Abstract
Multiple Myeloma (MM) is primarily a disease of old age with a median age of sixty-nine years at diagnosis. The development of novel therapies for induction and use of autologous stem cell transplantation has resulted in improved clinical outcomes and better quality of life for MM patients. Elderly patients, comprising the majority of MM population, have a higher incidence of age-related comorbidities, frailty and organ dysfunction which complicates the coordination of treatment and limits the selection of therapies. Even in the era of multiple chemotherapeutic options, the clinical heterogeneity of the myeloma patients’ demands personalized treatments which often require dose-adjustments or dose delays. The use of reduced-dose regimens and various comorbidity indices has improved clinical outcome and regimen tolerability in MM patients with renal, neurological and bone abnormalities. We focus on advancements in the treatment of multiple myeloma with the goal to guide clinicians towards patient-specific management.
Keywords: Multiple myeloma, personalized therapy, elderly, frailty, dose reduction, dose modifications, comorbidities
1. Introduction
Multiple Myeloma (MM), the second most common hematologic malignancy,(1) had an estimated incidence of more than 30,000 cases in the United States of America in 2018 (2). The median age for MM diagnosis is 69 years with the median age of death due to MM being 75 years (3). Increasing age, on one hand, confers a strong risk for acquiring malignancies, (mostly occurring in patients older than 65 years), on the other hand, increases the incidence of comorbidities and frailty risk (4). Presence of risk factors such as high-risk cytogenetics, advanced age, comorbidities and polypharmacy indicate management to be individualized to each patient’s unique needs (4).
Comorbidity, the presence of two or more distinct disease entities in the same individual, is common in elder (>65 years) patients. In the US, 27.5 % men and 26.9 % women of age > 65 years have three chronic conditions including arthritis, hypertension, and cancer (5). Nephropathy, neuropathy, venous thromboembolism, osteolytic bone lesions, anemia, increased risk of infections and hypercalcemia are the common MM-associated morbidities with increased prevalence in elderly patients (6). In addition, elderly patients have a higher incidence of diabetes mellitus (DM), hypertension (HTN), heart failure (HF), cardiac arrhythmia, stroke and hyperlipidemia in the same patient population (7). Moreover, factors like advanced patient age (≥ 75 years), renal failure at presentation, drug discontinuation secondary to toxicity and grade 3-4 treatment-related infectious, cardiac or gastrointestinal adverse events (AEs) are found to be associated with reduced overall survival (OS) in MM patients (8).
Published literature in the PubMed database and guidelines by National Comprehensive Cancer Network (NCCN),(9) International Myeloma Working Group (IMWG),(10) European Myeloma Network (EMN)(11) and Mayo Stratification for Myeloma and Risk-Adapted Therapy (mSMART)(12) highlight the importance of individualized management in comorbid elderly MM patients. The purpose of this review is to identify and summarize findings from original studies and guidelines for personalized therapy in the management of MM.
2. Selection of chemotherapeutic drug regimens in elderly MM patients
2.1. Frailty Assessment
Frailty is commonly assessed by the presence of a decrease in any three out of the following five parameters: weight, gait speed, hand-grip strength, self-reported energy and physical activity (13). Various assessment tools like Charlson Comorbidity Index (CCI), Activities of Daily Living (ADL) and Instrumental Activities of Daily Living (IADL) are used to stratify elderly MM patients on the basis of comorbidities and functional status. According to IMWG recommendations by Palumbo et al. (2015), age alone should not be used for dose-reduction. Utilization of the IMWG frailty score by considering other factors including medical comorbidities (assessed by CCI) and disabilities (assessed by ADL score and IADL score) for decision making in the elderly population is recommended. IMWG advised to categorize patients into groups of “fit” (cumulative IMWG frailty score = 0), “intermediate fit” (cumulative IMWG frailty score = 1) and “frail” (cumulative IMWG frailty score = 2-5 on the basis of age, CCI, ADL and IADL (10), as shown in Table 1 and is validated by many studies (10, 14).
Table 1:
IMWG and R-MCI classification of elderly MM patients according to fitness profile and frailty score
IMWG frailty score (2015) | ||
---|---|---|
Parameter | IMWG-frailty index points | |
Age (years) | ≤75 | 0 |
76- 80 | 1 | |
>80 | 2 | |
ADL | >4 | 0 |
≤4 | 1 | |
IADL | >5 | 0 |
≤5 | 1 | |
CCI | ≤1 | 0 |
>1 | 1 | |
Patient status | Cumulative IMWG frailty score | |
Fit | 0 | |
Intermediate-fit | 1 | |
Frail | 2-5 | |
R-MCI (2016) | ||
Parameter | R-MCI points | |
Age (years) | <60 | 0 |
60-69 | 1 | |
≥ 70 | 2 | |
KPS | 100% | 0 |
80-90% | 2 | |
<70% | 3 | |
eGFR (ml/min) | ≥60 | 0 |
<60 | 1 | |
Frailty* | Mild | 0 |
Moderate/Severe | 1 | |
Cytogenetics | Missing | 0 |
Favorable** | 0 | |
Unfavorable*** | 1 | |
Lung dysfunction | No/Mild | 0 |
Moderate/Severe | 1 | |
Patient status | Cumulative R-MCI frailty score | |
Fit | 0-3 | |
Intermediate-fit | 4-6 | |
Frail | 7-9 |
Abbreviations: ADL= Activities of daily life; CCI= Charlson Comorbidity Index; IADL= Instrumental activities of daily living; IMWG= International Myeloma Working Group; KPS Karnofsky Performance Status; R-MCI= Revised Myeloma Comorbidity Index.
Parameters: Karnofsky Index, Time Up/Go, IADL, Subjective fitness.
Hyperdiploidie, t(11; 14), NK, del(13q14)
t(4,14), t(14,16), t(14,20), del(17p), Hypodiploidie, c-myc, Chromosom1-aberrations
An IMWG pooled analysis by Palumbo et al. (2015) evaluated 869 newly diagnosed multiple myeloma (NDMM) patients in 3 clinical trials with a median age of 74 years, which evaluated the utility of IMWG frailty score. Patients were classified into fit, intermediate fit and frail categories based on the IMWG frailty score; the 3-year overall survival (OS) was 84% (95% CI 78-89 %), 76% (95% CI 67-82), and 57% (95% CI 45-68%) respectively. The 3-year progression free survival (PFS) was 48% (95% CI 41-56%) for fit patients, 41% (95% CI 32-49%) for intermediate fit and 33% (95% CI 25-41%) for frail patients. This propensity score validating the use of IMWG frailty score by showing its prognostic role and clinical predictability (10). Engelhardt et al. (2016) analyzed a cohort of 125 NDMM patients with a median age of 63 years who were classified into fit, intermediate fit and frail categories using IMWG frailty score. For these patients, 3-year OS was 91% (95% CI: 78-100%) for fit, 77% (95% CI: 55-95%) for intermediate-fit and 47% (95% CI: 26-67%) for frail patients (14). Engelhardt et al. (2017) developed a scoring system called revised Myeloma Comorbidity Index (R-MCI) for elderly MM patients on the basis of age, Karnofsky Performance Status (KPS), Estimated Glomerular Filtration Rate (eGFR), frailty, cytogenetics and lung dysfunction. Patients were classified into “fit” (R-MCI score = 0-3), “intermediate fit” (R-MCI score = 4-6) and “frail” (R-MCI score = 7-9). Eight hundred and one MM patients having the median age of 63 years, with 28% of patients aged 66-75 years and 13% aged > 75 years. 247, 446 and 108 of these patients were classified into fit, intermediate-fit and unfit categories based on R-MCI. The median overall survival rates of patients in these groups were 10.1 years, 4.4 years and 1.2 years; thereby validating the prognostic role of R-MCI for elderly MM patients (15). EMN also recommends IMWG frailty index and R-MCI as reliable parameters to classify elderly MM patients into fit, intermediate-fit and frail categories, with R-MCI having a relatively inferior outcome and a higher rate of treatment discontinuation (16).
2.2. Three drug versus two drug regimens in elderly myeloma patients
The FIRST trial performed by Benboubker et al. (2014) was the first randomized phase III study to compare the efficacy of two drug regimen containing lenalidomide and dexamethasone (Rd) with that of three-drug regimen containing melphalan, prednisone and thalidomide (MPT) in 1623 transplant-ineligible NDMM patients with median age of 73 years and 1531 (94.3%) patients with age ≥ 65 years. The patients were randomly assigned to three groups, 535 receiving continuous 28-day cycles of Rd until progression of the disease, 541 receiving 28-day cycles of Rd for 72 weeks (18 cycles) and 547 receiving MPT for 72 weeks. The median PFS was 25.5 months with continuous Rd, 20.7 months with 18 cycles of Rd, and 21.2 months with MPT. The OS at 4 years was 59% with continuous Rd, 56% with 18 cycles of Rd, and 51% with MPT. Eighty-five percent of patients in the continuous Rd group, 80% in the 18 cycle Rd group and 89% in the MPT group had at least one or more grade 3 or 4 AEs. This study showed a two-drug regimen of continuous Rd until disease progression having better efficacy and safety profile than MPT in transplant-ineligible patients, 94.3% of whom were ≥65 years (17). A phase 2 study by Larocca et al. (2016) in 152 NDMM patients aged ≥75 years, half of whom were frail, compared two-drug regimen of subcutaneous bortezomib with oral prednisone (VP) in 51 patients, three-drug regimen of VP plus cyclophosphamide (VCP) in 51 patients and three-drug regimen of VP plus melphalan (VMP) in 50 patients. Median PFS was 14.0 months, 15.2 months and 17.1 months while 2-year OS was 60%, 70%, and 76%, in patients receiving VP, VCP, and VMP. Twenty-two percent of patients treated with VP, 37% with VCP and 33% with VMP experienced at least one non-hematologic drug-related AE. The rate of drug discontinuation secondary to AEs was 12%, 14%, and 20% and the rate of deaths related to toxicity in 6 months was 4%, 4% and 8% with VP, VCP, and VMP respectively. No major difference in efficacy of VP, VCP and VMP regimens along with relatively better safety profile of VP compared to three-drug regimens favors the benefit of using two-drug regimen in very elderly and frail NDMM patients (18). A study performed by Magarotto et al. (2016) with 662 NDMM patients, who were either aged ≥ 65 years or ineligible for ASCT, compared the efficacy of three-drug vs. two-drug regimens containing lenalidomide. Patients were randomized into three groups, two groups receiving three-drug regimens melphalan-prednisone-lenalidomide (MPR) or cyclophosphamide-prednisone-lenalidomide (CPR) respectively and one group receiving two-drug regimen lenalidomide plus low-dose dexamethasone (Rd). A post hoc analysis classifying patients into fit, intermediate fit and frail on the basis of IMWG frailty score found 4 year OS to be 77% and 57% in fit patients receiving three-drug and two-drug regimens respectively while no significant benefit in 4 years OS with three-drug regimen was seen in intermediate fit and frail patients,(19) suggesting improved efficacy of three-drug regimens only in fit elderly patients. Another study by Bonomo et al. (2016) with 117 MM patients with a median age of 75 years (range 70-95) and significant comorbidities including a patient group with 36% cardiac, 20% renal and 5% pulmonary involvement. They compared the efficacy of bortezomib-based two drug and three drug regimens. Patients receiving RVD (n=34) showed an ORR of 94% and median PFS of 36 months as compared to the patients on VD (n=17) showing an ORR and median PFS of 65% and 24 months respectively (20). The 34 patients receiving RVD received attenuated, not full doses, of this regimen which favors dose reduction in elderly MM patients.
The latest EMN guidelines by Larocca et al. (2018) recommend to treat elderly MM patients based on their classification into “fit”, “intermediate fit” and “unfit” as directed by IMWG frailty score and R-MCI with primary goal of treatment being achieving deep response, balancing efficacy and toxicity, and conservative approach along with low toxicity in fit, intermediate fit, and frail patients, respectively. It suggests the use of ASCT along with full dose of triplet or doublet regimens in fit patients, full-dosed doublet or reduced-dose triplet regimens in intermediate fit patients and reduced-dose doublet regimens along with palliative and supportive care in frail patients (16).
3. Autologous Stem-Cell Transplantation (ASCT) in elderly MM patients
Almost all MM guidelines and studies consider patients aged ≤ 65 years suitable for ASCT, as evident from the phase III clinical trials (21, 22). In the recent years, many clinical studies performed regarding the clinical outcome of ASCT in MM patients aged > 65 years have shown promising results. Auner et al. (2015) compared the results of different studies regarding ASCT in elderly MM patients and established that eligibility for ASCT should not be decided on the basis of chronological age alone, other factors like overall health, functional reserve, and drug tolerability profile should also be considered (23). A study by Shah et al. (2015) for determining the cost-effectiveness of ASCT in MM patients aged ≥ 65 years compared the clinical outcome of ASCT by measuring the mOS and survival rate at five years in two groups of 270 MM patients each, one group received ASCT while the other did not. After a median follow-up of 3.5 years, mOS was 58 months vs. 37 months (p < 0.001) and 3-year survival rate was 73% vs. 50% (p < 0.001) in the ASCT group and non-ASCT group, respectively (24). A retrospective analysis by Desai et al. compared the toxicity of full dose melphalan (200 mg/m2) among two groups of ASCT patients, age <65 years (n=47) and age 65-69 years (n=49). No significant difference (p-value > 0.4) between cardiac, renal and hematological toxicity (infections, neutropenic fever) was seen between the two groups. The overall survival (OS) at 2 years and median progression free survival (mPFS) at 1.3 years were 88% and 60.5% respectively in these patients (n=68) with a median age of 67 years (25).
Bashir et al. (2011) analyzed the safety of ASCT in MM patients (n=84) age > 70 years with 21% of the patients ≥ 75 years of age. The 5 year OS and PFS was 67% (95% CI 54 – 82%) and 27 % (95% CI 16 – 44%), respectively. Three different doses of melphalan (140, 180 and 200 mg/m2) were used for conditioning regimen. No significant difference between response rate among the three doses (88%, 90%, and 82%) was noted. ASCT, if used as first line therapy, improves OS compared to use in patients with relapsed refractory disease (83% vs. 41% at 5 years, p-value=0.001) (26). Garderet et al. (2016) performed a study to analyze the efficacy and tolerability of high dose melphalan (200 mg/m2) followed by ASCT after induction treatment in 56 NDMM patients age > 65 years. Among 56 patients, 6 were not able to receive ASCT after induction due to different reasons but were considered for post-transplant analysis on the basis of intention-to-treat. Ten out of 56 patients received low-dose melphalan (140 mg/m2) while the remaining 46 patients received high dose melphalan (200 mg/m2). After a median follow up of 21 months, 2-year PFS and OS rates were 76% (CI 61.6-94.1%) and 88% (CI 76.7-100%). PFS was found to be relatively better in patients receiving high-dose melphalan (27). Straka et al. (2016) performed a multicenter trial on 434 NDMM patients randomized to 2 groups receiving ASCT, one with induction therapy and the other without induction. In 420 patients evaluable at the end of study for PFS and OS, the difference in median PFS and median OS was statistically insignificant in patients aged <65 years and ≥65 years. Median PFS was found to be 19.5 months and 22.1 months in patients aged <65 years and ≥65 years respectively (p-value=0.23). Median OS was found to be 56.3 months and 53.1 months in patients aged <65 years and ≥65 years respectively (p-value=0.58) (28). Ozaki et al. (2014) retrospectively analyzed the clinical outcome of different treatment modalities in NDMM patients aged 65-70 years. Among 318 patients, 192 patients were treated with conventional chemotherapy regimens, 88 with novel chemotherapy regimens, 21 with conventional chemotherapy regimens plus auto-SCT and 17 with conventional chemotherapy regimens plus auto-SCT. Patients receiving auto-SCT showed statistically significant improvement in OS compared to those not receiving auto-SCT (OS being not reached vs. 57.9 months with p < 0.02 in both groups respectively), showing auto-SCT as an effective treatment option in eligible elderly MM patients aged 65-70 years (29). These studies suggest the feasibility of ASCT in elderly MM patients, making it a reliable therapeutic option in this patient population and don’t consider advanced patient age as exclusion criteria for ASCT. Latest EMN guidelines by Gay et al. (2018) recommend age <65 years, Karnofsky Performance Status (PS) > 90%, Hematopoietic Cell Transplantation-Specific Comorbidity Index (HCT-CI) = 0 and Revised-Myeloma Comorbidity Index (R-MCI) 0-3 as a cut-off for receiving high-dose melphalan (200 mg/m2) for all MM patients receiving ASCT. These also recommend high-dose melphalan (200 mg/m2) for those aged 65-70 years with Karnofsky PS > 90% and those having Karnofsky PS < 90% related to the MM itself instead of other comorbidities, and consideration of melphalan dose reduction for more advanced age, Karnofsky PS < 90%, HCT-CI >1 or R-MCI 4-6 (30).
4. Comorbidities and management of multiple myeloma
Gregersen et al. (2017) reported the presence of comorbid conditions like congestive heart failure (OR 2.8), chronic pulmonary disease (OR 1.7), renal dysfunction (OR 11) and DM with complications (OR 2.3) in myeloma patients compared to general population (p-value < 0.05) during the 1 year to 1 month before the diagnosis of MM (6). Management of MM patients with comorbidities is complicated because of drug-drug interactions and potential side effect profile of each of the prescription medication. The addition of a third factor, old age with frailty, adds unique challenges to the treatment strategy. There is potential for pharmacokinetic or pharmacodynamics interactions among medications for comorbidities and anti-myeloma drugs.
4.1. Renal impairment
Renal impairment (RI) is present in 20%-25% MM patients at the time of diagnosis (31) and 55% of patients develop RI during the course of their treatment (32). Common factors that lead to renal impairment include hypercalcemia of malignancy, dehydration, amyloidosis, tumor lysis syndrome, age-related decline in renal function and exposure to nephrotoxic medications (33, 34). Renal impairment requires dose adjustment for therapies according to the degree of renal function, use of alternatives to avoid nephrotoxic drugs, and plasmapheresis or dialysis if needed. Effective treatment of underlying pathology is the best management strategy for the complicated kidney dysfunction.
Bortezomib-based regimens are the mainstay of the treatment in patients with RI (30). Bortezomib is metabolized in the liver by deboronation, therefore, it does not require dose adjustment in patients with RI (35, 36). Dimopoulos et al. (2009, n=227) compared the efficacy of VMP (bortezomib, melphalan, prednisone) to MP in patients with renal dysfunction. Among 34 patients with CrCl < 30 ml/min, response rate with VMP was better (74%) than MP (47%). Improvement in GFR from < 50 ml/min to > 60 ml/min was seen in 44% patients on VMP than 34% patients on MP (37). Similarly, objective response rate (ORR) of 73% (Morabito et al., n=117 with n=82 having CrCl < 30 ml/min)(38) and 67% (Ponisch et al., n=36 with 16 patients on dialysis)(39) was reported in patients with renal impairment receiving bortezomib-based regimens. Lenalidomide, when used in combination with dexamethasone, in renal adjusted doses can improve renal function in about 40% of patients (40). Renal dose adjustment of drugs used in multiple myeloma patients is summarized in Table 2.
Table 2:
Dose modification of drugs used for the treatment of multiple myeloma according to Creatinine Clearance (CrCl)
Drug | > 60 ml/min | 30-59 ml/min | 15-29 ml/min | <15 ml/min | On dialysis |
---|---|---|---|---|---|
Melphalan PO, mg/kg/d | 0.15-0.25 | 0.11-0.19 | 0.11-0.19 | 0.0175-0.125 | 0.0175-0.125 |
Cyclophosphamide | 300 mg/m2 | No dose adjustment required | |||
Doxorubicin | 30 mg/m2 | No dose adjustment required | |||
Carmustine | 150-200 mg/m2 (single dose or divided in 2 days) | No dose adjustment required | Discontinue at CrCl < 10 | ||
Plerixafor | 0.24 mg/kg/d | ≤50 CrCl, 0.16 mg/kg/d | |||
Dexamethasone | 20-40 mg | No dose adjustment required | |||
Prednisone | According to regimen | No dose adjustment required | |||
Thalidomide | 50-200 mg/d | No dose adjustment required | |||
Lenalidomide | 25 mg/d | 10 mg/d | 15 mg/48hr | 5 mg/d | 5 mg/d, post-dialysis |
Pomalidomide | 4 mg/d | No dose adjustment required | NA | 3 mg/d post-dialysis | |
Bortezomib | 1.3 mg/m2 | No dose adjustment required | |||
Carfilzomib | According to regimen (20/27/56 mg/m2) | No dose adjustment required | |||
Ixazomib | 4 mg/d | No dose adjustment required | 3 mg/d | 3 mg | 3 mg, pre or post dialysis |
Panobinostat | 20 mg/d | No dose adjustment required | Not studied | ||
Daratumumab | 16 mg/kg | No dose adjustment required | |||
Elotuzumab | 10 mg/kg | No dose adjustment required | |||
Pamidronate | 90 mg monthly | No dose adjustment required | Not recommended | ||
Zoledronic acid (monthly) | 4 mg | 3 - 3.5 mg CrCl 50-60 ml/min: 3.5mg, 40-49: 3.3 mg, 30-39: 3 mg. | Not recommended | ||
Denosumab | 120 mg monthly | No dose adjustment required | |||
Enoxaparin | 40 mg/d | No dose adjustment required | 30 mg/d |
4.1.1. ASCT in patients with renal impairment
ASCT is feasible in patients with renal impairment and can result in significant improvement in renal function. Fakih et al. (2015) analyzed the role of ASCT in patients on dialysis (21 on hemodialysis and 3 on peritoneal dialysis). Three year OS and PFS were reported to be 64% and 36% respectively. Regarding improvement of the renal function, 32% of the patients showed improvement in GFR (by 25% from baseline) (41). Among 54 dialysis dependent patients, 13 (24%) patients did not require dialysis at a median of 4 months after transplant (Lee at al., 2004) (42).
4.1.2. Supportive treatment
Along with the selection of appropriate chemotherapeutic drug and its dose adjustment, supportive therapy is also necessary. IMWG recommends high fluid intake (>3L/day) during myeloma treatment. Fluid challenge can be given to patients who are anuric as a way to reverse renal dysfunction. Nephrotoxic drugs like aminoglycosides, NSAIDs, iodine contrast, furosemide should be avoided (43).
4.2. Bone disease
4.2.1. Bisphosphonates
Osteolytic bone disease is present in 70% to 80% of MM patients at the time of diagnosis (44). Risk for skeletal-related events (pathological fractures and spinal cord compression) is increased in MM patients with bone disease (45). Bisphosphonates (BPs) not only decrease the skeletal-related events (SRE) but may also provide survival benefits in MM (46, 47). While personalizing the use of bisphosphonates in MM patients, the factors that need to be considered include grade of bone disease, stage of MM, route of administration, duration of treatment, adverse effects of bisphosphonates like osteonecrosis of jaw (ONJ) and renal function status of the patient. NCCN guidelines suggest the use of pamidronate (PAM) or zoledronic acid (ZOL) in MM patients with active disease at all stages with monitoring for renal function (CrCl) and osteonecrosis of jaw (9). The mSMART guidelines recommend the initiation of BPs in patients with evidence of bone disease by conventional radiography whereas IMWG suggests the use of advanced imaging modalities like MRI, CT and PET scan in addition to plain radiographs for detection and monitoring of lytic lesions for BP therapy (48, 49). The recommended dose of zoledronic acid (4 mg) is infused over 15 minutes whereas that of pamidronate (90mg) is infused over 2 to 4 hours. EMN suggests that BPs should be administered once every 3 to 4 weeks; the duration of use is 2 years for PAM while ZOL should be used continuously (11). Mayo consensus statement recommends the monthly dosing schedule of BPs for the first year and then decrease dosing frequency to once every 3 months for the second year. If disease relapse occurs, monthly dosing can be restarted (50).
Adverse effects associated with BP use are electrolyte imbalance (hypocalcemia and hypophosphatemia), inflammatory reactions at the injection site, and acute-phase reactions after IV administration. Renal impairment and osteonecrosis of jaw are infrequent, but serious adverse effects associated with BPs. Hypocalcemia can be prevented with the daily administration of oral calcium and vitamin D3. Around 40% of myeloma patients have low vitamin D levels (51). Therefore, baseline and annual measurement of vitamin D level are recommended (11, 52).
4.2.2. Management of bone disease in renal impairment
ZOL and PAM can cause renal impairment leading to acute renal failure (53). Renal impairment depends on the concentration of BPs in bloodstream at a given time, therefore, rapid infusion rate and high dosage can potentially lead to renal failure. Established guidelines recommend the monitoring of renal function while administering BPs. Dose of ZOL should be reduced in patients with decreased creatinine clearance according to the manufacturer guidelines (Table 2) (50). PAM may be administered over extended duration (>4 hours). No change in the duration of infusion of ZOL is advised. PAM and ZOL should not be used in patients with creatinine clearance less than 30 ml/min (44). Denosumab, a monoclonal antibody, is recently approved by FDA for use in MM patients. A phase III trial with 1718 patients showed non-inferiority of denosumab over zoledronic acid for time to development of first skeletal-related event (Hazard ratio = 0.98, p-value=0.010) (54). Denosumab is injected subcutaneously in a dose of 120 mg every 4 weeks. It is not renally cleared and thus its use can be considered in patients with RI (CrCl < 30 ml/min) (55). In a recent study by Symonds et al., data revealed an increased risk of rebound vertebral compression fractures in patients who stop taking denosumab. A long-term follow up of participants (n=1001) of FREEDOM trial showed a six-fold increased risk of vertebral fractures from 1.2 per 100 participant years while on the drug to 7.1 after discontinuation of denosumab, The risks and benefits of denosumab use must be reviewed with the patient before initiating or discontinuing therapy (56).
4.2.3. Monitoring and management of osteonecrosis of jaw
Osteonecrosis of jaw (ONJ) is a serious complication resulting from the use of bisphosphonates and denosumab. It presents with exposed bone with tissue swelling, loosening of teeth and dental infections (57). Incidence of ONJ is 4% to 11% in patients on BP therapy and risk is particularly increased in patients with use of ZOL (58). Longer duration of BP therapy, poor dental hygiene, dental infections, and corticosteroids are risk factors that lead to the development of ONJ. Utilization of BPs for more than 3 years causes increased incidence of ONJ (7.7%) compared to use for around 1 year (1.5%) (59). Prophylactic use of antibiotics before dental procedures decreases the risk of ONJ significantly (p-value = 0.012) (60). EMN, IMWG, mSMART and NCCN guidelines advise patients to get complete dental exam before starting BP therapy (9, 11, 44, 50). The purpose of this exam is to identify any dental pathology that may need surgical treatment like dental extraction, incision & drainage or pulpectomy. Once BP therapy is started, if any dental problem arises it should be treated conservatively. If necessary, surgical intervention should be performed by an experienced maxillofacial surgeon. Physicians should stop BP one month before dental surgery and restart once complete healing has occurred (48).
4.2.4. Role of kyphoplasty, radiation, and surgery
Balloon kyphoplasty (BKP) is an effective technique for immediate pain relief lasting up to 2 years in patients with painful vertebral fractures. BKP has shown better functional outcomes than vertebroplasty and prevented disability in MM (61). For uncontrolled pain, imminent pathologic fracture or imminent spinal cord compression, surgical intervention or palliative radiotherapy (10-30 Gy) is recommended (9).
4.3. Venous thromboembolism
Venous Thromboembolism (VTE) is a broad term that includes deep vein thrombosis (DVT), pulmonary embolism (PE), superficial vein thrombosis (SVT) and thrombosis in other vessels. Factors that contribute to the development of VTE in MM are hyperviscosity induced by MM, immune modulators (lenalidomide, thalidomide, pomalidomide), steroids (dexamethasone) and patient related factors (62). There is a strong risk of the development of VTE in the first year following the diagnosis of myeloma (63). Therefore it is imperative to prescribe VTE prophylaxis to multiple myeloma patients with risk factors. Drugs commonly used for VTE prophylaxis are aspirin, low molecular weight heparin (LMWH) and warfarin. Novel anticoagulants like apixaban (factor X inhibitor) are in the trial phase to be used as primary prevention of VTE in myeloma patients on IMiDs (64). Selection of VTE prophylaxis according to individual risk factors is necessary. Individual risk factors include smoking, obesity (BMI ≥ 30 kg/m2), family history of VTE, comorbidities like renal insufficiency, diabetes, immobilization, cardiac disease, recent surgery, polycythemia, medications like tamoxifen, steroids and erythropoietin (11). The summary of VTE prophylaxis guidelines by European Myeloma Network (EMN) and National Comprehensive Cancer Network (NCCN) is given in Table 3.
Table 3:
Recommendation for VTE prophylaxis in MM patients on Immunomodulator drugs (IMiDs)
Risk factors (RF) | Recommended Therapy | |
---|---|---|
Individual RFa Myeloma-associated RFb |
NCCN, 2018 | EMN, 2015 |
No or only one risk factor | ||
Aspirin 81 to 325 mg/d | Aspirin 100 mg/d | |
≥ 2 risk factors | ||
LMWH or Warfarind |
LWMH (Switch to aspirin after 4 months) or Warfarin |
|
IMiD based regimensc | LMWH or Warfarind |
LWMH (Switch to aspirin after 4 months) or Warfarin |
Abbreviations: EMN=European Myeloma Network; IMiD= Immunomodulator drugs; LMWH= low molecular weight heparin; MM=multiple myeloma; NCCN=National Comprehensive Cancer Network; RF=risk factor.
Individual risk factors are described in the text.
Myeloma associated risk factors include diagnosis of myeloma and hyperviscosity.
Immunomodulator drugs (lenalidomide, pomalidomide, thalidomide) in combination with dexamethasone (>480 mg/month), doxorubicin or combination chemotherapy.
Target INR 2-3.
IMWG recommends the use of aspirin in patients with no or one risk factor. MM patients with multiple risk factors should receive LMWH or full dose warfarin (65). NCCN recommends either monotherapy (LMWH, UFH, factor Xa inhibitors) or regimens (combinations of above with warfarin, dabigatran) if VTE develops. Selection of regimen should be based on factors like renal impairment (CrCl < 30 ml/min), mode of administration, bleeding risk and cost (66). IMWG and EMN agree to discontinue chemotherapy temporarily and start therapeutic anticoagulation if acute VTE develops (11, 65).
4.4. Diabetes mellitus
Approximately 20% of the people above the age of 65 years in the United States are diagnosed with Diabetes Mellitus (DM) (67). Hyperglycemia in the MM population can be due to either preexisting DM or steroid induced caused by the use of dexamethasone. Uncontrolled diabetes can cause nephropathy potentiating the renal dysfunction caused by MM. Wu et al. (2014, n=1240) analyzed the impact of diabetes and its management on overall survival (OS) as primary endpoint. Diabetic MM patients had decreased median OS of 65.4 months compared to 98.7 months in patients without diabetes. Use of metformin was associated with increased OS compared to no use (74.3 vs. 60.1 months, p=0.034). On the other hand, use of insulin resulted in shorter OS compared to no use (57 vs. 101 months, p <0.001) (68). The possible mechanisms behind decreased OS due to insulin include induction of resistance to chemotherapy in cancer cells and activation of cell cycle signaling pathways resulting in growth of cancer cells (69). It is recommended to screen patients before starting treatment and monitor glucose levels during dexamethasone or prednisone therapy (70). Dose reduction of dexamethasone should be considered in patients in uncontrolled hyperglycemia and those at risk of developing diabetes associated macrovascular and microvascular complications. Standard dose of dexamethasone 40 mg once per week can be changed to 20 mg twice weekly to achieve better glucose control. Further reduction to 20 mg once per week to 10 mg once per week can be done to decrease toxicity (71).
4.5. Cardiovascular diseases
Multiple myeloma patients have been noted to have a higher incidence of cardiovascular diseases. One study reported that the incidence of cardiac adverse event is higher (Hazard Ratio = 2.2) in MM treated patients as compared to patients without the diagnosis (72). Xiao and colleagues (2014, n= 4330) reported the overall incidence of all grade cardiotoxicity of 4.3% in bortezomib monotherapy versus 3.5% in bortezomib-based combination therapy for all cancer types. An analysis of six randomized clinical trials with bortezomib resulted in a pooled odds ratio for all grade cardiotoxicity of bortezomib is 1.15 with p-value=0.41. This meta-analysis concluded that bortezomib does not increases the risk of cardiotoxicity during treatment (73).
Cardiac adverse effects like dyspnea (2.8% vs. 1.8%), hypertension (4.3% vs. 1.8%) and cardiac failure (3.8% vs. 1.8%) have a higher incidence with carfilzomib based treatments (74). Mushtaq et al. reported the range of cardiovascular side effects in patients for carfilzomib based combination regimens for hypertension at 3-25% and of heart failure at 3.4-20% (75). In 24 prospective clinical trials, a total of 2594 MM patients were treated with carfilzomib from a range of doses of 15 to 88 mg/m2. Various cardiovascular adverse events (CVAE) including hypertension, heart failure, cardiac arrhythmia, cardiac ischemia and cardiac arrest were assessed in these patients and reported as an aggregate outcome. All grade CVAE were reported in 617 (18.1%) patients and grade 3 or higher CVAE were seen in 274 (8.2%) patients. Increased rate of high-grade CVAE (11.9%) was reported in patients receiving increased dose of carfilzomib (≥45mg/m2) compared to 6.4% in patients receiving lower dose (< 45 mg/m2), p=0.02. These results indicated positive correlation between dose of carfilzomib and rate of CVAE (76). Dimopoulos et al. (2017, n=60) reported the incidence of cardiac event (left ventricle ejection fraction ≥ 20%) in 7 (12%) patients on carfilzomib combination regimens. Cardiac function returned to normal in all patients after a median of 60 days after drug discontinuation and appropriate treatment (77). Before starting carfilzomib, it is recommended to assess for baseline cardiac function, and manage preexisting hypertension and cardiac failure (78). In patients presenting with suspected signs and symptoms of cardiac dysfunction, carfilzomib and excessive fluids should be stopped and appropriate interventions and monitoring should be initiated. Consultation with a cardiologist should be considered for better management (79).
4.6. Peripheral neuropathy
MM patients have a relatively higher incidence of peripheral neuropathy (PN) resulting in increased risk of falls, neuropathic pain along with the higher incidence of loss of bowel/bladder control, ultimately causing functional impairment. About two-thirds of MM patients report pain including neuropathic pain (80) and more than half report varying intensities of PN at diagnosis (81). Neurological assessment tools including the ‘Total Neuropathy Score’(82) and ‘National Cancer Institute (NCI) Common Toxicity Criteria’(83) can be used in elderly MM patients at diagnosis to decide suitable treatment and assessment should be repeated periodically during the course of treatment to determine the need for changing therapy. MM patients with pre-existing PN at diagnosis need suitable drug selection and dose reduction for treatment because of relatively commonly reported bortezomib-induced PN (BiPN) and thalidomide-induced PN (TiPN) (84). In a randomized controlled trial (RCT) by Palumbo et al. (2010), 511 elderly patients with newly diagnosed MM (NDMM) were randomly allocated to two groups, one receiving VMPT-VT and the other VMP. Initially, 134 patients in both groups were given twice-weekly bortezomib. Halfway through the trial, bortezomib dosing was reduced from twice-weekly to once-weekly in remaining 369 patients in both groups. In once-weekly vs. twice-weekly bortezomib arms, the rate of grade 3-4 non-hematological toxicities was 51% vs. 36% (p=0.003) and the rate of grade 3-4 sensory PN was 16% vs. 3% (p<0.001) respectively without any significant change in clinical outcome in both groups (85). In a phase III trial, 222 relapsed MM patients (half aged > 65 years) were randomized to receive either intravenous (IV) or subcutaneous (SC) bortezomib and the rate of grade 3 PN was 15% vs. 5% respectively,(86) suggesting better safety profile of SC bortezomib than IV bortezomib with no significant difference in clinical outcome measured as PFS and OS between the two groups. IMWG (87) and EMN (11) recommendations for dose-reduction in patients with drug-induced neuropathy is summarized in Table 4. The risk of peripheral neuropathy is lower as compared to the IMiDs such as lenalidomide (1.7%),(88) and pomalidomide (< 1% – 2% grade 3/4 PN) (89). Among proteasome inhibitors, the incidence of grade 3 treatment-induced PN with carfilzomib was 1.3% (89) and ixazomib-related grade 3 PN occurred in 2% of patients (90).
Table 4:
Recommended dose modifications in patients with peripheral neuropathy
Bortezomiba | Grade of PN | G1 | G1 (painful) or G2 | G2 (painful) or G3 | |
IMWGc | Twice-weekly | ↓ to 1 mg/m2 or shift to once-weekly | D/C | ||
Once-weekly | ↓ to 1mg/m2 | D/C temporarily or ↓ dose to 1mg/m2 | |||
EMNc | NA | ↓ to 0.7-1.0 mg/m2d | D/C till down-escalated to G1 Resume at 50% dose |
||
Thalidomideb | Grade of PN | G1 | G2 | G3-4 | |
IMWG | 50% dose reduction | D/C; Resume at 50% dose post resolution | D/C | ||
EMN | NA | 50% dose reduction | D/C till down-escalated to G1 Resume at 50% |
Abbreviations: D/C= discontinue; EMN=European Myeloma Network; G=grade; IMWG=International Myeloma Working Group; NA=not available; PN=peripheral neuropathy; ↓=decrease.
Dose of bortezomib is 1.3 mg/m2.
Dose of thalidomide is 100 mg.
Discontinue if G4 develops.
Preferably subcutaneous.
5. Conclusion
While making therapy decisions for MM patients, patient treatment should address a personalized approach with consideration for age, comorbidities, and frailty. In addition, ASCT should be considered in all MM patients based on their fitness profile and comorbidity index irrespective of the chronological age. Elderly patients, comprising the major part of MM patient population, are a clinically diverse group with specific and individual needs based on their drug tolerability profiles along with individual age-related and disease-related renal, neurological, endocrine and bone disorders. There is a need for clinical trials focusing on individualized treatment with special consideration for dose reduction and assessment of comorbidities in various subgroups of MM patients. More clinical trials should include very elderly MM patients as they are underrepresented in most of the completed clinical trials.
Highlights:
Myeloma patients need personalized therapy according to their comorbidities and age
Age, performance status and frailty guide the selection of treatment in elderly
Bortezomib regimens are the mainstay of therapy in patients with renal impairment
Denosumab should be used for bone disease in patients with CrCl < 30 ml/min
Doses of bortezomib and thalidomide should be reduced to manage neuropathy
Acknowledgments
Funding source
U.S. Department of Health & Human Services ∣ NIH ∣ National Cancer Institute (NCI) Grant number - P30 CA023074 [Faiz Anwer]
Biography
Muhammad Asad Fraz has his MD and is a graduate of King Edward Medical University (KEMU), Pakistan. Currently, he is working as research associate in Hematology/Oncology at University of Arizona Cancer Center (UACC).
Faiza Hassan Warraich has his MD and is a resident at Department of Internal Medicine, McLaren-Flint Medical Center, Flint, MI.
Sami Ullah Warraich has his MD and is a graduate of KEMU, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Muhammad Junaid Tariq has his MD and is a graduate of KEMU, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Zabih Warraich has his MD and is a resident at Department of Internal Medicine, United Health Services Wilson Memorial Regional Medical Center, Johnson City, NY.
Ali Younas Khan has his MD and is a graduate of Shifa College of Medicine, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Muhammad Usman has his MD and is a graduate of KEMU, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Awais Ijaz has his MD and is a graduate of Allama Iqbal Medical College, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Pavan Tenneti has his MD and is working as Clinical Assistant Professor at College of Medicine, University of Arizona.
Adeela Mushtaq has his MD and is a resident at Department of Medicine, University of Pittsburgh Medical Center, McKeesport.
Faisal Akbar has his MD and is working as Assistant Professor at Wake Forest Baptist Medical Center, North Carolina.
Zaina Shahid has her MD and working at Wilkes Medical Center - Wake Forest Baptist Health, North Carolina.
Zeeshan Ali has his MD and is working as Clinical Assistant Professor at College of Medicine, University of Arizona.
Hafiz Muhammad Fazeel has his MD and is a graduate of Services Institute of Medical Sciences, Pakistan. Currently, he is working as research associate in Hematology/Oncology at UACC.
Cesar Rodriguez has his MD and is working as Assistant Professor Hematology/Oncology at Wake Forest Baptist Medical Center, North Carolina.
Aboo Nasar has his MD and is working as geriatrician at Tri-City Medical Center, California.
Ali McBride has his PharmD and MS and is the Clinical Coordinator of Hematology/Oncology at UACC.
Faiz Anwer has his MD, is team leader for Multiple Myeloma program, Clinical Director of the adult Blood and Marrow Transplant Program and the Director of the Stem Cell Harvest Program in the Division of Hematology/Oncology at UACC and is an Associate Professor of Medicine at the University of Arizona, College of Medicine.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest
This manuscript is original research, has not been previously published and has not been submitted for publication elsewhere while under consideration. Authors declare no conflict of interest with this manuscript.
References:
- 1.Collins CD. Problems monitoring response in multiple myeloma. Cancer imaging : the official publication of the International Cancer Imaging Society. 2005;5 Spec No A:S119–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cancer Stat Facts: Myeloma 2018. [Available from: https://seer.cancer.gov/statfacts/html/mulmy.html.
- 3.Ma CKK, Clancy L, Simms R, Burgess J, Deo S, Blyth E, et al. Adjuvant Peptide Pulsed Dendritic Cell Vaccination in Addition to T Cell Adoptive Immunotherapy for Cytomegalovirus Infection in Allogeneic Hematopoietic Stem Cell Transplantation Recipients. Biology of Blood and Marrow Transplantation. 2018;24(1):71–7. [DOI] [PubMed] [Google Scholar]
- 4.Palumbo A, Bringhen S, Ludwig H, Dimopoulos MA, Blade J, Mateos MV, et al. Personalized therapy in multiple myeloma according to patient age and vulnerability: a report of the European Myeloma Network (EMN). Blood. 2011;118(17):4519–29. [DOI] [PubMed] [Google Scholar]
- 5.Ward BW, Schiller JS. Prevalence of multiple chronic conditions among US adults: estimates from the National Health Interview Survey, 2010. Preventing chronic disease. 2013;10:E65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gregersen H, Vangsted AJ, Abildgaard N, Andersen NF, Pedersen RS, Frolund UC, et al. The impact of comorbidity on mortality in multiple myeloma: a Danish nationwide population-based study. Cancer medicine. 2017;6(7):1807–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Beltrán-Sánchez H, Harhay MO, Harhay MM, McElligott S. Prevalence and trends of Metabolic Syndrome in the adult US population, 1999–2010. Journal of the American College of Cardiology. 2013;62(8):697–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bringhen S, Mateos MV, Zweegman S, Larocca A, Falcone AP, Oriol A, et al. Age and organ damage correlate with poor survival in myeloma patients: meta-analysis of 1435 individual patient data from 4 randomized trials. Haematologica. 2013;98(6):980–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kumar SK, Callander NS, Alsina M, Atanackovic D, Biermann JS, Chandler JC, et al. Multiple Myeloma, Version 3.2017, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network : JNCCN. 2017;15(2):230–69. [DOI] [PubMed] [Google Scholar]
- 10.Palumbo A, Bringhen S, Mateos MV, Larocca A, Facon T, Kumar SK, et al. Geriatric assessment predicts survival and toxicities in elderly myeloma patients: an International Myeloma Working Group report. Blood. 2015;125(13):2068–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Terpos E, Kleber M, Engelhardt M, Zweegman S, Gay F, Kastritis E, et al. European Myeloma Network guidelines for the management of multiple myeloma-related complications. Haematologica. 2015;100(10):1254–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mayo Stratification for Myeloma And Risk-adapted Therapy Newly Diagnosed Myeloma2016 April-19-2018. Available from: https://nebula.wsimg.com/e1520dd2009dae7c8ea5ca513775b8fa?AccessKeyId=A0994494BBBCBE4A0363&disposition=0&alloworigin=1.
- 13.Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older adults: evidence for a phenotype. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences. 2001;56(3):M146–M57. [DOI] [PubMed] [Google Scholar]
- 14.Engelhardt M, Dold SM, Ihorst G, Zober A, Moller M, Reinhardt H, et al. Geriatric assessment in multiple myeloma patients: validation of the International Myeloma Working Group (IMWG) score and comparison with other common comorbidity scores. Haematologica. 2016;101(9):1110–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Engelhardt M, Domm A-S, Dold SM, Ihorst G, Reinhardt H, Zober A, et al. A concise revised myeloma comorbidity index as a valid prognostic instrument in a large cohort of 801 multiple myeloma patients. Haematologica. 2017;102(5):910–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Larocca A, Dold SM, Zweegman S, Terpos E, Wasch R, D'Agostino M, et al. Patient-centered practice in elderly myeloma patients: an overview and consensus from the European Myeloma Network (EMN). Leukemia. 2018. [DOI] [PubMed] [Google Scholar]
- 17.Benboubker L, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M, et al. Lenalidomide and Dexamethasone in Transplant-Ineligible Patients with Myeloma. New England Journal of Medicine. 2014;371(10):906–17. [DOI] [PubMed] [Google Scholar]
- 18.Larocca A, Bringhen S, Teresa Petrucci M, Oliva S, Pia Falcone A, Caravita T, et al. A phase 2 study of three low-dose intensity subcutaneous Bortezomib regimens in elderly frail patients with untreated multiple myeloma 2016. [DOI] [PubMed] [Google Scholar]
- 19.Magarotto V, Bringhen S, Offidani M, Benevolo G, Patriarca F, Mina R, et al. Triplet vs doublet lenalidomide-containing regimens for the treatment of elderly patients with newly diagnosed multiple myeloma. Blood. 2016:blood-2015–08-662627. [DOI] [PubMed] [Google Scholar]
- 20.Bonomo L, Lue J, Jagannath S, Chari A. The outcomes of newly diagnosed elderly multiple myeloma patients treated at a single U.S. institution. Cancer medicine. 2016;5(3):500–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Palumbo A, Cavallo F, Gay F, Di Raimondo F, Ben Yehuda D, Petrucci MT, et al. Autologous Transplantation and Maintenance Therapy in Multiple Myeloma. New England Journal of Medicine. 2014;371(10):895–905. [DOI] [PubMed] [Google Scholar]
- 22.Gaynon PS. Treatment adherence and 6-mercaptopurine metabolites. Pediatric blood & cancer. 2006;46(2):120–1. [DOI] [PubMed] [Google Scholar]
- 23.Auner HW, Garderet L, Kroger N. Autologous haematopoietic cell transplantation in elderly patients with multiple myeloma. British journal of haematology. 2015;171(4):453–62. [DOI] [PubMed] [Google Scholar]
- 24.Shah GL, Winn AN, Lin PJ, Klein A, Sprague KA, Smith HP, et al. Cost-Effectiveness of Autologous Hematopoietic Stem Cell Transplantation for Elderly Patients with Multiple Myeloma using the Surveillance, Epidemiology, and End Results-Medicare Database. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2015;21(10):1823–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Desai A, Beitinjaneh A, Ramdial J, Ali R, Lekakis L, Pereira D, et al. Safety of High-Dose Melphalan (200 Mg/M2) as Conditioning for Autologous Stem Cell Transplantation for Myeloma in Elderly Patients. Biology of Blood and Marrow Transplantation. 2017;23(3):S133–S4. [Google Scholar]
- 26.Bashir Q, Shah N, Parmar S, Wei W, Rondon G, Weber DM, et al. Feasibility of autologous hematopoietic stem cell transplant in patients aged >/=70 years with multiple myeloma. Leukemia & lymphoma. 2012;53(1):118–22. [DOI] [PubMed] [Google Scholar]
- 27.Garderet L, Beohou E, Caillot D, Stoppa AM, Touzeau C, Chretien ML, et al. Upfront autologous stem cell transplantation for newly diagnosed elderly multiple myeloma patients: a prospective multicenter study. Haematologica. 2016;101(11):1390–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Straka C, Liebisch P, Salwender H, Hennemann B, Metzner B, Knop S, et al. Autotransplant with and without induction chemotherapy in older multiple myeloma patients: long-term outcome of a randomized trial. Haematologica. 2016;101(11):1398–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ozaki S, Harada T, Saitoh T, Shimazaki C, Itagaki M, Asaoku H, et al. Survival of multiple myeloma patients aged 65–70 years in the era of novel agents and autologous stem cell transplantation. A multicenter retrospective collaborative study of the Japanese Society of Myeloma and the European Myeloma Network. Acta haematologica. 2014;132(2):211–9. [DOI] [PubMed] [Google Scholar]
- 30.melphalnGay F, Engelhardt M, Terpos E, Wasch R, Giaccone L, Auner HW, et al. From transplant to novel cellular therapies in multiple myeloma: European Myeloma Network guidelines and future perspectives. Haematologica. 2018;103(2):197–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Knudsen LM, Hippe E, Hjorth M, Holmberg E, Westin J. Renal function in newly diagnosed multiple myeloma--a demographic study of 1353 patients. The Nordic Myeloma Study Group. European journal of haematology. 1994;53(4):207–12. [DOI] [PubMed] [Google Scholar]
- 32.Kyle RA, Gertz MA, Witzig TE, Lust JA, Lacy MQ, Dispenzieri A, et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clinic proceedings. 2003;78(1):21–33. [DOI] [PubMed] [Google Scholar]
- 33.Kleber M, Ihorst G, Deschler B, Jakob C, Liebisch P, Koch B, et al. Detection of renal impairment as one specific comorbidity factor in multiple myeloma: multicenter study in 198 consecutive patients. European journal of haematology. 2009;83(6):519–27. [DOI] [PubMed] [Google Scholar]
- 34.Kheder El-Fekih R, Izzedine H. [Lenalidomide nephrotoxicity]. Bulletin du cancer. 2016;103(5):499–506. [DOI] [PubMed] [Google Scholar]
- 35.Haynes R, Leung N, Kyle R, Winearls CG. Myeloma kidney: improving clinical outcomes? Advances in chronic kidney disease. 2012;19(5):342–51. [DOI] [PubMed] [Google Scholar]
- 36.Dimopoulos MA, Terpos E, Chanan-Khan A, Leung N, Ludwig H, Jagannath S, et al. Renal impairment in patients with multiple myeloma: a consensus statement on behalf of the International Myeloma Working Group. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(33):4976–84. [DOI] [PubMed] [Google Scholar]
- 37.Dimopoulos MA, Richardson PG, Schlag R, Khuageva NK, Shpilberg O, Kastritis E, et al. VMP (Bortezomib, Melphalan, and Prednisone) is active and well tolerated in newly diagnosed patients with multiple myeloma with moderately impaired renal function, and results in reversal of renal impairment: cohort analysis of the phase III VISTA study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(36):6086–93. [DOI] [PubMed] [Google Scholar]
- 38.Morabito F, Gentile M, Ciolli S, Petrucci MT, Galimberti S, Mele G, et al. Safety and efficacy of bortezomib-based regimens for multiple myeloma patients with renal impairment: a retrospective study of Italian Myeloma Network GIMEMA. European journal of haematology. 2010;84(3):223–8. [DOI] [PubMed] [Google Scholar]
- 39.Ponisch W, Moll B, Bourgeois M, Andrea M, Schliwa T, Heyn S, et al. Bendamustine and prednisone in combination with bortezomib (BPV) in the treatment of patients with relapsed or refractory multiple myeloma and light chain-induced renal failure. Journal of cancer research and clinical oncology. 2013;139(11):1937–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dimopoulos MA, Christoulas D, Roussou M, Kastritis E, Migkou M, Gavriatopoulou M, et al. Lenalidomide and dexamethasone for the treatment of refractory/relapsed multiple myeloma: dosing of lenalidomide according to renal function and effect on renal impairment. European Journal of Haematology. 2010;85(1):1–5. [DOI] [PubMed] [Google Scholar]
- 41.El Fakih R, Fox P, Popat U, Nieto Y, Shah N, Parmar S, et al. Autologous Hematopoietic Stem Cell Transplantation in Dialysis-Dependent Myeloma Patients. Clinical lymphoma, myeloma & leukemia. 2015;15(8):472–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lee CK, Zangari M, Barlogie B, Fassas A, van Rhee F, Thertulien R, et al. Dialysis-dependent renal failure in patients with myeloma can be reversed by high-dose myeloablative therapy and autotransplant. Bone marrow transplantation. 2004;33(8):823–8. [DOI] [PubMed] [Google Scholar]
- 43.Dimopoulos MA, Sonneveld P, Leung N, Merlini G, Ludwig H, Kastritis E, et al. International Myeloma Working Group Recommendations for the Diagnosis and Management of Myeloma-Related Renal Impairment. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(13):1544–57. [DOI] [PubMed] [Google Scholar]
- 44.Terpos E, Morgan G, Dimopoulos MA, Drake MT, Lentzsch S, Raje N, et al. International Myeloma Working Group recommendations for the treatment of multiple myeloma-related bone disease. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31(18):2347–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Coleman RE. Skeletal complications of malignancy. Cancer. 1997;80(8 Suppl):1588–94. [DOI] [PubMed] [Google Scholar]
- 46.Berenson JR, Lichtenstein A, Porter L, Dimopoulos MA, Bordoni R, George S, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. Myeloma Aredia Study Group. The New England journal of medicine. 1996;334(8):488–93. [DOI] [PubMed] [Google Scholar]
- 47.Morgan GJ, Child JA, Gregory WM, Szubert AJ, Cocks K, Bell SE, et al. Effects of zoledronic acid versus clodronic acid on skeletal morbidity in patients with newly diagnosed multiple myeloma (MRC Myeloma IX): secondary outcomes from a randomised controlled trial. The Lancet Oncology. 2011;12(8):743–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lacy MQ, Dispenzieri A, Gertz MA, Greipp PR, Gollbach KL, Hayman SR, et al. Mayo clinic consensus statement for the use of bisphosphonates in multiple myeloma. Mayo Clinic proceedings. 2006;81(8):1047–53. [DOI] [PubMed] [Google Scholar]
- 49.Durie BG. Use of bisphosphonates in multiple myeloma: IMWG response to Mayo Clinic consensus statement. Mayo Clinic proceedings. 2007;82(4):516–7; author reply 7–8. [DOI] [PubMed] [Google Scholar]
- 50.Bisphosphonates in Myeloma Mayo Consensus2012 April 20, 2018. Available from: https://nebula.wsimg.com/cbc2657c0a9d03db85556abfa41a2d29?AccessKeyId=A0994494BBBCBE4A0363&disposition=0&alloworigin=1.
- 51.Badros A, Goloubeva O, Terpos E, Milliron T, Baer MR, Streeten E. Prevalence and significance of vitamin D deficiency in multiple myeloma patients. British journal of haematology. 2008;142(3):492–4. [DOI] [PubMed] [Google Scholar]
- 52.Calcium and Vitamin D Supplementation in Myeloma Mayo Consensus2012 April 17, 2018. Available from: https://nebula.wsimg.com/4b8f7aa16474a6bad27a00fa84d90d6f?AccessKeyId=A0994494BBBCBE4A0363&disposition=0&alloworigin=1.
- 53.Morgan GJ, Davies FE, Gregory WM, Cocks K, Bell SE, Szubert AJ, et al. First-line treatment with zoledronic acid as compared with clodronic acid in multiple myeloma (MRC Myeloma IX): a randomised controlled trial. Lancet (London, England). 2010;376(9757):1989–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Raje N, Terpos E, Willenbacher W, Shimizu K, Garcia-Sanz R, Durie B, 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. 2018;19(3):370–81. [DOI] [PubMed] [Google Scholar]
- 55.Anderson K, Ismaila N, Flynn PJ, Halabi S, Jagannath S, Ogaily MS, et al. Role of Bone-Modifying Agents in Multiple Myeloma: American Society of Clinical Oncology Clinical Practice Guideline Update. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2018;36(8):812–8. [DOI] [PubMed] [Google Scholar]
- 56.Symonds C, Kline G. Warning of an increased risk of vertebral fracture after stopping denosumab. CMAJ : Canadian Medical Association journal = journal de l'Association medicale canadienne. 2018;190(16):E485–e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kalra S, Jain V. Dental complications and management of patients on bisphosphonate therapy: A review article. Journal of Oral Biology and Craniofacial Research. 2013;3(1):25–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Dimopoulos M, Kastritis E, Anagnostopoulos A, Melakopoulos I, Gika D, Moulopoulos L, et al. Osteonecrosis of the jaw in patients with multiple myeloma treated with bisphosphonates: evidence of increased risk after treatment with zoledronic acid. Haematologica. 2006;91(7):968–71. [PubMed] [Google Scholar]
- 59.Bamias A, Kastritis E, Bamia C, Moulopoulos LA, Melakopoulos I, Bozas G, et al. Osteonecrosis of the jaw in cancer after treatment with bisphosphonates: incidence and risk factors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23(34):8580–7. [DOI] [PubMed] [Google Scholar]
- 60.Montefusco V, Gay F, Spina F, Miceli R, Maniezzo M, Teresa Ambrosini M, et al. Antibiotic prophylaxis before dental procedures may reduce the incidence of osteonecrosis of the jaw in patients with multiple myeloma treated with bisphosphonates. Leukemia & lymphoma. 2008;49(11):2156–62. [DOI] [PubMed] [Google Scholar]
- 61.Bouza C, López-Cuadrado T, Cediel P, Saz-Parkinson Z, Amate JM. Balloon kyphoplasty in malignant spinal fractures: a systematic review and meta-analysis. BMC Palliative Care. 2009;8:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Klovaite J, Benn M, Nordestgaard BG. Obesity as a causal risk factor for deep venous thrombosis: a Mendelian randomization study. Journal of Internal Medicine. 2015;277(5):573–84. [DOI] [PubMed] [Google Scholar]
- 63.Kristinsson SY, Fears TR, Gridley G, Turesson I, Mellqvist UH, Bjorkholm M, et al. Deep vein thrombosis after monoclonal gammopathy of undetermined significance and multiple myeloma. Blood. 2008;112(9):3582–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Apixaban for Primary Prevention of Venous Thromboembolism in Patients With Multiple Myeloma [Available from: https://ClinicalTrials.gov/show/NCT02958969. [DOI] [PMC free article] [PubMed]
- 65.Palumbo A, Rajkumar SV, San Miguel JF, Larocca A, Niesvizky R, Morgan G, et al. International Myeloma Working Group consensus statement for the management, treatment, and supportive care of patients with myeloma not eligible for standard autologous stem-cell transplantation. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32(6):587–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Cancer-Associated Venous Thromboembolic Disease, Version 1.2018. 2018 April 20, 2018. Available from: https://www.nccn.org/professionals/physiciangls/pdf/vte.pdf. [DOI] [PubMed]
- 67.National Diabetes Statistics Report, 2017. [Google Scholar]
- 68.Wu W, Merriman K, Nabaah A, Seval N, Seval D, Lin H, et al. The association of diabetes and anti-diabetic medications with clinical outcomes in multiple myeloma. British journal of cancer. 2014;111(3):628–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Feng YH, Velazquez-Torres G, Gully C, Chen J, Lee MH, Yeung SC. The impact of type 2 diabetes and antidiabetic drugs on cancer cell growth. Journal of cellular and molecular medicine. 2011;15(4):825–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ahmed YA, Eltayeb A. Clinical challenges: myeloma and concomitant type 2 diabetes. International journal of hematology-oncology and stem cell research. 2013;7(1):34–41. [PMC free article] [PubMed] [Google Scholar]
- 71.Wildes TM, Rosko A, Tuchman SA. Multiple Myeloma in the Older Adult: Better Prospects, More Challenges. Journal of Clinical Oncology. 2014;32(24):2531–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kistler KD, Kalman J, Sahni G, Murphy B, Werther W, Rajangam K, et al. Incidence and Risk of Cardiac Events in Patients With Previously Treated Multiple Myeloma Versus Matched Patients Without Multiple Myeloma: An Observational, Retrospective, Cohort Study. Clinical lymphoma, myeloma & leukemia. 2017;17(2):89–96.e3. [DOI] [PubMed] [Google Scholar]
- 73.Xiao Y, Yin J, Wei J, Shang Z. Incidence and risk of cardiotoxicity associated with bortezomib in the treatment of cancer: a systematic review and meta-analysis. PloS one. 2014;9(l):e87671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Stewart AK, Rajkumar SV, Dimopoulos MA, Masszi T, Špička I, Oriol A, et al. Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. New England Journal of Medicine. 2015;372(2):142–52. [DOI] [PubMed] [Google Scholar]
- 75.Mushtaq A, Kapoor V, Latif A, Iftikhar A, Zahid U, McBride A, et al. Efficacy and toxicity profile of carfilzomib based regimens for treatment of multiple myeloma: A systematic review. Critical reviews in oncology/hematology. 2018;125:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Waxman AJ, Clasen S, Hwang WT, Garfall A, Vogl DT, Carver J, et al. Carfilzomib-Associated Cardiovascular Adverse Events: A Systematic Review and Meta-analysis. JAMA oncology. 2018;4(3):e174519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Dimopoulos MA, Roussou M, Gavriatopoulou M, Psimenou E, Ziogas D, Eleutherakis-Papaiakovou E, et al. Cardiac and renal complications of carfilzomib in patients with multiple myeloma. Blood advances. 2017;1(7):449–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Mikhael J Management of Carfilzomib-Associated Cardiac Adverse Events. Clinical lymphoma, myeloma & leukemia. 2016;16(5):241–5. [DOI] [PubMed] [Google Scholar]
- 79.Jakubowiak AJ, DeCara JM, Mezzi K. Cardiovascular events during carfilzomib therapy for relapsed myeloma: practical management aspects from two case studies. Hematology (Amsterdam, Netherlands). 2017;22(10):585–91. [DOI] [PubMed] [Google Scholar]
- 80.Kariyawasan C, Hughes D, Jayatillake M, Mehta A. Multiple myeloma: causes and consequences of delay in diagnosis. QJM: An International Journal of Medicine. 2007;100(10):635–40. [DOI] [PubMed] [Google Scholar]
- 81.Plasmati R, Pastorelli F, Cavo M, Petracci E, Zamagni E, Tosi P, et al. Neuropathy in multiple myeloma treated with thalidomide. Neurology. 2007;69(6):573.17679676 [Google Scholar]
- 82.Cavaletti G, Frigeni B, Lanzani F, Piatti M, Rota S, Briani C, et al. The Total Neuropathy Score as an assessment tool for grading the course of chemotherapy-induced peripheral neurotoxicity: Comparison with the National Cancer Institute-Common Toxicity Scale. Journal of the Peripheral Nervous System. 2007;12(3):210–5. [DOI] [PubMed] [Google Scholar]
- 83.Trotti A, Colevas AD, Setser A, Rusch V, Jaques D, Budach V, et al. , editors. CTCAE v3. 0: development of a comprehensive grading system for the adverse effects of cancer treatment Seminars in radiation oncology; 2003: Elsevier. [DOI] [PubMed] [Google Scholar]
- 84.Delforge M, Bladé J, Dimopoulos MA, Facon T, Kropff M, Ludwig H, et al. Treatment-related peripheral neuropathy in multiple myeloma: the challenge continues. The Lancet Oncology. 2010;11(11):1086–95. [DOI] [PubMed] [Google Scholar]
- 85.Palumbo A, Bringhen S, Rossi D, Cavalli M, Larocca A, Ria R, et al. Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: a randomized controlled trial. Journal of Clinical Oncology. 2010;28(34):5101–9. [DOI] [PubMed] [Google Scholar]
- 86.Moreau P, Pylypenko H, Grosicki S, Karamanesht I, Leleu X, Grishunina M, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. The lancet oncology. 2011;12(5):431–40. [DOI] [PubMed] [Google Scholar]
- 87.Ludwig H, Miguel J, Dimopoulos M, Palumbo A, Sanz RG, Powles R, et al. International Myeloma Working Group recommendations for global myeloma care. Leukemia. 2014;28(5):981. [DOI] [PubMed] [Google Scholar]
- 88.Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. The New England journal of medicine. 2007;357(21):2133–42. [DOI] [PubMed] [Google Scholar]
- 89.Martin TG. Peripheral neuropathy experience in patients with relapsed and/or refractory multiple myeloma treated with carfilzomib. Oncology (Williston Park, NY). 2013;27 Suppl 3:4–10. [PubMed] [Google Scholar]
- 90.Offidani M, Corvatta L, Caraffa P, Gentili S, Maracci L, Leoni P. An evidence-based review of ixazomib citrate and its potential in the treatment of newly diagnosed multiple myeloma. OncoTargets and therapy. 2014;7:1793–800. [DOI] [PMC free article] [PubMed] [Google Scholar]