Multiple myeloma is a disease typical of elderly people, with a median age at diagnosis of 70 years. Much progress has been made in the past few years thanks to the introduction of new drugs. However, increases in survival were much less pronounced in patients aged 60 to 69 years, and no improvement was seen in older patients. Furthermore, the currently approved treatment regimens were tested in clinical trials with stringent inclusion criteria. Aging is associated with a high prevalence of frailty, that is, a state of increased vulnerability to stressors due to a critical decline in physiologic reserves. Elderly people may be categorized as fit or frail according to clinical, functional, cognitive, and socioeconomic criteria. The presence of frailty may complicate the management and outcome of myeloma patients. To date, the choice of treatment of myeloma patients has focused primarily on chronological age and performance status as markers of frailty. However, the elderly population is highly heterogeneous, and improved assessment strategies are needed to define the frailty profile of patients and provide them with the most adequate treatment, thus avoiding the overtreatment of frail patients and the undertreatment of fit patients. The geriatric assessment is a fundamental tool for the evaluation of cognitive and functional status.

Multiple myeloma (MM) is largely a disease of older adults, with a median age at diagnosis of 70 years. A consistent and continuous increase in life expectancy is recorded worldwide and the global population is rapidly aging. Consequently, the number of patients with MM is expected to considerably increase by nearly 80% each year in the next 2 decades.1-3 

Although effective novel agents and supportive care have substantially improved outcome, patients ≥75 years have a shorter survival.4-6  Thus, a deeper knowledge of the factors causing such poor outcomes is needed.

Aging is a complex process characterized by a gradual, progressive decrease in physiological reserve, with changes in body composition and clinically significant reductions in renal, gastric, hepatic, and cardiovascular functions.7,8  It is commonly associated with the concomitant occurrence of multiple diseases (comorbidity),9  and an increased risk of developing physical and cognitive decline (disability).10,11 

In addition, older people are at high risk of developing cancer and frailty. Frailty is a state of increased vulnerability, with cumulative deficits in several physiological systems, which results in diminished resistance to stressors such as MM and its treatment. This state has a negative effect on patients’ quality of life (QoL) and on treatment efficacy and tolerability, with a consequent increase in health care costs.

Approximately one-third of MM patients at diagnosis are frail.1,12  Yet, these patients are poorly characterized, underrepresented, or even excluded from clinical trials due to stringent eligibility criteria or the presence of comorbidities, abnormal laboratory test results, and physical disability.13  Thus, frail patients usually receive regimens tested in fit patients, which may be too toxic for them and lead to early treatment discontinuation, low efficacy, and poor QoL. On the contrary, fit elderly patients may receive reduced-dose treatments based solely on their age. An appropriate definition of frailty is fundamental to better assess patients and provide them with effective, tailored treatments.

An 82-year-old woman presenting with anemia (hemoglobin [Hb], 9.8 g/dL) and high immunoglobulin G level (4630 mg/dL) was diagnosed with immunoglobulin Gk MM in December 2005. Her medical history was significant for hypertension and colic diverticula; her Eastern Cooperative Oncology Group (ECOG) performance status was 1. Treatment at diagnosis consisted of 6 cycles of reduced-dose melphalan-prednisone-thalidomide (MPT; melphalan, 4 mg/m2 on days 1-4; prednisone, 40 mg/m2 on days 1-4; thalidomide, 50 mg/d), followed by thalidomide maintenance, resulting in a partial response. The first relapse occurred in January 2009 when the patient was 86 years old. The disease was symptomatic for anemia (Hb, 8.3 g/dL) and pathological fractures of tibia and fibula requiring surgical stabilization. Her ECOG performance status was 3. The patient received 9 cycles of reduced-dose bortezomib-melphalan-prednisone, with intravenous weekly bortezomib (1.3 mg/m2), melphalan (2 mg), and prednisone (25 mg) every other day. The patient achieved a complete response and experienced a significant improvement in her QoL and performance status (ECOG 1), without any serious adverse event (AE), except for grade 3 diarrhea and hyperglycemia. In March 2011, the patient was 88 years old and experienced a second relapse. She had anemia (Hb, 8.8 g/dL) and a mild renal impairment (creatinine, 1.6 mg/dL). Her ECOG performance status was 2. The patient was treated with reduced-dose lenalidomide (15 mg every other day according to renal function) and prednisone (25 mg every other day). Treatment was well tolerated: no significant toxicities occurred, except for grade 3 hyperglycemia, grade 1 thrombocytopenia, and grade 1 cramps, and no lenalidomide dose reduction was required. The patient achieved a partial response, anemia resolved (Hb, 11.6 g/dL), and renal function improved (creatinine, 1.2 mg/dL). In February 2012, the patient was 89 years old and experienced a third relapse. Due to her rapidly worsening clinical conditions, she received palliative care and she died in April 2012.

This case shows that when frail patients are recognized and treatment is appropriately tailored, a long-term control of the disease and of the related symptoms may be obtained.

Elderly MM patients are highly heterogeneous; chronological age, performance status, or clinician judgment is not sufficient to properly differentiate them. In MM, the term “frail” often refers to a person >75 years old, which sometimes leads to improper undertreatment when the treatment is based only on age.

The geriatric assessment (GA) is a more sensitive predictor of frailty. A comprehensive GA is a systematic procedure to objectively appraise the health status of older people, focusing on somatic, functional, and psychosocial domains.14  Because a full comprehensive GA is a time-consuming procedure that is difficult to use in everyday clinical practice, a simplified GA that includes Katz and Akpom’s basic activities of daily living (ADL) scale,15  Lawton and Brody’s instrumental ADL (IADL) scale,16  and the Charlson Comorbidity Index (CCI),17  should be adopted for elderly patients. The ADL and IADL scores are used to assess self-care activities, tasks of household management, and independence status; the CCI is used to estimate the number and severity of comorbidities (Tables 1-3).15-18 

Table 1

Basic ADL scale

Activities
Bathing: Bathes self completely or needs help in bathing only a single part of the body. 
Dressing: Gets clothes from closets and drawers and puts on clothes. Some help with tying shoes may be needed. 
Toileting: Goes to toilet (may use cane or walker), gets on and off, arranges clothes, cleans genital area without help (may use bedpan/urinal at night). 
Transferring: Moves in and out of bed or chair unassisted. Mechanical transferring aides are acceptable. 
Continence: Exercises complete self-control over urination and defecation. 
Feeding: Gets food from plate into mouth without help. Food may be prepared by another person. 
Activities
Bathing: Bathes self completely or needs help in bathing only a single part of the body. 
Dressing: Gets clothes from closets and drawers and puts on clothes. Some help with tying shoes may be needed. 
Toileting: Goes to toilet (may use cane or walker), gets on and off, arranges clothes, cleans genital area without help (may use bedpan/urinal at night). 
Transferring: Moves in and out of bed or chair unassisted. Mechanical transferring aides are acceptable. 
Continence: Exercises complete self-control over urination and defecation. 
Feeding: Gets food from plate into mouth without help. Food may be prepared by another person. 

If the patient can perform an activity without supervision, direction, or assistance, then a score of 1 should be assigned for that activity. If the patient can perform the activity with supervision, direction, personal assistance, or total care, then a score of 0 should be assigned for that activity.

Table 2

IADL scale

Activities
Ability to use telephone 
Shopping 
 Completely unable to shop 
Food preparation 
Housekeeping 
Laundry 
Mode of transportation 
 Travels independently on public transportation or drives own car 
Responsibility for own medications 
Ability to handle finances 
Activities
Ability to use telephone 
Shopping 
 Completely unable to shop 
Food preparation 
Housekeeping 
Laundry 
Mode of transportation 
 Travels independently on public transportation or drives own car 
Responsibility for own medications 
Ability to handle finances 

A score of 1 or 0 should be assigned to each activity based on the patient's ability to perform that activity. The sum of the points provides the final IADL score.

Table 3

Charlson Comorbidity Index

Comorbidity
Myocardial infarction 
Congestive heart failure 
Peripheral vascular disease 
Cerebrovascular disease 
Dementia 
Chronic pulmonary disease 
Connective tissue disease 
Ulcer 
Mild liver disease 
Diabetes 
Diabetes with end-organ damage 
Ictus 
Moderate-to-severe renal failure 
Nonmetastatic solid tumor 
Leukemia 
Lymphoma, MM 
Moderate-to-severe liver disease 
Metastatic solid tumor 
AIDS 
Comorbidity
Myocardial infarction 
Congestive heart failure 
Peripheral vascular disease 
Cerebrovascular disease 
Dementia 
Chronic pulmonary disease 
Connective tissue disease 
Ulcer 
Mild liver disease 
Diabetes 
Diabetes with end-organ damage 
Ictus 
Moderate-to-severe renal failure 
Nonmetastatic solid tumor 
Leukemia 
Lymphoma, MM 
Moderate-to-severe liver disease 
Metastatic solid tumor 
AIDS 

Frail patients are at high risk of nonhematologic AEs and treatment discontinuation, regardless of other prognostic factors, and should be appropriately evaluated to determine their ability to tolerate treatment.

Age and GA are the fundamental determinants of our approach. The cutoff age that we consider to define frail patients is 80 years.19  However, irrespective of age, the presence of either a functional decline on ADL and IADL, or the presence of comorbidities, may identify frail patients. The Web site http://www.myelomafrailtyscorecalculator.net/ offers a quick and easy way to calculate the frailty score (Table 4).19 

Table 4

GA to identify frail patients with MM

Age, yGA
>80  
76-80 Plus at least 1 of the following:
 ADL score ≤4
 IADL score ≤5
 CCI score ≥2 
≤75 Plus at least 2 of the following:
 ADL score ≤4
 IADL score ≤5
 CCI score ≥2 
Age, yGA
>80  
76-80 Plus at least 1 of the following:
 ADL score ≤4
 IADL score ≤5
 CCI score ≥2 
≤75 Plus at least 2 of the following:
 ADL score ≤4
 IADL score ≤5
 CCI score ≥2 

The GA score can be calculated through the Web site http://www.myelomafrailtyscorecalculator.net.

In frail patients, treatment should be delayed in cases of nonspecific symptoms, and it should be started based on the presence of confirmed CRAB (hypercalcemia, renal failure, anemia, bone lesions) criteria.20-22 

Particular attention is needed with comorbid conditions. Frail patients may present with CRAB-like symptoms that do not lead to actual organ dysfunction and require no immediate antimyeloma treatment but only close monitoring.23  For instance, patients may have age-related osteopenia, mild renal impairment due to hypertension or diabetes, or mild anemia secondary to iron or vitamin deficiency, renal failure, chronic inflammatory diseases, or concomitant dyserythropoietic/myelodysplastic syndrome.

Clear clinical manifestations of serious end-organ damage that can be attributed to myeloma should be considered CRAB, such as a progressive worsening of serum creatinine caused by light-chain cast nephropathy or a decrease in Hb levels from baseline. Creatinine clearance should be considered with caution. Many of the methods used to estimate glomerular filtration rate have not been well validated at the extremes of age. For example, using the Cockcroft-Gault method, a serum creatinine level of 1.5 mg/dL in a patient weighing 70 kg corresponds to an estimated glomerular filtration rate of 38 and 33 mL/min in an 80-year-old man and woman, respectively. In this case, progressive renal impairment rather than a fixed concentration cutoff should be considered to confirm MM diagnosis.

Recently, the International Myeloma Working Group provided updated criteria for MM diagnosis.24  The changes are based on the identification of biomarkers associated with near inevitable development of CRAB features. They include clonal bone marrow plasma cell percentage ≥60%, involved/uninvolved serum free light-chain ratio ≥100, and >1 focal lesion on magnetic resonance imaging studies. Even if no data are present for this special patient population, the assessment of these parameters may avoid organ damage that could inevitably worsen patients’ condition. We use a skeletal survey and a low-dose computed tomography scan as routine investigations to assess bone disease. More complex investigations such as magnetic resonance imaging or positron emission tomography can be unnecessary on a routine basis in frail patients but should be considered in selected cases.

Although the achievement of a complete response is highly important and has become an attainable aim also in the elderly, toxicity may cancel the benefits derived from such a response.25 

In frail patients facing a serious disease such as MM, the choice of therapy should focus not on the quality of response but rather on controlling symptoms, maintaining an independence status, and preserving cognitive function and QoL.

The achievement of stable disease without symptoms related to myeloma is an acceptable goal, and therapy should not be changed.

Treatment should not cause excessive toxicities, which may lead to treatment interruption and reduced efficacy. If toxicities occur, it is likely that inappropriate drug doses are being used and dose reductions are needed. Therefore, keeping the balance between disease control and toxicity is fundamental.

The treatment of elderly MM patients has evolved substantially over the last decade.12  New effective treatments including thalidomide, bortezomib, and lenalidomide have replaced the former standard melphalan-prednisone (MP). MPT and melphalan-prednisone-bortezomib are the reference treatments for elderly patients ineligible for high-dose therapy.26-33  Recently, continuous lenalidomide-dexamethasone (Rd) was shown to be more effective than MPT at diagnosis.34  Of note, currently approved combinations were validated in studies that included highly selected elderly patients, and a geriatric evaluation was not performed.

The major advances obtained with new drugs were limited in older patients, primarily due to an increased treatment-related toxicity. Advanced age (≥75 years); the occurrence of severe cardiac, gastrointestinal, and infective AEs; and drug discontinuation predicted shorter survival in newly diagnosed MM patients treated with MP, either alone or in combination with thalidomide and/or bortezomib. The negative effect of such factors was particularly evident with the use of more complex combinations including both bortezomib and thalidomide.35,36  Moreover, drug discontinuation due to AEs was correlated with an increased risk of death within the first 6 months of treatment.

These data underline the importance of avoiding treatment interruption and reducing the risk of side effects during the initial phase of therapy; thus, low dose–intensity treatments should be preferred for frail patients.

After the diagnosis of symptomatic MM, an appropriate treatment should be determined on the basis of several factors, particularly patient age and GA (Table 5). Based on the results of the GA, it is possible to stratify patients into fit patients suitable for full-dose therapy with 3-drug combinations, and frail patients requiring dose-adjusted therapies. Treatment strategies for frail patients should have minimal cumulative toxicity, which will not exacerbate any preexisting comorbid conditions. In particular, in this setting, 2-drug regimens showed similar efficacy and lower toxicity compared with multidrug combinations.37 

Table 5

Factors to consider in the clinical decision making for frail patients with MM

FactorsAim
Age To assess frailty 
GA 
CRAB criteria To start treatment 
 Hypercalcemia 
 Renal failure 
 Anemia 
 Bone lesions 
Biomarkers of malignancy 
 Clonal bone marrow plasma cell percentage ≥60% 
 Involved/uninvolved serum free light-chain ratio ≥100 
 >1 focal lesion (≥5 mm) on MRI studies 
Cardiovascular history To choose treatment 
History of diabetes 
Renal function 
Neuropathy 
Psychosocial status 
Preferences of the patient and the caregiver 
FactorsAim
Age To assess frailty 
GA 
CRAB criteria To start treatment 
 Hypercalcemia 
 Renal failure 
 Anemia 
 Bone lesions 
Biomarkers of malignancy 
 Clonal bone marrow plasma cell percentage ≥60% 
 Involved/uninvolved serum free light-chain ratio ≥100 
 >1 focal lesion (≥5 mm) on MRI studies 
Cardiovascular history To choose treatment 
History of diabetes 
Renal function 
Neuropathy 
Psychosocial status 
Preferences of the patient and the caregiver 

MRI, magnetic resonance imaging.

A recent phase 3 trial compared Rd, melphalan-prednisone-lenalidomide (MPR), and cyclophosphamide-prednisone-lenalidomide in newly diagnosed elderly MM patients.38  The addition of an alkylating agent to the lenalidomide-steroid combination did not show any significant advantage in patients >75 years. In another trial, the doublet bortezomib-dexamethasone proved to be less toxic and equally effective in comparison with the triplets bortezomib-melphalan-prednisone and bortezomib-thalidomide-dexamethasone in an elderly and frail population.37 

Dosing, schedule, and route of administration can make a substantial difference in the safety profile of therapy. Different trials showed that once-weekly bortezomib significantly reduced the incidence of grade 3-4 AEs (35% vs 51%) and the rate of discontinuation due to toxicity (17% vs 23%) compared to the twice-weekly schedule. In addition, bortezomib administered subcutaneously showed to be as effective as the IV administration39 ; this, combined with an improved safety profile, had a very positive effect on the QoL. Similarly, lenalidomide plus low-dose dexamethasone was better tolerated than lenalidomide plus high-dose dexamethasone and proved to be even more effective.40 

In frail patients, we prefer the oral regimen Rd in cases of nonaggressive disease. Bortezomib has the inconveniences of hospitalization and subcutaneous injection (Table 6). Lenalidomide has the advantage of oral administration, and patients do not need to attend frequent hospital visits. In addition, Rd may be the treatment of choice in patients with preexisting neuropathy.34,40  We use bortezomib-dexamethasone in cases of aggressive disease, which needs a rapid cytoreduction and symptom control.37,41  Similarly, with acute renal dysfunction, bortezomib exerts a faster and more potent action and has a greater chance to reverse renal failure.

Table 6

Treatment strategy in frail patients with MM

Specific characteristicsTreatmentMaintenance
Absence of renal failure Lenalidomide based Low-dose lenalidomide 
No aggressive disease 
Presence of peripheral neuropathy 
Difficult access to hospital 
Presence of renal failure Bortezomib based Twice-monthly subcutaneous bortezomib 
Aggressive disease 
Presence of extramedullary disease 
Easy access to hospital 
Specific characteristicsTreatmentMaintenance
Absence of renal failure Lenalidomide based Low-dose lenalidomide 
No aggressive disease 
Presence of peripheral neuropathy 
Difficult access to hospital 
Presence of renal failure Bortezomib based Twice-monthly subcutaneous bortezomib 
Aggressive disease 
Presence of extramedullary disease 
Easy access to hospital 
Table 7

Treatment schedule in frail patients with MM

Frontline treatmentSecond-line treatmentFollowing lines of treatment
Lenalidomide-steroid
 R*: 10-15 mg/d, d 1-21
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Bortezomib-steroid
 V: 1.3 mg/m2 once weekly
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Melphalan-prednisone
 M: 2 mg every other d
 P: 25 mg/d every other d 
Bortezomib-steroid
 V: 1.3 mg/m2 once weekly
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Lenalidomide-steroid
 R*: 10-15 mg/d, d 1-21
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Cyclophosphamide-prednisone
 C: 50 mg every other d
 P: 25 mg/d every other d 
 Re-treatment Thalidomide-prednisone
 T: 50 mg every other d
 P: 25 mg/d every other d 
Frontline treatmentSecond-line treatmentFollowing lines of treatment
Lenalidomide-steroid
 R*: 10-15 mg/d, d 1-21
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Bortezomib-steroid
 V: 1.3 mg/m2 once weekly
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Melphalan-prednisone
 M: 2 mg every other d
 P: 25 mg/d every other d 
Bortezomib-steroid
 V: 1.3 mg/m2 once weekly
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Lenalidomide-steroid
 R*: 10-15 mg/d, d 1-21
 d: 10 mg/d once weekly or P: 25 mg/d every other d 
Cyclophosphamide-prednisone
 C: 50 mg every other d
 P: 25 mg/d every other d 
 Re-treatment Thalidomide-prednisone
 T: 50 mg every other d
 P: 25 mg/d every other d 

C, cyclophosphamide; d, dexamethasone; M, melphalan; P, prednisone; R, lenalidomide; T, thalidomide; V, bortezomib.

*

Or according to renal function.

In frail patients, we also administer modified drug doses to minimize toxicity. The initial dose of lenalidomide should not exceed 10 to 15 mg/d, and the dose can be adjusted on the basis of renal function and blood counts to avoid profound and prolonged myelosuppression. In this setting, it is reasonable to use prophylactic growth factors and an antimicrobial (at a fixed dose and timing) to avoid myelosuppression and infections, and to permit the patient to stay longer on treatment. A previous history of cardiovascular disease or thromboembolism does not preclude the use of lenalidomide when an adequate thromboprophylaxis is associated.

Corticosteroids may cause elevation of blood pressure and fluid retention, and should therefore be reduced, particularly in cardiac patients. Other adverse effects include hyperglycemia, gastritis, mood swings, insomnia, and increased risk of opportunistic infections. The dose of dexamethasone should be reduced to as little as 10 mg once a week. Alternatively, prednisone, 25 mg every other day, is a valid option.

Subcutaneous weekly bortezomib can be used without dose reductions, although thrombocytopenia could be a concern.42,43  This regimen is especially relevant in frail patients, in whom the better tolerability may allow them to continue treatment, with a higher chance of disease control.

Bortezomib can cause peripheral neuropathy differently from novel proteasome inhibitors.44,45  In frail patients, the use of weekly carfilzomib and oral ixazomib and oprozomib may be implemented in the future.

A conservative approach should be suggested in frail patients: an initial, gentler therapy may be used, and dose escalation may be considered in subsequent cycles if the treatment is optimally tolerated, in the absence of significant toxicities, or in the case of inadequate response.

Finally, in patients with severe impairment of cognitive function or social dependency, palliation is suggested. In these cases, we treat patients with reduced-dose corticosteroids, MP, or cyclophosphamide-prednisone to induce only a relief of disease symptoms.

Evidence of clear benefit of maintenance therapy has only recently emerged in elderly MM patients.12,46  Continuous Rd improved progression-free survival (PFS) compared to fixed-duration Rd for 18 cycles (Rd18) and significantly prolonged PFS and overall survival compared to MPT.34  The superiority of continuous Rd over Rd18 was achieved at the expense of a modest increase in toxicity, and most toxicities occurred within the first 18 months and decreased over time. In this study, 35% of patients were >75 years old, and 9% had severe renal impairment. The superiority of continuous Rd over MPT for both PFS and overall survival was noted even in patients >75 years. Although toxicities were more common in patients >75 years than in younger patients, there was no marked difference in AE rates with continuous Rd and Rd18 within this age subgroup.

In contrast, the more complex combination of MPR followed by lenalidomide maintenance compared to MPR or MP followed by placebo was associated with a reduced rate of progression in patients 65 to 75 years old but not in patients >75 years old.47  This observation can be explained by the increased rate of AEs associated with MPR and the need for more frequent dose modifications in patients >75 years old. In that study, the major benefit to PFS was achieved with lenalidomide maintenance therapy. A landmark analysis showed that lenalidomide maintenance reduced the rate of progression by 66% compared to placebo, regardless of age.

Maintaining an asymptomatic disease status is particularly relevant for frail patients, because advanced age and coexisting morbidities may compromise subsequent salvage therapies at disease relapse. Nevertheless, the benefits associated with continuous therapy should be balanced against the toxicity due to prolonged drug exposure, particularly in frail patients, who are more susceptible to treatment-related toxicities. We use continuous therapy, provided that it does not cause toxicity or worsening of a patient’s condition. Frail patients whose disease is responding slowly and who tolerate therapy well may benefit from maintenance therapy. However, in the case of significant adverse effects, dose reductions or treatment interruption should be considered (Table 6).

In patients with asymptomatic serological relapse, treatment should be delayed. It can be started when clinical relapse, defined by CRAB criteria, occurs. A significant paraprotein relapse with doubling M-component within 2 months is an indirect indicator of increasing disease.48  Close monitoring of paraprotein levels is required, and treatment should be started at the first sign of clinical disease progression.

Similarly to diagnosis, rather than achieving a deep response, the aim of therapy at relapse for frail patients is to maintain the disease asymptomatic and preserve the independency of patients, without excessive toxicity.

Novel agents are the mainstay of treatment at relapse. Lenalidomide has proved to be an effective option, and the addition of steroids even at low doses had a synergistic activity.49-51  Bortezomib combined with steroids is effective and well tolerated,52,53  particularly when the subcutaneous weekly administration is adopted.43,54 

The patient’s overall condition and previous toxicities should be considered to determine the patient’s ability to tolerate treatment and, consequently, to choose the appropriate treatment. Time of relapse and type of previous therapy are also fundamental aspects to take into account.

Bortezomib and lenalidomide re-treatment can be a valuable option, with no cumulative toxicity.48  Rechallenge with a previously used agent may be considered if the treatment produced a substantial benefit and had an acceptable toxicity. In newly diagnosed patients, we consider re-treatment with the previous agents when a response lasted at least 12 months. In frail relapsed patients, we rechallenge with previous drugs when a benefit lasted at least 6 months.

For short-term response duration (<6-12 months, based on the number of previous lines of therapy) or progression while on therapy, we propose an alternative regimen.48  Sequencing of drugs is preferable: if lenalidomide is used as frontline treatment, a drug-class switch is necessary and bortezomib may be used. In frail patients, continuous therapy until progression could be recommended if therapy has an acceptable long-term safety profile. Alkylator-based regimens such as low-dose cyclophosphamide-prednisone, MP, or thalidomide may still play a role in frail patients relapsing after lenalidomide and bortezomib. Cyclophosphamide (50 mg every other day), melphalan (2 mg every other day), or thalidomide (50 mg every other day) may be used.

In the case of aggressive relapse or a short treatment-free interval, a more palliative approach is needed in frail patients with poor condition. In such instances, palliative care aims to optimize the comfort, function, and social support of patients and their families. To relieve the disabling myeloma-related symptoms, low doses of cyclophosphamide, melphalan, corticosteroids, or thalidomide may be used. Treatment of pain should be always introduced.

Frail patients are more susceptible to AEs and treatment interruption. Therefore, appropriate supportive care and early identification of toxicities are needed.

Proteasome inhibitors necessitate antiviral prophylaxis for herpes zoster reactivation; immunomodulatory agents require an appropriate risk-based thromboprophylaxis. Corticosteroids may need gastrointestinal prophylaxis, and a more accurate glycemic control in diabetic patients should be recommended. Antibacterial prophylaxis may be warranted in cases of severe myelosuppression. Neutropenia and anemia may necessitate growth factors.

A careful review of the patient’s previous medications and attention to potential drug interactions are essential in frail patients.

Prompt action, particularly in frail patients, is needed when AEs occur. When a grade 3-4 toxicity occurs while on treatment, therapy should be stopped. It can be restarted when toxicity decreases to at least grade 1, and appropriate dose reductions should be applied.12  Lenalidomide dose may be decreased from 15 to 10 mg/d, from 10 to 5 mg/d, or, if required, to 5 mg every other day on days 1 to 21 every 4 weeks. Bortezomib may be reduced from 1.3 mg/m2 weekly to 1.0 mg/m2 once weekly or even to 0.7 mg/m2 once weekly.

New targeted agents have improved the treatment options for MM. Carfilzomib is a novel ketoepoxide tetrapeptide proteasome inhibitor recently approved by the US Food and Drug Administration for the treatment of relapsed/refractory MM in patients previously treated with bortezomib and lenalidomide. The most common grade 3-4 AEs reported with this agent were fatigue, anemia, nausea, and thrombocytopenia; peripheral neuropathy was mainly limited to grade 1-2. In a recent phase 3 study, carfilzomib plus Rd was more effective than Rd.55  Carfilzomib was also effective in newly diagnosed patients.56,57 

Ixazomib (MLN9708) is another novel proteasome inhibitor that showed promising results in both relapsed and newly diagnosed settings. Two phase 2 studies are evaluating oral ixazomib as monotherapy in relapsed/refractory patients previously exposed to proteasome inhibitors, and one trial has adopted a weekly administration of the drug.58  The most frequent AEs included thrombocytopenia, fatigue, nausea, and diarrhea.59  Positive results were also reported in a phase 1/2 study evaluating weekly ixazomib in combination with Rd in newly diagnosed myeloma patients, followed by maintenance with ixazomib alone.60 

Oprozomib, an orally available carfilzomib structural analog, has been tested in a phase 1b/2 trial including patients with relapsed MM. The most common grade 3-4 toxicities included diarrhea, nausea, neutropenia, hypophosphatemia, thrombocytopenia, vomiting, and fatigue.61 

The monoclonal antibodies elotuzumab and daratumumab are other promising agents. Daratumumab is an anti-CD38 monoclonal antibody. Infusion reactions were the most common toxicity.62-64  Elotuzumab is an anti-CS1 fully humanized monoclonal antibody. Elotuzumab was not so effective as monotherapy, and treatment-related toxicities were largely limited to grade 1-2 infusion reactions.65  In two phase 1 studies, elotuzumab plus bortezomib66  or elotuzumab plus lenalidomide and low-dose dexamethasone proved effective. Two phase 3 trials are comparing elotuzumab plus Rd vs Rd in relapsed and newly diagnosed patients.

Based on the available results, in frail patients, weekly carfilzomib can be a valuable option due to its reduced toxicity. Ixazomib can be an option because of its low toxicity and has the advantage of oral administration. Elotuzumab and daratumumab are effective options and have a very good tolerability.

The growing number of older adults with myeloma is increasing the need for practical strategies to recognize and appropriately manage frail patients, and to avoid the undertreatment of fit patients and the overtreatment of frail patients. The GA is a sensitive assessment tool and should be introduced in everyday clinical practice.

Frail patients need effective tailored treatments to better control the disease while minimizing the risk of toxicity and treatment discontinuation. Improving survival may not be the primary goal of therapy in frail patients. The real aim in this setting should be to keep patients asymptomatic as long as possible, preserve their functional status and independence, and improve their QoL.

The selection of therapy should be based on the risk of toxicity and the capacity of patients to tolerate treatment. Lenalidomide and bortezomib have an essential role in the treatment of frail patients. Two-drug regimens including low-dose steroid in combination with lenalidomide or bortezomib are indicated. Second-generation proteasome inhibitors or immunomodulatory drugs or even monoclonal antibodies are potential candidates to improve the care of frail patients with MM in the future.

The authors thank their assistant, Giorgio Schirripa.

Contribution: A.P. and A.L. collected the data and wrote the manuscript.

Conflict-of-interest disclosure: A.P. has received consultancy fees from Bristol-Myers Squibb, Celgene, Janssen-Cilag, Millennium Pharmaceuticals Inc, and Onyx Pharmaceuticals; and honoraria from Bristol-Myers Squibb, Genmab A/S, Celgene, Janssen-Cilag, Millennium Pharmaceuticals Inc, and Onyx Pharmaceuticals. A.L. has received honoraria from Celgene and Janssen-Cilag.

Correspondence: Antonio Palumbo, Myeloma Unit, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Via Genova 3 Torino, 10126 Italy; e-mail: appalumbo@yahoo.com.

1
National Cancer Institute
 
Surveillance, Epidemiology, and End Results (SEER) Program Research Data (1973-2010), National Cancer Institute, Division of Cancer Control and Population Sciences, Surveillance Research Program, Surveillance Systems Branch, released April 2013, based on the November 2012 submission. Available at: www.seer.cancer.gov. Accessed January 11, 2015
2
Turesson
 
I
Velez
 
R
Kristinsson
 
SY
Landgren
 
O
Patterns of multiple myeloma during the past 5 decades: stable incidence rates for all age groups in the population but rapidly changing age distribution in the clinic.
Mayo Clin Proc
2010
, vol. 
85
 
3
(pg. 
225
-
230
)
3
Smith
 
BD
Smith
 
GL
Hurria
 
A
Hortobagyi
 
GN
Buchholz
 
TA
Future of cancer incidence in the United States: burdens upon an aging, changing nation.
J Clin Oncol
2009
, vol. 
27
 
17
(pg. 
2758
-
2765
)
4
Kumar
 
SK
Rajkumar
 
SV
Dispenzieri
 
A
, et al. 
Improved survival in multiple myeloma and the impact of novel therapies.
Blood
2008
, vol. 
111
 
5
(pg. 
2516
-
2520
)
5
Kumar
 
SK
Dispenzieri
 
A
Lacy
 
MQ
, et al. 
Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients.
Leukemia
2014
, vol. 
28
 
5
(pg. 
1122
-
1128
)
6
Schaapveld
 
M
Visser
 
O
Siesling
 
S
Schaar
 
CG
Zweegman
 
S
Vellenga
 
E
Improved survival among younger but not among older patients with Multiple Myeloma in the Netherlands, a population-based study since 1989.
Eur J Cancer
2010
, vol. 
46
 
1
(pg. 
160
-
169
)
7
Yuen
 
GJ
Altered pharmacokinetics in the elderly.
Clin Geriatr Med
1990
, vol. 
6
 
2
(pg. 
257
-
267
)
8
Sotaniemi
 
EA
Arranto
 
AJ
Pelkonen
 
O
Pasanen
 
M
Age and cytochrome P450-linked drug metabolism in humans: an analysis of 226 subjects with equal histopathologic conditions.
Clin Pharmacol Ther
1997
, vol. 
61
 
3
(pg. 
331
-
339
)
9
Fried
 
LP
Ferrucci
 
L
Darer
 
J
Williamson
 
JD
Anderson
 
G
Untangling the concepts of disability, frailty, and comorbidity: implications for improved targeting and care.
J Gerontol A Biol Sci Med Sci
2004
, vol. 
59
 
3
 
M255-M263
10
Pope
 
AM
Tarlov
 
AR
Disability in America: Toward A National Agenda for Prevention
 
Washington, DC: Institute of Medicine; 1991. Available at: http://www.nap.edu/openbook.php?isbn=0309043786. Accessed January 11, 2015
11
Adams
 
PF
Hendershot
 
GE
Marano
 
MA
Centers for Disease Control and Prevention/National Center for Health Statistics
 
Current estimates from the National Health Interview Survey, 1996. Vital Health Stat. 1999;10(200):1-203
12
Palumbo
 
A
Anderson
 
K
Multiple myeloma.
N Engl J Med
2011
, vol. 
364
 
11
(pg. 
1046
-
1060
)
13
Hutchins
 
LF
Unger
 
JM
Crowley
 
JJ
Coltman
 
CA
Albain
 
KS
Underrepresentation of patients 65 years of age or older in cancer-treatment trials.
N Engl J Med
1999
, vol. 
341
 
27
(pg. 
2061
-
2067
)
14
Hamaker
 
ME
Jonker
 
JM
de Rooij
 
SE
Vos
 
AG
Smorenburg
 
CH
van Munster
 
BC
Frailty screening methods for predicting outcome of a comprehensive geriatric assessment in elderly patients with cancer: a systematic review.
Lancet Oncol
2012
, vol. 
13
 
10
(pg. 
e437
-
e444
)
15
Katz
 
S
Akpom
 
CA
 
A measure of primary sociobiological functions. Int J Health Serv. 1976;6:493-508
16
Lawton
 
MP
Brody
 
EM
 
Assessment of older people: Self-maintaining and instrumental activities of daily living. Gerontologist. 1969;9(3 part 1):179–186
17
Charlson
 
ME
Pompei
 
P
Ales
 
KL
MacKenzie
 
CR
A new method of classifying prognostic comorbidity in longitudinal studies: development and validation.
J Chronic Dis
1987
, vol. 
40
 
5
(pg. 
373
-
383
)
18
Lawton
 
MP
Scales to measure competence in everyday activities.
Psychopharmacol Bull
1988
, vol. 
24
 
4
(pg. 
609
-
614
)
19
Palumbo
 
A
Bringhen
 
S
Mateos
 
MV
, et al. 
Geriatric assessment predicts survival and toxicities in elderly myeloma patients: an International Myeloma Working Group report.
Blood
2015
, vol. 
125
 
13
(pg. 
2068
-
2074
)
20
Durie
 
BG
Kyle
 
RA
Belch
 
A
, et al. 
Scientific Advisors of the International Myeloma Foundation
Myeloma management guidelines: a consensus report from the Scientific Advisors of the International Myeloma Foundation.
Hematol J
2003
, vol. 
4
 
6
(pg. 
379
-
398
)
21
Durie
 
BG
Harousseau
 
JL
Miguel
 
JS
, et al. 
International Myeloma Working Group
International uniform response criteria for multiple myeloma.
Leukemia
2006
, vol. 
20
 
9
(pg. 
1467
-
1473
)
22
Kyle
 
RA
Rajkumar
 
SV
Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma.
Leukemia
2009
, vol. 
23
 
1
(pg. 
3
-
9
)
23
Mehta
 
J
Cavo
 
M
Singhal
 
S
How I treat elderly patients with myeloma.
Blood
2010
, vol. 
116
 
13
(pg. 
2215
-
2223
)
24
Rajkumar
 
SV
Dimopoulos
 
MA
Palumbo
 
A
, et al. 
International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma.
Lancet Oncol
2014
, vol. 
15
 
12
(pg. 
e538
-
e548
)
25
Gay
 
F
Larocca
 
A
Wijermans
 
P
, et al. 
Complete response correlates with long-term progression-free and overall survival in elderly myeloma treated with novel agents: analysis of 1175 patients.
Blood
2011
, vol. 
117
 
11
(pg. 
3025
-
3031
)
26
Palumbo
 
A
Bringhen
 
S
Caravita
 
T
, et al. 
Italian Multiple Myeloma Network, GIMEMA
Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial.
Lancet
2006
, vol. 
367
 
9513
(pg. 
825
-
831
)
27
Palumbo
 
A
Bringhen
 
S
Liberati
 
AM
, et al. 
Oral melphalan, prednisone, and thalidomide in elderly patients with multiple myeloma: updated results of a randomized controlled trial.
Blood
2008
, vol. 
112
 
8
(pg. 
3107
-
3114
)
28
Facon
 
T
Mary
 
JY
Hulin
 
C
, et al. 
Intergroupe Francophone du Myélome
Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial.
Lancet
2007
, vol. 
370
 
9594
(pg. 
1209
-
1218
)
29
Hulin
 
C
Facon
 
T
Rodon
 
P
, et al. 
Efficacy of melphalan and prednisone plus thalidomide in patients older than 75 years with newly diagnosed multiple myeloma: IFM 01/01 trial.
J Clin Oncol
2009
, vol. 
27
 
22
(pg. 
3664
-
3670
)
30
Wijermans
 
P
Schaafsma
 
M
Termorshuizen
 
F
, et al. 
Dutch-Belgium Cooperative Group HOVON
Phase III study of the value of thalidomide added to melphalan plus prednisone in elderly patients with newly diagnosed multiple myeloma: the HOVON 49 Study.
J Clin Oncol
2010
, vol. 
28
 
19
(pg. 
3160
-
3166
)
31
Waage
 
A
Gimsing
 
P
Fayers
 
P
, et al. 
Nordic Myeloma Study Group
Melphalan and prednisone plus thalidomide or placebo in elderly patients with multiple myeloma.
Blood
2010
, vol. 
116
 
9
(pg. 
1405
-
1412
)
32
Beksac
 
M
Haznedar
 
R
Firatli-Tuglular
 
T
, et al. 
Addition of thalidomide to oral melphalan/prednisone in patients with multiple myeloma not eligible for transplantation: results of a randomized trial from the Turkish Myeloma Study Group.
Eur J Haematol
2011
, vol. 
86
 
1
(pg. 
16
-
22
)
33
San Miguel
 
JF
Schlag
 
R
Khuageva
 
NK
, et al. 
VISTA Trial Investigators
Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma.
N Engl J Med
2008
, vol. 
359
 
9
(pg. 
906
-
917
)
34
Benboubker
 
L
Dimopoulos
 
MA
Dispenzieri
 
A
, et al. 
FIRST Trial Team
Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma.
N Engl J Med
2014
, vol. 
371
 
10
(pg. 
906
-
917
)
35
Palumbo
 
A
Bringhen
 
S
Rossi
 
D
, 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.
J Clin Oncol
2010
, vol. 
28
 
34
(pg. 
5101
-
5109
)
36
Palumbo
 
A
Bringhen
 
S
Larocca
 
A
, et al. 
Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: updated follow-up and improved survival.
J Clin Oncol
2014
, vol. 
32
 
7
(pg. 
634
-
640
)
37
Niesvizky
 
R
Flinn
 
IW
Rifkin
 
R
, et al. 
Efficacy and safety of three bortezomib-based combinations in elderly, newly diagnosed multiple myeloma patients: results from all randomized patients in the community-based, phase 3b UPFRONT Study [abstract].
Blood
2011
, vol. 
118
 
21
 
Abstract 478
38
Magarotto
 
V
Bringhen
 
S
Musto
 
P
, et al. 
Doublet vs triplet lenalidomide-containing regimens in newly diagnosed myeloma patients, younger or older than 75 years: subgroup analysis of a phase III study [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 2110
39
Moreau
 
P
Pylypenko
 
H
Grosicki
 
S
, et al. 
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study.
Lancet Oncol
2011
, vol. 
12
 
5
(pg. 
431
-
440
)
40
Rajkumar
 
SV
Jacobus
 
S
Callander
 
NS
, et al. 
Eastern Cooperative Oncology Group
Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial.
Lancet Oncol
2010
, vol. 
11
 
1
(pg. 
29
-
37
)
41
Larocca
 
A
Cavallo
 
F
Magarotto
 
V
, et al. 
Reduced dose-intensity subcutaneous bortezomib plus prednisone (VP) or plus cyclophosfamide (VCP) or plus melphalan (VMP) for newly diagnosed multiple myeloma patients older than 75 years of age [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 539
42
Petrucci
 
MT
Finsinger
 
P
Chisini
 
M
Gentilini
 
F
Subcutaneous bortezomib for multiple myeloma treatment: patients’ benefits.
Patient Prefer Adherence
2014
, vol. 
8
 (pg. 
939
-
946
)
43
Mateos
 
MV
San Miguel
 
JF
Safety and efficacy of subcutaneous formulation of bortezomib versus the conventional intravenous formulation in multiple myeloma.
Ther Adv Hematol
2012
, vol. 
3
 
2
(pg. 
117
-
124
)
44
Moreau
 
P
Richardson
 
PG
Cavo
 
M
, et al. 
Proteasome inhibitors in multiple myeloma: 10 years later.
Blood
2012
, vol. 
120
 
5
(pg. 
947
-
959
)
45
Bringhen
 
S
Gay
 
F
Donato
 
F
Troia
 
R
Mina
 
R
Palumbo
 
A
Current Phase II investigational proteasome inhibitors for the treatment of multiple myeloma.
Expert Opin Investig Drugs
2014
, vol. 
23
 
9
(pg. 
1193
-
1209
)
46
Ludwig
 
H
Durie
 
BG
McCarthy
 
P
, et al. 
International Myeloma Working Group
IMWG consensus on maintenance therapy in multiple myeloma.
Blood
2012
, vol. 
119
 
13
(pg. 
3003
-
3015
)
47
Palumbo
 
A
Hajek
 
R
Delforge
 
M
, et al. 
MM-015 Investigators
Continuous lenalidomide treatment for newly diagnosed multiple myeloma.
N Engl J Med
2012
, vol. 
366
 
19
(pg. 
1759
-
1769
)
48
Kastritis
 
E
Palumbo
 
A
Dimopoulos
 
MA
Treatment of relapsed/refractory multiple myeloma.
Semin Hematol
2009
, vol. 
46
 
2
(pg. 
143
-
157
)
49
Dimopoulos
 
M
Spencer
 
A
Attal
 
M
, et al. 
Multiple Myeloma (010) Study Investigators
Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma.
N Engl J Med
2007
, vol. 
357
 
21
(pg. 
2123
-
2132
)
50
Weber
 
DM
Chen
 
C
Niesvizky
 
R
, et al. 
Multiple Myeloma (009) Study Investigators
Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America.
N Engl J Med
2007
, vol. 
357
 
21
(pg. 
2133
-
2142
)
51
Touzeau
 
C
Blin
 
N
Clavert
 
A
, et al. 
Efficacy of lenalidomide plus dexamethasone in patients older than 75 years with relapsed multiple myeloma.
Leuk Lymphoma
2012
, vol. 
53
 
7
(pg. 
1318
-
1320
)
52
Jagannath
 
S
Richardson
 
PG
Barlogie
 
B
, et al. 
SUMMIT/CREST Investigators
Bortezomib in combination with dexamethasone for the treatment of patients with relapsed and/or refractory multiple myeloma with less than optimal response to bortezomib alone.
Haematologica
2006
, vol. 
91
 
7
(pg. 
929
-
934
)
53
Jagannath
 
S
Barlogie
 
B
Berenson
 
JR
, et al. 
Updated survival analyses after prolonged follow-up of the phase 2, multicenter CREST study of bortezomib in relapsed or refractory multiple myeloma.
Br J Haematol
2008
, vol. 
143
 
4
(pg. 
537
-
540
)
54
Arnulf
 
B
Pylypenko
 
H
Grosicki
 
S
, et al. 
Updated survival analysis of a randomized phase III study of subcutaneous versus intravenous bortezomib in patients with relapsed multiple myeloma.
Haematologica
2012
, vol. 
97
 
12
(pg. 
1925
-
1928
)
55
Stewart
 
AK
Rajkumar
 
SV
Dimopoulos
 
MA
, et al. 
ASPIRE Investigators
Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma.
N Engl J Med
2015
, vol. 
372
 
2
(pg. 
142
-
152
)
56
Bringhen
 
S
Petrucci
 
MT
Larocca
 
A
, et al. 
Carfilzomib, cyclophosphamide, and dexamethasone in patients with newly diagnosed multiple myeloma: a multicenter, phase 2 study.
Blood
2014
, vol. 
124
 
1
(pg. 
63
-
69
)
57
Moreau
 
P
Kolb
 
B
Hulin
 
C
, et al. 
Carfilzomib plus melphalan and prednisone (CMP) is a promising combination therapy for the treatment of elderly patients with newly diagnosed multiple myeloma: results of a phase I/II trial in 68 cases [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 1933
58
Kumar
 
S
Bensinger
 
W
Zimmerman
 
TM
, et al. 
Weekly MLN9708, an investigational oral proteasome inhibitor (PI), in relapsed/ refractory multiple myeloma (MM): results from a phase I study after full enrollment [abstract].
J Clin Oncol
2013
, vol. 
31
 
suppl
 
Abstract 8514
59
Kumar
 
SK
Roy
 
V
Reeder
 
C
, et al. 
Phase 2 trial of single agent MLN9708 in patients with relapsed multiple myeloma not refractory to bortezomib [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 1944
60
Kumar
 
SK
Berdeja
 
JG
Niesvizky
 
R
, et al. 
A phase 1/2 study of weekly MLN9708, an investigational oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma (MM) [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 332
61
Ghobrial
 
IM
Kaufman
 
JL
Siegel
 
DS
, et al. 
Clinical profile of single-agent modified-release oprozomib tablets in patients (Pts) with hematologic malignancies: updated results from a multicenter, open-label, dose escalation phase 1b/2 study [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3184
62
Plesner
 
T
Lokhorst
 
H
Gimsing
 
P
, et al. 
Daratumumab, a CD38 monoclonal antibody in patients with multiple myeloma: data from a dose-escalation phase I/II study [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 78
63
Lokhorst
 
H
Plesner
 
T
Gimsing
 
P
Nahi
 
H
Lisby
 
S
Richardson
 
P
Daratumubab, a CD38 monoclonal antibody study in advanced multiple myeloma: an open-label, dose-escalation followed by open-label extension in a single-arm phase I/II study [abstract].
Haematologica
2013
, vol. 
98
 
suppl 1
 
Abstract 576
64
Plesner
 
T
Arkenau
 
T
Lokhorst
 
H
, et al. 
Preliminary safety and efficacy data of daratumumab in combination with lenalidomide and dexamethasone in relapsed or refractory multiple myeloma [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 1986
65
Zonder
 
JA
Mohrbacher
 
AF
Singhal
 
S
, et al. 
A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma.
Blood
2012
, vol. 
120
 
3
(pg. 
552
-
559
)
66
Jakubowiak
 
AJ
Benson
 
DM
Bensinger
 
W
, et al. 
Phase I trial of anti-CS1 monoclonal antibody elotuzumab in combination with bortezomib in the treatment of relapsed/refractory multiple myeloma.
J Clin Oncol
2012
, vol. 
30
 
16
(pg. 
1960
-
1965
)
Sign in via your Institution