Multiple Myeloma

Multiple Myeloma

Last Section Update: 11/2020

Contributor(s): Tina Kaczor, ND/FABNO; Shayna Sandhaus, PhD

1 Introduction

Multiple myeloma (MM) is a cancer of mature plasma cells, which are a type of white blood cell that produces immunoglobulins (antibodies). Antibodies are normally produced to protect the body from microbes, such as bacteria. In multiple myeloma, the monoclonal plasma cells produce a monoclonal immunoglobulin (M-spike) that is dysfunctional, may inhibit the immune system, and can lead to damage to different organs. Advancing MM generally manifests some or all of a constellation of defining events referred to as the CRAB signs: high serum Calcium, Renal failure, Anemia, and Bone lesions and fractures. Myeloma also puts patients at an increased risk for serious infections.1,2

Multiple myeloma is preceded by precursor conditions known as monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM). These three conditions together represent a continuum of disease. Determining which patients will progress from lower-risk precursor conditions to active MM is a challenge for physicians and comprises an active area of research. Most patients with MGUS and some with SMM never progress to active MM. Overall, MM and its precursor conditions have a variable clinical course, and treatment-related decisions must be tailored to each patient’s individual risk profile.3

Myeloma is the second-most-common blood cancer in the United States4: an estimated 32,000 patients are diagnosed with myeloma every year.5 Over a span of five years, there are approximately 230,000 cases worldwide.6 Myeloma is more commonly diagnosed in older patients, with a median age at diagnosis between 66 and 70 years.7 About 3% of the general population over age 50 is thought to have MGUS.8,9 Of these individuals, about 1% per year progress to myeloma or a related cancer.3

Myeloma is considered incurable with current therapies. However, the number of available treatment options has expanded substantially since the mid-1900s. Recent advances in therapeutic strategies, including novel classes of drugs such as proteasome inhibitors, immunomodulatory drugs, immunotherapy, and broader application of autologous stem cell transplantation have all extended the typical survival of patients with myeloma. Today, the likelihood of surviving five years following a myeloma diagnosis is over 70%.10

2 Biology and Development of Myeloma

Blood Cells and Genes

Blood largely consists of fluid (primarily water) and a combination of three different cell types: platelets that help form clots and control bleeding, red blood cells that deliver oxygen throughout the body, and white blood cells, which are part of the immune system.11 Blood cells grow and develop within bone marrow, the spongy tissue inside bones. Immature blood cells are referred to as hematopoietic stem cells, which develop into young precursor blood cells that eventually mature into platelets, red blood cells, and white blood cells before leaving the bone marrow and entering the bloodstream.

The two types of white blood cells produced in the bone marrow are B cells and T cells. When an invading organism enters the body, B cells mature to plasma cells, which are cells that produce antibodies.12 Antibodies (also referred to as immunoglobulins) are proteins that circulate in the bloodstream and neutralize invading organisms, such as bacteria and viruses. An antibody is made up of two different kinds of proteins that are named according to their relative size. The smaller protein is called the light chain and the larger protein is called the heavy chain. Each antibody is composed of two heavy chains and two light chains. The heavy chain defines the antibody subtype. There are five different subtypes of antibodies—IgM, IgG, IgA, IgE, and IgD. There are two types of light chains: kappa (κ) and lambda (λ), and either of them can be bound to any of the heavy chains, leading to 10 different types of immunoglobulins. In addition, light chains can be secreted independently of the heavy chain and can circulate as free light chains.13

All cells in the body contain genes, which are instructions for how cells function and build new cells.11 Genes are stored in the form of deoxyribonucleic acid (DNA) and organized into long strands called chromosomes. Changes in DNA sequence, which are referred to as mutations, have the potential to turn normal cells into cancer cells. In addition to mutations, a variety of other changes can occur in the chromosomes, including loss, gain, and rearrangements, as well as changes in the way genes are expressed (epigenetic changes).

Basics of Myeloma

Myeloma pathobiology is complex, and many aspects of the development of the disease are poorly understood. In general, myeloma develops as a result of genetic alterations in plasma cells. Unlike healthy mature cells, myeloma cells continue to divide and produce more myeloma cells. This expansion of the cancer cell population occurs in the bone marrow microenvironment and can crowd out healthy cells, reducing the production of other blood cells. Phenomena such as genetic instability and the breakdown of immune surveillance contribute to the expansion of malignant plasma cell populations and progression of MM. Additionally, the expansion of myeloma cell populations inside the bone interferes with typical bone turnover through a variety of complex mechanisms, resulting in bone pain and pathologic fractures.3

Myeloma can inhibit healthy antibody production in the bone marrow microenvironment, thereby suppressing the part of the immune response that is mediated by antibodies.14 Because antibodies play an important role in protecting the body from bacteria and viruses, patients with myeloma are susceptible to these infections.15,16

The production of myeloma cells causes an increase in monoclonal proteins in the body. Monoclonal proteins are antibodies that are identical because they were produced by identical (clonal) plasma cells. Another term for monoclonal proteins is M proteins. The majority of patients develop myeloma cells that produce the IgG and IgA subtype of M protein.17 The remaining subtypes are less common. In addition to the two heavy chains and two light chains required to make a functional antibody, myeloma cells may produce extra light chains unbound to heavy chains called free light chains.18 This type of myeloma is called light-chain myeloma. Clinicians may use the type of myeloma to monitor disease status and predict the response to treatment and overall course of disease.

Metabolic Processes

All cells in the body require energy to function and survive. Glucose and glutamine are two critical nutrients that provide this energy.19 Glycolysis is the process of breaking down glucose inside the cell into energy that is usable by the cell.20 Glycolysis can occur under either aerobic (with oxygen) or anaerobic (without oxygen) conditions, although aerobic glycolysis is more efficient. Glutamine is also important for cellular function, as it maintains cellular metabolism and serves as a precursor in the synthesis of nucleic acids.21 Glutamine enters the cell and is metabolized by a process called glutaminolysis to produce glutamate, citrate, aspartate, and other amino acids.22,23

Compared with healthy cells, cancer cells have greater metabolic requirements because they are constantly proliferating and avoiding cell death. They meet this increased demand by altering the metabolic processes related to glycolysis and glutaminolysis.24 Anaerobic glycolysis is enhanced to provide additional energy to malignant cells, a phenomena known as the “Warburg effect.”25 As a result, glycolysis no longer serves as a source of sufficient biosynthetic precursors. To compensate for this, cancer cells enhance the rates of glutaminolysis to increase the conversion of glutamine to α-ketoglutarate and other amino acids required for cellular function.26

Myeloma cells are no exception to altered metabolic pathways present in other cancer cells.19 Hexokinase II (HKII) catalyzes the first step in glycolysis and has been shown to be upregulated in multiple myeloma cells.27 Conversion of phosphoenolpyruvate into pyruvate and adenosine triphosphate (ATP) is the last step in glycolysis and is mediated by pyruvate kinase (PK). An isoform of PK, known as PKM2, is profuse in cancer cells and has increased expression in myeloma cells, further promoting increased energy production.28 Myeloma cells also show signals of increased glutaminolysis. The myelocytomatosis oncogene (MYC) is often overexpressed in myeloma cells29 and promotes enhanced cellular uptake of glutamine, resulting in increased tumor growth and protein synthesis.30 Researchers are investigating different ways these metabolic alterations could be leveraged as drug targets.31

Symptoms

Anemia is present in almost all myeloma patients at some time during the course of disease17,32 and can manifest as fatigue or generalized weakness. The cause of anemia is often related to bone marrow suppression or kidney damage, which results in reduced number of red blood cells. Myeloma can also reduce the number of platelets in the blood. Platelets stop bleeding when the body is injured, and reduced levels can result in increased bleeding or bruising.

Bone pain occurs in the majority of patients with myeloma and is commonly localized to the axial skeleton (spine, rib cage, and pelvis).17 This happens when myeloma cells crowd and damage healthy bone, making it more susceptible to fractures. Destruction of bone can result in high calcium blood levels, which may cause excessive thirst, nausea, confusion, and constipation.33

Patients with MM are at increased risk of bacterial infections due to multifactorial immune deficiency related to the disease itself and to common treatment regimens.34,35 Patients should monitor themselves for fever, since this is an early sign that the body is fighting an infection.

Around half of patients with MM will experience kidney dysfunction.36 This damage can be caused by high levels of calcium in the blood or damage from the monoclonal proteins filtering through the kidneys into the urine (monoclonal proteins detected in the urine of MM patients are called Bence Jones proteins).37 The use of non-steroidal anti-inflammatory drugs (NSAIDs), like ibuprofen or aspirin, should be avoided in patients with myeloma-induced kidney injury as these types of medications may worsen kidney function.

Neurological problems sometimes arise in MM due to breakdown of bone surrounding the spinal cord or nerve compression by a plasmacytoma, although these are relatively rare manifestations.38

3 Causes and Risk Factors

Age

Myeloma is most commonly diagnosed in older adults. The median age at diagnosis of active myeloma is 67‒70 years.39 The incidence of the early-stage myeloma precursor condition, monoclonal gammopathy of undetermined significance (MGUS), increases more than 4-fold between the ages of 50 and 90 years in males.40 The prevalence of MGUS was found in one U.S. county to be 5.3% in people over 70 years of age and 7.5% in people over 85 years.41

Ethnicity

The risk of MGUS is approximately twice as high in blacks compared with whites,41-43 and the incidence of developing active myeloma in blacks is two to three times higher than in whites.44 People of Asian descent may be at lower risk for developing myeloma.45

Genetics

Genetic alterations play a role in the development and progression of myeloma and myeloma precursor conditions. While alterations in certain chromosomal regions and specific genes have been identified as playing key roles in the development of MM, research is ongoing to better define the genetic landscape of the disease and clarify potential therapeutic targets.3,46

The acquisition of new mutations over time can shape how cases of MGUS, SMM, and MM evolve and progress. In some cases, mutations can contribute to the rapid progression from MGUS to SMM and MM. Myeloma cells sometimes acquire new mutations that make them more resistant to treatment. In fact, some evidence suggests that some treatments may select for resistant clones, which subsequently expand and contribute to relapse.3,47

There is some evidence for heritability of myeloma risk. The risk of developing MGUS or myeloma is about 2- to 4-fold higher in those with a first-degree relative who has been diagnosed.48-50 However, the number of cases of familial myeloma is believed to be small.

Environmental Exposures

A number of environmental factors are thought to contribute to myeloma, including exposure to certain pesticides and radiation.51 One study showed that radiologists with high exposure to radiation were at increased risk for developing myeloma.52 Another study showed that occupational exposure to some chemicals was also associated with increased risk.53 Agricultural workers who come into contact with livestock or pesticides may be more likely to develop the disease; however, the evidence for these associations is relatively weak.54-56

Modifiable Risk Factors

Adults with a higher body mass index (BMI) are more susceptible to developing cancer, including myeloma.57-59 In one study, obesity, defined as a BMI of 30 kg/m2 or greater, was associated with a 2.4-fold increased risk of myeloma in men, compared with non-obese males, after accounting for age and physical activity.60 A more modest 1.6-fold increase was identified for women with BMIs over 30 kg/m2. A strong association between tobacco use and myeloma has also been identified.61 This effect seems to be most pronounced in women and those smoking more than 20 cigarettes per day.62

Immune Senescence and Myeloma

Myeloma is most commonly diagnosed in people over 60 years of age. This age-related increase in myeloma incidence coincides with another physiological phenomena called immune senescence that can be described as an age-related immune system decline which increases the risk of infection and autoimmune disorders.63

Emerging evidence suggests a relationship between immune senescence and increased risk of some cancers, including myeloma.64,65 There are several mechanisms by which this may occur. Immune senescence results in increased inflammatory signals circulating in the blood. This is thought to create a favorable environment for cancer cells, allowing them to grow and spread. Another impact of immune senescence is reduced production of immune cells from bone marrow.63,66,67 This hinders the body’s ability to detect harmful agents, such as cancer cells. Moreover, MM treatment regimens can further compromise immune function. Therapies are being developed to leverage the body’s immune system against cancers.

More information about maintaining healthy immune function during aging is available in Life Extension’s Immune Senescence protocol.

4 Diagnosis and Testing for Myeloma

Diagnosis

MGUS, the precursor to myeloma, is an asymptomatic, non-cancerous condition where abnormal levels of M proteins are found in the blood.68 Only a small number of patients with MGUS develop myeloma. There are two main stages of myeloma: smoldering (or asymptomatic) and symptomatic or active. The difference between smoldering myeloma and active myeloma is the presence of end-organ damage or certain biomarkers indicating high risk of development of end-organ damage. Rarely, active myeloma may progress to a terminal plasma cell dyscrasia known as secondary plasma cell leukemia, which is very aggressive and has a poor prognosis.69 Table 1 summarizes typical diagnostic criteria for MGUS, smoldering myeloma, and active myeloma.

Table 1: Definitions of MGUS, Smoldering Myeloma, and Symptomatic Myeloma*

Monoclonal Gammopathy of Undetermined Significance (MGUS)

Non-IgM monoclonal gammopathy of undetermined significance

  • Serum monoclonal protein (non-IgM type) <30 mg/dL (<30 g/L)
  • Clonal bone marrow plasma cells <10%
  • Absence of end-organ damage such as hypercalcemia, renal insufficiency, anemia, and bone lesions (CRAB) or amyloidosis that can be attributed to the plasma cell proliferative disorder

IgM monoclonal gammopathy of undetermined significance

  • Serum IgM monoclonal protein <30 mg/dL (<30 g/L)
  • Bone marrow lymphoplasmacytic infiltration <10%
  • No evidence of anemia, constitutional symptoms, hyperviscosity, lymphadenopathy, hepatosplenomegaly, or other end-organ damage that can be attributed to the underlying lymphoproliferative disorder

Light-chain monoclonal gammopathy of undetermined significance

  • Abnormal FLC ratio (<0.26 or >1.65)
  • Increased level of the appropriate involved light chain (increased κ FLC in patients with ratio >1.65 and increased λ FLC in patients with ratio <0.26)
  • No immunoglobulin heavy chain expression on immunofixation
  • Absence of end-organ damage such as hypercalcemia, renal insufficiency, anemia, and bone lesions (CRAB) or amyloidosis that can be attributed to the plasma cell proliferative disorder
  • Clonal bone marrow plasma cells <10%
  • Urinary monoclonal protein <500 mg/24 h

Smoldering (Asymptomatic) Myeloma

Serum monoclonal protein (IgG or IgA) ≥30 mg/dL (≥30 g/L) or urinary monoclonal protein ≥500 mg per 24 h and/or clonal bone marrow plasma cells 10–60%

Absence of myeloma defining events or amyloidosis

Active Myeloma

Clonal bone marrow plasma cells ≥10% or biopsy-proven bony or extramedullary plasmacytoma** and any one or more of the following myeloma-defining events:

  • Myeloma defining events:
    • Evidence of end-organ damage that can be attributed tothe underlying plasma cell proliferative disorder (CRAB features), specifically:
      • Hypercalcemia: serum calcium >0.25 mmol/L (>1 mg/dL) higher than the upper limit of normal or >2.75 mmol/L (>11 mg/dL)
      • Renal insufficiency: creatinine clearance <40 mL per min or serum creatinine >177 μmol/L (>2 mg/dL)
      • Anemia: hemoglobin value >2 g/dL (>20 g/L) below the lower limit of normal, or hemoglobin value <10 g/dL (<100 g/L)
      • Bone lesions: one or more osteolytic lesions on skeletal radiography, CT, or PET-CT
    • Any one or more of the following biomarkers of malignancy:
      • Clonal bone marrow plasma cell percentage ≥60%
      • Involved:uninvolved serum free light chain ratio ≥100
      • >1 focal lesion on MRI studies

*To be interpreted with the help of a clinician skilled in the management of myeloma.

Derived from National Comprehensive Cancer Network (NCCN) Guidelines and 2014 International Myeloma Working Group (IMWG) consensus criteria.70,71

**Clonality should be established by showing κ/λ-light-chain restriction on flow cytometry, immunohistochemistry, or immunofluorescence. Bone marrow plasma cell percentage should preferably be estimated from a core biopsy specimen; in case of a disparity between the aspirate and core biopsy, the highest value should be used.

Testing for Myeloma

Evaluation of a potential myeloma case should involve a complete medical history and physical exam, including a neurological exam. Examination should focus on any signs or symptoms related to bone pain and infections. On occasion, tests for myeloma will be conducted when a separate routine blood test identifies something unusual. If myeloma is suspected, tests should be conducted to examine the blood and bone marrow for evidence of myeloma.72,73

Complete blood count with differential. A complete blood count (CBC) provides information on the amount of red blood cells, white blood cells, and platelets in the body.74 Inclusion of a differential measures each type of white blood cell. These tests help determine the status of a patient’s immune system and can help diagnose anemia.

Blood chemistry panel. A blood chemistry panel can provide information on blood electrolytes and markers of kidney function. High levels of calcium may represent a marker for bone damage in myeloma.75 Abnormalities in blood creatinine or urea nitrogen may be indicative of kidney damage.

Tests for serum protein. Serum quantitative immunoglobulin tests measure the amount and type of immunoglobulins in the blood. Typically IgG, IgM, and IgA subtypes are included.76 Serum immunofixation electrophoresis shows the type of M proteins present in the blood, while serum protein electrophoresis (SPEP) measures the amount of M proteins in the blood.77 Serum free light chain assay allows for estimating the amount of free light chain in the serum and is particularly useful in patients with light chain myeloma.

Tests for urine protein. Similar to tests for blood proteins, there are tests for urine proteins that measure the amount and types of M protein in the urine.77

Bone marrow aspiration and biopsy. To confirm diagnosis of myeloma, a small piece of bone marrow and a small amount of bone marrow aspirate are sampled for testing, typically from the hip bone. These tests can determine the number of myeloma cells in the bone marrow and also allow for additional tests on the myeloma cells such as proliferation rate and presence of genetic abnormalities.78,79

Imaging. X-rays may be used to check for bone damage or fractures resulting from myeloma; however, they are less accurate than other types of imaging. A low-dose computed tomography (CT) scan, which is a series of X-rays put together to assemble a single image,80 is sometimes used in combination with a positron emission tomography (PET) scan. A PET scan uses a drug that is injected into the bloodstream to identify possible sites of cancer with high sensitivity.81 The use of magnetic resonance imaging (MRI), or pictures taken using a series of magnets, has also increased recently as it is more accurate than traditional imaging approaches in identifying abnormalities in both tissue and bone marrow.82-85

5 Staging and Prognostics

Staging

After diagnosis of myeloma, staging is used to determine how much cancer is in the body. There are two main methods for staging myeloma. The International Staging System (ISS),86 which is the more commonly used method, classifies patients into one of three stages based on the amount of beta-2 microglobulin and albumin in the blood. The ISS was revised to include prognostic information based on blood lactate dehydrogenase and genetic abnormalities.7 This system is known as the Revised ISS (R-ISS). The second staging system, known as Durie-Salmon Staging,87 determines prognosis by classifying patients into one of three stages based on bone marrow tumor density and other markers of organ damage.

Table 2: Comparison of International Staging System and Revised International Staging System*

Method

Stage

Criteria

International Staging System (ISS)

1
 

Serum β2-microglobulin <3.5 mg/L,
Serum albumin >3.5 g/dL

2

Not meeting criteria for ISS Stage 1 or 3

3

Serum β2-microglobulin >5.5 mg/L

Revised International Staging System (R-ISS)

1

Criteria for ISS Stage 1, and
No high-risk chromosomal abnormality [(t4;14), t(14;16) del 17p], and
Serum LDH at or under upper limit of normal

2

Not meeting criteria for R-ISS Stage 1 or 3

3

Criteria for ISS Stage 3, and

High-risk chromosomal abnormality, or
Serum LDH over upper limit of normal

*To be interpreted with the help of a clinician skilled in the management of myeloma.
Derived from Palumbo et al.7 and National Comprehensive Cancer Network (NCCN) Guidelines.70
LDH, lactate dehydrogenase.

Prognostic Scoring and Risk Groups

Prognosis and risk stratification are important to help predict the course of disease and determine the likelihood that a patient progresses from MGUS to smoldering myeloma or from smoldering myeloma to active myeloma.88 Risk of disease progression is most commonly determined using the amount of myeloma cells in the body, blood labs, and other tests that look at genetic abnormalities.89 Additionally, risk stratification is used to guide myeloma treatment. In general, more aggressive treatment approaches are reserved for high-risk patients, while patients stratified into lower risk categories are usually considered for less aggressive treatments that limit exposure to side effects.90 However, how to best stratify patients and determine which patients are likely to benefit from early treatment is a matter of ongoing debate and research.

Risk-based stratification for smoldering myeloma. Smoldering myeloma is defined by low levels of M proteins in the blood and lack of end-organ damage at time of diagnosis; however, disease severity and progression risk in patients with smoldering myeloma can vary greatly. Therefore, many attempts have been made to develop methods to accurately categorize patients according to progression risk.

Developing better methods of identifying patients at high risk of progressing from smoldering MM to active MM and stratifying patients according to progression risk are very active areas of MM research. The International Myeloma Working Group (IMWG) is one of the groups at the forefront of myeloma research and has played an influential role in helping establish currently accepted risk stratification classifications. At a presentation at the 2019 American Society of Clinical Oncology annual meeting, researchers associated with the IMWG presented their latest model for risk stratification.91 The model assigns one point for each of the following risk parameters:

  • Free light chain ratio > 20,
  • Serum M spike > 2 g/dL,
  • Bone marrow plasma cell percentage >20%, and
  • Presence of a high-risk genetic factor [t(4,14), t(14,16), 1q gain, or del13q]

After the points have been assigned appropriately, scores are tabulated and assigned to progression risk categories as shown in Table 3.

Table 3: Risk of Progressing from Smoldering Myeloma to Active Multiple Myeloma

Risk Stratification Group

Number of risk factors (points)

Risk of Progression at 2 years

Low risk

0

8%

Low-Intermediate risk

1

21%

Intermediate risk

2

37%

High risk

≥3

59%

New strategies for risk-stratifying that utilize, for example, genomic analyses are being investigated. Recent evidence suggests mutations in genes involved in certain cell-signaling pathways (eg, MAPK, APOBEC) may help identify patients at higher risk of progressing from SMM to MM.92 However, these kinds of analyses are not yet generally included in standard risk-stratification assessments because they may not be available in all facilities and more research is needed to validate their predictive value. Hopefully, these kinds of emerging techniques will improve risk-stratification algorithms in the not-too-distant future.

Risk-based stratification for active myeloma. Bone marrow cytogenetics and fluorescence in situ hybridization (FISH) are two tests that can be used to measure genetic abnormalities in patients with active myeloma to evaluate the aggressiveness of disease.93 Abnormalities might occur when genetic material is lost from a chromosome, gained by a chromosome, or moves from one chromosome to another. The human body has 23 pairs of chromosomes. Examples of abnormalities that may put myeloma patients at higher risk for progression include translocation of genetic material between chromosomes 4 and 14 [abbreviated t(4;14)] or loss of genetic material from chromosome 17 [abbreviated del(17p)].88 It is recommended that all patients undergo cytogenetic evaluation at the time of diagnosis.90 Patients can be assigned to risk groups based on cytogenetic abnormalities as shown in Table 4.

Table 4: Risk-Based Stratification for Active Myeloma

Risk

Description

Standard

All other abnormalities, including t(11;14), t(6;14)

Intermediate

t(4;14), del(13), hypodiploidy, PCLI >3%

High

del(17p), t(14;16), t(14;20)

*To be interpreted with the help of a clinician skilled in the management of myeloma.
Derived from Mikhael, et al.90
IgH, immunoglobulin heavy chain; t(), translocation; del(), deletion; PCLI, plasma cell labeling index.

6 Conventional Treatments

Chemotherapy

Chemotherapy uses intravenous or oral drugs to kill abnormal cells or stop new ones from being made. Chemotherapeutics for treatment of myeloma are commonly used in combination with other drugs, such as steroids. These medications can also affect normal cells, so they are given in varying cycles of treatment days followed by rest. Chemotherapeutic agents may be used alone, prior to stem cell transplantation, or after stem cell transplantation for maintenance.

Proteasome inhibitors, one class of commonly used drugs, work by disrupting mechanisms that cancer cells use to avoid normal cell death.94 Bortezomib (Velcade) is one example of a commonly used proteasome inhibitor.95-98 It is used with lenalidomide (Revlimid), another anti-cancer agent, and dexamethasone, a steroid medication, for first-line treatment of myeloma.70,99-101 Bortezomib is given as four doses spread over a 21- or 28-day cycle. Side effects observed with this class of medications are similar to other chemotherapeutic agents and include numbness in the hands and feet, nausea, fatigue, constipation, and headache. It should be administered subcutaneously to reduce the risk of peripheral neuropathy.102

Preventing Proteasome Inhibitor-Induced Peripheral Neuropathy (PN)

Peripheral neuropathy from proteasome inhibitors is a treatment limiting side effect, meaning patients often must discontinue the drug due to the severity of the PN. Preventing PN from anti-MM drugs is ideal so the patient does not need to stop an otherwise effective treatment.

A phase II prospective study on 33 newly diagnosed patients with MM that were to begin treatment containing bortezomib was done to assess whether a combination of 400 mg docosahexaenoic acid (DHA), 600 mg α-lipoic acid, 60 mg vitamin C, and 10 mg vitamin E twice daily for six months could reduce the neurotoxicity of the treatment.103 At the time of analysis, there were 25 patients who completed the study. After six months of using the nutrient combination, 10 patients were free of symptoms and no one needed to discontinue treatment due to painful neuropathy.

Because vitamin C may interfere with bortezomib, at this time vitamin C is contraindicated in those taking bortezomib.104 Similarly, some in vitro evidence suggests lipoic acid may interfere with the anti-myeloma activity of bortezomib.105 Because some evidence suggests lipoic acid may be useful in the context of bortezomib-induced peripheral neuropathy, more studies are needed to assess the safety and utility of lipoic acid in the clinical setting among myeloma patients.


Potential Bortezomib Interactions with Natural Substances

Green Tea Extract

A study published in 2009 found green tea catechins, particularly epigallocatechin-3-gallate (EGCG), blocked the action of boronic acid-based proteasome inhibitor drugs (eg, bortezomib) in myeloma cells both in cell cultures as well as in mice.106 This interaction was not seen with non-boronic-acid-based proteasome inhibitors. Several other studies have reported that green tea antagonizes the action of bortezomib as well. Green tea extract should therefore be avoided by those taking bortezomib.104,107-109

Vitamin C

As mentioned in the vitamin C section later in this protocol, there was one animal study that suggested vitamin C may interfere with the anti-MM action of bortezomib.110 Several other in vitro studies have also identified some evidence that vitamin C may interfere with the action of bortezomib.110-112 On the other hand, a small clinical study found that a formulation of natural agents, including low-dose vitamin C, helped curb PN associated with proteasome inhibitors, so more studies are needed to assess the potential interaction between vitamin C and bortezomib. Nevertheless, most oncologists will err on the side of caution and recommend against the use of vitamin C in combination with bortezomib.103,113

The potential for interference by oral vitamin C with boronic-acid-based proteasome inhibitors appears to be unique to this class of drug only. Intravenous (pharmacologic) ascorbate has been safely combined with chemotherapies for many different cancers, including MM.114 There are also studies that have combined intravenous vitamin C with bortezomib and noted efficacy and safety rather than interference. Several trials have studied chemotherapy regimens for multiple myeloma using intravenous vitamin C combined with different regimens including arsenic trioxide (Trisenox), melphalan (Evomela), dexamethasone, and bortezomib, and found good tolerability as well as favorable efficacy.115,116

Lipoic Acid

Preliminary evidence from an in vitro study suggests lipoic acid may interfere with the anti-myeloma effects of bortezomib.105 When myeloma cells were incubated with alpha-lipoic acid (ALA) plus bortezomib, their viability was about twice that of myeloma cells incubated with bortezomib alone. Moreover, this same study showed that incubating myeloma cells with lipoic acid alone stimulated their growth and increased their viability compared with untreated control myeloma cells. The researchers who conducted the study concluded, “Our data suggest that ALA has the potential to antagonize the anti-MM effects of bortezomib based on our in vitro results using MM cell lines. Thus, it is possible that ALA could negatively impact the therapeutic benefit of bortezomib for MM patients and this requires further study especially if ALA is accepted as an intervention in bortezomib-related neuropathy.”

Immunomodulators

Immunomodulatory drugs (IMiDs) work against cancer through several mechanisms. They boost the immune system to allow the body to fight cancer. They can also slow cancer growth directly by cutting off the blood supply to the tumor and leveraging inflammatory pathways, though it is unclear how much these mechanisms contribute to the efficacy of IMiDs in myeloma.117,118

Thalidomide (Thalomid) was the first of this class of drug to be tested in myeloma, but its use has waned recently given its considerable severe side effect profile. At the time of this writing, lenalidomide, a more potent immunomodulator that has greater tolerability, is a mainstay of myeloma therapy.119,120 It can be used in combination with other drugs for the initial treatment of myeloma, or by itself for long-term maintenance therapy.70 Lenalidomide is given by mouth once daily for 21 days out of a 28-day cycle. Side effects of lenalidomide include low red blood cell, white blood cell, and platelet counts, diarrhea, and fatigue. Lenalidomide is extremely toxic to unborn children and can cause birth deformities. Women of child-bearing potential, as well as their partners, must use contraception when taking lenalidomide.121 Pomalidomide (Pomalyst) is another IMiD used in myeloma that has relapsed after prior treatment.

Glucocorticoids

Glucocorticoids are a type of steroid known to modulate gene expression and induce cancer cell death through various mechanisms.122 They are an important part of myeloma treatment.123

The most commonly used glucocorticoid is dexamethasone, which is used in combination with chemotherapeutic agents, immunosuppressants, and targeted therapies, such as bortezomib, lenalidomide, cyclophosphamide (Cytoxan), and daratumumab (Darzalex).124,125 Dexamethasone is part of most recommended drug regimens for both first-line and relapsed treatment of myeloma. Side effects of glucocorticoids include weight gain, fluid retention, increased blood sugar, weakened bones, and increased risk of infections.126,127 Due to these side effects, long-term use of glucocorticoids is not recommended.

Targeted Therapies

Targeted therapies are drugs that target specific mechanisms of myeloma. Unlike chemotherapy, these drugs are generally directed towards cancer cells and have less of an effect on healthy cells; therefore, they have fewer side effects.128

One example of a targeted therapy is daratumumab. Daratumumab is a therapeutic antibody that binds to proteins on myeloma cells and induces cell death.129,130 It is most commonly used in combination with other drugs and is part of a preferred regimen for patients who are not eligible to undergo a hematopoietic stem cell transplant (SCT).70 It can be administered weekly, every two weeks, or monthly, depending on how long the patient has been receiving it.131 Side effects of daratumumab may include fatigue, nausea, low blood cell counts, and infusion-site reactions when the medication is administered.132 Patients should receive other medications before taking daratumumab to reduce the risk of experiencing a skin reaction.

Stem Cell Transplant

Stem cell transplant (SCT) is a transplantation of hematopoietic stem cells, usually derived from bone marrow, peripheral blood, or umbilical cord blood of a donor to a recipient whose bone marrow was destroyed with high-dose chemotherapy prior to SCT. The goal of SCT is to fight the cancer by using high doses of chemotherapy to wipe out bone marrow cells, both cancerous and normal, while allowing healthy blood stem cells to grow from the infused stem cells and form new blood cells. This process, called engraftment, takes about 2 to 4 weeks.133-137 These new cells grow into healthy bone marrow and can produce non-cancerous blood cells.

There are two types of stem cell transplants—autologous and allogeneic. “Autologous” means the patient’s own stem cells are transplanted back into his or her body. “Allogeneic” means the stem cells of another person with similar characteristics are transplanted into the patient. Allogeneic transplantation is much riskier and patients receiving allogeneic transplants typically have more side effects than those receiving autologous transplants.138 It can also be difficult to find an appropriate donor. For these reasons, autologous transplants are considered the standard of care in patients with myeloma.70

Autologous SCT is an important part of myeloma treatment and should be considered in all eligible patients; however, it is an intensive procedure that not everyone can tolerate.70,139-141 Patients take medication before the procedure to wipe out the existing cancerous cells and prepare their body for transplantation, and again after the procedure to maintain control of the tumor.70,138 Whether SCT should take place early in the disease course or upon relapse is a matter of some debate. As of late 2020, evidence suggests early SCT results in deeper responses, but may not improve overall survival. Thus, the decision to undergo transplantation is an individualized choice and must be based upon several factors taken into consideration by the treating oncology team and the patient.142

After transplantation, response is typically measured by assessing serum or urinary monoclonal M-protein levels. Free light chain measurements can be used in patients with undetectable M-protein levels in their serum or urine. Bone marrow immunohistochemistry or immunofluorescence may be utilized as well. An approach taken by some experts is to assess patients 100 days after SCT, then reassess every three to four months thereafter if the patient is doing well. The degree of response following therapy may be predictive of survival, but this is controversial in the myeloma research community (see section titled “Evaluating Response” below).142

Serious side effects (eg, nausea, vomiting, weakened immune system) may result from medication. Eligibility for transplant depends on patients’ age, fitness, and other disease-related factors, but the decision to have a transplant should involve a discussion between the patient and his or her doctor.

Radiation

Radiation therapy uses strong beams of energy to kill cancer cells or stop their growth. The most commonly used radiation therapy in myeloma is external beam radiation therapy. It is predominantly used where there is a single, localized mass of myeloma cells called a plasmacytoma.70,143,144 Radiation therapy can also damage healthy cells and cause side effects. Common side effects of radiation-based therapies include pain or other changes to the skin at the site of radiation. Patients may also develop fatigue and nausea over time in response to radiation.

Supportive Care

Myeloma can cause damage to several organs, including the kidneys. It is important to stay hydrated and avoid nephrotoxic medications, such as NSAIDs, if a patient is experiencing kidney injury.145 Because myeloma reduces production of red blood cells, patients with low red blood cell numbers may develop anemia. Doctors may suggest treatment with erythropoietin (EPO) to help regenerate red blood cells.146,147 Myeloma also puts patients at increased risk for infection by suppressing the immune system. Vaccines and prophylactic antibiotics may be administered to reduce the risk of infection.148-150

Addressing Bone Health

Bone disease affects almost all myeloma patients. Approximately 60% of patients present with bone pain (particularly in the back or chest) at the time of diagnosis,17,38 and approximately 20‒25% present with pathologic fractures, compression fractures, or osteoporosis at diagnosis.17 These bone-related events represent a huge burden to myeloma patients, decreasing survival rates, increasing treatment costs, and reducing quality of life.38,151,152

Bone remodeling is an important part of maintaining healthy bone in all individuals. It is achieved through a delicate balance of the resorption of old bone (by cells called osteoclasts) and formation of new bone (by cells called osteoblasts).153 Increased expression of cytokines in myeloma patients contribute to an imbalance in bone turnover, characterized by increased osteoclast and reduced osteoblast activity.154,155 This means bone is being resorbed but not being replaced by new bone,156 which manifests as osteolytic bone lesions, weakened bone, and increased calcium release into the blood.

Due to the morbidity and mortality associated with myeloma bone disease, most experts recommend that MM patients with lytic bone lesions and/or osteopenia or osteoporosis be treated with an osteoclast inhibitor. However, the potential for benefit is less clear for patients with no apparent bone lesions or osteopenia/osteoporosis, so experts disagree on the best approach. Some recommend osteoclast inhibitors to all patients with MM requiring treatment regardless of whether they have bone disease, while others avoid these drugs in patients without bone disease. The decision to use an osteoclast inhibitor should be made on a case-by-case basis.70

Pamidronate and zoledronic acid (Reclast) are two bisphosphonates that work by reducing bone resorption and have both demonstrated a similar reduction of skeletal-related events in myeloma.157-161 Denosumab, a therapeutic antibody that also works by reducing bone resorption, has similar bone health benefits to bisphosphonates but is preferred in patients with kidney dysfunction.162 All subjects receiving either bisphosphonates or denosumab should receive calcium, vitamin D, and have regular dental monitoring for osteonecrosis of the jaw.70

Vitamin K and Bone Health in the Context of Multiple Myeloma

Vitamin K is a fat-soluble vitamin found in many foods and also available as a dietary supplement. There are two types of naturally occurring vitamin K: vitamin K1 and vitamin K2. Another form, vitamin K3, is a synthetic form that is converted to vitamin K2 in the liver after ingestion.163 Vitamin K has been shown to reduce bone resorption by inhibiting osteoclast activity through various pathways, resulting in improved bone health.164,165 One study showed Japanese women with osteoporosis receiving daily doses of 45 mg vitamin K2 (menatetrenone) had fewer fractures compared with those not receiving supplementation.166 Findings from two randomized controlled trials showed vitamin K2 provided a similar reduction in fractures, as well as favorable effects on bone mineral density, especially in Japanese patients.167,168 Daily supplementation with vitamin K1 (phytonadione) may also reduce the risk of fractures.169

Preliminary evidence from an in vitro study suggests vitamin K2 may exert some anti-myeloma effects in addition to its benefits for bone health (see vitamin K discussion in the “Integrative Interventions” section, below).170

It should be noted that the majority of evidence supporting positive effects of vitamin K on bone health are in women with existing osteoporosis or osteopenia, and even within this population, results from other studies are conflicting.171,172 Additionally, studies in men and premenopausal female athletes have shown no improvement in bone health.167,173 Rigorous studies of the effects of vitamin K on bone health in the context of multiple myeloma are needed.

7 Treatment Options

Treatment of Solitary Plasmacytoma

A solitary plasmacytoma is a single mass of plasma cells that has not yet developed into myeloma. This is a rare condition. Since this mass is isolated, it can be treated with radiation therapy and/or surgery, which may have curative potential. There are two types of solitary plasmacytomas: those originating in bone (osseous) and those involving soft tissue (extraosseous).70,174 Treatment of both types of solitary plasmacytomas with radiation and surgery (if needed) has resulted in favorable outcomes and disease control.175,176 Patients are closely watched after treatment to monitor for disease recurrence.

Treatment of Smoldering Myeloma

In general, progression from smoldering myeloma to active myeloma occurs in about 60% of patients within 10 years.177 However, the risk of progression varies greatly between patients, with some very high-risk patients having an 80‒90% likelihood of progression within two years. Treatment decisions depend on the likelihood that the patient will progress. Patients determined to be at low risk of progression may not require treatment and will instead be closely monitored at regular time intervals. On the other hand, those deemed to be at high risk of progression may undergo treatment with some of the same drugs used to treat active myeloma. There is much ongoing research attempting to clarify the best ways to identify which SMM patients are at high risk of progression as well as determine the ideal treatment regimen.178

The National Comprehensive Cancer Network (NCCN) guidelines70 recommend all patients with smoldering myeloma be monitored at three to six month intervals; however, the Mayo Clinic provides recommendations179 to determine the frequency of monitoring based on risk of disease progression. According to these recommendations, patients with low-risk disease may be monitored every six months to test for disease progression. Intermediate-risk patients can be re-tested for disease progression every three to four months. Patients with high-risk smoldering myeloma should be monitored every two to three months and should also be considered for enrollment in clinical trials to prevent or delay progression.

Some emerging evidence suggests early treatment can delay progression to active myeloma and extend overall survival, although this is very controversial as of late 2020 and more trials are needed to clarify overall benefits.70 One study on 119 patients with high-risk smoldering myeloma showed treatment with a combination of dexamethasone and lenalidomide prolonged the time to progression to active myeloma and extended overall survival at three years, compared with observation only. A subsequent analysis at a median of about six years showed lenalidomide continued to provide benefit in terms of time to progression: the treatment group had not yet reached their median time to progression, while the observation-only group had a median time to progression of 23 months. Eighty-six percent of participants in the observation-only group progressed to active myeloma, compared with only 39% of those in the lenalidomide plus dexamethasone group.180,181 A more recent study, published in 2020, showed lenalidomide alone delayed progression to symptomatic disease and lengthened progression-free survival among 182 patients with high-risk smoldering multiple myeloma. Half of the participants in the lenalidomide group exhibited a treatment response, and one-, two-, and three-year survival in the lenalidomide group was 98%, 93%, and 91%, compared with 89%, 76%, and 66% in the observation-only group. Subgroup analysis showed that the progression-free survival benefit was clear in high-risk patients, but not as apparent in those with intermediate-risk disease. Serious adverse events occurred in 41% of participants in the lenalidomide group.182

Based on promising results from these studies, some experts now recommend either lenalidomide plus dexamethasone or lenalidomide alone, rather than observation, for patients with high-risk smoldering multiple myeloma. It should be noted that the criteria used to identify patients at high risk for progression to active myeloma was not uniformly defined in these studies, and many experts continue to recommend that more intensive treatment for lower-risk SMM patients should be reserved for the clinical trial setting as opposed to routine practice.70 Importantly, this is a very active area of research and is a matter of much debate in the myeloma research community. Decisions to initiate early treatment must be made on a case-by-case basis and must involve careful consideration of risks and benefits by physicians experienced in treating myeloma.

Treatment of Symptomatic Myeloma

Patients with newly diagnosed symptomatic myeloma should receive treatment with a combination of medicines. Initial treatment depends on whether the patient is eligible for autologous SCT. Preferred drug regimens based on NCCN guidelines for patients that are eligible and those that are ineligible to receive a transplant are listed below. Patients should also receive a medication for bone health and other supportive care based on symptoms.

Table 5: Preferred Initial Treatment Regimens for Myeloma Based on NCCN Guidelines*

Myeloma Treatment for SCT-Eligible Patients

Bortezomib, lenalidomide, dexamethasone

Bortezomib, cyclophosphamide, dexamethasone

Myeloma Treatment for SCT-Ineligible Patients

Bortezomib, lenalidomide, dexamethasone

Daratumumab, lenalidomide, dexamethasone

Lenalidomide, dexamethasone (low dose)

Bortezomib, cyclophosphamide, dexamethasone

*To be interpreted with the help of a clinician skilled in the management of myeloma.
Derived from NCCN Guidelines70
SCT, stem cell transplant.

A risk-based treatment algorithm for patients eligible for autologous SCT is provided below. Initial treatment consists of three to four cycles of induction therapy using bortezomib, lenalidomide, and dexamethasone (VRd) in standard-risk and intermediate-risk patients or carfilzomib (Kyprolis), lenalidomide, and dexamethasone (KRd) in high-risk patients. All initial treatment regimens should include low-dose dexamethasone (40 mg once weekly) as it has been associated with higher overall survival and lower rates of toxicity compared with high-dose dexamethasone.183 Induction therapy is then followed by either early or delayed transplantation. Maintenance therapy with either lenalidomide or a proteasome inhibitor is provided after transplantation to reduce the risk of disease progression.70 Maintenance therapy can prolong progression-free survival and may improve overall survival. The selection of drug(s) used during maintenance therapy will depend on risk stratification and other patient-specific characteristics. Maintenance therapy is generally recommended for at least two years. Research is ongoing to determine if longer maintenance therapy provides benefits that outweigh drug toxicity risks.142

Table 6: Risk-Based Treatment Algorithm for SCT-Eligible Patients*

Standard Risk

Intermediate Risk

High Risk

Initial Treatment

VRd x3 to 4 cycles

VRd x3 to 4 cycles

VRd or KRd x3 to 4 cycles

Transplantation#

Early or Delayed ASCT

Early ASCT

Early ASCT

Maintenance Therapy

Lenalidomide

Bortezomib-based

Carfilzomib or bortezomib-based

*To be interpreted with the help of a clinician skilled in the management of myeloma.
Derived from Rajkumar, et al.88 and NCCN Guidelines70
#The decision to initiate ASCT early or delay the procedure until first signs of progression must be made on an individual basis. Whether early transplantation is superior to delayed transplantation is somewhat controversial, although early ASCT is considered standard of care by most institutions. More research is needed.
Abbreviations: VRd, bortezomib, lenalidomide, dexamethasone; KRd, carfilzomib, lenalidomide, dexamethasone; ASCT autologous stem cell transplantation.

Patients diagnosed with myeloma may be ineligible for SCT due to frailty or age. High-risk patients that are ineligible for SCT may receive primary treatment with bortezomib, lenalidomide, dexamethasone (VRd) followed by maintenance therapy with bortezomib or a bortezomib-based regimen. It is recommended that patients categorized as standard-risk receive daratumumab, lenalidomide, and dexamethasone (DRd) or bortezomib, lenalidomide, dexamethasone (VRd) followed by maintenance therapy with either lenalidomide and daratumumab or lenalidomide alone. Frail patients or patients over age 75 may receive lenalidomide and dexamethasone until disease progression.70

Table 7: Risk-Based Treatment Algorithm for SCT-Ineligible Patients*

Standard Risk

Intermediate Risk

High Risk

Initial Treatment

VRd or DRd x8 to 12 cycles**

VRd x8 to 12 cycles

Maintenance Therapy

Lenalidomide or lenalidomide-daratumumab

Bortezomib or bortezomib-based

*To be interpreted with the help of a clinician skilled in the management of myeloma.
**If patient is frail, treat with Rd until progression.
Derived from Rajkumar, et al.88 and NCCN Guidelines70
VRd, bortezomib, lenalidomide, dexamethasone; Rd, lenalidomide, dexamethasone.

Due to high rates of organ damage associated with active myeloma, all patients initially diagnosed with myeloma should be considered for appropriate supportive care. Patients receiving treatment for myeloma may also be prescribed medication to manage potential side effects. For example, patients receiving immunomodulatory agents (eg, lenalidomide), particularly in combination with high-dose glucocorticoids, are at significantly greater risk of thrombotic complications and should receive preventative aspirin therapy. Similarly, those receiving treatment with proteasome inhibitors should receive preventative antiviral therapy to prevent viral infectious diseases common in MM patients, such as shingles.70

Evaluating Response

After treatment, the doctor will evaluate how well the patient has responded based on symptoms and how many myeloma cells remain in the body. Response to treatment is generally categorized as complete, partial, minimal, or progressive, as defined below. Relapse is defined as an initial positive response to treatment followed by worsening of disease.

Table 8: International Myeloma Working Group Myeloma Response Criteria*

Category

Definition

Complete Response

No M proteins in the blood or urine, and
Under 5% plasma cells in the bone marrow
No evidence of plasmacytoma, if previously present

Partial Response

50% or greater reduction in M proteins in the blood , and
90% or greater reduction in 24-hour urinary M proteins, and
50% or greater reduction in plasma cells in the bone marrow, and
50% or greater reduction in the size of solitary plasmacytoma, if previously present

Minimal Response

25% to less than 50% reduction in M proteins in the blood, and
50% to less than 89% reduction in M proteins in the urine, and
25% to less than 50% reduction in size of plasmacytoma, if previously present

Progressive Disease

25% or greater increase in M proteins in the blood (at least 0.5 g/dL), or
25% or greater increase in M proteins in the urine (at least 200 mg/24 hours), or
50% or greater increase the number of plasma cells in the blood, or
10% or greater increase in the percentage of plasma cells in the bone marrow, or
50% or greater increase in the size of existing lesion or presence of new lesions.

*To be interpreted with the help of a clinician skilled in the management of myeloma.
Derived from Kumar, et al.184 and NCCN Guidelines70

Treatment of Relapsed Myeloma

The majority of myeloma patients will relapse at some point during the course of their disease.185 Treatment for relapsed disease should consider previous treatments, time to relapse, and disease severity. Patients that underwent successful autologous SCT for initial therapy and had a durable response can be considered for a second transplant.70 Preferred treatments for patients experiencing their first relapse include three-drug regimens containing either a proteasome inhibitor (eg, bortezomib, carfilzomib)186 or monoclonal antibody (eg, daratumumab, elotuzumab [Empliciti]).187,188 Treatment of subsequent relapses should focus on therapeutics not previously used to treat the initial relapse.

8 Novel and Emerging Therapies

This section outlines several clinical developments in the myeloma treatment pipeline. None of the therapeutics described in this section are approved for first-line myeloma treatment as of mid-2020, though some are approved for use in patients who have undergone prior therapies. These clinical developments represent some important advancements in myeloma research, but more research is required to translate these developments into routine clinical care.

Participating in a Clinical Trial

New treatment approaches for myeloma are currently being developed and researched. Especially for certain patients who do not meet the criteria for available treatments or see benefit when using current therapies, clinical trials may provide suitable options and should be considered when available.

The Multiple Myeloma Research Foundation provides information on finding and participating in clinical trials for investigative therapies.

Second-Generation Proteasome Inhibitors

First-generation proteasome inhibitors, such as bortezomib, are associated with serious adverse effects, such as numbness in the extremities. They are also associated with high rates of relapse. Second-generation proteasome inhibitors work the same way as first-generation therapies, but boast higher response rates, new routes of administration, and lower incidence of side effects.189 Carfilzomib is an approved second-generation proteasome inhibitor that is administered by intravenous (IV) infusion as part of combination therapy in patients previously treated for myeloma.70,190 Ixazomib (Ninlaro), an oral proteasome inhibitor administered weekly, is approved for treatment of myeloma.191 Oprozomib, an analog of carfilzomib that can be administered orally, is currently under investigation192; the FDA has designated it an orphan drug for the treatment of Waldenström’s macroglobulinemia. Oral medications for myeloma are becoming increasingly important as clinical advancements are made and patients live longer on therapies. Marizomib is another novel second-generation proteasome inhibitor that has shown promising results in patients with relapsed/refractory multiple myeloma.193,194

Monoclonal Antibodies

Daratumumab is a monoclonal antibody approved as monotherapy for multiple myeloma as of the time of this writing. Another monoclonal antibody, elotuzumab, is approved as part of combination therapy for relapsed or refractory MM. Several additional monoclonal antibodies are in various stages of clinical development. Because monoclonal antibodies can target many different parts of cancer cells, there are numerous possibilities for the development of effective therapies. For example, GSK2857916 (belantamab) is an antibody that targets the BCMA receptor on myeloma cells. It has been studied in a Phase I study and found to be well tolerated and efficacious in patients with relapsed/refractory myeloma.195,196 In August 2020, The FDA granted belantamab accelerated approval for use in relapsed MM patients who have undergone a minimum of four prior therapies.197

Interim analysis of a phase III clinical study (IKEMA) indicated the combination of a monoclonal antibody, isatuximab (Sarclisa), with carfilzomib and dexamethasone (known as Isa-Kd) increased progression-free survival in patients with relapsed or refractory MM compared with carfilzomib and dexamethasone without isatuximab (Kd) after a median follow-up of 20.7 months. In the Isa-Kd arm, nearly 30% of patients were negative for minimal residual disease (a measure of whether myeloma cells are still detectable in the body after treatment), while only 13% in the Kd arm were negative. The researchers concluded that Isa-Kd “represents a possible new standard of care in patients with relapsed MM.”198

One of the major research challenges that currently faces monoclonal antibody-based therapies is identifying adjuvants or combination therapies that effectively enhance response while maintaining a reasonable safety profile.196 Many clinical trials of monoclonal antibodies are recruiting or are in the pre-recruiting phase as of the time of this writing. More information about these trials is available at ClinicalTrials.gov.

CAR-T Therapies

Chimeric antigen receptor T-cell (CAR-T) therapy is a technique that removes immune cells from the body and modifies them to recognize cancer cells. The immune cells are then put back in the body to fight cancer. CAR-T treatments have been recently approved for other cancers and have the potential for long-term disease control, but are still under investigation for myeloma.199-201 The results from a Phase I trial investigating bb2121, a CAR-T therapy that targets B-cell maturation antigen (BCMA), were recently published.202 Myeloma patients with refractory disease responded well to the treatment; however, a large number of subjects experienced side effects, including neurotoxicity. A clinical trial is underway to compare bb2121 to standard triplet therapy in a Phase III study of patients with relapsed and refractory myeloma.203

Vaccines

Studies are currently investigating vaccines that target proteins highly expressed by myeloma cells. Vaccines are generally well-tolerated with fewer side effects compared with conventional therapies. One promising vaccine candidate (ImMucin) under development was recently studied in a Phase I/II clinical trial204 that included MM patients with disease that progressed following autologous SCT. Of the 15 patients in the trial that received the vaccine, 11 either demonstrated improvement or had disease that remained stable through the follow-up period of 41 months.

Histone Deacetylase Inhibitors (HDACis): Panobinostat

Due to the propensity of multiple myeloma to relapse and progress to refractory disease, many anti-cancer drugs typically used in other cancers have been tested specifically in the setting of relapsed or refractory myeloma in hopes of mitigating treatment failure and relapse. Histone deacetylase inhibitors (HDACis) are one class of drug that has shown promise in this setting, with panobinostat (Farydak) in particular demonstrating evidence of efficacy and tolerability. Panobinostat received accelerated FDA approval in 2015 for use in MM patients who have already received at least two prior treatment regimens, including bortezomib and an IMiD.205 A 2019 meta-analysis of data from 19 trials and nearly 2,200 patients found that panobinostat led to a more robust overall response rate (0.64) than two other HDACis, vorinostat (Zolinza) and ricolinostat (ORRs 0.51 and 0.38, respectively). The ORR was 0.36 for patients whose myeloma was refractory to bortezomib and 0.43 for those whose myeloma was refractory to lenalidomide. The researchers concluded panobinostat-containing regimens were effective and tolerable for patients with relapsed, refractory MM.206 More trials are needed to further validate the superiority of panobinostat over other HDACis in the setting of refractory MM.

Repurposed Drugs with Potential Utility in MGUS/SMM/MM

Interest in repurposing existing drugs for use in the prevention and/or treatment of cancer has increased over the last few decades. Repurposed drugs typically have a cost advantage and more thoroughly documented safety profile than new cancer drugs recently brought to market. The disadvantage is usually less robust efficacy data due to a lack of financial incentive for the private sector to conduct clinical trials of existing drugs for off-label use.207 Nevertheless, some drugs have sound biological plausibility for use in cancer and are supported by observational evidence or preliminary trial data suggesting potential benefit in the context of certain cancers. Below are brief summaries of several such repurposed drugs that may be promising in the context of multiple myeloma and its precursor conditions.

Table 9: Repurposed Drugs with Potential Utility in MGUS/SMM/MM

 

Potential for Use in MGUS/SMM/MM

Drug

Standard Use

Preclinical Data/ Biological Plausibility?

Observational Data in MGUS/SMM/MM?

Interventional Data in MGUS/SMM/MM?

Beta-Blockers (propranolol)

Primarily Cardiovascular

Yes208-217

Yes218

No

Statins (eg, simvastatin, lovastatin, atorvastatin)

Cholesterol Management

Yes219-223

Yes224-227

Yes, but preliminary228,229

Metformin

Diabetes/ Glucose Control

Yes230,231

Yes232-235

No

PDE-5 Inhibitors (eg, sildenafil, tadalafil)

Male sexual dysfunction, benign prostatic hyperplasia, pulmonary hypertension

Yes236-246

No

Yes, but preliminary, with one small, unpublished trial (n=13) in MM demonstrating lack of response resulting in early termination.247 One case report in MM,248 and one phase II study in Waldenström’s macroglobulinemia,249 which is a distinct cancer but similar to MM.

COX-2 Inhibitors (eg, celecoxib)

Analgesic, anti-inflammatory, antipyretic

Yes239,250-261

No

Yes, but results available as of mid-2020 are not suggestive of benefit262,263

All-trans Retinoic Acid (ATRA)

Acute promyelocytic leukemia

Yes237,264-277

No

Yes, but preliminary.278-280 One study showed ATRA was only effective in combination with chemotherapy, not alone.281

Antihistamines (eg, cimetidine, loratadine, ranitidine)

Allergic conditions

Yes282-305

No

Yes, but preliminary306

Interleukin-2 (eg, low-dose Proleukin)

Immunotherapy in some cancer treatment contexts, but use is limited

Yes307-312

No

Yes, but preliminary.313,314 One study found no benefit of rIL-2 in combination with rHuman-IFNα-2a315

Clarithromycin

Macrolide antibiotic used in H. pylori eradication and some other infectious contexts

Yes316-318

Yes (limited)319

Yes, but preliminary320-325

*Although the drugs presented in this table are supported by varying degrees of preliminary evidence suggesting they could theoretically be beneficial in the context of MGUS/SMM/MM, none have been validated in rigorous clinical trials. Moreover, some of these drugs may cause drug interactions or side effects, some of which could be serious. Adding off-label repurposed drugs to a standard myeloma treatment regimen should not be undertaken without the consent and oversight of a qualified healthcare provider.

More Information about repurposing drugs for use in the context of cancer in general is available at Life Extension’s Drug Repurposing in Cancer protocol.

9 Potential Targets of Opportunity to Control MM or MGUS and SMM Progression to MM

As research progresses, the molecular biology of MGUS, SMM, and MM is continually being elucidated, and targeted approaches have helped improve treatment of MM, particularly over the past decade. Some of the pathways involved in the perpetuation and progression of MGUS/SMM/MM may also be targeted by natural agents or repurposed drugs. Below are some pathways that are appealing as potential intervention targets with natural agents or repurposed drugs.

Nuclear Factor-Kappa B (NF-κB)

Nuclear factor-kappa B (NF-κB) regulates the transcription of numerous genes involved in proliferation, angiogenesis, survival, and apoptosis, making NF-κB an attractive target for control of the disease.326 Some drugs currently approved to treat MM suppress NF-κB, including thalidomide and bortezomib.327

Constantly activated NF-κB in myeloma results in higher levels of inflammatory mediators [eg, insulin-like growth factor, interleukin-6, vascular endothelial growth factor, and macrophage inflammatory protein-1alpha (MIP-1α)] in the microenvironment of the bone marrow.328 The net effect of these mediators is increased bone degradation and proliferation of myeloma cells.

NF-κB is involved in the production of osteoclasts (osteoclastogenesis) through the action of cytokine receptor activator of NF-κB ligand (RANKL). Denosumab, a monoclonal antibody, is used to control bone loss in MM patients through the inhibition of RANKL binding to the receptor activator of NF-κB (RANK), which normally stimulates osteoclast-driven bone loss. Inhibition of NF-κB may result in interference of RANK/RANKL osteoclastic bone loss as well.

There is emerging information that suggests there is a positive feedback loop between osteoclasts and myeloma cells, creating a vicious cycle of proliferation and activation of both cell types.329 NF-κB is an integral part of this cycle. Thus, suppressing NF-κB has the potential to simultaneously disrupt osteoclast generation and myeloma cell proliferation/activity.

Among the numerous natural agents that suppress NF-κB,curcumin is one of the most well studied.330,331 Additional natural suppressors of NF-κB activation include resveratrol, ursolic acid,capsaicin, silibinin,silymarin, guggulsterone, and plumbagin.332-334 Quercetin has evidence of interfering with NF-κB activation in addition to simultaneously increasing the action of osteoblasts.335 Honokiol, a polyphenolic compound from Magnolia officinalis, has demonstrated a dual action of both being anabolic as well as anti-catabolic on bone through the disruption of NF-κB mediated bone loss.336-338 Glycyrrhetinic acid, a compound found in licorice root, also inhibits RANKL-induced osteoclastogenesis through suppression of NF-κB.339,340

MAPK Pathway

Mitogen-activated protein kinase (MAPK) pathways play critical roles in cellular proliferation and differentiation.341 Mutations in genes involved the MAPK pathway are among the most common in MM, present in up to half of newly diagnosed MM patients.342,343 MAPKs are grouped into three families: ERKs, JNKs, and p38. One of the most frequent ways the MAPK pathway is overactivated is via mutations in RAS proteins, which regulate cell growth, proliferation, and differentiation.344 Recent studies using next-generation sequencing (NGS) have shown that the RAS protein family mutations accumulate during myeloma progression. NRAS and BRAF mutations that result in their constitutive activation are found in both MGUS and MM cells.345 Thus, the dysregulated MAPK pathway represents an appealing target through which novel therapies might disrupt the progression from MGUS to MM.

Interestingly, statin drugs have piqued the interest of cancer researchers due to evidence suggesting they can inhibit some processes that activate the MAPK pathway. Statins appear to inhibit prenylation of small GTPases such as Ras and Rho, which activate the mitogen-activated protein (MAP) kinase MEK/ERK cascade regulating proliferation, survival, and apoptosis.346-349 In bone and prostate cancer cells, statins have been shown to decrease proliferation and induce apoptosis through inhibition of the ERK/Bcl-2 pathway.350-352 Observational evidence suggested statin use may be associated with lower MM incidence as well as better outcomes in MM patients.224,226,227,353 Some preliminary interventional evidence suggests statins may be helpful in the context of MM,228,229 but more rigorous research is needed to clarify whether statin use meaningfully affects the clinical course of MM or its precursor conditions.

PI3K/AKT/mTOR Signaling Pathway

The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is aberrantly activated in MM cells, and its inhibition induces apoptosis.354,355 Because it plays a critical role in regulating proliferation, growth, survival, and migration of malignant plasma cells, the PI3K/AKT/mTOR pathway is central in MM pathophysiology and disease progression. As such, the mTOR pathway has become an emerging therapeutic target in MM.354,356-358

Metformin, a first-line diabetes drug with many potentially beneficial off-target effects, has emerged as a potentially viable therapeutic agent in some cancers.359-365 Because metformin targets multiple signaling pathways in cancer cells, many questions remain unanswered concerning relevant mechanisms of action.363,366 However, two potential accepted anti-cancer mechanisms of metformin have been proposed. First, metformin activates AMP-activated protein kinase (AMPK) resulting in inhibition of downstream AKT/mTOR signaling and consequent suppression of cell proliferation.367-369 Consistent with this postulate, metformin was shown to induce cell cycle arrest without apoptosis by activating AMPK and inhibiting mTORC1 and mTORC2 and downstream pro-survival signaling pathways including AKT in human MM cell lines.370 Metformin was also shown to inhibit IL-6 signaling and increase cell death via AMPK activation and mTOR inhibition in human MM primary cells and cell lines.230 Second, metformin’s anti-cancer effects appear to also be driven by reduced circulating levels of growth factors insulin and insulin-like growth factor 1 (IGF-1), which prevents activation of AKT/mTOR downstream signaling to inhibit cell proliferation and induce apoptosis.371,372 Indeed, metformin has been shown to inhibit proliferation and induce apoptosis of MM cells by inhibiting the PI3K/AKT/mTOR downstream signaling pathway.373

Angiogenesis

Angiogenesis is the growth of new blood vessels. These blood vessels provide blood and nutrients for tumor growth.374 Angiogenesis is needed to create a conducive microenvironment for the perpetuation of MM in the bone marrow.375 Studies suggest an increase in angiogenesis (ie, the angiogenic switch) is an essential step in the progression from plasmacytoma or MGUS to MM.376 Angiogenic mediators arising from clonal plasma cells as well as resident osteoclasts and stromal cells in the bone marrow are associated with increased aggressivity and worse prognosis. Several drugs used to treat MM are primarily anti-angiogenic agents (eg, thalidomide),377 suggesting this may be a valuable therapeutic tactic in the control of MM.

Natural compounds with anti-angiogenic effects deserve to be investigated for those with MGUS or SMM, where the intent is not necessarily to treat but to prevent progression to MM. The types of naturally occurring compounds that have antiangiogenic effects include polyphenols, alkaloids, phytohormones, and terpenes that can be found in fruits, vegetables, herbs, and spices.378 Some of the best researched antiangiogenics from nature include ellagic acid from berries,genestein from soy, resveratrol, silymarin, and epigallocatechin 3-galate (EGCG).377

Immune-related Targets

In addition to aberrant signaling, malignant cells exploit a wide range of immune escape mechanisms, including induction of an immunosuppressive tumor microenvironment to evade immune destruction. Myeloid-derived suppressor cells (MDSCs), a diverse population of immature myeloid cells that expand during cancer and have potent immunosuppressive activity, play a key role in the pathophysiology of MGUS and MM.379,380 The metabolism of the non-essential amino acid L-arginine was the first identified mechanism for MDSC immunosuppression.236,381 Specifically, L-arginine serves as a substrate for inducible nitric oxide synthase (iNOs, generating NO) and arginase 1 (Arg-1). The upregulation of both enzymes in MDSCs leads to a shortage of L-arginine in the tumor microenvironment, and consequently to the impairment of T-cell function.380 Furthermore, the increased NO production leads to suppression of T-cell function through the inhibition of the IL-2 downstream pathway.382,383

From a systemic perspective, there are many natural agents that support immune function and may lessen the risk of infection. General immune support to prevent infection should begin with ensuring the nutrients supportive of immune function (eg, vitamin D, zinc) are replete.384 Then, extracts from plants or mushrooms that have been used traditionally for immune support as well as validated with current research may be considered. A few of the most well-known immune supportive natural agents include echinacea, elderberry, reishi, and maitake.385 In a study of mice with plasmacytoma, echinacea added to their chow led to an increase in natural killer (NK) cells in the bone marrow and spleen, partly negating the immunosuppression caused by the plasmacytoma cells without affecting either T- or B-cell immune lineages (ie, humoral immunity).386

Interleukin-6 (IL-6) is an immune cytokine that perpetuates clonal populations of plasma cells in an autocrine and paracrine fashion.387 IL-6 is a key messenger in the microenvironment of the bone in MM and inhibition of its signaling has recently been revived as a potential therapy for MM.388 While direct inhibition of IL-6 through monoclonal antibody binding has not led to benefit, targeting nearby stromal cell (eg, adipocyte) production of IL-6 may offer a more effective strategy.389 IL-6 may be inhibited by natural agents such as gossypol, parthenolide, and curcumin.390

There have been two case reports of complete remission, and as is the case with most spontaneous remissions, it appears they were immune-mediated. The first case is from 1955, in which a male with hepatitis had resolution of his underlying multiple myeloma after his illness.391 The second case was of a woman who began using a Chinese medicine combination, Huangqi Guizhi Wuwu Tang (HGWT), a formula with a large amount of astragalus.392 Astragalus is an immune tonic commonly used in Asian medicine. The patient had stable disease until the published report in 2017. During the course of her 18 years with stable disease, she did have progression at one point. Her dose of astragalus was quadrupled at that time from 30 grams to 120 grams daily, and stabilization resumed.

10 Dietary and Lifestyle Considerations

Dietary Considerations

A number of studies have evaluated associations between certain dietary habits and risk of developing multiple myeloma. A study published in 2007 found that greater consumption of specific foods was associated with reduced risk of multiple myeloma among women in Connecticut. These foods included cooked tomatoes, cruciferous vegetables, fresh fish, and food-derived vitamin A. On the other hand, this same study found that greater consumption of cream-based soups, jello, ice cream, and pudding was associated with increased multiple myeloma risk. This study also showed trends suggesting that increased carbohydrate consumption may be associated with increased risk, whereas consuming more vitamin D and calcium may be associated with reduced risk.393

A 2001 study examined dietary patterns among 539 people with multiple myeloma and 1,989 control subjects. The researchers found that greater consumption of fish and cruciferous vegetables was associated with reduced risk of multiple myeloma. Interestingly, this study also found that use of vitamin C supplements was associated with lower myeloma risk.394

In 2016, researchers in Italy published a study in which they examined the associations between various foods of animal origin and risk of non-Hodgkin lymphoma and multiple myeloma. They included data from 33 independent studies representing over 16,000 people with non-Hodgkin lymphoma and over 3,600 people with multiple myeloma. The analysis showed that greater consumption of fish and seafood was associated with lower multiple myeloma risk, while increased red meat and dairy consumption was associated with increased non-Hodgkin lymphoma risk. The researchers concluded that “Foods of animal origin likely play a role in the aetiology of non-Hodgkin lymphoma and multiple myeloma, with red meat and dairy tending to increase the risk, and fish that tends to decrease it. Our findings reinforce the recommendations to reduce the consumption of red meat by replacing it with vegetables, legumes and fish.395

A remarkable study published in February 2020 by a collaborative group of researchers from prestigious universities and hospitals across the United States found that diet may not only influence the risk of developing myeloma, but also survival after diagnosis. This study included prospective survival analyses of 423 cases of MM from the Nurses’ Health Study and the Health Professionals Follow-Up Study. The researchers examined how closely the subjects adhered to healthy eating indices prior to diagnosis and their subsequent survival and cause of death. They found that for each standard deviation increase in adherence to a healthy eating index, death specifically related to MM was reduced by 15‒24%. Conversely, MM-specific death was 16‒24% more common for each standard deviation increase in adherence to an unhealthy dietary pattern. The researchers remarked, “… our consistent findings for multiple dietary patterns provide the first evidence that MM patients with healthier pre-diagnosis dietary habits may have longer survival than those with less healthy diets.”396

The study described in the previous paragraph was a follow-up to a 2019 study conducted by the same research group in which they evaluated dietary patterns and myeloma risk. In this study, the scientists found that proinflammatory dietary patterns and dietary patterns associated with insulin resistance were linked with increased myeloma risk among men. Specifically, adherence to an inflammatory dietary pattern was associated with a 16% increased multiple myeloma risk. Suggestive (though not statistically significant) positive associations were also observed between myeloma risk and greater adherence to dietary patterns linked to insulin resistance. For the inflammatory dietary pattern, adherence was determined by assessing frequency of consumption of foods predictive of three inflammatory markers: IL-6, CRP, and TNF-α. Foods associated with increases in these inflammatory biomarkers included processed red meat, refined grains, and high-sugar beverages. Conversely, foods associated with lower levels of these inflammatory biomarkers included green leafy vegetables and coffee.397

Other evidence suggests greater fruit intake may be associated with lower risk of progressing from MGUS to MM. In a study published in 2018, researchers analyzed data on over 5,700 participants who had submitted food frequency questionnaires as part of a population-based study in Iceland. The analysis showed people with MGUS who ate fruit at least three times per week during late life were much less likely to progress from MGUS to MM than those who ate fruit less frequently.398

Mental Health

Myeloma is a disease that suppresses the body’s ability to fight infections. Mental health is an important part of maintaining a healthy immune system.399 Patients should focus on reducing stress and getting enough sleep to support immune health. A myeloma diagnosis can be overwhelming. There are support groups available that can help patients better understand their condition and help them through this difficult time.

Stop Smoking

There is a strong association between smoking and the development of myeloma, especially in women and heavy smokers.60,61 For patients with myeloma who smoke, it is imperative to stop. Various tobacco cessation programs are available through nonprofit organizations that offer helpful tips and resources.149,160 The Centers for Disease Control and Prevention (CDC) also provides resources to aid in quitting tobacco smoking.

Body Mass and Physical Activity

Studies have shown that people who are obese (defined as a BMI of 30 kg/m2 or greater)400 are at increased risk for myeloma.60 Eating healthy to maintain an appropriate weight is an important lifestyle modification not only in the context of multiple myeloma, but in general. Exercise is also effective for helping maintain a healthy BMI.60 It is important to note that bone disease from myeloma puts patients at increased risk of fracture when exercising. Patients should have a discussion with their physician about the risks and benefits of physical activity before initiating exercise regimens.

Table 10: Body Mass Index (BMI) Calculations*

Measurement Units

Formula and Calculation

Example

Kilograms and meters

weight (kg) = BMI kg/m2
[height (m)]2

The formula for BMI is weight in kilograms divided by height in meters squared. If height has been measured in centimeters, divide by 100 to convert this to meters.

81 kg = 25 kg/m2
1.8 m2

Pounds and inches

703 x weight (lbs) = BMI kg/m2
[height (in)]2

When using American measurements, pounds should be divided by inches squared. This should then be multiplied by 703 to convert from lbs/inches2 to kg/m2.

703 x 180 lbs = 25 kg/m2
71 in2

*To be interpreted with the help of a skilled clinician.

11 Nutrients

The integrative interventions described below may complement conventional myeloma treatments. Although these interventions have been shown to positively influence certain disease-related parameters or modify biological pathways involved in myeloma pathobiology, patients should always consult clinicians skilled in the management of myeloma before starting a new regimen with any agent.

Note that some of the interventions described here have sound biological plausibility to be supportive in the context of MM or its precursor conditions but have not necessarily been validated in rigorous clinical trials. The natural agents discussed in this section are covered in detail due to at least some research indicating their usefulness for MGUS/SMM/MM specifically. Most of the data on the agents is preliminary and trials, when available, are small. Regardless, the following natural agents may have potential benefits in those with MGUS/SMM/MM.

Agaricus Mushroom

Agaricus blazei Murill (AbM) is traditionally used as both an edible and medicinal mushroom. AbM has immunomodulating properties that may benefit those with MGUS/MM.401 Mushrooms generally contain immune-supportive polysaccharides, and AbM possesses potent anti-angiogenic effects that are attributable to several compounds.402 AbM is rich in beta-glucans, which are known to modulate the immune system by activating the complement system.403,404 AbM also contains unique compounds such as agaritine, which was shown to induce death of leukemia and myeloma cells in culture.327 The mushroom’s antitumor properties have been demonstrated in mouse models of several different cancers.405

In addition to the well-characterized immune effects, AbM has anticlastogenic action, meaning it protects against the occurrence of chromosomal damage.406,407 This may be particularly relevant for MM given that genetic translocations and deletions are integral to the progressive pathogenesis from MGUS to MM to refractory MM.

The potential use of AbM for MGUS/MM is based on a small study that needs verification through larger clinical trials.331 In the study, 40 patients were randomized to receive placebo or a product with three mushroom extracts called AndoSan, (82.4% AbM, 14.7 % Hericium erinaceus, and 2.9% Grifola frondosa) in combination with chemotherapy and SCT. Patients receiving AndoSan (n=19) had enhanced immune responses based on lab findings. However, there was no difference in clinical outcomes between the treatment and placebo groups. Given the preliminary data that indicates it may be useful for those with MM, AbM is an intriguing option for immune support. Further research may elucidate the form, dose, and patient populations that may derive the most benefit from its use.408

Arabinoxylan

Arabinoxylan, a hemicellulose (ie, a class of compounds found in plant cell walls) derived from the nutrient-rich hard outer layer of rice, has been shown to stimulate the immune system. Biobran, or MGN-3, is a patented product containing arabinoxylan that uses enzymes from shitake mushroom (Lentinus edodes mycelia) to break down a particular hemicellulose compound (hemicellulose B) and render a unique immunomodulatory product. MGN-3 has demonstrated immunomodulatory effects both in lab tests and in humans.332,333,409 Several animal cancer models showed arabinoxylan has antitumor and chemopreventive properties—effects possibly attributable to immune modulation and induction of cancer cell apoptosis.410-412

Preclinical studies have shown arabinoxylan can stimulate macrophage phagocytotic activity, enhance NK cell activity, and promote production of IL-2 (a cytokine involved in modulating white blood cells).410,413-416 MGN-3 was also shown to boost NK cell activity in a small clinical trial that enrolled older participants.417 In addition, a clinical study in 80 healthy participants demonstrated that arabinoxylan supplementation increased production of interferon (IFN)-γ, a cytokine essential for innate and adaptive immunity.418 These results are intriguing because immune suppression is a hallmark of MGUS/SMM and myeloma.

In a 2018 review of 11 clinical studies with cancer patients (including myeloma), the researchers concluded that MGN-3 can “complement the conventional cancer treatment through upregulating the patient’s immune system, especially in boosting the NK cell activity… It may be used as a complementary immune therapy to reduce side effects, improve treatment outcomes, and enhance long-term survival rate.”419 One study found supplementation with MGN-3 improved survival in patients with various malignancies. The investigators noted that most of the studies included were small and of short duration.

MGN-3 has been tested specifically in myeloma. An in vitro study using a cell line of human multiple myeloma (U266) cells found inhibition of MM proliferation when either MGN-3 or curcumin was added to the cells’ medium. When the two were combined (100 µg/mL MGN-3 plus 10 µM curcumin) there was a synergistic effect that resulted in an 87% decrease in U266 myeloma cell count and 2.6-fold increase in apoptosis.420 A small pilot study assessed the use of curcumin (6 grams/day of 95% curcuminoids) and MGN-3 (2 grams/day) on the complete blood counts of 10 MGUS/MM patients and 10 patients with stage 0/1 chronic lymphocytic leukemia (CLL) over a six-month period.421 Half of the MGUS/MM patients were neutropenic at baseline. The intervention increased neutrophil counts in eight of the 10 participants with MGUS/MM. In addition, four of the 10 MGUS/MM patients experienced a reduction in erythrocyte sedimentation rate (ESR), indicating lower systemic inflammation. Additionally, in a small randomized study of 48 myeloma patients (32 receiving 2 grams MGN-3 daily and 16 receiving placebo), MGN-3 increased the activity of the innate immune system, including increasing NK cell activity, myeloid dendritic cell level, and Th1-related cytokine activation, all of which target imbalances inherent to MM.332 Further studies on the use of MGN-3 in those with MGUS/MM are warranted.

Curcumin

“Curcumin” is a term often used loosely to refer to turmeric root (Curcuma longa) extracts containing naturally occurring curcuminoids (eg, curcumin, desmethoxycurcumin and bisdesmethoxycurcumin). Studies on curcumin are generally done using a highly concentrated extract from the turmeric root containing >90% curcumin rather than the whole turmeric root, which contains only 3‒5% curcumin. This is an essential consideration since studies suggesting curcumin’s benefit in MGUS/MM have resulted from very high doses of curcumin.

Studies of curcumin in potentially modulating MGUS/MM have shown possible mechanisms including suppressing proliferation, inducing apoptosis, and inhibiting osteoclast formation.384,422,423

The anti-MM effects of curcumin are intriguing enough to elicit ongoing research into curcumin analogs (drugs) in the search for a novel multi-targeted anti-myeloma agent.424-426 Many of the well-established pathways of myeloma growth and progression, including STAT3, MAPK, IL-6, NF-κB, and RANKL are affected by curcumin either directly or indirectly.

The transcription factor NF-κB is often involved in myeloma, and its liberation from the cytosol to the nucleus is suppressed by curcumin.326 Specific to MM, the addition of curcumin (as diferuloylmethane) reestablished apoptosis through suppression of NF-κB.427 Myeloma is one of many cancer types in which the pro-apoptotic effect of curcumin has been demonstrated.428

STAT3 must be phosphorylated to be activated, a process that can be induced by IL-6, a cytokine often high in those with MM. A cell study showed curcumin rapidly and reversibly blocked the activation of STAT3 by IL-6 at doses that can be achieved in humans (10 μM).429

Two small studies evaluated high doses of curcumin in people with MGUS/MM. In one study of participants with MGUS (n=26), daily supplementation with curcumin (4 grams/day) led to favorable effects, including a reduction in M protein for those with the highest levels (>20 grams/L) and a reduction in markers of bone resorption (urinary N-telopeptide of type 1 collagen) in 27% of patients.430

In the second study, patients with MGUS and SMM were randomized to receive either 4 grams curcumin per day or placebo.431 The study groups crossed-over after three months, switching from the initial group to which they were randomized to the other group. So, subjects who started the study taking curcumin crossed over to taking placebo at three months. After the completion of the six-month crossover study, participants were given the option of continuing in a open-label study using higher-dose curcumin (8 grams daily). Curcumin supplementation led to improved markers of disease progression and bone turnover. The authors concluded that “… curcumin might have the potential to slow the disease process in patients with MGUS and SMM.”

In a dose-ranging study (2, 4, 6, 8, 10, and 12 grams/day) of 12 weeks duration, a minimum of six patients with MM were recruited to each dose either with or without 10 mg/d of Bioperine.432 Patients with at least stable disease at 12 weeks were allowed to continue in the trial up to one year. While there were no objective responses (n=29 participants), 12 patients continued treatment for more than 12 weeks and five were stable at one year. There were significant reductions in constitutively active NF-κB, STAT3, and COX-2 at each monthly time point for most of the participants. The authors proposed that curcumin should be “further investigated either alone or as a modulator of chemo-resistance in combination with other active agents.”

There is a published case of stabilization of disease with the use of high-dose curcumin as a single agent.433 A 57-year-old female who had been diagnosed with ISS stage III MM and treated with two prior lines of standard therapy was given 8 grams/night of curcumin. All objective indicators of MM disease (M protein, bone imaging) stabilized, and the patient had no further disease progression at the time of the publication, which was five years.

Multi-drug resistance is a major hurdle in the control of MM, with eventual clonal expansion of drug resistant populations for every targeted drug currently used for the disease. In addition to direct anti-MM effects of curcumin, there is preliminary data on its role in sensitizing MM cells to anti-MM drugs. For example, curcumin enhanced the cytotoxicity of carfilzomib on MM cells (U266 cells) through multiple mechanisms, including NF-κB and induction of cell cycle arrest.434 In a mouse model of MM, curcumin prevented chemoresistance of bortezomib and thalidomide, potentiating their cytotoxic effects through NF-κB‒mediated mechanisms and increased apoptosis.435 Clinical trials using curcumin alongside targeted agents for MM should be considered to ensure the combination is safe and determine an optimal dose for this application.

Sea Cucumber

Sea cucumbers belong to the phylum Echinodermata (which also include starfish and sand dollars). Various species of sea cucumber have been valued for centuries as a traditional medicine and functional food with various bioactivities. These organisms contain a multitude of compounds with therapeutic properties, including triterpene glycosides, carotenoids, chondroitin sulfates, and collagen, as well as many vitamins, minerals, and more.436 One sea cucumber formulation, TBL-12, has already undergone phase II clinical trials for asymptomatic multiple myeloma.437

Various sea cucumber extracts have demonstrated properties that may be useful for patients with MGUS/SMM or active myeloma. Preclinical in vitro and animal studies have shown sea cucumber extracts have potent antitumor effects.438,439 The extracts appear to exert their effects through many mechanisms that are important in the context of myeloma, including inhibiting angiogenic factors,440,441 activating immune NK cells,442,443 and stimulating macrophages via the NF-κB and MAPK signaling pathways.444,445 The immune-enhancing effects may be helpful to slow down the progression of MGUS/SMM to active myeloma, while the antitumor effects may be beneficial for active disease. Sea cucumber extracts have also demonstrated anti-inflammatory activity in preclinical studies. This is important, as inflammation is a critical driver in the pathogenesis of MGUS/SMM and myeloma.446 In immune-suppressed rats, a sea cucumber extract reduced serum levels of inflammatory factors like IL-6 and TNF-α.447 Similar results were seen in insulin-resistant mice.448 Additionally, sea cucumber extracts appear to have some benefits for bone as well: in vitro studies indicate they can inhibit the development of osteoclasts which break down bone, and promote differentiation of bone marrow-derived stem cells into bone-building cells.449,450 These effects may be helpful both in slowing the progression from MGUS/SMM to active myeloma and managing bone lesions associated with active myeloma.

In an open-label clinical trial of TBL-12, 20 patients with asymptomatic multiple myeloma were given a total of 80 mL daily of the sea cucumber extract. The patients were all at high or intermediate risk of disease progression. After follow-up of up to 72 months, the treatment was well-tolerated, and median progression-free survival rates compared favorably to expected outcomes.437 Further clinical trials are necessary to determine whether early intervention, and sea cucumber extracts, may offer overall survival and health benefits for patients with MGUS/SMM or active myeloma.

Omega-3 Fatty Acids (EPA and DHA)

Omega-3 polyunsaturated fatty acids (PUFAs) play a critical role in cell structure and function, including cell signaling and resolution of inflammation, and they have more direct anti-inflammatory effects as well. Notably, they inhibit the inflammatory regulator and transcriptional factor NF-κB. Omega-3 PUFAs have been studied extensively, clinically and preclinically, in the context of cancer complications.451 Intake of omega-3 PUFAs has been shown in human studies to improve survival of cancer patients.451,452 The omega-3 PUFA docosahexaenoic acid (DHA), primarily derived from marine sources such as fish, has been reported in preclinical studies to increase sensitivity of cancer cells to anti-neoplastic agents in drug-resistant cell lines when combined with anti-cancer agents.453

Exposure of human MM cells to eicosapentaenoic acid (EPA) and DHA in vitro inhibited constitutive NF-κB activity and induced cell apoptosis through mitochondrial perturbation and caspase-3 activation. EPA and DHA also increased sensitivity of human MM cells to the proteasome inhibitor bortezomib.454 Stimulation of human U266 MM cells with EPA and DHA in the presence of dexamethasone increased MM cell apoptosis indicating omega‐3 fatty acids increased the sensitivity of MM cells to dexamethasone.454 The increase in sensitivity of MM cells to dexamethasone correlated with the increased expression of tumor suppressor p53 and miR-34a and reduced expression of Bcl-2. Thus, EPA and DHA increase sensitivity of MM cells to dexamethasone through the NF-κB-dependent p53/miR-34a/Bcl-2 axis.455

As of mid-2020, no clinical studies have investigated the effects of omega‐3 PUFAs in MGUS or SMM patients. However, a few clinical studies have been conducted to study the effects of EPA and DHA on hematological malignancies and side effects of treatment.

A prospective, randomized, single-center, open-label clinical trial including 12 acute lymphoblastic leukemia and six acute myeloid leukemia patients investigated the effects of EPA as part of an energy- and protein-dense supplement. At the end of the trial, alleviation of cancer-induced weight loss and improvement of the overall conditions of pediatric patients was observed. Decrease in levels of acute phase proteins were attributed to the beneficial effects of EPA.456

A randomized clinical trial investigated the effects of EPA and DHA supplementation on inflammatory markers and long-term survival of 22 patients with acute and chronic leukemia and lymphomas (Hodgkin and non-Hodgkin) receiving chemotherapy. The study found that consumption of 2 grams/day fish oil (containing EPA and DHA) for nine weeks resulted in greater reduction in C-reactive protein (CRP) and CRP/albumin ratio in patients receiving the supplement and chemotherapy compared with those receiving chemotherapy only. The overall long-term survival time of patients receiving fish oil was also greater (465 days after the start of chemotherapy) relative to controls. These findings indicate an improved nutritional‐inflammatory risk and suggest potential for long‐term survival in patients with hematological malignancies receiving chemotherapy who also supplement with fish oil.457

A multi-institutional phase II cooperative group study to examine the potential of fish oil supplements administered at high doses to slow weight loss and improve quality of life in patients with malignancy‐related cachexia was conducted in 43 patients diagnosed with advanced hematological malignancies including leukemia, lymphoma, and MM. Patients were supplemented with high‐dose omega‐3 PUFA capsules containing 4.7 grams EPA and 2.8 grams DHA per day for about 1.2 months. Study data show 24 patients had weight stabilization, six gained > 5% of their body weight, and six lost ≥ 5% of their body weight. Quality of life scores were superior for patients who gained weight. Thus, supplementation with high-dose omega‐3 PUFAs supported weight stabilization or weight gain in a subset of patients, which resulted in improved quality of life in this group.458

Genistein

Genistein, an isoflavone found predominantly in soybeans, has been shown to exert a wide range of bioactivities, including anti-cancer activities.459 In MM cells, genistein (40 mM) increased the expression of miR-29b and suppressed NF-κB. The suppression of NF-κB resulted in reduced cellular proliferation, increased caspase-3 activity, and apoptosis.460

Bone marrow stromal cells (BMSCs) are critical for promoting myeloma cell survival and proliferation. Genistein was shown to downregulate NF-κB in BMSCs, which decreased the expansion of MM cells when these two cell types were co-cultured.461 Furthermore, genistein inhibited cellular proteasome activity,462 which could be beneficial for MM patients as proteasome inhibitors are a standard drug therapy for MM. In clinical settings, genistein was shown to strengthen NK cell activity.463

An epidemiological study of 220 patients with MM compared with 220 matched controls suggested dietary soy intake was associated with a reduced risk of developing MM.464 Genistein (60 mg daily for seven days every two weeks) was tested in 13 patients with metastatic colorectal cancer undergoing chemotherapy and found to be safe and tolerable, with potential positive impacts on efficacy that include increased NK cell activity.463 In 16 patients with pancreatic cancer, a novel crystalline form of genistein administered at doses up to 1600 mg/day in combination with standard treatment resulted in maximum serum genistein concentrations of 1 µM.465 As genistein appears to follow a linear dose-response curve,466 these results suggest that clinically relevant doses of genistein up to 800 mg/day should not lead to serum levels exceeding 0.5 mM. At this dose genistein may help promote NK cell-mediated antitumor response in cancer patients. However, MM patients may wish to avoid genistein doses greater than 800 mg/day as genistein concentrations above 0.5 mM have been shown to adversely affect NK cell function in vitro.467

Green Tea Extract (EGCG)

Green tea’s (Camellia sinensis) main chemical components are tea polyphenols, which have been shown to exhibit various therapeutic effects against several human pathologies, including cancer. Tea polyphenols in green tea are comprised of catechins including epicatechin (EC), epicatechin-3-gallate (ECG), epigallocatechin (EGC), and epigallocatechin-3-gallate (EGCG). Among these components, EGCG is the most abundant tea polyphenol.468,469

Effects of EGCG on cell death mechanisms via the induction of apoptosis, necrosis, and autophagy are well-documented in in vitro studies and several preclinical models.470-472 The anti-cancer effects of EGCG have been demonstrated in MM cells as well.473

A phase I trial found that a concentrated green tea preparation containing EGCG, known as polyphenon E, was well tolerated at doses up to 2,000 mg twice daily in CLL patients and led to some positive clinical effects.474,475

Note: Green tea may interact with bortezomib. See the sidebar titled “Potential Bortezomib Interactions with Natural Substances” for more information.

Icaritin

Icaritin is a flavonoid (prenylflavonoid) extracted from a traditional Chinese herb, Epimedium grandiflorum, also known as horny goat weed. Its cytotoxic effects are thought to induce tumor cell killing and inhibit the spread of several types of cancer cells through various mechanisms, including inhibiting proliferation, blocking the cell cycle, and inducing apoptosis.338 Studies into icaritin for control of MM have largely focused on its ability to inhibit signal transduction pathways, leading to improved apoptosis of MM cells.340,476

Icaritin has been shown to reverse drug resistance exhibited by some myeloma cells.339 There is also potential for icaritin to regenerate bone in those with MM by increasing bone formation through increasing bone morphogenic proteins (BMPs) among other osteogenic mechanisms.477 Clinical trials are needed to further explore the potential of icaritin in patients with MM.

Parthenolide

Parthenolide, a sesquiterpene lactone naturally occurring in the feverfew (Tanacetum parthenium) plant, is known to have multiple anti-cancer effects.478 In an experiment using a 3D tissue culture that mimics the microenvironment of bone marrow, myeloma stem cells were cultured. Both parthenolide and andrographolide (from the plant Andrographis paniculata) inhibited the growth of the myeloma stem cells.479 However, once cells found in bone marrow stroma were added, only parthenolide maintained its cytotoxic effect.

In addition to the effect of parthenolide on myeloma stem cells, it also suppresses activation of NF-κB by binding TRAF6 in myeloma cells, effectively suppressing proliferation and inducing apoptosis.480

Parthenolide induced apoptosis in four separate myeloma cell lines while not harming normal lymphocytes.481 In this cell experiment, apoptosis was completely stopped by the addition of N-acetyl cysteine, indicating the effect is mediated by reactive oxygen species (ROS). This was confirmation of an earlier cell study that showed parthenolide induced apoptosis through ROS generation.482

Proteolytic Enzymes

Enzymes have been used extensively alongside conventional cancer treatments worldwide.483 The most commonly used preparations for add-on therapy to cancer regimens are proteolytic or pancreatic enzymes. Research on the benefits of enzymes in cancer is fairly scant, but the predominant benefit reported in the available literature has been in the reduction in side effects from chemotherapy and radiation.484

One small study suggested better outcomes with proteolytic enzymes for MM patients undergoing chemotherapy. In this trial, oral enzyme tablets consisting of 100 mg papain, 40 mg trypsin, and 40 mg chymotrypsin were studied in 265 myeloma patients (stages I‒III). Patients received either standard chemotherapy alone or standard chemotherapy with two oral enzyme tablets three times daily.485 Dosing began on the first day of chemotherapy and was reduced to one tablet three times daily after 12 months. Across all stages of disease, the number of patients that achieved either remission or stable disease was significantly higher in the enzyme plus chemotherapy group versus chemotherapy alone. Participants with stage III myeloma (n=54) that received enzyme supplements plus chemotherapy survived longer compared with chemotherapy alone (83 months vs. 47 months; p= 0.0014). While this small trial was intriguing, placebo-controlled studies are needed before any recommendation regarding enzymes can be made.

Intravenous Vitamin C (Ascorbate)

Vitamin C insufficiency may be present in those with MM. One study of 50 patients with MM (ISS stage II) found higher oxidative stress (per malondialdehyde levels) as well as lower status of both enzymatic (glutathione peroxidase, superoxide dismutase, and catalase) and nonenzymatic (vitamins C and E) antioxidants in MM patients versus control subjects.486

Intravenous vitamin C, sometimes called pharmacologic ascorbate, has been studied extensively in cancer, including hematologic cancers, and in preclinical settings against multiple myeloma.487 Pharmacologic ascorbate’s anti-cancer activity has a different mechanism of action than oral antioxidant vitamin C, which has not been shown to be effective at killing cancer cells or tumors.488,489 Multiple clinical trials of pharmacologic ascorbate are planned, underway, or completed. Most such trials have combined intravenous vitamin C with chemotherapy, and several of them have reported encouraging results.487,490

Safety note: Some in vivo data suggests vitamin C may interfere with the efficacy of boronic-acid-based proteasome inhibitors such as bortezomib.110 Therefore, MM patients on bortezomib should talk with their oncologist before taking vitamin C in supplemental form (see sidebar titled “Potential Bortezomib Interactions with Natural Agents”).

Vitamin D

Vitamin D is a steroidal compound that is made in the skin as a result of direct exposure to sunshine. Vitamin D receptors are found in cells throughout the body. Given this widespread distribution, it is not surprising that vitamin D is involved in physiological homeostasis of nearly every system in the body.491 Some foods (eg, fatty fish, mushrooms) contain small amounts of vitamin D, but repletion of circulating levels usually requires oral supplementation or intramuscular injection.

Clinical consideration of vitamin D use in those with MGUS/MM should be approached cautiously given its effect on calcium homeostasis. One of the classic presenting symptoms of MM is hypercalcemia, and the normal physiological role of vitamin D in preserving calcium by optimizing intestinal absorption and reducing renal clearance of calcium must be taken into consideration. In contrast, vitamin D deficiency portends poorer outcomes in MM patients. Given that vitamin D deficiency and hypercalcemia are each a risk for the patient, the prudent approach may be to monitor both circulating vitamin D and calcium on a routine basis in those with MGUS/MM.

Recent studies suggest low vitamin D status is associated with a higher risk and poorer prognosis of a variety of cancers, including MM.492,493

Vitamin D deficiency has been associated with MM, even in tropical climates with ample sunshine.494 In a single-institution study that assessed vitamin D levels of incoming patients with bone metastasis or multiple myeloma, vitamin D deficiency was “alarmingly common” according to the authors.495 In the MM cohort, average vitamin D level was just under 15 ng/mL, a value low enough that bone loss is expected even in healthy individuals.

There is some controversy regarding optimal levels of circulating vitamin D (as 25-hydroxycholicalciferol). However, there is consensus that levels below 20 ng/mL represent a gross deficiency that results in bone loss.496,497

In a study that assessed 83 myeloma patients and vitamin D levels, deficiency (<10 ng/mL) was associated with higher levels of plasma cells in the bone marrow.498 Furthermore, supplementation with vitamin D led to significant increases in hemoglobin, leukocyte, and RBC levels, and improvement in platelets.

Vitamin D deficiency (serum levels <50 nmol/L or 20 ng/mL) is associated with more inflammation (as measured by CRP levels), lower serum albumin, and higher creatinine when compared with MM subjects without vitamin D deficiency.499 In a small single clinic study of 31 MM patients (12 female, 19 male), the median level of circulating vitamin D was just below 12 ng/mL with 93.5% of patients deficient at the time of diagnosis.500 The clinic also reported that vitamin D levels were lower after each chemotherapy session.

While there is no clinical data to determine whether adequate vitamin D status is preventative of the progression from MGUS to MM, it is plausible to expect that the effects of vitamin D on preventing bone resorption, regulating immune function, and acting as a pro-differentiating agent may result in reduction of transformation from benign MGUS to MM.501

A study of 158 patients scheduled to receive their first autologous stem cell transplant found pre-transplant vitamin D levels below 23 ng/mL were associated with inferior survival time after autologous stem cell transplant (25 months vs. 32 months; p=0.03).502 They also found lower vitamin D levels were significantly associated with younger age at diagnosis of MM.

In one report of 108 MM patients followed for one year, lower vitamin D levels were associated with myeloma activity (paraprotein concentration), higher markers of bone turnover (urine deoxypyridinoline), and worse bone mineral density (dual-energy X-ray absorptiometry, or DEXA).503

Vitamin D repletion may benefit those taking bortezomib. Bortezomib has been shown to improve bone health through a mechanism that involves vitamin D and its receptor (VDR).504 One mechanism of bone loss in myeloma is disruption of vitamin D dependent differentiation of osteoblasts, a process that is inhibited by bortezomib. While bortezomib alone partially overcomes this, the addition of vitamin D had an additive effect, allowing for improved maturation of osteoblasts co-cultured with myeloma cells. As of this writing, there are no clinical trials published on the bone effects of repletion of vitamin D in those taking bortezomib.

Vitamin D may complement some MM drugs known to induce peripheral neuropathy. Vitamin D deficiency is associated with neuropathy induced by drugs used to treat MM such as bortezomib and thalidomide.505 Given the possible benefits to those taking such drugs, vitamin D should be closely monitored and supplemented as needed.

It appears that correcting vitamin D deficiency could improve outcomes in those with MGUS/MM. This simple intervention is important, since vitamin D levels are frequently low and infrequently assessed in MM patients.506

Vitamin K

Vitamin K is required to maintain homeostasis of osteoblasts and osteoclasts in normal bone turnover. While human data on the outcome of supplemental vitamin K in MGUS/MM is lacking, many of the mechanisms of bone degradation in MM may be targets for the action of vitamin K in maintaining bone health.

There are many other types of proliferative disorders of the bone marrow (eg, dyscrasias and leukemias), but only the clonal populations of plasma cells associated with MM are marked by the destruction of bone in the nearby microenvironment. This characteristic trait has been proposed as a crux of the plasma cell proliferation process in plasmacytomas/MM. While a singular definitive mechanism shared by all MM cases has yet to be found, it is clear that there is an imbalance in the expression of bone regulating proteins that stimulate or inhibit osteoclastic and osteoblastic actions (Table 11).

Table 11: Overexpressed Proteins Correlating with Severity of Bone Degradation in Those with MM*

Produced by

Inhibit nearby Osteoblasts

Stimulate nearby Osteoclasts

MM cells

DKK1, SRP-3, HGF, MIP-1alpha

MIP 1-alpha, IL-32, BDNF

Stromal cells

Activin A

RANKL, GDF15, BDNF

*Compiled from Why Do Myeloma Patients Have Bone Disease: A Historical Perspective by Borset et al.507

Vitamin K also appears to affect bone turnover favorably through dual actions on osteoclasts and osteoblasts derived from its inhibition of NF-κB activation.508

Preliminary evidence from an in vitro study suggests vitamin K2 may exert some anti-myeloma effects in addition to its benefits for bone health. When myeloma cells were incubated with various concentrations of vitamin K2, their growth was inhibited and apoptosis increased.170

Plumbagin, an analogue of vitamin K derived from Plumbago zeylanica, an Ayurvedic medical plant, has been shown to inhibit RANKL and suppress osteoclastogenesis in a rodent model of multiple myeloma.509 Further studies should be done in humans before any conclusions are drawn on its use.

2020

  • Nov: Comprehensive update & review

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. Albagoush SA, Azevedo AM. Cancer, Multiple Myeloma. StatPearls. 2019.
  2. Kyle RA, Rajkumar SV. Multiple myeloma. Blood. Mar 15 2008;111(6):2962-72. doi:10.1182/blood-2007-10-078022
  3. Rajkumar SV, Kyle RA, Connor RF. Multiple myeloma: Pathobiology. UpToDate. Updated 03/05/2020. Accessed 08/21/2020, https://www.uptodate.com/contents/multiple-myeloma-pathobiology?search=myeloma &source=search_result&selectedTitle=3~150&usage_type=default&display_rank=3
  4. Kazandjian D. Multiple myeloma epidemiology and survival: A unique malignancy. Seminars in oncology. Dec 2016;43(6):676-681. doi:10.1053/j.seminoncol.2016.11.004
  5. Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA: a cancer journal for clinicians. Nov 12 2016;66(6):443-459. doi:10.3322/caac.21357
  6. Cid Ruzafa J, Merinopoulou E, Baggaley RF, et al. Patient population with multiple myeloma and transitions across different lines of therapy in the USA: an epidemiologic model. Pharmacoepidemiology and drug safety. Aug 2016;25(8):871-9. doi:10.1002/pds.3927
  7. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group. J Clin Oncol. Sep 10 2015;33(26):2863-9. doi:10.1200/JCO.2015.61.2267
  8. Kyle RA, Larson DR, Therneau TM, et al. Long-Term Follow-up of Monoclonal Gammopathy of Undetermined Significance. New England Journal of Medicine. 2018;378(3):241-249. doi:10.1056/NEJMoa1709974
  9. Go RS, Rajkumar SV. How I manage monoclonal gammopathy of undetermined significance. Blood. 2018;131(2):163-173. doi:10.1182/blood-2017-09-807560
  10. Fonseca R, Abouzaid S, Bonafede M, et al. Trends in overall survival and costs of multiple myeloma, 2000-2014. Leukemia. Sep 2017;31(9):1915-1921. doi:10.1038/leu.2016.380
  11. Greenberg P. The myelodysplastic syndromes. In: Hoffman R BE, Shattil S, ed. Hematology: Basic Principles and Practice. 3rd ed. Churchill Livingstone; 2000:1106-1129.
  12. Justiz Vaillant AA, Ramphul K. Immunoglobulin. StatPearls. 2019.
  13. Janeway CA, Travers P, Walport M, Shlomchik MJ. Immunobiology: The Immune System in Health and Disease, 5th ed. New York: Garland Science; 2001. The production of armed effector T cells. Available at: http://www.ncbi.nlm.nih.gov/books/NBK27118/ .
  14. ASCO. American Society of Clinical Oncology. Multiple Myeloma: Introduction. Updated 5/2020. Accessed 9/24/2020, https://www.cancer.net/cancer-types/multiple-myeloma/introduction
  15. Laing K. Immune responses to viruses. British Society for Immunology. Accessed 9/24/2020, https://www.immunology.org/public-information/bitesized-immunology/pathogens-and-disease/immune-responses-viruses
  16. Blimark C, Holmberg E, Mellqvist UH, et al. Multiple myeloma and infections: a population-based study on 9253 multiple myeloma patients. Haematologica. Jan 2015;100(1):107-13. doi:10.3324/haematol.2014.107714
  17. Kyle RA, Gertz MA, Witzig TE, et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc. Jan 2003;78(1):21-33. doi:10.4065/78.1.21
  18. Rafae A, Malik MN, Abu Zar M, Durer S, Durer C. An Overview of Light Chain Multiple Myeloma: Clinical Characteristics and Rarities, Management Strategies, and Disease Monitoring. Cureus. Aug 15 2018;10(8):e3148. doi:10.7759/cureus.3148
  19. El Arfani C, De Veirman K, Maes K, De Bruyne E, Menu E. Metabolic Features of Multiple Myeloma. International journal of molecular sciences. Apr 14 2018;19(4)doi:10.3390/ijms19041200
  20. Chaudhry R, Varacallo M. Biochemistry, Glycolysis. StatPearls. 2019.
  21. Newsholme P, Procopio J, Lima MM, Pithon-Curi TC, Curi R. Glutamine and glutamate--their central role in cell metabolism and function. Cell Biochem Funct. Mar 2003;21(1):1-9. doi:10.1002/cbf.1003
  22. Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. Oct 2016;16(10):619-34. doi:10.1038/nrc.2016.71
  23. Hirschey MD, DeBerardinis RJ, Diehl AME, et al. Dysregulated metabolism contributes to oncogenesis. Seminars in cancer biology. Dec 2015;35 Suppl:S129-S150. doi:10.1016/j.semcancer.2015.10.002
  24. Akins NS, Nielson TC, Le HV. Inhibition of Glycolysis and Glutaminolysis: An Emerging Drug Discovery Approach to Combat Cancer. Current topics in medicinal chemistry. 2018;18(6):494-504. doi:10.2174/1568026618666180523111351
  25. Warburg O. On the origin of cancer cells. Science. Feb 24 1956;123(3191):309-14. doi:10.1126/science.123.3191.309
  26. Erickson JW, Cerione RA. Glutaminase: a hot spot for regulation of cancer cell metabolism? Oncotarget. Dec 2010;1(8):734-40. doi:10.18632/oncotarget.208
  27. Nakano A, Miki H, Nakamura S, et al. Up-regulation of hexokinaseII in myeloma cells: targeting myeloma cells with 3-bromopyruvate. J Bioenerg Biomembr. Feb 2012;44(1):31-8. doi:10.1007/s10863-012-9412-9
  28. Tamada M, Suematsu M, Saya H. Pyruvate kinase M2: multiple faces for conferring benefits on cancer cells. Clin Cancer Res. Oct 15 2012;18(20):5554-61. doi:10.1158/1078-0432.CCR-12-0859
  29. Effenberger M, Bommert KS, Kunz V, et al. Glutaminase inhibition in multiple myeloma induces apoptosis via MYC degradation. Oncotarget . Oct 17 2017;8(49):85858-85867. doi:10.18632/oncotarget.20691
  30. Gabay M, Li Y, Felsher DW. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harbor perspectives in medicine. Jun 2 2014;4(6)doi:10.1101/cshperspect.a014241
  31. Kouidhi S, Ben Ayed F, Benammar Elgaaied A. Targeting Tumor Metabolism: A New Challenge to Improve Immunotherapy. Front Immunol. 2018;9:353. doi:10.3389/fimmu.2018.00353
  32. Hussain A, Almenfi HF, Almehdewi AM, Hamza MS, Bhat MS, Vijayashankar NP. Laboratory Features of Newly Diagnosed Multiple Myeloma Patients. Cureus. May 22 2019;11(5):e4716. doi:10.7759/cureus.4716
  33. Seccareccia D. Cancer-related hypercalcemia. Canadian family physician Medecin de famille canadien. Mar 2010;56(3):244-6, e90-2.
  34. Doughney KB, Williams DM, Penn RL. Multiple myeloma: infectious complications. Southern medical journal. Jul 1988;81(7):855-8. doi:10.1097/00007611-198807000-00012
  35. Nucci M, Anaissie E. Infections in patients with multiple myeloma in the era of high-dose therapy and novel agents. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America . Oct 15 2009;49(8):1211-25. doi:10.1086/605664
  36. Winearls CG. Acute myeloma kidney. Kidney international. Oct 1995;48(4):1347-61. doi:10.1038/ki.1995.421
  37. Korbet SM, Schwartz MM. Multiple myeloma. Journal of the American Society of Nephrology : JASN. Sep 2006;17(9):2533-45. doi:10.1681/ASN.2006020139
  38. Laubach JP, Kyle RA, Rajkumar V, Connor RF. Multiple myeloma: Clinical features, laboratory manifestations, and diagnosis. UpToDate. Updated 08/13/2020. Accessed 08/21/2020, https://www.uptodate.com/contents/multiple-myeloma-clinical-features-laboratory-manifestations-and-diagnosis?search=myeloma &source=search_result&selectedTitle=1~150&usage_type=default&display_rank=1#H8
  39. Smith A, Howell D, Patmore R, Jack A, Roman E. Incidence of haematological malignancy by sub-type: a report from the Haematological Malignancy Research Network. British journal of cancer. Nov 22 2011;105(11):1684-92. doi:10.1038/bjc.2011.450
  40. Therneau TM, Kyle RA, Melton LJ, 3rd, et al. Incidence of monoclonal gammopathy of undetermined significance and estimation of duration before first clinical recognition. Mayo Clin Proc. Nov 2012;87(11):1071-9. doi:10.1016/j.mayocp.2012.06.014
  41. Kyle RA, Therneau TM, Rajkumar SV, et al. Prevalence of monoclonal gammopathy of undetermined significance. The New England journal of medicine. Mar 30 2006;354(13):1362-9. doi:10.1056/NEJMoa054494
  42. Konstantinopoulos PA, Pantanowitz L, Dezube BJ. Higher prevalence of monoclonal gammopathy of undetermined significance in African Americans than whites--the unknown role of underlying HIV infection. Journal of the National Medical Association. Nov 2006;98(11):1860-1.
  43. Landgren O, Katzmann JA, Hsing AW, et al. Prevalence of monoclonal gammopathy of undetermined significance among men in Ghana. Mayo Clin Proc. Dec 2007;82(12):1468-73. doi:10.1016/S0025-6196(11)61089-6
  44. Waxman AJ, Mink PJ, Devesa SS, et al. Racial disparities in incidence and outcome in multiple myeloma: a population-based study. Blood. Dec 16 2010;116(25):5501-6. doi:10.1182/blood-2010-07-298760
  45. Huang SY, Yao M, Tang JL, et al. Epidemiology of multiple myeloma in Taiwan: increasing incidence for the past 25 years and higher prevalence of extramedullary myeloma in patients younger than 55 years. Cancer. Aug 15 2007;110(4):896-905. doi:10.1002/cncr.22850
  46. Hoang PH, Dobbins SE, Cornish AJ, et al. Whole-genome sequencing of multiple myeloma reveals oncogenic pathways are targeted somatically through multiple mechanisms. Leukemia. 2018/11/01 2018;32(11):2459-2470. doi:10.1038/s41375-018-0103-3
  47. Landau HJ, Yellapantula V, Diamond BT, et al. Accelerated single cell seeding in relapsed multiple myeloma. Nature communications. 2020/07/17 2020;11(1):3617. doi:10.1038/s41467-020-17459-z
  48. Lynch HT, Sanger WG, Pirruccello S, Quinn-Laquer B, Weisenburger DD. Familial multiple myeloma: a family study and review of the literature. Journal of the National Cancer Institute. Oct 3 2001;93(19):1479-83. doi:10.1093/jnci/93.19.1479
  49. Ogmundsdottir HM, Haraldsdottirm V, Johannesson GM, et al. Familiality of benign and malignant paraproteinemias. A population-based cancer-registry study of multiple myeloma families. Haematologica. Jan 2005;90(1):66-71.
  50. Landgren O, Kristinsson SY, Goldin LR, et al. Risk of plasma cell and lymphoproliferative disorders among 14621 first-degree relatives of 4458 patients with monoclonal gammopathy of undetermined significance in Sweden. Blood. Jul 23 2009;114(4):791-5. doi:10.1182/blood-2008-12-191676
  51. Riedel DA, Pottern LM. The epidemiology of multiple myeloma. Hematol Oncol Clin North Am. Apr 1992;6(2):225-47.
  52. Lewis EB. Leukemia, Multiple Myeloma, and Aplastic Anemia in American Radiologists. Science. Dec 13 1963;142(3598):1492-4. doi:10.1126/science.142.3598.1492
  53. Gold LS, Stewart PA, Milliken K, et al. The relationship between multiple myeloma and occupational exposure to six chlorinated solvents. Occup Environ Med. Jun 2011;68(6):391-9. doi:10.1136/oem.2009.054809
  54. Eriksson M, Karlsson M. Occupational and other environmental factors and multiple myeloma: a population based case-control study. Br J Ind Med. Feb 1992;49(2):95-103. doi:10.1136/oem.49.2.95
  55. Svec MA, Ward MH, Dosemeci M, Checkoway H, De Roos AJ. Risk of lymphatic or haematopoietic cancer mortality with occupational exposure to animals or the public. Occup Environ Med. Oct 2005;62(10):726-35. doi:10.1136/oem.2005.021550
  56. Sonoda T, Ishida T, Mori M, et al. A case-control study of multiple myeloma in Japan: association with occupational factors. Asian Pacific journal of cancer prevention : APJCP. Jan-Mar 2005;6(1):33-6.
  57. Blair CK, Cerhan JR, Folsom AR, Ross JA. Anthropometric characteristics and risk of multiple myeloma. Epidemiology (Cambridge, Mass). Sep 2005;16(5):691-4. doi:10.1097/01.ede.0000172135.61188.2d
  58. Pan SY, Johnson KC, Ugnat AM, Wen SW, Mao Y, Canadian Cancer Registries Epidemiology Research G. Association of obesity and cancer risk in Canada. American journal of epidemiology. Feb 1 2004;159(3):259-68. doi:10.1093/aje/kwh041
  59. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. The New England journal of medicine. Apr 24 2003;348(17):1625-38. doi:10.1056/NEJMoa021423
  60. Birmann BM, Giovannucci E, Rosner B, Anderson KC, Colditz GA. Body mass index, physical activity, and risk of multiple myeloma. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jul 2007;16(7):1474-8. doi:10.1158/1055-9965.EPI-07-0143
  61. Stagnaro E, Ramazzotti V, Crosignani P, et al. Smoking and hematolymphopoietic malignancies. Cancer Causes Control. May 2001;12(4):325-34. doi:10.1023/a:1011216102871
  62. Nieters A, Deeg E, Becker N. Tobacco and alcohol consumption and risk of lymphoma: results of a population-based case-control study in Germany. International journal of cancer Journal international du cancer. Jan 15 2006;118(2):422-30. doi:10.1002/ijc.21306
  63. Fulop T, Montgomery RR. Editorial overview: Immune senescence: known knowns and unknown unknowns. Curr Opin Immunol. Aug 2014;29:vii-ix. doi:10.1016/j.coi.2014.06.005
  64. Cooke RE, Koldej R, Ritchie D. Immunotherapeutics in Multiple Myeloma: How Can Translational Mouse Models Help? Journal of oncology. 2019;2019:2186494. doi:10.1155/2019/2186494
  65. Kourelis TV, Villasboas JC, Jessen E, et al. Mass cytometry dissects T cell heterogeneity in the immune tumor microenvironment of common dysproteinemias at diagnosis and after first line therapies. Blood cancer journal. Aug 28 2019;9(9):72. doi:10.1038/s41408-019-0234-4
  66. Joshua D, Suen H, Brown R, et al. The T Cell in Myeloma. Clin Lymphoma Myeloma Leuk. Oct 2016;16(10):537-542. doi:10.1016/j.clml.2016.08.003
  67. Weksler ME. Immune senescence. Ann Neurol. 1994;35 Suppl:S35-7. doi:10.1002/ana.410350711
  68. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. The Lancet Oncology. Nov 2014;15(12):e538-48. doi:10.1016/S1470-2045(14)70442-5
  69. Gundesen MT, Lund T, Moeller HEH, Abildgaard N. Plasma Cell Leukemia: Definition, Presentation, and Treatment. Curr Oncol Rep. Jan 28 2019;21(1):8. doi:10.1007/s11912-019-0754-x
  70. National Comprehensive Cancer Network (NCCN). NCCN Guidelines - Multiple Myeloma. 12/27/2019, Updated 10/9/2019 (version 2.2020). Accessed 12/27/2019, https://pubmed.ncbi.nlm.nih.gov/31590151/
  71. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. The Lancet Oncology. 2014;15(12):e538-e548. doi:10.1016/S1470-2045(14)70442-5
  72. Smith A, Wisloff F, Samson D, Forum UKM, Nordic Myeloma Study G, British Committee for Standards in H. Guidelines on the diagnosis and management of multiple myeloma 2005. Br J Haematol. Feb 2006;132(4):410-51. doi:10.1111/j.1365-2141.2005.05867.x
  73. Dimopoulos M, Kyle R, Fermand JP, et al. Consensus recommendations for standard investigative workup: report of the International Myeloma Workshop Consensus Panel 3. Blood. May 5 2011;117(18):4701-5. doi:10.1182/blood-2010-10-299529
  74. George-Gay B, Parker K. Understanding the complete blood count with differential. J Perianesth Nurs. Apr 2003;18(2):96-114; quiz 115-7. doi:10.1053/jpan.2003.50013
  75. Oyajobi BO. Multiple myeloma/hypercalcemia. Arthritis research & therapy. 2007;9 Suppl 1:S4. doi:10.1186/ar2168
  76. Loh RK, Vale S, McLean-Tooke A. Quantitative serum immunoglobulin tests. Australian family physician. Apr 2013;42(4):195-8.
  77. Protein Testing in Patients with Multiple Myeloma: A Review of Clinical Effectiveness and Guidelines . 2015. CADTH Rapid Response Reports.
  78. Rajkumar SV, Fonseca R, Dispenzieri A, et al. Methods for estimation of bone marrow plasma cell involvement in myeloma: predictive value for response and survival in patients undergoing autologous stem cell transplantation. Am J Hematol. Dec 2001;68(4):269-75. doi:10.1002/ajh.10003
  79. Stifter S, Babarovic E, Valkovic T, et al. Combined evaluation of bone marrow aspirate and biopsy is superior in the prognosis of multiple myeloma. Diagnostic pathology. May 18 2010;5:30. doi:10.1186/1746-1596-5-30
  80. Derlin T, Bannas P. Imaging of multiple myeloma: Current concepts. World J Orthop. Jul 18 2014;5(3):272-82. doi:10.5312/wjo.v5.i3.272
  81. Bannas P, Kroger N, Adam G, Derlin T. [Modern imaging techniques in patients with multiple myeloma]. Rofo. Jan 2013;185(1):26-33. Moderne Bildgebungsverfahren beim Multiplen Myelom. doi:10.1055/s-0032-1325405
  82. Hanrahan CJ, Christensen CR, Crim JR. Current concepts in the evaluation of multiple myeloma with MR imaging and FDG PET/CT. Radiographics. Jan 2010;30(1):127-42. doi:10.1148/rg.301095066
  83. Baur-Melnyk A, Buhmann S, Durr HR, Reiser M. Role of MRI for the diagnosis and prognosis of multiple myeloma. European journal of radiology. Jul 2005;55(1):56-63. doi:10.1016/j.ejrad.2005.01.017
  84. Moulopoulos LA, Dimopoulos MA, Alexanian R, Leeds NE, Libshitz HI. Multiple myeloma: MR patterns of response to treatment. Radiology. Nov 1994;193(2):441-6. doi:10.1148/radiology.193.2.7972760
  85. Moulopoulos LA, Dimopoulos MA, Smith TL, et al. Prognostic significance of magnetic resonance imaging in patients with asymptomatic multiple myeloma. J Clin Oncol. Jan 1995;13(1):251-6. doi:10.1200/JCO.1995.13.1.251
  86. Greipp PR, San Miguel J, Durie BG, et al. International staging system for multiple myeloma. J Clin Oncol. May 20 2005;23(15):3412-20. doi:10.1200/JCO.2005.04.242
  87. Durie BG, Salmon SE. A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer. Sep 1975;36(3):842-54. doi:10.1002/1097-0142(197509)36:3<842::aid-cncr2820360303>3.0.co;2-u
  88. Rajkumar SV, Gupta V, Fonseca R, et al. Impact of primary molecular cytogenetic abnormalities and risk of progression in smoldering multiple myeloma. Leukemia. Aug 2013;27(8):1738-44. doi:10.1038/leu.2013.86
  89. Hanbali A, Hassanein M, Rasheed W, Aljurf M, Alsharif F. The Evolution of Prognostic Factors in Multiple Myeloma. Advances in hematology. 2017;2017:4812637. doi:10.1155/2017/4812637
  90. Mikhael JR, Dingli D, Roy V, et al. Management of newly diagnosed symptomatic multiple myeloma: updated Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) consensus guidelines 2013. Mayo Clin Proc. Apr 2013;88(4):360-76. doi:10.1016/j.mayocp.2013.01.019
  91. Miguel JS, Mateos M-V, Gonzalez V, et al. Updated risk stratification model for smoldering multiple myeloma (SMM) incorporating the revised IMWG diagnostic criteria. Journal of Clinical Oncology. 2019;37(15_suppl):8000-8000. doi:10.1200/JCO.2019.37.15_suppl.8000
  92. Bustoros M, Sklavenitis-Pistofidis R, Park J, et al. Genomic Profiling of Smoldering Multiple Myeloma Identifies Patients at a High Risk of Disease Progression. J Clin Oncol. Jul 20 2020;38(21):2380-2389. doi:10.1200/jco.20.00437
  93. Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple myeloma for clinical practice. Blood cancer journal. Oct 30 2015;5:e365. doi:10.1038/bcj.2015.92
  94. Okazuka K, Ishida T. Proteasome inhibitors for multiple myeloma. Japanese journal of clinical oncology. Sep 1 2018;48(9):785-793. doi:10.1093/jjco/hyy108
  95. Field-Smith A, Morgan GJ, Davies FE. Bortezomib (Velcadetrade mark) in the Treatment of Multiple Myeloma. Therapeutics and clinical risk management. Sep 2006;2(3):271-9. doi:10.2147/tcrm.2006.2.3.271
  96. Grosicki S, Barchnicka A, Jurczyszyn A, Grosicka A. Bortezomib for the treatment of multiple myeloma. Expert Rev Hematol. Apr 2014;7(2):173-85. doi:10.1586/17474086.2014.899144
  97. Kouroukis TC, Baldassarre FG, Haynes AE, Imrie K, Reece DE, Cheung MC. Bortezomib in multiple myeloma: systematic review and clinical considerations. Current oncology (Toronto, Ont). Aug 2014;21(4):e573-603. doi:10.3747/co.21.1798
  98. Reece D, Imrie K, Stevens A, Smith CA, Hematology Disease Site Groupof Cancer Care Ontario's Program in Evidence-based C. Bortezomib in multiple myeloma and lymphoma: a systematic review and clinical practice guideline. Current oncology (Toronto, Ont). Oct 2006;13(5):160-72.
  99. Richardson PG, Weller E, Lonial S, et al. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. Aug 5 2010;116(5):679-86. doi:10.1182/blood-2010-02-268862
  100. Rosinol L, Oriol A, Rios R, et al. Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma. Blood. Oct 17 2019;134(16):1337-1345. doi:10.1182/blood.2019000241
  101. Roussel M, Lauwers-Cances V, Robillard N, et al. Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: a phase II study by the Intergroupe Francophone du Myelome. J Clin Oncol. Sep 1 2014;32(25):2712-7. doi:10.1200/JCO.2013.54.8164
  102. Moreau P, Pylypenko H, Grosicki S, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. The Lancet Oncology. May 2011;12(5):431-40. doi:10.1016/S1470-2045(11)70081-X
  103. Maschio M, Zarabla A, Maialetti A, et al. The Effect of Docosahexaenoic Acid and alpha-Lipoic Acid as Prevention of Bortezomib-Related Neurotoxicity in Patients With Multiple Myeloma. Integrative cancer therapies. Jan-Dec 2019;18:1534735419888584. doi:10.1177/1534735419888584
  104. Jia L, Liu F-T. Why bortezomib cannot go with 'green'? Cancer Biol Med. 2013;10(4):206-213. doi:10.7497/j.issn.2095-3941.2013.04.004
  105. Steinberg JA, Shen J, Sanchez E, et al. Alpha Lipoic Acid (ALA) Inhibits the Anti-Myeloma Effects of Bortezomib. Blood. 2009;114(22):3832-3832. doi:10.1182/blood.V114.22.3832.3832
  106. Golden EB, Lam PY, Kardosh A, et al. Green tea polyphenols block the anticancer effects of bortezomib and other boronic acid-based proteasome inhibitors. Blood. Jun 4 2009;113(23):5927-37. doi:10.1182/blood-2008-07-171389
  107. Le CT, Leenders WPJ, Molenaar RJ, van Noorden CJF. Effects of the Green Tea Polyphenol Epigallocatechin-3-Gallate on Glioma: A Critical Evaluation of the Literature. Nutrition and cancer. 2018;70(3):317-333. doi:10.1080/01635581.2018.1446090
  108. Glynn SJ, Gaffney KJ, Sainz MA, Louie SG, Petasis NA. Molecular characterization of the boron adducts of the proteasome inhibitor bortezomib with epigallocatechin-3-gallate and related polyphenols. Organic & biomolecular chemistry. 2015;13(13):3887-3899. doi:10.1039/c4ob02512a
  109. Modernelli A, Naponelli V, Giovanna Troglio M, et al. EGCG antagonizes Bortezomib cytotoxicity in prostate cancer cells by an autophagic mechanism. Scientific reports. 2015;5:15270-15270. doi:10.1038/srep15270
  110. Perrone G, Hideshima T, Ikeda H, et al. Ascorbic acid inhibits antitumor activity of bortezomib in vivo. Leukemia. Sep 2009;23(9):1679-86. doi:10.1038/leu.2009.83
  111. Llobet D, Eritja N, Encinas M, et al. Antioxidants block proteasome inhibitor function in endometrial carcinoma cells. Anti-cancer drugs. 2008;19(2):115-124. doi:10.1097/CAD.0b013e3282f24031
  112. Zou W, Yue P, Lin N, et al. Vitamin C inactivates the proteasome inhibitor PS-341 in human cancer cells. Clinical cancer research : an official journal of the American Association for Cancer Research . 2006;12(1):273-280. doi:10.1158/1078-0432.CCR-05-0503
  113. Maschio M, Zarabla A, Maialetti A, et al. Prevention of Bortezomib-Related Peripheral Neuropathy With Docosahexaenoic Acid and α-Lipoic Acid in Patients With Multiple Myeloma: Preliminary Data. Integrative cancer therapies. 2018;17(4):1115-1124. doi:10.1177/1534735418803758
  114. Nauman G, Gray JC, Parkinson R, Levine M, Paller CJ. Systematic Review of Intravenous Ascorbate in Cancer Clinical Trials. Antioxidants (Basel, Switzerland). Jul 12 2018;7(7)doi:10.3390/antiox7070089
  115. Held LA, Rizzieri D, Long GD, et al. A Phase I study of arsenic trioxide (Trisenox), ascorbic acid, and bortezomib (Velcade) combination therapy in patients with relapsed/refractory multiple myeloma. Cancer investigation. Mar 2013;31(3):172-6. doi:10.3109/07357907.2012.756109
  116. Berenson JR, Boccia R, Siegel D, et al. Efficacy and safety of melphalan, arsenic trioxide and ascorbic acid combination therapy in patients with relapsed or refractory multiple myeloma: a prospective, multicentre, phase II, single-arm study. Br J Haematol. Oct 2006;135(2):174-83. doi:10.1111/j.1365-2141.2006.06280.x
  117. Gorgun G, Calabrese E, Soydan E, et al. Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood. Oct 28 2010;116(17):3227-37. doi:10.1182/blood-2010-04-279893
  118. Holstein SA, McCarthy PL. Immunomodulatory Drugs in Multiple Myeloma: Mechanisms of Action and Clinical Experience. Drugs. Apr 2017;77(5):505-520. doi:10.1007/s40265-017-0689-1
  119. Chen C, Baldassarre F, Kanjeekal S, Herst J, Hicks L, Cheung M. Lenalidomide in multiple myeloma-a practice guideline. Current oncology (Toronto, Ont). Apr 2013;20(2):e136-49. doi:10.3747/co.20.1252
  120. Raza S, Safyan RA, Lentzsch S. Immunomodulatory Drugs (IMiDs) in Multiple Myeloma. Curr Cancer Drug Targets. 2017;17(9):846-857. doi:10.2174/1568009617666170214104426
  121. Reece D, Kouroukis CT, Leblanc R, Sebag M, Song K, Ashkenas J. Practical approaches to the use of lenalidomide in multiple myeloma: a canadian consensus. Advances in hematology. 2012;2012:621958. doi:10.1155/2012/621958
  122. Sharma S, Lichtenstein A. Dexamethasone-induced apoptotic mechanisms in myeloma cells investigated by analysis of mutant glucocorticoid receptors. Blood. Aug 15 2008;112(4):1338-45. doi:10.1182/blood-2007-11-124156
  123. Burwick N, Sharma S. Glucocorticoids in multiple myeloma: past, present, and future. Annals of hematology. Jan 2019;98(1):19-28. doi:10.1007/s00277-018-3465-8
  124. Facon T, Kumar S, Plesner T, et al. Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. The New England journal of medicine. May 30 2019;380(22):2104-2115. doi:10.1056/NEJMoa1817249
  125. Kumar S, Flinn I, Richardson PG, et al. Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma. Blood. May 10 2012;119(19):4375-82. doi:10.1182/blood-2011-11-395749
  126. Ericson-Neilsen W, Kaye AD. Steroids: pharmacology, complications, and practice delivery issues. Ochsner J. Summer 2014;14(2):203-7.
  127. Yasir M, Sonthalia S. Corticosteroid Adverse Effects. StatPearls. 2019.
  128. NCI. National Cancer Institute. Targeted Therapy to Treat Cancer. Updated 3/11/2020. Accessed 9/24/2020, https://www.cancer.gov/about-cancer/treatment/types/targeted-therapies
  129. Costello C. An update on the role of daratumumab in the treatment of multiple myeloma. Therapeutic advances in hematology. Jan 2017;8(1):28-37. doi:10.1177/2040620716677523
  130. Plesner T, Krejcik J. Daratumumab for the Treatment of Multiple Myeloma. Front Immunol. 2018;9:1228. doi:10.3389/fimmu.2018.01228
  131. Richardson PG, Laubach J, Gandolfi S, Facon T, Weisel K, O'Gorman P. Maintenance and continuous therapy for multiple myeloma. Expert review of anticancer therapy. Aug 2018;18(8):751-764. doi:10.1080/14737140.2018.1490181
  132. Kalis JA. Daratumumab: Dawn of a New Paradigm in Multiple Myeloma? Journal of the advanced practitioner in oncology. Jan-Feb 2017;8(1):82-90. doi:10.6004/jadpro.2017.8.1.7
  133. Bierings M, Nachman JB, Zwaan CM. Stem cell transplantation in pediatric leukemia and myelodysplasia: state of the art and current challenges. Curr Stem Cell Res Ther. Jan 2007;2(1):53-63. doi:10.2174/157488807779317035
  134. Eissa H, Gooley TA, Sorror ML, et al. Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia: relapse-free survival is determined by karyotype and comorbidities. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation . Jun 2011;17(6):908-15. doi:10.1016/j.bbmt.2010.09.018
  135. Festuccia M, Deeg HJ, Gooley TA, et al. Minimal Identifiable Disease and the Role of Conditioning Intensity in Hematopoietic Cell Transplantation for Myelodysplastic Syndrome and Acute Myelogenous Leukemia Evolving from Myelodysplastic Syndrome. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation . Jul 2016;22(7):1227-1233. doi:10.1016/j.bbmt.2016.03.029
  136. Shaw PJ, Kan F, Woo Ahn K, et al. Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors. Blood. Nov 11 2010;116(19):4007-15. doi:10.1182/blood-2010-01-261958
  137. Strahm B, Nollke P, Zecca M, et al. Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG-MDS 98 study. Leukemia. Mar 2011;25(3):455-62. doi:10.1038/leu.2010.297
  138. Bjorkstrand BB, Ljungman P, Svensson H, et al. Allogeneic bone marrow transplantation versus autologous stem cell transplantation in multiple myeloma: a retrospective case-matched study from the European Group for Blood and Marrow Transplantation. Blood. Dec 15 1996;88(12):4711-8.
  139. Gonsalves WI, Buadi FK, Ailawadhi S, et al. Utilization of hematopoietic stem cell transplantation for the treatment of multiple myeloma: a Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) consensus statement. Bone marrow transplantation. Mar 2019;54(3):353-367. doi:10.1038/s41409-018-0264-8
  140. Shah N, Callander N, Ganguly S, et al. Hematopoietic Stem Cell Transplantation for Multiple Myeloma: Guidelines from the American Society for Blood and Marrow Transplantation. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation . Jul 2015;21(7):1155-66. doi:10.1016/j.bbmt.2015.03.002
  141. Sobh M, Michallet M, Gahrton G, et al. Allogeneic hematopoietic cell transplantation for multiple myeloma in Europe: trends and outcomes over 25 years. A study by the EBMT Chronic Malignancies Working Party. Leukemia. Oct 2016;30(10):2047-2054. doi:10.1038/leu.2016.101
  142. Rajkumar SV, Negrin RS, Kyle RA, Connor RF. Multiple myeloma: Use of autologous hematopoietic cell transplantation. UpToDate. Updated 08/10/2020. Accessed 11/03/2020, https://www.uptodate.com/contents/multiple-myeloma-use-of-autologous-hematopoietic-cell-transplantation?search=myeloma%20treatment &topicRef=6643&source=see_link#H5
  143. Talamo G, Dimaio C, Abbi KK, et al. Current role of radiation therapy for multiple myeloma. Frontiers in oncology. 2015;5:40. doi:10.3389/fonc.2015.00040
  144. Tsang RW, Campbell BA, Goda JS, et al. Radiation Therapy for Solitary Plasmacytoma and Multiple Myeloma: Guidelines From the International Lymphoma Radiation Oncology Group. International journal of radiation oncology, biology, physics. Jul 15 2018;101(4):794-808. doi:10.1016/j.ijrobp.2018.05.009
  145. Yussim E, Schwartz E, Sidi Y, Ehrenfeld M. Acute renal failure precipitated by non-steroidal anti-inflammatory drugs (NSAIDs) in multiple myeloma. Am J Hematol. Jun 1998;58(2):142-4. doi:10.1002/(sici)1096-8652(199806)58:2<142::aid-ajh10>3.0.co;2-d
  146. Beguin Y. Erythropoiesis and erythropoietin in multiple myeloma. Leuk Lymphoma. Aug 1995;18(5-6):413-21. doi:10.3109/10428199509059639
  147. Prutchi-Sagiv S, Golishevsky N, Oster HS, et al. Erythropoietin treatment in advanced multiple myeloma is associated with improved immunological functions: could it be beneficial in early disease? Br J Haematol. Dec 2006;135(5):660-72. doi:10.1111/j.1365-2141.2006.06366.x
  148. Pandit A, Leblebjian H, Hammond SP, et al. Safety of live-attenuated measles-mumps-rubella and herpes zoster vaccination in multiple myeloma patients on maintenance lenalidomide or bortezomib after autologous hematopoietic cell transplantation. Bone marrow transplantation. Jul 2018;53(7):942-945. doi:10.1038/s41409-018-0112-x
  149. Oken MM, Pomeroy C, Weisdorf D, Bennett JM. Prophylactic antibiotics for the prevention of early infection in multiple myeloma. Am J Med. Jun 1996;100(6):624-8. doi:10.1016/s0002-9343(95)00043-7
  150. Renaud L, Schraen S, Fouquet G, et al. Response to pneumococcal vaccination in multiple myeloma. Cancer Med. Jul 2019;8(8):3822-3830. doi:10.1002/cam4.2253
  151. Terpos E, Berenson J, Cook RJ, Lipton A, Coleman RE. Prognostic variables for survival and skeletal complications in patients with multiple myeloma osteolytic bone disease. Leukemia. May 2010;24(5):1043-9. doi:10.1038/leu.2010.62
  152. Terpos E, Ntanasis-Stathopoulos I, Gavriatopoulou M, Dimopoulos MA. Pathogenesis of bone disease in multiple myeloma: from bench to bedside. Blood cancer journal. Jan 12 2018;8(1):7. doi:10.1038/s41408-017-0037-4
  153. Hameed A, Brady JJ, Dowling P, Clynes M, O'Gorman P. Bone disease in multiple myeloma: pathophysiology and management. Cancer growth and metastasis. 2014;7:33-42. doi:10.4137/CGM.S16817
  154. Roodman GD. Role of the bone marrow microenvironment in multiple myeloma. J Bone Miner Res. Nov 2002;17(11):1921-5. doi:10.1359/jbmr.2002.17.11.1921
  155. Borset M, Sundan A, Waage A, Standal T. Why do myeloma patients have bone disease? A historical perspective. Blood reviews. Nov 29 2019:100646. doi:10.1016/j.blre.2019.100646
  156. Panaroni C, Yee AJ, Raje NS. Myeloma and Bone Disease. Current osteoporosis reports. Oct 2017;15(5):483-498. doi:10.1007/s11914-017-0397-5
  157. Berenson JR, Lichtenstein A, Porter L, et al. Long-term pamidronate treatment of advanced multiple myeloma patients reduces skeletal events. Myeloma Aredia Study Group. J Clin Oncol. Feb 1998;16(2):593-602. doi:10.1200/JCO.1998.16.2.593
  158. Major P, Lortholary A, Hon J, et al. Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials. J Clin Oncol. Jan 15 2001;19(2):558-67. doi:10.1200/JCO.2001.19.2.558
  159. Lee OL, Horvath N, Lee C, et al. Bisphosphonate guidelines for treatment and prevention of myeloma bone disease. Internal medicine journal. Aug 2017;47(8):938-951. doi:10.1111/imj.13502
  160. Mhaskar R, Kumar A, Miladinovic B, Djulbegovic B. Bisphosphonates in multiple myeloma: an updated network meta-analysis. The Cochrane database of systematic reviews. Dec 18 2017;12:CD003188. doi:10.1002/14651858.CD003188.pub4
  161. Mhaskar R, Redzepovic J, Wheatley K, et al. Bisphosphonates in multiple myeloma: a network meta-analysis. The Cochrane database of systematic reviews. May 16 2012;(5):CD003188. doi:10.1002/14651858.CD003188.pub3
  162. Raje N, Terpos E, Willenbacher W, et al. Denosumab versus zoledronic acid in bone disease treatment of newly diagnosed multiple myeloma: an international, double-blind, double-dummy, randomised, controlled, phase 3 study. The Lancet Oncology. Mar 2018;19(3):370-381. doi:10.1016/S1470-2045(18)30072-X
  163. Fusaro M, Mereu MC, Aghi A, Iervasi G, Gallieni M. Vitamin K and bone. Clin Cases Miner Bone Metab. May-Aug 2017;14(2):200-206. doi:10.11138/ccmbm/2017.14.1.200
  164. Akbari S, Rasouli-Ghahroudi AA. Vitamin K and Bone Metabolism: A Review of the Latest Evidence in Preclinical Studies. BioMed research international. 2018;2018:4629383-4629383. doi:10.1155/2018/4629383
  165. Weber P. Vitamin K and bone health. Nutrition (Burbank, Los Angeles County, Calif). Oct 2001;17(10):880-7. doi:10.1016/s0899-9007(01)00709-2
  166. Shiraki M, Shiraki Y, Aoki C, Miura M. Vitamin K2 (menatetrenone) effectively prevents fractures and sustains lumbar bone mineral density in osteoporosis. J Bone Miner Res. Mar 2000;15(3):515-21. doi:10.1359/jbmr.2000.15.3.515
  167. Cockayne S, Adamson J, Lanham-New S, Shearer MJ, Gilbody S, Torgerson DJ. Vitamin K and the prevention of fractures: systematic review and meta-analysis of randomized controlled trials. Arch Intern Med. Jun 26 2006;166(12):1256-61. doi:10.1001/archinte.166.12.1256
  168. Huang ZB, Wan SL, Lu YJ, Ning L, Liu C, Fan SW. Does vitamin K2 play a role in the prevention and treatment of osteoporosis for postmenopausal women: a meta-analysis of randomized controlled trials. Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA . Mar 2015;26(3):1175-86. doi:10.1007/s00198-014-2989-6
  169. Cheung AM, Tile L, Lee Y, et al. Vitamin K supplementation in postmenopausal women with osteopenia (ECKO trial): a randomized controlled trial. PLoS Med. Oct 14 2008;5(10):e196. doi:10.1371/journal.pmed.0050196
  170. Tsujioka T, Miura Y, Otsuki T, et al. The mechanisms of vitamin K2-induced apoptosis of myeloma cells. Haematologica. May 2006;91(5):613-9.
  171. Booth SL, Dallal G, Shea MK, Gundberg C, Peterson JW, Dawson-Hughes B. Effect of vitamin K supplementation on bone loss in elderly men and women. J Clin Endocrinol Metab. Apr 2008;93(4):1217-23. doi:10.1210/jc.2007-2490
  172. Inoue T, Fujita T, Kishimoto H, et al. Randomized controlled study on the prevention of osteoporotic fractures (OF study): a phase IV clinical study of 15-mg menatetrenone capsules. Journal of bone and mineral metabolism. 2009;27(1):66-75. doi:10.1007/s00774-008-0008-8
  173. Booth SL, Broe KE, Gagnon DR, et al. Vitamin K intake and bone mineral density in women and men. Am J Clin Nutr. Feb 2003;77(2):512-6. doi:10.1093/ajcn/77.2.512
  174. Grammatico S, Scalzulli E, Petrucci MT. Solitary Plasmacytoma. Mediterranean journal of hematology and infectious diseases. 2017;9(1):e2017052-e2017052. doi:10.4084/MJHID.2017.052
  175. Knobel D, Zouhair A, Tsang RW, et al. Prognostic factors in solitary plasmacytoma of the bone: a multicenter Rare Cancer Network study. BMC cancer. May 5 2006;6:118. doi:10.1186/1471-2407-6-118
  176. Tournier-Rangeard L, Lapeyre M, Graff-Caillaud P, et al. Radiotherapy for solitary extramedullary plasmacytoma in the head-and-neck region: A dose greater than 45 Gy to the target volume improves the local control. International journal of radiation oncology, biology, physics. Mar 15 2006;64(4):1013-7. doi:10.1016/j.ijrobp.2005.09.019
  177. Cesana C, Klersy C, Barbarano L, et al. Prognostic factors for malignant transformation in monoclonal gammopathy of undetermined significance and smoldering multiple myeloma. J Clin Oncol. Mar 15 2002;20(6):1625-34. doi:10.1200/JCO.2002.20.6.1625
  178. Rajkumar SV, Kyle RA, Connor RF. Smoldering multiple myeloma. UpToDate. Updated 03/03/2020. Accessed 08/24/2020, https://www.uptodate.com/contents/smoldering-multiple-myeloma?search=smoldering%20multiple%20myeloma &source=search_result&selectedTitle=1~56&usage_type=default&display_rank=1
  179. Lakshman A, Rajkumar SV, Buadi FK, et al. Risk stratification of smoldering multiple myeloma incorporating revised IMWG diagnostic criteria. Blood cancer journal. Jun 12 2018;8(6):59. doi:10.1038/s41408-018-0077-4
  180. Mateos MV, Hernandez MT, Giraldo P, et al. Lenalidomide plus dexamethasone versus observation in patients with high-risk smouldering multiple myeloma (QuiRedex): long-term follow-up of a randomised, controlled, phase 3 trial. The Lancet Oncology. Aug 2016;17(8):1127-1136. doi:10.1016/S1470-2045(16)30124-3
  181. Mateos MV, Hernandez MT, Giraldo P, et al. Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. The New England journal of medicine. Aug 1 2013;369(5):438-47. doi:10.1056/NEJMoa1300439
  182. Lonial S, Jacobus S, Fonseca R, et al. Randomized Trial of Lenalidomide Versus Observation in Smoldering Multiple Myeloma. J Clin Oncol. Apr 10 2020;38(11):1126-1137. doi:10.1200/JCO.19.01740
  183. Rajkumar SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. The Lancet Oncology. Jan 2010;11(1):29-37. doi:10.1016/S1470-2045(09)70284-0
  184. Kumar DK, Choi SH, Washicosky KJ, et al. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Science translational medicine. May 25 2016;8(340):340ra72. doi:10.1126/scitranslmed.aaf1059
  185. Kumar SK, Therneau TM, Gertz MA, et al. Clinical course of patients with relapsed multiple myeloma. Mayo Clin Proc. Jul 2004;79(7):867-74. doi:10.4065/79.7.867
  186. Stewart AK, Rajkumar SV, Dimopoulos MA, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. The New England journal of medicine. Jan 8 2015;372(2):142-52. doi:10.1056/NEJMoa1411321
  187. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. The New England journal of medicine. Oct 6 2016;375(14):1319-1331. doi:10.1056/NEJMoa1607751
  188. Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. The New England journal of medicine. Aug 13 2015;373(7):621-31. doi:10.1056/NEJMoa1505654
  189. Hari P, Matous JV, Voorhees PM, et al. Oprozomib in patients with newly diagnosed multiple myeloma. Blood cancer journal. Aug 16 2019;9(9):66. doi:10.1038/s41408-019-0232-6
  190. Boccia RV, Bessudo A, Agajanian R, et al. A Multicenter, Open-Label, Phase 1b Study of Carfilzomib, Cyclophosphamide, and Dexamethasone in Newly Diagnosed Multiple Myeloma Patients (CHAMPION-2). Clin Lymphoma Myeloma Leuk. Jul 2017;17(7):433-437. doi:10.1016/j.clml.2017.05.009
  191. Ito S. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Cancers. 2020;12(2):E265. doi:10.3390/cancers12020265
  192. Zhou HJ, Aujay MA, Bennett MK, et al. Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). J Med Chem. May 14 2009;52(9):3028-38. doi:10.1021/jm801329v
  193. Richardson PG, Zimmerman TM, Hofmeister CC, et al. Phase 1 study of marizomib in relapsed or relapsed and refractory multiple myeloma: NPI-0052-101 Part 1. Blood. Jun 2 2016;127(22):2693-700. doi:10.1182/blood-2015-12-686378
  194. Spencer A, Harrison S, Zonder J, et al. A phase 1 clinical trial evaluating marizomib, pomalidomide and low-dose dexamethasone in relapsed and refractory multiple myeloma (NPI-0052-107): final study results. Br J Haematol. Jan 2018;180(1):41-51. doi:10.1111/bjh.14987
  195. Trudel S, Lendvai N, Popat R, et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy from dose expansion phase I study. Blood cancer journal. Mar 20 2019;9(4):37. doi:10.1038/s41408-019-0196-6
  196. Iftikhar A, Hassan H, Iftikhar N, et al. Investigational Monoclonal Antibodies in the Treatment of Multiple Myeloma: A Systematic Review of Agents under Clinical Development. Antibodies (Basel). 2019;8(2):34. doi:10.3390/antib8020034
  197. FDA. Food and Drug Administration. FDA granted accelerated approval to belantamab mafodotin-blmf for multiple myeloma. Updated 8/6/2020. Accessed 9/29/2020, https://www.fda.gov/drugs/drug-approvals-and-databases/fda-granted-accelerated-approval-belantamab-mafodotin-blmf-multiple-myeloma
  198. Moreau P, Mikhael J. Isatuximab Plus Carfilzomib and Dexamethasone vs Carfilzomib and Dexamethasone in Relapsed/Refractory Multiple Myeloma (IKEMA): Interim Analysis of A Phase 3 Randomized, Open-Label Study. 2020:
  199. Cho SF, Anderson KC, Tai YT. BCMA CAR T-cell therapy arrives for multiple myeloma: a reality. Ann Transl Med. Dec 2018;6(Suppl 2):S93. doi:10.21037/atm.2018.11.14
  200. Lin Q, Zhao J, Song Y, Liu D. Recent updates on CAR T clinical trials for multiple myeloma. Molecular cancer. Nov 5 2019;18(1):154. doi:10.1186/s12943-019-1092-1
  201. Timmers M, Roex G, Wang Y, et al. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front Immunol. 2019;10:1613. doi:10.3389/fimmu.2019.01613
  202. Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. The New England journal of medicine. May 2 2019;380(18):1726-1737. doi:10.1056/NEJMoa1817226
  203. Novick S. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). Identifier: NCT03651128. A Phase 3, Multicenter, Randomized, Open-label Study to Compare the Efficacy and Safety of bb2121 Versus Standard Regimens in Subjects With Relapsed and Refractory Muleiple Myeloma (RRMM) (MarMMa-3). Last updated 8/11/2020. Available from: https://clinicaltrials.gov/ct2/show/NCT03651128 . Accessed 9/29/2020,
  204. Carmon L, Avivi I, Kovjazin R, et al. Phase I/II study exploring ImMucin, a pan-major histocompatibility complex, anti-MUC1 signal peptide vaccine, in multiple myeloma patients. Br J Haematol. Apr 2015;169(1):44-56. doi:10.1111/bjh.13245
  205. Summary Review for Regulatory Action: Farydak (2015).
  206. Gao X, Shen L, Li X, Liu J. Efficacy and toxicity of histone deacetylase inhibitors in relapsed/refractory multiple myeloma: Systematic review and meta-analysis of clinical trials. Experimental and therapeutic medicine. 2019;18(2):1057-1068. doi:10.3892/etm.2019.7704
  207. Dresser R, Frader J. Off-label prescribing: a call for heightened professional and government oversight. The Journal of law, medicine & ethics : a journal of the American Society of Law, Medicine & Ethics . Fall 2009;37(3):476-396. doi:10.1111/j.1748-720X.2009.00408.x
  208. Kozanoglu I, Yandim MK, Cincin ZB, Ozdogu H, Cakmakoglu B, Baran Y. New indication for therapeutic potential of an old well-known drug (propranolol) for multiple myeloma. Journal of cancer research and clinical oncology. Feb 2013;139(2):327-35. doi:10.1007/s00432-012-1331-y
  209. Ji Y, Chen S, Xiao X, Zheng S, Li K. beta-blockers: a novel class of antitumor agents. OncoTargets and therapy. 2012;5:391-401. doi:10.2147/ott.S38403
  210. Chim H, Armijo BS, Miller E, Gliniak C, Serret MA, Gosain AK. Propranolol induces regression of hemangioma cells through HIF-1alpha-mediated inhibition of VEGF-A. Ann Surg. Jul 2012;256(1):146-56. doi:10.1097/SLA.0b013e318254ce7a
  211. Yang EV, Sood AK, Chen M, et al. Norepinephrine up-regulates the expression of vascular endothelial growth factor, matrix metalloproteinase (MMP)-2, and MMP-9 in nasopharyngeal carcinoma tumor cells. Cancer Res. Nov 1 2006;66(21):10357-64. doi:10.1158/0008-5472.Can-06-2496
  212. Yang EV, Kim SJ, Donovan EL, et al. Norepinephrine upregulates VEGF, IL-8, and IL-6 expression in human melanoma tumor cell lines: implications for stress-related enhancement of tumor progression. Brain Behav Immun . Feb 2009;23(2):267-75. doi:10.1016/j.bbi.2008.10.005
  213. Guo K, Ma Q, Wang L, et al. Norepinephrine-induced invasion by pancreatic cancer cells is inhibited by propranolol. Oncology reports. Oct 2009;22(4):825-30. doi:10.3892/or_00000505
  214. Hajighasemi F, Hajighasemi S. Effect of propranolol on angiogenic factors in human hematopoietic cell lines in vitro. Iran Biomed J. Oct 2009;13(4):223-8.
  215. Wolter NE, Wolter JK, Enepekides DJ, Irwin MS. Propranolol as a novel adjunctive treatment for head and neck squamous cell carcinoma. J Otolaryngol Head Neck Surg. Oct 2012;41(5):334-44.
  216. Pasquier E, Street J, Pouchy C, et al. beta-blockers increase response to chemotherapy via direct antitumour and anti-angiogenic mechanisms in neuroblastoma. British journal of cancer. Jun 25 2013;108(12):2485-94. doi:10.1038/bjc.2013.205
  217. Madden KS, Szpunar MJ, Brown EB. beta-Adrenergic receptors (beta-AR) regulate VEGF and IL-6 production by divergent pathways in high beta-AR-expressing breast cancer cell lines. Breast cancer research and treatment. Dec 2011;130(3):747-58. doi:10.1007/s10549-011-1348-y
  218. Hwa YL, Shi Q, Kumar SK, et al. Beta-blockers improve survival outcomes in patients with multiple myeloma: a retrospective evaluation. Am J Hematol. Jan 2017;92(1):50-55. doi:10.1002/ajh.24582
  219. Trojan PJ, Bohatch-Junior MS, Otuki MF, et al. Pravastatin induces cell cycle arrest and decreased production of VEGF and bFGF in multiple myeloma cell line. Brazilian journal of biology = Revista brasleira de biologia. Feb 2016;76(1):59-65. doi:10.1590/1519-6984.11914
  220. van de Donk NW, Kamphuis MM, Lokhorst HM, Bloem AC. The cholesterol lowering drug lovastatin induces cell death in myeloma plasma cells. Leukemia. Jul 2002;16(7):1362-71. doi:10.1038/sj.leu.2402501
  221. Slawinska-Brych A, Zdzisinska B, Mizerska-Dudka M, Kandefer-Szerszen M. Induction of apoptosis in multiple myeloma cells by a statin-thalidomide combination can be enhanced by p38 MAPK inhibition. Leuk Res. May 2013;37(5):586-94. doi:10.1016/j.leukres.2013.01.022
  222. Perucha E, Melchiotti R, Bibby JA, et al. The cholesterol biosynthesis pathway regulates IL-10 expression in human Th1 cells. Nature communications. Jan 30 2019;10(1):498. doi:10.1038/s41467-019-08332-9
  223. Tsubaki M, Mashimo K, Takeda T, et al. Statins inhibited the MIP-1alpha expression via inhibition of Ras/ERK and Ras/Akt pathways in myeloma cells. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie . Mar 2016;78:23-29. doi:10.1016/j.biopha.2015.12.017
  224. Sanfilippo KM, Keller J, Gage BF, et al. Statins Are Associated With Reduced Mortality in Multiple Myeloma. J Clin Oncol. Nov 20 2016;34(33):4008-4014. doi:10.1200/jco.2016.68.3482
  225. Chiu BC-H, Chen J-H, Yen Y-C, et al. Long Term Statin Use and Risk of Multiple Myeloma Among 15.5 Million Taiwanese Adults: A Retrospective Cohort Study. Blood. 2015;126(23):4198-4198. doi:10.1182/blood.V126.23.4198.4198
  226. Epstein MM, Divine G, Chao CR, et al. Statin use and risk of multiple myeloma: An analysis from the cancer research network. International journal of cancer Journal international du cancer. Aug 1 2017;141(3):480-487. doi:10.1002/ijc.30745
  227. Ponvilawan B, Charoenngam N, Rittiphairoj T, Ungprasert P. Receipt of Statins Is Associated With Lower Risk of Multiple Myeloma: Systematic Review and Meta-analysis. Clin Lymphoma Myeloma Leuk. Feb 26 2020;doi:10.1016/j.clml.2020.02.011
  228. Sondergaard TE, Pedersen PT, Andersen TL, et al. A phase II clinical trial does not show that high dose simvastatin has beneficial effect on markers of bone turnover in multiple myeloma. Hematol Oncol. Mar 2009;27(1):17-22. doi:10.1002/hon.869
  229. Hus M, Grzasko N, Szostek M, et al. Thalidomide, dexamethasone and lovastatin with autologous stem cell transplantation as a salvage immunomodulatory therapy in patients with relapsed and refractory multiple myeloma. Annals of hematology. Oct 2011;90(10):1161-6. doi:10.1007/s00277-011-1276-2
  230. Mishra AK, Dingli D. Metformin inhibits IL-6 signaling by decreasing IL-6R expression on multiple myeloma cells. Leukemia. Nov 2019;33(11):2695-2709. doi:10.1038/s41375-019-0470-4
  231. Xu P, Yin K, Tang X, et al. Metformin inhibits the function of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie . Dec 2019;120:109458. doi:10.1016/j.biopha.2019.109458
  232. Chang SH, Luo S, O'Brian KK, et al. Association between metformin use and progression of monoclonal gammopathy of undetermined significance to multiple myeloma in US veterans with diabetes mellitus: a population-based retrospective cohort study. Lancet Haematol. Jan 2015;2(1):e30-6. doi:10.1016/s2352-3026(14)00037-4
  233. Boursi B, Mamtani R, Yang YX, Weiss BM. Impact of metformin on the progression of MGUS to multiple myeloma. Leuk Lymphoma. May 2017;58(5):1265-1267. doi:10.1080/10428194.2016.1236375
  234. Wu W, Merriman K, Nabaah A, et al. The association of diabetes and anti-diabetic medications with clinical outcomes in multiple myeloma. British journal of cancer. Jul 29 2014;111(3):628-36. doi:10.1038/bjc.2014.307
  235. Duma N, Vera Aguilera J, Paludo J, et al. Impact of metformin use in the outcomes of multiple myeloma patients post stem cell transplant. J Clin Oncol. 2017;35(15)
  236. Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. Nov 27 2006;203(12):2691-702. doi:10.1084/jem.20061104
  237. Malek E, de Lima M, Letterio JJ, et al. Myeloid-derived suppressor cells: The green light for myeloma immune escape. Blood reviews. Sep 2016;30(5):341-8. doi:10.1016/j.blre.2016.04.002
  238. Peak TC, Richman A, Gur S, Yafi FA, Hellstrom WJ. The Role of PDE5 Inhibitors and the NO/cGMP Pathway in Cancer. Sexual medicine reviews. Jan 2016;4(1):74-84. doi:10.1016/j.sxmr.2015.10.004
  239. Wesolowski R, Markowitz J, Carson WE, 3rd. Myeloid derived suppressor cells - a new therapeutic target in the treatment of cancer. J Immunother Cancer. 2013;1:10. doi:10.1186/2051-1426-1-10
  240. Lad D, Huang Q, Hoeppli R, et al. Evaluating the role of Tregs in the progression of multiple myeloma. Leuk Lymphoma. Sep 2019;60(9):2134-2142. doi:10.1080/10428194.2019.1579324
  241. D'Arena G, Rossi G, Laurenti L, et al. Circulating Regulatory T-Cells in Monoclonal Gammopathies of Uncertain Significance and Multiple Myeloma: In Search of a Role. J Immunol Res. 2016;2016:9271469. doi:10.1155/2016/9271469
  242. Wang JN, Cao XX, Zhao AL, Cai H, Wang X, Li J. Increased activated regulatory T cell subsets and aging Treg-like cells in multiple myeloma and monoclonal gammopathy of undetermined significance: a case control study. Cancer Cell Int. 2018;18:187. doi:10.1186/s12935-018-0687-8
  243. Weed DT, Vella JL, Reis IM, et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. Jan 1 2015;21(1):39-48. doi:10.1158/1078-0432.Ccr-14-1711
  244. Zhang N, Bevan MJ. CD8(+) T cells: foot soldiers of the immune system. Immunity. Aug 26 2011;35(2):161-8. doi:10.1016/j.immuni.2011.07.010
  245. International Myeloma Foundation. Understanding the immune system in myeloma. Accessed April 23, 2020, https://www.myeloma.org/sites/default/files/resource/u-immune.pdf
  246. Tai LH, Alkayyal AA, Leslie AL, et al. Phosphodiesterase-5 inhibition reduces postoperative metastatic disease by targeting surgery-induced myeloid derived suppressor cell-dependent inhibition of Natural Killer cell cytotoxicity. Oncoimmunology. 2018;7(6):e1431082. doi:10.1080/2162402x.2018.1431082
  247. Ghosh N, Rudraraju L, Ye X, Noonan K, Huff CA, Borrello IM. Administration Of An Oral PDE5 Inhibitor, Tadalafil In Conjunction With a Lenalidomide Containing Regimen In Patients With Multiple Myeloma. Blood. 2013;122(21):1959-1959. doi:10.1182/blood.V122.21.1959.1959
  248. Noonan KA, Ghosh N, Rudraraju L, Bui M, Borrello I. Targeting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer immunology research. 2014;2(8):725-731. doi:10.1158/2326-6066.CIR-13-0213
  249. Patterson CJ, Soumerai J, Hunter Z, Leleu X, Ghobrial IM, Treon SP. Sildenafil citrate suppresses disease progression in patients with Waldenstrom’s macroglobulinemia. J Clin Oncol. 2006;24(18):7556.
  250. Zhang M, Abe Y, Matsushima T, Nishimura J, Nawata H, Muta K. Selective cyclooxygenase 2 inhibitor NS-398 induces apoptosis in myeloma cells via a Bcl-2 independent pathway. Leuk Lymphoma. Mar 2005;46(3):425-33. doi:10.1080/10428190400015691
  251. Ladetto M, Vallet S, Trojan A, et al. Cyclooxygenase-2 (COX-2) is frequently expressed in multiple myeloma and is an independent predictor of poor outcome. Blood. Jun 15 2005;105(12):4784-91. doi:10.1182/blood-2004-11-4201
  252. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. May 1 2007;67(9):4507-13. doi:10.1158/0008-5472.Can-06-4174
  253. Rodriguez PC, Hernandez CP, Quiceno D, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. Oct 3 2005;202(7):931-9. doi:10.1084/jem.20050715
  254. Talmadge JE, Hood KC, Zobel LC, Shafer LR, Coles M, Toth B. Chemoprevention by cyclooxygenase-2 inhibition reduces immature myeloid suppressor cell expansion. International immunopharmacology. Feb 2007;7(2):140-51. doi:10.1016/j.intimp.2006.09.021
  255. Veltman JD, Lambers ME, van Nimwegen M, et al. COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC cancer. Aug 30 2010;10:464. doi:10.1186/1471-2407-10-464
  256. Rosas C, Sinning M, Ferreira A, Fuenzalida M, Lemus D. Celecoxib decreases growth and angiogenesis and promotes apoptosis in a tumor cell line resistant to chemotherapy. Biol Res. Jun 16 2014;47:27. doi:10.1186/0717-6287-47-27
  257. Ninomiya I, Nagai N, Oyama K, et al. Antitumor and anti-metastatic effects of cyclooxygenase-2 inhibition by celecoxib on human colorectal carcinoma xenografts in nude mouse rectum. Oncology reports. Sep 2012;28(3):777-84. doi:10.3892/or.2012.1885
  258. Minter HA, Eveson JW, Huntley S, Elder DJ, Hague A. The cyclooxygenase 2-selective inhibitor NS398 inhibits proliferation of oral carcinoma cell lines by mechanisms dependent and independent of reduced prostaglandin E2 synthesis. Clinical cancer research : an official journal of the American Association for Cancer Research . May 2003;9(5):1885-97.
  259. Yao M, Lam EC, Kelly CR, Zhou W, Wolfe MM. Cyclooxygenase-2 selective inhibition with NS-398 suppresses proliferation and invasiveness and delays liver metastasis in colorectal cancer. Br J Cancer. Feb 9 2004;90(3):712-9. doi:10.1038/sj.bjc.6601489
  260. Waskewich C, Blumenthal RD, Li H, Stein R, Goldenberg DM, Burton J. Celecoxib exhibits the greatest potency amongst cyclooxygenase (COX) inhibitors for growth inhibition of COX-2-negative hematopoietic and epithelial cell lines. Cancer Res. Apr 1 2002;62(7):2029-33.
  261. Zhang GS, Liu DS, Dai CW, Li RJ. Antitumor effects of celecoxib on K562 leukemia cells are mediated by cell-cycle arrest, caspase-3 activation, and downregulation of Cox-2 expression and are synergistic with hydroxyurea or imatinib. Am J Hematol. Apr 2006;81(4):242-55. doi:10.1002/ajh.20542
  262. Prince HM, Mileshkin L, Roberts A, et al. A multicenter phase II trial of thalidomide and celecoxib for patients with relapsed and refractory multiple myeloma. Clin Cancer Res. Aug 1 2005;11(15):5504-14. doi:10.1158/1078-0432.Ccr-05-0213
  263. Kalaycio M. Celecoxib in Preventing Multiple Myeloma in Patients With Monoclonal Gammopathy or Smoldering Myeloma, NCT00099047. U.S. National Library of Medicine. Updated 12/30/2016. Accessed 09/01/2020, https://clinicaltrials.gov/ct2/show/study/NCT00099047
  264. Ogata A, Nishimoto N, Shima Y, Yoshizaki K, Kishimoto T. Inhibitory effect of all-trans retinoic acid on the growth of freshly isolated myeloma cells via interference with interleukin-6 signal transduction. Blood. Nov 1 1994;84(9):3040-6.
  265. Sidell N, Taga T, Hirano T, Kishimoto T, Saxon A. Retinoic acid-induced growth inhibition of a human myeloma cell line via down-regulation of IL-6 receptors. J Immunol. Jun 1 1991;146(11):3809-14.
  266. Taetle R, Dos Santos B, Akamatsu K, Koishihara Y, Ohsugi Y. Effects of all-trans retinoic acid and antireceptor antibodies on growth and programmed cell death of human myeloma cells. Clin Cancer Res. Feb 1996;2(2):253-9.
  267. Otsuki T, Sakaguchi H, Hatayama T, Wu P, Takata A, Hyodoh F. Effects of all-trans retinoic acid (ATRA) on human myeloma cells. Leuk Lymphoma. Oct 2003;44(10):1651-6. doi:10.1080/1042819031000099652
  268. Huang H, Wu D, Fu J, et al. All-trans retinoic acid can intensify the growth inhibition and differentiation induction effect of rosiglitazone on multiple myeloma cells. Eur J Haematol. Sep 2009;83(3):191-202. doi:10.1111/j.1600-0609.2009.01277.x
  269. van de Donk W. Daratumumab in Combination With ATRA (DARA/ATRA). NIH. Accessed 04/29/2020, https://clinicaltrials.gov/ct2/show/NCT02751255
  270. Hengesbach LM, Hoag KA. Physiological concentrations of retinoic acid favor myeloid dendritic cell development over granulocyte development in cultures of bone marrow cells from mice. The Journal of nutrition. Oct 2004;134(10):2653-9. doi:10.1093/jn/134.10.2653
  271. Kusmartsev S, Su Z, Heiser A, et al. Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma. Clin Cancer Res. Dec 15 2008;14(24):8270-8. doi:10.1158/1078-0432.Ccr-08-0165
  272. Kusmartsev S, Cheng F, Yu B, et al. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res. Aug 1 2003;63(15):4441-9.
  273. Braga WM, Atanackovic D, Colleoni GW. The role of regulatory T cells and TH17 cells in multiple myeloma. Clin Dev Immunol. 2012;2012:293479. doi:10.1155/2012/293479
  274. Bryant C, Suen H, Brown R, et al. Long-term survival in multiple myeloma is associated with a distinct immunological profile, which includes proliferative cytotoxic T-cell clones and a favourable Treg/Th17 balance. Blood Cancer J. Sep 13 2013;3:e148. doi:10.1038/bcj.2013.34
  275. Park H, Li Z, Yang XO, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. Nov 2005;6(11):1133-41. doi:10.1038/ni1261
  276. Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol. Apr 2007;8(4):345-50. doi:10.1038/ni0407-345
  277. Wu S, Wang W, Le Q. All-trans Retinoic Acid Regulates the Balance of Treg-Th17 Cells through ERK and P38 Signaling Pathway. Iran J Immunol. Mar 2019;16(1):1-10. doi:10.22034/iji.2019.39402
  278. Musto P, Sajeva MR, Sanpaolo G, D'Arena G, Scalzulli PR, Carotenuto M. All-trans retinoic acid in combination with alpha-interferon and dexamethasone for advanced multiple myeloma. Haematologica. May-Jun 1997;82(3):354-6.
  279. Avilés A, Rosas A, Huerta-Guzmán J, Talavera A, Cleto S. Dexamethasone, all trans retinoic acid and interferon alpha 2a in patients with refractory multiple myeloma. Cancer Biother Radiopharm. Feb 1999;14(1):23-6. doi:10.1089/cbr.1999.14.23
  280. Juturi J, Bukowski RA, Bocock K, Bloom T, Finke J, Hussein MA. High, intermittent dose of all-trans retinoic acid in combination with alpha-interferon for advanced multiple myeloma. Haematologica. Jul 2001;86(7):776-7.
  281. Koskela K, Pelliniemi TT, Pulkki K, Remes K. Treatment of multiple myeloma with all-trans retinoic acid alone and in combination with chemotherapy: a phase I/II trial. Leuk Lymphoma. Apr 2004;45(4):749-54. doi:10.1080/10428190310001628158
  282. Mao X, Liang S-b, Hurren R, et al. Cyproheptadine displays preclinical activity in myeloma and leukemia. Blood, The Journal of the American Society of Hematology. 2008;112(3):760-769.
  283. Ellegaard AM, Dehlendorff C, Vind AC, et al. Repurposing Cationic Amphiphilic Antihistamines for Cancer Treatment. EBioMedicine. Jul 2016;9:130-139. doi:10.1016/j.ebiom.2016.06.013
  284. Tutton PJ, Barkla DH. Comparison of the tumor inhibiting effects of three histamine H2-receptor antagonists. Anticancer research. Jan-Feb 1983;3(1):7-10.
  285. Zheng Y, Xu M, Li X, Jia J, Fan K, Lai G. Cimetidine suppresses lung tumor growth in mice through proapoptosis of myeloid-derived suppressor cells. Mol Immunol. May 2013;54(1):74-83. doi:10.1016/j.molimm.2012.10.035
  286. Dana P, Vaeteewoottacharn K, Kariya R, Matsuda K, Wongkham S, Okada S. Repurposing cimetidine for cholangiocarcinoma: Antitumor effects in vitro and in vivo. Oncology letters. Mar 2017;13(3):1432-1436. doi:10.3892/ol.2017.5563
  287. Jiang CG, Liu FR, Yu M, Li JB, Xu HM. Cimetidine induces apoptosis in gastric cancer cells in vitro and inhibits tumor growth in vivo. Oncology reports. Mar 2010;23(3):693-700. doi:10.3892/or_00000686
  288. Chen JS, Lin SY, Tso WL, et al. Checkpoint kinase 1-mediated phosphorylation of Cdc25C and bad proteins are involved in antitumor effects of loratadine-induced G2/M phase cell-cycle arrest and apoptosis. Molecular carcinogenesis. Jul 2006;45(7):461-78. doi:10.1002/mc.20165
  289. Soule BP, Simone NL, DeGraff WG, Choudhuri R, Cook JA, Mitchell JB. Loratadine dysregulates cell cycle progression and enhances the effect of radiation in human tumor cell lines. Radiation oncology (London, England). Feb 3 2010;5:8. doi:10.1186/1748-717x-5-8
  290. O'Mahony L, Akdis M, Akdis CA. Regulation of the immune response and inflammation by histamine and histamine receptors. J Allergy Clin Immunol. Dec 2011;128(6):1153-62. doi:10.1016/j.jaci.2011.06.051
  291. Li Y, Yang GL, Yuan HY, et al. Effects of perioperative cimetidine administration on peripheral blood lymphocytes and tumor infiltrating lymphocytes in patients with gastrointestinal cancer: results of a randomized controlled clinical trial. Hepatogastroenterology. Mar-Apr 2005;52(62):504-8.
  292. Martin RK, Saleem SJ, Folgosa L, et al. Mast cell histamine promotes the immunoregulatory activity of myeloid-derived suppressor cells. Journal of leukocyte biology. Jul 2014;96(1):151-9. doi:10.1189/jlb.5A1213-644R
  293. Elenkov IJ, Webster E, Papanicolaou DA, Fleisher TA, Chrousos GP, Wilder RL. Histamine potently suppresses human IL-12 and stimulates IL-10 production via H2 receptors. J Immunol. Sep 1 1998;161(5):2586-93.
  294. Ogawara H, Handa H, Yamazaki T, et al. High Th1/Th2 ratio in patients with multiple myeloma. Leuk Res. Feb 2005;29(2):135-40. doi:10.1016/j.leukres.2004.06.003
  295. Murakami H, Ogawara H, Hiroshi H. Th1/Th2 cells in patients with multiple myeloma. Hematology. Feb 2004;9(1):41-5. doi:10.1080/10245330310001652437
  296. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology. Feb 2013;138(2):105-15. doi:10.1111/imm.12036
  297. Vila-Leahey A, Oldford SA, Marignani PA, Wang J, Haidl ID, Marshall JS. Ranitidine modifies myeloid cell populations and inhibits breast tumor development and spread in mice. Oncoimmunology. Jul 2016;5(7):e1151591. doi:10.1080/2162402x.2016.1151591
  298. Kubota T, Fujiwara H, Ueda Y, et al. Cimetidine modulates the antigen presenting capacity of dendritic cells from colorectal cancer patients. British journal of cancer. Apr 22 2002;86(8):1257-61. doi:10.1038/sj.bjc.6600233
  299. Adams WJ, Morris DL. Pilot study--cimetidine enhances lymphocyte infiltration of human colorectal carcinoma: results of a small randomized control trial. Cancer. Jul 1 1997;80(1):15-21. doi:10.1002/(sici)1097-0142(19970701)80:1<15::aid-cncr3>3.0.co;2-e
  300. Lin CY, Bai DJ, Yuan HY, et al. Perioperative cimetidine administration promotes peripheral blood lymphocytes and tumor infiltrating lymphocytes in patients with gastrointestinal cancer: Results of a randomized controlled clinical trial. World J Gastroenterol. Jan 2004;10(1):136-42.
  301. Tomita K, Izumi K, Okabe S. Roxatidine- and cimetidine-induced angiogenesis inhibition suppresses growth of colon cancer implants in syngeneic mice. J Pharmacol Sci. Nov 2003;93(3):321-30. doi:10.1254/jphs.93.321
  302. Chihara Y, Fujimoto K, Miyake M, Hiasa Y, Hirao Y. Anti-tumor effect of cimetidine via inhibiting angiogenesis factors in N-butyl-N-(4-hydroxybutyl) nitrosamine-induced mouse and rat bladder carcinogenesis. Oncol Rep. Jul 2009;22(1):23-8. doi:10.3892/or_00000401
  303. Leurs R, Church MK, Taglialatela M. H1-antihistamines: inverse agonism, anti-inflammatory actions and cardiac effects. Clin Exp Allergy. Apr 2002;32(4):489-98. doi:10.1046/j.0954-7894.2002.01314.x
  304. Hunto ST, Kim HG, Baek KS, et al. Loratadine, an antihistamine drug, exhibits anti-inflammatory activity through suppression of the NF-kB pathway. Biochem Pharmacol. Apr 3 2020;177:113949. doi:10.1016/j.bcp.2020.113949
  305. Lazze MC, Pizzala R, Savio M, Stivala LA, Prosperi E, Bianchi L. Anthocyanins protect against DNA damage induced by tert-butyl-hydroperoxide in rat smooth muscle and hepatoma cells. Mutation research. Feb 5 2003;535(1):103-15.
  306. Nielsen HJ, Nielsen H, Moesgaard F, et al. The effect of ranitidine on cellular immunity in patients with multiple myeloma. Cancer immunology, immunotherapy : CII. 1990;32(3):201-5. doi:10.1007/bf01771458
  307. Shimazaki C, Atzpodien J, Wisniewski D, et al. Cell-mediated toxicity of interleukin-2-activated lymphocytes against autologous and allogeneic human myeloma cells. Acta haematologica. 1988;80(4):203-9. doi:10.1159/000205638
  308. Maekawa R, Matsumoto M, Kitagawa T, Harada M, Sato K. Effect of recombinant interleukin 2 (R-IL2) on in vivo growth of murine myeloma X5563. Cancer Immunology, Immunotherapy. 1986/09/01 1986;23(1):25-30. doi:10.1007/BF00205551
  309. Peest D, de Vries I, Hölscher R, Leo R, Deicher H. Effect of interleukin-2 on the ex vivo growth of human myeloma cells. Cancer immunology, immunotherapy : CII. 1989;30(4):227-32. doi:10.1007/bf01665009
  310. Ross SH, Cantrell DA. Signaling and Function of Interleukin-2 in T Lymphocytes. Annu Rev Immunol. Apr 26 2018;36:411-433. doi:10.1146/annurev-immunol-042617-053352
  311. Cacalano NA, Johnston JA. Interleukin-2 signaling and inherited immunodeficiency. Am J Hum Genet. Aug 1999;65(2):287-93. doi:10.1086/302518
  312. de la Rosa M, Rutz S, Dorninger H, Scheffold A. Interleukin-2 is essential for CD4+CD25+ regulatory T cell function. Eur J Immunol. Sep 2004;34(9):2480-8. doi:10.1002/eji.200425274
  313. Peest D, Leo R, Bloche S, et al. Low-dose recombinant interleukin-2 therapy in advanced multiple myeloma. British Journal of Haematology. 1995;89(2):328-337. doi:10.1111/j.1365-2141.1995.tb03308.x
  314. Morecki S, Revel-Vilk S, Nabet C, et al. Immunological evaluation of patients with hematological malignancies receiving ambulatory cytokine-mediated immunotherapy with recombinant human interferon-alpha 2a and interleukin-2. Cancer immunology, immunotherapy : CII. 1992;35(6):401-11. doi:10.1007/bf01789019
  315. Belch AR, Eisenhauer EA, Muldal A, Browman G, Klasa R, Osterwalder B. Phase II study of subcutaneous rHu-interleukin-2 and rHu-interferon alpha-2a in previously treated patients with multiple myeloma. Ann Oncol. Sep 1995;6(7):721-3. doi:10.1093/oxfordjournals.annonc.a059290
  316. Qiu XH, Zhai YP. [Progress of research on clarithromycin for treatment of multiple myeloma]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. Feb 2015;23(1):246-9. doi:10.7534/j.issn.1009-2137.2015.01.046
  317. Qiu XH, Shao JJ, Mei JG, Li HQ, Cao HQ. Clarithromycin Synergistically Enhances Thalidomide Cytotoxicity in Myeloma Cells. Acta haematologica. 2016;135(2):103-9. doi:10.1159/000438855
  318. Nakamura M, Kikukawa Y, Takeya M, Mitsuya H, Hata H. Clarithromycin attenuates autophagy in myeloma cells. International journal of oncology. 2010;37(4):815-820.
  319. Lai T, Zhao Q, Li F, et al. [Cost-Effectiveness Analysis of Different Chemotherapy Regimens in the Treatment of Patients with Multiple Myeloma]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. Jun 2018;26(3):824-828. doi:10.7534/j.issn.1009-2137.2018.03.032
  320. Van Nuffel AM, Sukhatme V, Pantziarka P, Meheus L, Sukhatme VP, Bouche G. Repurposing Drugs in Oncology (ReDO)-clarithromycin as an anti-cancer agent. Ecancermedicalscience. 2015;9:513. doi:10.3332/ecancer.2015.513
  321. Puig N, Hernández MT, Rosinol Dachs L, et al. Randomized Trial of Lenalidomide and Dexamethasone Versus Clarythromycin, Lenalidomide and Dexamethasone As First Line Treatment in Patients with Multiple Myeloma Not Candidates for Autologous Stem Cell Transplantation: Results of the GEM-Claridex Clinical Trial. Blood. 2019;134(Supplement_1):694-694. doi:10.1182/blood-2019-123997
  322. Holmberg LA, Becker PS, Bensinger W. Results from Two Consecutive Studies of Consolidation Therapy after Autologous Transplant for Multiple Myeloma: Thalidomide, Dexamethasone, and Clarithromycin or Lenalidomide, Dexamethasone, and Clarithromycin. Acta haematologica. 2017;137(3):123-131. doi:10.1159/000455937
  323. Gay F, Rajkumar SV, Coleman M, et al. Clarithromycin (Biaxin)-lenalidomide-low-dose dexamethasone (BiRd) versus lenalidomide-low-dose dexamethasone (Rd) for newly diagnosed myeloma. Am J Hematol. Sep 2010;85(9):664-9. doi:10.1002/ajh.21777
  324. Rossi A, Mark T, Jayabalan D, et al. BiRd (clarithromycin, lenalidomide, dexamethasone): an update on long-term lenalidomide therapy in previously untreated patients with multiple myeloma. Blood. Mar 14 2013;121(11):1982-5. doi:10.1182/blood-2012-08-448563
  325. Mark TM, Forsberg PA, Rossi AC, et al. Phase 2 study of clarithromycin, pomalidomide, and dexamethasone in relapsed or refractory multiple myeloma. Blood Adv. Feb 26 2019;3(4):603-611. doi:10.1182/bloodadvances.2018028027
  326. Fuchs O. Transcription Factor NF-B Inhibitors as Single Therapeutic Agents or in Combination with Classical Chemotherapeutic Agents for the Treatment of Hematologic Malignancies. 2010. p. 98-122.
  327. Endo M, Beppu H, Akiyama H, et al. Agaritine purified from Agaricus blazei Murrill exerts anti-tumor activity against leukemic cells. Biochim Biophys Acta. Jul 2010;1800(7):669-73. doi:10.1016/j.bbagen.2010.03.016
  328. Vrábel D, Pour L, Ševčíková S. The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Churchill Livingstone; 2019. p. 56-66.
  329. Monroe DG, McGee-Lawrence ME, Oursler MJ, Westendorf JJ. Update on Wnt signaling in bone cell biology and bone disease. Gene. Jan 15 2012;492(1):1-18. doi:10.1016/j.gene.2011.10.044
  330. Tangen JM, Holien T, Mirlashari MR, Misund K, Hetland G. Cytotoxic Effect on Human Myeloma Cells and Leukemic Cells by the Agaricus blazei Murill Based Mushroom Extract, Andosan. Biomed Res Int. 2017;2017:2059825. doi:10.1155/2017/2059825
  331. Tangen JM, Tierens A, Caers J, et al. Immunomodulatory effects of the Agaricus blazei Murrill-based mushroom extract AndoSan in patients with multiple myeloma undergoing high dose chemotherapy and autologous stem cell transplantation: a randomized, double blinded clinical study. Biomed Res Int. 2015;2015:718539. doi:10.1155/2015/718539
  332. Cholujova D, Jakubikova J, Czako B, et al. MGN-3 arabinoxylan rice bran modulates innate immunity in multiple myeloma patients. Cancer immunology, immunotherapy : CII. Mar 2013;62(3):437-45. doi:10.1007/s00262-012-1344-z
  333. Ghoneum M, Badr El-Din NK, Ali DA, El-Dein MA. Modified arabinoxylan from rice bran, MGN-3/biobran, sensitizes metastatic breast cancer cells to paclitaxel in vitro. Anticancer research. Jan 2014;34(1):81-7.
  334. Ghoneum M, Gollapudi S. Modified arabinoxylan rice bran (MGN-3/Biobran) sensitizes human T cell leukemia cells to death receptor (CD95)-induced apoptosis. Cancer letters. Nov 10 2003;201(1):41-9. doi:10.1016/s0304-3835(03)00458-0
  335. Yamaguchi M, Weitzmann MN. Quercetin, a potent suppressor of NF-kappaB and Smad activation in osteoblasts. Int J Mol Med. Oct 2011;28(4):521-5. doi:10.3892/ijmm.2011.749
  336. Guo Y, Zhang X, Meng J, Wang ZY. An anticancer agent icaritin induces sustained activation of the extracellular signal-regulated kinase (ERK) pathway and inhibits growth of breast cancer cells. European journal of pharmacology. May 11 2011;658(2-3):114-22. doi:10.1016/j.ejphar.2011.02.005
  337. Wu Z, Ou L, Wang C, et al. Icaritin induces MC3T3-E1 subclone14 cell differentiation through estrogen receptor-mediated ERK1/2 and p38 signaling activation. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie . Oct 2017;94:1-9. doi:10.1016/j.biopha.2017.07.071
  338. Yang XJ, Xi YM, Li ZJ. Icaritin: A Novel Natural Candidate for Hematological Malignancies Therapy. Biomed Res Int. 2019;2019:4860268. doi:10.1155/2019/4860268
  339. Li ZY, Li ZJ, Chen X, et al. [Icaritin Reverses Multidrug Resistance of Multiple Myeloma Cell Line KM3/BTZ]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. Dec 2017;25(6):1690-1695. doi:10.7534/j.issn.1009-2137.2017.06.020
  340. Zhu S, Wang Z, Li Z, et al. Icaritin suppresses multiple myeloma, by inhibiting IL-6/JAK2/STAT3. Oncotarget. Apr 30 2015;6(12):10460-72. doi:10.18632/oncotarget.3399
  341. Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell research. 2002/03/01 2002;12(1):9-18. doi:10.1038/sj.cr.7290105
  342. Rossi A, Voigtlaender M, Janjetovic S, et al. Mutational landscape reflects the biological continuum of plasma cell dyscrasias. Blood cancer journal. 2017;7(2):e537-e537.
  343. Heuck CJ, Jethava Y, Khan R, et al. Inhibiting MEK in MAPK pathway-activated myeloma. Leukemia. 2016;30(4):976-980. doi:10.1038/leu.2015.208
  344. Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. May 2003;4(5):373-84. doi:10.1038/nrm1105
  345. Xu J, Pfarr N, Endris V, et al. Molecular signaling in multiple myeloma: association of RAS/RAF mutations and MEK/ERK pathway activation. Oncogenesis. 2017;6(5):e337-e337.
  346. Garcia-Ruiz C, Morales A, Fernandez-Checa JC. Statins and protein prenylation in cancer cell biology and therapy. Anti-cancer agents in medicinal chemistry. May 2012;12(4):303-15. doi:10.2174/187152012800228715
  347. Islam M, Sharma S, Kumar B, Teknos TN. Atorvastatin inhibits RhoC function and limits head and neck cancer metastasis. Oral oncology. Aug 2013;49(8):778-86. doi:10.1016/j.oraloncology.2013.04.003
  348. Graaf MR, Richel DJ, van Noorden CJ, Guchelaar HJ. Effects of statins and farnesyltransferase inhibitors on the development and progression of cancer. Cancer treatment reviews. Nov 2004;30(7):609-41. doi:10.1016/j.ctrv.2004.06.010
  349. Sarrabayrouse G, Pich C, Teiti I, Tilkin-Mariame AF. Regulatory properties of statins and rho gtpases prenylation inhibitiors to stimulate melanoma immunogenicity and promote anti-melanoma immune response. International journal of cancer Journal international du cancer. Feb 15 2017;140(4):747-755. doi:10.1002/ijc.30422
  350. Fromigue O, Hay E, Modrowski D, et al. RhoA GTPase inactivation by statins induces osteosarcoma cell apoptosis by inhibiting p42/p44-MAPKs-Bcl-2 signaling independently of BMP-2 and cell differentiation. Cell Death Differ. Nov 2006;13(11):1845-56. doi:10.1038/sj.cdd.4401873
  351. Deng JL, Zhang R, Zeng Y, Zhu YS, Wang G. Statins induce cell apoptosis through a modulation of AKT/FOXO1 pathway in prostate cancer cells. Cancer management and research. 2019;11:7231-7242. doi:10.2147/cmar.S212643
  352. Fujiwara D, Tsubaki M, Takeda T, et al. Statins induce apoptosis through inhibition of Ras signaling pathways and enhancement of Bim and p27 expression in human hematopoietic tumor cells. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine . Oct 2017;39(10):1010428317734947. doi:10.1177/1010428317734947
  353. Chiu BCH, Chen JH, Yen YC, et al. Long Term Statin Use and Risk of Multiple Myeloma Among 15.5 Million Taiwanese Adults: A Retrospective Cohort Study. Blood. 2015;126(23):4198.
  354. Hoang B, Frost P, Shi Y, et al. Targeting TORC2 in multiple myeloma with a new mTOR kinase inhibitor. Blood. Nov 25 2010;116(22):4560-8. doi:10.1182/blood-2010-05-285726
  355. Zhang J, Choi Y, Mavromatis B, Lichtenstein A, Li W. Preferential killing of PTEN-null myelomas by PI3K inhibitors through Akt pathway. Oncogene. Sep 18 2003;22(40):6289-95. doi:10.1038/sj.onc.1206718
  356. Li J, Zhu J, Cao B, Mao X. The mTOR signaling pathway is an emerging therapeutic target in multiple myeloma. Curr Pharm Des. 2014;20(1):125-35. doi:10.2174/13816128113199990638
  357. Younes H, Leleu X, Hatjiharissi E, et al. Targeting the phosphatidylinositol 3-kinase pathway in multiple myeloma. Clin Cancer Res. Jul 1 2007;13(13):3771-5. doi:10.1158/1078-0432.Ccr-06-2921
  358. Hemmings BA, Restuccia DF. PI3K-PKB/Akt pathway. Cold Spring Harb Perspect Biol. Sep 1 2012;4(9):a011189. doi:10.1101/cshperspect.a011189
  359. Leone A, Di Gennaro E, Bruzzese F, Avallone A, Budillon A. New perspective for an old antidiabetic drug: metformin as anticancer agent. Cancer treatment and research. 2014;159:355-76. doi:10.1007/978-3-642-38007-5_21
  360. Saini N, Yang X. Metformin as an anti-cancer agent: actions and mechanisms targeting cancer stem cells. Acta biochimica et biophysica Sinica. Feb 1 2018;50(2):133-143. doi:10.1093/abbs/gmx106
  361. Wang YW, He SJ, Feng X, et al. Metformin: a review of its potential indications. Drug design, development and therapy. 2017;11:2421-2429. doi:10.2147/dddt.S141675
  362. Rojas LB, Gomes MB. Metformin: an old but still the best treatment for type 2 diabetes. Diabetol Metab Syndr. Feb 15 2013;5(1):6. doi:10.1186/1758-5996-5-6
  363. Aljofan M, Riethmacher D. Anticancer activity of metformin: a systematic review of the literature. Future Sci. 2019;5(8):FSO410.
  364. Saraei P, Asadi I, Kakar MA, Moradi-Kor N. The beneficial effects of metformin on cancer prevention and therapy: a comprehensive review of recent advances. Cancer management and research. 2019;11:3295-3313. doi:10.2147/cmar.S200059
  365. Decensi A, Puntoni M, Goodwin P, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer prevention research (Philadelphia, Pa). Nov 2010;3(11):1451-61. doi:10.1158/1940-6207.capr-10-0157
  366. Lei Y, Yi Y, Liu Y, et al. Metformin targets multiple signaling pathways in cancer. Chinese journal of cancer. Jan 26 2017;36(1):17. doi:10.1186/s40880-017-0184-9
  367. Gong J, Kelekar G, Shen J, Shen J, Kaur S, Mita M. The expanding role of metformin in cancer: an update on antitumor mechanisms and clinical development. Target Oncol. Aug 2016;11(4):447-67. doi:10.1007/s11523-016-0423-z
  368. Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. Oct 2001;108(8):1167-74. doi:10.1172/jci13505
  369. Hawley SA, Gadalla AE, Olsen GS, Hardie DG. The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism. Diabetes. Aug 2002;51(8):2420-5. doi:10.2337/diabetes.51.8.2420
  370. Wang Y, Xu W, Yan Z, et al. Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. Journal of experimental & clinical cancer research : CR. Mar 20 2018;37(1):63. doi:10.1186/s13046-018-0731-5
  371. Xie Y, Wang JL, Ji M, et al. Regulation of insulin-like growth factor signaling by metformin in endometrial cancer cells. Oncology letters. Nov 2014;8(5):1993-1999. doi:10.3892/ol.2014.2466
  372. Sarfstein R, Friedman Y, Attias-Geva Z, Fishman A, Bruchim I, Werner H. Metformin downregulates the insulin/IGF-I signaling pathway and inhibits different uterine serous carcinoma (USC) cells proliferation and migration in p53-dependent or -independent manners. PLoS One. 2013;8(4):e61537. doi:10.1371/journal.pone.0061537
  373. Zi FM, He JS, Li Y, et al. Metformin displays anti-myeloma activity and synergistic effect with dexamethasone in in vitro and in vivo xenograft models. Cancer letters. Jan 28 2015;356(2 Pt B):443-53. doi:10.1016/j.canlet.2014.09.050
  374. NCI. National Cancer Institute. Angiogenesis Inhibitors. Updated 4/2/2018. Accessed 9/24/2020, https://www.cancer.gov/about-cancer/treatment/types/immunotherapy/angiogenesis-inhibitors-fact-sheet
  375. Palumbo A, Anderson K. Multiple myeloma. Massachussetts Medical Society; 2011. p. 1046-1060.
  376. Mondello P, Cuzzocrea S, Navarra M, Mian M. Bone marrow micro-environment is a crucial player for myelomagenesis and disease progression. Impact Journals LLC; 2017. p. 20394-20409.
  377. Li WW, Li VW, Hutnik M, Chiou AS. Tumor angiogenesis as a target for dietary cancer prevention. Journal of oncology. 2012;2012:879623. doi:10.1155/2012/879623
  378. Khalid EB, Ayman ELMELK, Rahman H, Abdelkarim G, Najda A. Natural products against cancer angiogenesis. Springer Netherlands; 2016. p. 14513-14536.
  379. Ostrand-Rosenberg S, Fenselau C. Myeloid-Derived Suppressor Cells: Immune-Suppressive Cells That Impair Antitumor Immunity and Are Sculpted by Their Environment. J Immunol. Jan 15 2018;200(2):422-431. doi:10.4049/jimmunol.1701019
  380. Botta C, Gulla A, Correale P, Tagliaferri P, Tassone P. Myeloid-derived suppressor cells in multiple myeloma: pre-clinical research and translational opportunities. Frontiers in oncology. 2014;4:348. doi:10.3389/fonc.2014.00348
  381. Rodriguez PC, Quiceno DG, Zabaleta J, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. Aug 15 2004;64(16):5839-49. doi:10.1158/0008-5472.Can-04-0465
  382. Bingisser RM, Tilbrook PA, Holt PG, Kees UR. Macrophage-derived nitric oxide regulates T cell activation via reversible disruption of the Jak3/STAT5 signaling pathway. J Immunol. Jun 15 1998;160(12):5729-34.
  383. Mazzoni A, Bronte V, Visintin A, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. Jan 15 2002;168(2):689-95. doi:10.4049/jimmunol.168.2.689
  384. Gomez-Bougie P, Halliez M, Maiga S, et al. Curcumin induces cell death of the main molecular myeloma subtypes, particularly the poor prognosis subgroups. Cancer Biol Ther. 2015;16(1):60-5. doi:10.4161/15384047.2014.986997
  385. Valverde ME, Hernandez-Perez T, Paredes-Lopez O. Edible mushrooms: improving human health and promoting quality life. International journal of microbiology. 2015;2015:376387. doi:10.1155/2015/376387
  386. Brousseau M, Miller SC. The Effects ofEchinaceaRoot Extract in Plasmacytoma-Bearing Mice: Enhancement of Non-Specific Immunity. Journal of Herbs, Spices & Medicinal Plants. 2008;13(2):11-23. doi:10.1300/J044v13n02_02
  387. Klein B, Zhang XG, Jourdan M, et al. Interleukin-6 is the central tumor growth factor in vitro and in vivo in multiple myeloma. 1990. p. 193-201.
  388. Matthes T, Manfroi B, Huard B. Revisiting IL-6 antagonism in multiple myeloma. Elsevier Ireland Ltd; 2016. p. 1-4.
  389. Harmer D, Falank C, Reagan MR. Interleukin-6 Interweaves the Bone Marrow Microenvironment, Bone Loss, and Multiple Myeloma. Frontiers in endocrinology. 2018;9(JAN):788. doi:10.3389/fendo.2018.00788
  390. Sadahira K, Sagawa M, Nakazato T, et al. Gossypol induces apoptosis in multiple myeloma cells by inhibition of interleukin-6 signaling and Bcl-2/Mcl-1 pathway. International journal of oncology. Dec 2014;45(6):2278-86. doi:10.3892/ijo.2014.2652
  391. London RE. Multiple myeloma: report of a case showing unusual remission lasting two years following severe hepatitis. Ann Intern Med. Jul 1955;43(1):191-201. doi:10.7326/0003-4819-43-1-191
  392. Tian M, Huang H. The therapeutic effect of modified Huangqi Guizhi Wuwu Tang for multiple myeloma: An 18-year follow-up case report. Medicine. Dec 2017;96(49):e9074. doi:10.1097/MD.0000000000009074
  393. Hosgood HD, 3rd, Baris D, Zahm SH, Zheng T, Cross AJ. Diet and risk of multiple myeloma in Connecticut women. Cancer causes & control : CCC. 2007;18(10):1065-1076. doi:10.1007/s10552-007-9047-z
  394. Brown LM, Gridley G, Pottern LM, et al. Diet and nutrition as risk factors for multiple myeloma among blacks and whites in the United States. Cancer causes & control : CCC. 2001;12(2):117-125. doi:10.1023/a:1008937901586
  395. Caini S, Masala G, Gnagnarella P, et al. Food of animal origin and risk of non-Hodgkin lymphoma and multiple myeloma: A review of the literature and meta-analysis. Critical reviews in oncology/hematology. 2016;100:16-24. doi:10.1016/j.critrevonc.2016.02.011
  396. Lee DH, Fung TT, Tabung FK, et al. Pre-diagnosis dietary pattern and survival in patients with multiple myeloma. International journal of cancer. 2020:10.1002/ijc.32928. doi:10.1002/ijc.32928
  397. Lee DH, Fung TT, Tabung FK, et al. Dietary Pattern and Risk of Multiple Myeloma in Two Large Prospective US Cohort Studies. JNCI Cancer Spectr. 2019;3(2):pkz025-pkz025. doi:10.1093/jncics/pkz025
  398. Thordardottir M, Lindqvist EK, Lund SH, et al. Dietary intake is associated with risk of multiple myeloma and its precursor disease. PloS one. 2018;13(11):e0206047-e0206047. doi:10.1371/journal.pone.0206047
  399. Pariante CM. Neuroscience, mental health and the immune system: overcoming the brain-mind-body trichotomy. Epidemiol Psychiatr Sci. Apr 2016;25(2):101-5. doi:10.1017/S204579601500089X
  400. Overweight & Obesity. Accessed November 22, 2019, https://www.cdc.gov/obesity/adult/defining.html
  401. Tangen J-M. Antitumor and immunomodulating effects of the mushroom product AndosanTM, based on the Basidiomycetes mushroom Agaricus blazei Murill, with special focus on multiple myeloma. 2019;
  402. Kimura Y, Kido T, Takaku T, Sumiyoshi M, Baba K. Isolation of an anti-angiogenic substance from Agaricus blazei Murill: its antitumor and antimetastatic actions. Cancer science. Sep 2004;95(9):758-64. doi:10.1111/j.1349-7006.2004.tb03258.x
  403. Miyazawa K, Nishimaki J, Ohyashiki K, et al. Vitamin K2 therapy for myelodysplastic syndromes (MDS) and post-MDS acute myeloid leukemia: information through a questionnaire survey of multi-center pilot studies in Japan. Leukemia. Jun 2000;14(6):1156-7. doi:10.1038/sj.leu.2401790
  404. Nishimaki J, Miyazawa K, Yaguchi M, et al. Vitamin K2 induces apoptosis of a novel cell line established from a patient with myelodysplastic syndrome in blastic transformation. Leukemia. Sep 1999;13(9):1399-405. doi:10.1038/sj.leu.2401491
  405. Hetland G, Johnson E, Lyberg T, Bernardshaw S, Tryggestad AM, Grinde B. Effects of the medicinal mushroom Agaricus blazei Murill on immunity, infection and cancer. Scand J Immunol. Oct 2008;68(4):363-70. doi:10.1111/j.1365-3083.2008.02156.x
  406. Luiz R. Mechanism of anticlastogenicity of Agaricus blazei Murill mushroom organic extracts in wild type CHO (K1) and repair deficient (xrs5) cells by chromosome aberration and sister chromatid exchange assays. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis . 2003;528(1-2):75-79. doi:10.1016/s0027-5107(03)00098-8
  407. Bellini MF, Giacomini NL, Eira AF, Ribeiro LR, Mantovani MS. Anticlastogenic effect of aqueous extracts of Agaricus blazei on CHO-k1 cells, studying different developmental phases of the mushroom. Toxicol In Vitro. Aug 2003;17(4):465-9. doi:10.1016/s0887-2333(03)00043-2
  408. Tangen J-M. Antitumor and immunomodulating effects of the mushroom product Andosan TM , based on the Basidiomycota mushroom Agaricus blazei Murill, with special focus on multiple myeloma . 2019.
  409. Zhang S, Li W, Smith CJ, Musa H. Cereal-derived arabinoxylans as biological response modifiers: extraction, molecular features, and immune-stimulating properties. Critical reviews in food science and nutrition. 2015;55(8):1035-1052. doi:10.1080/10408398.2012.705188
  410. Cao L, Liu X, Qian T, et al. Antitumor and immunomodulatory activity of arabinoxylans: a major constituent of wheat bran. International journal of biological macromolecules. 2011;48(1):160-164. doi:10.1016/j.ijbiomac.2010.10.014
  411. Badr El-Din NK, Noaman E, Ghoneum M. In vivo tumor inhibitory effects of nutritional rice bran supplement MGN-3/Biobran on Ehrlich carcinoma-bearing mice. Nutrition and cancer. 2008;60(2):235-244. doi:10.1080/01635580701627285
  412. Badr El-Din NK, Abdel Fattah SM, Pan D, Tolentino L, Ghoneum M. Chemopreventive Activity of MGN-3/Biobran Against Chemical Induction of Glandular Stomach Carcinogenesis in Rats and Its Apoptotic Effect in Gastric Cancer Cells. Integrative cancer therapies. 2016;15(4):NP26-NP34. doi:10.1177/1534735416642287
  413. Savitha Prashanth MR, Shruthi RR, Muralikrishna G. Immunomodulatory activity of purified arabinoxylans from finger millet (Eleusine coracana, v. Indaf 15) bran. Journal of food science and technology. 2015;52(9):6049-6054. doi:10.1007/s13197-014-1664-4
  414. Ghoneum M, Matsuura M. Augmentation of macrophage phagocytosis by modified arabinoxylan rice bran (MGN-3/biobran). Int J Immunopathol Pharmacol. Sep-Dec 2004;17(3):283-292. doi:10.1177/039463200401700308
  415. Ogawa K, Takeuchi M, Nakamura N. Immunological effects of partially hydrolyzed arabinoxylan from corn husk in mice. Bioscience, biotechnology, and biochemistry. 2005;69(1):19-25. doi:10.1271/bbb.69.19
  416. Ghoneum M, Abedi S. Enhancement of natural killer cell activity of aged mice by modified arabinoxylan rice bran (MGN-3/Biobran). J Pharm Pharmacol. 2004;56(12):1581-1588. doi:10.1211/0022357044922
  417. Elsaid AF, Shaheen M, Ghoneum M. Biobran/MGN-3, an arabinoxylan rice bran, enhances NK cell activity in geriatric subjects: A randomized, double-blind, placebo-controlled clinical trial. Experimental and therapeutic medicine. 2018;15(3):2313-2320. doi:10.3892/etm.2018.5713
  418. Choi J-Y, Paik D-J, Kwon DY, Park Y. Dietary supplementation with rice bran fermented with Lentinus edodes increases interferon-γ activity without causing adverse effects: a randomized, double-blind, placebo-controlled, parallel-group study. Nutrition journal. 2014;13:35-35. doi:10.1186/1475-2891-13-35
  419. Ooi SL, McMullen D, Golombick T, Nut D, Pak SC. Evidence-Based Review of BioBran/MGN-3 Arabinoxylan Compound as a Complementary Therapy for Conventional Cancer Treatment. Integrative cancer therapies. 2018;17(2):165-178. doi:10.1177/1534735417735379
  420. Ghoneum M, Gollapudi S. Synergistic apoptotic effect of arabinoxylan rice bran (MGN-3/Biobran) and curcumin (turmeric) on human multiple myeloma cell line U266 in vitro. Neoplasma. 2011;58(2):118-123. doi:10.4149/neo_2011_02_118
  421. Golombick T, Diamond TH, Manoharan A, Ramakrishna R. Addition of Rice Bran Arabinoxylan to Curcumin Therapy May Be of Benefit to Patients With Early-Stage B-Cell Lymphoid Malignancies (Monoclonal Gammopathy of Undetermined Significance, Smoldering Multiple Myeloma, or Stage 0/1 Chronic Lymphocytic Leukemia): A Preliminary Clinical Study. Integrative cancer therapies. Jun 2016;15(2):183-9. doi:10.1177/1534735416635742
  422. Bharti AC, Donato N, Singh S, Aggarwal BB. Curcumin down-regulates the constitutive activation of nuclear factor kappa B and IkB-a kinase in human multiple myeloma cells leading to suppression of proliferation and induction of apoptosis. Blood. 2003;101(3):1052-1062.
  423. Bharti AC, Takada Y, Aggarwal BB. Curcumin (diferuloylmethane) inhibits receptor activator of NF-kappa B ligand-induced NF-kappa B activation in osteoclast precursors and suppresses osteoclastogenesis. J Immunol . May 15 2004;172(10):5940-7. doi:10.4049/jimmunol.172.10.5940
  424. Kudo C, Yamakoshi H, Sato A, et al. Novel curcumin analogs, GO-Y030 and GO-Y078, are multi-targeted agents with enhanced abilities for multiple myeloma. Anticancer research. Nov 2011;31(11):3719-26.
  425. Mujtaba T, Kanwar J, Wan SB, Chan TH, Dou QP. Sensitizing human multiple myeloma cells to the proteasome inhibitor bortezomib by novel curcumin analogs. Int J Mol Med. Jan 2012;29(1):102-6. doi:10.3892/ijmm.2011.814
  426. Wan SB, Yang H, Zhou Z, et al. Evaluation of curcumin acetates and amino acid conjugates as proteasome inhibitors. International journal of molecular medicine. 2010;26(4):447-455.
  427. Bharti AC, Shishodia S, Reuben JM, et al. Nuclear factor-kappaB and STAT3 are constitutively active in CD138+ cells derived from multiple myeloma patients, and suppression of these transcription factors leads to apoptosis. Blood. Apr 15 2004;103(8):3175-84. doi:10.1182/blood-2003-06-2151
  428. Reuter S, Eifes S, Dicato M, Aggarwal BB, Diederich M. Modulation of anti-apoptotic and survival pathways by curcumin as a strategy to induce apoptosis in cancer cells. Biochemical pharmacology. Dec 1 2008;76(11):1340-51. doi:10.1016/j.bcp.2008.07.031
  429. Bharti AC, Donato N, Aggarwal BB. Curcumin (diferuloylmethane) inhibits constitutive and IL-6-inducible STAT3 phosphorylation in human multiple myeloma cells. J Immunol. Oct 1 2003;171(7):3863-71. doi:10.4049/jimmunol.171.7.3863
  430. Golombick T, Diamond TH, Badmaev V, Manoharan A, Ramakrishna R. The potential role of curcumin in patients with monoclonal gammopathy of undefined significance--its effect on paraproteinemia and the urinary N-telopeptide of type I collagen bone turnover marker. Clin Cancer Res. Sep 15 2009;15(18):5917-22. doi:10.1158/1078-0432.CCR-08-2217
  431. Golombick T, Diamond TH, Manoharan A, Ramakrishna R. Monoclonal gammopathy of undetermined significance, smoldering multiple myeloma, and curcumin: a randomized, double-blind placebo-controlled cross-over 4g study and an open-label 8g extension study. Am J Hematol. May 2012;87(5):455-60. doi:10.1002/ajh.23159
  432. Vadhan-Raj S, Weber D, Wang M. Curcumin downregulates NF-B and related genes in patients with multiple myeloma: results of a phase I/II study. Blood. 2007;110(11):1177-1177.
  433. Zaidi A, Lai M, Cavenagh J. Long-term stabilisation of myeloma with curcumin. BMJ case reports. Apr 16 2017;2017doi:10.1136/bcr-2016-218148
  434. Allegra A, Speciale A, Molonia MS, et al. Curcumin ameliorates the in vitro efficacy of carfilzomib in human multiple myeloma U266 cells targeting p53 and NF-kappaB pathways. Toxicol In Vitro. Mar 2018;47:186-194. doi:10.1016/j.tiv.2017.12.001
  435. Sung B, Kunnumakkara AB, Sethi G, Anand P, Guha S, Aggarwal BB. Curcumin circumvents chemoresistance in vitro and potentiates the effect of thalidomide and bortezomib against human multiple myeloma in nude mice model. Molecular cancer therapeutics. Apr 2009;8(4):959-70. doi:10.1158/1535-7163.Mct-08-0905
  436. Pangestuti R, Arifin Z. Medicinal and health benefit effects of functional sea cucumbers. Journal of traditional and complementary medicine. 2017;8(3):341-351. doi:10.1016/j.jtcme.2017.06.007
  437. Chari A, Mazumder A, Lau K, Catamero D, Galitzeck Z, Jagannath S. A phase II trial of TBL-12 sea cucumber extract in patients with untreated asymptomatic myeloma. Br J Haematol. Jan 2018;180(2):296-298. doi:10.1111/bjh.14314
  438. Mohamed AS, Mahmoud SA, Soliman AM, Fahmy SR. Antitumor activity of saponin isolated from the sea cucumber, holothuria arenicola against ehrlich ascites carcinoma cells in swiss albino mice. Nat Prod Res. Jul 25 2019:1-5. doi:10.1080/14786419.2019.1644633
  439. Janakiram NB, Mohammed A, Zhang Y, et al. Chemopreventive effects of Frondanol A5, a Cucumaria frondosa extract, against rat colon carcinogenesis and inhibition of human colon cancer cell growth. Cancer prevention research (Philadelphia, Pa). Jan 2010;3(1):82-91. doi:10.1158/1940-6207.Capr-09-0112
  440. Yuan L, Huang X, Zhou K, et al. Sea cucumber extract TBL-12 inhibits the proliferation, migration, and invasion of human prostate cancer cells through the p38 mitogen-activated protein kinase and intrinsic caspase apoptosis pathway. Prostate. 2019;79(8):826-839. doi:10.1002/pros.23788
  441. Janakiram NB, Mohammed A, Bryant T, et al. Improved innate immune responses by Frondanol A5, a sea cucumber extract, prevent intestinal tumorigenesis. Cancer prevention research (Philadelphia, Pa). 2015;8(4):327-337. doi:10.1158/1940-6207.CAPR-14-0380
  442. Surayot U, Lee S, You S. Effects of sulfated fucan from the sea cucumber Stichopus japonicus on natural killer cell activation and cytotoxicity. International journal of biological macromolecules. 2018;108:177-184. doi:10.1016/j.ijbiomac.2017.11.102
  443. Song Y, Jin S-J, Cui L-H, Ji X-J, Yang F-G. Immunomodulatory effect of Stichopus japonicus acid mucopolysaccharide on experimental hepatocellular carcinoma in rats. Molecules (Basel, Switzerland). 2013;18(6):7179-7193. doi:10.3390/molecules18067179
  444. Monmai C, Park SH, You S, Park WJ. Immuno-enhancement effect of polysaccharide extracted from Stichopus japonicus on cyclophosphamide-induced immunosuppression mice. Food Sci Biotechnol. 2017;27(2):565-573. doi:10.1007/s10068-017-0248-2
  445. Wang H, Xu L, Yu M, et al. Glycosaminoglycan from Apostichopus japonicus induces immunomodulatory activity in cyclophosphamide-treated mice and in macrophages. International journal of biological macromolecules. 2019;130:229-237. doi:10.1016/j.ijbiomac.2019.02.093
  446. Bosseboeuf A, Allain-Maillet S, Mennesson N, et al. Pro-inflammatory State in Monoclonal Gammopathy of Undetermined Significance and in Multiple Myeloma Is Characterized by Low Sialylation of Pathogen-Specific and Other Monoclonal Immunoglobulins. Frontiers in immunology. 2017;8:1347-1347. doi:10.3389/fimmu.2017.01347
  447. Saad DY, Soliman MM, Mohamed AA, Youssef GB. Protective effects of sea cucumber (Holothuria atra) extract on testicular dysfunction induced by immune suppressant drugs in Wistar rats. Andrologia. 2018;50(6):e13017-e13017. doi:10.1111/and.13017
  448. Li S, Jiang W, Hu S, et al. Fucosylated chondroitin sulphate from Cusumaria frondosa mitigates hepatic endoplasmic reticulum stress and inflammation in insulin resistant mice. Food & function. 2015;6(5):1547-1556. doi:10.1039/c4fo01153h
  449. Kariya Y, Mulloy B, Imai K, et al. Isolation and partial characterization of fucan sulfates from the body wall of sea cucumber Stichopus japonicus and their ability to inhibit osteoclastogenesis. Carbohydr Res. 2004;339(7):1339-1346. doi:10.1016/j.carres.2004.02.025
  450. Baharara J, Amini E, Kerachian MA, Soltani M. The osteogenic differentiation stimulating activity of Sea cucumber methanolic crude extraction on rat bone marrow mesenchymal stem cells. Iranian journal of basic medical sciences. 2014;17(8):626-631.
  451. Freitas RDS, Campos MM. Protective Effects of Omega-3 Fatty Acids in Cancer-Related Complications. Nutrients. 2019;11(5):945. doi:10.3390/nu11050945
  452. Engeset D, Braaten T, Teucher B, et al. Fish consumption and mortality in the European Prospective Investigation into Cancer and Nutrition cohort. European journal of epidemiology. Jan 2015;30(1):57-70. doi:10.1007/s10654-014-9966-4
  453. Siddiqui RA, Harvey KA, Xu Z, Bammerlin EM, Walker C, Altenburg JD. Docosahexaenoic acid: a natural powerful adjuvant that improves efficacy for anticancer treatment with no adverse effects. BioFactors (Oxford, England). Nov-Dec 2011;37(6):399-412. doi:10.1002/biof.181
  454. Abdi J, Garssen J, Faber J, Redegeld FA. Omega-3 fatty acids, EPA and DHA induce apoptosis and enhance drug sensitivity in multiple myeloma cells but not in normal peripheral mononuclear cells. The Journal of Nutritional Biochemistry. 2014/12/01/ 2014;25(12):1254-1262. doi: https://doi.org/10.1016/j.jnutbio.2014.06.013
  455. Dai X, Li M, Geng F. Omega-3 Polyunsaturated Fatty Acids Eicosapentaenoic Acid and Docosahexaenoic Acid Enhance Dexamethasone Sensitivity in Multiple Myeloma Cells by the p53/miR-34a/Bcl-2 Axis. Biochemistry Biokhimiia. Jul 2017;82(7):826-833. doi:10.1134/s0006297917070082
  456. Bayram I, Erbey F, Celik N, Nelson JL, Tanyeli A. The use of a protein and energy dense eicosapentaenoic acid containing supplement for malignancy-related weight loss in children. Pediatr Blood Cancer. May 2009;52(5):571-4. doi:10.1002/pbc.21852
  457. Chagas TR, Borges DS, de Oliveira PF, et al. Oral fish oil positively influences nutritional-inflammatory risk in patients with haematological malignancies during chemotherapy with an impact on long-term survival: a randomised clinical trial. J Hum Nutr Diet. Dec 2017;30(6):681-692. doi:10.1111/jhn.12471
  458. Burns CP, Halabi S, Clamon G, et al. Phase II study of high-dose fish oil capsules for patients with cancer-related cachexia. Cancer. 2004;101(2):370-378. doi:10.1002/cncr.20362
  459. Tuli HS, Tuorkey MJ, Thakral F, et al. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Frontiers in pharmacology. 2019;10:1336. doi:10.3389/fphar.2019.01336
  460. Xie J, Wang J, Zhu B. Genistein inhibits the proliferation of human multiple myeloma cells through suppression of nuclear factor-κB and upregulation of microRNA-29b. Molecular medicine reports. Feb 2016;13(2):1627-32. doi:10.3892/mmr.2015.4740
  461. He H, Chen L, Zhai M, Chen JZ. Genistein down-regulates the constitutive activation of nuclear factor-kappaB in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Phytother Res. Jun 2009;23(6):868-73. doi:10.1002/ptr.2715
  462. Landis-Piwowar KR, Milacic V, Chen D, et al. The proteasome as a potential target for novel anticancer drugs and chemosensitizers. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy . Dec 2006;9(6):263-73. doi:10.1016/j.drup.2006.11.001
  463. Pintova S, Dharmupari S, Moshier E, Zubizarreta N, Ang C, Holcombe RF. Genistein combined with FOLFOX or FOLFOX-Bevacizumab for the treatment of metastatic colorectal cancer: phase I/II pilot study. Cancer chemotherapy and pharmacology. Sep 2019;84(3):591-598. doi:10.1007/s00280-019-03886-3
  464. Wang Q, Wang Y, Ji Z, et al. Risk factors for multiple myeloma: A hospital-based case–control study in Northwest China. Cancer epidemiology. 2012/10/01/ 2012;36(5):439-444. doi: https://doi.org/10.1016/j.canep.2012.05.002
  465. Löhr JM, Karimi M, Omazic B, et al. A phase I dose escalation trial of AXP107-11, a novel multi-component crystalline form of genistein, in combination with gemcitabine in chemotherapy-naive patients with unresectable pancreatic cancer. Pancreatology : official journal of the International Association of Pancreatology (IAP) [et al] . Jul-Aug 2016;16(4):640-5. doi:10.1016/j.pan.2016.05.002
  466. Ullmann U, Metzner J, Frank T, Cohn W, Riegger C. Safety, tolerability, and pharmacokinetics of single ascending doses of synthetic genistein (Bonistein) in healthy volunteers. Adv Ther. Jan-Feb 2005;22(1):65-78. doi:10.1007/bf02850186
  467. Zhang Y, Song TT, Cunnick JE, Murphy PA, Hendrich S. Daidzein and genistein glucuronides in vitro are weakly estrogenic and activate human natural killer cells at nutritionally relevant concentrations. J Nutr. Feb 1999;129(2):399-405. doi:10.1093/jn/129.2.399
  468. Tomas-Barberan FA, Andres-Lacueva C. Polyphenols and health: current state and progress. ACS Publications; 2012.
  469. Corcoran MP, McKay DL, Blumberg JB. Flavonoid basics: chemistry, sources, mechanisms of action, and safety. Journal of Nutrition in Gerontology and Geriatrics. 2012;31(3):176-189.
  470. Sharifi-Rad M, Pezzani R, Redaelli M, et al. Preclinical Pharmacological Activities of Epigallocatechin-3-gallate in Signaling Pathways: An Update on Cancer. Molecules (Basel, Switzerland). 2020;25(3):467. doi:10.3390/molecules25030467
  471. Lambert JD, Elias RJ. The antioxidant and pro-oxidant activities of green tea polyphenols: a role in cancer prevention. Archives of biochemistry and biophysics. Sep 1 2010;501(1):65-72. doi:10.1016/j.abb.2010.06.013
  472. Ahmad N, Mukhtar H. Green tea polyphenols and cancer: biologic mechanisms and practical implications. Nutrition reviews. Mar 1999;57(3):78-83. doi:10.1111/j.1753-4887.1999.tb06927.x
  473. Wang Q, Li J, Gu J, et al. Potentiation of (−)-epigallocatechin-3-gallate-induced apoptosis by bortezomib in multiple myeloma cells. Acta biochimica et biophysica Sinica. 2009;41(12):1018-1026. doi:10.1093/abbs/gmp094
  474. Shanafelt TD, Call TG, Zent CS, et al. Phase I trial of daily oral Polyphenon E in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology . 2009;27(23):3808-3814. doi:10.1200/JCO.2008.21.1284
  475. Shanafelt TD, Call TG, Zent CS, et al. Phase 2 trial of daily, oral Polyphenon E in patients with asymptomatic, Rai stage 0 to II chronic lymphocytic leukemia. Cancer. 2013;119(2):363-370. doi:10.1002/cncr.27719
  476. Catlett-Falcone R, Landowski TH, Oshiro MM, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. Jan 1999;10(1):105-15. doi:10.1016/s1074-7613(00)80011-4
  477. Wu T, Shu T, Kang L, et al. Icaritin, a novel plant-derived osteoinductive agent, enhances the osteogenic differentiation of human bone marrow- and human adipose tissue-derived mesenchymal stem cells. Int J Mol Med . Apr 2017;39(4):984-992. doi:10.3892/ijmm.2017.2906
  478. Mathema VB, Koh YS, Thakuri BC, Sillanpaa M. Parthenolide, a sesquiterpene lactone, expresses multiple anti-cancer and anti-inflammatory activities. Inflammation. Apr 2012;35(2):560-5. doi:10.1007/s10753-011-9346-0
  479. Gunn EJ, Williams JT, Huynh DT, et al. The natural products parthenolide and andrographolide exhibit anti-cancer stem cell activity in multiple myeloma. Leuk Lymphoma. Jun 2011;52(6):1085-97. doi:10.3109/10428194.2011.555891
  480. Kong FC, Zhang JQ, Zeng C, et al. Inhibitory effects of parthenolide on the activity of NF-kappaB in multiple myeloma via targeting TRAF6. J Huazhong Univ Sci Technolog Med Sci. Jun 2015;35(3):343-349. doi:10.1007/s11596-015-1435-0
  481. Wang W, Adachi M, Kawamura R, et al. Parthenolide-induced apoptosis in multiple myeloma cells involves reactive oxygen species generation and cell sensitivity depends on catalase activity. Apoptosis. Dec 2006;11(12):2225-35. doi:10.1007/s10495-006-0287-2
  482. Wen J, You KR, Lee SY, Song CH, Kim DG. Oxidative stress-mediated apoptosis. The anticancer effect of the sesquiterpene lactone parthenolide. J Biol Chem. Oct 11 2002;277(41):38954-64. doi:10.1074/jbc.M203842200
  483. Beuth J. Proteolytic enzyme therapy in evidence-based complementary oncology: Fact or fiction? 2008. p. 311-316.
  484. Leipner J, Saller R. Systemic enzyme therapy in oncology: effect and mode of action. Drugs. Apr 2000;59(4):769-80. doi:10.2165/00003495-200059040-00004
  485. Sakalova A, Bock PR, Dedik L, et al. Retrolective cohort study of an additive therapy with an oral enzyme preparation in patients with multiple myeloma. Cancer chemotherapy and pharmacology. Jul 2001;47 Suppl(7):S38-44. doi:10.1007/s002800170008
  486. Sharma A, Tripathi M, Satyam A, Kumar L. Study of antioxidant levels in patients with multiple myeloma. Leuk Lymphoma. May 2009;50(5):809-15. doi:10.1080/10428190902802323
  487. Shenoy N, Creagan E, Witzig T, Levine M. Ascorbic Acid in Cancer Treatment: Let the Phoenix Fly. Cancer cell. Nov 12 2018;34(5):700-706. doi:10.1016/j.ccell.2018.07.014
  488. Parrow NL, Leshin JA, Levine M. Parenteral ascorbate as a cancer therapeutic: a reassessment based on pharmacokinetics. Antioxid Redox Signal. Dec 10 2013;19(17):2141-56. doi:10.1089/ars.2013.5372
  489. Lee B, Oh SW, Myung SK. Efficacy of Vitamin C Supplements in Prevention of Cancer: A Meta-Analysis of Randomized Controlled Trials. Korean journal of family medicine. Nov 2015;36(6):278-85. doi:10.4082/kjfm.2015.36.6.278
  490. ClinicalTrials.gov. Search: pharmacologic ascorbate and cancer. U.S. National Library of Medicine. Accessed 9/24/2020, https://clinicaltrials.gov/ct2/results?cond=Cancer &term=pharmacologic+ascorbate
  491. Lappe JM. The Role of Vitamin D in Human Health: A Paradigm Shift. Journal of evidence-based complementary & alternative medicine . 2011;16(1):58-72. doi:10.1177/1533210110392952
  492. Vaughan-Shaw PG, O'Sullivan F, Farrington SM, et al. The impact of vitamin D pathway genetic variation and circulating 25-hydroxyvitamin D on cancer outcome: systematic review and meta-analysis. British journal of cancer. Apr 11 2017;116(8):1092-1110. doi:10.1038/bjc.2017.44
  493. Badros A, Goloubeva O, Terpos E, Milliron T, Baer MR, Streeten E. Prevalence and significance of vitamin D deficiency in multiple myeloma patients. 2008. p. 492-494.
  494. Nath K, Ganeshalingam V, Ewart B, et al. A retrospective analysis of the prevalence and clinical outcomes of vitamin D deficiency in myeloma patients in tropical Australia. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer . Jun 21 2019;doi:10.1007/s00520-019-04942-7
  495. Maier GS, Horas K, Kurth AA, Lazovic D, Seeger JB, Maus U. Prevalence of Vitamin D Deficiency in Patients with Bone Metastases and Multiple Myeloma. Anticancer research. Nov 2015;35(11):6281-5.
  496. Vitamin D — Health Professional Fact Sheet.
  497. National Institutes and Health (NIH). Vitamin D: Fact Sheet for Health Professionals. Updated 8/7/2019. Accessed 1/27/2020, https://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional/
  498. Lauter B, Schmidt-Wolf IG. Prevalence, Supplementation, and Impact of Vitamin D Deficiency in Multiple Myeloma Patients. Cancer investigation. 2015;33(10):505-9. doi:10.3109/07357907.2015.1081690
  499. Ng AC, Kumar SK, Rajkumar SV, Drake MT. Impact of vitamin D deficiency on the clinical presentation and prognosis of patients with newly diagnosed multiple myeloma. Am J Hematol. Jul 2009;84(7):397-400. doi:10.1002/ajh.21412
  500. Yokus O, Hilmi Dogu M, Aydinli F, Altindal S. Incidence of Vitamin D Deficiency in Multiple Myeloma Patients and The Relationship Between Chemotherapies, Autologous Stem Cell Transplantation, Bone Density and Fractures. Clinical Lymphoma Myeloma and Leukemia. 2015;15:S50-S50. doi:10.1016/j.clml.2015.07.103
  501. Burwick N. Vitamin D and plasma cell dyscrasias: reviewing the significance. Springer Verlag; 2017. p. 1271-1277.
  502. V Rakhee SACRZSLDDH. Low pre-transplant vitamin D levels predict an inferior survival in patients with multiple myeloma undergoing an autologous stem cell transplant. Blood. 2016;128(22):5655-5655.
  503. Diamond T, Golombick T, Manoharan A. Vitamin D status may effect the skeletal complications of multiple myeloma. Am J Hematol. Apr 2010;85(4):302-3. doi:10.1002/ajh.21619
  504. Kaiser MF, Heider U, Mieth M, Zang C, von Metzler I, Sezer O. The proteasome inhibitor bortezomib stimulates osteoblastic differentiation of human osteoblast precursors via upregulation of vitamin D receptor signalling. Eur J Haematol. Apr 2013;90(4):263-72. doi:10.1111/ejh.12069
  505. Wang J, Udd KA, Vidisheva A, et al. Low serum vitamin D occurs commonly among multiple myeloma patients treated with bortezomib and/or thalidomide and is associated with severe neuropathy. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer . Jul 2016;24(7):3105-10. doi:10.1007/s00520-016-3126-1
  506. N Ravenborg KUABFCJRB. Vitamin D levels are frequently below normal in multiple myeloma patients and are infrequently assessed by their treating physicians. Blood. 2014;124(21):5769-5769.
  507. Borset M, Sundan A, Waage A, Standal T. Why do myeloma patients have bone disease? A historical perspective. Blood reviews. Nov 29 2019;(xxxx):100646. doi:10.1016/j.blre.2019.100646
  508. Yamaguchi M, Weitzmann MN. Vitamin K2 stimulates osteoblastogenesis and suppresses osteoclastogenesis by suppressing NF-kappaB activation. Int J Mol Med. Jan 2011;27(1):3-14. doi:10.3892/ijmm.2010.562
  509. Sung B, Oyajobi B, Aggarwal BB. Plumbagin inhibits osteoclastogenesis and reduces human breast cancer-induced osteolytic bone metastasis in mice through suppression of RANKL signaling. Molecular cancer therapeutics. Feb 2012;11(2):350-9. doi:10.1158/1535-7163.MCT-11-0731