A user-friendly, frequently updated reference guide that aligns with international guidelines and protocols.
Plasma Cell Neoplasms
This category includes:
Plasma Cell Myeloma
Smouldering (asymptomatic) myeloma
Non-secretory myeloma
Plasma cell leukemia
Plasmacytoma
Solitary Plasmacytoma of Bone
Extramedullary Plasmacytoma
Plasma cell neoplasms with associated paraneoplastic syndrome
POEMS Syndrome
TEMPI Syndrome
AESOP Syndrome
Monoclonal gammopathy of unknown significance
Monoclonal gammopathy of clinical significance
Heavy chain diseases
Idiopathic Capillary Leak Syndrome
Plasma cell Myeloma
(Multiple myeloma)
Introduction:
It is a bone marrow based, multifocal plasma cell neoplasm characterized by elevated serum monoclonal proteins, skeletal destruction, hypercalcemia and anemia.
Epidemiology:
It accounts for 1-2% of all malignancies
In US it is most common lymphoid malignancy in blacks and second most common in whites.
Men are commonly affected than women. (M:F- 1.4:1)
It represents 15% of all hematological malignancies
Median age- 70 years
3-4 cases per 1lac population
Etiology: Exact cause is not known
Environmental agents – pesticides, petroleum products, asbestos, rubber, plastic and wood products, dioxins, solvents, cleaners etc.
High dose irradiation (100 cGy)
Chronic inflammation (chronic antigenic stimulation) such as osteomyelitis, rheumatoid arthritis, HHV-8 infections, EBV, HIV, hepatitis virus infection, infection with “Stealth adapted” viruses such as mutated CMV.
It is increasingly seen in cosmetologists, farmers and laxative users.
Pathogenesis:
2 hit hypothesis
1st hit – Initial antigenic stimulation which leads to multiple benign clones
2nd hit – Mutational change which leads to malignant transformation
Bone marrow microenvironment in myeloma
Homing in of plasma cells in BM involves
Selective adhesion to BM endothelial cells with expression of CXCR4, CXCR3, CCR1, CCR2, CCR5
Trans endothelial migration
Adhesion to stromal cells through the production of stromal derived factor1 (SDF1) and insulin like GF1 (ILGF1)
Adhesion of myeloma cells to BM stromal cells through a4 b1 integrin / VCAM1 induces production of several cytokines (mostly by NF-KB pathway)
RANKL- Binds to RANK on osteoclasts leading to enhanced differentiation, proliferation and survival of osteoclasts. (normally osteoprotegerin binds to RANK, but in myeloma its level is decreased)
Some cytokines are directly produced by plasma cells
VEGF and Beta FGF cause enhance angiogenesis
MIP 1alfa (RANTES family) activates osteoclasts
dickkop -1 protein which leads to inhibition of Wnt- mediated osteoblast differentiation and reduced bone formation
Growth signals for myeloma cells are mediated through following pathways
PITK/AKT
STAT3
RAS/MAPK
NFk-beta
Myeloma cells may secrete immunoglobulins (Paraproteins)- They get deposited in kidney and cause renal damage
Single heavy and light chain – known as monoclonal protein / paraprotein
Only light chains – In urine they are known as Bence Jones proteins
IgG – 60%
IgA – 20%
Light chain only - 15-20% (Risk of renal disease is high)
IgD or IgE – 1%
None (Non secreting) – 1% (Normal Serum, urine immunofixation electrophoresis and normal FLC ratio. M protein can be detected by IHC)
Oligosecretory- Serum M Protein <1g/dL and Urine M Protein <200mg/24hrs
Clinical Features:
20% are asymptomatic
Weight loss, malaise and fatigue
Bone pain
Especially in ribs and back (Due to osteoporosis, lytic lesions and pathological fractures)
Even with adequate therapy, lytic lesions remain and new bone formation does not occur.
Hypercalcemia
It may be due to hyperactivity of osteoclasts and suppression of osteoblasts.
May cause anorexia, vomiting, diarrhea / constipation, polydipsia, polyuria, muscle weakness, shortening of QT etc.
Repeated infection (URTI, LRTI, UTI):
Occurs due to
Decreased protective immunoglobulins
Neutropenia
Hypoventilation due to pathological fractures
Unproductive Th2 cell response due to cytokines such as IL6, TGF beta, IL 10 and beta 2 microglobulin
Side effect of antimyeloma treatment (Cytotoxic drugs and glucocorticoids)
Infection with encapsulated organisms such as streptococcus pneumonia, Haemophilus influenza etc are common
Anemia:
Seen in 75% of patients.
Occur due to
Myelophthiasis – Replacement of marrow by tumor cells
Tumor production of inhibitory factors and auto antibodies (IL6 stimulates Hepcidin release)
Expanded plasma volume due to increased paraproteins
Renal failure- Decreased EPO production
Chronic infection
Bleeding
Nutritional deficiency
Renal failure
Seen in 25% of patients at presentation
30-50% patients have some degree of renal impairment and 10% need hemodialysis
It occurs due to
Light chain nephropathy: Tubular damage by excretion of light chains (Lambda chains are more nephrotoxic)
Cast nephropathy: Tubular casts block distal convoluted tubule leading to interstitial nephritis
Hypercalcemia resulting in nephrocalcinosis
Glomerular deposition of amyloid leading to nephrotic syndrome
Hyperuricemia resulting in urate nephropathy
Recurrent urinary infections
Infiltration of kidney by myeloma cells
Use of NSAIDs for pain control
Nephrotoxic antibiotics and imaging contrast agents and use of bisphosphonates.
Hyperviscosity
May present as headache, confusion, breathlessness, visual disturbance and bleeding.
Normal relative serum viscosity is 1.8 mpascals, and symptoms occur at 5-6 mpascals
Common with IgA and IgG3 type paraproteinemia
Fundoscopy: Retinal vein distension, hemorrhage and papilledema
Amyloid deposition (AL type amyloidosis)
May manifest as – Carpal tunnel syndrome, macroglossia, nephrotic syndrome, cardiac failure, peripheral neuropathy, subcutaneous nodules, hepatosplenomegaly etc.
Bleeding manifestations due to
Paraprotein causes abnormal platelet function
Thrombocytopenia in advanced disease
Paraprotein may act as anticoagulant by inhibition of fibrin monomer polymerization.
Hyperviscosity
Perivascularamyloidosis
Acquired states such as factor 10 deficiency
Tumor formation especially in ribs
They are firm, tender and are associated with egg shell crackling
Extramedullaryplasmacytoma
Seen in 7% of patients at diagnosis
Cord compression- Sensory loss, paresthesia, limb weakness, walking difficulty, sphincter disturbance leading to incontinence
Neuropathy due to
Regional myeloma cell growth compressing spinal cord or cranial nerves
Perineural or perivasa nervosum amyloid deposition
Metabolic causes related to hypercalcemia or hyperviscosity
IgM related neuropathy- Antibodies to myeline associated globulin
Thrombosis : Due to
Hyperviscosity
Defective fibrin structure and fibrinolysis because of increased IgG levels
Increased acquired protein C resistance
Increased pro-inflammatory cytokines such as IL6
Sometimes lupus anticoagulants
Thalidomide, lenalidomide and EPO use
Investigations:
Hemogram:
Normocyticnormochromic anemia
Marked red cell rouleaux formation
Abnormally blue stained background of blood film due to presence of paraproteins
Moderate leucopenia
Few atypical plasma cells may be present
Sometimes there can be leucoerythroblastic blood picture
Thrombocytopenia is rare and late, as IL6 stimulates megakaryopoiesis
Thrombocytosis may indicate hyposplenism because of amyloidosis of spleen.
BM aspiration
If possible, should be done from radiologically abnormal site
Hypercellular marrow
Erythropoiesis and myelopoiesis are generally suppressed
Megakaryocytes are generally adequate
Plasma cells are increased in number
>10% Clonal plasma cells is a major criteria for diagnosis (In aspirate/ biopsy). Clonality can be assessed by flow cytometry/ IHC.
Diagnosis of MM can be confirmed with less than 10% plasma cells in BM, if other diagnostic criteria are fulfilled and if there is histopathological confirmation of a soft tissue or bony plasmacytoma. If disease is patchy, some patients need BM aspiration from 2-3 different sites/ imaging guided biopsy.
They can be mature/ immature/ pleomorphic/ anaplastic (Greipp plasma cell grading system)
Mature type- Dense chromatin clumping (“Spoke wheel” or “Clock face” chromatin), nucleus <8microns, Nucleolus- <1 micron, Cytoplasm well developed, nucleus is eccentric with prominent perinuclearhof .
Immature type- Diffuse chromatin pattern, nucleus >10 microns, Nucleolus >2micron, abundant cytoplasm, nucleus is eccentric with hof
Plasmablastic type- Very high N:C ratio, scanty cytoplasm, central immature large nucleus with reticular chromatin pattern, prominent nucleolus, little/ no hof
Intermediate type- Not meeting criteria of other types
Nuclear immaturity and pleomorphism are rarely seen in reactive plasma cells. Hence they are reliable indicators of neoplastic plasma cells.
Morphologically distinctive varieties may be seen due to crystallized cytoplasmic immunoglobulin in endoplasmic reticulum
Russel bodies- Cherry red refractile round bodies.
Flame cells- Vermilion staining glycogen rich IgA
Goucher like cells (Thesaurocytes)- Over stuffed fibrils.
Dutcher bodies- Nuclear accumulation of immunoglobulins
Number counted on morphology is considered gold standard, as on flow, there can be loss of plasma cells during processing or they may nor express required antigens.
Considerable site to site variation in plasma cell density is present (Macrofocal myeloma- Multiple plasmacytoma with normal bone marrow aspiration and biopsy)
Bone marrow biopsy
Excess of marrow plasma cells
Normally, plasma cells are present in small clusters of 5/6 cells around marrow arterioles
In myeloma, they occur in large foci, nodules / sheets, and later displaces normal marrow elements.
IHC on biopsy gives higher percentage of plasma cells compared to BM aspiration, due to relative difference in "aspirability" between plasma cells and other marrow cells.
IHC on biopsy is helpful in assessing monoclonality of cells
Kidney biopsy:
Interstitial infiltrates of abnormal plasma cells
Proteinaceous casts in distal convoluted tubules (they contain BenceJones proteins, normal plasma proteins, immunoglobulins, Tamm Horsfall protein etc)
Casts are often surrounded by multinucleate giant cells
Cells lining the cast containing tubules become necrotic
S. Protein electrophoresis and paraprotein quantitation
Prominent M spike (M component) in the region of gamma globulin
"M"- Earlier stood for "Malignant/ Myeloma", now it stands for "monoclonal"
It can be whole immunoglobulin (with heavy and light chain) or only immunoglobulin light chain
It is the most common screening test
Done on support medium such as cellulose acetate. After passing current, proteins get separated based on their charge.
Stained with Ponceaus blue and proteins are measured with densitometry.
Malignant/ Progressive- Plasma cell myeloma, solitary plasmacytoma, light chain amyloidosis, Waldenstrom macroglobulinemia, heavy chain disease, CLL and related lymphomas
S. Immunofixation electrophoresis
It is required to determine the immunoglobulin class
Other indications- Narrow band in beta or gamma region
Done on agarose gel with impregnated antiserum to specific heavy or light chains (anti alfa, beta, gamma, kappa and lambda) is used.
Only immunoprecipitates remain on the gel and non precipitated proteins wash out
Plasma cell dyscrasias secreting only light chains are usually not detectable as limit of immunofixation sensitivity is more than 10 x Normal FLC levels
S. Free Light Chain Assay
Measure low levels of FLC in serum
Measurement is done by nephalometry (based on observation that antigen-antibody complexes form cloudy precipitates). Precipitates can be detected photoelectrically. Varying dilutions of serum are incubated with antibodies specific for anyone of the different immunoglobulin heavy and light chains. After precipitate forms the amount of precipitate is determined by comparison with standard curves produced by precipitating immunoglobulins of known concentration.
Done when there is strong suspicion of myeloma, but in whom SPE is negative
It is useful in monitoring as well.
Abnormal kappa/lambda SFLC ratio is used as surrogate marker for the secretion of monoclonal free light chain
Abnormal ratio can be observed in SLE and HIV infection and during immune reconstitution following stem cell transplantation
Normal values (This can differ in different instruments)
Kappa- 3.3-19.4 mg/L
Lambda- 5.71-26.3 mg/L
Kappa:Lambda ratio- 0.26-1.65
Patients with FLC ratio >100, have high risk of progression to end stage organ damage
S. Immunoglobulin levels
Levels of uninvolved immunoglobulin is reduced
Electrophoresis, Immunofixation electrophoresis and FLC assay can be done on urine and CSF. Test on CSF helps in diagnosis of plasmacytoma of CNS secreting paraprotein.
Urine microscopic examination: Large, waxy, laminated casts in case of cast nephropathy
Urine-Bence Jones proteins.
They are free monoclonal light chains
They can be detected by
Heat precipitation – Urine flocculates when heated slowly to 50-60oC and flocculated proteins dissolve on boiling
Electrophoresis of concentrated urine specimen on cellulose acetate – BJ proteins migrated to globulin region.
Other conditions in which Bence Jones proteinuria is seen
Macroglobulinemia
Amyloidosis
Lymphoma
Leukemia
Immunophenotyping
Gating: Low side scatter with dim/negative CD45
Abnormal phenotype and/or monoclonality confirms the diagnosis
Express monotypic cytoplasmic immunoglobulin- Kappa or Lambda
Lack surface immunoglobulin
Markers positive on both normal plasma cells and myeloma cells: CD138, CD38, CD79a, CD40, CD44, CD54, VLA4, VLA5
Positive only on normal cells: CD19, CD45 (Positive on mature myeloma cells), CD11a
Negative on both normal and plasma cells: CD11b, LFA1, CD20
Cytogenetics/ FISH:
Cytogeneticsis difficult due to low proliferative fraction. But FISH can be done.
Most common abnormality is partial deletion of 13q14 or monosomy 13
Other abnormalities include:
Hyperdiploidy with gains in chromosomes – 3, 6, 7, 9, 11, 15 & 19
Losses from chromosomes - 8, 13, 14, X
Deletions from chromosomes 6q, 16q, 17p
Translocations involving IGH gene located on chromosome 14q32 is seen in 60% of patients. These translocations are associated with poor prognosis.
Molecular studies:
Antigen receptor genes: Clonal rearrangement of immunoglobulin genes. Single monoclonal rearranged Ig band is the rule in myeloma. High frequency of Ig VH gene somatic mutations is noted. This test is done using PCR.
Transposition of Cyclin D1 gene into on IgH gamma switch region
Over expression of Cyclin – D1
Altered expression of PAX-5 gene on chromosome 9 (This results in loss of CD19).
Deletion of 17p13 which results in loss of p53
Activation of NRAS or KRAS - Seen in 40% of cases
Other mutations seen- myc, FGFR-3, p18, RB1, CDKN2A, PTEN
Epigenetic changes manifested by DNA methylation are associated with tumor progression
Deletion of long arm of chromosome 7 (This confers multidrug resistance)
Quantitative PCR using allele specific oligonucleotides if the gold standard for MRD detection. But this is not practical as laboratory has to synthesize probe specifically for each patient.
Gene expression profile:
Helps in
Better understanding of pathobiology
Developing new prognostic models
Identifying new targets for drug development
Developing personalized medicine approaches
Technique:
RNA extracted from CD138+ myeloma cells
This is hybridized on high density arrays to evaluate expression of genes from the whole transcribed genome
There are about 10 subclasses of myeloma based on this technique
X-ray –
2 types of lesions
Diffuse decalcification
Localized area of bone destruction – Multiple, rounded, discrete, punched out area with no sclerosis at the margin (most commonly seen in skull)
Skeletal survey must include:
Chest (PA view)
AP and lateral view of cervical spine, thoracic and lumbar spine
AP and lateral - Humeri and femora
AP and lateral skull
AP of pelvis
Order of skeletal involvement: Vertebra (66%), ribs (44%), Skull (41%), Pelvis (28%), Femur (24%), Clavicle (21%)
Less sensitive compared to cross sectional imaging methods (Whole body CT, MRI, or PET/CT)
Whole body low dose CT
CT has improved sensitivity over X ray, as it picks up lytic lesions measuring <5mm, which are missed on X ray.
It can be done, even if patient cannot sit/ stand.
Helps in identifying soft tissue lesions and guides for taking biopsies and doing surgical/ radiotherapy interventions.
Whole body PET/CT
To be done if low dose CT is not available.
More sensitive in picking up extramedullary lesions
MRI
It can directly visualise the disease within the bone marrow, rather than its secondary effects on the cortical bone.
It is technique of choice in case of spinal cord compression
Avoid Gadolinium if GFR <30mL/min
Fat pad staining for amyloid
S. Viscosity- If hyperviscosity is suspected
HLA typing- If Allo HSCT is planned
DEXA scan – For measurement of bone density
Serum calcium level- Elevated
Serum uric acid level- Elevated
Serum albumin – Decreased
Serum CRP- Raised
Serum beta 2 microglobulin – Raised (reflects tumor burden and high value is associated with poor prognosis)
Serum Urea, Creatinine – elevated in case of renal dysfunction
Serum LDH level – Raised
Markers for bone destruction – Elevated levels of
TRACP – 5b
Collagen degradation products – NTX, 1CTP, CTX
Markers of bone formation – Decreased levels of bone alkaline phosphatase and osteocalcin
Criteria for Diagnosis:
Clonal bone marrow plasma cells ≥ 10% or biopsy proven bony or extramedullaryplasmacytoma (Clonality by flow/IHC)
AND
Any one or more of of following myeloma defining events(CRAB):
Calcium >11mg/dL
Renal dysfunction: Creatinine- >2mg/dL or creatinine clearance <40ml/min
Anemia- Hemoglobin <10gm/dL
Bone lesions: One or more osteolytic bone lesions on skeletal survey/ CT/PET-CT
OR
Clonal BM plasma cells >60%
OR
Abnormal FLC ratio >100 (involved kappa) or <0.01 (involved lambda)
Normal hemoglobin, serum calcium, Ig levels (Non-M component).
> 60 months
Stage III : Any one or more of the following
High M-component IgG>7 g/dl, IgA>5 g/dl; Urine BJ >12g/24hr
Advanced, multiple lytic bone lesions
Hemoglobin < 8.5 g/dl, serum calcium > 12 mg/dl
23 months
Stage II : Overall values between I and III
41 months
Subclassification : Based on renal function
A = serum creatinine< 2 mg/dl
B = serum creatinine> 2 mg/dl
International Staging System (ISS) stage:
Stage I-
Serum beta 2 microglobulin- <3500ng/mL
Serum albumin >3.5g/dL
Stage II
Not ISS I or III
Stage III
Serum beta 2 microglobulin >5500ng/mL
Revised ISS
Stage I- (All)
Serum beta 2 microglobulin- <3500ng/mL
Serum albumin >3.5g/dL
Standard risk cytogenetics by FISH
Serum LDH- Normal
Stage II
Not ISS I or III
Stage III (Any one)
Serum beta 2 microglobulin >5500ng/mL
High risk chromosomal abnormalities by FISH- t(4:14), t (14:16), del 17p, 1q gains, 1p del
Raised LDH levels
Prognosis:
Multiple myeloma is not considered a curable malignancy with current approaches
Median survival with present treatment- 7 years
Median survival in untreated symptomatic patients- 6 months
Poor prognostic markers
Advanced age with poor PS and presence of co-morbidities
Raised beta-2 microglobulin levels (which indicates high tumor burden)
Hemoglobin – Less than 7 gm/dl
Severe hypoalbuminemia
High LDH
Extramedullary disease
Intractable renal failure
Thrombocytopenia
>50% plasma cells in bone marrow
Plasmablastic morphology
Circulating plasma cells
High Ki 67 positivity
t(4:14), t (14:16), del 17p, 1q gains, 1p del, 13q del
Some gene expression profiles
High serum levels of IL6, CRP, hepatocyte growth factor, C terminal cross linked telopeptide of collagen I, TGFbetaand Syndecan
Tumor resistance
Risk stratification:
High risk: Any one of the following
t(4;14), t(14;16), t(14;20), del17p13, or gain 1q
LDH- >2 x ULN
Features of plasma cell leukemia (>5% circulating plasma cells)
Standard risk: Rest all patients
Differential Diagnosis: For increased number of plasma cells in bone marrow
Aplastic anemia
Rheumatoid arthritis
Cirrhosis of liver
Sarcoidosis
Secondary carcinoma
SLE
Chronic inflammation.
Primary amyloidosis- Generally havenephrotic syndrome, heart failure, hepatomegaly etc which are not found in multiple myeloma. They have usually less than 20% plasma cells in BM, no lytic lesions on imaging and BJ proteins in urine)
Chemotherapy consent after informing about disease, prognosis, cost of therapy, side effects, hygiene, food and contraception
Fertility preservation
PICC line insertion and Chest X ray after line insertion
Tumor board meeting and decision
Attach supportive care drug sheet
Inform primary care physician
Treatment Plan:
Goals of treatment:
Control of disease
Maximize quality of life
Prolong survival
Affordable patient with high risk disease: Daratumumab-bortezomib-lenalidomide-dexamethasone (DVRd) is the preferred regimen(3-6 cycles), followed by early tandem ASCT/Allo SCT in select cases, followed by dual drug maintenance using Lenalidomide and bortezomib.
Extremely frail patient unable to tolerate 3 drugs: 2 drugs (Lenalidomide with Dexa) may be given given, followed by Lenalidomide maintenance.
For all others:
First line regimens:
BorLenDex
CyBorD
BorThalDex
DaraLenDex
CarfilLenDex
Second Line regimens (Avoid Bortezomib or Lenalidomide if used previously):
If CNS is involved- RT to localized disease, IT chemo, along with systemic therapy
Response Criteria:
Complete remission (CR)
Negative immunofixation of serum and urine
Disappearance of any soft tissue plasmacytomas
<5% plasma cells in BMA
Stringent CR
CR with normal FLC ratio
Partial response:
≥ 50% reduction in Serum M protein or ≥ 50% decrease in difference between involved and uninvolved FLC levels
≥ 50% reduction in number of plasma cells in BM (Provided baseline plasma cell percentage is >30%)
Monitoring when on maintenance therapy/otherwise:
Once a month: History, examination, CBC, Creat, Calcium
Once in 3 months: SPE
If clinically indicate: BMA and biopsy imaging, Immunofixation electrophoresis, SFLC Assay
Progressive disease is identified by rise in monoclonal protein/ worsening SFLC ratio. This does not require immediate change of treatment.
Therapy for relapsed disease is indicated if:
Clinical relapse (Development of CRAB features)
Extramedullary disease
Rapid rise of paraproteins: Doubling of M Protein over 2-3 months with an increase in the absolute levels of M protein of >1gm/dL, confirmed by 2 consecutive tests.
Absence of myeloma-defining events or amyloidosis
Bone marrow plasma cells- >20%
M Protein- >2gm/dL
SFLC ratio- >20
About Each Modality of Treatment:
Chemotherapy Regimens for treatment of myeloma
Triplet regimens are usually used as standard therapy, however elderly/ frail patients may be treated with doublet therapy
Usually 4-6 cycles are given prior to reassessment for response
In case of pre-existing severe neuropathy- Avoid thalidomide, bortezomib and Vincristine.
CyBorD
Frequency: 28 days
Inj. Dexamethasone 40mg in 100ml NS over 1hr- on day 1, day 8, day 15 and day 22
Inj. Bortezomib- 1.3mg/m2- in running saline- on day 1, day 8, day 15 and day 22
Inj. Cyclophosphamide- 500mg- in 500ml D5% over 2hrs- on day 1, day 8, day 15 and day 22
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
ANC- <1000/cmm or Platelet count- <50,000/cmm- Hold chemo until recovery and restart at dose- Cyclophosphamide 400mg/300mg and Bortezomib- 1mg/m2
Creatinine- >3.4mg/dL despite of vigorous hydration- Omit cyclophosphamide
Creatinine clearance- <30ml/min- Bortezomib- Give 50% of dose
Peripheral neuropathy
Grade 2- Bortezomib- Give 1mg/m2
Grade 3- Stop bortezomib till symptoms subside and then restart at 0.7mg/m2
Grade 4- Discontinue bortezomib
Severe diarrhea- Delay until diarrhea resolves. Restart bortezomib at 1gm/m2
LenBorD
Frequency: 21 days
Inj. Dexamethasone 40mg in 100ml NS over 1hr- From Day 1 to Day 4. (4 days)
Inj. Bortezomib- 1.3mg/m2- in running saline- on day 1, day 4, day 8 and day 11
Cap. Lenalidomide- 25mg- PO- OD- From day 1 to day 14. (14 days)
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
Dexamethasone
Bortezomib
Lenalidomide
Zolendronic acid
ANC (/cmm)
<500
--
Give 1mg/m2
Stop till recovery and restart at 10mg/day
Platelet count (/cmm)
<30,000
Stop till recovery and restart at 10mg/day
Creatinine clerance (ml/min)
30-50
Give 10mg/day
Give 3mg
<30
Give 50% of dose
Give 5mg/day
Omit
Peripheral neuropathy
Grade 2
Give 1mg/m2
Grade 3
Stop till recovery and restart at 0.7mg/m2
Grade 4
Omit
Severe diarrhea
Stop till diarrhea subsides and ten start at 1mg/m2
MPT
Frequency: 28 days
Tab. Melphalan- 7mg/m2- OD- From Day 1 to Day 4 (4 days)
Tab. Prednisolone- 50mg- OD- From Day 1 to Day 4 (4 days)
Tab. Thalidomide 50mg HS, then gradually increase to 200mg HS- From Day 1 to Day 28
Tab. Aspirin- 75mg- OD- From Day 1 to Day 28
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
Melphalan
Prednisolone
Thalidomide
Zolendronic acid
ANC (/cmm) and / or Platelet count (/cmm)
(Monitor every week)
Hold chemo till ANC >1300 and Platelet count >75,000 and decrease the dose in subequent cycles.
If no improvement till 6 weeks- change to alternate chemotherapy.
500-1000/ 25,000-75,000
Give for 3 days
<25,000/ <500
Give for 2 days
Creatinineclerance (ml/min)
30-50
Give 50% of dose
Give 3mg
<30
Avoid
Omit
Peripheral neuropathy
Grade 2 or more
Stop
Carfil-Len-Dex
Frequency: 28 days
Inj. Dexamethasone- 40mg in 100ml NS over 30min- On Day 1, Day, 8, Day 15 and Day 22.
Inj. Carfilzomib- 27mg/m2 in 100ml D5% over 10min (start within 30min of Dexamethasone)-
For Cycles 1- 12- On Day 1, Day 2, Day 8, Day 9, Day 15 and Day 16.
For Cycles 13- 18- On Day 1, Day 2, Day 15 and Day 16.
After 18 cycles- discontinue Carfolzomib and continue Len Dexuntil disease progression/ acceptable toxicity.
Tab. Lenalidomide- 25mg- OD- From day 1 to Day 21 (21 days)
Tab. Aspirin- 75mg- OD- From day 1 to Day 21 (21 days)
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
Cap BSA at 2..2m2
Dexamethasone
Carfilzomib
Lenalidomide
Zolendronic acid
Elderly
Give 20mg
ANC (/cmm) or Platelet count (/cmm)
Delay until ANC >1000 and Platelet count >30,000
<1000/ <30,000
First time- Restart in same dose
Recurrence- Restart at 20mg/m2
Again recurrs- 15mg/m2
Again recurs- Discontinue
First time- Restart at 20mg/day
Same counts on day 15- Omit for rest of cycle and next cycle give 15mg/day
Recurs again on day 15- Omit for rest of cycle and next cycle give 10mg/day
Recurs again on day 15- Omit for rest of cycle and next cycle give 5mg/day
Recurs again on day 15- Omit for rest of cycle and next cycle give 2.5mg/day
Creatinineclerance (ml/min)
30-60
10mg/day
Give 3mg
15-30 but no HD
15mg/day
Omit
15-30 and requires HD
5mg/day- after HD
Omit
<15
Hold till >15
5mg/day
Omit
Pomalidomide- Cyclo-Dex
Frequency: 28 days
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Tab. Pomalidomide- Start with 2mg- OD and gradually increase to 4mg OD- From days 1 to 21
Tab. Dexamethasone- 40mg- On days 1, 8, 15 and 22.
Tab. Cyclophosphamide- 400mg- on days 1, 8, and 15 (Premedicate Tab. Emeset- 8mg)
Tab. Aspirin- 75mg- OD
Dose adjustments:
ANC- <1000/cmm or Platelet count- <50,000/cmm- Hold chemo until recovery and restart at dose- Cyclophosphamide 300mg and Pomalidomide at 3mg. If this repeats hold until recovery and restart at dose- Cyclophosphamide 300mg and Pomalidomide at 2mg. If this repeats hold until recovery and restart at dose- Cyclophosphamide 300mg and Pomalidomide at 1mg. If this repeats discontinue pomalidomide.
Creatinine- >3.4mg/dL despite of vigorous hydration- Omit cyclophosphamide. Pomalidomide has no renal dose adjustment.
Severe rash- Discontinue pomalidomide
VAD
Frequency: 21 days
Inj. Vincristine (0.4mg/m2) + Inj. Doxorubicin (9mg/m2)- in 500ml NS over 24 hrs- From Day 1 to Day 4 (Continuous infusion for 4 days)
Tab. Dexamethasone- 40mg- OD from Day 1 to Day 4 and then from Day 12 to Day 15.
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
ANC- <1000/cmm or Platelet count <50,000/cmm- Hold next cycle until ANC- >1000/cmm and Platelet count >50,000/cmm (Unless cytopenia is thought to be due to myeloma per se)
Significant neuropathy- Omit vincristine
Bilirubin- >3gm/dL- Consider decreasing the dose of vincristine and doxorubicin.
Severe steroid related side effects- Omit dexamethasone from day 12 to day 15.
Creatinine clearance (ml/min)
30-50- Zolendronic acid- Give 3mg
<30- Omit Zolendronic acid
Dara-Len-Dex
Frequency: 28 days
Premedication- Tab. Montelukast- 10mg- Stat,Tab. Paracetamol- 1gm-Stat, Tab. Avil- Stat
Tab. Dexamethasone 20mg
Cycle- 1 and 2- Days- 1&2, 8&9, 15&16, 22&23
Cycle- 3 to 6- Days- 1&2, 8&9, 15&16, 22&23
Cycle 7 onwards- Days 1 and 2, 40mg on days 8, 15, and 22.
Inj. Daratumumab- 1800mg- SC- over abdomen- over 3-5min
Cycle- 1 and 2- Days- 1, 8, 15, 22
Cycle- 3 to 6- Days- 1 and 15
Cycle 7 onwards- Day 1
Cap. Lenalidomide- 25mg- PO- OD- From day 1 to day 21
Inj. Zolendronic acid- 4mg- in 100ml NS over 30min- on day 1
Dose adjustments:
Dexamethasone
Dara
Lenalidomide
Zolendronic acid
ANC (/cmm)
<1000
--
Stop till recovery and restart at 10mg/day
Platelet count (/cmm)
<30,000
Stop till recovery and restart at 10mg/day
Creatinine clerance (ml/min)
30-50
Give 10mg/day
Give 3mg
<30
Give 5mg/day
Omit
Autologous Stem Cell Transplantation
It is associated with long treatment free survival and excellent quality of life.
Renal dysfunction or advanced age are not contraindications for auto SCT
Compared to chemotherapy alone, ASCT has superior progression free survival (median >20 months) but same overall survival.
Should be done if patient is in CR or PR
2 types:
Early HCT: Done as a part of initial therapy
Late HCT: Delayed until first relapse
PBSCs are harvested after mobilization with growth factors. Sometimes cyclophosphamide (1.5-4gm/m2- especially if lenalidomide is used in induction) and plerixafor are used.
Generally sufficient stem cells to support 2 ASCTs are collected. Second ASCT is to be done in case of relapse.
After high dose melphalan (usually 200mg/m2, needs renal dose adjustment), stem cell infusion is done
High response rate – CR up to 30%, 7% molecular remission is seen even in patients who are refractory to conventional treatment
Event free survival- 13 months
Median survival – 50 months
TRM < 2%
Tandem transplant- Second course of high dose chemotherapy and autologous stem cell transplant, done within 6 months of first course. There is no clear data on benefit of this on improving overall survival.
Allogeneic SCT
Myeloablative conditioning- only for age <40years who have achieved atleast PR with initial therapy.
Non myeloablative/ RIC regimens may be used for elderly patients.
Too risky but has advantage of Graft Vs Myeloma effect. Hence long term disease free survival
Median survival – 50 months
TRM – 30 – 50%
Chances of CR- 50%
Limited role as more than 75% patients of myeloma are aged >55years and have multiple comorbidities.
It is the only treatment option with potential of cure
DLI can be used for relapse or for mixed chimerism
MUD/Haplo SCT is not recommended, except in the context of a clinical trial.
Maintenance therapy
Prolongs remission duration, thereby life expectancy
Must be combined with bisphosphonates once a month
Supportive Care:
Bone disease:
Bisphosphonates- Given to all patients and continued indefinitely even after treatment except those who are in CR following transplant.
Due to concerns of osteonecrosis of jaw, do dental examination prior to starting.
Calcium and vitamin D supplementation also should be given.
Stabilization and subsequent radiotherapy for fractures (8Gy single fraction- Improves pain control and promotes healing of fracture site)
Pain:
67% of myeloma patients have pain at diagnosis.
Refer to supportive care section
Hypercalcemia: Refer to supportive care section
Tumor lysis prophylaxis and treatment: Refer to supportive care section
Renal impairment:
Rehydration- To achieve urine flow of 3lit/day
Correct hypercalcemia
Discontinue nephrotoxic drugs like NSAIDS
Start antimyeloma therapy immediately (High dose dexamethasone to be started while decision about chemotherapy is being made)
Dialysis
New dialysis filters are available that can remove light chains.
Nephropathy is reversible in 50% of patients.
For renal transplant myeloma is not a contraindication
Radiotherapy –
Indications:
Extramedullaryplasmacytoma
Painful destruction of vertebral body/ pathological fracture
Spinal cord compression
Doses:
Low dose- RT- 8Gy x 1 fraction
10-30Gy in 2-3Gy fractions
Surgery: Indications:
Spinal cord compression
Unstable vertebral fractures
Anemia:
Transfusion support
Erythropoietin- 5000 U- Twice a week – SC or Darbepoetin 6.25 microgm/kg every 3 weeks
Decreases transfusion requirement
Given to maintain Hemoglobin around 11gm/dL. With higher hemoglobin, risk of thrombosis increases.
Hyperviscosity – Plasma exchange
Infections
Antibiotics
Prophylactic IV- immunoglobulins if patient has recurring infections- 500mg/kg every month, for up to 6 months.
Pneumococcal and influenza vaccines
PCP, herpes and antifungal prophylaxis
Coagulation
Prophylactic anticoagulation for patients receiving thalidomide/ lenalidomide
Aspirin- 75mg- OD if patient is receiving thalidomide/ lenalidomide
If additional risk factors forthrombosis(personal/family history of DVT, Obesity, co-morbidities, recent surgery) are present, avoid Thal/Len or give LMWH/Warfarin (target INR-2-3)/ Rivaroxaban-10mg-OD as thromboprophylaxis. It should be continued till disease is under control (generally for 4-6 months).
Avoid anticoagulation/ antiplatelet agents, if there is risk of bleeding.
Cord compression: Refer supportive care section
Peripheral neuropathy: Neuromodulatory drugs may be used
Gabapentin- 800mg/day. It suppresses BM, hence must be used with caution if AutoSCT is planned
Pregabalin
Oxcarbamazepine
IgD Myeloma
1.8% of all myelomas
Very small or no M band
BJ Proteins, extramedullary involvement, lytic lesions and amyloidosis are common
IgM Myeloma
0.4% of all myelomas
Must be differentiated from Waldenstrom'smacroglobulinemia
High incidence of t(11:14)
Poor prognosis
Smouldering myeloma
Fulfills the criteria for multiple myeloma but no myeloma defining (CRAB) events/ amyloidosis
It progresses to multiple myeloma at a rate of approximately 10% per year.
Criteria for diagnosis: Essential:
Serum monocional protein (IgG or IgA) ≥ 3 g/dL, or urinary monoclonal protein ≥ 500 mg per 24 h and/or clonal bone marrow plasma cells 10-60%
AND
High risk: If 2 or more of following features are present-
Management
Observe only- 3-6 monthly CBC, Creatinine, Calcium, SPEP, SIFE and SFLC Assay. If clinically indicated do BMA and biopsy and WB MRI or PET-CT.
If there is progression, treat like multiple myeloma
For high risk patients: Same as above/ Lenalidmide monotherapy may be considered
Indolent myeloma
Criteria for diagnosis of multiple myeloma are met, but
Patients have up to 3 lytic bone lesions, without bone pain
M component is at intermediate level.
Normal Hb, serum calcium and creatinine.
No evidence of infection
Non secretory myeloma (1% cases)
In 85% of cases, there is synthesis, but no secretion of immunoglobulin
Presentation is same as multiple myeloma but renal insufficiency is not seen.
Fulfills all criteria of multiple myeloma (including >10% clonal plasma cells in bone marrow) but no monoclonal immunoglobulin and normal SFLC assay.
Plasma cell leukemia
Presence of ≥ 5% circulating plasma cells in peripheral blood smears in patients otherwise diagnosed with plasma cell myeloma
Unlike myeloma, CD56 is negative. Some are cyclin D1-Positive.
Can be de novo or may appear as late feature in the course of multiple myeloma.
Higher incidence in Light chain only, IgD or IgE myelomas
Morphology: Plasma cells may be small with little cytoplasm and may resemble plasmacytoid lymphocytes.
Presentation- Osteolytic lesions are less frequent, Lymphadenopathy and organomegaly are common.
Prognosis: Poor
Treatment: Treat as ver high risk myeloma. Bortezomib containing chemotherapy followed by autologous stem cell transplant
Solitary plasmacytoma
Localized osseous or extraosseous growth of plasma cells, identical to those seen in plasma cell myeloma without evidence of multiple myeloma or end organ damage due to plasma cell neoplasia
Account for 3-5% of plasma cell neoplasms
M:F= 1.87
Mean age- 60 years
Types
Solitary plasmacytoma of bone: Involves vertebrae, ribs, skull, pelvis and femur. There is lytic bone lesion but without bone marrow involvement. Present with bone pain at the site of the lesion and pathological fracture
Markers in differentiating plasma cells from B cells: CD138, MUM1/IRF4, CD20, PAX5
Bone marrow aspiration and biopsy
Skeletal survey/ MRI spine with pelvis
Diagnostic criteria- Each of the following must be present
Biopsy proven plasmacytoma with biopsy proven clonal plasma cells
Absence of clonal plasma cells in BM aspirate (<10% )
Absence of end organ damage that can be attributed to underlying plasma cell neoplasm (CRAB features)
Absence of other lytic lesions on WB-PET-CT or WB- MRI.
Prognostic categories:
Variables
Risk Group
5 year progression rate to multiple myeloma
Normal SFLC ratio Monoclonal protein <5gm/L
Low
13%
Either variable abnormal
Intermediate
26%
Abnormal SFLC ratio Monoclonal protein >5gm/L
High
62%
>50% of patients with plasmacytoma may progress to multiple myeloma in 2-4 years. This chance is 30% with extra osseous plasmacytomas.
Plasmacytoma with minimal BM involvement (<10% clonal plasma cells) have high risk of developing multiple myeloma
Treatment:
Radiation – 40 – 50 Gy in 1.8-2Gy/fraction to involved field (with margin of atleast 2cm) with/without surgery
Surgery is advised if there is structural instability or if there is neurological compromise due to mass effect. This should be done prior to radiation.
Decompression laminectomy
Spinal fusion
Intramedullary rod fixation of long bones
Plasmacytoma with minimal BM involvement may be treated similar to multiple myeloma
Local recurrence chance is < 5%
Follow-up: Every 3-6 monthly with CBC, Creatinine, Calcium, LDH, SPEP, SIFE and SFLC Assay- If clinically indicated BMA and Biopsy- If there is progressive disease treat as multiple myeloma
Multiple Plasmacytomas
Criteria for diagnosis:
No M protein in serum/ urine
>1 localized area of bone destruction/ extramedullary tumor of clonal plasma cells
Normal bone marrow
Normal skeletal survey/ MRI spine
No related organ/ tissue impairment
Treatment - Not clear. Depends on patient's age, sites involved, number of lesions, disease free survival. Options of treatment are
Systemic therapy +/- autoSCT
Radiotherapy alone
Plasma cell neoplasms with associated paraneoplastic syndrome
POEMS Syndrome
(Osteosclerotic Myeloma, Crow – Fukase syndrome)
It is associated with:
Polyneuropathy- Sensory motor demyelination
Organomegaly- Hepatosplenomegaly
Endocrinopathy- Diabetes, Testicular atrophy
Monoclonal gammopathy- Usually IgG lambda or IgA lambda
<10% bone marrow infiltration by clonal neoplastic cells
No evidence of anaemia, constitutional symptoms, hyperviscosity, lymphadenopathy, lytic bone lesions, renal insufficiency, or hepatosplenomegaly that can be attributed to the underlying lymphoproliferative disorder
If paraproteins are >30gm/L or Marrow plasma cells are >10%, with no CRAB features, it is categorized as smoldering myeloma
Investigations:
Evaluate thoroughly to rule out myeloma- CBC, RFT, Ca, SIFE. SFLC assay
Skeletal survey
Bone marrow aspiration and biopsy
Normal mature plasma cells, without nucleoli which constitute<10% of nucleated cells in BM
Express monotypic cIG of the same isotype as the M component in the serum and urine
Flow cytoemtry: 2 populations of plasma cells
Normal (Polyclonal)- CD38+ (bright), CD19+, CD56-
Abnormal (Monoclonal)- CD19-, CD56+/-, Weak CD38+
Cytogenetics- Difficult to get metaphases
Risk of transformation to multiple myeloma is 1% per year
High risk features:
M Protein- >1.5gm/dL
Non IgG MGUS
Abnormal SFLC ratio
Risk
Number of risk factors
Absolute risk of progression at 20years
Low
0
5%
Low-Int
1
21%
Int-High
2
37%
High
3
58%
No treatment needed (Early intervention with len-dexa is being tried in high risk patients)
Follow up every 6-12 months: With History, SPE, CBC, RFT and Calcium.
Monoclonal gammopathy of clinical significance
It is same as MGUS, but is associated with potentially severe organ damage, due to toxicity of the monoclonal immunoglobulin or to other mechanisms
They include
Monoclonal gammopathy of renal significance
Atypical bullouspephigoid with MGUS
MGUS with corneal crystallopathy
Paraprotein related neuropathy
They need treatment to preserve involved organ function
Monoclonal gammopathy of renal significance
It includes any B-Cell or plasma cell clonal disorder that does not fulfill criteria for cancer yet produces a nephrotoxic monoclonal immunoglobulin that leads to kidney injury.
In this condition, there is low tumor burden, yet, there is renal injury due to presence of monoclonal immunoglobulins.
Mechanisms of renal damage include:
Cast nephropathy (this requires high levels of serum free light chains and often patients with this findings have overt multiple myeloma)
Amyloid deposition
Intracytoplasmic crystals of light chains
C3 Glomerulopathy
Activation of alternate complement pathway: Proliferative glomerulonephritis with monoclonal immunoglobulin deposits (PGNMID)
Thrombotic microangiopathy
40-45% of patients with MGUS have MGRS related diseases if renal biopsy is done. Renal biopsy is indicated in MGUS is there is
High urinary protein level (>1.5gm/day)
Abnormal free light chain ratio
Microscopic hematuria
AKI-Stage 3
eGFR <60mL/min
Subtypes of MGRS
Always associated with MGRS
Crystal-storing histiocytosis
Crystalglobulin-induced nephropathy
Ig-related amyloidosis
LC proximal tubulopathy
Monoclonal Ig deposition disease
Proliferative glomerulonephritis with monoclonal Ig deposits
Frequently associated with MGRS
C3 glomerulopathy with MG
Cryoglobulinaemic glomerulonephritis (type I and [mostly] type II)
Monoclonal immunotactoid glomerulonephritis
Thrombotic microangiopathy with MG
Rarely associated with MGRS
Monotypic membranous nephropathy
Monotypic anti-GBM disease
Monotypic IgA nephropathy / Henoch Schönlein purpura nephritis
If MGRS is idenfied on renal biopsy, these patients must undergo
S. Protein electrophoresis
S. Immunofixation electrophoresis
S. Free light chain assay
Bone marrow aspiration and biopsy
Flow cytometry on BM sample for identification of clonal population
PET CT to detect focal lesion outside BM
Criteria for diagnosis:
Essential
Kidney biopsy demonstrating injury as a result of a monoclonal immunoglobulin
Proteinuria > 1 g/d comprised mostly of albuminuria
Progressive acute or subacute kidney injury
Desirable
Lack of lytic bone lesions
No extramedullary plasmacytoma
No hypercalcemia secondary to bone lesions
No anaemia with haemoglobin < 10 g/dl
Bone marrow plasma cells < 60%
Involved to uninvolved free light chain ratio < 100
No hyperviscosity
No bulky lymphadenopathy
No thrombocytopenia (< 1lac/cmm)
Treatment (Depends on nature of the clone detected)
If there is <1gm/day proteinuria and no renal damage: Wait and watch along with ACE inhibitors as anti-proteinuric therapy
CLL clone/ IgM Deposits: Chemotherapy along with rituximab
Plasma cell clone/ non-IgM Deposits: Myeloma type of treatment (Bortezomib based)
If there is ESRD- Renal transplantation is the best option, after eradicating monoclonal gammopathy
If there is no favourable response after 2-3 cycles of chemotherapy, change the therapy to target the alternate clone.
Heavy chain diseases
Characterized by the production of a shortened monoclonal immunoglobulin heavy chain, with typically no light chain.
Heavy chain is often truncated with absence of portions of the variable heavy chain domain
As variable sized proteins are produced, M spike is is often not seen, but immunofixation can identify abnormal chains.
3 types:
Mu HCD: Extremely rare, resembles CLL/SLL, Some associated with MYD88 p. L265P mutation, presents with splenomegaly, hepatomegaly, lymphadenopathy or lytic bone lesion, BM shows IgM positive small lymphocytes, wait and watch, if causing problems- treat as CLL.
Gamma HCD (Franklin disease): Rare, resembles lymphoplasmacytic lymphoma/ SMZL, may be associated with autoimmune diseases, presents with fever, weight loss, and generalized lymphadenopathy, IgG is elevated, wait and watch, if causing problems- Rituximab based therapy
Alpha HCD (Selingmann’s disease, Mediterranean abdominal lymphoma, immuno proliferative small intestinal disease): Caused by infections such as Helicobacter pylori, Vibrio cholera, Campylobacter jejuni, or intestinal parasites, Presents with chronic watery diarrhea, weight loss, malabsorption, abdominal pain, finger clubbing, vomiting and fever, IgA is elevated, duodenum is commonly involved, Treatment includes 6 months course of antibiotics. If no response/ systemic disease/ large cell transformation use R-CHOP.
Idiopathic Capillary Leak Syndrome
Introduction:
It is a rare disease charectorised by episodes of severe hypotension, hypoalbuminemia and hemoconcentration
Thromboprophylaxis in patients receiving treatment for multiple myeloma
There is limited data on use of DOACs as thromboprophylaxis in patients with multiple myeloma. In a study by Katrina Piedra, which involved 305 newly diagnosed myeloma patients, the incidence of VTE in patients receiving carfilzomib, lenalidomide, dexamethasone + aspirin, bortezomib, lenalidomide, dexamethasone with Aspirin and carfilzomib, lenalidomide, dexamethasone + rivaroxaban were 16·1%, 4·8%, and 4·8%, respectively. This shows risk of VTE is higher in KRD induction compared to RVD induction. This higher risk can be mitigated with use of low dose rivaroxaban.
Novel PBX1-FOXM1 axis detected in chr1q-amp high risk myeloma
Clinical multi-omics study has showed that in patients with chr1q-amp associated high risk myeloma, there is involvement of core PBX1-FOXM1 regulatory axis. In view of availability of PBX1 inhibitor such as T417 and thiostrepton, these findings are said to be clinically significant. Chr1q-amp is a major copy number aberration (CNA) which confers adverse prognosis in several types of cancers. The technology used in identifying such pathways is called “systems medicine approach”.
PIM2 kinase has pivotal role in plasmablast generation
Differentiation of B cells into plasmablasts and then plasma cells involves extensive cell reprogramming. By using specific inhibition strategies (including a novel morpholino RNA antisense approach) researchers have found novel PIM2 kinase, which is highly expressed in malignant plasma cells. Experimental studies have also showed that pan-PIM inhibitors are useful in reducing proliferation of plasma cells. PIM2 may be used as therapeutic target in multiple myeloma.
Impact of maintenance therapy post autologous stem cell transplantation for multiple myeloma in early and delayed transplant
ASCT within 12 months of diagnosis is called early ASCT, whereas after 12 months is called as late ASCT. Most common maintenance approach was an IMiD (61%), followed by a PI (31%). PFS was superior with maintenance for early ASCT vs. for delayed ASCT. OS from diagnosis was also better for the whole cohort with maintenance therapy compared to patients not receiving maintenance therapy.
Triplet Therapy, Transplantation, and Maintenance until Progression in Myeloma
Among 357 patients in the RVD-alone group and 365 in the transplantation group, at a median follow-up of 76.0 months, 328 events of disease progression or death occurred. The risk was 53% higher in the RVD-alone group than in the transplantation group. 5-year survival was 79.2% and 80.7% respectively.
Teclistamab in Relapsed or Refractory Multiple Myeloma
Teclistamab is a T-cell–redirecting bispecific antibody that targets both CD3 expressed on the surface of T cells and B-cell maturation antigen expressed on the surface of myeloma cells. Patients with relapsed/ refractory disease received a weekly subcutaneous injection of teclistamab (at a dose of 1.5 mg per kilogram of body weight) after receiving step-up doses of 0.06 mg and 0.3 mg per kilogram. With a median follow-up of 14.1 months, the overall response rate was 63.0%.
G protein–coupled receptor, class C, group 5, member D (GPRC5D) has been identified as an immunotherapeutic target in multiple myeloma. In a recent study GPRC5D-targeted CAR T-cell therapy (MCARH109) was administered to patients with heavily pretreated multiple myeloma, including patients with relapse after BCMA CAR T-cell therapy. A response was reported in 71% of the patients in the entire cohort.
Data from a large phase III trial (Myeloma XI) were examined to determine the relationship between MRD status, progression-free survival (PFS), and overall survival (OS) in post-ASCT patients randomly assigned to lenalidomide maintenance or no maintenance at 3 months after ASCT. MRD status was assessed by flow cytometry. MRD-negative status was associated with improved PFS and OS. Patients randomly assigned to lenalidomide maintenance were more likely to convert from being MRD-positive before maintenance random assignment to MRD-negative 6 months later (lenalidomide 30%, observation 17%).
Identification of High-Risk Multiple Myeloma With a Plasma Cell Leukemia-Like Transcriptomic Profile
A molecular marker for primary plasma cell leukemia is currently lacking. In this study a transcriptomic classifier for PCL-like disease was bioinformatically constructed. Study concluded that primary plasma cell leukemia can be identified on the basis of a specific tumor transcriptome, which was also present in patients with high-risk newly detected multiple myeloma, despite not being clinically leukemic. Incorporating PCL-like status into current risk models in NDMM may improve prognostic accuracy.
Talquetamab, a T-Cell–Redirecting GPRC5D Bispecific Antibody for Multiple Myeloma
G protein–coupled receptor, family C, group 5, member D (GPRC5D) is an orphan receptor expressed in malignant plasma cells. Talquetamab, a bispecific antibody against CD3 and GPRC5D, redirects T cells to mediate killing of GPRC5D-expressing myeloma cells. In the prresent study talquetamab administered intravenously weekly or every other week (in doses from 0.5 to 180 μg per kilogram of body weight) or subcutaneously weekly, every other week, or monthly (5 to 1600 μg per kilogram) in patients who had heavily pretreated relapsed or refractory multiple myeloma. 232 patients had received talquetamab. Cytokine release syndrome, skin-related events, and dysgeusia were common with talquetamab treatment. A response rate of approximately 65% was observed.
Abnormal metaphase cytogenetics predicts venous thromboembolism in myeloma
The aim of this study was to develop a new risk prediction model for VTE in the context of modern antimyeloma therapy. A 5-component risk prediction tool, named the PRISM score, was developed, including the following variables: prior VTE, prior surgery, immunomodulatory drug use, abnormal metaphase cytogenetics, and Black race. The model stratified patients into low, intermediate, and high risk, with 12-month cumulative VTE incidence of 2.7%, 10.8%, and 36.5%, respectively. Risk of VTE increased significantly with increasing score.
Efficacy and safety of cilta-cel in patients with progressive multiple myeloma after exposure to other BCMA-targeting agents
B-cell maturation antigen (BCMA)–targeting therapies, including bispecific antibodies (BsAbs) and antibody-drug conjugates (ADCs), are promising treatments for multiple myeloma (MM), but disease may progress after their use. Cilta-cel, is an anti-BCMA chimeric antigen receptor T therapy. In the present study single cilta-cel infusion was given after lymphodepletion. Overall response rate was 60.0%. Median duration of response and progression-free survival were 11.5and 9.1 months, respectively.
Ide-cel or Standard Regimens in Relapsed and Refractory Multiple Myeloma
Idecabtagene vicleucel (ide-cel) is a B-cell maturation antigen–directed chimeric antigen receptor (CAR) T-cell therapy. Present study involved adults with relapsed and refractory multiple myeloma who had received two to four regimens previously. They were randomly assigned to receive either ide-cel (dose range, 150×106 to 450×106 CAR-positive T cells) or one of five standard regimens. Ide-cel therapy significantly prolonged progression-free survival and improved response as compared with standard regimens.
Elotuzumab Plus Pomalidomide and Dexamethasone for Relapsed/Refractory Multiple Myeloma: ELOQUENT-3 Trial
Patients with RRMM who had received ≥ 2 prior lines of therapy, with disease refractory to last therapy and either refractory or relapsed and refractory to lenalidomide and a PI were randomly assigned (1:1) to receive EPd or Pd. EPd demonstrated a statistically significant improvement in OS versus Pd.
Ciltacabtagene Autoleucel, an Anti–B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE Study.
Present study presents updated results 2 years after last patient in CARTITUDE Study. At a median follow up of 27.7 months, the overall response rate was 97.9% 27-month PFS and OS rates were 54.9% and 70.4%, respectively. Duration of response, PFS, and/or OS were shorter in patients with high-risk cytogenetics, International Staging System stage III, high tumor burden, or plasmacytomas.
Bendamustine with pomalidomide and dexamethasone in relapsed/refractory multiple myeloma
In this phase II study, patients received bendamustine 120 mg/m2day 1, pomalidomide 3 mg days 1–21, and dexamethasone 40 mg days 1, 8, 11, 22. Regimen given for a maximum of six cycles. An ORR of 57.6% was achieved. At a median follow-up of 8.6 months, the median PFS and OS were 6.2 and 9.7 months respectively. Toxicity was manageable.
Daratumumab: Results from the Canadian Myeloma Research Group Database
This study aimed to evaluate the outcomes of dara-containing regimens in the Canadian real-world setting among relapsed and refractory MM available within the national Canadian Myeloma Research Group Database. A total of 583 MM patients who received dara-based therapy in second-line or later treatment were included. After a median follow-up of 17.5 months, the median progression-free survival (PFS) and overall survival (OS) for the entire cohort were 13.1 and 32.9 months, respectively. The addition of bortezomib, lenalidomide or pomalidomide to dara resulted in an improved median PFS and OS of 8.3 and 26.2 months; 26.8 and 43.0 months; and 9.7 and 31.4 months respectively.
Present study investigated early mortality in a prospective cohort study of all patients with newly diagnosed myeloma registered on the Australian and New Zealand Myeloma and Related Diseases Registry at 36 institutions between July 2011 and March 2020. Variables that were independent predictors of early mortality were age, Eastern Cooperative Oncology Group performance status, serum albumin, cardiac disease and International Staging System.
Belantamab mafodotin therapy for relapsed/refractory multiple myeloma
Belantamab mafodotin, an immuno-conjugate targeting B-cell maturation antigen and was recently approved for heavily pretreated relapsed/refractory multiple myeloma patients. This real world data showed response rate of 45.5%. Median progression-free survival was 4.7 months.
Ultra-High-Risk Multiple Myeloma: Induction with Daratumumab, Cyclophosphamide, Bortezomib, Lenalidomide and Dexamethasone
The OPTIMUM phase II trial assessed Dara-CVRd treatment in high-risk multiple myeloma patients before and after autologous stem-cell transplant (ASCT), comparing outcomes with the MyeXI trial. UHiR patients received Dara-CVRd induction, ASCT, and Dara-VRd consolidation. Comparison showed OPTIMUM's superiority (99.5% probability) in 18-month progression-free survival (PFS18m) over MyeXI. At 30 months, OPTIMUM demonstrated improved PFS (77% vs. 39.8%) and overall survival (OS) (83.5% vs. 73.5%). Extended Dara-VRd post-ASCT consolidation was well-tolerated.
DOI: 10.1200/JCO.22.02567
Role of PET/CT after autologous stem cell transplantation in myeloma patients
This phase II study evaluated the impact of 18F-Fluorodeoxyglucose PET positivity after autologous stem cell transplantation (ASCT) in multiple myeloma patients. PET-positive patients achieving PET negativity after treatment displayed comparable outcomes to initially PET-negative patients. The study screened 159 post-ASCT patients with very good partial response (VGPR) or better. Of PET-positive patients, 57% were MRD-negative, indicating complementary response assessment. Carfilzomib-lenalidomide-dexamethasone (KRd) consolidation converted 33% of PET-positive patients to PET negativity, with better success among MRD-negative patients. Overall, PET effectively identified residual disease after ASCT, and KRd consolidation altered PET status in a subset of patients.
https://doi.org/10.1038/s41375-023-01998-7
Mezigdomide plus Dexamethasone in Relapsed and Refractory Multiple Myeloma
This study focused on evaluating mezigdomide, a novel cereblon E3 ubiquitin ligase modulator, in combination with dexamethasone for patients with relapsed and refractory multiple myeloma. In the phase 1 part of the study, 77 patients were enrolled, and the recommended phase 2 dose was determined to be 1.0 mg of mezigdomide given once daily with dexamethasone for 21 days, followed by 7 days off, in each 28-day cycle. In the phase 2 portion, 101 patients with triple-class–refractory multiple myeloma were treated. Common adverse events included neutropenia (77%) and infections (65%). An overall response rate of 41% was observed, with a median duration of response of 7.6 months and a median progression-free survival of 4.4 months. This combination demonstrated promising efficacy in heavily pretreated multiple myeloma patients, with manageable adverse effects primarily related to bone marrow suppression.
Weekly carfilzomib 70 mg/m2 and dexamethasone with or without cyclophosphamide in relapsed and/or refractory multiple myeloma patients
In this randomized phase II study for relapsed/refractory multiple myeloma (RRMM) patients with 1-3 prior lines of treatment, the combination of weekly carfilzomib, cyclophosphamide, and dexamethasone (KCd) was compared to carfilzomib and dexamethasone (Kd). Out of 197 patients, 97 received KCd, and 100 received Kd. Median progression-free survival (PFS) was similar at 19.1 months for KCd and 16.6 months for Kd. However, in a post hoc analysis of lenalidomide-refractory patients, the addition of cyclophosphamide to Kd significantly improved PFS to 18.4 months versus 11.3 months. Both groups achieved similar overall response rates and complete response rates (around 70% and 20%, respectively). Safety profiles were manageable, but severe infections were more common with the KCd combination. In conclusion, KCd did not improve outcomes overall but showed promise for lenalidomide-refractory patients in this RRMM study.
Single-agent belantamab mafodotin versus pomalidomide plus low-dose dexamethasone in patients with relapsed or refractory multiple myeloma
Belantamab is a humanised anti-BCMA afucosylated monoclonal antibody. In the DREAMM-3 phase 3 study for relapsed or refractory multiple myeloma, belantamab mafodotin was compared to the standard of care, pomalidomide-dexamethasone. While belantamab mafodotin showed no statistically significant improvement in progression-free survival, it demonstrated a manageable safety profile. Thrombocytopenia and anaemia were the most common grade 3-4 adverse events for belantamab mafodotin, while neutropenia and anaemia were predominant for pomalidomide-dexamethasone. Serious adverse events occurred in both groups, but there were no treatment-related deaths with belantamab mafodotin. Further research is ongoing to explore belantamab mafodotin in combination regimens for multiple myeloma.
https://doi.org/10.1016/S2352-3026(23)00243-0
Efficacy and safety of isatuximab plus bortezomib, lenalidomide, and dexamethasone in patients with newly diagnosed multiple myeloma ineligible/with no immediate intent for autologous stem cell transplantation
This Phase 1b study assessed the combination of isatuximab with bortezomib-lenalidomide-dexamethasone (Isa-VRd) in newly diagnosed multiple myeloma (NDMM) patients ineligible forASCT. Isatuximab is a chimeric monoclonal antibody targeted against surface CD38 glycoproteins. Among 73 patients, the overall response rate was 98.6%, with 56.3% achieving a complete response or better. Minimal residual disease negativity (10^-5 sensitivity) was attained by 50.7% of patients. Grade ≥3 treatment-emergent adverse events occurred in 79.5% of patients, but discontinuation of study treatment due to adverse events was observed in 19.2% of patients.
Post-transplant relapse prediction score in multiple myeloma
This study aimed to predict early relapse (ER) following Autologous Hematopoietic Cell Transplantation (AHCT) and developed a novel scoring system based on various factors. A total of 14,367 AHCT-1 patients were transplanted between 2014 and 2019, with training and validation cohorts. The scoring system assigned points for factors including ISS stages, disease status, and Karnofsky performance score. The risk score distribution ranged from 0 to ≥4 points. The lowest risk group had a 12-month progression-free survival (PFS-12) of 91.7%, while the highest risk group had a PFS-12 of 57.1%.
Pomalidomide and dexamethasone until progression after first salvage therapy in multiple myeloma
This study investigated the use of pomalidomide as a maintenance therapy in multiple myeloma patients in their first relapse. Patients received pomalidomide, cyclophosphamide, and dexamethasone salvage therapy, followed by transplantation or consolidation cycles. Out of 100 patients, 75 reached therapy and showed a median treatment duration of 23.7 months. One third of the patients experienced an improved treatment response, with median progression-free survival of 33.2 months. Adverse events, mainly hematological, were observed, but long-term administration of pomalidomide and dexamethasone was feasible and demonstrated the potential for response improvement in some patients.
Lenalidomide and dexamethasone maintenance with or without ixazomib, tailored by residual disease status in myeloma
In a study from November 2014 to May 2017, 332 patients treated with bortezomib, lenalidomide, and dexamethasone (VRD) were randomized for maintenance therapy with lenalidomide and dexamethasone (RD) or RD plus ixazomib (IRD) after autologous stem cell transplant and VRD consolidation. After a median follow-up of 69 months, progression-free survival (PFS) was similar between RD and IRD groups (61.3% vs. 55.6% at 6 years), supporting the efficacy of RD maintenance. Discontinuation of maintenance in patients with negative measurable residual disease (MRD) at 2 years showed low progression rates (17.2% at 4 years), even in those with high-risk features.
The MASTER trial investigated the use of daratumumab, carfilzomib, lenalidomide, and dexamethasone (Dara-KRd) therapy in newly diagnosed multiple myeloma patients, where minimal residual disease (MRD) status was utilized to modulate treatment duration and cessation. The study included 123 participants, and after the induction phase followed by autologous stem-cell transplantation and consolidation with Dara-KRd, 81% achieved MRD negativity. Of those, 71% reached MRD-SURE and treatment cessation. Progression-free survival at 36 months varied based on the number of high-risk chromosome abnormalities (HRCAs). While participants without HRCAs had an 88% progression-free survival, those with two or more HRCAs had a 50% progression-free survival. Notably, 61 participants remain free of therapy and MRD-negative as of February 7, 2023.
Thromboprophylaxis in patients with multiple myeloma
The meta-analysis comparing direct oral anticoagulants (DOACs) with aspirin for primary thromboprophylaxis in multiple myeloma patients undergoing anti-myeloma therapy (n=1026) found that DOACs were associated with a significantly lower incidence of venous thromboembolism (VTE) . Major bleeding, clinically relevant non-major bleeding, and minor bleeding event rates did not significantly differ between DOACs and aspirin. While DOACs may be preferable for MM-related thrombosis prevention, further research is needed to establish optimal thromboprophylaxis strategies, considering heterogeneous baseline characteristics and potential bias from observational studies.
Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone for the Treatment of High-Risk Newly Diagnosed Multiple Myeloma
The GMMG-CONCEPT trial investigated the combination of isatuximab, carfilzomib, lenalidomide, and dexamethasone (Isa-KRd) in patients with newly diagnosed multiple myeloma (NDMM) with exclusively high-risk disease. The trial enrolled both transplant-eligible (TE) and transplant-noneligible (TNE) patients with high-risk cytogenetic aberrations (HRCAs). The study aimed to induce minimal residual disease (MRD) negativity. The interim analysis (IA) included 125 patients with high-risk NDMM. The trial met its primary endpoint, with MRD negativity rates after consolidation of 67.7% in TE patients and 54.2% in TNE patients. MRD negativity was sustained for ≥1 year in 62.6% of patients. With a median follow-up of 44 months for TE patients and 33 months for TNE patients, median progression-free survival (PFS) was not reached in either arm. The combination of Isa-KRd demonstrated efficacy in inducing sustainable MRD negativity in patients with high-risk NDMM, regardless of transplant status, and showed promising PFS outcomes.
Elotuzumab plus pomalidomide and dexamethasone in relapsed/refractory multiple myeloma
The real-world study involving 200 cases of relapsed/refractory multiple myeloma (RRMM) treated with the combination of elotuzumab, pomalidomide, and dexamethasone (EloPd) in 35 Italian centers outside of clinical trials provides insights into the treatment's efficacy and safety. The overall response rate was 55.4%, consistent with the pivotal trial results. The toxicity profile in the real-world cohort was similar to the ELOQUENT-3 trial, with no significant differences between younger and older patients.
Daratumumab, Bortezomib, Lenalidomide, and Dexamethasone for Multiple Myeloma
In a phase 3 trial involving 709 transplantation-eligible patients with newly diagnosed multiple myeloma, the efficacy of subcutaneous daratumumab combined with bortezomib, lenalidomide, and dexamethasone (D-VRd) was compared to VRd alone. The D-VRd group showed a lower risk of disease progression or death, with an estimated 84.3% progression-free survival at 48 months, compared to 67.7% in the VRd group. The D-VRd group also exhibited higher rates of complete response (87.9% vs. 70.1%) and minimal residual disease (MRD)-negative status (75.2% vs. 47.5%). The addition of subcutaneous daratumumab demonstrated a significant benefit in progression-free survival for transplantation-eligible patients with newly diagnosed multiple myeloma.
Idecabtagene vicleucel chimeric antigen receptor T-cell therapy for relapsed/refractory multiple myeloma with renal impairment
Idecabtagene vicleucel (ide-cel) is a B-cell-maturation antigen (BCMA)-directed chimeric antigen receptor T cell therapy. Patients with relapsed multiple myeloma and renal impairment treated with ide-cel showed comparable safety and efficacy outcomes to those without renal impairment. While rates of cytokine release syndrome and neurotoxicity were similar between groups, patients with renal impairment had a higher incidence of short-term severe cytopenias. However, renal function did not deteriorate following CAR T-cell therapy, and response rates and survival outcomes were comparable between the two groups, highlighting the feasibility of ide-cel treatment in patients with renal impairment.
Melflufen plus dexamethasone and daratumumab or bortezomib in relapsed/refractory multiple myeloma
The ANCHOR study assessed melflufen (Melphalanflufenamide, alkylating peptide-drug conjugate) in combination with dexamethasone and daratumumab or bortezomib in triple-class refractory multiple myeloma (MM). No dose-limiting toxicities were observed, and thrombocytopenia and neutropenia were the most common grade ≥3 adverse events. In the daratumumab arm, the overall response rate was 73%, with a median progression-free survival of 12.9 months, while in the bortezomibarm, the overall response rate was 78%, with a median progression-free survival of 14.7 months. Based on these findings, melflufen 30 mg was determined as the recommended dose for future studies in relapsed/refractory MM when used with dexamethasone and daratumumab or bortezomib.
Best treatment strategy before autologous peripheral blood stem cell transplantation in POEMS syndrome
This study conducted in ten Italian centers evaluated the impact of prior therapies and mobilization regimen on outcomes in patients with POEMS syndrome undergoing autologous peripheral blood stem cell transplantation (aPBSCT) from 1998 to 2020. Patients were divided into three groups based on their treatment history before transplant. Regardless of prior treatment received, all 45 patients underwent aPBSCT after a high-dose melphalan conditioning regimen. The study found no statistically significant differences in responses, progression-free survival, overall survival, or transplant-related mortality among the three groups, suggesting that pre-treatment and disease status at transplant did not affect major transplant outcomes in patients with POEMS syndrome.
CAR-T and BITE therapies in multiple myeloma patients with prior allogeneic transplantation
In a cohort of 33 multiple myeloma (MM) patients with prior allogeneic hematopoietic cell transplantation (allo-HCT), chimeric antigen receptor T-cell (CAR-T) therapy (n = 24) demonstrated an overall response rate (ORR) of 92%, with 67% achieving a stringent complete response (CR), and 73% achieving minimal residual disease (MRD) negativity. Bispecific T-cell engagers (BITE) therapy (n = 9) resulted in an ORR of 44%, with 44% achieving a CR, and 44% achieving MRD negativity. Safety analysis revealed grade ≥3 adverse events (AEs) in 92% of CAR-T and 56% of BITE recipients, with cytokine release syndrome (CRS) occurring in 83% of CAR-T and 78% of BITE recipients. Notably, no exacerbation of graft-versus-host disease occurred except in one BITE recipient.
Teclistamab in Relapsed/ Refractory multiple myeloma
Teclistamab, a B-cell maturation antigen (BCMA) × CD3 directed bispecific antibody. In a retrospective analysis of 123 patients with relapsed and refractory multiple myeloma (RRMM) treated with teclistamab at 18 German centers, outcomes were assessed to determine real-world efficacy and tolerability. Most patients had triple-class or penta-drug refractory disease, with a subset previously treated with BCMA-directed therapies. The overall response rate (ORR) was 59.3% with a median progression-free survival (PFS) of 8.7 months. Subgroup analyses revealed lower ORR and PFS in patients with extramedullary disease, ISS 3, and those previously treated with idecabtagenevicleucel (ide-cel) CAR-T cell therapy, although duration of response in ide-celpretreated patients was comparable to anti-BCMA naive patients. Adverse events primarily included infections and grade ≥3 cytopenias.
Artificial intelligence in prognosticating multiple myeloma
This study aimed to decipher and predict the diverse outcomes seen in newly diagnosed multiple myeloma (NDMM) patients by analyzing clinical, genomic, and therapeutic data from 1,933 individuals. Through this analysis, the researchers identified 12 distinct groups based on genomic drivers, surpassing previous classifications. They developed a predictive model called IRMMa, integrating clinical, genomic, and treatment variables, which outperformed existing prognostic models in accuracy. Key genomic features, such as 1q21 gain/amp and TP53 loss, contributed to the model's precision. The study underscores the importance of personalized therapeutic decisions in NDMM based on individualized risk assessment, potentially optimizing treatment outcomes.
Autologous-allogeneic versus autologous tandem stem cell transplantation and maintenance therapy with thalidomide for multiple myeloma patients
This clinical trial compared autologous-allogeneic tandem stem cell transplantation (alloTSCT) to autologous tandem transplantation (autoTSCT) followed by thalidomide maintenance therapy in patients with multiple myeloma. Among the 217 patients enrolled, 178 underwent a second transplant, with alloTSCT showing a non-significant trend towards better progression-free survival (PFS) at 4 years (47% vs. 35%, P=0.26) and a more substantial difference at 8 years (P=0.10). AlloTSCT significantly reduced relapse rates at 4 years (40% vs. 63%, P=0.04) and at 8 years (44% vs. 77%, P=0.002). However, overall survival at 4 and 8 years was similar between both groups (66% vs. 66% at 4 years; 52% vs. 50% at 8 years, P=0.91 and P=0.87, respectively).
ChatGPT as an educational resource for patients with multiple myeloma
This study evaluated ChatGPT's potential as an educational tool for multiple myeloma (MM) patients by assessing its responses to a 21-question questionnaire. ChatGPT demonstrated a high accuracy rate (95%), with no harmful responses, though one treatment-related question was flagged for incompleteness. Simplifying medical terminology and ensuring up-to-date, comprehensive responses were noted as areas for improvement. Overall, ChatGPT shows promise as a supplementary educational resource, enhancing patient understanding while emphasizing the need for ongoing refinements and human medical expertise.
Linvoseltamab for Treatment of Relapsed/Refractory Multiple Myeloma
In a phase I/II trial, linvoseltamab, a B-cell maturation antigen × CD3 bispecific antibody, was evaluated for safety and efficacy in relapsed/refractory multiple myeloma (RRMM) patients. Among 117 patients treated with the 200 mg dose, the overall response rate (ORR) was 71%, with 50% achieving a complete response or better. The median duration of response was 29.4 months. The most common adverse events were cytokine release syndrome, neutropenia, and infections, but these events were manageable. Linvoseltamab 200 mg showed deep and durable responses with an acceptable safety profile in heavily pretreated RRMM patients.
Pembrolizumab and low-dose, single-fraction radiotherapy for patients with relapsed or refractory multiple myeloma
This phase 2 trial at Winship Cancer Institute evaluated the safety and activity of pembrolizumab combined with low-dose radiotherapy (8 Gy/1 fx) in 25 patients with relapsed/refractory multiple myeloma. The primary outcome was acceptable toxicity, with no grade 4 or 5 adverse events reported. At 3 months, 32% of patients showed treatment benefits, including partial and complete responses, with no severe radiation-related toxicity.
Belantamab Mafodotin, Bortezomib, and Dexamethasone for Multiple Myeloma
This phase 3 trial compared belantamab mafodotin, bortezomib, and dexamethasone (BVd) versus daratumumab, bortezomib, and dexamethasone (DVd) in relapsed or refractory multiple myeloma. BVd significantly improved progression-free survival (36.6 vs. 13.4 months) and overall survival at 18 months (84% vs. 73%). BVd also led to higher rates of complete response and minimal residual disease negativity (25% vs. 10%). However, BVd was associated with more grade 3 or higher adverse events (95% vs. 78%), particularly ocular events. BVd demonstrated strong efficacy but with notable safety concerns.
Curative Strategy for High-Risk Smoldering Myeloma
This study evaluated a curative approach for high-risk smoldering myeloma using carfilzomib, lenalidomide, and dexamethasone (KRd), followed by autologous stem-cell transplantation (ASCT), consolidation, and maintenance. Among 90 patients, 62% achieved undetectable measurable residual disease (uMRD) post-ASCT, and 31% maintained uMRD at 4 years. The overall survival rate at 70 months was 92%, with 5 patients progressing to multiple myeloma. Neutropenia and infections were common adverse events. These findings suggest a promising outcome for early aggressive treatment in high-risk smoldering myeloma.
Efficacy and safety of teclistamab in patients with relapsed/refractory multiple myeloma after BCMA-targeting therapies
Teclistamab, a BCMA-directed bispecific antibody, showed promising efficacy and safety in heavily pretreated relapsed/refractory multiple myeloma (R/RMM) patients with prior anti-BCMA therapy, according to the MajesTEC-1 trial cohort. Among 40 patients with a median of six prior treatment lines, the overall response rate was 52.5%, with 47.5% achieving very good partial response or better. The median duration of response, progression-free survival, and overall survival were 14.8, 4.5, and 15.5 months, respectively. Common treatment-emergent adverse events included neutropenia, infections, and cytokine release syndrome, with infections being the most frequent grade ≥3 adverse events. These results support teclistamab as an effective option for this challenging patient population.
Anti-BCMA/GPRC5D bispecific CAR T cells in patients with relapsed or refractory multiple myeloma
This phase 1 trial evaluated anti-BCMA/GPRC5D bispecific CAR T cells in 21 patients with relapsed/refractory multiple myeloma, identifying 2.0 × 10⁶ CAR T cells per kg as the maximum tolerated dose. At this dose, no severe organ toxicities or grade ≥3 ICANS occurred, and cytokine release syndrome was mild (grades 1–2). The overall response rate was 92% at the MTD, with 75% achieving complete response or better and 81% attaining measurable residual disease negativity. These findings highlight the promising safety and efficacy of bispecific CAR T cells as a potential new treatment option for this challenging patient population.
Disclaimer: Information provided on this website is only for medical education purposes and not intended as medical advice. Although authors have made every effort to provide up-to-date information, the recommendations should not be considered standard of care. Responsibility for patient care resides with the doctors on the basis of their professional license, experience, and knowledge of the individual patient. For full prescribing information, including indications, contraindications, warnings, precautions, and adverse effects, please refer to the approved product label. Neither the authors nor publisher shall be liable or responsible for any loss or adverse effects allegedly arising from any information or suggestion on this website. This website is written for use of healthcare professionals only; hence person other than healthcare workers is advised to refrain from reading the content of this website.