826
Views
5
CrossRef citations to date
0
Altmetric
Review

Managing multiple myeloma in the elderly: are we making progress?

, &
Pages 301-315 | Published online: 10 Jan 2014

Abstract

Treatment of multiple myeloma has evolved rapidly over the last decade due to novel therapeutic agents. Improved upfront and salvage options have resulted in enhanced survival; however, this has been less pronounced in elderly patients compared with their younger counterparts. Indeed, treatment-related toxicities in older patients may have subverted the survival benefit made by newer treatment modalities. However, owing to the immaturity of current published data, the true survival impact made by novel agents in the elderly patient subgroup is far from being fully appreciated. Improved responses, along with increased salvage options, imply that progress for elderly patients is being made. The current challenge to improve survival for elderly patients not only rests with continued research into tolerable novel treatment regimens, but also, scrupulous supportive care and the judicious use of current novel agents in appropriate dosing, combinations and sequence. Here, we review the outcomes of elderly patients with multiple myeloma over recent years and focus on the current treatment options available for this group.

Medscape: Continuing Medical Education Online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint sponsorship of Medscape, LLC and Expert Reviews Ltd. Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit(s). Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test and/or complete the evaluation at www.medscape.org/journal/experthematology; (4) view/print certificate.

Release date: 14 June 2011; Expiration date: 14 June 2012

Learning objectives

Upon completion of this activity, participants will be able to:

  • • Describe advances in treatment and survival for patients with MM over the past decade

  • • Identify predictors of short- and long-term outcomes in patients with MM

  • • Compare best upfront treatment options for older patients with MM

  • • Describe best options for treatment of MM in older patients with poorer prognosis

  • • Describe factors in the elderly that reduce benefits of MM treatments compared with younger patients

Financial & competing interests disclosure

EDITOR

Elisa Manzotti

Editorial Director, Future Science Group, London, UK.

Disclosure: Elisa Manzotti has disclosed no relevant financial relationships.

CME AUTHOR

Désirée Lie, MD, MSEd

Clinical Professor, Director of Research and Faculty Development, Department of Family Medicine, University of California, Irvine, CA, USA.

Disclosure: Désirée Lie, MD, MSEd, has served as a nonproduct speaker for: “Topics in Health” for Merck Speaker Services.

AUTHORS AND CRENDENTIALS

Hang Quach

Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia; Monash Medical Centre, Clayton, Victoria, Australia; Malignant Hematology and Stem Cell Transplantation, Alfred Health-Southern Health Hematology Consortium, Victoria, Australia; Department of Medicine, University of Melbourne, Victoria, Australia.

Disclosure: Hang Quach has disclosed no relevant financial relationships.

H Miles Prince

Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia; Department of Medicine, University of Melbourne, Victoria, Australia; Hematology Service, Peter MacCallum Cancer Centre, Victoria, Australia.

Disclosure: H Miles Prince has disclosed no relevant financial relationships.

Andrew Spencer

Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia; Malignant Hematology and Stem Cell Transplantation, Alfred Health-Southern Health Hematology Consortium, Victoria, Australia.

Disclosure: Andrew Spencer has disclosed no relevant financial relationships.

Over the last decade, new therapeutic options together with improved supportive care in autologous stem-cell transplantation (ASCT) have substantially increased the survival expectations for patients with multiple myeloma (MM). Patients diagnosed within the last 10 years can now expect to live for a median of approximately 5–8 years, compared with less than 3 years several decades ago Citation[1,2]. Improvement in survival has certainly been more pronounced in the younger population, compared with the elderly subgroup, in whom only a minor improvement has been documented over the last decade, as reported in recent cohort studies Citation[1,3]. Despite this, no convincing evidence exists to suggest that MM is biologically different in the elderly Citation[4,5]. Thus, the apparent lack in survival progress in older patients over the last decade Citation[1,3] may relate to a combination of treatment-related toxicities and the inability to deliver effective therapy. On the other hand, given the immaturity of published data on novel treatment regimens for elderly patients and their only recent routine use in the clinical setting in this patient subgroup, the true survival impact made by novel agents may have yet to be fully appreciated. Nonetheless, significant progress in the treatment of elderly patients is already evidenced by our ability to induce better quality responses with newer therapeutic modalities that, in turn, correlate with better clinical outcomes Citation[6,7]. Older patients are no longer necessarily assigned to a palliative treatment approach and the therapeutic goal of attaining a state of minimal residual disease can now be feasibly applied to patients irrespective of age group or transplant eligibility Citation[8]. With increasing upfront treatment and salvage options, the challenge of treating elderly patients with MM rests with juggling the different therapeutic combinations and sequence of novel agents, so as to maximize survival outcome and minimize treatment-related toxicities, while offering concurrent best supportive care. Here, we review the outcomes of elderly myeloma patients over the last decade and focus on the current treatment options available for this group of patients.

Survival outcomes for elderly patients

The incidence of MM is approximately five cases per 100,000 persons/year Citation[9]. MM is a disease of the elderly, with a median age of onset of 65–70 years, with one study reporting an increase of median age at diagnosis from 70 to 74 years over the last 50 years Citation[10]. Several reports have noted the increased incidence of MM over time Citation[10,201]. Whilst these data may be prejudiced by improved reporting strategies from cancer registries and better healthcare access, the increasing incidence seen in elderly patients are probably real, given the improved life expectancy of the normal population.

Until the turn of the century, no improvement was made with respect to overall survival (OS) in patients with MM. In fact, an increased mortality rate was generally observed over time Citation[11–13]. A modest improvement in survival was achieved upon the introduction of high-dose melphalan with ASCT in the 1990s. Although MM remained incurable, ASCT improved median OS for younger patients in the order of 4–5 years Citation[14,15], while OS for elderly patients remained unsatisfactory at 2–3 years Citation[16]. The next wave of substantial improvement in long-term prognosis occurred upon the introduction of thalidomide in the 1990s, followed by bortezomib in 2003 and lenalidomide in 2005 Citation[1,17]. However, in the elderly patient subgroup, the impact on survival appeared to be more modest, according to recent reports Citation[1,3]. Recently, Kumar and colleagues attributed the clear improvement in OS from both initial diagnosis (44.8 vs 29.9 months; p < 0.001) and time of relapse (23.9 vs 11.8 months; p < 0.001) to novel therapeutic agents in 2981 patients diagnosed with MM after the year 2000 Citation[1]. However, the degree of improvement in survival over the last 10 years appeared to progressively diminish when assessed according to increasing age group; the greatest increase in 10-year relative survival was seen in patients younger than 50 years of age, whereas only a modest increase was seen in patients 60–69 years of age and virtually no improvement in survival was seen in patients aged 70 years or over. Similarly, via a novel model-based projection method using epidemiological data from 1973 to 2005, one group reported much lower 5- and 10-year relative-survival projections for elderly patients compared with young patients diagnosed with MM between 2006 and 2010 Citation[3]. Of note, the aforementioned analysis for survival improvement in elderly patients with MM was extrapolated from data from the first half of this decade, when in fact the widespread and routine use of novel therapeutic agents in the upfront treatment of MM in elderly patients has only occurred in the latter half of this decade. As such, these survival estimates may not adequately reflect the survival expectations for elderly patients diagnosed in the current era.

Many clinical trials utilizing novel therapeutic agents demonstrate improvements in clinical outcomes irrespective of age Citation[18,19]. Despite this, the application of innovative treatment strategies in elderly patients has generally lagged behind that of young patients in recent years. This, combined with the tendency to opt for a nonintensive or palliative approach in older patients, has led to a proportion of patients being undertreated Citation[20]. Conversely, with improved supportive care, the upper age limit criteria for intensive treatments have progressively increased Citation[21–23]. In addition, we now have a number of trials showing that tolerance to novel therapeutic agents is similar between young and elderly patients Citation[18,19,24]. One would hope that with time, the delivery of a new standard of care, such as chemotherapy with novel agents, will not be compromised by age. Indeed, it will be some time before any survival advantage is fully appreciated.

Importance of complete response with upfront treatment: does this apply to the elderly population?

The achievement of complete response (CR) in MM, has so far been difficult with conventional-dose chemotherapy and its positive correlation with improved survival has been difficult to establish. CR rates were typically in the order of 5% with conventional-dose chemotherapy, and many studies historically demonstrated no correlation between depth of response and long-term survival Citation[25,26]. The potential prognostic impact of CR (as stringently defined by immunofixation negativity as per either the European Bone Marrow Transplant [EBMT] Citation[27] or International Response Criteria Citation[28]) was realized when such a depth of response became more achievable with high-dose therapy (HDT) and ASCT Citation[29–33] and even more so with the incorporation of novel therapeutic agents Citation[34–36]. Indeed, a recent review of 4990 patients confirmed that achievement of CR translated to improved long-term outcome Citation[37]. This holds true in the era of novel agents in both the upfront and relapse setting; CR has emerged as a predictor for longer progression-free survival (PFS) and, in some cases, OS Citation[6,7,18,34–36,38,39]. It now represents an important goal in the treatment of MM Citation[7]. However, does such an endeavor carry the same degree of importance for elderly patients in whom treatment-related mortality might subvert the survival benefit conferred by the achievement of CR?

Recent clinical trials that have focused on elderly patients demonstrate the benefit in achieving a CR with respect to PFS and treatment-free interval (TFI), but not necessarily OS. In the Phase III Velcade as Initial Standard Therapy in Multiple Myeloma: Assessment with Melphalan and Prednisone (VISTA) trial, where a combination of bortezomib, melphalan and prednisone (VMP) was compared with melphalan and prednisone (MP) in a group of patients with a median age of 71 years Citation[18], CR was associated with longer time to progression (TTP; hazard ratio [HR]: 0.45; p = 0.004) and TFI (HR: 0.38; p < 0.0001) Citation[6]. The numerical improvement in OS in patients who achieved CR (HR: 0.87; p = 0.54) was not statistically significant Citation[6]. Possible reasons include short follow-up period and a small number of deaths in each arm. Nonetheless, the significant prolongation of TFI in the VMP arm (from 15 months [MP] to 30 months [VMP]) is itself an important clinical benefit and could be argued as a justification for a more intensive approach in elderly patients Citation[8]. In studies that have much longer follow-up, such as the recent analysis of 1175 patients who received MP in combination with thalidomide (MPT), achievement of a CR was an independent predictor of longer PFS and OS, regardless of age. Patients aged above 75 years derived similar survival benefits from the achievement of CR Citation[40].

In other studies, the potential survival benefit conferred by a higher rate of CR was counterbalanced by a higher rate of toxicity-related deaths in elderly patients. This was observed in a randomized trial comparing thalidomide and dexamethasone (TD) with MP in a group of 289 patients aged over 65 years Citation[41]. Despite a higher proportion of CR/very good partial response (VGPR) in the TD group (26 vs 13%; p = 0.006), OS was slightly shorter in the TD group due to a higher rate of toxicities, especially in patients above 75 years of age with poor performance status.

Irrespective of age, the impact of CR on survival is not pronounced in patients whose symptomatic MM has progressed from monoclonal gammopathy of uncertain significance (MGUS) or smouldering myeloma Citation[42–44]. Via microarray analysis, one group demonstrated that MM patients with MGUS-like molecular signatures had prolonged survival despite a lower incidence of CR following intensive therapy with tandem transplantation Citation[43]. Similarly, the impact of CR on survival was not seen for International Staging System stage I disease in the long-term analysis of the Intergroupe Francophone du Myelome (IFM) trials, in which patients underwent tandem ASCT Citation[44]. This may relate to the inherently indolent biology of these subgroups such that a ‘reversal back to their smouldering state’ is sufficient to achieve a durable response.

Overall, the two groups that may not derive a significant survival advantage from achieving CR are frail elderly patients with poor performance status and patients who have progressed from prior MGUS/smouldering myeloma or International Staging System stage I disease. For the remainder of the elderly population, with improved supportive therapy, CR may still be a valid endeavor, especially with the advent of less toxic but more efficacious treatment options.

Upfront treatment options for elderly patients

It has been increasingly recognized that chronological age does not always reflect biological age and, as such, the traditional blanket omission of HDT for patients aged over 65 years has been superseded by a risk-adapted approach based on biological assessments Citation[45]. The options for induction therapy in elderly patients have expanded over the last decade to incorporate novel therapeutic agents, with or without consolidative HDT and ASCT postinduction therapy.

Elderly patients eligible for HDT & ASCT

Currently, HDT and ASCT are valid upfront treatment strategies for elderly patients who are deemed suitable based on good performance status and limited comorbidities. Generally, such patients are limited to those under the age of 70 years. Although younger patients have better OS with ASCT, age per se is not an independent prognostic variable with ASCT Citation[46]. Thus, older patients may stand to benefit as much from ASCT as younger patients Citation[46,47]. Treatment-related mortality appeared to be reduced with the use of lower-dose melphalan conditioning (100–140 mg/m2) in elderly patients Citation[23,48] and has rendered this treatment modality feasible in selected patients between the ages of 65 and 70 years, especially with the improvement in supportive care. Patients eligible for ASCT should receive stem cell-sparing induction therapy (i.e., regimens not containing melphalan) for three to four cycles prior to stem cell collection. This generally consists of a combination of one or two chemotherapy agents with or without corticosteroids and novel therapeutic agents including thalidomide, lenalidomide or bortezomib Citation[49–55]. The choice often depends on local treatment guidelines and access to novel agents.

Given the efficacy of novel agent-containing induction regimens for MM, the role of ASCT has been questioned, especially when the IFM99 study demonstrated superior OS with MPT compared with vincristine, doxorubicin and dexamethasone induction therapy followed by ASCT for elderly patients Citation[39]. However, the question remains as to whether ASCT will further improve the outcome for patients receiving induction therapy that incorporate novel agents upfront. Indeed, thalidomide, lenalidomide or bortezomib-based induction therapy have achieved impressive CR rates in the order of 20–44% Citation[32,33,55,56], a result comparable to, if not better than, that seen post-ASCT in the era of conventional-dose chemotherapy with infusional vincristine, doxorubicin and dexamethasone. These promising results have led to the current era of trials comparing novel induction agents with or without consolidative ASCT Citation[55,57]. In the subset of patients who achieve CR post-novel agent-based induction therapy, the question of whether deferring ASCT until relapse will result in similar survival outcomes remains uncertain. The depth of CR in such patients may be further improved with ASCT; indeed, early data seem to suggest that such patients still benefit further with HDT and ASCT in consolidation Citation[58,59].

Elderly patients not eligible for HDT & ASCT

In patients not eligible for ASCT, the addition of thalidomide, lenalidomide and bortezomib to conventional MP have improved response and survival outcomes . Results from the six studies comparing MPT with MP are outlined in . In all six studies, MPT resulted in a significantly superior PFS, TTP and event-free survival compared with MP. However, the Italian Citation[19,38] and Nordic Citation[60] studies, which utilized ongoing thalidomide maintenance, did not demonstrate superiority in OS in the MPT arm compared with MP. There are at least two reasons for the lack of OS benefit: first, the successful salvage with thalidomide or other novel agents in the MP group upon progression, and second, patients in the MPT group appeared to be more resistant to salvage therapy upon relapse. By contrast, the two French trials (IFM 99 Citation[39] and IFM 01 Citation[61]), in which thalidomide maintenance was not incorporated, did demonstrate superiority in both PFS and OS in the MPT arm compared with MP. This differs to recent results from the Turkish group Citation[62], which showed that eight cycles of MPT, without ongoing thalidomide maintenance, resulted in a superior overall response rate (ORR) and disease-free survival, but not OS, compared with MP in patients aged over 55 years. The median follow-up of this study was a short 23 months. Overall, a meta-analysis of all six trials recently confirmed HRs for PFS and OS of 0.68 and 0.80, respectively, in favor of MPT Citation[63]. The superior efficacy of MPT comes at a cost of greater toxicity; mainly myelosuppression, thromboembolism and peripheral neuropathy. Higher doses of thalidomide, up to 400 mg, as used in the Nordic trial, resulted in greater toxicity, whereas lower doses (100–200 mg) were better tolerated and did not compromise survival outcome Citation[60]. As such, there is a move towards lower doses of thalidomide (50–100 mg) and melphalan when MPT is applied to patients 75 years of age or older Citation[63].

Like MPT, combination VMP is impressive. An initial interim analysis of the randomized Phase III VISTA trial showed convincing superiority of VMP compared with MP with regards to ORR (70 vs 44% in the MP arm), CR (35 vs 4%), TTP (24 vs 16 months) and 2-year OS (83 vs 70%) Citation[18]. A more recent report showed that the quality of responses improved with prolonged VMP treatment, with 28% of CRs achieved during cycles five to nine Citation[6]. Importantly, the superior efficacy of VMP was demonstrated in poor prognostic groups, including patients with high-risk cytogenetics, renal impairment and advanced age. However, this was not confirmed in the Spanish study comparing VMP with bortezomib, thalidomide and prednisone (VTP) in patients aged 65 years or older Citation[32]. Here, patients with high-risk cytogenetics (t4;14, t14;16 or del17p) had shorter OS, despite a similar ORR compared with patients with standard-risk cytogenetics. There was no difference in efficacy between VMP and VTP, with impressive CR rates of 20 and 28%, respectively, postinduction therapy (p = 0.2). VTP was associated with a higher rate of serious adverse events – in particular, 8% of serious cardiac events, leading to discontinuation. Importantly, the rate of CR was further improved with ongoing bortezomib and prednisone or bortezomib and thalidomide (VT) maintenance therapy, to 39 and 44%, respectively. Weekly dosing of bortezomib (as opposed to twice weekly in the classic scheduling of day 1, 4, 8 and 11 every 21 days) appears to reduce bortezomib-induced peripheral neuropathy, constipation and thrombocytopenia Citation[32,64] and should be considered for elderly patients.

The main contender for MPT and MPV as upfront treatment for elderly patients is MP combined with lenalidomide (MPR; Revlimid™). In an interim report of the international Phase III randomized trial (MM-015) comparing MPR with MPR followed by lenalidomide maintenance (MPR-R) and MP, the response was highest in the MPR-R arm (MP: ORR 77 vs 50%, p < 0.001; MP: ≥VGPR 32 vs 12%, p < 0.001) Citation[65]. Ongoing therapy with lenalidomide (i.e., MPR-R) resulted in a 69% risk reduction of progression (p < 0.001) compared with MPR alone. Grade 3 or 4 neutropenia, thrombocytopenia and anemia occurred in 71, 38 and 24% of patients, respectively. However, the incidence of myelosuppression was low during single-agent lenalidomide maintenance therapy.

Other non-melphalan-containing induction regimens could also be used in patients not eligible for ASCT. However, these have yet to demonstrate superior survival compared with MPT. In the Phase III Eastern Cooperative Oncology Group E4A03 trial Citation[55], lenalidomide and dexamethasone (len–dex) combination appeared efficacious as induction therapy. However, the use of lenalidomide with high-dose dexamethasone (RD; dexamethasone 40 mg daily on days 1–4, 9–12 and 17–20 every 28 days) was more toxic compared with lenalidomide and low-dose dexamethasone (Rd; dexamethasone 40 mg weekly). This was especially true for patients aged over 65 years, in whom increased early deaths, venous thromboembolism (VTE) complications and other serious adverse events were encountered. The increased treatment-related mortality in the high-dose dexamethasone plus lenalidomide arm resulted in an inferior OS despite a superior response rate Citation[55]. A study comparing MPT with len–dex, MM-020, is nearing completion. It is worth reiterating that unlike len–dex, combination TD has only modest efficacy as an induction therapy, with an ORR between 64 and 76% Citation[66–68] and an inferior OS compared with MP in elderly patients Citation[41].

Combinations of two novel agents with chemotherapy, such as the addition of thalidomide to VMP (VMPT) followed by maintenance VT, appear to induce an impressive response, with a CR rate of 38% Citation[33,64] and superior PFS compared with VMP (p = 0.008) However, toxicities were significant, with rates of grade 3 and 4 hematological and nonhematological toxicities being up to 45 and 51%, respectively. Similarly, lenalidomide, bortezomib and dexamethasone is emerging as an effective combination for elderly patients Citation[56,57]. Preliminary data from a Phase I/II trial Citation[57], which included patients up to the age of 86 years, showed an ORR of 100%, with a CR of 37%. The incidence of grade 3 or 4 neutropenia and thrombocytopenia were 10% each and nonhematological toxicities were low. This non-chemotherapy-containing regimen is impressive, but requires further study to ascertain the outcome in the elderly-patient subgroup in the ongoing Phase II trial.

Maintenance treatment

Maintenance therapy postinduction or ASCT aims to improve PFS and OS, but must be weighed up against the potential treatment-related toxicities in a group that would otherwise enjoy a treatment-free period of remission. Furthermore, the potential resistance to subsequent salvage therapy as a result of ongoing drug exposure remains an unresolved issue Citation[19].

In the setting of post-ASCT, thalidomide maintenance has shown a trend towards improvement in OS in one meta-analysis Citation[69]. Two Citation[70,71] out of the five randomized studies Citation[70–74] of thalidomide maintenance following ASCT have shown benefits with respect to OS. However, these studies pertain to a younger patient population and are extrapolated to older patients undergoing ASCT. Thalidomide maintenance, especially when applied to elderly patients, is not without side effects, which include lethargy, sedation, peripheral neuropathy and constipation. The incidence of thalidomide-induced neuropathy is both cumulative and dose-related and, generally, patients only tolerate lower doses of thalidomide (∼100 mg) for 12–24 months. The benefit of maintenance therapy with other novel agents such as lenalidomide Citation[75] or bortezomib Citation[76] following ASCT is being evaluated in ongoing trials.

In patients not proceeding towards ASCT, the ability of maintenance therapy post-induction treatment to improve OS is still unclear in the era of novel therapeutic agents. In a randomized comparison between maintenance thalidomide (100 mg/day) plus dexamethasone (20 mg daily on days 1–4 every 28 days) or interferon three-times weekly, following induction with thalidomide, dexamethasone and pegylated liposomal doxorubicin, Offidani and colleagues demonstrated a superior 2-year PFS and OS with thalidomide-based maintenance (PFS 63 vs 32%; p = 0.02; OS 84 vs 68%; p = 0.03) Citation[77]. However, in the setting of MPT, survival postrelapse appeared to be shorter in patients with ongoing thalidomide maintenance compared with patients who received MP Citation[19]. This was not observed in MPT trials that ceased thalidomide after induction therapy Citation[39], raising the question of whether thalidomide maintenance could induce drug resistance, which would potentially compromise duration of response and survival postrelapse.

Like thalidomide, maintenance therapy with bortezomib Citation[32] or lenalidomide Citation[78] post-induction appears to improve responses, but the impact on OS remains to be seen. On preliminary analysis, the randomized trial of MPR with or without lenalidomide maintenance versus MP alone, showed that the addition of lenalidomide maintenance improved the quality of responses and reduced the risk of progression (HR: 0.42; p < 0.001) Citation[65]. Similarly, in a recent Spanish study, ongoing maintenance with VT after induction with VTP or VMP improved CR rates from 24% after induction to an impressive 42% in patients randomized to the maintenance arm Citation[32]. This was echoed by the recent Italian study, which showed that maintenance therapy for at least 6 months with VT after VMPT induction further improved the CR rate from 58 to 62% Citation[33]. Such a CR rate is unprecedented for elderly patients with MM and will hopefully translate to improve OS with longer-term follow-up. Importantly, maintenance with VT was well tolerated with a low incidence of grade 3 and 4 toxicity including peripheral neuropathy Citation[32].

Treatment of relapsed elderly patients with myeloma

The principles guiding the choice and sequence of salvage therapy for elderly patients, like young patients, are dependent on the nature and response to past treatments and speed of relapse, but with emphasis placed on current comorbidities that may impinge on treatment tolerance. Many effective salvage options have emerged that combine one or more of thalidomide, lenalidomide and bortezomib, with or without chemotherapy and with or without corticosteroids Citation[51,54,79–85]. Owing to the heterogeneity and effectiveness of salvage therapy at subsequent relapses, it is difficult to compare OS outcome between different treatment regimens. However, incorporation of novel agents at first relapse appears to produce superior outcomes compared with their use as later lines of salvage treatment Citation[86,87].

Often, the choice of which novel agent to use at relapse not only depends on availability, but also on individual preference (oral vs intravenous route of administration) and, importantly, comorbidities. In patients with pre-existing neuropathy, exacerbation may occur with bortezomib or thalidomide. Peripheral neuropathy is dose related and is usually reversible if prompt dose reduction or temporary cessation of bortezomib or thalidomide is applied upon the onset of grade ≥2 severity Citation[88,89]. For patients with a previous history of VTE or who are at high risk of VTE events, thalidomide and lenalidomide, although not absolutely contraindicated, should be avoided if alternate effective options are available. VTE risks are highest when these immunomodulatory drugs are used with high-dose dexamethasone or anthracycline chemotherapy. Prophylactic or therapeutic-dose anticoagulation should therefore be instituted as appropriate Citation[90]. Lenalidomide is cleared via the renal route and is not generally the first choice of treatment in patients with moderate-to-severe renal impairment. Thalidomide or bortezomib are preferred in such patients, especially as the latter has been shown to potentially reverse the poor prognosis implicated by renal impairment Citation[18].

In patients with rapidly progressive disease, intensive regimens combining novel agents with chemotherapy can be considered in selected elderly patients with good performance status and organ function Citation[54,79,81–83]. By contrast, patients with slow disease progression may do well with single-agent novel therapy with or without dexamethasone, especially if they cannot tolerate more intensive treatment. Similarly, HDT and ASCT could be considered at early relapses for a selected group of well-functioning elderly patients, particularly as this option is not viable with the decline in performance status and advancing age at subsequent relapses. If relapse occurs over 6 months following cessation of the last treatment regimen, the same regimen can be reconsidered, however, an inferior duration and quality of response is to be expected. When all different novel agent combinations have been exhausted, conventional moderate doses of cyclophosphamide Citation[91], nonmyeloablative doses of melphalan Citation[92] or low-to-modest doses of corticosteroids remain viable options, as is palliation in patients who cannot tolerate any further therapy.

Special treatment considerations for elderly patients

The management of elderly patients with MM must focus on issues beyond that of disease control given the likelihood of multiple comorbidities. The chance of succumbing to any complications is higher and the stepwise clinical deterioration associated with each complication is often substantial, underlining the importance of preventative and supportive therapy. For instance, elderly patients, especially those with concomitant cardiorespiratory disease, tolerate anemia poorly. Apart from blood transfusions, recombinant human erythropoietin is beneficial and has been shown to improve fatigue score and quality of life in patients with MM Citation[93]. Vigilance regarding VTE risks is warranted, especially when recombinant human erythropoietin is used in combination with thalidomide or lenalidomide. VTE prophylaxis with either aspirin, prophylactic-dose low-molecular-weight heparin or therapeutic-dose warfarin should be used according to VTE risk stratification Citation[90].

Infectious complications can result in significant clinical decline and must be treated promptly if not prevented. Elderly patients are more susceptible to varicella-zoster virus reactivation, which can potentially result in debilitating post-herpetic neuralgia. Patients treated with bortezomib should be on prophylaxis against Varicella Zoster virus with aciclovir, especially when used in combination with high-dose corticosteroids Citation[94]. Prophylaxis against Pneumocystis jirovecii (carinii) with trimethoprim and sulfamethoxazole should be considered in patients receiving high-dose corticosteroids. In patients with recurrent pneumonia and marked hypoglobulinemia, monthly infusions of intravenous immunoglobulin may help to prevent infections. Impaired renal function is more common in elderly patients, necessitating the dose reduction of medications that are renally cleared, such as lenalidomide.

Increasingly, it is recognized that dose reduction of many antimyeloma therapies may benefit elderly patients in whom the delivery of standard-dose therapy is difficult. In the MM-009 and MM-010 trials, treatment-related toxicities requiring lenalidomide dose reduction (from a starting dose of 25 mg) were more common in elderly patients and patients with renal impairment. Lower doses of lenalidomide are better tolerated in this patient group. Our preliminary data of 15 mg of lenalidomide in combination with lower-dose dexamethasone (20 mg orally daily on days 1–4, 9–12 and 17–20 every 28 days) appear to reduce myelosuppression, without reduced efficacy in a group of elderly patients with or without renal impairment compared with standard-dose lenalidomide (25 mg) and dexamethasone Citation[95]. Similarly, the use of lower-dose bortezomib (weekly as opposed to twice weekly) appear to result in less neurological and other nonhematological toxicities without compromising efficacy Citation[64]. Dose reduction of corticosteroids is often warranted in elderly patients, in whom diabetes, mood disorders and increased risk of infections are more common Citation[55].

Finally, the issue of polypharmacy and drug interactions is especially relevant to elderly patients. For example, thalidomide-induced constipation may be exacerbated by opiod analgesics; bradycardia associated with both thalidomide and lenalidomide warrants caution when used in conjunction with β-blockers for cardiovascular diseases.

Are we making progress in the treatment of MM for elderly patients?

Undoubtedly, the addition of novel therapeutic agents to conventional therapy and the expansion of effective salvage options have increased the outcome expectation for elderly patients with MM. Unprecedented CR rates can now be achieved in elderly patients, for example, 38% CR with VMPT–VT Citation[33], compared with 24–33% with MPV and <5% within the MP era. Practically however, the standard incorporation of novel agents in the treatment of elderly patients has trailed behind the publications of clinical trials that have shown improved outcome for this older subgroup. Even in so-called developed countries, limitations of access to certain novel agents, either due to economic or regulatory constraints, means that some treatment options are still either restricted to certain phases of disease or not available at all. Thus, survival estimates predicted by clinical trials may not reflect what we see in real-life clinical practice. This partially accounts for the apparent lack of survival improvements in elderly patients in recent reports Citation[3] compared with younger patients in whom clinical trials and the application of innovative approaches have been established much earlier in the historical timeline.

Expert commentary

Increasingly, the clinical approach for older patients with MM requires individualization due to the expanding array of treatment options. ‘Elderly patients’ are no longer synonymous with persons not eligible for HDT and ASCT, given that the enhanced safety of this treatment modality has made it feasible in selected patients up to 70 years of age Citation[47,96]. Indeed, induction therapies that combine novel agents with corticosteroids, with or without chemotherapy, have induced impressive rates of CR even prior to ASCT. Deferring ASCT in this situation is debatable. However, at present, we prefer to proceed to ASCT following up-front induction in eligible patients up to the age of 70 years, as this further improves the depth of responses Citation[59,97] and may not be feasible at future relapse upon declining performance status and further advancing age. In patients not eligible for ASCT, we would use a novel therapeutic agent in combination with chemotherapy and corticosteroids. Currently, either MPT Citation[39,61] or MPV Citation[6] are appropriate induction regimens and are clearly superior to MP. MPR Citation[65], as well as more intensive regimens combining two novel agents Citation[32,33], appears promising and we eagerly await final results. Vigilance regarding treatment-related toxicity is especially relevant in older patients in whom multiple comorbidities may limit the intensity of deliverable treatment. In particular, recognition is required of a small subgroup of elderly patients above 80–85 years of age who cannot tolerate toxicities and in whom reduced doses of MP, monotherapy with novel agents or corticosteroids, or palliation therapy is more appropriate. The wide spectrum of treatment-intensity options for elderly patients with MM is a result of our growing recognition of the clinical heterogeneity of this population. Increasingly, a balancing act is required from the treating physician to recognize on the one hand, ‘fit’ elderly patients who will be potentially disadvantaged by undertreatment and on the other, frailer elderly patients in whom the toxicities of intensive treatment may outweigh any potential survival benefit.

Five-year view

Treatment of MM for elderly patients will continue to evolve over the next 5 years, as long-term data materialize and more clinical trials emerge focusing on this subgroup of patients. Long-term survival data will probably confirm the positive impact on survival in elderly patients with MM, made possible by the use of novel therapeutic agents. Questions regarding the optimal front-line treatment, in particular the role of HDT and ASCT in improving depth of CRs in the era of novel agents, will be elucidated upon longer follow-up and recommendations can then be made based on robust survival data.

Complete response is becoming an increasingly achievable goal for elderly patients Citation[6,32,40] and has emerged as an independent predictor of long-term survival regardless of age Citation[40]. This fact has, and will, prompt further studies utilizing complex combinations of two or more novel agents with or without chemotherapy and maintenance, in an attempt to increase the rate of CR for elderly patients. The future will see fine-tuning of dosage and schedule of such multiple drug combinations to minimize toxicity.

The use of free-light-chain assay, multiparameter flow cytometry and PCR may be incorporated into routine practice in order to detect a deeper level of CR compared with immunofixation Citation[7,32]. This will probably create a new standard of CR and a more ambitious treatment goal for both young and old patients with MM. As these techniques improve, a role for minimal disease detection in deciding treatment intensity and maintenance therapy may emerge.

Finally, new classes of antimyeloma agents will continue to surface. Histone deacetylase inhibitors Citation[98] have already shown potential synergism with bortezomib or lenalidomide Citation[99,100] or chemotherapy Citation[101] in relapsed MM. Other families, including heat shock protein-90 inhibitors Citation[102,103], promise synergistic activities with the current novel therapeutic agents. The side-effect profile of these novel agents, especially when used in combination with currently available therapy, needs to be carefully assessed. Importantly, assessments of quality of life, which hitherto have been lacking in MM studies, need to be incorporated into future studies and are especially relevant for studies involving older patients with MM. Indeed, the challenge in enhancing survival for elderly patients not only rests with ongoing research into tolerable novel treatment regimens, but importantly, scrupulous supportive care and the judicious use of the currently available novel therapeutic agents in appropriate dosing, combinations and sequence, so as to minimize toxicities while maximizing disease control and so improving patient outcome.

Table 1. Examples of induction regimens for elderly patients not eligible for high-dose therapy and autologous stem-cell transplantation.

Table 2. Phase III studies comparing melphalan and prednisone with melphalan, prednisone and thalidomide for the upfront treatment of multiple myeloma.

Key issues

  • • Improved survival in patients with multiple myeloma is a direct consequence of novel therapeutic agents.

  • • Treatment-related toxicities have retrenched this improvement in the elderly patient population.

  • • Achievement of a complete response is an independent predictor of long-term survival irrespective of age; however, the goal of achieving complete response must be balanced with treatment-related toxicities for elderly patients; thus, individualization of treatment based on biological age is required.

  • • Ongoing research into tolerable treatment regimens, effective supportive care and correct dosing will optimize disease control, minimize toxicities and continue to improve survival for elderly patients with multiple myeloma in the future.

References

  • Kumar SK, Rajkumar SV, Dispenzieri A et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood111(5), 2516–2520 (2008).
  • Barlogie B, Pineda-Roman M, van Rhee F et al. Thalidomide arm of total therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities. Blood112(8), 3115–3121 (2008).
  • Brenner H, Gondos A, Pulte D. Expected long-term survival of patients diagnosed with multiple myeloma in 2006–2010. Haematologica94(2), 270–275 (2009).
  • Clavio M, Casciaro S, Gatti AM et al. Multiple myeloma in the elderly: clinical features and response to treatment in 113 patients. Haematologica81(3), 238–344 (1996).
  • Froom P, Quitt M, Aghai E. Multiple myeloma in the geriatric patient. Cancer66(5), 965–967 (1990).
  • Harousseau JL, Palumbo A, Richardson PG et al. Superior outcomes associated with complete response in newly diagnosed multiple myeloma patients treated with nonintensive therapy: analysis of the Phase 3 VISTA study of bortezomib plus melphalan–prednisone versus melphalan–prednisone. Blood116(19), 3743–3750 (2010).
  • Chanan-Khan AA, Giralt S. Importance of achieving a complete response in multiple myeloma, and the impact of novel agents. J. Clin. Oncol.28(15), 2612–2624 (2010).
  • Spencer A. No longer the poor relations. Blood116(19), 3685–3686 (2010).
  • Landgren O, Weiss BM. Patterns of monoclonal gammopathy of undetermined significance and multiple myeloma in various ethnic/racial groups: support for genetic factors in pathogenesis. Leukemia23(10), 1691–1697 (2009).
  • 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 Clinic Proc.85(3), 225–230 (2010).
  • Devesa SS, Silverman DT. Cancer incidence and mortality trends in the United States: 1935–74. J. Natl Cancer Inst.60(3), 545–571 (1978).
  • Velez R, Beral V, Cuzick J. Increasing trends of multiple myeloma mortality in England and Wales; 1950–79, are the changes real? J. Natl Cancer Inst.69(2), 387–392 (1982).
  • Levi F, Lucchini F, Negri E, Boyle P, La Vecchia C. Cancer mortality in Europe, 1995–1999, and an overview of trends since 1960. Int. J. Cancer110(2), 155–169 (2004).
  • Attal M, Harousseau JL, Stoppa AM et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N. Engl. J. Med.335(2), 91–97 (1996).
  • Child JA, Morgan GJ, Davies FE et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N. Engl. J. Med.348(19), 1875–1883 (2003).
  • Anagnostopoulos A, Gika D, Symeonidis A et al. Multiple myeloma in elderly patients: prognostic factors and outcome. Eur. J. Hematol.75(5), 370–375 (2005).
  • Kristinsson SY, Landgren O, Dickman PW, Derolf AR, Bjorkholm M. Patterns of survival in multiple myeloma: a population-based study of patients diagnosed in Sweden from 1973 to 2003. J. Clin. Oncol.25(15), 1993–1999 (2007).
  • San Miguel JF, Schlag R, Khuageva NK et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N. Engl. J. Med.359(9), 906–917 (2008).
  • 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. Blood112(8), 3107–3114 (2008).
  • Rodon P, Linassier C, Gauvain JB et al. Multiple myeloma in elderly patients: presenting features and outcome. Eur. J. Hematol.66(1), 11–17 (2001).
  • Quach H, Prince HM, Mileshkin L. Treatment strategies in elderly patients with multiple myeloma: current status. Drugs Aging24(10), 829–850 (2007).
  • Palumbo A, Triolo S, Baldini L et al. Dose-intensive melphalan with stem cell support (CM regimen) is effective and well tolerated in elderly myeloma patients. Haematologica85(5), 508–513 (2000).
  • Badros A, Barlogie B, Siegel E et al. Autologous stem cell transplantation in elderly multiple myeloma patients over the age of 70 years. Br. J. Hematol.114(3), 600–607 (2001).
  • Richardson PG, Sonneveld P, Schuster MW et al. Safety and efficacy of bortezomib in high-risk and elderly patients with relapsed multiple myeloma. Br. J. Hematol.137(5), 429–435 (2007).
  • Blade J, Lopez-Guillermo A, Bosch F et al. Impact of response to treatment on survival in multiple myeloma: results in a series of 243 patients. Br. J. Hematol.88(1), 117–121 (1994).
  • Oivanen TM, Kellokumpu-Lehtinen P, Koivisto AM, Koivunen E, Palva I. Response level and survival after conventional chemotherapy for multiple myeloma: a Finnish Leukaemia Group study. Eur. J. Hematol.62(2), 109–116 (1999).
  • Blade J, Samson D, Reece D et al. Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation. Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant. Br. J. Hematol.102(5), 1115–1123 (1998).
  • Durie BG, Harousseau JL, Miguel JS et al. International uniform response criteria for multiple myeloma. Leukemia20(12), 2220 (2006).
  • Barlogie B, Jagannath S, Vesole DH et al. Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood89(3), 789–793 (1997).
  • Davies FE, Forsyth PD, Rawstron AC et al. The impact of attaining a minimal disease state after high-dose melphalan and autologous transplantation for multiple myeloma. Br. J. Hematol.112(3), 814–819 (2001).
  • Alexanian R, Weber D, Giralt S et al. Impact of complete remission with intensive therapy in patients with responsive multiple myeloma. Bone Marrow Transplant.27(10), 1037–1043 (2001).
  • Mateos MV, Oriol A, Martinez-Lopez J et al. Bortezomib, melphalan, and prednisone versus bortezomib, thalidomide, and prednisone as induction therapy followed by maintenance treatment with bortezomib and thalidomide versus bortezomib and prednisone in elderly patients with untreated multiple myeloma: a randomised trial. Lancet Oncol.11(10), 934–941 (2010).
  • 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.28(34), 5101–5109 (2010).
  • Niesvizky R, Richardson PG, Rajkumar SV et al. The relationship between quality of response and clinical benefit for patients treated on the bortezomib arm of the international, randomized, Phase 3 APEX trial in relapsed multiple myeloma. Br. J. Hematol.143(1), 46–53 (2008).
  • Harousseau J, Weber D, Dimopoulos M et al. Relapsed/refractory multiple myeloma patients treated with lenalidomide/dexamethasone who achieve a complete or near complete response have longer overall survival and time to progression compared with patients achieving a partial response. Blood110, 3598 (2007) (Abstract).
  • Quach H, Mileshkin L, Seymour JF et al. Predicting durable remissions following thalidomide therapy for relapsed myeloma. Leuk. Lymph.50(2), 223–229 (2009).
  • van de Velde HJ, Liu X, Chen G, Cakana A, Deraedt W, Bayssas M. Complete response correlates with long-term survival and progression-free survival in high-dose therapy in multiple myeloma. Haematologica92(10), 1399–1406 (2007).
  • Palumbo A, Bringhen S, Caravita T et al. Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial. Lancet367(9513), 825–831 (2006).
  • Facon T, Mary JY, Hulin C et al. 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. Lancet370(9594), 1209–1218 (2007).
  • Gay F, Larocca A, Wijermans P et al. Complete response correlates with long-term progression-free and overall survivals in elderly myeloma treated with novel agents: analysis of 1175 patients. Blood117(11), 3025–3031 (2011).
  • Ludwig H, Hajek R, Tothova E et al. Thalidomide–dexamethasone compared to melphalan–prednisolone in elderly patients with multiple myeloma. Blood113(15), 3435–3442 (2009).
  • Pineda-Roman M, Bolejack V, Arzoumanian V et al. Complete response in myeloma extends survival without, but not with history of prior monoclonal gammopathy of undetermined significance or smouldering disease. Br. J. Hematol.136(3), 393–399 (2007).
  • Zhan F, Barlogie B, Arzoumanian V et al. Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis. Blood109(4), 1692–1700 (2007).
  • Harousseau JL, Avet-Loiseau H, Attal M et al. Achievement of at least very good partial response is a simple and robust prognostic factor in patients with multiple myeloma treated with high-dose therapy: long-term analysis of the IFM 99-02 and 99-04 trials. J. Clin. Oncol.27(34), 5720–5726 (2009).
  • Jantunen E. Autologous stem cell transplantation beyond 60 years of age. Bone Marrow Transplant.38(11), 715–720 (2006).
  • Mehta J, Singhal S. High-dose chemotherapy and autologous hematopoietic stem cell transplantation in myeloma patients under the age of 65 years. Bone Marrow Transplant.40(12), 1101–1114 (2007).
  • Sirohi B, Powles R, Treleaven J et al. The role of autologous transplantation in patients with multiple myeloma aged 65 years and over. Bone Marrow Transplant.25(5), 533–539 (2000).
  • Palumbo A, Triolo S, Argentino C et al. Dose-intensive melphalan with stem cell support (MEL100) is superior to standard treatment in elderly myeloma patients. Blood94(4), 1248–1253 (1999).
  • Spencer A, Seldon M, Deveridge S et al. Induction with oral chemotherapy (CID) followed by early autologous stem cell transplantation for de novo multiple myeloma patients. Hematol. J.5(3), 216–221 (2004).
  • Wu P, Davies FE, Horton C et al. The combination of cyclophosphomide, thalidomide and dexamethasone is an effective alternative to cyclophosphamide–vincristine–doxorubicin–methylprednisolone as induction chemotherapy prior to autologous transplantation for multiple myeloma: a case-matched analysis. Leuk. Lymph.47(11), 2335–2338 (2006).
  • Kyriakou C, Thomson K, D’Sa S et al. Low-dose thalidomide in combination with oral weekly cyclophosphamide and pulsed dexamethasone is a well tolerated and effective regimen in patients with relapsed and refractory multiple myeloma. Br. J. Hematol.129(6), 763–770 (2005).
  • Harousseau JL, Attal M, Avet-Loiseau H et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 Phase III trial. J. Clin. Oncol.28(30), 4621–4629 (2010).
  • Popat R, Oakervee HE, Hallam S et al. Bortezomib, doxorubicin and dexamethasone (PAD) front-line treatment of multiple myeloma: updated results after long-term follow-up. Br. J. Hematol.141(4), 512–516 (2008).
  • Morgan GJ, Schey SA, Wu P et al. Lenalidomide (Revlimid), in combination with cyclophosphamide and dexamethasone (RCD), is an effective and tolerated regimen for myeloma patients. Br. J. Hematol.137(3), 268–269 (2007).
  • Rajkumar SV, Jacobus S, Callander NS et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol.11(1), 29–37 (2010).
  • Wang M, Delasalle K, Giralt S, Alexanian R. Rapid control of previously untreated multiple myeloma with bortezomib–lenalidomide–dexamethasone (BLD). Hematology15(2), 70–73 (2010).
  • Richardson PG, Weller E, Lonial S et al. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood116(5), 679–686 (2010).
  • Dingli D, Pacheco JM, Nowakowski GS et al. Relationship between depth of response and outcome in multiple myeloma. J. Clin. Oncol.25(31), 4933–4937 (2007).
  • Dingli D, Gay F, Buadi F, Dispenzieri A, Gertz M. Depth of response with stem cell transplantation and outcome for multiple myeloma in the era of novel agents. Blood114, 506 (2009) (Abstract 1228).
  • Waage A, Gimsing P, Fayers P et al. Melphalan and prednisone plus thalidomide or placebo in elderly patients with multiple myeloma. Blood116(9), 1405–1412 (2010).
  • 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.27(22), 3664–3670 (2009).
  • 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. Hematol.86(1), 16–22 (2011).
  • Kapoor P, Rajkumar SV, Dispenzieri A et al. Melphalan and prednisone versus melphalan, prednisone and thalidomide for elderly and/or transplant ineligible patients with multiple myeloma: a meta-analysis. Leukemia25(4), 689–696 (2011).
  • Bringhen S, Larocca A, Rossi D et al. Efficacy and safety of once weekly bortezomib in multiple myeloma patients. Blood116(23), 4745–4753 (2010).
  • Palumbo A, Delforge M, Catalano J, Hajek R, Dimopoulos M. A Phase 3 study evaluating the efficacy and safety of lenalidomide combined with melphalan and prednisone in patients ≥65 years with newly diagnosed multiple myeloma (NDMM), continous use of lenalidomide vs. fixed-duration regimens. Blood116, 662 (2010).
  • Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR. Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J. Clin. Oncol.24(3), 431–436 (2006).
  • Cavo M, Zamagni E, Tosi P et al. Superiority of thalidomide and dexamethasone over vincristine-doxorubicindexamethasone (VAD) as primary therapy in preparation for autologous transplantation for multiple myeloma. Blood106(1), 35–39 (2005).
  • Gay F, Hayman SR, Lacy MQ et al. Lenalidomide plus dexamethasone versus thalidomide plus dexamethasone in newly diagnosed multiple myeloma: a comparative analysis of 411 patients. Blood115(7), 1343–1350 (2010).
  • Hicks LK, Haynes AE, Reece DE et al. A meta-analysis and systematic review of thalidomide for patients with previously untreated multiple myeloma. Cancer Treatment Rev.34(5), 442–452 (2008).
  • Spencer A, Prince HM, Roberts AW et al. Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure. J. Clin. Oncol.27(11), 1788–1793 (2009).
  • Attal M, Harousseau JL, Leyvraz S et al. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood108(10), 3289–3294 (2006).
  • Lokhorst HM, van der Holt B, Zweegman S et al. A randomized Phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma. Blood115(6), 1113–1120 (2010).
  • Barlogie B, Tricot G, Anaissie E et al. Thalidomide and hematopoietic-cell transplantation for multiple myeloma. N. Engl. J. Med.354(10), 1021–1030 (2006).
  • Feyler S, Rawstron A, Jackson G, Snowden JA, Cocks K, Johnson RJ. Thalidomide maintenance following high-dose therapy in multiple myeloma: a UK myeloma forum Phase 2 study. Br. J. Hematol.139(3), 429–433 (2007).
  • McCarthy P, Owzar K, Stadtmauer E, Giralt S, Anderson KC. Phase III intergroup study of lenalidomide (CC-5013) versus placebo maintenace therapy following single autologous stem cell transplant for multiple myeloma (CALBG 100104), initial report of patient accrual and averse events. Blood114, 3416a (2009).
  • Sahebi F, Krishnan A, Forman S. High rate of complete remission (CR) and upgraded response with weekly maintenance bortezomib post single autologous peripheral stem cell transplant (PSCT) in patients with multiple myeloma. Results of a Phase II prospective study. Blood116 (2010) (Abstract 2399).
  • Offidani M, Corvatta L, Polloni C et al. Thalidomide–dexamethasone versus interferon-α–dexamethasone as maintenance treatment after ThaDD induction for multiple myeloma: a prospective, multicentre, randomised study. Br. J. Hematol.144(5), 653–659 (2009).
  • Palumbo A, Dimopoulos M, Delforge M, Kropff M, Foa R, Catalano J. A Phase III study to determine the efficacy and safety of lenalidomide in combination with melphalan and prednisolone (mpr) in elderly patients with newly diagnosed multiple myeloma. Blood114 (2009) (Abstract 613a).
  • Palumbo A, Ambrosini MT, Benevolo G et al. Bortezomib, melphalan, prednisone, and thalidomide for relapsed multiple myeloma. Blood109(7), 2767–2772 (2007).
  • Anagnostopoulos A, Weber D, Rankin K, Delasalle K, Alexanian R. Thalidomide and dexamethasone for resistant multiple myeloma. Br. J. Hematol.121(5), 768–771 (2003).
  • Offidani M, Bringhen S, Corvatta L et al. Thalidomide–dexamethasone plus pegylated liposomal doxorubicin vs. thalidomide–dexamethasone: a case-matched study in advanced multiple myeloma. Eur. J. Hematol.78(4), 297–302 (2007).
  • Kropff M, Bisping G, Schuck E et al. Bortezomib in combination with intermediate-dose dexamethasone and continuous low-dose oral cyclophosphamide for relapsed multiple myeloma. Br. J. Hematol.138(3), 330–337 (2007).
  • Knop S, Gerecke C, Liebisch P et al. Lenalidomide, adriamycin, and dexamethasone (RAD) in patients with relapsed and refractory multiple myeloma: a report from the German Myeloma Study Group DSMM (Deutsche Studiengruppe Multiples Myelom). Blood113(18), 4137–4143 (2009).
  • Weber DM, Chen C, Niesvizky R et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N. Engl. J. Med.357(21), 2133–2142 (2007).
  • Dimopoulos M, Spencer A, Attal M et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N. Engl. J. Med.357(21), 2123–2132 (2007).
  • Stadtmauer EA, Weber DM, Niesvizky R et al. Lenalidomide in combination with dexamethasone at first relapse in comparison with its use as later salvage therapy in relapsed or refractory multiple myeloma. Eur. J. Hematol.82(6), 426–432 (2009).
  • Vogl DT, Stadtmauer EA, Richardson PG et al. Impact of prior therapies on the relative efficacy of bortezomib compared with dexamethasone in patients with relapsed/refractory multiple myeloma. Br. J. Hematol.147(4), 531–534 (2009).
  • Richardson PG, Sonneveld P, Schuster MW et al. Reversibility of symptomatic peripheral neuropathy with bortezomib in the Phase III APEX trial in relapsed multiple myeloma: impact of a dose-modification guideline. Br. J. Hematol.144(6), 895–903 (2009).
  • Dimopoulos MA, Mateos MV, Richardson PG et al. Risk factors for, and reversibility of, peripheral neuropathy associated with bortezomib–melphalan–prednisone in newly diagnosed patients with multiple myeloma: subanalysis of the Phase 3 VISTA study. Eur. J. Hematol.86(1), 23–31 (2011).
  • Lenhard RE Jr, Oken MM, Barnes JM, Humphrey RL, Glick JH, Silverstein MN. High-dose cyclophosphamide. An effective treatment for advanced refractory multiple myeloma. Cancer53(7), 1456–1460 (1984).
  • Sonneveld P, van der Holt B, Segeren CM et al. Intermediate-dose melphalan compared with myeloablative treatment in multiple myeloma: long-term follow-up of the Dutch Cooperative Group HOVON 24 trial. Haematologica92(7), 928–935 (2007).
  • Dammacco F, Castoldi G, Rodjer S. Efficacy of epoetin alfa in the treatment of anaemia of multiple myeloma. Br. J. Hematol.113(1), 172–179 (2001).
  • Palumbo A, Rajkumar SV, Dimopoulos MA et al. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia22(2), 414–423 (2008).
  • Vickrey E, Allen S, Mehta J, Singhal S. Acyclovir to prevent reactivation of varicella zoster virus (herpes zoster) in multiple myeloma patients receiving bortezomib therapy. Cancer115(1), 229–232 (2009).
  • Quach H, Fernyhough L, Henderson R, Corbett G, Lynch K, Prince HM. Lower-dose lenalidomide and dexamethasone reduces toxicity without compromising efficacy in patients with relapse/refractory myeloma, who are aged ≥60 years or have renal impairment: planned interim results of a prospective multicentre Phase II trial. Blood116 (2010) (Abstract 1961).
  • Siegel DS, Desikan KR, Mehta J et al. Age is not a prognostic variable with autotransplants for multiple myeloma. Blood93(1), 51–54 (1999).
  • Fermand JP, Katsahian S, Divine M et al. High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. J. Clin. Oncol.23(36), 9227–9233 (2005).
  • Niesvizky R, Ely S, Mark T et al. Phase 2 trial of the histone deacetylase inhibitor romidepsin for the treatment of refractory multiple myeloma. Cancer117(2), 336–342 (2011).
  • Kaiser M, Lamottke B, Mieth M et al. Synergistic action of the novel HSP90 inhibitor NVP-AUY922 with histone deacetylase inhibitors, melphalan, or doxorubicin in multiple myeloma. Eur. J. Hematol.84(4), 337–344 (2010).
  • Ocio EM, Vilanova D, Atadja P et al. In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica95(5), 794–803 (2010).
  • Sanchez E, Shen J, Steinberg J et al. The histone deacetylase inhibitor LBH589 enhances the anti-myeloma effects of chemotherapy in vitro and in vivo. Leuk.Res.35(3), 373–379 (2010).
  • Richardson PG, Mitsiades CS, Laubach JP, Lonial S, Chanan-Khan AA, Anderson KC. Inhibition of heat shock protein 90 (HSP90) as a therapeutic strategy for the treatment of myeloma and other cancers. Br. J. Hematol.152(4), 367–379 (2011).
  • Allegra A, Sant’antonio E, Penna G et al. Novel therapeutic strategies in multiple myeloma: role of the heat shock protein inhibitors. Eur. J. Hematol.86(2), 93–110 (2011).
  • Wijermans P, Schaafsma M, Termorshuizen F et al. 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.28(19), 3160–3166 (2010).

Website

Managing multiple myeloma in the elderly: are we making progress?

To obtain credit, you should first read the journal article. After reading the article, you should be able to answer the following, related, multiple-choice questions. To complete the questions and earn continuing medical education (CME) credit, please go to www.medscape.org/journal/experthematology. Credit cannot be obtained for tests completed on paper, although you may use the worksheet below to keep a record of your answers. You must be a registered user on Medscape.com. If you are not registered on Medscape.com, please click on the New Users: Free Registration link on the left hand side of the website to register. Only one answer is correct for each question. Once you successfully answer all post-test questions you will be able to view and/or print your certificate. For questions regarding the content of this activity, contact the accredited provider, [email protected]. For technical assistance, contact [email protected]. American Medical Association’s Physician’s Recognition Award (AMA PRA) credits are accepted in the US as evidence of participation in CME activities. For further information on this award, please refer to http://www.ama-assn.org/ama/pub/category/2922.html. The AMA has determined that physicians not licensed in the US who participate in this CME activity are eligible for AMA PRA Category 1 Credits™. Through agreements that the AMA has made with agencies in some countries, AMA PRA credit is acceptable as evidence of participation in CME activities. If you are not licensed in the US and want to obtain an AMA PRA CME credit, please complete the questions online, print the certificate and present it to your national medical association.

Activity Evaluation: Where 1 is strongly disagree and 5 is strongly agree

1. A 55-year-old man was diagnosed with multiple myeloma (MM) in 2010. Compared with being diagnosed in 2000, which of the following best describes his expected median survival?

  • A Similar

  • B Improved by less than 2 years

  • C Improved by 2–5 years

  • D Improved by 6–8 years

2. For the patient in question 1, which of the following is most likely to be predictive of progression-free survival and long-term outcome?

  • A Complete response

  • B Performance status

  • C Presence of comorbidities

  • D Response to melphalan

3. A 66-year-old woman is diagnosed with MM and has good performance status with one comorbidity. Which of the following is considered the most optimal upfront treatment for her?

  • A Stem cell-sparing induction treatment followed by ASCT

  • B High-dose melphalan and dexamethasone

  • C Conventional dose vincristine and doxorubicin

  • D All of the above are equally efficacious

4. In a 75-year-old man with high-risk cytogenetics MM and renal impairment, not eligible for ASCT, which of the following is likely to be the most optimal treatment strategy?

  • A Melphalan and prednisone

  • B High-dose melphalan, prednisone, and thalidomide

  • C Bortezomib, melphalan, and prednisone

  • D All of the above are similar in efficacy and toxicity

5. Beyond disease control, which of the following factor(s) is/are likely to poorly impact outcomes of MM treatment in elderly compared with younger patients?

  • A Poor tolerance of anemia

  • B Varicella zoster reactivation

  • CPneumocystis carinii infection

  • D All of the above

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.