Next Article in Journal
Temporal Dynamics and Clinical Predictors of Brain Metastasis in Breast Cancer: A Two-Decade Cohort Analysis Toward Tailored CNS Screening
Previous Article in Journal
Updates on the Treatment of Richter’s Syndrome, Including Novel Combination Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Challenges in Multiple Myeloma Therapy in Older and Frail Patients

by
Anna Aureli
1,*,
Beatrice Marziani
2,†,
Tommaso Sconocchia
3,†,
Gianmario Pasqualone
4,
Luca Franceschini
4,
Giulio Cesare Spagnoli
1,
Adriano Venditti
4 and
Giuseppe Sconocchia
1,*
1
CNR Institute of Translational Pharmacology, 67100 L’Aquila, Italy
2
Emergency Medicine Department, Sant’Anna University Hospital, Via A. Moro, 8, Cona, 44124 Ferrara, Italy
3
San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy
4
Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(6), 944; https://doi.org/10.3390/cancers17060944
Submission received: 10 January 2025 / Revised: 19 February 2025 / Accepted: 7 March 2025 / Published: 11 March 2025
(This article belongs to the Section Cancer Immunology and Immunotherapy)

Simple Summary

MM incidence is increasing globally, particularly in high-income countries and in the male population aged ≥ 50. While advances in treatment strategies have led to improvement of survival over the past decades, MM remains incurable in a large percentage of cases. Importantly, about one third of patients diagnosed with MM are >75 years old and are characterized by relatively low fitness or frailty. Few older and frail patients diagnosed with multiple myeloma (MM) are currently enrolled in pivotal clinical trials, and little data are presently available on appropriate management. These patients are more vulnerable to stressors and present lower resistance to cancer and related treatments. Here, we reviewed relevant studies analyzing myeloma treatments in frail and older patients, and we analyzed their effectiveness and safety.

Abstract

Every year, more than 150,000 cases of multiple myeloma (MM) are diagnosed worldwide, and over 100,000 deaths caused by this malignancy are recorded. MM incidence is increasing globally, particularly in high-income countries and in the male population aged ≥ 50. While advances in treatment strategies have led to the improvement of survival over the past decades, MM remains incurable in a large percentage of cases. Importantly, about one-third of patients diagnosed with MM are >75 years old and are characterized by relatively low fitness or frailty. These patients are more vulnerable to stressors and present lower resistance to cancer and related treatments. Therefore, patients’ fitness and frailty should become part of the standard assessment in MM, and flexible therapeutic options should apply. A careful review of studies investigating the management of frail patients with MM strongly supports the use of a dynamic evaluation of effectiveness and adverse events associated with current treatments in the context of patient-specific frailty and co-morbidities.

1. Introduction

Multiple myeloma (MM) is an aggressive hematologic malignancy characterized by an excess of plasma cells (PCs) in the bone marrow (BM). Its progression starts with a pre-malignant monoclonal gammopathy of undetermined significance (MGUS) [1], followed by an intermediate asymptomatic stage, smoldering multiple myeloma (SMM) [2], which preludes MM development in high percentages of patients [2].
Malignant transformation begins inside lymph nodes’ germinal centers, where B-lymphocytes undergo genetic and epigenetic alterations before moving into BM, where they differentiate into PCs by interacting with the immune microenvironment [3]. Malignant PCs produce serum immunoglobulins and activate osteoclasts, resulting in bone tissue degradation, and focal injuries, leading to bone fractures [4].
Cytogenetic anomalies such as immunoglobulin heavy-chain locus translocations and hyperdiploid karyotypes are already detectable in premalignant phases [5], but only 1% of MGUS cases progress to MM [6], suggesting that the switch to symptomatic disease may be due to sudden, low-frequency, randomly acquired genomic events [7].
While progression to SMM does not usually produce end-organ damage [8], in some cases, MM features might already be detectable [9] with malignant plasma cell levels > 10% in the BM and unusually large numbers of “M spike” serum proteins. The weakening of the immune system, anemia, bleeding disorders, hypercalcemia, kidney failure, and bone lesions are some of the symptoms observed in advanced MM stages [10,11,12].
In 2014, the International Myeloma Working Group (IMWG) updated MM diagnostic criteria for MM to allow for earlier diagnosis and the initiation of treatment prior to the occurrence of organ damage. Important progress has been made since, with significant improvements in survival [11,12]. Nevertheless, further efforts and additional resources are needed to successfully treat MM, particularly in the relevant percentages of unfit and frail patients who do not tolerate and/or recover from systemic therapy and require modifications of treatment protocols. Evaluating the functional health status of patients with MM is as important as that of myeloma-related risk factors. Indeed, patients aged ≥ 65 and those with long-term disabilities are known to be highly heterogeneous and require proper assessment strategies to define their frailty profile and most appropriate treatments. Usually, they do not participate in clinical trials and are not eligible to receive treatments of choice, including hematopoietic stem cell transplantation, myeloablative chemotherapies, and enhanced supportive care, but undergo unconventional therapies. To provide strategies of potential use in the selection of relevant therapeutic options, in this review, we analyze treatment protocols recommended for “frail” and older patients with MM.

2. Frailty: Definition, Categorization, and Impact on Disease Outcomes

Aging is a gradual and progressive physiological process, associated with several modifications in vital organs and metabolism and increased susceptibility to multiple vulnerabilities. About one-third of newly diagnosed MM patients are >75 years old at diagnosis. Multiple comorbidities, frequently observed in older adults, including chronic kidney disease, osteoporosis, diabetes, obesity, high blood pressure, and various forms of heart disease, may contribute to confounding the clinical picture and delaying diagnosis, which is often reached when MM-related organ dysfunction is already present [13].
While different subgroups of older and frail patients may be identified based on specific characteristics, such as functional status, cognition, socio-economic factors, nutritional status, independence in daily activities, and fitness, they overall represent a poorly investigated population in clinical trials. Indeed, due to the normal decline of organ and system functions, older adults are at a higher risk of experiencing iatrogenic adverse events [14]. Therefore, extensive expertise and comprehensive geriatric assessment (GA) are required to correctly identify personalized management and therapy and to predict tolerance and adverse outcomes [15].
Several GA scoring systems have been proposed. The IMWG has designed a tool to stratify patients considering age, comorbidities, cognitive, and physical conditions [14,16]. The Freiburg Comorbidity Index (Initial Myeloma Comorbidity Index—I-MCI) is based on Karnofsky performance status and on lung and renal impairment [17]. The Revised Myeloma Comorbidity Index (R-MCI) has extended its assessment to disease cytogenetics [18,19]. Moreover, the Geriatric Assessment in Hematology (GAH), a 30-item scale directed at older patients diagnosed with hematological malignancies, dedicates special attention to psychometric evaluation, including, in addition to standard comorbidities, polypharmacy, gait speed, mood, daily activities, health status, nutrition, and mental status [17,20]. Additional GA tools and scoring systems have also been developed more recently [21,22,23].
The goal of personalized treatment in older patients should be to provide a therapy that effectively prolongs survival without affecting patients’ quality of life. A balance between benefits in disease control and the conservation of quality of life, physical independence, and preventing hospitalization should be pursued. A global assessment should also include palliative therapies and hospice care. In this vein, multiple recommendations have been formulated to guide the use of specific drugs [15,24]. As an example, dexamethasone plays a key role in MM treatment. However, despite its effectiveness, high-dose therapy may be associated with neurological and psychiatric disorders, iatrogenic diabetes mellitus, gastrointestinal bleeding, higher risk of infection, and glucocorticoid-induced osteoporosis [15]. Moreover, its cardiovascular impact is non-negligible, particularly in frail and elderly patients. Prolonged use is related to the development or worsening of hypertension, fluid retention, and metabolic dysregulation, increasing the risk of arrhythmias and the decompensation of prior chronic heart failure [25]. The toxicity profile of this key player in MM treatment has led to the development of low-dose dexamethasone administration schedules intended for older and unfit patients. The formulation of fixed-duration treatment schedules to reduce toxicity while preserving effectiveness is currently under investigation [15,26,27].

3. Choice of Treatment Regimen in Older MM Patients Ineligible to Receive Transplantation

Frailty status represents a key factor in the management of older patients with MM. Those above 75 years of age are immediately classified as intermediately fit or frail [28] since the use of aggressive treatments in these patients may lead to negative outcomes. Indeed, it has been demonstrated that frail older patients with MM have worse progression-free (PFS) and overall survival (OS) in comparison to fit individuals and show higher rates of infection and treatment toxicity [19,29]. Therefore, personalized treatments should be administered according to the assigned frailty score. However, the use of different tools leads to high variability in the definition of frailty in MM, with prevalence rates ranging between 17.2% and 66.0% [21], and treatment is usually determined on a case-by-case basis. Nevertheless, patients aged over 75 are typically deemed transplant-ineligible.
The recognition of the importance of frailty status has led to reconsiderations of MM treatment. While Melphalan and prednisone with or without bortezomib or thalidomide (MPT, VMP, or MP) previously represented the standard of care for these patients [17], VRd (bortezomib–lenalidomide–dexamethasone) triplet therapy has subsequently been utilized based on the results of the SWOG S0777 phase 3 study, showing that the addition of bortezomib to lenalidomide–dexamethasone results in improved PFS and OS, irrespective of age [30,31]. These results were also confirmed by O’Donnell and colleagues, who reported that in patients older than 75 years requiring a reduction in the intensity of the VRd regimen, modifications to doses and schedules did not compromise treatment effectiveness [32]. Indeed, their data, obtained from a phase 2 study, underline that the modified VRd regimen was not only well tolerated and highly effective but also more appropriate for older patients [33].
More recently, the addition of anti-CD38 monoclonal antibodies (mAb), such as daratumumab, to similar drug combinations has led to the achievement of good and long-lasting responses in several patient groups. Treatment with DRd (daratumumab–lenalidomide–dexamethasone) or D-VMP (daratumumab plus bortezomib, melphalan, and prednisone) [11,13] has resulted in longer than 4/5-year survival [34]. Therefore, DRd and D-VMP should now be considered the gold standard first-line therapy for elderly and/or frail patients. A phase 3 clinical trial supported the first-line use of DRd for patients with MM ineligible for transplantation, regardless of frailty status [35]. Moreover, a further subgroup analysis, including fit, intermediate, and frail patients, has demonstrated that treatment with DRd results in improved PFS, OS, and minimal/measurable residual disease (MRD), as compared to Rd alone, regardless of age and frailty [36,37]. Remarkably, approval of subcutaneous daratumumab—which is considered non-inferior to intravenous formulation in terms of efficacy and pharmacokinetics and provides an improved safety profile also in relapsed/refractory MM patients—has marked a turning point in the management of frail patients with MM [38], with positive effects on patients’ quality of life as well [39]. Other studies have also underlined both a prolonged OS [40] and a 4-fold-higher MRD-negativity rate in patients treated with D-VMP compared to those treated with VMP alone, even in a frail subgroup [41,42,43]. However, DRd appears to be preferable to DVMP, except for patients with severe renal impairment and/or a recurrent history of thrombosis.
Most recently, the EMA has approved the use of isatuximab plus VRd for newly diagnosed MM in transplant-ineligible settings based on the results of a phase 3 trial (IMROZ ClinicalTrials.gov number, NCT03319667). Facon et al. demonstrated that the addition of the anti-CD38 monoclonal antibody isatuximab to the VRd regimen slowed disease progression in patients with MM from 18 to 80 years of age [44].
Comparing isatuximab plus VRd and VRd alone, a better PFS at 60 months was in fact highlighted in an isatuximab-VRd group compared with a VRd group (63.2% vs. 45.2%), with a similar safety profile [44].
Notably, however, the use of multiple-agent treatments in MM patients over 75 has also been shown to be associated with higher toxicity [45]. Within this context, it is important to consider that high-dose dexamethasone, included for almost 40 years in several drug combinations for MM treatment [46], is associated with a variety of adverse effects, including neurological disorders and, most importantly, increased susceptibility to infections, cardiovascular complications, and secondary diabetes mellitus [47,48,49].
Therefore, to allow older, intermediately fit patients to receive and tolerate sufficiently prolonged dexamethasone treatments, dose adjustments based on clinical, functional status and comorbidities have been proposed, aimed at managing potentially occurring adverse events [36]. In this context, Stege and colleagues [50] reported that in frail patients with a median age of 81 years, a low-dose dexamethasone treatment of up to 10 mg every 2/4 weeks had potential advantages and was not less effective. Moreover, in these patients, short-term treatments would be preferable [47,48], as also demonstrated by a randomized phase 3 study where a dose/schedule-adjusted Rd treatment without dexamethasone was used in the maintenance (Rd-R) of intermediately fit patients [26]. Other trials are underway aimed at determining the possibility of omitting dexamethasone in older and frail patients. Preliminary data from the IFM 2017_03 trial anticipate that in frail NDMM patients, the early discontinuation of dexamethasone (after two cycles) in the context of daratumumab and lenalidomide treatment is associated with effective and durable responses with a manageable safety profile [15].
The choice of treatment for relapsing elderly patients is even more challenging. A series of factors, including front-line treatment, patient age, long-term side effects, comorbidities, and the presence of cytogenetic alterations, needs to be considered [51,52]. Data from relapsed/refractory MM frail patients enrolled in the CANDOR trial have shown that daratumumab in combination with Kd56 (D-Kd) is effective and safe and results in improved PFS without additional toxicity [53]. Importantly, cardiac failure has been shown to occur less frequently in frail patients treated with dexamethasone [53].
Table 1 summarizes suggestions for the tailored treatment of patients with MM, with different levels of frailty or defined co-morbidities.
Altogether, the data available from real-world practice regarding older and frail patients are far from optimal. A retrospective analysis of real-world outcomes in about 5000 MM European patients has underlined that only 61% of these patients reach second-line treatment, and only 38% reach the third line [54]. Patients aged ≥ 70 years remain at the highest risk of early mortality in the presence of a variety of aging-related vulnerabilities, resulting in a highly heterogeneous population with diverse clinical outcomes. Therefore, tailored therapeutic approaches are required, carefully balancing effectiveness and treatment burden.

4. CAR-T Cell Therapy in Older Patients

Most recently, MM treatment has been revolutionized by the application of targeted therapies, including those with chimeric antigen receptor (CAR) T cells, currently also used in relapsed or refractory MM [55]. Data from the phase 3 CARTITUDE-4 clinical trial have shown that the administration of CAR-T cells targeting the B cell maturation antigen (BCMA) reduces the risk of disease progression or death by 74% compared to two standard-of-care regimens in patients with relapsed and lenalidomide-refractory MM who have previously received one to three lines of treatment [55].
The safety and effectiveness of CAR-T have also been evaluated in elderly patients with R/R MM [56]. Tolerable safety and reasonable effectiveness have been observed [56], although frailer patients have higher treatment-related mortality. Moreover, all grades of immune-effector-cell-associated neurotoxicity syndrome (ICANS) are slightly higher in the elderly population, even if no differences in grade 3 or higher ICANS are observed [56]. Nevertheless, reports on the use of CAR-T cell therapies in older patients are still scarce, the most relevant question being, “How old is too old for CAR-T cell therapies in multiple myeloma?” [57]. Clearly, although age, per se, should not automatically exclude patients from such treatments, related toxicities have to be considered, including not only the most severe events but also low-grade ones impacting quality of life and clinical outcomes.
As underlined by Rajeeve and Usmani [57], to correctly manage treatment-related toxicities, it is critically important to define a correct sequence of therapies. However, to be eligible for CAR-T clinical trials, patients have to meet the strict criteria of performance status and homeostasis regulation that were established to de-risk the development of treatment-related toxicities. Unfortunately, very few elderly patients meet these requirements. Accordingly, the oldest patients included in two important CAR-T phase 2 clinical trials, namely, KarMMa [58] and CARTITUDE-1 [59], were 78 and 70 years old, respectively. Since the median age at MM diagnosis is 69 years, while the median age of patients admitted to phase 2 clinical trials is 61 years, clearly, older patients tend to be excluded from CAR-T therapy. However, interestingly, retrospective meta-analyses underline that anti-BCMA CAR-T cell therapy in older patients with relapsed MM is as safe and effective as in younger patients [60]. Consistently, in a cohort of 83 patients, including 22 ≥70-year-olds, Reyes et al. observed that CRS and ICANS rates, days to absolute neutrophil count recovery, and the incidence of hypogammaglobulinemia were comparable in older and younger patients, without specific geriatric complications [61]. An optimistic view of CAR-T cell therapy in older patients also emerges from a post hoc analysis of the ZUMA-1 trial, documenting similar clinical effectiveness and safety to that of younger patients [62].
On the other hand, Davis et al. reported that although older/frail patients with RRMM treated with CAR-T cell therapy in a real-world setting do not show excess high-grade toxicities, therapy is associated with lower efficacy, as compared to non-frail patients [56].
Taken together, these data support the concept that advanced chronological age, per se, cannot represent an absolute contraindication for CAR-T cell therapy and that physiologic age should instead be assessed as in defined US therapy programs. As an example, Yates et al. used a geriatric-assessment-guided multidisciplinary clinic (GA-MDC) as a tool to evaluate and select older adults for CAR-T cell therapy and to provide an effective risk stratification [63]. Therefore, it would be desirable to enclose geriatric assessments and frailty scores into cellular therapy risk assessment to standardize the selection processes, thereby possibly increasing the number of older patients with MM eligible for CAR-T cell therapy.

5. Bispecific Antibodies

Bispecific antibodies (BsAbs) are effectively used in RRMM treatment to promote the elimination of malignant cells using immune effector cells [64]. They include bispecific T cell engagers (BiTEs) and DuoBodies. The main targets are BCMA, G protein-coupled receptor class C group 5 member D (GPRC5D), and Fc receptor-like 5 (FcRH5) (Table 2).
BiTEs combine into one molecule, with a mAb binding a tumor-associated surface antigen and another binding CD3 in the absence of an Fc fragment. They mainly trigger CD8+ T cell cytotoxic activity against targeted tumor cells [65]. However, while they easily penetrate tumors thanks to their small size, they are usually characterized by a short half-life, requiring frequent/continuous infusion [66].
DuoBodies represent evolutions of BiTEs and include two different single-chain variable fragments linked by an Fc domain mediating antibody-dependent cytotoxicity (ADCC), phagocytosis, and complement-dependent cytotoxicity. Their increased half-life also allows intermittent dosing [67].
Table 2. BsAbs used in the management of patients with R/R MM.
Table 2. BsAbs used in the management of patients with R/R MM.
TargetsBsAbsStudyPatient NumberDosing Schedule/Efficacy (Ref.)
BCMATeclistamab (JNJ-64007957)Phase 1/2 MajesTEC-1 trial (NCT04557098); (NCT03145181)165
Patients ≥ 75 yr: 24 (14.5%).
Subcutaneous injection: 0.06 mg–0.3 mg–1.5 mg/kg once weekly. Deep and durable response [68,69].
Elranatab (PF-06863135)Phase 2 MagnetisMM-3 trial (NCT04649359cohort A)123
Patients ≥ 75 yr: 24 (19.5%).
Subcutaneous injection: 12–32–76 mg once weekly. Good efficacy and safety [70,71].
Linvoseltamab (REGN5458)Phase 1/2 LINKER-MM1 trial (NCT03761108)200 mg.: 117
Patients ≥ 75 yr: 31 (26.4%).
Intravenous injection: 5–25–200 mg once weekly. Consistent efficacy across high-risk subgroups and induced responses in pts who progressed on 50 ng [72,73].
Alnuctamab (CC-93269)Phase 1 trial (NCT03486067)73
Patients ≥ 75 yr: n.a.
Target dose: subcutaneous injection of 30 mg once weekly (cycle 1 to 3) every other week (cycle 4 to 6) and every 4 weeks from cycle 7. Favorable safety profile [74].
ABBV-383 (TNB-383B)Phase 1 trial (NCT03933735)124
Patients ≥ 75 yr: n.a.
Intravenous injection: 60 mg every three weeks. Good tolerance and durable response [75,76].
REGN-5459Phase 1/2 trial (NCT04083534)43
Patients ≥ 75 yr: n.a.
Target dose: intravenous injection of 480 mg once weekly. Acceptable safety/tolerability [77].
GPRC5DTalquetamabPhase 1/2 MonumenTAL-1 trial. (NCT03399799); (NCT04634552)375
Patients ≥ 75 yr: n.a.
Subcutaneous injection: 0.4 mg/kg once weekly or 0.8 mg/kg every other week, with step-up doses. The safety profile was consistent with previous results [78].
Forimtamig (RG6234)Phase 1 trial (NCT04557150)108
Patients ≥ 75 yr: n.a.
Intravenous injection dose range: 6–10,000 µg (51 pts). Subcutaneous injection dose range: 30–7200 µg (57 pts). High response rate across all tested doses for both IV and SC dosing [79].
FcRH5CevostamabPhase 1 trial (NCT03275103)160
Patients ≥ 75 yr: n.a.
Intravenous infusion in 21-day cycles. In the single-step-up cohorts, the step dose (0.05–3.6 mg) is provided on C1 Day (D) 1, and the target dose (0.15–198 mg) on C1D8. In the double-step-up cohorts, the step doses are provided on C1D1 (0.3–1.2 mg) and C1D8 (3.6 mg) and the target dose (60–160 mg) on C1D15. In both regimens, the target dose is provided on D1 of subsequent cycles. Cevostamab is continued for a total of 17 cycles unless progressive disease or unacceptable toxicity occurs. Clinically meaningful activity and no increase in CRS rate [80].
Abbreviation: n.a.: not available.
These T cell-redirecting therapies are associated with >60% ORR and a median PFS of around 1 year [67]. However, even if first reports are encouraging, the use of BsAbs requires a careful examination of patients > 75 years old, particularly because of potential immune adverse events due to excessive cytokine production. Indeed, Dieterle et al. successfully used teclistamab—a humanized BCMAxCD3 IgG-like BsAb characterized by Fc mutations that stabilize and minimize its immunological effector functions—on octogenarian patients with RRMM, with promising efficacy and reasonable safety [81]. However, the frequent infectious events evidenced in clinical trials negatively impact patient management [82], and preventive measures are required to mitigate this risk, particularly in frail and elderly patients.

6. Treatment of Complications and Adverse Events

Frailty often results in a high incidence of adverse events and treatment discontinuation. Therefore, supportive care, prevention, and early identification of complications are crucial for treatment success. Bone disease should be conservatively managed with calcium and vitamin D supplementation; bisphosphonates; and anti-RANKL antibodies, such as denosumab, but may require radiotherapy or orthopedic surgery [13,24]. Infections are a major cause of death among older patients with MM due to multiple factors, including the disease itself, corticosteroid and treatment-related immunosuppression, and neutropenia. Vaccine prophylaxis against influenza, pneumococcal and varicella-zoster virus infections, and pharmacological prophylaxis should be judiciously added to the ongoing treatment. Anemia, present as a prior comorbidity or MM-related, could also affect quality of life and impact old and frail patient equilibrium. This condition may worsen with the initiation of chemotherapy and should be managed by supporting patients with iron supplementation in case of deficiency, erythropoietin-stimulating agents, and red cell transfusions [26].
Clinical signs and symptoms of pain and neuropathy should be carefully evaluated in MM patients. After the initial assessment, pain might represent a warning sign because of its relationship with disease progression or drug-related adverse events [83]. In particular, pain might present in acute and/or chronic form. Its etiology is often multifactorial and associated with bone disease, fractures, nerve compression, peripheral neuropathy, and infectious [VZV] neuralgias. The management of this disabling condition should be patient and symptom-tailored, considering the patient’s characteristics and comorbidities, such as chronic kidney disease. In 2018, guidelines for the management of pain in cancer patients (https://www.who.int/publications/i/item/9789241550390, accessed on 17 June 2024) were provided by the World Health Organization. Depending on its etiology, pain could be treated pharmacologically with corticosteroids; acetaminophen; neuromodulators, including pregabalin and gabapentin; and opioids. Tricyclic antidepressants, such as serotonin uptake inhibitors, and local anesthetics, including lidocaine and capsaicin, could also be of use [13,83,84]. More specifically, bone-active agents, including bisphosphonates and denosumab, or interventional procedures, such as radiotherapy, vertebroplasty, neurolysis, radiofrequency neurotomy, and spinal cord decompression, should be evaluated [85]. Most importantly, frailty and fitness in patients with MM should be considered dynamic features potentially modified by the initiation of treatment and supportive care [86,87]. Therefore, a flexible approach should be adopted to face treatment modification and to warrant patient-tailored management [14]. Notably, ongoing prospective clinical trials are analyzing the clinical significance of the application of a dynamic frailty assessment to guide therapeutic decisions [88] (https://classic.clinicaltrials.gov/ct2/show/NCT06099912, accessed on 17 June 2024).

7. Conclusions

Our analysis of published studies suggests that, at least in intermediately fit, elderly, newly diagnosed MM patients, treatment intensity during continuous therapy can be de-escalated without a negative impact on outcomes. However, in subsequent phases, the choice of best therapeutic options, enabling the reduction of treatment-related toxicities and preserving a good quality of life, is more difficult since treatment effectiveness may indeed be affected by the presence of co-morbidities, often related to advanced age, and by defective compliance [57]. As a result, few older and frail patients are currently enrolled in pivotal clinical trials, and few data are presently available on appropriate management (Table 3).
Within this context, it is remarkable that in a large majority of clinical studies, analysis of potential patient frailty is limited to age reports, without mentioning the use of available though imperfect evaluation tools.
On the other hand, while emerging targeted treatments, including CAR-T cells and BsAbs, are powerfully expanding the range of available MM therapeutic options, with increasing effectiveness, their application is also accompanied by the occurrence of specific adverse events [88], with potentially greater impacts on frail and older patients. For the safer use of these treatments, standardized frailty assessments are urgently required based not only on clinical scoring but, possibly, also on the characterization of innovative markers and laboratory tests [88]. In their absence, dynamic assessments and patient-tailored treatments should be recommended.

Author Contributions

Conceptualization, A.A. and G.S.; methodology, B.M. and T.S.; software, B.M. and T.S.; validation, A.A., G.S. and G.C.S.; formal analysis, A.A.; investigation, A.A. and G.S.; resources, G.S.; data curation, G.C.S.; writing—original draft preparation, A.A., B.M., T.S. and G.P.; writing—review and editing, A.A., G.S. and G.C.S.; visualization, L.F.; supervision, A.V. and G.C.S.; project administration, G.S.; funding acquisition, G.S. All authors have read and agreed to the published version of the manuscript.

Funding

GS is supported by the Italian Association for Cancer Research (AIRC), Investigator Grant (IG) 2020-24440, and the National Operational Program (PON), Ministry of the University and Research (MUR)-EU ARS01_00906.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kyle, R.A.; Greipp, P.R. Smoldering Multiple Myeloma. N. Engl. J. Med. 1980, 302, 1347–1349. [Google Scholar] [CrossRef] [PubMed]
  2. Landgren, O.; Kyle, R.A.; Pfeiffer, R.M.; Katzmann, J.A.; Caporaso, N.E.; Hayes, R.B.; Dispenzieri, A.; Kumar, S.; Clark, R.J.; Baris, D.; et al. Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: A prospective study. Blood 2009, 113, 5412–5417. [Google Scholar] [CrossRef]
  3. Tricot, G. New insights into role of microenvironment in multiple myeloma. Lancet 2000, 355, 248–250. [Google Scholar] [CrossRef] [PubMed]
  4. Antoine-Pepeljugoski, C.; Braunstein, M.J. Management of Newly Diagnosed Elderly Multiple Myeloma Patients. Curr. Oncol. Rep. 2019, 21, 64. [Google Scholar] [CrossRef] [PubMed]
  5. Bolli, N.; Martinelli, G.; Cerchione, C. The Molecular Pathogenesis of Multiple Myeloma. Hematol. Rep. 2020, 12, 9054. [Google Scholar] [CrossRef]
  6. Kyle, R.A.; Therneau, T.M.; Rajkumar, S.V.; Offord, J.R.; Larson, D.R.; Plevak, M.F.; Melton, L.J. A Long-Term Study of Prognosis in Monoclonal Gammopathy of Undetermined Significance. N. Engl. J. Med. 2002, 346, 564–569. [Google Scholar] [CrossRef]
  7. Kyle, R.A.; Buadi, F.; Vincent Rajkumar, S. Management of Monoclonal Gammopathy of Undetermined Significance (MGUS) and Smoldering Multiple Myeloma (SMM). Oncology 2011, 25, 578. [Google Scholar]
  8. da Vià, M.C.; Ziccheddu, B.; Maeda, A.; Bagnoli, F.; Perrone, G.; Bolli, N. A Journey Through Myeloma Evolution: From the Normal Plasma Cell to Disease Complexity. HemaSphere 2020, 4, e502. [Google Scholar] [CrossRef]
  9. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.-V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  10. Bolli, N.; Maura, F.; Minvielle, S.; Gloznik, D.; Szalat, R.; Fullam, A.; Martincorena, I.; Dawson, K.J.; Samur, M.K.; Zamora, J.; et al. Genomic patterns of progression in smoldering multiple myeloma. Nat. Commun. 2018, 9, 3363. [Google Scholar] [CrossRef]
  11. Rajkumar, S.V.; Kumar, S. Multiple Myeloma: Diagnosis and Treatment. Mayo Clin. Proc. 2016, 91, 101–119. [Google Scholar] [CrossRef]
  12. Rajkumar, S.V. Multiple myeloma: 2022 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2022, 97, 1086–1107. [Google Scholar] [CrossRef]
  13. Kaweme, N.M.; Changwe, G.J.; Zhou, F. Approaches and Challenges in the Management of Multiple Myeloma in the Very Old: Future Treatment Prospects. Front. Med. 2021, 8, 612696. [Google Scholar] [CrossRef] [PubMed]
  14. Dimopoulos, M.A.; Moreau, P.; Terpos, E.; Mateos, M.V.; Zweegman, S.; Cook, G.; Delforge, M.; Hájek, R.; Schjesvold, F.; Cavo, M.; et al. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2021, 32, 309–322. [Google Scholar] [CrossRef] [PubMed]
  15. Mina, R.; Bringhen, S.; Wildes, T.M.; Zweegman, S.; Rosko, A.E. Approach to the Older Adult with Multiple Myeloma. Am. Soc. Clin. Oncol. Educ. B 2019, 39, 500–518. [Google Scholar] [CrossRef] [PubMed]
  16. Facon, T.; Leleu, X.; Manier, S. How I treat multiple myeloma in geriatric patients. Blood 2024, 143, 224–232. [Google Scholar] [CrossRef]
  17. Palumbo, A.; Bringhen, S.; Mateos, M.V.; Larocca, A.; Facon, T.; Kumar, S.K.; Offidani, M.; McCarthy, P.; Evangelista, A.; Lonial, S.; et al. Geriatric assessment predicts survival and toxicities in elderly myeloma patients: An International Myeloma Working Group report. Blood 2015, 125, 2068–2074. [Google Scholar] [CrossRef]
  18. Kleber, M.; Ihorst, G.; Terhorst, M.; Koch, B.; Deschler, B.; Wäsch, R.; Engelhardt, M. Comorbidity as a prognostic variable in multiple myeloma: Comparative evaluation of common comorbidity scores and use of a novel MM—Comorbidity score. Blood Cancer J. 2011, 1, e35. [Google Scholar] [CrossRef] [PubMed]
  19. Engelhardt, M.; Domm, A.S.; Dold, S.M.; Ihorst, G.; Reinhardt, H.; Zober, A.; Hieke, S.; Baayen, C.; Müller, S.J.; Einsele, H.; et al. A concise revised Myeloma Comorbidity Index as a valid prognostic instrument in a large cohort of 801 multiple myeloma patients. Haematologica 2017, 102, 910–921. [Google Scholar] [CrossRef]
  20. Engelhardt, M.; Dold, S.M.; Ihorst, G.; Zober, A.; Möller, M.; Reinhardt, H.; Hieke, S.; Schumacher, M.; Wäsch, R. Geriatric assessment in multiple myeloma patients: Validation of the International Myeloma Working Group (IMWG) score and comparison with other common comorbidity scores. Haematologica 2016, 101, 1110–1119. [Google Scholar] [CrossRef]
  21. Bonanad, S.; De la Rubia, J.; Gironella, M.; Persona, E.P.; González, B.; Lago, C.F.; Arnan, M.; Zudaire, M.; Rivas, J.H.; Soler, A.; et al. Development and psychometric validation of a brief comprehensive health status assessment scale in older patients with hematological malignancies: The GAH Scale. J. Geriatr. Oncol. 2015, 6, 353–361. [Google Scholar] [CrossRef]
  22. Mian, H.S.; Wildes, T.M.; Fiala, M.A. Development of a Medicare Health Outcomes Survey Deficit-Accumulation Frailty Index and Its Application to Older Patients with Newly Diagnosed Multiple Myeloma. JCO Clin. Cancer Inform. 2018, 2, 1–13. [Google Scholar] [CrossRef]
  23. Wildes, T.M.; Tuchman, S.A.; Klepin, H.D.; Mikhael, J.; Trinkaus, K.; Stockerl-Goldstein, K.; Vij, R.; Colditz, G. Geriatric Assessment in Older Adults with Multiple Myeloma. J. Am. Geriatr. Soc. 2019, 67, 987–991. [Google Scholar] [CrossRef]
  24. Rosko, A.E.; Huang, Y.; Benson, D.M.; Efebera, Y.A.; Hofmeister, C.; Jaglowski, S.; Devine, S.; Bhatt, G.; Wildes, T.M.; Dyko, A.; et al. Use of a comprehensive frailty assessment to predict morbidity in patients with multiple myeloma undergoing transplant. J. Geriatr. Oncol. 2019, 10, 479–485. [Google Scholar] [CrossRef] [PubMed]
  25. de Ramon Ortiz, C.; Beauverd, Y.; Samii, K. Assessment and Treatment of Elderly Patients with Multiple Myeloma. Health TIMES Oncol. Hematol. 2024, 20, 86–93. [Google Scholar]
  26. Ludwig, H.; Delforge, M.; Facon, T.; Einsele, H.; Gay, F.; Moreau, P.; Avet-Loiseau, H.; Boccadoro, M.; Hajek, R.; Mohty, M.; et al. Prevention and management of adverse events of novel agents in multiple myeloma: A consensus of the European Myeloma Network. Leukemia 2018, 32, 1542–1560. [Google Scholar] [CrossRef] [PubMed]
  27. Larocca, A.; Bonello, F.; Gaidano, G.; D’agostino, M.; Offidani, M.; Cascavilla, N.; Capra, A.; Benevolo, G.; Tosi, P.; Galli, M.; et al. Dose/schedule-adjusted Rd-R vs continuous Rd for elderly, intermediate-fit patients with newly diagnosed multiple myeloma. Blood 2021, 137, 3027–3036. [Google Scholar] [CrossRef] [PubMed]
  28. Manier, S.; Corre, J.; Hulin, C.; Laribi, K.; Araujo, C.; Pica, G.-M.; Touzeau, C.; Godmer, P.; Slama, B.; Karlin, L.; et al. A Dexamethasone Sparing-Regimen with Daratumumab and Lenalidomide in Frail Patients with Newly-Diagnosed Multiple Myeloma: Efficacy and Safety Analysis of the Phase 3 IFM2017-03 Trial. Blood 2022, 140, 1369–1370. [Google Scholar] [CrossRef]
  29. Larocca, A.; Cani, L.; Bertuglia, G.; Bruno, B.; Bringhen, S. New Strategies for the Treatment of Older Myeloma Patients. Cancers 2023, 15, 2693. [Google Scholar] [CrossRef]
  30. Durie, B.G.M.; Hoering, A.; Abidi, M.H.; Rajkumar, S.V.; Epstein, J.; Kahanic, S.P.; Thakuri, M.; Reu, F.; Reynolds, C.M.; Sexton, R.; et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): A randomised, open-label, phase 3 trial. Lancet 2017, 389, 519–527. [Google Scholar] [CrossRef]
  31. Durie, B.G.M.; Hoering, A.; Sexton, R.; Abidi, M.H.; Epstein, J.; Rajkumar, S.V.; Dispenzieri, A.; Kahanic, S.P.; Thakuri, M.C.; Reu, F.J.; et al. Longer term follow-up of the randomized phase III trial SWOG S0777: Bortezomib, lenalidomide and dexamethasone vs. lenalidomide and dexamethasone in patients (Pts) with previously untreated multiple myeloma without an intent for immediate autologous stem cell transplant (ASCT). Blood Cancer J. 2020, 10, 53. [Google Scholar]
  32. O’Donnell, E.K.; Laubach, J.P.; Yee, A.J.; Chen, T.; Huff, C.A.; Basile, F.G.; Raje, N.S. A phase 2 study of modified lenalidomide, bortezomib and dexamethasone in transplant-ineligible multiple myeloma. Br. J. Haematol. 2018, 182, 222–230. [Google Scholar] [CrossRef]
  33. O’Donnell, E.K.; Laubach, J.P.; Yee, A.J.; Redd, R.; Huff, C.A.; Basile, F.; Wade, P.M.; Paba-Prada, C.E.; Ghobrial, I.M.; Schlossman, R.L.; et al. Updated Results of a Phase 2 Study of Modified Lenalidomide, Bortezomib, and Dexamethasone (RVd-lite) in Transplant-Ineligible Multiple Myeloma. Blood 2019, 134, 3178. [Google Scholar] [CrossRef]
  34. Hulin, C.; Facon, T.; Rodon, P.; Pegourie, B.; Benboubker, L.; Doyen, C.; Moreau, P. 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, 27, 3664–3670. [Google Scholar] [CrossRef]
  35. Facon, T.; Kumar, S.K.; Plesner, T.; Orlowski, R.Z.; Moreau, P.; Bahlis, N.; Basu, S.; Nahi, H.; Hulin, C.; Quach, H.; et al. Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): Overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 1582–1596. [Google Scholar] [CrossRef] [PubMed]
  36. Facon, T.; Cook, G.; Usmani, S.Z.; Hulin, C.; Kumar, S.; Plesner, T.; Touzeau, C.; Bahlis, N.J.; Basu, S.; Nahi, H.; et al. Daratumumab plus lenalidomide and dexamethasone in transplant-ineligible newly diagnosed multiple myeloma: Frailty subgroup analysis of MAIA. Leukemia 2022, 36, 1066–1077. [Google Scholar] [CrossRef] [PubMed]
  37. Durie, B.G.M.; Kumar, S.K.; Usmani, S.Z.; Nonyane, B.A.S.; Ammann, E.M.; Lam, A.; Kobos, R.; Maiese, E.M.; Facon, T. Daratumumab-lenalidomide-dexamethasone vs standard-of-care regimens: Efficacy in transplant-ineligible untreated myeloma. Am. J. Hematol. 2020, 95, 1486–1494. [Google Scholar] [CrossRef]
  38. Mateos, M.V.; Nahi, H.; Legiec, W.; Grosicki, S.; Vorobyev, V.; Spicka, I.; Hungria, V.; Korenkova, S.; Bahlis, N.; Flogegard, M.; et al. Subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma (COLUMBA): A multicentre, open-label, non-inferiority, randomised, phase 3 trial. Lancet Haematol. 2020, 7, e370–e380. [Google Scholar] [CrossRef]
  39. Anderson, K.C.; Landgren, O.; Arend, R.C.; Chou, J.; Jacobs, I.A. Humanistic and economic impact of subcutaneous versus intravenous administration of oncology biologics. Future Oncol. 2019, 15, 3267–3281. [Google Scholar] [CrossRef]
  40. Fonseca, R.; Facon, T.; Hashim, M.; Nair, S.; He, J.; Ammann, E.; Lam, A.; Wildgust, M.; Kumar, S. Impact of Treatment Sequencing on Overall Survival in Patients with Transplant-Ineligible Newly Diagnosed Myeloma. Oncologist 2023, 28, e263–e269. [Google Scholar] [CrossRef]
  41. Mateos, M.-V.; Dimopoulos, M.A.; Cavo, M.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lúcio, P.; Nagy, Z.; Kaplan, P.; et al. Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma. N. Engl. J. Med. 2018, 378, 518–528. [Google Scholar] [CrossRef] [PubMed]
  42. Mateos, M.V.; Dimopoulos, M.A.; Cavo, M.; Suzuki, K.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; Pour, L.; Grosicki, S.; et al. Daratumumab Plus Bortezomib, Melphalan, and Prednisone Versus Bortezomib, Melphalan, and Prednisone in Transplant-Ineligible Newly Diagnosed Multiple Myeloma: Frailty Subgroup Analysis of ALCYONE. Clin. Lymphoma Myeloma Leuk. 2021, 21, 785–798. [Google Scholar] [CrossRef]
  43. San-Miguel, J.; Avet-Loiseau, H.; Paiva, B.; Kumar, S.K.; Dimopoulos, M.A.; Facon, T.; Mateos, M.-V.; Touzeau, C.; Jakubowiak, A.J.; Usmani, S.Z.; et al. Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE. Blood 2022, 139, 492–501. [Google Scholar] [CrossRef] [PubMed]
  44. Facon, T.; Dimopoulos, M.-A.; Leleu, X.P.; Beksac, M.; Pour, L.; Hájek, R.; Liu, Z.; Minarik, J.; Moreau, P.; Romejko-Jarosinska, J.; et al. Isatuximab, Bortezomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2024, 391, 1597–1609. [Google Scholar] [CrossRef]
  45. Mateos, M.V.; Cavo, M.; Blade, J.; Dimopoulos, M.A.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; et al. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): A randomised, open-label, phase 3 trial. Lancet 2020, 395, 132–141. [Google Scholar] [CrossRef]
  46. Barlogie, B.; Smith, L.; Alexanian, R. Effective Treatment of Advanced Multiple Myeloma Refractory to Alkylating Agents. N. Engl. J. Med. 1984, 310, 1353–1356. [Google Scholar] [CrossRef] [PubMed]
  47. Rajkumar, S.V.; Blood, E.; Vesole, D.; Fonseca, R.; Greipp, P.R. 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. 2006, 24, 431–436. [Google Scholar] [CrossRef]
  48. Facon, T.; Mary, J.Y.; Pégourie, B.; Attal, M.; Renaud, M.; Sadoun, A.; Voillat, L.; Dorvaux, V.; Hulin, C.; Lepeu, G.; et al. Dexamethasone-based regimens versus melphalan-prednisone for elderly multiple myeloma patients ineligible for high-dose therapy. Blood 2006, 107, 1292–1298. [Google Scholar] [CrossRef]
  49. Bringhen, S.; Milan, A.; Ferri, C.; Wäsch, R.; Gay, F.; LaRocca, A.; Salvini, M.; Terpos, E.; Goldschmidt, H.; Cavo, M.; et al. Cardiovascular adverse events in modern myeloma therapy—Incidence and risks. A review from the European Myeloma Network (EMN) and Italian Society of Arterial Hypertension (SIIA). Haematologica 2018, 103, 1422–1432. [Google Scholar] [CrossRef]
  50. Stege, C.A.M.; Nasserinejad, K.; van der Spek, E.; Bilgin, Y.M.; Kentos, A.; Sohne, M.; van Kampen, R.J.W.; Ludwig, I.; Thielen, N.; Durdu-Rayman, N.; et al. Ixazomib, Daratumumab, and Low-Dose Dexamethasone in Frail Patients with Newly Diagnosed Multiple Myeloma: The Hovon 143 Study. J. Clin. Oncol. 2021, 39, 2758–2767. [Google Scholar] [CrossRef]
  51. Chim, C.S.; Kumar, S.K.; Orlowski, R.Z.; Cook, G.; Richardson, P.G.; Gertz, M.A.; Giralt, S.; Mateos, M.V.; Leleu, X.; Anderson, K.C. Management of relapsed and refractory multiple myeloma: Novel agents, antibodies, immunotherapies and beyond. Leukemia 2018, 32, 252–262. [Google Scholar] [CrossRef] [PubMed]
  52. Sonneveld, P. Management of multiple myeloma in the relapsed/refractory patient. Hematology 2017, 2017, 508–517. [Google Scholar] [CrossRef]
  53. Quach, H.; Nooka, A.; Samoylova, O.; Venner, C.P.; Kim, K.; Facon, T.; Spencer, A.; Usmani, S.Z.; Grosicki, S.; Suzuki, K.; et al. Carfilzomib, dexamethasone and daratumumab in relapsed or refractory multiple myeloma: Results of the phase III study CANDOR by prior lines of therapy. Br. J. Haematol. 2021, 194, 784–788. [Google Scholar] [CrossRef] [PubMed]
  54. Yong, K.; Delforge, M.; Driessen, C.; Fink, L.; Flinois, A.; Gonzalez-McQuire, S.; Safaei, R.; Karlin, L.; Mateos, M.; Raab, M.S.; et al. Multiple myeloma: Patient outcomes in real-world practice. Br. J. Haematol. 2016, 175, 252–264. [Google Scholar] [CrossRef]
  55. Rodríguez-Otero, P.; San Miguel, J.F. Post-CAR-T Cell Therapy (Consolidation and Relapse): Multiple Myeloma. In The EBMT/EHA CAR-T Cell Handbook; Springer: Cham, Switzerland, 2022; pp. 173–176. [Google Scholar]
  56. Davis, J.A.; Dima, D.; Ahmed, N.; DeJarnette, S.; McGuirk, J.; Jia, X.; Raza, S.; Khouri, J.; Valent, J.; Anwer, F.; et al. Impact of Frailty on Outcomes after Chimeric Antigen Receptor T Cell Therapy for Patients with Relapsed/Refractory Multiple Myeloma. Transplant. Cell Ther. 2024, 30, 298–305. [Google Scholar] [CrossRef]
  57. Rajeeve, S.; Usmani, S.Z. How Old is Too Old for CAR-T Cell Therapies in Multiple Myeloma? Transplant. Cell Ther. 2023, 29, 343–344. [Google Scholar] [CrossRef] [PubMed]
  58. Munshi, N.C.; Anderson, L.D.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef]
  59. Berdeja, J.G.; Madduri, D.; Usmani, S.Z.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Stewart, A.K.; Hari, P.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet 2021, 398, 314–324. [Google Scholar] [CrossRef]
  60. Akhtar, O.S.; Sheeba, B.A.; Azad, F.; Alessi, L.; Hansen, D.; Alsina, M.; Baz, R.; Shain, K.; Cruz, A.G.; Puglianini, O.C.; et al. Safety and efficacy of anti-BCMA CAR-T cell therapy in older adults with multiple myeloma: A systematic review and meta-analysis. J. Geriatr. Oncol. 2024, 15, 101628. [Google Scholar] [CrossRef]
  61. Reyes, K.R.; Huang, C.Y.; Lo, M.; Arora, S.; Chung, A.; Wong, S.W.; Wolf, J.; Olin, R.L.; Martin, T.; Shah, N.; et al. Safety and Efficacy of BCMA CAR-T Cell Therapy in Older Patients with Multiple Myeloma. Transplant. Cell Ther. 2023, 29, 350–355. [Google Scholar] [CrossRef]
  62. Neelapu, S.S.; Jacobson, C.A.; Oluwole, O.O.; Munoz, J.; Deol, A.; Miklos, D.B.; Bartlett, N.L.; Braunschweig, I.; Jiang, Y.; Kim, J.J.; et al. Outcomes of older patients in ZUMA-1, a pivotal study of axicabtagene ciloleucel in refractory large B-cell lymphoma. Blood 2020, 135, 2106–2109. [Google Scholar] [CrossRef] [PubMed]
  63. Yates, S.J.; Cursio, J.F.; Artz, A.; Kordas, K.; Bishop, M.R.; Derman, B.A.; Kosuri, S.; Riedell, P.A.; Kline, J.; Jakubowiak, A.; et al. Optimization of older adults by a geriatric assessment–guided multidisciplinary clinic before CAR T-cell therapy. Blood Adv. 2024, 8, 3785–3797. [Google Scholar] [CrossRef]
  64. Zhou, X.; Einsele, H.; Danhof, S. Bispecific Antibodies: A New Era of Treatment for Multiple Myeloma. J. Clin. Med. 2020, 9, 2166. [Google Scholar] [CrossRef] [PubMed]
  65. Walter, R.B. Biting back: BiTE antibodies as a promising therapy for acute myeloid leukemia. Expert Rev. Hematol. 2014, 7, 317–319. [Google Scholar] [CrossRef]
  66. Ellerman, D. Bispecific T-cell engagers: Towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2019, 154, 102–117. [Google Scholar] [CrossRef] [PubMed]
  67. Tacchetti, P.; Barbato, S.; Mancuso, K.; Zamagni, E.; Cavo, M. Bispecific Antibodies for the Management of Relapsed/Refractory Multiple Myeloma. Cancers 2024, 16, 2337. [Google Scholar] [CrossRef]
  68. Moreau, P.; Garfall, A.L.; van de Donk, N.W.C.J.; Nahi, H.; San-Miguel, J.F.; Oriol, A.; Nooka, A.K.; Martin, T.; Rosinol, L.; Chari, A.; et al. Teclistamab in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2022, 387, 495–505. [Google Scholar] [CrossRef]
  69. Usmani, S.Z.; Garfall, A.L.; van de Donk, N.W.C.J.; Nahi, H.; San-Miguel, J.F.; Oriol, A.; Rosinol, L.; Chari, A.; Bhutani, M.; Karlin, L.; et al. Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): A multicentre, open-label, single-arm, phase 1 study. Lancet 2021, 398, 665–674. [Google Scholar] [CrossRef]
  70. Raje, N.S.; Leleu, X.P.; Lesokhin, A.M.; Mohty, M.; Nooka, A.K.; Leip, E.; Conte, U.; Viqueira, A.; Manier, S. Efficacy and safety of elranatamab by age and frailty in patients (pts) with relapsed/refractory multiple (RRMM): A subgroup analysis from MagnetisMM-3. J. Clin. Oncol. 2023, 41, 8040. [Google Scholar] [CrossRef]
  71. Lesokhin, A.M.; Tomasson, M.H.; Arnulf, B.; Bahlis, N.J.; Prince, H.M.; Niesvizky, R.; Rodrίguez-Otero, P.; Martinez-Lopez, J.; Koehne, G.; Touzeau, C.; et al. Elranatamab in relapsed or refractory multiple myeloma: Phase 2 MagnetisMM-3 trial results. Nat. Med. 2023, 29, 2259–2267. [Google Scholar] [CrossRef]
  72. Lee, H.C.; Bumma, N.; Richter, J.R.; Dhodapkar, M.V.; Hoffman, J.E.; Suvannasankha, A.; Zonder, J.A.; Shah, M.R.; Lentzsch, S.; Maly, J.J.; et al. LINKER-MM1 study: Linvoseltamab (REGN5458) in patients with relapsed/refractory multiple myeloma. J. Clin. Oncol. 2023, 41, 8006. [Google Scholar] [CrossRef]
  73. Jagannath, S.; Richter, J.; Dhodapkar, M.V.; Hoffman, J.E.; Lee, H.C.; Suvannasankha, A.; Shah, M.R.; Lentzsch, S.; Zonder, J.A.; Baz, R.; et al. Patterns of Response to 200 Mg Linvoseltamab in Patients with Relapsed/Refractory Multiple Myeloma: Longer Follow-Up of the Linker-MM1 Study. Blood 2023, 142, 4746. [Google Scholar] [CrossRef]
  74. Bar, N.; Mateos, M.V.; Ribas, P.; Hansson, M.; Paris, L.; Hofmeister, C.C.; Otero, P.R.; Bermúdez, M.A.; Martin, T.; Santoro, A.; et al. Alnuctamab (ALNUC.; BMS-986349; CC-93269), a 2+1 B-Cell Maturation Antigen (BCMA) × CD3 T-Cell Engager (TCE), Administered Subcutaneously (SC) in Patients (Pts) with Relapsed/Refractory Multiple Myeloma (RRMM): Updated Results from a Phase 1 First-in-Human Clinical Study. Blood 2023, 142, 2011. [Google Scholar]
  75. D’souza, A.; Shah, N.; Rodriguez, C.; Voorhees, P.M.; Weisel, K.; Bueno, O.F.; Kumar, S. A Phase I First-in-Human Study of ABBV-383, a B-Cell Maturation Antigen × CD3 Bispecific T-Cell Redirecting Antibody, in Patients with Relapsed/Refractory Multiple Myeloma. J. Clin. Oncol. 2022, 40, 3576–3586. [Google Scholar] [CrossRef]
  76. Voorhees, P.M.; D’Souza, A.; Weisel, K.; Hurd, D.D.; Teipel, R.; Chung, A.; Rodriguez, C.; Tuchman, S.A.; Korde, N.; Safah, H.; et al. A Phase 1 First-in-Human Study of Abbv-383, a BCMA × CD3 Bispecific T-Cell-Redirecting Antibody, As Monotherapy in Patients with Relapsed/Refractory Multiple Myeloma. Blood 2022, 140, 4401–4404. [Google Scholar] [CrossRef]
  77. Suvannasankha, A.; Kapoor, P.; Pianko, M.J.; Richter, J.; D’Souza, A.; Anderson, L.D.; Lee, H.C. Abstract CT013: Safety and efficacy from the phase 1/2 first-in-human study of REGN5459, a BCMA×CD3 bispecific antibody with low CD3 affinity, in patients with relapsed/refractory multiple myeloma. Cancer Res. 2023, 83, CT013. [Google Scholar] [CrossRef]
  78. Rasche, L.; Schinke, C.; Touzeau, C.; Minnema, M.C.; van de Donk, N.W.; Rodríguez-Otero, P.; Chari, A. Long-Term Efficacy and Safety Results From the Phase 1/2 MonumenTAL-1 Study of Talquetamab, a GPRC5D×CD3 Bispecific Antibody, in Patients with Relapsed/Refractory Multiple Myeloma (RRMM). Clin. Lymphoma Myeloma Leuk. 2024, 24, S240. [Google Scholar] [CrossRef]
  79. Carlo-Stella, C.; Mazza, R.; Manier, S.; Facon, T.; Yoon, S.-S.; Koh, Y.; Harrison, S.J.; Er, J.; Pinto, A.; Volzone, F.; et al. RG6234, a GPRC5DxCD3 T-Cell Engaging Bispecific Antibody, Is Highly Active in Patients (pts) with Relapsed/Refractory Multiple Myeloma (RRMM): Updated Intravenous (IV) and First Subcutaneous (SC) Results from a Phase I Dose-Escalation Study. Blood 2022, 140, 397–399. [Google Scholar] [CrossRef]
  80. Trudel, S.; Cohen, A.D.; Krishnan, A.Y.; Fonseca, R.; Spencer, A.; Berdeja, J.G.; Lesokhin, A.; Forsberg, P.A.; Laubach, J.P.; Costa, L.J.; et al. Cevostamab Monotherapy Continues to Show Clinically Meaningful Activity and Manageable Safety in Patients with Heavily Pre-Treated Relapsed/Refractory Multiple Myeloma (RRMM): Updated Results from an Ongoing Phase I Study. Blood 2021, 138, 157. [Google Scholar] [CrossRef]
  81. Dieterle, M.P.; Mostufi-Zadeh-Haghighi, G.; Kus, J.W.; Wippel, C.; Brugger, Z.; Miething, C.; Wäsch, R.; Engelhardt, M. Safe and successful teclistamab treatment in very elderly multiple myeloma (MM) patients: A case report and experience from a total of three octogenarians. Ann. Hematol. 2023, 102, 3639–3641. [Google Scholar] [CrossRef]
  82. Jourdes, A.; Cellerin, E.; Touzeau, C.; Harel, S.; Denis, B.; Escure, G.; Faure, E.; Jamard, S.; Danion, F.; Sonntag, C.; et al. Characteristics and incidence of infections in patients with multiple myeloma treated by bispecific antibodies: A national retrospective study. Clin. Microbiol. Infect. 2024, 30, 764–771. [Google Scholar] [CrossRef] [PubMed]
  83. Terpos, E.; Mikhael, J.; Hajek, R.; Chari, A.; Zweegman, S.; Lee, H.C.; Mateos, M.-V.; Larocca, A.; Ramasamy, K.; Kaiser, M.; et al. Management of patients with multiple myeloma beyond the clinical-trial setting: Understanding the balance between efficacy, safety and tolerability, and quality of life. Blood Cancer J. 2021, 11, 40. [Google Scholar] [CrossRef]
  84. Mohty, B.; El-Cheikh, J.; Yakoub-Agha, I.; Moreau, P.; Harousseau, J.L.; Mohty, M. Peripheral neuropathy and new treatments for multiple myeloma: Background and practical recommendations. Haematologica 2010, 95, 311–319. [Google Scholar] [CrossRef] [PubMed]
  85. Du, J.S.; Yen, C.H.; Hsu, C.M.; Hsiao, H.H. Management of Myeloma Bone Lesions. Int. J. Mol. Sci. 2021, 22, 3389. [Google Scholar] [CrossRef] [PubMed]
  86. Mach, S.; Javed, S.; Chen, G.H.; Huh, B.K. Peripheral Nerve Stimulation for Back Pain in Patients with Multiple Myeloma as Bridge Therapy to Radiation Treatment: A Case Series. Neuromodulation 2023, 26, 694–699. [Google Scholar] [CrossRef]
  87. Mian, H.S.; Wildes, T.; Vij, R.; Major, A.; Fiala, M.A. Need for Dynamic Frailty Risk Assessment Among Older Adults with Multiple Myeloma: A Population-Based Cohort Study. Blood 2022, 140, 423–424. [Google Scholar] [CrossRef]
  88. Cook, G.; Pawlyn, C.; Royle, K.-L.; Senior, E.R.; Everritt, D.; Bird, J.; Bowcock, S.J.; Dawkins, B.; Drayson, M.; Gillson, S.; et al. Dynamic Frailty Assessment in Transplant Non-Eligible Newly Diagnosed Myeloma Patients: Initial Data from UK Myeloma Research Alliance (UK-MRA) Myeloma XIV (FiTNEss): A Frailty-Adjusted Therapy Study. Blood 2023, 142, 4748. [Google Scholar] [CrossRef]
  89. Kumar, S.K.; Jacobus, S.J.; Cohen, A.D.; Weiss, M.; Callander, N.; Singh, A.K.; Parker, T.L.; Menter, A.; Yang, X.; Parsons, B.; et al. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020, 21, 1317–1330. [Google Scholar]
  90. Facon, T.; Lee, J.H.; Moreau, P.; Niesvizky, R.; Dimopoulos, M.; Hajek, R.; Pour, L.; Jurczyszyn, A.; Qiu, L.; Klippel, Z.; et al. Carfilzomib or bortezomib with melphalan-prednisone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood 2019, 133, 1953–1963. [Google Scholar] [CrossRef]
  91. Usmani, S.Z.; Nahi, H.; Legiec, W.; Grosicki, S.; Vorobyev, V.; Spicka, I.; Hungria, V.; Korenkova, S.; Bahlis, N.J.; Flogegard, M.; et al. Final analysis of the phase III non-inferiority COLUMBA study of subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma. Haematologica 2022, 107, 2408–2417. [Google Scholar] [CrossRef]
  92. Richardson, P.G.; Kumar, S.K.; Masszi, T.; Grzasko, N.; Bahlis, N.J.; Hansson, M.; Pour, L.; Sandhu, I.; Ganly, P.; Baker, B.W.; et al. Final Overall Survival Analysis of the TOURMALINE-MM1 Phase III Trial of Ixazomib, Lenalidomide, and Dexamethasone in Patients with Relapsed or Refractory Multiple Myeloma. J. Clin. Oncol. 2021, 39, 2430–2442. [Google Scholar] [CrossRef] [PubMed]
  93. Facon, T.; Niesvizky, R.; Mateos, M.V.; Siegel, D.; Rosenbaum, C.; Bringhen, S.; Weisel, K.; Ho, P.J.; Ludwig, H.; Kumar, S.; et al. Efficacy and safety of carfilzomib-based regimens in frail patients with relapsed and/or refractory multiple myeloma. Blood Adv. 2020, 4, 5449–5459. [Google Scholar] [CrossRef] [PubMed]
  94. Berdeja, J.G.; Raje, N.S.; Siegel, D.S.; Lin, Y.; Anderson, L.D.; Rodriguez-Otero, P.; Manier, S.; Einsele, H.; Cavo, M.; Truppel-Hartmann, A.; et al. Efficacy and Safety of Idecabtagene Vicleucel (ide-cel, bb2121) in Elderly Patients with Relapsed and Refractory Multiple Myeloma: KarMMa Subgroup Analysis. Blood 2020, 136, 16–17. [Google Scholar] [CrossRef]
Table 1. Tailoring treatments for MM patients with different IMWG frailty levels and co-morbidities (1–18).
Table 1. Tailoring treatments for MM patients with different IMWG frailty levels and co-morbidities (1–18).
IMWG-FI FitIMWG-FI Intermediate FitIMWG-FI FrailRenal InsufficiencyPeripheral NeuropathyCardiovascular Disease
First-Line TherapyDRd,
IsaVRd,
or
D-VMP
DRd (reduced),
D-VMP (reduced),
or
VRd (reduced)
Rd (reduced) or try
Dara-based regimens (reduced)
Prefer Bortezomib-based regimensReduce or avoid Bortezomib
-
Prefer
Lena-based regimens
Prefer Dara-based therapy
-
Avoid Carfilzomib
-
Reduce Dexa
R/R MM TherapyKRd, IxaRd
(if Lena naïve)
Or DaraPd, PVd, SVd,
or
CAR-T, BsAbs
DaraPd (consider reduction),
PVd (consider reduction), or
SVd
Low-dose therapies:
DRd (reduced if Lena-naïve); DaraPd (reduced)
-
Supportive care
Prefer Dara-based or regimens (without Lena)IxaRd
-
Avoid Carfilzomib
Avoid Carfilzomib
-
Reduce or discontinue Dexa
LenalidomideStandard
(25 mg/day)
Dose reduction to 15 mg/dayDose reduction to 10 mg/dayCrCl 30–50 mL/min: 10 mg/day
CrCl < 30 mL/min (no dialysis): 15 mg/2 days
CrCl < 30 mL/min (yes dialysis): 5 mg/day post-dialysis
No modification
needed
No modification
-
Thrombo-prophylaxis
PomalidomideStandard (4 mg as per therapy schedule)Dose reduction to 3 mg if toxicityDose reduction to 3 mg or 2 mgNo modificationNo modificationNo modification
-
Thrombo-prophylaxis
Dexamethasone40 mg/weekDose reduction to 20 mg/weekDose reduction to 10 mg/week or
early discontinuation
Dose reduction
or discontinuation (if concomitant CHF or DM)
No modification
needed
Reduction to 20–10 mg/week or early discontinuation
(if CHF, arrhythmia, HTN, or ischemia)
BortezomibTwice weekly (1.3 mg/m2)Weekly (1.3 mg/m2)
or
reduce to weekly 1.0 mg/m2 if neuropathy
Weekly (1.3 mg/m2)
or
Reduce to weekly 1.0 mg/m2 or less if neuropathy
Preferred over Lena
-
No reduction needed
Reduce to 1.0 mg/m2 or less; avoid if severe neuropathyAvoid in significant cardiopathy and monitor closely if pulmonary hypertension
DaratumumabStandardStandardStandardNo modification No modificationNo modification
CarfilzomibStandardDose reduction to 36 mg/m2, weekly administrationAvoid if possibleAvoid if CrCl
<30 mL/min
Avoid if severe neuropathyAvoid in pre-existing CHF and severe HTN
Abbreviations: IMWG-FI: International Myeloma Working Group Frailty Index; DRd: daratumumab + lenalidomide + dexamethasone; IsaVRd: isatuximab + bortezomib + lenalidomide + dexamethasone; D-VMP: daratumumab + bortezomib + melphalan + prednisone; Rd: lenalidomide + dexamethasone; Dara: daratumumab; Lena: lenalidomide; Dexa: dexamethasone; R/R MM: relapsed/refractory multiple myeloma; KRd: carfilzomib + lenalidomide + dexamethasone; IxaRd: ixazomib + lenalidomide + dexamethasone.
Table 3. Approved immunotherapy options for older, frail patients ineligible for transplantation at initial diagnosis and at relapse.
Table 3. Approved immunotherapy options for older, frail patients ineligible for transplantation at initial diagnosis and at relapse.
Newly Diagnosed Patients
MAIA study [35]n = 737 pts (D-Rd, n = 368; Rd, n = 369)
396 non-frail pts (D-Rd, 196; Rd, 200)
341 frail pts (D-Rd, 172; Rd, 169)
Clinical benefit irrespective of frailty in newly diagnosed, transplant-ineligible patients
ALCYONE [45]n = 706 pts (D-VMP, n = 350; VMP, n = 356)
391 non-frail pts (D-VMP, 187; VMP, 204)
315 frail pts (D-VMP, 163; VMP,152)
Clinical benefit of D-VMP irrespective of frailty in newly diagnosed transplant-ineligible patients enrolled in ALCYONE, regardless of frailty status.
SWOG S0777 [30,31]n = 460 pts (VRd, n = 235; Rd, n = 225)
91/235 pts in the VRd arm aged ≥65 years
Addition of bortezomib to standard lenalidomide–dexamethasone clinically advantageous irrespective of age in previously untreated patients.
ENDURANCE [89]n = 1087 pts (VRd, n = 542; KRd, n = 545)
VRd lite in older pts [32,33]
Addition of Carfilzomib to VRd in newly diagnosed multiple myeloma patients is not more effective and characterized by higher toxicity.
A modified VRd treatment effective in >65 years old, newly diagnosed, transplant-ineligible patients
CLARION [90]n = 955 pts (KMP, n = 478; VMP, n = 477).
KMP is not more effective than VMP in newly diagnosed patients with multiple myeloma ineligible for transplant, irrespective of age.
HOVON 143 [50]n = 65 frail newly diagnosed multiple myeloma pts, treated with Ixa-Dara-Dex.
High response rate but toxicity and early mortality.
Treatment Options at Relapse
COLUMBA [91]n = 522 pts (DARA SC, n = 263; DARA IV, n = 259)
Similar effectiveness to daratumumab upon subcutaneous or intravenous administration in relapsed or refractory multiple myeloma.
TOURMALINE [92]n = 722 (IRd, n = 360; Rd, n = 362)
Improved PFS upon treatment with IRd than with Rd in patients with relapsed or refractory multiple myeloma.
ASPIRE-ENDEAVOR-ARROW [93]ASPIRE n = 792 pts (KRd27, n = 396 vs. Rd n = 396)
ENDEAVOR n = 929 pts (Kd56, n = 464 vs. Vd, n = 465)
ARROW (once-weekly) n = 478 pts (Kd70, n = 240 vs. Kd27, n = 238)
Efficacy and safety were consistent across frailty subgroups with KRd27, Kd56, and weekly Kd70 in relapsed and/or refractory MM.
KarMMa study [94]CAR-T Therapy (n = 128; n = 45 ≥65 years; n = 20 ≥70 years)
Durable responses and manageable safety profile in patients with relapsed/recurrent multiple myeloma aged ≥65 years and ≥70 years.
Abbreviations: pts = patients; D-Rd = daratumumab plus lenalidomide and dexamethasone; Rd = lenalidomide and dexamethasone; D-VMP = daratumumab, plus bortezomib, melphalan, and prednisone; VMP = bortezomib, melphalan, and prednisone; VRd = bortezomib + lenalidomide + dexamethasone; Rd = lenalidomide–dexamethasone; KRd = carfilzomib, lenalidomide, and dexamethasone; KMP = carfilzomib–melphalan–prednisone; Ixa-Dara-Dex = ixazomib–daratumumab–low-dose dexamethasone; DARA = daratumumab; SC = subcutaneous; IV = intravenous; IRd = ixazomib–lenalidomide–dexamethasone; KRd27 = (carfilzomib [27 mg/m2]–lenalidomide–dexamethasone; Kd56 = carfilzomib [56 mg/m2–dexamethasone; Vd = bortezomib–dexamethasone; Kd70 = carfilzomib [70 mg/m2]–dexamethasone.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Aureli, A.; Marziani, B.; Sconocchia, T.; Pasqualone, G.; Franceschini, L.; Spagnoli, G.C.; Venditti, A.; Sconocchia, G. Challenges in Multiple Myeloma Therapy in Older and Frail Patients. Cancers 2025, 17, 944. https://doi.org/10.3390/cancers17060944

AMA Style

Aureli A, Marziani B, Sconocchia T, Pasqualone G, Franceschini L, Spagnoli GC, Venditti A, Sconocchia G. Challenges in Multiple Myeloma Therapy in Older and Frail Patients. Cancers. 2025; 17(6):944. https://doi.org/10.3390/cancers17060944

Chicago/Turabian Style

Aureli, Anna, Beatrice Marziani, Tommaso Sconocchia, Gianmario Pasqualone, Luca Franceschini, Giulio Cesare Spagnoli, Adriano Venditti, and Giuseppe Sconocchia. 2025. "Challenges in Multiple Myeloma Therapy in Older and Frail Patients" Cancers 17, no. 6: 944. https://doi.org/10.3390/cancers17060944

APA Style

Aureli, A., Marziani, B., Sconocchia, T., Pasqualone, G., Franceschini, L., Spagnoli, G. C., Venditti, A., & Sconocchia, G. (2025). Challenges in Multiple Myeloma Therapy in Older and Frail Patients. Cancers, 17(6), 944. https://doi.org/10.3390/cancers17060944

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop