence of an amyloid-related organ-involvement syndrome, detection in tissues of amyloid deposition based on birefringence-staining with Congo red, and detection in tissues of κ or λ deposition based on electron microscopy or mass spectrometry.[1] Reflecting the widespread systemic nature of the disease, patient median survival is only 8 months dating from the time of diagnosis. Treatment commonly improves this poor survival. In a Mayo Clinic study, for example, AL amyloidosis assigned stage 1, 2, 3, or 4 based on the presence of 0, 1, 2, or 3 prognostic signs (high blood levels of [cardiac troponin T]), blood levels of a marker for congestive heart failure (viz., NT-ProBNP), or free light chain ratios) had median survivals of 94.1, 40.3, 14, and 5.8 months respectively. Additional factors indicating a worse prognosis include the involvement of multiple organs, ≥ 10% bone marrow plasma cells, presence of a translocation between chromosomes 11 and 14 [i.e. t(11;14)], and chromosomal trisomy.[20]

AH Amyloidosis

AH and AHL amyloidosis are extremely rare forms of systemic amyloidosis in which the amyloid deposit is a free heavy chain (AH amyloidosis) or a free heavy chain plus free light chain (AHL amyloidosis).[26] Case reports have detected amyloid deposits containing a free γ, α, or μ heavy chain (or portions of one of these chains) accompanied in many cases by a free κ or λ light chain in primarily in the kidney but also the spleen and other tissues. AH plus AHL amyloidosis cases are ~17-fold less common than AL amyloidosis cases.[26] The disease often presents late in its course with signs and/or symptoms of kidney failure such as those associated with the nephrotic syndrome and is therefore treated as a malignant condition.[24][27] In a small study of 16 patients with renal amyloidosis, the 5 patients with AH amyloidosis and the 11 patients with AHL amyloidosis had less frequent concurrent cardiac involvement and better overall survival than 202 patients with renal AL amyloidosis. The hematological response to chemotherapy of the AH and AHL renal amyloidosis patients was comparable to those with renal AL amyloidosis.[24]

POEMS syndrome

POEMS syndrome (also known as Crow–Fukase syndrome, Takatsuki disease, or PEP syndrome) is a rare and complex medical syndrome that involves a combination of syndrome-defining signs and symptoms due to the dysfunction of multiple organs. The syndrome is associated with a plasma cell dyscrasia in almost 100% of cases, pathological overexpression of certain cytokines in >95% of cases, and the lymphoproliferative disorder termed Castleman's disease in ~15% of cases. (Rare cases of POEMS have been associated with polyclonal rather than clonal plasma cells; these cases are not plasma cell dyscrasias but rather appear to be caused by the over-activity of non-malignant immune cell responses in chronic infections or autoimmune diseases.) POEMS is an acronym standing for the characteristic signs or symptoms of the syndrome: Polyneuropathy, Organomegaly, Endocrinopathy, Plasma cell disorder (typically, the plasma cell burden is low in POEMS patients), and Skin changes (e.g. hemangioma, hyperpigmentation). The syndrome is defined by the presence of; both of two major criteria, peripheral neuropathy and a clonal plasma cell dyscrasia (increased bone marrow plasma cells in ~67% of cases; ≥1 plasmacytoma in ~33% of cases); at least one other major criteria (Castleman's disease, sclerotic bone lesions, elevated serum levels of the cytokine VEGF); and at least one minor criterion (organomegaly, extravascular volume overload [e.g. ascites, edema, pleural effusion, and/or pericardial effusion], endocrinopathy [i.e. hypogonadism, defects in the hypothalamic–pituitary–adrenal axis], skin changes, papilledema, and/or hematological manifestations [i.e. thrombocytosis or polycythemia]).[28] The monoclonal protein in POEMS patients is typically identified as IgA or IgG which in >95% of cases contains a λ chain that is restricted to either of two members of the V lambda 1 subfamily viz., IGLV1-40*01 and IGLV1-44*01 (there are 29 other members in the V lambda family). That is, the myeloma protein in POEMS is almost invariably a clonal λ light chain variant. Deletion of chromosome 13 and chromosomal translocations but not increases in chromosome number have also been reported to occur in POEMS patients.[29]

Patients with 1 or 2 isolated plasmacytomas have been successfully treated with targeted radiotherapy to obtain relief of symptoms and sometimes complete remission of disease. (Isolated plasmacytomas may regress spontaneously.) Patients with >2 plasmacytomas or symptomatic disseminated disease have been treated with chemotherapy often followed by autologous stem-cell transplantation; these treatments have been found to reduce symptoms of the disease and lead to long-term partial remissions of disease.[28][29] The overall survival of POEMS patients who have been treated for their disease is relatively good for a disease occurring in patients with an average age of 50 years; one estimate of median overall survival is 14 years. POEMS patients evaluated to be in low and intermediate risk groups had ≥>85% survival at 10 years; those in the high risk group had a 40% survival over this time period.[30]

Cryoglobulinemia

Cryoglobulins are proteins, principally immunoglobulins, that circulate in the blood, precipitate at temperatures <37 °C (98.6 °F), and re-solubilize upon restoring physiological blood temperatures. They are made and secreted into the blood as a result of underlying pathological conditions viz., inflammation, infection, or malignancies. Rarely, cryoglobulinemia (i.e. essential cryoglobulinemia) occurs in patients without these or other identifiable conditions. Non-essential cryoglobulonemia is classified into three types. Type 1 cryoglobulinemia (10-25% of cases) involves a circulating myeloma protein, typically IgM or IgG but in rare case reports IgA. The condition is associated with Waldenström macroglobulinemia or multiple myeloma in ~40% of type I cases, the MGUS or smoldering predecessors to these diseases in ~44% of type I cases, and other B cell lymphoproliferative disorders in ~16% of type I cases.[31] Type II cryglobulinemia (50-60% of cases) involves circulating IgM myeloma protein with rheumatoid factor activity and therefore bound to polyclonal IgG and protein components of the blood complement system; hepatitis C virus and, far more rarely, hepatitis B virus or human immunodeficiency virus infections are the major causes of this cryoglobulinemia. Type III cryoglobulinemia (15-30% of cases) involves circulating polyclonal IgM protein with rheumatoid factor activity bound to polyclonal IgG and blood complement components; autoimmune diseases and, less commonly, hepatitis virus C infection or lymphoproliferative disorders are the cause of this type of croglobulinemia. Only types I and II are defined as plasma cell dyscrasias.[32]

Patients suffering type 1 cryoglobulinemia present with symptoms due to cold temperature-induce blood hyperviscosity and consequential interruptions of blood flow, e.g. skin lesions (lower extremity purpuric spots and papules, acrocyanosis, necrosis skin ulcers, livedo reticularis urticaria), peripheral neuropathy, blurred vision, loss of vision, hearing loss, headaches, confusion, transient ischemic attacks, chest pain, heart failure, glomerulonephritis, kidney failure, oral bleeding, and nasal bleeding. Rarely, patients may present with catastrophic decreases in blood flow to vital tissues and require emergency treatment. Symptomatic patients typically exhibit levels of a myeloma protein >5 gram/liter and can be diagnosed by simple observing the temperature-induced, reversible induction of serum precipitate formation. Patients, particularly those with catastrophic presentations, are treated with plasma exchange and/or plasmapharesis to reduce the load of circulating myeloma proteins and relieve acute symptoms. Patients with an overt malignancy are treated with the chemotherapy regimens used for Waldenstroms macroglobulinemia or multiply myeloma; patients with MGUS precursors to these diseases appear less responsive to these chemotherapeutic regimens. These patients as well as patients with overt malignancy may be treated with rituximab (kills normal and malignant B cells that bear the CD20 antigen or the proteasome inhibitor, Bortezomib.[31]

Patients suffering type II (or type III) cryoglobulinemia present with many of the symptoms of type I disease plus those of inflammatory vasculitis. Their treatments are tailored to the underlying infectious, autoimmune, or malignant disease. Type II patients associated with a monoclonal antibody and clonal plasma cells or other types of clonal B cells, are typically treated with regimens used for Walsdenstorms macroglobulonemia or multiple myeloma.[31]

Malignant stage

In the malignant stage of plasma cell dyscrasias, a clearly excessive tumor cell burden causes symptoms and findings predictive of rapid, life-threatening progression of disease. These dyscrasias fall into several distinct categories.

Solitary plasmacytoma

Solitary plasmacytoma is an early stage malignancy with a clinical course that lies between MGUS and multiple myeloma in the spectrum of plasma cell dyscrasias.[5] Solitary plasmacytomas typically present with local symptoms due to the growing mass of plasma cells such as the bone pain or pathologic bone fractures occurring in solitary plasmacytomas of bone or the headache, focal neurological deficits, and cranial nerve palsies occurring in extramedullary plasmacytomas of sellar and parasellar compartments of the brain.[33] Its diagnoses must meet all four of the following criteria: biopsy-proven tumor consisting of clonal plasma cells; no evidence of any other plasmacytomas based on bone survey and MRI (or in place of MRI, CT scan); normal bone marrow examination; and absence end organ damage, CRAB features, or other signs or symptoms of systemic disease attributable to a plasma cell dyscrasia.[5] Blood or urine myeloma proteins are usually undetectable or low in solitary plasmacytomas. Solitary plasmacytoma is a rare disease with an incidence in the USA of <450 cases per year. In a review of 1,691 cases in the US, the median age at diagnosis was 63 with males representing ~60% of all cases. The most common site of plasmacytoma involvement was bone (~58%) followed by upper or lower airway tract (~16%), soft tissue or connective tissue (~5%), central nervous system (~3%), gastrointestinal tract (~3%), skin (~1%), and all other sites (~3%). Overall median survival was 8.12 years with survival decreasing with age from 12.4 years for patients <40 to 5.2 years for patients of 60 years or older.[34] Risk of its recurrence or progression to overt multiple myeloma within 3 years is ~10%.[5]

A subset of solitary plasmacytomas, termed solitary plasmacytoma with minimal bone marrow involvement, has the same criteria for diagnosis as solitary plasmacytoma except that bone marrow examination shows an increase in plasma cells from a normal value of ~0% to 1.5% to >~1.6% but less than 10% of total nucleated cells. While its presentations and findings are similar to solitary plasmacytoma, solitary plasmacytoma with minimal bone marrow involvement is more likely to progress, i.e. it recurs or becomes overt multiple myeloma in 20% to 60% of cases within 3 years. Solitary plasmacytomas associated with 10% or more plasma cells are diagnosed as overt multiple myeloma.[5]

Non-secretory multiple myeloma

Non-secretory multiple myeloma represents a class of plasma cell dyscrasias where no myeloma protein is detected in serum or urine of patients with evidence of increased clonal bone marrow plasma cells and/or multiple plasmacytomas, particularly of the bone but also of soft tissues. While a pre-malignant phase is likely, most new cases of non-secretory multiple myeloma are brought to attention not because of incidental M protein detection which by definition is absent but because of patient symptoms indicative of malignancy possibly of plasma cell origin. The condition has been diagnosed based on biopsy-proved clonal plasma cell tumors and/or the presence in bone marrow of plasma cells at ≥10% of nucleated cells in individuals who have evidence of end organ damage attributable to an underlying plasma cell disorder. These patients typically also show one or more CRAB signs and lack evidence of a myeloma protein as measured by protein electrophoresis and immunofixation. However, more sensitive methods of detecting urinary and serum light chain myeloma proteins using enzyme-linked immunosorbent assays indicate that >60% of cases initially diagnosed as non-secretory multiple myeloma had abnormal levels of either a clonal κ or λ light chain in their urine or serum and therefore were better diagnosed as having light chain multiple myeloma.[35][36][37] Based on the latter definition, non-secretory multiple myeloma represents ~1% of all multiple myeloma cases with formerly diagnosed non-secretory myelomas considered to be cases primarily of light chain multiple myeloma but on occasion "false non-secretors", i.e. cases in which there is evidence of myeloma protein secretion such as renal myeloma protein deposits.[36]

A Mayo Clinic study of 124 patients initially diagnosed as having non-secretory multiple myeloma were later found to be composed of 65% free light chain secretors and 35% true non-secretors. As a group, these patients response to therapy, time to disease recurrence, and overall survival were similar to typical myeloma patients. However, in a subset of patients diagnosed after 2001 and therefore treated with more effective therapy that included autologous stem-cell transplantation, prognosis was significantly better in non-secretory multiple myeloma patients (median survival 8.3 years) compared to typical myeloma patients (median survival 5.4 years). In addition, non-secretory patients exhibited a better prognosis than light chain-secretory patients.[35]

Plasma Cell Myeloma with concomitant chronic Lymphocytic Leukemia/monoclonal B-Cell Lymphocytosis

Multiple myeloma occurring concurrently with chronic Lymphocytic Leukemia or its pre-malignant precursor, monoclonal B-cell lymphocytosis, is an extremely rare condition in which patients evidence findings of the plasma cell dyscrasia plus either one of the cited clonal lymphocytic diseases. Patients are typically elderly (median age of 74, range 42–91 years old) males (51 of 66 case reports) and commonly present with a combination of symptoms related to chronic lymphocytic leukemia symptoms (fatigue, autoimmune hemolytic anemia, enlargements of liver and/or spleen and lymphadenopathy) plus symptoms of multiple myelomas. Patients exhibit two distinct populations of clonal cells in their bone marrow, blood, and/or other tissues: plasma cells, which may have an immature plasmablastic morphology and small lymphocytes, which have a morphology typical of chronic lymphocytic leukemia cells. Patients blood and/or urine evidences a plasma cell-derived myeloma proteins, either IgG, IgA, or free light chain in ~50%, 20%, and 20% of cases, respectively, but may also have a second myeloma protein made by the lymphocytic cells, either an IgM or IgG. Signs and symptoms of chronic lymphocytic leukemia commonly precede those of multiple myeloma, sometimes by years.[38] The relationship between the two clones of cells in this combined disease has not been established although one study suggests that the clonal plasma cells and clonal lymphocytes arise from a common hematological stem cell.[39] In general, patients with plasma cell myeloma with concomitant chronic Lymphocytic Leukemia/monoclonal B-cell Lymphocytosis have been treated with the same regimens used for multiple myeloma patients unless significant complications related to the lymphocytic component of their disease (e.g. autoimmune hemolytic anemia) require treatments used in chronic lymphocytic leukemia. Some patients who lack appreciable symptoms have been followed with no specific treatment of their disease.[38]

Waldenström macroglobulinemia

According to the International Workshop on Waldenström's Macroglobulinemia, the disease is diagnosed in patients that have a serum IgM monoclonal protein and a bone marrow that contains ≥10% of its nucleated cells as lymphoplasmacytic cells. There is no requirement for symptomatic disease, a particular level of IgM protein, or presence of extramedullary (i.e. non-bone) lymphoplasmacytic cell infiltrates. The overall survival for this malignancy at 5 and 10 years among >5,000 patients is 62% and 39%, respectively, with newer treatment regimens anticipated to improve these survival rates in the future.[8]

Multiple myeloma

Multiple myeloma is diagnosed in patients that (except for non-secretory multiple myeloma patients) have a clonal IgG, IgA, IgD, or IgE myeloma protein in their serum and/or a clonal κ or λ light chain in their serum or urine plus either one of two sets of criteria. In the first criteria set, patients must have ≥10% bone marrow clonal plasma cells plus ≥1 of the CRAB criteria; in the second criteria set, patients must have ≥10 bone marrow clonal plasma cells plus ≥1 of the following findings, ≥60% bone marrow clonal plasma cells, a free κ/λ or λ/κ light chain ratio in serum of ≥100 (the involved clonal light chain concentration must be ≥100 milligrams/liter), and/or >1 focal bone lesion on magnetic resonance imaging.[4] The 5 year medium survival of patients with multiple myeloma treated with currently used treatment regiments is 48.5%.[40]

Light chain multiple myeloma

Light chain multiple myeloma is diagnosed in patients who have: a) the criteria for diagnosis of multiple myeloma except having a serum free light chain ratio outside the normal range of 0.26 to 1.65 without evidence of an intact immunoglobulin or free heavy chain; or b) an extreme free light chain ratio, i.e. outside the range of 0.02 to 100 (with the light chain having the lower concentration being present at >10 milligrams/liter) regardless of the stage of their plasma cell dyscrasia.[41] At the time of diagnosis, 30% to 50% of light chain multiple myeloma patients have severe renal dysfunction or kidney failure due to light chain myeloma cast nephropathy or the nephrotoxic effects of free light chains on renal tubular cells. Patients are treated similarly to patients suffering the counterparts those with multiple myeloma except that the focus is treating or preventing kidney damage using chemotherapy to reduce production of the monoclonal light chain and thereby stopping, reversing, or preventing kidney injury.[42]

Plasma cell leukemia

Plasma cell leukemia is a form of multiple myeloma in which significant numbers of typically immature appearing plasma cells, i.e. plasmablasts, circulate in the blood. Very small numbers of plasma cells may reach the circulation in non-IgM multiple myeloma, non-IgM SMM, and, exceptionally, non-IgM MGUS. In these plasma cell dyscrasias, the presence of even very small numbers of circulating plasma cells is a poor prognostic indicator. In plasma cell leukemia, however, circulating plasma cells reach far higher numbers and at these circulating levels are associated with exceptionally poor survival rates. The International Myeloma Working Group has defined the diagnostic criteria for plasma cell leukemia as the presence in blood of >2x109 plasma cells per liter or, alternatively, >20% of nucleated blood cells being plasma cells. More recently, the Group has suggested that values of 0.5x109 or 5%, respectively, may be more appropriate from a therapeutic viewpoint and therefore should be studied as a definitive criterion for the disease.[43] A recent study supported this suggestion in finding that multiple myeloma patients with >5% circulating plasma cells had a prognosis much worse than that for multiple myeloma and similar to that for plasma cell leukemia.[44] Flow cytometry immunophenotyping of blood cells to detect clonal phenotypes of plasma cells seen in multiple myeloma (e.g. the CD138+, CD38+, CD19, CD4+/- phenotype) may be a more sensitive method to enumerate circulating clonal plasma cells and diagnose plasma cell leukemia.[45]

There are two forms of plasma cell leukemia: Primary plasma cell leukemia in which patients without a history of multiple myeloma present with diagnostically high levels of circulating plasma cells and Secondary plasma cell leukemia in which patients with multiple myeloma suffer their dyscrasia's progression by the expansion of large numbers of their malignant plasma cells into the circulation and distant tissues. Historically, primary plasma cell leukemia was more common than the secondary form but with the increased survival of multiple myeloma patients due to new treatment regiments, more cases of secondary plasma cell leukemia are occurring; currently, the two forms occur in approximately equal numbers.[43] Patients with primary plasma cell leukemia present with clinical findings that are less commonly found in multiple myeloma, e.g. they often have hepatomegaly, splenomegaly, lymphadenopathy, nerve and central nervous system defects, bleeding tendencies secondary to thrombocytopenia, and pleural effusions. They are less likely than multiple myeloma patients to have lytic bone lesions. In several studies of patients with either form of plasma cell leukemia, the disease was associated with clonal IgG in 28% to 56% of cases, IgA in 4% to 7% of cases, and a light chain in 23% to 44% of cases; 0-12% of patients had no myeloma protein. Medium survival for primary and secondary plasma cell dyscrasias have been 7–13 months and 2–7 months, respectively, but appear to be improving with new treatment regimens.[18][45]

Heavy chain disease

The four heavy chain diseases are exceedingly rare conditions associated with the production, circulation in blood, and often presence in urine of a free clonal heavy chain with no detected clonal light chains. The heavy chain is non-functional and altered by having deletions, insertions, and point mutations due to somatic mutations in their respective coding genes. Beyond this commonality, however, these diseases have very different clinical differences. Furthermore, each of the heavy chain diseases appears to be due to rare variants of lymphoma and therefore is sometimes regarded as a B cell dyscrasia[11] However, heavy chain diseases are still often classified with plasma cell dyscrasias.[46] The heavy chain diseases are classified as α, γ, and μ heavy chain diseases and are based respectively on >400, 130, and 30-40 case reports as reviewed in a 2014 publication.[11]

α Heavy chain disease

α Heavy chain disease (also termed immunoproliferative small intestinal disease or IPSID, Mediterranean lymphoma, and Seligmann disease) afflicts primarily individuals of Mediterranean, North African, and Middle Eastern descent of lower economic status. Many cases are centered in the Middle East and associated with relatively unsanitary living conditions. The disease usually appears between the ages 10 and 30 and in some cases may be an aberrant immune response to a parasite or other microorganism.[47] The disease commonly effects the gastrointestinal tract leading to signs and symptoms of a malabsorption syndrome or, far less commonly, the respiratory tract with signs and symptoms of respiratory dysfunction. Involved tissues usually include mucosa-associated lymphoid tissues and evidence a histology of lymphoplasmacytoid infiltrates accompanied by large numbers of plasma cells and small lymphocytes. The plasma cells therein express the monoclonal α chain and therefore are clonal in nature and the sole or contributing producer of the α chain myeloma protein. Some 57% to 66% of patients present with disseminated lymphoma, 17% to 36% of patients present with a localized lymphoma, and 9% to 17% of patients lack any evidence of a lymphoplasmacytic neoplasm. A majority of the latter patients have an autoimmune disease or a chronic infection which may be responsible for, or contribute to, production of the α heavy chain. Studies indicated that a sub-set particularly of the digestive form of heavy chain disease is caused by infection. This is based on findings that the majority of α heavy chain disease patients are in the lower economic class living under unsanitary conditions, that gastrointestinal bacterial and parasitic infections have been documented in many of these patients, and that long-term (>6 months) appropriately selected antibiotic therapy has improved the condition in 33% to 71% of patients who are at an early stage of the disease and documented to be infected. However, these patients frequently relapse. Patients resistant to antibiotic trials have been treated with multiple drug chemotherapy to obtain complete remission rates of 64% and an overall 5 year survival of 67%.[11]

γ heavy chain disease

γ Heavy chain disease (also termed Franklin disease or Franklin's disease) presents in three patterns: a) aggressive lymphoma (57% to 66% of cases) associated with constitutional symptoms and in 50% of cases with enlargement of lymph nodes, spleen, and/or liver; b) localized lymphoma (~25% of cases) with lymphoma limited to the bone marrow or an extra-nodal site, usually the skin but sometimes the thyroid gland, parotid gland, oropharyngeal cavity, conjunctiva, or gastrointestinal tract; and c) no lymphoma (9% to 17% of cases) associated typically with a preexistent autoimmune disease but no evidence of lymphoma. Involved lymphoma-infiltrated tissues typically show a mixture of lymphoplamsmacytoid cells, plasma cells, lymphocytes, and sometimes variable numbers of eosinophils and hystiocytes. Treatment of the disease varies with its clinical severity. Patients with aggressive lymphoma have been treated with multiple drug chemotherapy, patients with limited lymphoma have been monitored for disease progression or treated locally (e.g. radiation therapy, surgical removal), and patients with no lymphoma have been monitored for progression in their diseases while being treated for any autoimmune disease that they bear. Spontaneous remissions in γ heavy chain disease have occurred. Regardless of presentation pattern, these patients may have an aggressive or indolent disease with courses ranging from the asymptomatic presence of a stable monoclonal heavy chain in the serum or urine (e.g. MGUS) to a rapid, downhill progression of a few weeks' duration. γ Heavy chain disease survivorship ranged form 1 month to >20 years (medium survival 7.4 years) in a Mayo Clinic study.[11][48]

μ Heavy chain disease

μ Heavy chain disease presents with a picture of a lymphoid neoplasm resembling either chronic lymphocytic leukemia or small lymphocytic lymphoma. This picture includes splenomegaly in virtually all cases, hepatomegaly in ~75% of cases, lymphadenopathy in ~40% of cases, and lytic bone lesions in ~20% of cases. Patients often have hypogammaglobulinemia, increases in urinary free light chains, and a bone marrow containing vacuolated plasma cells or lymphoid cells. Treatment of μ heavy chain disease had varied form observation only in asymptomatic patients to single drug or and multiple drug chemotherapy in symptomatic patients. Survival with this disease varies between <1 month to >10 years with a median survival rate of ~ 2years.[11][47][48]

See also

References

  1. ^ a b c Castillo JJ (2016). "Plasma Cell Disorders". Primary Care. 43 (4): 677–691. doi:10.1016/j.pop.2016.07.002. PMID 27866585.
  2. ^ a b c d e f g h i j k Willrich MA, Murray DL, Kyle RA (2017). "Laboratory testing for monoclonal gammopathies: Focus on monoclonal gammopathy of undetermined significance and smoldering multiple myeloma". Clinical Biochemistry. 51: 38–47. doi:10.1016/j.clinbiochem.2017.05.001. PMID 28479151.
  3. ^ a b c d e van de Donk NW, Mutis T, Poddighe PJ, Lokhorst HM, Zweegman S (2016). "Diagnosis, risk stratification and management of monoclonal gammopathy of undetermined significance and smoldering multiple myeloma". International Journal of Laboratory Hematology. 38 Suppl 1: 110–22. doi:10.1111/ijlh.12504. PMID 27161311. S2CID 21348701.
  4. ^ a b Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, Kumar S, Hillengass J, Kastritis E, Richardson P, Landgren O, Paiva B, Dispenzieri A, Weiss B, LeLeu X, Zweegman S, Lonial S, Rosinol L, Zamagni E, Jagannath S, Sezer O, Kristinsson SY, Caers J, Usmani SZ, Lahuerta JJ, Johnsen HE, Beksac M, Cavo M, Goldschmidt H, Terpos E, Kyle RA, Anderson KC, Durie BG, Miguel JF (2014). "International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma". The Lancet. Oncology. 15 (12): e538–48. doi:10.1016/S1470-2045(14)70442-5. PMID 25439696.
  5. ^ a b c d e f Rajkumar SV (2016). "Updated Diagnostic Criteria and Staging System for Multiple Myeloma". American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Meeting. 35: e418–23. doi:10.14694/EDBK_159009. PMID 27249749.
  6. ^ a b c d e f g Dutta AK, Hewett DR, Fink JL, Grady JP, Zannettino AC (2017). "Cutting edge genomics reveal new insights into tumour development, disease progression and therapeutic impacts in multiple myeloma". British Journal of Haematology. 178 (2): 196–208. doi:10.1111/bjh.14649. PMID 28466550. S2CID 25838916.
  7. ^ a b c d El-Ayoubi A, Wang JQ, Hein N, Talaulikar D (2017). "Role of plasma cells in Waldenström macroglobulinaemia". Pathology. 49 (4): 337–345. doi:10.1016/j.pathol.2017.02.004. PMID 28483372.
  8. ^ a b c d e Abeykoon JP, Yanamandra U, Kapoor P (2017). "New developments in the management of Waldenström macroglobulinemia". Cancer Management and Research. 9: 73–83. doi:10.2147/CMAR.S94059. PMC 5354523. PMID 28331368.
  9. ^ Pandey S, Kyle RA (2013). "Unusual myelomas: a review of IgD and IgE variants". Oncology (Williston Park, N.Y.). 27 (8): 798–803. PMID 24133829.
  10. ^ Ribourtout B, Zandecki M (2015). "Plasma cell morphology in multiple myeloma and related disorders". Morphologie: Bulletin de l'Association des Anatomistes. 99 (325): 38–62. doi:10.1016/j.morpho.2015.02.001. PMID 25899140.
  11. ^ a b c d e f g h Bianchi G, Anderson KC, Harris NL, Sohani AR (2014). "The heavy chain diseases: clinical and pathologic features". Oncology (Williston Park, N.Y.). 28 (1): 45–53. PMID 24683718.
  12. ^ "KIF2B kinesin family member 2B [Homo sapiens (human)] - Gene - NCBI".
  13. ^ "CIDEC cell death inducing DFFA like effector c [Homo sapiens (human)] - Gene - NCBI".
  14. ^ Simeon V, Todoerti K, La Rocca F, Caivano A, Trino S, Lionetti M, Agnelli L, De Luca L, Laurenzana I, Neri A, Musto P (2015). "Molecular Classification and Pharmacogenetics of Primary Plasma Cell Leukemia: An Initial Approach toward Precision Medicine". International Journal of Molecular Sciences. 16 (8): 17514–34. doi:10.3390/ijms160817514. PMC 4581206. PMID 26263974.
  15. ^ a b c Al-Hussain T, Hussein MH, Al Mana H, Akhtar M (2015). "Renal involvement in monoclonal gammopathy". Advances in Anatomic Pathology. 22 (2): 121–34. doi:10.1097/PAP.0000000000000056. PMID 25664947. S2CID 5137203.
  16. ^ Itoh T (2016). "Multiple myeloma developing in a patient with immune thrombocytopenia". [Rinsho Ketsueki] the Japanese Journal of Clinical Hematology. 57 (5): 630–3. doi:10.11406/rinketsu.57.630. PMID 27263790.
  17. ^ a b c d e f g h Mailankody S, Landgren O (2016). "Monoclonal gammopathy of undetermined significance and Waldenström's macroglobulinemia". Best Practice & Research. Clinical Haematology. 29 (2): 187–193. doi:10.1016/j.beha.2016.08.015. PMID 27825465.
  18. ^ a b Bommannan K, Sachdeva MU, Malhotra P, Kumar N, Sharma P, Naseem S, Ahluwalia J, Das R, Varma N, Prakash G, Khadwal A, Srinivasan R, Varma S (2016). "Plasma cell leukemia in North India: retrospective analysis of a distinct clinicohematological entity from a tertiary care center and review of literature". Blood Research. 51 (1): 23–30. doi:10.5045/br.2016.51.1.23. PMC 4828524. PMID 27104188.
  19. ^ Khouri J, Samaras C, Valent J, Mejia Garcia A, Faiman B, Mathur S, Hamilton K, Nakashima M, Kalaycio M (January 2019). "Monoclonal gammopathy of undetermined significance: A primary care guide". Cleveland Clinic Journal of Medicine. 86 (1): 39–46. doi:10.3949/ccjm.86a.17133. PMID 30624183. S2CID 58554384.
  20. ^ a b c d e f Muchtar E, Kumar SK, Magen H, Gertz MA (2017). "Diagnosis and management of smoldering multiple myeloma: the razor's edge between clonality and cancer". Leukemia & Lymphoma. 59 (2): 1–12. doi:10.1080/10428194.2017.1334124. PMID 28592156. S2CID 11195536.
  21. ^ a b Ciocchini M, Arbelbide J, Musso CG (2017). "Monoclonal gammopathy of renal significance (MGRS): the characteristics and significance of a new meta-entity". International Urology and Nephrology. 49 (12): 2171–2175. doi:10.1007/s11255-017-1594-y. PMID 28425076. S2CID 1318744.
  22. ^ Kyle RA, Larson DR, Therneau TM, Dispenzieri A, Melton LJ, Benson JT, Kumar S, Rajkumar SV (2014). "Clinical course of light-chain smouldering multiple myeloma (idiopathic Bence Jones proteinuria): a retrospective cohort study". The Lancet. Haematology. 1 (1): e28–e36. doi:10.1016/S2352-3026(14)70001-8. PMC 4266993. PMID 25530988.
  23. ^ Małyszko J, Kozłowska K, Małyszko JS (2017). "Amyloidosis: A cancer-derived paraproteinemia and kidney involvement". Advances in Medical Sciences. 62 (1): 31–38. doi:10.1016/j.advms.2016.06.004. PMID 28153807.
  24. ^ a b c Nasr SH, Said SM, Valeri AM, Sethi S, Fidler ME, Cornell LD, Gertz MA, Dispenzieri A, Buadi FK, Vrana JA, Theis JD, Dogan A, Leung N (2013). "The diagnosis and characteristics of renal heavy-chain and heavy/light-chain amyloidosis and their comparison with renal light-chain amyloidosis". Kidney International. 83 (3): 463–70. doi:10.1038/ki.2012.414. PMID 23302715.
  25. ^ Elsaman AM, Radwan AR, Akmatov MK, Della Beffa C, Walker A, Mayer CT, Dai L, Nativ S, Rygg M, Atsali E, Saijo K, Ogdie AR, Srinivasulu N, Fathi N, Schumacher HR, Pessler F (2013). "Amyloid arthropathy associated with multiple myeloma: a systematic analysis of 101 reported cases". Seminars in Arthritis and Rheumatism. 43 (3): 405–12. doi:10.1016/j.semarthrit.2013.07.004. PMID 23992800.
  26. ^ a b Said SM, Sethi S, Valeri AM, Leung N, Cornell LD, Fidler ME, Herrera Hernandez L, Vrana JA, Theis JD, Quint PS, Dogan A, Nasr SH (2013). "Renal amyloidosis: origin and clinicopathologic correlations of 474 recent cases". Clinical Journal of the American Society of Nephrology. 8 (9): 1515–23. doi:10.2215/CJN.10491012. PMC 3805078. PMID 23704299.
  27. ^ "UpToDate".
  28. ^ a b Warsame R, Yanamandra U, Kapoor P (2017). "POEMS Syndrome: an Enigma". Current Hematologic Malignancy Reports. 12 (2): 85–95. doi:10.1007/s11899-017-0367-0. PMID 28299525. S2CID 31324035.
  29. ^ a b Dispenzieri A (2015). "POEMS syndrome: update on diagnosis, risk-stratification, and management". American Journal of Hematology. 90 (10): 951–62. doi:10.1002/ajh.24171. PMID 26331353. S2CID 5167507.
  30. ^ Kourelis TV, Dispenzieri A (2017). "Validation of a prognostic score for patients with POEMS syndrome: a mayo clinic cohort". Leukemia. 31 (5): 1251. doi:10.1038/leu.2017.68. PMID 28280263. S2CID 2521267.
  31. ^ a b c Retamozo S, Brito-Zerón P, Bosch X, Stone JH, Ramos-Casals M (2013). "Cryoglobulinemic disease". Oncology (Williston Park, N.Y.). 27 (11): 1098–1105, 1110–6. PMID 24575538.
  32. ^ Ghetie D, Mehraban N, Sibley CH (2015). "Cold hard facts of cryoglobulinemia: updates on clinical features and treatment advances". Rheumatic Disease Clinics of North America. 41 (1): 93–108, viii–ix. doi:10.1016/j.rdc.2014.09.008. PMID 25399942.
  33. ^ Lee J, Kulubya E, Pressman BD, Mamelak A, Bannykh S, Zada G, Cooper O (2017). "Sellar and clival plasmacytomas: case series of 5 patients with systematic review of 65 published cases". Pituitary. 20 (3): 381–392. doi:10.1007/s11102-017-0799-5. PMC 5429193. PMID 28251542.
  34. ^ Thumallapally N, Meshref A, Mousa M, Terjanian T (2017). "Solitary plasmacytoma: population-based analysis of survival trends and effect of various treatment modalities in the USA". BMC Cancer. 17 (1): 13. doi:10.1186/s12885-016-3015-5. PMC 5216567. PMID 28056880.
  35. ^ a b Chawla SS, Kumar SK, Dispenzieri A, Greenberg AJ, Larson DR, Kyle RA, Lacy MQ, Gertz MA, Rajkumar SV (2015). "Clinical course and prognosis of non-secretory multiple myeloma". European Journal of Haematology. 95 (1): 57–64. doi:10.1111/ejh.12478. PMID 25382589. S2CID 22409325.
  36. ^ a b Dupuis MM, Tuchman SA (2016). "Non-secretory multiple myeloma: from biology to clinical management". OncoTargets and Therapy. 9: 7583–7590. doi:10.2147/OTT.S122241. PMC 5171196. PMID 28008276.
  37. ^ Heaney JL, Campbell JP, Griffin AE, Birtwistle J, Shemar M, Child JA, Gregory WM, Cairns DA, Morgan G, Jackson G, Drayson MT (2017). "Diagnosis and monitoring for light chain only and oligosecretory myeloma using serum free light chain tests" (PDF). British Journal of Haematology. 178 (2): 220–230. doi:10.1111/bjh.14753. PMID 28573706. S2CID 38301916.
  38. ^ a b Alley CL, Wang E, Dunphy CH, Gong JZ, Lu CM, Boswell EL, Burchette J, Lagoo AS (2013). "Diagnostic and clinical considerations in concomitant bone marrow involvement by plasma cell myeloma and chronic lymphocytic leukemia/monoclonal B-cell lymphocytosis: a series of 15 cases and review of literature". Archives of Pathology & Laboratory Medicine. 137 (4): 503–17. doi:10.5858/arpa.2011-0696-OA. PMID 23544940.
  39. ^ Trudel S, Ghamlouch H, Dremaux J, Delette C, Harrivel V, Marolleau JP, Gubler B (2016). "The Importance of an In-depth Study of Immunoglobulin Gene Rearrangements When Ascertaining the Clonal Relationship between Concomitant Chronic Lymphocytic Leukemia and Multiple Myeloma". Frontiers in Immunology. 7: 625. doi:10.3389/fimmu.2016.00625. PMC 5187371. PMID 28082975.
  40. ^ Bustoros M, Mouhieddine TH, Detappe A, Ghobrial IM (2017). "Established and Novel Prognostic Biomarkers in Multiple Myeloma". American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Meeting. 37: 548–560. doi:10.14694/EDBK_175175. PMID 28561668.
  41. ^ Raza S, Safyan RA, Rosenbaum E, Bowman AS, Lentzsch S (2017). "Optimizing current and emerging therapies in multiple myeloma: a guide for the hematologist". Therapeutic Advances in Hematology. 8 (2): 55–70. doi:10.1177/2040620716680548. PMC 5298389. PMID 28203342.
  42. ^ Tessenow H, Holzvogt M, Holzvogt B, Andrea M, Heyn S, Schliwa T, Schwarz M, Zehrfeld T, Becker C, Pfrepper C, Franke GN, Krahl R, Jentzsch M, Leiblein S, Schwind S, Bill M, Vucinic V, Lange T, Niederwieser D, Pönisch W (2017). "Successful treatment of patients with newly diagnosed/untreated light chain multiple myeloma with a combination of bendamustine, prednisone and bortezomib (BPV)". Journal of Cancer Research and Clinical Oncology. 143 (10): 2049–2058. doi:10.1007/s00432-017-2439-x. PMID 28534173. S2CID 22739588.
  43. ^ a b Fernández de Larrea C, Kyle RA, Durie BG, Ludwig H, Usmani S, Vesole DH, Hajek R, San Miguel JF, Sezer O, Sonneveld P, Kumar SK, Mahindra A, Comenzo R, Palumbo A, Mazumber A, Anderson KC, Richardson PG, Badros AZ, Caers J, Cavo M, LeLeu X, Dimopoulos MA, Chim CS, Schots R, Noeul A, Fantl D, Mellqvist UH, Landgren O, Chanan-Khan A, Moreau P, Fonseca R, Merlini G, Lahuerta JJ, Bladé J, Orlowski RZ, Shah JJ (2013). "Plasma cell leukemia: consensus statement on diagnostic requirements, response criteria and treatment recommendations by the International Myeloma Working Group". Leukemia. 27 (4): 780–91. doi:10.1038/leu.2012.336. PMC 4112539. PMID 23288300.
  44. ^ Granell M, Calvo X, Garcia-Guiñón A, Escoda L, Abella E, Martínez CM, Teixidó M, Gimenez MT, Senín A, Sanz P, Campoy D, Vicent A, Arenillas L, Rosiñol L, Sierra J, Bladé J, de Larrea CF (2017). "Prognostic impact of circulating plasma cells in patients with multiple myeloma: implications for plasma cell leukemia definition". Haematologica. 102 (6): 1099–1104. doi:10.3324/haematol.2016.158303. PMC 5451342. PMID 28255016.
  45. ^ a b Gonsalves, Wilson I. (2017-04-10). "Primary Plasma Cell Leukemia: A Practical Approach to Diagnosis and Clinical Management". American Journal of Hematology / Oncology. 13 (3).
  46. ^ Willrich MA, Katzmann JA (2016). "Laboratory testing requirements for diagnosis and follow-up of multiple myeloma and related plasma cell dyscrasias". Clinical Chemistry and Laboratory Medicine. 54 (6): 907–19. doi:10.1515/cclm-2015-0580. PMID 26509779. S2CID 2706697.
  47. ^ a b "Heavy Chain Diseases - Hematology and Oncology".
  48. ^ a b "UpToDate".

External links

plasma, cell, dyscrasias, also, termed, plasma, cell, disorders, plasma, cell, proliferative, diseases, spectrum, progressively, more, severe, monoclonal, gammopathies, which, clone, multiple, clones, malignant, malignant, plasma, cells, sometimes, association. Plasma cell dyscrasias also termed plasma cell disorders and plasma cell proliferative diseases are a spectrum of progressively more severe monoclonal gammopathies in which a clone or multiple clones of pre malignant or malignant plasma cells sometimes in association with lymphoplasmacytoid cells or B lymphocytes over produce and secrete into the blood stream a myeloma protein i e an abnormal monoclonal antibody or portion thereof The exception to this rule is the disorder termed non secretory multiple myeloma this disorder is a form of plasma cell dyscrasia in which no myeloma protein is detected in serum or urine at least as determined by conventional laboratory methods of individuals who have clear evidence of an increase in clonal bone marrow plasma cells and or evidence of clonal plasma cell mediated tissue injury e g plasmacytoma tumors Here a clone of plasma cells refers to group of plasma cells that are abnormal in that they have an identical genetic identity and therefore are descendants of a single genetically distinct ancestor cell Plasma cell dyscrasiaOther namesPlasma cell proliferative diseasesSpecialtyHematology oncologyAt one end of this spectrum of hematological disorders detection of one of these myeloma proteins in an individual s blood or urine is due to a common and clinically silent disorder termed MGUS i e monoclonal gammopathy of undetermined significance At the other end of this spectrum detection of the myeloid protein is due to a hematological malignancy i e multiple myeloma Waldenstrom macroglobulinemia or other B cell associated neoplasm that has developed often in a stepwise manner from their MGUS precursors 1 2 The clinical importance of understanding this spectrum of diseases is that it can be used to a advise individuals on the likelihood of their condition progressing to a malignant phase b monitor individuals for the many complications that may occur at any stage of the dyscrasias so that they can be treated to avoid or reduce their clinical impacts and c monitor patients for transitions to malignancy so that the malignancy can be treated at an early stage when treatment results are best 3 Unless otherwise noted the advice and monitoring given here are those recommended by the International Myeloma Working Group in 2014 4 and updated in 2016 5 Contents 1 Clonal plasma cells 2 Myeloma protein toxicity 3 Stages 3 1 MGUS stage 3 1 1 Non IgM MGUS 3 1 2 IgM MGUS 3 1 3 Light chain MGUS 3 1 4 Monoclonal gammopathy of renal significance 3 2 Smoldering multiple myeloma stage 3 2 1 Non IgM SMM 3 2 2 Smoldering Waldenstrom macroglobulinemia 3 2 3 Light chain SMM 3 3 Paraneoplastic complications 3 3 1 Amyloidosis 3 3 1 1 AL Amyloidosis 3 3 1 2 AH Amyloidosis 3 3 2 POEMS syndrome 3 3 3 Cryoglobulinemia 3 4 Malignant stage 3 4 1 Solitary plasmacytoma 3 4 2 Non secretory multiple myeloma 3 4 3 Plasma Cell Myeloma with concomitant chronic Lymphocytic Leukemia monoclonal B Cell Lymphocytosis 3 4 4 Waldenstrom macroglobulinemia 3 4 5 Multiple myeloma 3 4 6 Light chain multiple myeloma 3 4 7 Plasma cell leukemia 4 Heavy chain disease 4 1 a Heavy chain disease 4 2 g heavy chain disease 4 3 m Heavy chain disease 5 See also 6 References 7 External linksClonal plasma cells EditPlasma cells are key effector elements of the adaptive immune system They contribute to immunity by making antibodies that bind with and thereby initiate the process of neutralizing specific antigens that usually are found on the surface of invading pathogens and foreign substances Plasma cells develop from B lymphocytes i e B cells which are stimulated to undergo this maturational development by T lymphocytes during the latter cells processing of these antigens As they are stimulated to become plasma cells B cells refashion parts of their genome in efforts to create a new gene that encodes a functional antibody In humans antibodies are composed of two identical heavy chains which are of the gamma g alpha a epsilon e delta d or mu m subtypes and two identical light chains which are of the kappa k or lambda l subtypes Antibodies are classified as IgG IgA IgE IgD and IgM based on their being made up of g a e d or m heavy chains respectively Formation of the genes that make these antibodies requires B cells and or their descendent plasma cells to mutate break and recombine various genes at the immunoglobulin heavy chain antigen binding locus on the long i e q arm of human chromosome 14 at position 32 33 notated as 14q32 33 and the immunoglobulin light chain antigen binding locus on the q arm of chromosome 22 at position 11 2 i e 22 q11 2 by processes termed V D J recombination somatic hypermutation and immunoglobulin class switching These genomic changes can go awry by placing a gene that controls cell growth an or survival adjacent to a normally highly active antibody gene promoter and or by causing the formation of extra chromosomes see trisomy or chromosomes with large deletions that result in the overexpression or under expression respectively of genes that control cell growth and or survival In consequence of these primary genomic changes an expanding clone of cells develops overproduces and secretes a monoclonal IgM IgG IgA IgE or IgD antibody a k or l light chain an a g or m heavy chain or very rarely fragments of these proteins and may accumulate secondary genomic changes that cause them to become malignant The overproduced monoclonal proteins termed myeloma proteins commonly circulate in blood may accumulate in urine and are the hallmarks of plasma cell dyscrasias including their most malignant forms viz multiple myeloma light chain multiple myeloma and plasma cell leukemia 6 7 8 IgG secretory IgA secretory and light chain secretory multiple myeloma represent 52 21 and 16 respectively of all multiple myeloma cases these myelomas are associated with various types of chromosomal aberrancies and mutations IgD secretory multiple myeloma occurs in only 1 to 2 of multiple myeloma cases and is commonly associated with somatic mutations in the gene encoding the gV i e variable region of the monoclonal antibody IgE secretory multiple myeloma has been reported in lt 50 cases as of 2013 and is characteristically associated with translocations between the q arms of chromosome 11 and 14 i e t 11 14 q13 q32 translocations 9 In other cases plasma cells and or lymphoplasmacytoid cells a type of B cell resembling and possible precursor to plasma cells suffer other kinds of mutations that lead to the production of an IgM myeloma protein Overproduction of this myeloma protein may progress to a different form of plasma cell lymphoplasmacytoid cell malignancy Waldenstrom macroglobulinaemia Genetic mutations thought to be involved in the development and or progression of the latter disease include the L265P mutation in the MYD88 gene found in gt 90 of Waldenstrom macroglobulinaemia patients as well as various mutations in the CXCR gene found in 27 to 40 of Waldenstrom macroglobulinaemia patients 6 7 10 11 The clonal plasma cells involved in plasma cell dyscrasias exhibit a high degree of genetic instability For example the clonal plasma cell population formed by initial genetic alterations that lead to multiple myeloma contains cells that develop further genetic changes that enhance their survival proliferation tissue injuring and metastatic capacities This allows the new cell clones to crowd out older cell clones and thereby establish a more malignant disease Repetition of such genetic changes underlie the evolution of a clinically silent plasma cell dyscrasia to an overt malignancy 6 7 11 The progressive genetic changes in clonal plasma cells include accumulating numerous single nucleotide polymorphisms increases and decreases in gene and chromosome copy numbers and chromosomal translocations Genes affected include those regulating genome stability itself e g KIF2B 12 as well as cellular activation proliferation and apoptosis e g CIDEC 13 TP52 ATM KRAS NRAS Wnt and NF kB In the most malignant form of plasma cell dyscrasias primary plasma cell leukemia the plasma cell population contains gt 1900 distinct DNA alterations in gt 600 genes 14 In general the plasma cell dyscrasias are defined by 1 the presence of these genetically unstable clonal plasma cells lymphoplasmacytoid cells or B cells infiltrating the bone marrow or forming distinct masses in bone and or other tissues as defined by biopsy of involved tissues and 2 the presence of these cells myeloma proteins i e intact monoclonal antibody free light chain free heavy chain shortened version of these proteins or any combination of these proteins in blood and or urine as defined by various types of gel electrophoresis Obviously the latter criterion does not apply to the rare cases of true non secretory myeloma 6 Myeloma protein toxicity EditMyeloma proteins form as a result of gene mutations rather than physiological gene remodeling responses to an instigating foreign antigen typically these proteins are non functional However they sometimes cause serious tissue damage with the kidney being a particularly vulnerable target The toxic effects of monoclonal proteins may occur at early stages in the plasma cell dyscrasia spectrum and require treatment independently of the mass or tissue destructive effects of the myeloma protein producing cells Myeloma protein toxicities include Monoclonal free light chains free heavy chains or a mixture of these chains can deposit in the kidney and other organs to cause systemic monoclonal immunoglobulin deposition disease free k or l light chains can deposit selectively in the kidney s proximal tubule to cause light chain proximal tubulopathy or in the kidneys distal tubule to cause light chain myeloma cast nephropathy and various myeloma proteins can deposit selectively in the kidneys glomeruli to cause various forms of organized deposit and non organized deposit glomerulonephritis diseases 15 Free k or l light chains can aggregate with each other to cause extracellular amyloid deposits and a disease termed amyloidosis in which the deposits injure and ultimately lead to the failure of such organs as the kidney heart liver stomach and intestines 15 these deposits can also cause peripheral and autonomic neuropathies 1 IgM myeloma proteins or in rare cases other myeloma proteins such as IgA free k light chains or free l light chains may increase blood viscosity deposit in peripheral blood vessels and thereby cause vascular occlusion and gangrene of the extremities in the syndrome termed cryoglobulinemia 7 Monoclonal IgM myeloma proteins operating through their effects on increasing blood hyperviscosity can reduce blood flow to the central nervous system to cause blurred vision headaches vertigo ataxia and cold induced hemolytic anemia 8 IgM IgG and to lesser extents k and l free light chain myeloma proteins can cause Immune thrombocytopenic purpura with extensive bleeding tendencies 16 Stages EditMGUS stage Edit Monoclonal gammopathy of undetermined significance MGUS is defined as the presence in the blood or urine of a monoclonal antibody antibody heavy chain or antibody light chain in a person lacking symptoms or signs of a more serious plasma cell dyscrasia The condition is typically discovered as an incidental finding when serum protein electrophoresis is done for various reasons unrelated to plasma cell dyscrasias Protein electrophoresis generally detects one of the following patterns of monoclonal myeloma protein spikes representing a intact IgG IgA IgE IgE or IgM b intact IgG IgA IgE IgD or IgM plus high concentrations of a free i e not bound to a heavy chain k or l light chain c a free k chain in great excess of a l chain or a free l chain in great excess of a k chain and d free g d or m heavy chains unbound to a light chain free a and e heavy chain myeloma protein spikes have not been reported Among MGUS cases expressing an intact antibody 70 15 12 and 3 express either IgG IgM IgA or two of these M proteins respectively with or without excessive levels of a light chain these cases represent 80 of all MGUS About 20 of MGUS cases express either k or l light chains As a group these MGUS findings occur more commonly in men and are 2 fold more common in individuals of African descent than Caucasians 2 17 MGUS cases expressing free g d or m heavy chains are extremely rare 17 11 MGUS is categorized into the following sub types based upon the identity and levels of the myeloma proteins detected as well as the prognoses for progressive disease indicated by these myeloma protein findings citation needed Non IgM MGUS Edit Main article Monoclonal gammopathy of undetermined significance Non IgM MGUS commonly termed MGUS is diagnosed in individuals who exhibit a serum IgG IgD IgA or IgE monoclonal protein with or without increased levels of blood and or urine free k or l light chains These patients typically also show small increases in bone marrow plasma cells Further requirements for the diagnosis of non IgM MGUS are a bone marrow clonal plasma cells lt 10 of total nucleated cells b absence of any of the four CRAB criteria CRAB criteria are C Calcium serum levels gt 1 milligram deciliter above normal values and or a serum level gt 11 milligram deciliter R Renal insufficiency as defined by a glomerular filtration rate lt 40 milliliter minute and or a serum creatinine gt 2 gram deciliter due to myeloma protein induced kidney damage A Anemia as defined by a blood hemoglobin level gt 2 gram deciliter below normal and or lt 10 gram deciliter due to the plasma cell dyscrasia rather than e g iron deficiency or blood loss B Bone lesions i e 1 lytic i e bone re adsorbing bone lesion due to a plasmacytoma as detected by skeletal radiography computed tomography or positron emission tomography computed tomography c no evidence of a plasmacytoma in bone or soft tissues of amyloidosis or of another plasma cell disorder d a ratio of free serum light chains i e free k l or l k light chain ratio less than 100 providing that the higher light chain concentration is gt 100 milligram liter and e a circulating blood plasma cell absolute count of lt 2x109 and or lt 20 of total circulating nucleated cells 2 18 19 Presence of any one of the latter findings indicates that the plasma cell dyscrasia has progressed beyond the MGUS stage citation needed Non IgM MGUS is a relatively stable condition afflicting 3 of people aged 50 and 5 of people aged 70 on average it progresses to multiple myeloma at a rate of 0 5 1 cases per year as defined in studies following patients over a 25 year period A study conducted by the Mayo Clinic found that MGUS associated with non IgG myeloma proteins or with serum myeloma protein levels greater than 15 gram liter had a greater risk per year of progression to multiple myeloma A Spanish study group found that MGUS patients demonstrating aneuploidy i e abnormal number of chromosomes in bone marrow cells or gt 95 of resident bone marrow plasma cells that are clonal in nature also have a greater risk per year of progression to myeloma 6 In a more recent study MGUS patients that had the presence of none 1 2 or 3 of the three following risk factors serum M protein levels gt 15 gram liter a non IgG isotype and abnormal free light chain ratios had 5 32 37 and 58 chances respectively of progressing to multiple myeloma within 20 years In another study MGUS patients with none 1 or 2 of the following risk factors gt 95 of bone marrow plasma cells that are clonal in nature and a 10 or greater rise in the levels of their monoclonal proteins within 3 years had 2 16 and 72 risks respectively of progression within 7 years 3 However estimates on the risks of progression for some of these parameters are tentative and subject to change For example the IgA form of MGUS while once considered to have a poorer prognosis than IgG MGUS was found to have a prognosis similar to IgG MGUS in a more recent study 20 IgM MGUS Edit Main article Waldenstrom macroglobulinemia While traditionally classified as such it is not clear that IgM MGUS is a clonal plasma cell dyscrasia IgM MGUS involves an increase in a B cell derivative with morphological features of both plasma cells and lymphocytes viz lymphoplasmacytic cells Studies indicate that both plasma cells and lymphoblastic cells infiltrate involved tissues and that one or perhaps both cell types harbor mutations in a the MYD88 gene 20 in IgM MGUS and gt 90 in IgM related malignancies almost all of which are L265P mutations i e changing leucine to proline at the 265th amino acid position of the MYK88 protein thereby causing the protein to be continuously active in stimulating the same cell activating pathways that Toll like receptors activate intermittently and on a physiologically basis b the CXCR4 gene 8 in IgM MGUS 25 in IgM related malignancies and c increased gene copy number due to chromosomal rearrangements 36 in IgM MGUS 82 in IgM related malignancies 17 It is clear that each cell type contributes to different features of IgM malignancies but not clear that clonal plasma cells are critical to the development or progression of IgM MGUS 17 In all events IgM MGUS is diagnosed in individuals who have serum IgM levels less than 30 gram liter have less than 10 of nucleated bone marrow cells with the lymphoplasmacytic morphology and have no symptoms or findings of end organ dysfunction attributed to Waldenstrom macroglobulinemia such as anemia decreases in any white blood cell count cold agglutinin disease hyperviscosity of blood lymphadenopathy hepatomegaly splenomegaly peripheral neuropathy cryoglobulinemia or constitutional symptoms 2 8 17 There may be a modest increase in the incidence of IgM MGUS in people of African descent A study of 213 individuals diagnosed with IgM MGUS found that 10 at 5 years and 24 in 15 years progressed to more serious IgM related diseases including non Hodgkin lymphoma Waldenstorm s macroglobulinemia systemic amyloidosis and chronic lymphocytic leukemia A second long term study of 116 individuals with IgM MGUS found a 15 fold increased risk of progressing to a lymphoid malignancy mostly to Waldenstorm s macroglobulinemia In general progression to one of these malignant outcomes occurs at a rate of 2 to 3 per year Individuals with higher serum IgM or lower serum albumin levels progress at faster rates than those with normal levels of these parameters 17 Light chain MGUS Edit Individuals diagnosed with light chain MGUS typically do not express detectable levels of an IgG IgA IgD IgE or IgM intact myeloma protein in their blood Rather they overexpress a monoclonal aberrant free k i e kappa or l i e lambda immunoglobulin light chain For diagnosis the k and l free light chains are quantified by immunological methods and the ratio of k to l light chains is used to detect unbalanced light chain synthesis that is indicative of a monoclonal light chain plasma cell dyscrasia Light chain MGUS is defined as a disorder in which a serum k to l free light chain ratio falls outside the normal range of 0 26 1 65 mean 0 9 provided that it is not associated with a any of the CRAB criteria b a bone marrow plasma cell count of 10 or a higher percentage of nucleated cells c evidence of amyloid deposition see Light chain deposition disease and d an accumulation of 0 5 or more grams of the monoclonal light chain in the urine over a 24 hour period As so defined light chain MGUS comprises 19 of all MGUS cases occurs in 0 8 of the general population and progresses to light chain multiple myeloma at the very slow rate of 0 3 cases per 100 years 2 6 3 Some early studies have reported that a very rapid rate of progression occurs in light chain MGUS patients who have free light chain k l or l k ratios equal to or greater than 100 i e and k l ratio outside of 0 02 to 100 About 80 of individuals bearing these light chain ratios were found to progress to light chain multiple myeloma within 2 years In consequence these individuals were recommended for being diagnosed and treated as having light chain multiple myeloma 3 However two more recent studies reported a 2 year progression rate for these patients of 64 and 30 It is therefore suggested that the diagnosis of light chain multiple myeloma based solely on a free k l light chain ratio of 0 02 to 100 may be premature 20 Monoclonal gammopathy of renal significance Edit Monoclonal gammopathy of renal significance or MGRS designates any MGUS disorder that has a clinically significant impact on renal function MGRS can be caused by the deposition of a monoclonal immunoglobulin in and consequent injury to the kidneys The diagnosis of this form of MGRS is made based on the presence of 1 a disorder meeting the criteria for MGUS b decreased kidney function as evidence by e g a Glomerular filtration rate of lt 40 and c biopsy confirmed or suspicion of cast nephropathy glomerulonephritis of other morphological expressions of clonal immunoglobulin induced kidney injury Increased excretion of a urinary monoclonal light chain typically gt 0 5 gram day which suggests the presence of a particularly severe form of kidney injury myeloma cast nephropathy supports but is not a requirement for the diagnosis of MGRS 5 15 21 The disorder can also be caused by a monoclonal immunoglobulin that acts as an autoantibody that activates the blood complement system to cause complement related kidney injury This form of MGRS is usually associated with other syndromes like glomerulopathy associated with a monoclonal immunoglobulin or C4 dense deposit disease associated with a monoclonal immunoglobulin Diagnosis depends or identifying these other syndromes and the identification of complement components on kidney biopsy Regardless of the exact pathophysiology causing monoclonal immunoglobulin induced kidney injury MGRS has a greater morbidity and mortality than other forms of MGUS Since renal dysfunction usually improves with therapy directed at the underlying plasma cell dyscrasia MGRS may warrant treatment even when other parameters of plasma cell dyscrasia severity e g low levels of serum monoclonal immunoglobulin and bone marrow plasma cells suggest the presence of minimal non malignant disease 21 Smoldering multiple myeloma stage Edit Main article Smoldering multiple myeloma Smoldering multiple myeloma or SMM also termed smoldering myeloma is the next stage following MGUS in the spectrum of plasma cell dyscrasias While still considered a pre malignant condition its chances of progressing to a malignant plasma cell dyscrasia are generally greater than that for MGUS 20 SMM consists of the following subtypes which represent progression of their corresponding MGUS subtypes citation needed Non IgM SMM Edit Non IgM SMM also termed IgG and IgA SMM because of the rarity of IgD and IgE SMM is diagnosed in asymptomatic individuals based on criteria identical to those listed above for Non IgM MGUS except that their intact IgG or IgA myeloma protein levels are equal to or greater than 30 grams liter rather than 15 grams liter their bone marrow shows plasma cells comprise between 10 and lt 60 rather than lt 10 of nucleated cells and or their 24 hour urine contains 0 5 gram or greater levels of Bence Jones i e light chain myeloma proteins individuals must also lack evidence of more recently established multiple myeloma defining criteria viz CRAB features amyloidosis more than one solitary plasmacytoma and or serum or urine free light chain k to l or l to k ratios of 100 or greater 2 20 Overall the risk of Non IgM SMM progressing to multiple myeloma is 10 per year for the first 5 years but falls off sharply to 3 per year for the next 5 years and thereafter to 1 per year 2 Smoldering Waldenstrom macroglobulinemia Edit Smoldering Waldenstrom macroglobulinemia is diagnosed in asymptomatic individuals that have a serum IgM level 30 gram liter and or a bone marrow lymphoplasmacytoid cell infiltrate gt 10 of total nucleated cells These cases should have no symptoms or findings of end organ dysfunction attributed to Waldenstrom macroglobulinemia such as anemia decreases in any white blood cell count cold agglutinin disease hyperviscosity of blood lymphadenopathy hepatomegaly splenomegaly peripheral neuropathy cryoglobulinemia or constitutional symptoms 17 As determined by a Mayo Clinic study of 48 individuals smoldering Waldenstrom macroglobulinemia s risk of progression to Waldenstrom macroglobulinemia is estimated to be 12 per year and then falling of sharply for at least the next 5 years to 2 per year In this study the only factor predictive of a more rapid progress was a finding of anemia hemoglobin level lt 115 grams liter During a 15 year follow up the Clinic subsequently reported that patients progressed to Waldenstrom macroglobulinemia amyloidosis or a related IgM associated neoplasm at a rate of 6 39 59 and 68 after the first third fifth and tenth year respectively However the Southwest Oncology Group in a study on 231 individuals reported that the smoldering disease progressed to overt Waldenstrom macroglobulinemia over 9 years in only 26 of cases 2 8 17 Light chain SMM Edit Light chain smoldering multiple myeloma light chain SMM was previously termed idiopathic Bence Jones proteinuria The condition is currently diagnosed in asymptomatic individuals who have a 24 hour urinary Bence Jones i e light chain myeloma protein level that is gt 0 5 grams and or bone marrow plasma cells that are 10 to lt 60 of nucleated cells These individuals must also lack detectable IgG IgA IgD IgE or IgM myeloma proteins in sera have a free k l or l k light chain ratio outside of 0 26 to 1 65 range but less than 100 and or have no evidence for the presence of any one of the CRAB criteria amyloidosis or end organ damage attributable to the myeloma proteins or plasma cells 2 3 6 In a Mayo clinic study of 101 individuals with light chain SMM the cumulative probability of progression to active multiple myeloma or light chain amyloidosis in patients with light chain SMM was 28 45 and 56 after 5 10 and 15 years respectively The major risk factors for progression were the level of urinary excretion of M protein percentage of bone marrow plasma cells and immunoparesis i e reduced serum levels of intact immunoglobulins 2 22 Paraneoplastic complications Edit Serious and potentially life threatening paraneoplastic complications can occur in plasma cell dyscrasias regardless of tumor cell burden myeloma protein levels or the presence of other criteria suggesting the dyscrasia has entered a malignant phase Many of these complications are caused by the tissue destructive effects of the myeloma proteins are predictive of a rapidly progressive disease and require chemotherapeutic or other treatments directed at lowering the burden of the myeloma protein producing cells The serious paraneoplasitic diseases that complicate the plasma cell dyscrasias and may require such treatments include the following citation needed Amyloidosis Edit Main article Amyloidosis Main article Light chain deposition disease Amyloidosis is a general term for a protein misfolding syndrome that involves the deposition of a low molecular weight beta pleated sheet containing protein in extracellular tissues These proteins normally circulate in the blood but may undergo conformational changes that cause them to auto aggregate along their beta pleated sheets to become insoluble and form fibril deposits in and outside of the circulation These deposits disrupt tissue architecture and in the case of light chains directly injure cells thereby causing potentially cataclysmic organ failures There are 31 types of circulating proteins that can become misfolded and lead to distinctly different types of amyloidosis among these myeloma proteins particularly free light chains are the predominant cause of the disease 20 23 Increases in the levels of free k or l light chains are a common feature of plasma cell dyscrasias These increases occur in 40 of IgM MGUS IgM SMM and Waldenstroms macroglbulonemia cases 60 to 70 of non secretory multiple myelom cases 90 to 95 of intact immunoglobulin multiple myeloma cases and by definition 100 of light chain multiple myeloma cases 2 There are two different types of plasma cell dyscrasia associated amyloidosis syndromes amyloid light chain amyloidosis AL amyloidosis in which amyloid deposits consist of free light chains and amyloid heavy chain amyloidosis AH amyloidosis in which amyloid deposits contain only free heavy chains The deposits in a third type AHL amyloidosis consists of both free light chains and free heavy chains AHL amyloidosis is here as in some recent reports grouped with AH amyloidosis 24 AL Amyloidosis Edit Main article AL Amyloidosis Main article Light chain deposition disease AL amyloidosis can occur at any stage in the plasma cell dyscrasia spectrum Typically patients developing this type of amyloidosis have had excess k or l free light chains in their urine for years before diagnosis At diagnosis however they typically have a relatively small plasma cell burden bone marrow plasma cells lt 5 to 7 of total nucleated cells and in only lt 5 to 10 of cases do other findings indicate the presence of a malignant condition i e definitive signs of multiple myeloma Waldenstrom macroglobulinemia or chronic lymphocytic leukemia associated with over production of a clonal light chain Nonetheless these individuals often evidence serious involvement of the kidney proteinuria nephrotic syndrome or heart restrictive cardiomyopathy arrhythmias in 70 or 60 of cases respectively and of dysfunction in the peripheral nervous system numbness paresthesias or autonomic nervous system orthostatic hypotension in 20 or 15 of cases respectively They may also exhibit evidence of liver involvement liver failure increases in circulating liver enzymes bleeding due to factor X deficiency gastrointestinal track deficiencies malabsorption and amyloid deposition in surface tissues macroglossia shoulder pad masses cutaneous nodules Arthritis in multiple joints often manifested before diagnosis is also a common feature of AL amyloidosis and has led to initial misdiagnoses of rheumatoid arthritis 25 Diagnosis of the disease requires evidence of increased levels of a k or l myeloma protein in blood and or blood presence of an amyloid related organ involvement syndrome detection in tissues of amyloid deposition based on birefringence staining with Congo red and detection in tissues of k or l deposition based on electron microscopy or mass spectrometry 1 Reflecting the widespread systemic nature of the disease patient median survival is only 8 months dating from the time of diagnosis Treatment commonly improves this poor survival In a Mayo Clinic study for example AL amyloidosis assigned stage 1 2 3 or 4 based on the presence of 0 1 2 or 3 prognostic signs high blood levels of cardiac troponin T blood levels of a marker for congestive heart failure viz NT ProBNP or free light chain ratios had median survivals of 94 1 40 3 14 and 5 8 months respectively Additional factors indicating a worse prognosis include the involvement of multiple organs 10 bone marrow plasma cells presence of a translocation between chromosomes 11 and 14 i e t 11 14 and chromosomal trisomy 20 AH Amyloidosis Edit AH and AHL amyloidosis are extremely rare forms of systemic amyloidosis in which the amyloid deposit is a free heavy chain AH amyloidosis or a free heavy chain plus free light chain AHL amyloidosis 26 Case reports have detected amyloid deposits containing a free g a or m heavy chain or portions of one of these chains accompanied in many cases by a free k or l light chain in primarily in the kidney but also the spleen and other tissues AH plus AHL amyloidosis cases are 17 fold less common than AL amyloidosis cases 26 The disease often presents late in its course with signs and or symptoms of kidney failure such as those associated with the nephrotic syndrome and is therefore treated as a malignant condition 24 27 In a small study of 16 patients with renal amyloidosis the 5 patients with AH amyloidosis and the 11 patients with AHL amyloidosis had less frequent concurrent cardiac involvement and better overall survival than 202 patients with renal AL amyloidosis The hematological response to chemotherapy of the AH and AHL renal amyloidosis patients was comparable to those with renal AL amyloidosis 24 POEMS syndrome Edit Main article POEMS syndrome Main article Castleman s disease POEMS syndrome also known as Crow Fukase syndrome Takatsuki disease or PEP syndrome is a rare and complex medical syndrome that involves a combination of syndrome defining signs and symptoms due to the dysfunction of multiple organs The syndrome is associated with a plasma cell dyscrasia in almost 100 of cases pathological overexpression of certain cytokines in gt 95 of cases and the lymphoproliferative disorder termed Castleman s disease in 15 of cases Rare cases of POEMS have been associated with polyclonal rather than clonal plasma cells these cases are not plasma cell dyscrasias but rather appear to be caused by the over activity of non malignant immune cell responses in chronic infections or autoimmune diseases POEMS is an acronym standing for the characteristic signs or symptoms of the syndrome Polyneuropathy Organomegaly Endocrinopathy Plasma cell disorder typically the plasma cell burden is low in POEMS patients and Skin changes e g hemangioma hyperpigmentation The syndrome is defined by the presence of both of two major criteria peripheral neuropathy and a clonal plasma cell dyscrasia increased bone marrow plasma cells in 67 of cases 1 plasmacytoma in 33 of cases at least one other major criteria Castleman s disease sclerotic bone lesions elevated serum levels of the cytokine VEGF and at least one minor criterion organomegaly extravascular volume overload e g ascites edema pleural effusion and or pericardial effusion endocrinopathy i e hypogonadism defects in the hypothalamic pituitary adrenal axis skin changes papilledema and or hematological manifestations i e thrombocytosis or polycythemia 28 The monoclonal protein in POEMS patients is typically identified as IgA or IgG which in gt 95 of cases contains a l chain that is restricted to either of two members of the V lambda 1 subfamily viz IGLV1 40 01 and IGLV1 44 01 there are 29 other members in the V lambda family That is the myeloma protein in POEMS is almost invariably a clonal l light chain variant Deletion of chromosome 13 and chromosomal translocations but not increases in chromosome number have also been reported to occur in POEMS patients 29 Patients with 1 or 2 isolated plasmacytomas have been successfully treated with targeted radiotherapy to obtain relief of symptoms and sometimes complete remission of disease Isolated plasmacytomas may regress spontaneously Patients with gt 2 plasmacytomas or symptomatic disseminated disease have been treated with chemotherapy often followed by autologous stem cell transplantation these treatments have been found to reduce symptoms of the disease and lead to long term partial remissions of disease 28 29 The overall survival of POEMS patients who have been treated for their disease is relatively good for a disease occurring in patients with an average age of 50 years one estimate of median overall survival is 14 years POEMS patients evaluated to be in low and intermediate risk groups had gt 85 survival at 10 years those in the high risk group had a 40 survival over this time period 30 Cryoglobulinemia Edit Main article Cryoglobulinemia Cryoglobulins are proteins principally immunoglobulins that circulate in the blood precipitate at temperatures lt 37 C 98 6 F and re solubilize upon restoring physiological blood temperatures They are made and secreted into the blood as a result of underlying pathological conditions viz inflammation infection or malignancies Rarely cryoglobulinemia i e essential cryoglobulinemia occurs in patients without these or other identifiable conditions Non essential cryoglobulonemia is classified into three types Type 1 cryoglobulinemia 10 25 of cases involves a circulating myeloma protein typically IgM or IgG but in rare case reports IgA The condition is associated with Waldenstrom macroglobulinemia or multiple myeloma in 40 of type I cases the MGUS or smoldering predecessors to these diseases in 44 of type I cases and other B cell lymphoproliferative disorders in 16 of type I cases 31 Type II cryglobulinemia 50 60 of cases involves circulating IgM myeloma protein with rheumatoid factor activity and therefore bound to polyclonal IgG and protein components of the blood complement system hepatitis C virus and far more rarely hepatitis B virus or human immunodeficiency virus infections are the major causes of this cryoglobulinemia Type III cryoglobulinemia 15 30 of cases involves circulating polyclonal IgM protein with rheumatoid factor activity bound to polyclonal IgG and blood complement components autoimmune diseases and less commonly hepatitis virus C infection or lymphoproliferative disorders are the cause of this type of croglobulinemia Only types I and II are defined as plasma cell dyscrasias 32 Patients suffering type 1 cryoglobulinemia present with symptoms due to cold temperature induce blood hyperviscosity and consequential interruptions of blood flow e g skin lesions lower extremity purpuric spots and papules acrocyanosis necrosis skin ulcers livedo reticularis urticaria peripheral neuropathy blurred vision loss of vision hearing loss headaches confusion transient ischemic attacks chest pain heart failure glomerulonephritis kidney failure oral bleeding and nasal bleeding Rarely patients may present with catastrophic decreases in blood flow to vital tissues and require emergency treatment Symptomatic patients typically exhibit levels of a myeloma protein gt 5 gram liter and can be diagnosed by simple observing the temperature induced reversible induction of serum precipitate formation Patients particularly those with catastrophic presentations are treated with plasma exchange and or plasmapharesis to reduce the load of circulating myeloma proteins and relieve acute symptoms Patients with an overt malignancy are treated with the chemotherapy regimens used for Waldenstroms macroglobulinemia or multiply myeloma patients with MGUS precursors to these diseases appear less responsive to these chemotherapeutic regimens These patients as well as patients with overt malignancy may be treated with rituximab kills normal and malignant B cells that bear the CD20 antigen or the proteasome inhibitor Bortezomib 31 Patients suffering type II or type III cryoglobulinemia present with many of the symptoms of type I disease plus those of inflammatory vasculitis Their treatments are tailored to the underlying infectious autoimmune or malignant disease Type II patients associated with a monoclonal antibody and clonal plasma cells or other types of clonal B cells are typically treated with regimens used for Walsdenstorms macroglobulonemia or multiple myeloma 31 Malignant stage Edit In the malignant stage of plasma cell dyscrasias a clearly excessive tumor cell burden causes symptoms and findings predictive of rapid life threatening progression of disease These dyscrasias fall into several distinct categories Solitary plasmacytoma Edit Main article Plasmacytoma Solitary plasmacytoma is an early stage malignancy with a clinical course that lies between MGUS and multiple myeloma in the spectrum of plasma cell dyscrasias 5 Solitary plasmacytomas typically present with local symptoms due to the growing mass of plasma cells such as the bone pain or pathologic bone fractures occurring in solitary plasmacytomas of bone or the headache focal neurological deficits and cranial nerve palsies occurring in extramedullary plasmacytomas of sellar and parasellar compartments of the brain 33 Its diagnoses must meet all four of the following criteria biopsy proven tumor consisting of clonal plasma cells no evidence of any other plasmacytomas based on bone survey and MRI or in place of MRI CT scan normal bone marrow examination and absence end organ damage CRAB features or other signs or symptoms of systemic disease attributable to a plasma cell dyscrasia 5 Blood or urine myeloma proteins are usually undetectable or low in solitary plasmacytomas Solitary plasmacytoma is a rare disease with an incidence in the USA of lt 450 cases per year In a review of 1 691 cases in the US the median age at diagnosis was 63 with males representing 60 of all cases The most common site of plasmacytoma involvement was bone 58 followed by upper or lower airway tract 16 soft tissue or connective tissue 5 central nervous system 3 gastrointestinal tract 3 skin 1 and all other sites 3 Overall median survival was 8 12 years with survival decreasing with age from 12 4 years for patients lt 40 to 5 2 years for patients of 60 years or older 34 Risk of its recurrence or progression to overt multiple myeloma within 3 years is 10 5 A subset of solitary plasmacytomas termed solitary plasmacytoma with minimal bone marrow involvement has the same criteria for diagnosis as solitary plasmacytoma except that bone marrow examination shows an increase in plasma cells from a normal value of 0 to 1 5 to gt 1 6 but less than 10 of total nucleated cells While its presentations and findings are similar to solitary plasmacytoma solitary plasmacytoma with minimal bone marrow involvement is more likely to progress i e it recurs or becomes overt multiple myeloma in 20 to 60 of cases within 3 years Solitary plasmacytomas associated with 10 or more plasma cells are diagnosed as overt multiple myeloma 5 Non secretory multiple myeloma Edit Non secretory multiple myeloma represents a class of plasma cell dyscrasias where no myeloma protein is detected in serum or urine of patients with evidence of increased clonal bone marrow plasma cells and or multiple plasmacytomas particularly of the bone but also of soft tissues While a pre malignant phase is likely most new cases of non secretory multiple myeloma are brought to attention not because of incidental M protein detection which by definition is absent but because of patient symptoms indicative of malignancy possibly of plasma cell origin The condition has been diagnosed based on biopsy proved clonal plasma cell tumors and or the presence in bone marrow of plasma cells at 10 of nucleated cells in individuals who have evidence of end organ damage attributable to an underlying plasma cell disorder These patients typically also show one or more CRAB signs and lack evidence of a myeloma protein as measured by protein electrophoresis and immunofixation However more sensitive methods of detecting urinary and serum light chain myeloma proteins using enzyme linked immunosorbent assays indicate that gt 60 of cases initially diagnosed as non secretory multiple myeloma had abnormal levels of either a clonal k or l light chain in their urine or serum and therefore were better diagnosed as having light chain multiple myeloma 35 36 37 Based on the latter definition non secretory multiple myeloma represents 1 of all multiple myeloma cases with formerly diagnosed non secretory myelomas considered to be cases primarily of light chain multiple myeloma but on occasion false non secretors i e cases in which there is evidence of myeloma protein secretion such as renal myeloma protein deposits 36 A Mayo Clinic study of 124 patients initially diagnosed as having non secretory multiple myeloma were later found to be composed of 65 free light chain secretors and 35 true non secretors As a group these patients response to therapy time to disease recurrence and overall survival were similar to typical myeloma patients However in a subset of patients diagnosed after 2001 and therefore treated with more effective therapy that included autologous stem cell transplantation prognosis was significantly better in non secretory multiple myeloma patients median survival 8 3 years compared to typical myeloma patients median survival 5 4 years In addition non secretory patients exhibited a better prognosis than light chain secretory patients 35 Plasma Cell Myeloma with concomitant chronic Lymphocytic Leukemia monoclonal B Cell Lymphocytosis Edit Multiple myeloma occurring concurrently with chronic Lymphocytic Leukemia or its pre malignant precursor monoclonal B cell lymphocytosis is an extremely rare condition in which patients evidence findings of the plasma cell dyscrasia plus either one of the cited clonal lymphocytic diseases Patients are typically elderly median age of 74 range 42 91 years old males 51 of 66 case reports and commonly present with a combination of symptoms related to chronic lymphocytic leukemia symptoms fatigue autoimmune hemolytic anemia enlargements of liver and or spleen and lymphadenopathy plus symptoms of multiple myelomas Patients exhibit two distinct populations of clonal cells in their bone marrow blood and or other tissues plasma cells which may have an immature plasmablastic morphology and small lymphocytes which have a morphology typical of chronic lymphocytic leukemia cells Patients blood and or urine evidences a plasma cell derived myeloma proteins either IgG IgA or free light chain in 50 20 and 20 of cases respectively but may also have a second myeloma protein made by the lymphocytic cells either an IgM or IgG Signs and symptoms of chronic lymphocytic leukemia commonly precede those of multiple myeloma sometimes by years 38 The relationship between the two clones of cells in this combined disease has not been established although one study suggests that the clonal plasma cells and clonal lymphocytes arise from a common hematological stem cell 39 In general patients with plasma cell myeloma with concomitant chronic Lymphocytic Leukemia monoclonal B cell Lymphocytosis have been treated with the same regimens used for multiple myeloma patients unless significant complications related to the lymphocytic component of their disease e g autoimmune hemolytic anemia require treatments used in chronic lymphocytic leukemia Some patients who lack appreciable symptoms have been followed with no specific treatment of their disease 38 Waldenstrom macroglobulinemia Edit Main article Waldenstrom macroglobulinemia According to the International Workshop on Waldenstrom s Macroglobulinemia the disease is diagnosed in patients that have a serum IgM monoclonal protein and a bone marrow that contains 10 of its nucleated cells as lymphoplasmacytic cells There is no requirement for symptomatic disease a particular level of IgM protein or presence of extramedullary i e non bone lymphoplasmacytic cell infiltrates The overall survival for this malignancy at 5 and 10 years among gt 5 000 patients is 62 and 39 respectively with newer treatment regimens anticipated to improve these survival rates in the future 8 Multiple myeloma Edit Main article Multiple myeloma Multiple myeloma is diagnosed in patients that except for non secretory multiple myeloma patients have a clonal IgG IgA IgD or IgE myeloma protein in their serum and or a clonal k or l light chain in their serum or urine plus either one of two sets of criteria In the first criteria set patients must have 10 bone marrow clonal plasma cells plus 1 of the CRAB criteria in the second criteria set patients must have 10 bone marrow clonal plasma cells plus 1 of the following findings 60 bone marrow clonal plasma cells a free k l or l k light chain ratio in serum of 100 the involved clonal light chain concentration must be 100 milligrams liter and or gt 1 focal bone lesion on magnetic resonance imaging 4 The 5 year medium survival of patients with multiple myeloma treated with currently used treatment regiments is 48 5 40 Light chain multiple myeloma Edit Main article Light chain deposition disease Light chain multiple myeloma is diagnosed in patients who have a the criteria for diagnosis of multiple myeloma except having a serum free light chain ratio outside the normal range of 0 26 to 1 65 without evidence of an intact immunoglobulin or free heavy chain or b an extreme free light chain ratio i e outside the range of 0 02 to 100 with the light chain having the lower concentration being present at gt 10 milligrams liter regardless of the stage of their plasma cell dyscrasia 41 At the time of diagnosis 30 to 50 of light chain multiple myeloma patients have severe renal dysfunction or kidney failure due to light chain myeloma cast nephropathy or the nephrotoxic effects of free light chains on renal tubular cells Patients are treated similarly to patients suffering the counterparts those with multiple myeloma except that the focus is treating or preventing kidney damage using chemotherapy to reduce production of the monoclonal light chain and thereby stopping reversing or preventing kidney injury 42 Plasma cell leukemia Edit Main article Plasma cell leukemia Plasma cell leukemia is a form of multiple myeloma in which significant numbers of typically immature appearing plasma cells i e plasmablasts circulate in the blood Very small numbers of plasma cells may reach the circulation in non IgM multiple myeloma non IgM SMM and exceptionally non IgM MGUS In these plasma cell dyscrasias the presence of even very small numbers of circulating plasma cells is a poor prognostic indicator In plasma cell leukemia however circulating plasma cells reach far higher numbers and at these circulating levels are associated with exceptionally poor survival rates The International Myeloma Working Group has defined the diagnostic criteria for plasma cell leukemia as the presence in blood of gt 2x109 plasma cells per liter or alternatively gt 20 of nucleated blood cells being plasma cells More recently the Group has suggested that values of 0 5x109 or 5 respectively may be more appropriate from a therapeutic viewpoint and therefore should be studied as a definitive criterion for the disease 43 A recent study supported this suggestion in finding that multiple myeloma patients with gt 5 circulating plasma cells had a prognosis much worse than that for multiple myeloma and similar to that for plasma cell leukemia 44 Flow cytometry immunophenotyping of blood cells to detect clonal phenotypes of plasma cells seen in multiple myeloma e g the CD138 CD38 CD19 CD4 phenotype may be a more sensitive method to enumerate circulating clonal plasma cells and diagnose plasma cell leukemia 45 There are two forms of plasma cell leukemia Primary plasma cell leukemia in which patients without a history of multiple myeloma present with diagnostically high levels of circulating plasma cells and Secondary plasma cell leukemia in which patients with multiple myeloma suffer their dyscrasia s progression by the expansion of large numbers of their malignant plasma cells into the circulation and distant tissues Historically primary plasma cell leukemia was more common than the secondary form but with the increased survival of multiple myeloma patients due to new treatment regiments more cases of secondary plasma cell leukemia are occurring currently the two forms occur in approximately equal numbers 43 Patients with primary plasma cell leukemia present with clinical findings that are less commonly found in multiple myeloma e g they often have hepatomegaly splenomegaly lymphadenopathy nerve and central nervous system defects bleeding tendencies secondary to thrombocytopenia and pleural effusions They are less likely than multiple myeloma patients to have lytic bone lesions In several studies of patients with either form of plasma cell leukemia the disease was associated with clonal IgG in 28 to 56 of cases IgA in 4 to 7 of cases and a light chain in 23 to 44 of cases 0 12 of patients had no myeloma protein Medium survival for primary and secondary plasma cell dyscrasias have been 7 13 months and 2 7 months respectively but appear to be improving with new treatment regimens 18 45 Heavy chain disease EditMain article Heavy chain disease The four heavy chain diseases are exceedingly rare conditions associated with the production circulation in blood and often presence in urine of a free clonal heavy chain with no detected clonal light chains The heavy chain is non functional and altered by having deletions insertions and point mutations due to somatic mutations in their respective coding genes Beyond this commonality however these diseases have very different clinical differences Furthermore each of the heavy chain diseases appears to be due to rare variants of lymphoma and therefore is sometimes regarded as a B cell dyscrasia 11 However heavy chain diseases are still often classified with plasma cell dyscrasias 46 The heavy chain diseases are classified as a g and m heavy chain diseases and are based respectively on gt 400 130 and 30 40 case reports as reviewed in a 2014 publication 11 a Heavy chain disease Edit a Heavy chain disease also termed immunoproliferative small intestinal disease or IPSID Mediterranean lymphoma and Seligmann disease afflicts primarily individuals of Mediterranean North African and Middle Eastern descent of lower economic status Many cases are centered in the Middle East and associated with relatively unsanitary living conditions The disease usually appears between the ages 10 and 30 and in some cases may be an aberrant immune response to a parasite or other microorganism 47 The disease commonly effects the gastrointestinal tract leading to signs and symptoms of a malabsorption syndrome or far less commonly the respiratory tract with signs and symptoms of respiratory dysfunction Involved tissues usually include mucosa associated lymphoid tissues and evidence a histology of lymphoplasmacytoid infiltrates accompanied by large numbers of plasma cells and small lymphocytes The plasma cells therein express the monoclonal a chain and therefore are clonal in nature and the sole or contributing producer of the a chain myeloma protein Some 57 to 66 of patients present with disseminated lymphoma 17 to 36 of patients present with a localized lymphoma and 9 to 17 of patients lack any evidence of a lymphoplasmacytic neoplasm A majority of the latter patients have an autoimmune disease or a chronic infection which may be responsible for or contribute to production of the a heavy chain Studies indicated that a sub set particularly of the digestive form of heavy chain disease is caused by infection This is based on findings that the majority of a heavy chain disease patients are in the lower economic class living under unsanitary conditions that gastrointestinal bacterial and parasitic infections have been documented in many of these patients and that long term gt 6 months appropriately selected antibiotic therapy has improved the condition in 33 to 71 of patients who are at an early stage of the disease and documented to be infected However these patients frequently relapse Patients resistant to antibiotic trials have been treated with multiple drug chemotherapy to obtain complete remission rates of 64 and an overall 5 year survival of 67 11 g heavy chain disease Edit g Heavy chain disease also termed Franklin disease or Franklin s disease presents in three patterns a aggressive lymphoma 57 to 66 of cases associated with constitutional symptoms and in 50 of cases with enlargement of lymph nodes spleen and or liver b localized lymphoma 25 of cases with lymphoma limited to the bone marrow or an extra nodal site usually the skin but sometimes the thyroid gland parotid gland oropharyngeal cavity conjunctiva or gastrointestinal tract and c no lymphoma 9 to 17 of cases associated typically with a preexistent autoimmune disease but no evidence of lymphoma Involved lymphoma infiltrated tissues typically show a mixture of lymphoplamsmacytoid cells plasma cells lymphocytes and sometimes variable numbers of eosinophils and hystiocytes Treatment of the disease varies with its clinical severity Patients with aggressive lymphoma have been treated with multiple drug chemotherapy patients with limited lymphoma have been monitored for disease progression or treated locally e g radiation therapy surgical removal and patients with no lymphoma have been monitored for progression in their diseases while being treated for any autoimmune disease that they bear Spontaneous remissions in g heavy chain disease have occurred Regardless of presentation pattern these patients may have an aggressive or indolent disease with courses ranging from the asymptomatic presence of a stable monoclonal heavy chain in the serum or urine e g MGUS to a rapid downhill progression of a few weeks duration g Heavy chain disease survivorship ranged form 1 month to gt 20 years medium survival 7 4 years in a Mayo Clinic study 11 48 m Heavy chain disease Edit m Heavy chain disease presents with a picture of a lymphoid neoplasm resembling either chronic lymphocytic leukemia or small lymphocytic lymphoma This picture includes splenomegaly in virtually all cases hepatomegaly in 75 of cases lymphadenopathy in 40 of cases and lytic bone lesions in 20 of cases Patients often have hypogammaglobulinemia increases in urinary free light chains and a bone marrow containing vacuolated plasma cells or lymphoid cells Treatment of m heavy chain disease had varied form observation only in asymptomatic patients to single drug or and multiple drug chemotherapy in symptomatic patients Survival with this disease varies between lt 1 month to gt 10 years with a median survival rate of 2years 11 47 48 See also EditParaproteinemia GammaglobulinsReferences Edit a b c Castillo JJ 2016 Plasma Cell Disorders Primary Care 43 4 677 691 doi 10 1016 j pop 2016 07 002 PMID 27866585 a b c d e f g h i j k Willrich MA Murray DL Kyle RA 2017 Laboratory testing for monoclonal gammopathies Focus on monoclonal gammopathy of undetermined significance and smoldering multiple myeloma Clinical Biochemistry 51 38 47 doi 10 1016 j clinbiochem 2017 05 001 PMID 28479151 a b c d e van de Donk NW Mutis T Poddighe PJ Lokhorst HM Zweegman S 2016 Diagnosis risk stratification and management of monoclonal gammopathy of undetermined significance and smoldering multiple myeloma International Journal of Laboratory Hematology 38 Suppl 1 110 22 doi 10 1111 ijlh 12504 PMID 27161311 S2CID 21348701 a b Rajkumar SV Dimopoulos MA Palumbo A Blade J Merlini G Mateos MV Kumar S Hillengass J Kastritis E Richardson P Landgren O Paiva B Dispenzieri A Weiss B LeLeu X Zweegman S Lonial S Rosinol L Zamagni E Jagannath S Sezer O Kristinsson SY Caers J Usmani SZ Lahuerta JJ Johnsen HE Beksac M Cavo M Goldschmidt H Terpos E Kyle RA Anderson KC Durie BG Miguel JF 2014 International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma The Lancet Oncology 15 12 e538 48 doi 10 1016 S1470 2045 14 70442 5 PMID 25439696 a b c d e f Rajkumar SV 2016 Updated Diagnostic Criteria and Staging System for Multiple Myeloma American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Meeting 35 e418 23 doi 10 14694 EDBK 159009 PMID 27249749 a b c d e f g Dutta AK Hewett DR Fink JL Grady JP Zannettino AC 2017 Cutting edge genomics reveal new insights into tumour development disease progression and therapeutic impacts in multiple myeloma British Journal of Haematology 178 2 196 208 doi 10 1111 bjh 14649 PMID 28466550 S2CID 25838916 a b c d El Ayoubi A Wang JQ Hein N Talaulikar D 2017 Role of plasma cells in Waldenstrom macroglobulinaemia Pathology 49 4 337 345 doi 10 1016 j pathol 2017 02 004 PMID 28483372 a b c d e Abeykoon JP Yanamandra U Kapoor P 2017 New developments in the management of Waldenstrom macroglobulinemia Cancer Management and Research 9 73 83 doi 10 2147 CMAR S94059 PMC 5354523 PMID 28331368 Pandey S Kyle RA 2013 Unusual myelomas a review of IgD and IgE variants Oncology Williston Park N Y 27 8 798 803 PMID 24133829 Ribourtout B Zandecki M 2015 Plasma cell morphology in multiple myeloma and related disorders Morphologie Bulletin de l Association des Anatomistes 99 325 38 62 doi 10 1016 j morpho 2015 02 001 PMID 25899140 a b c d e f g h Bianchi G Anderson KC Harris NL Sohani AR 2014 The heavy chain diseases clinical and pathologic features Oncology Williston Park N Y 28 1 45 53 PMID 24683718 KIF2B kinesin family member 2B Homo sapiens human Gene NCBI CIDEC cell death inducing DFFA like effector c Homo sapiens human Gene NCBI Simeon V Todoerti K La Rocca F Caivano A Trino S Lionetti M Agnelli L De Luca L Laurenzana I Neri A Musto P 2015 Molecular Classification and Pharmacogenetics of Primary Plasma Cell Leukemia An Initial Approach toward Precision Medicine International Journal of Molecular Sciences 16 8 17514 34 doi 10 3390 ijms160817514 PMC 4581206 PMID 26263974 a b c Al Hussain T Hussein MH Al Mana H Akhtar M 2015 Renal involvement in monoclonal gammopathy Advances in Anatomic Pathology 22 2 121 34 doi 10 1097 PAP 0000000000000056 PMID 25664947 S2CID 5137203 Itoh T 2016 Multiple myeloma developing in a patient with immune thrombocytopenia Rinsho Ketsueki the Japanese Journal of Clinical Hematology 57 5 630 3 doi 10 11406 rinketsu 57 630 PMID 27263790 a b c d e f g h Mailankody S Landgren O 2016 Monoclonal gammopathy of undetermined significance and Waldenstrom s macroglobulinemia Best Practice amp Research Clinical Haematology 29 2 187 193 doi 10 1016 j beha 2016 08 015 PMID 27825465 a b Bommannan K Sachdeva MU Malhotra P Kumar N Sharma P Naseem S Ahluwalia J Das R Varma N Prakash G Khadwal A Srinivasan R Varma S 2016 Plasma cell leukemia in North India retrospective analysis of a distinct clinicohematological entity from a tertiary care center and review of literature Blood Research 51 1 23 30 doi 10 5045 br 2016 51 1 23 PMC 4828524 PMID 27104188 Khouri J Samaras C Valent J Mejia Garcia A Faiman B Mathur S Hamilton K Nakashima M Kalaycio M January 2019 Monoclonal gammopathy of undetermined significance A primary care guide Cleveland Clinic Journal of Medicine 86 1 39 46 doi 10 3949 ccjm 86a 17133 PMID 30624183 S2CID 58554384 a b c d e f Muchtar E Kumar SK Magen H Gertz MA 2017 Diagnosis and management of smoldering multiple myeloma the razor s edge between clonality and cancer Leukemia amp Lymphoma 59 2 1 12 doi 10 1080 10428194 2017 1334124 PMID 28592156 S2CID 11195536 a b Ciocchini M Arbelbide J Musso CG 2017 Monoclonal gammopathy of renal significance MGRS the characteristics and significance of a new meta entity International Urology and Nephrology 49 12 2171 2175 doi 10 1007 s11255 017 1594 y PMID 28425076 S2CID 1318744 Kyle RA Larson DR Therneau TM Dispenzieri A Melton LJ Benson JT Kumar S Rajkumar SV 2014 Clinical course of light chain smouldering multiple myeloma idiopathic Bence Jones proteinuria a retrospective cohort study The Lancet Haematology 1 1 e28 e36 doi 10 1016 S2352 3026 14 70001 8 PMC 4266993 PMID 25530988 Malyszko J Kozlowska K Malyszko JS 2017 Amyloidosis A cancer derived paraproteinemia and kidney involvement Advances in Medical Sciences 62 1 31 38 doi 10 1016 j advms 2016 06 004 PMID 28153807 a b c Nasr SH Said SM Valeri AM Sethi S Fidler ME Cornell LD Gertz MA Dispenzieri A Buadi FK Vrana JA Theis JD Dogan A Leung N 2013 The diagnosis and characteristics of renal heavy chain and heavy light chain amyloidosis and their comparison with renal light chain amyloidosis Kidney International 83 3 463 70 doi 10 1038 ki 2012 414 PMID 23302715 Elsaman AM Radwan AR Akmatov MK Della Beffa C Walker A Mayer CT Dai L Nativ S Rygg M Atsali E Saijo K Ogdie AR Srinivasulu N Fathi N Schumacher HR Pessler F 2013 Amyloid arthropathy associated with multiple myeloma a systematic analysis of 101 reported cases Seminars in Arthritis and Rheumatism 43 3 405 12 doi 10 1016 j semarthrit 2013 07 004 PMID 23992800 a b Said SM Sethi S Valeri AM Leung N Cornell LD Fidler ME Herrera Hernandez L Vrana JA Theis JD Quint PS Dogan A Nasr SH 2013 Renal amyloidosis origin and clinicopathologic correlations of 474 recent cases Clinical Journal of the American Society of Nephrology 8 9 1515 23 doi 10 2215 CJN 10491012 PMC 3805078 PMID 23704299 UpToDate a b Warsame R Yanamandra U Kapoor P 2017 POEMS Syndrome an Enigma Current Hematologic Malignancy Reports 12 2 85 95 doi 10 1007 s11899 017 0367 0 PMID 28299525 S2CID 31324035 a b Dispenzieri A 2015 POEMS syndrome update on diagnosis risk stratification and management American Journal of Hematology 90 10 951 62 doi 10 1002 ajh 24171 PMID 26331353 S2CID 5167507 Kourelis TV Dispenzieri A 2017 Validation of a prognostic score for patients with POEMS syndrome a mayo clinic cohort Leukemia 31 5 1251 doi 10 1038 leu 2017 68 PMID 28280263 S2CID 2521267 a b c Retamozo S Brito Zeron P Bosch X Stone JH Ramos Casals M 2013 Cryoglobulinemic disease Oncology Williston Park N Y 27 11 1098 1105 1110 6 PMID 24575538 Ghetie D Mehraban N Sibley CH 2015 Cold hard facts of cryoglobulinemia updates on clinical features and treatment advances Rheumatic Disease Clinics of North America 41 1 93 108 viii ix doi 10 1016 j rdc 2014 09 008 PMID 25399942 Lee J Kulubya E Pressman BD Mamelak A Bannykh S Zada G Cooper O 2017 Sellar and clival plasmacytomas case series of 5 patients with systematic review of 65 published cases Pituitary 20 3 381 392 doi 10 1007 s11102 017 0799 5 PMC 5429193 PMID 28251542 Thumallapally N Meshref A Mousa M Terjanian T 2017 Solitary plasmacytoma population based analysis of survival trends and effect of various treatment modalities in the USA BMC Cancer 17 1 13 doi 10 1186 s12885 016 3015 5 PMC 5216567 PMID 28056880 a b Chawla SS Kumar SK Dispenzieri A Greenberg AJ Larson DR Kyle RA Lacy MQ Gertz MA Rajkumar SV 2015 Clinical course and prognosis of non secretory multiple myeloma European Journal of Haematology 95 1 57 64 doi 10 1111 ejh 12478 PMID 25382589 S2CID 22409325 a b Dupuis MM Tuchman SA 2016 Non secretory multiple myeloma from biology to clinical management OncoTargets and Therapy 9 7583 7590 doi 10 2147 OTT S122241 PMC 5171196 PMID 28008276 Heaney JL Campbell JP Griffin AE Birtwistle J Shemar M Child JA Gregory WM Cairns DA Morgan G Jackson G Drayson MT 2017 Diagnosis and monitoring for light chain only and oligosecretory myeloma using serum free light chain tests PDF British Journal of Haematology 178 2 220 230 doi 10 1111 bjh 14753 PMID 28573706 S2CID 38301916 a b Alley CL Wang E Dunphy CH Gong JZ Lu CM Boswell EL Burchette J Lagoo AS 2013 Diagnostic and clinical considerations in concomitant bone marrow involvement by plasma cell myeloma and chronic lymphocytic leukemia monoclonal B cell lymphocytosis a series of 15 cases and review of literature Archives of Pathology amp Laboratory Medicine 137 4 503 17 doi 10 5858 arpa 2011 0696 OA PMID 23544940 Trudel S Ghamlouch H Dremaux J Delette C Harrivel V Marolleau JP Gubler B 2016 The Importance of an In depth Study of Immunoglobulin Gene Rearrangements When Ascertaining the Clonal Relationship between Concomitant Chronic Lymphocytic Leukemia and Multiple Myeloma Frontiers in Immunology 7 625 doi 10 3389 fimmu 2016 00625 PMC 5187371 PMID 28082975 Bustoros M Mouhieddine TH Detappe A Ghobrial IM 2017 Established and Novel Prognostic Biomarkers in Multiple Myeloma American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Meeting 37 548 560 doi 10 14694 EDBK 175175 PMID 28561668 Raza S Safyan RA Rosenbaum E Bowman AS Lentzsch S 2017 Optimizing current and emerging therapies in multiple myeloma a guide for the hematologist Therapeutic Advances in Hematology 8 2 55 70 doi 10 1177 2040620716680548 PMC 5298389 PMID 28203342 Tessenow H Holzvogt M Holzvogt B Andrea M Heyn S Schliwa T Schwarz M Zehrfeld T Becker C Pfrepper C Franke GN Krahl R Jentzsch M Leiblein S Schwind S Bill M Vucinic V Lange T Niederwieser D Ponisch W 2017 Successful treatment of patients with newly diagnosed untreated light chain multiple myeloma with a combination of bendamustine prednisone and bortezomib BPV Journal of Cancer Research and Clinical Oncology 143 10 2049 2058 doi 10 1007 s00432 017 2439 x PMID 28534173 S2CID 22739588 a b Fernandez de Larrea C Kyle RA Durie BG Ludwig H Usmani S Vesole DH Hajek R San Miguel JF Sezer O Sonneveld P Kumar SK Mahindra A Comenzo R Palumbo A Mazumber A Anderson KC Richardson PG Badros AZ Caers J Cavo M LeLeu X Dimopoulos MA Chim CS Schots R Noeul A Fantl D Mellqvist UH Landgren O Chanan Khan A Moreau P Fonseca R Merlini G Lahuerta JJ Blade J Orlowski RZ Shah JJ 2013 Plasma cell leukemia consensus statement on diagnostic requirements response criteria and treatment recommendations by the International Myeloma Working Group Leukemia 27 4 780 91 doi 10 1038 leu 2012 336 PMC 4112539 PMID 23288300 Granell M Calvo X Garcia Guinon A Escoda L Abella E Martinez CM Teixido M Gimenez MT Senin A Sanz P Campoy D Vicent A Arenillas L Rosinol L Sierra J Blade J de Larrea CF 2017 Prognostic impact of circulating plasma cells in patients with multiple myeloma implications for plasma cell leukemia definition Haematologica 102 6 1099 1104 doi 10 3324 haematol 2016 158303 PMC 5451342 PMID 28255016 a b Gonsalves Wilson I 2017 04 10 Primary Plasma Cell Leukemia A Practical Approach to Diagnosis and Clinical Management American Journal of Hematology Oncology 13 3 Willrich MA Katzmann JA 2016 Laboratory testing requirements for diagnosis and follow up of multiple myeloma and related plasma cell dyscrasias Clinical Chemistry and Laboratory Medicine 54 6 907 19 doi 10 1515 cclm 2015 0580 PMID 26509779 S2CID 2706697 a b Heavy Chain Diseases Hematology and Oncology a b UpToDate External links Edit Retrieved from https en wikipedia org w index php title Plasma cell dyscrasias amp oldid 1134825233, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.