Next Article in Journal
In Vitro Percutaneous Absorption of Permeation-Enhancing Estrogen Formulations
Previous Article in Journal
Enhanced Topical Delivery of Methotrexate via Transferosome-Loaded Microneedle Array Patch: Formulation, Optimization, and In Vitro–In Vivo Assessment
Previous Article in Special Issue
Minimal Residual Disease Significance in Multiple Myeloma Patients Treated with Anti-CD38 Monoclonal Antibodies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study

by
Nicola Sgherza
1,
Olga Battisti
1,
Paola Curci
1,
Concetta Conticello
2,
Salvatore Palmieri
3,
Daniele Derudas
4,
Candida Germano
5,
Enrica Antonia Martino
6,
Giuseppe Mele
7,
Roberta Della Pepa
8,
Francesca Fazio
9,
Anna Mele
10,
Bernardo Rossini
11,
Giulia Palazzo
12,
Daniela Roccotelli
13,
Simona Rasola
14,
Maria Teresa Petrucci
9,
Domenico Pastore
7,
Giuseppe Tarantini
5,
Fabrizio Pane
8,
Massimo Gentile
6,15,
Francesco Di Raimondo
2,16,
Emanuela Resta
17 and
Pellegrino Musto
1,14,*
add Show full author list remove Hide full author list
1
Hematology and Stem Cell Transplantation Unit, AOUC Policlinico, 70124 Bari, Italy
2
Division of Hematology and Stem Cell Transplantation, Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
3
Hematology Unit, Ospedale Cardarelli, 80131 Napoli, Italy
4
Department of Hematology, Businco Hospital, 09121 Cagliari, Italy
5
Hematology Unit, “Dimiccoli” Hospital, 70051 Barletta, Italy
6
Department of Onco-Hematology, Hematology Unit, Azienda Ospedaliera Annunziata, 87100 Cosenza, Italy
7
Hematology Unit, “Perrino” Hospital, 72100 Brindisi, Italy
8
Hematology—Department of Clinical Medicine and Surgery, University Hospital “Federico II”, 80131 Napoli, Italy
9
Department of Translational and Precision Medicine Hematology, Sapienza University, 00185 Roma, Italy
10
Hematology Unit, “Cardinale Panico” Hospital, 70039 Tricase, Italy
11
Hematology and Cell Therapy Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy
12
Haematology Unit, Ospedale G. Moscati, 74010 Taranto, Italy
13
Department of Hematology and Bone Marrow Transplant, IRCSS “Casa Sollievo Della Sofferenza”, 71013 Foggia, Italy
14
Department of Precision and Regenerative Medicine and Ionian Area, “Aldo Moro” University School of Medicine, 70121 Bari, Italy
15
Department of Pharmacy, Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
16
Department of General Surgery and Medical-Surgical Specialties, Hematology Section, University of Catania, 95124 Catania, Italy
17
Department of Clinical and Molecular Medicine, Sapienza University, 00185 Roma, Italy
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(4), 595; https://doi.org/10.3390/ph18040595
Submission received: 26 February 2025 / Revised: 10 April 2025 / Accepted: 14 April 2025 / Published: 19 April 2025

Abstract

:
Background: In the randomized, phase-3 IKEMA trial, the triplet isatuximab, carfilzomib, and dexamethasone (IsaKd) demonstrated superior clinical benefit compared to those of carfilzomib and dexamethasone alone in patients with relapsed/refractory multiple myeloma after 1–3 prior treatments. Methods: Our real-world, AENEID study aimed to evaluate the efficacy and safety of IsaKd in patients who relapsed after frontline lenalidomide treatment, poorly represented in the IKEMA trial. Specifically, in the present multicenter analysis, we enrolled eighty-two patients who received, between April 2022 and September 2024 and outside of clinical trials, at least one cycle of IsaKd as a second-line treatment at the first relapse after induction therapy, autologous stem cell transplantation (ASCT), and lenalidomide maintenance. Results: After a median follow-up time of 12.9 months (range, 1–77), the overall response rate, at least a very good partial response rate, and median progression-free survival time were 79.3%, 56.1%, and 24.4 months, respectively. This slightly lower performance compared to that in the IKEMA study may be attributed to the well-known poor prognostic impact of lenalidomide refractoriness (len-R), developed by all our patients during maintenance therapy, and to a higher proportion of patients with extramedullary disease present in our series, which was identified as the only factor significantly affecting the PFS in multivariable analysis. The median overall survival was not reached, as in the pivotal trial, while the 1-year survival probability was 85.1%. Regarding the safety profile, our findings were consistent with those of the IKEMA trial, with no new safety signals reported. Conclusions: These real-world data support the use of IsaKd as a valuable option for len-R MM patients relapsing after the first-line therapy, including ASCT and lenalidomide maintenance.

1. Introduction

Patients with refractory or relapsed multiple myeloma (MM) after first-line treatments, including lenalidomide—either in combination with dexamethasone alone (Rd), as a part of triplets, such as bortezomib–lenalidomide–dexamethasone (VRd) or daratumumab–lenalidomide–dexamethasone (DRd), or as a maintenance therapy following autologous stem cell transplantation (ASCT)—represent a growing and clinically relevant population [1,2,3]. In this context, most combination treatments currently available in daily practice as second-line therapies were approved based on pivotal trials evaluating triplet regimens incorporating daratumumab or isatuximab (in patients not refractory to anti-CD38 antibodies), pomalidomide, and bortezomib or carfilzomib [4,5,6,7,8,9]. However, in most of these studies, detailed data on the clinical outcomes of patients who are truly refractory to lenalidomide (len-R) after only one prior line of therapy remain limited.
Promising results come from the prospective, open-label, phase-3 IKEMA trial (clinicaltrials.gov identifier: NCT03275285) [8,9], in which 302 patients with refractory/relapsed MM (RRMM) were randomly assigned to receive isatuximab (Isa) in combination with carfilzomib (K) and dexamethasone (d) (IsaKd) (179 patients) versus Kd (123 patients) after a median of two prior lines of therapy (range: 1–3). Isatuximab is an IgG1 monoclonal antibody that targets a specific epitope of CD38, inducing myeloma cell death through multiple mechanisms, including antibody-dependent cell-mediated cytotoxicity, complement-mediated cytotoxicity, and direct apoptosis [10,11]. Carfilzomib, a second-generation proteasome inhibitor, irreversibly binds the active sites of the 20S proteasome, as well as the core component within the 26S proteasome; by selectively inhibiting these proteasomes, carfilzomib delays proliferation and induces apoptosis in malignant plasma cells [12].
In the most recent update of the IKEMA trial [9], after a median follow-up time of 44 months, the median progression-free survival (mPFS) time was 35.7 months in the intention-to-treat population treated with IsaKd, with 72 patients (40%) considered as lenalidomide exposed and 57 patients (32%) as len-R. However, the number of patients who had progressed on frontline lenalidomide was, indeed, very small (n = 8), and a specific mPFS for this subgroup could not be evaluated.
Therefore, to further contribute to this clinically relevant issue, the AENEID study (A rEtrospective, observatioNal, multicEnter study of Isatuximab, car-filzomib and Dexamethasone as second line treatment in multiple myeloma patients relapsed/refractory after an initial therapy including lenalidomide) aimed to evaluate the efficacy and safety of IsaKd in this specific subset of patients, who were underrepresented in the IKEMA trial.

2. Results

2.1. Patients

Eighty-two patients (median age: 62 years), treated, at the first relapse, with IsaKd between April 2022 and September 2024, after induction therapy, ASCT, and lenalidomide maintenance, were included in this study. At the data cut-off point, the median follow-up time was 12.9 months (range: 1–77.1). The baseline characteristics of the patients are summarized in Table 1.
Most of the patients (86.6%) received bortezomib–thalidomide–dexamethasone as induction therapy and a single ASCT (53.7%). The median duration of the lenalidomide maintenance was 20 months (range: 1–60). Guideline-based dose reduction of lenalidomide was opted for in 31 patients (37.8%) mainly because of hematological toxicities. Sixty-seven patients (81.7%) showed symptomatic relapse; 15 (18.3%), a biochemical relapse. At the start of the IsaKd, thirty-eight patients (46.3%) were in ISS stage I; twenty-three (28.1%), in stage II; twelve (14.6%), in stage III; while in nine patients (11%), ISS was not available. Cytogenetic data were available for 50 patients (61%), 13 of whom were classified at high risk (26%), considering the same criteria of the IKEMA trial (the presence of del(17p), t(4;14), or t(14;16)), while in 15 patients (30%), chromosome 1q abnormalities were observed. Extramedullary disease (EMD) was reported in 16 cases (19.5%). Twenty-seven patients (32.9%) showed cardiac comorbidities (Table 1) before starting IsaKd therapy.

2.2. Efficacy

At the last follow-up time, the median number of IsaKd cycles administered was 7 (range: 1–24). Sixty-five patients (79.3%) achieved at least a partial response (PR) (Figure 1), with a median of three cycles (range: 1–11); more specifically, three patients (3.6%) achieved a stringent complete response (sCR); twenty-four (29.3%), a complete response (CR); nineteen (23.2%), a very good partial response (VGPR); and nineteen (23.2%), a PR.
Moreover, the median time from the initial treatment to the initiation of the second-line IsaKd was 40.5 months (range: 20–105). The median times to the first response and the best response were 2 months (range: 1–4) and 3 months (range: 1–12), respectively.
After a median follow-up time of 12.9 months, twenty-four (29.3%) patients experienced disease progression or death. The mPFS time was 24.4 months (95% CI: 15.27-NA), and the 1-year PFS probability rate was 65.76% (Figure 2A).
No differences in terms of PFS were observed considering the age (<62 vs. ≥62 years; HR = 0.98, 95% CI: 0.93–1.04; p = 0.360), the ISS stage (II + III vs. I; HR = 2.15, 95% CI: 0.95–4.87; p = 0.062), the duration of maintenance ((≤12 vs. > 12 months; HR = 0.87, 95% CI: 0.29–2.59; p = 0.810)/(≤24 vs. > 24 months; HR = 0.77, 95% CI: 0.36–1.66; p = 0.510)), the number of previous ASCTs (single vs. tandem; HR = 1.51, 95% CI: 0.7–3.26; p = 0.290), the cytogenetic risk (high vs. standard; HR = 0.5, 95% CI: 0.14–1.74, p = 0.260) or the 1q21+ status (absent vs. present; HR = 0.5, 95% CI: 0.14–1.78; p = 0.282), the type of relapse (clinical vs. laboratory; HR = 0.65, 95% CI: 0.19–2.17; p = 0.480), and LDH values (elevated vs. normal; HR = 0.52, 95% CI: 0.24–1.15; p = 0.100). Notably, multivariable analysis showed that EMD was the only parameter significantly associated with an inferior PFS (HR = 0.24, 95% CI: 0.11–0.53; p = 0.00013).
A total of 28 (34.1%) patients discontinued the IsaKd treatment, primarily because of disease progression (24 patients, 85.7%). Following IsaKd discontinuation, 19 patients (23.2%) received a subsequent treatment. The median time to the next treatment (TTNT) was 26.3 months (95% CI: 17.2-NA), with a 1-year probability rate of requiring retreatment of 67.9%. More specifically, eleven patients (57.9%) received EloPd (elotuzumab, pomalidomide, and dexamethasone), five patients (26.3%) were treated with D-PACE (dexamethasone, cisplatin, doxorubicin, cyclophosphamide, and etoposide), and the remaining three patients received Ixa-Rd (ixazomib, lenalidomide, and dexamethasone), teclistamab, and Pd, respectively. A total of 13 deaths (15.8%) were recorded. The median overall survival (OS) time was not reached (95% CI, NA-NA), and the 1-year OS probability rate was 85.12% (Figure 2B).

2.3. Safety

Infusion-related reactions occurred during the first administration of isatuximab in nine patients (10.9%, all grades 1–2) and were promptly resolved in all the cases, with no treatment discontinuation reported. Hematological toxicity included grades 3/4 thrombocytopenia (30.5%), lymphocytopenia (19.5%), neutropenia (17.1%), and anemia (10.9%) (Table 2).
Among the all-grade infectious events, upper respiratory tract infections and pneumonia were each reported in 11 patients (13.4%), while bacteremia occurred in 3.6% of the subjects (n = 3). Four cases of SARS-CoV-2 infections were also reported. Cardiac toxicity consisted of grades I and II hypertension (13.4% of the patients). Guideline-based dose reduction of carfilzomib was opted for in 26 patients (31.7%) because of age, hypertension onset, and hematological toxicity, without compromising the treatment efficacy. Overall, the treatment was permanently discontinued in four patients (4.9%) because of infectious complications. Certainly, because this is a retrospective study, some lower-grade toxicities may have been underreported.

3. Discussion

Despite the availability of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 antibodies, which have significantly improved the prognosis of MM patients in recent years, most still experience a pattern of alternating remissions and relapses [13,14], generally with progressively shorter durations of response to each subsequent regimen [15]. Furthermore, relapses within 12 months after the initial therapy with novel agents are associated with worse prognoses. These patients, currently considered to be at “functional” high risk [16,17], represent an unmet clinical need. Lenalidomide, an IMiD having the CBRL (cereblon) protein as the target for its anti-neoplastic activities, represents a key component of most first-line multi-agent regimens, both in transplant-eligible patients, as a part of induction treatment (VRd) and/or in maintenance therapy following ASCT, and in transplant-ineligible MM patients (VRd, Rd, and DRd) [18]. Because lenalidomide is usually continued until disease progression or intolerance, most MM patients become len-R. Therefore, this aspect represents a relevant issue for the choice of appropriate approaches in the context of patients with RRMM disease, particularly at the first relapse. Notably, no phase-3 trials have been exclusively conducted in len-R patients in this setting, and real-world data remain limited [19,20,21,22].
In reviewing the literature data [23], the phase-3 OPTIMISMM [5,6] and phase-2 EMN011 [24] trials (investigating pomalidomide- and dexamethasone-based combinations with bortezomib or carfilzomib, respectively) are among the few that report outcomes in len-R patients after a single prior line of therapy, particularly in terms of mPFS.
Focusing on patient populations comparable to ours (i.e., len-R patients after the first-line ASCT treatment and lenalidomide maintenance), the OPTIMISMM trial (with a median follow-up time of 16.4 months) showed a mPFS time of 22 months and an overall response rate (ORR) of 91.1% with second-line PVd. Regarding the EMN011 trial, with a longer median follow-up time of 40 months, the mPFS time with carfilzomib–pomalidomide–dexamethasone (KPd) was 32 months, while the ORR and ≥VGPR were 92% and 75%, respectively. Overall, the clinical outcome of the patients treated with IsaKd reported in our analysis was slightly better than that reported in the OPTIMISMM trial but inferior to that observed in the EMN011 study.
In real-world settings, a recent single-center report [19] included 138 len-R MM patients who initiated various salvage regimens after the first-line therapy. The results were disappointing: The ORR to the second-line therapy in len-R patients was 53%, the mPFS time was 10.7 months, and the mOS time was 23.8 months. However, in this study, the percentage of len-R patients receiving ASCT as a part of their first-line treatment was only 26%, with the best estimated mPFS time (18.4 months) obtained using a not otherwise specified combination of PI and IMIDs.
Other real-world experiences in len-R patients treated after one prior therapy have reported quite short mPFS times: 11.9 months (in 11 patients treated with elotuzumab–carfilzomib–lenalidomide–dexamethasone at median follow-up time of 28.2 months) [20] and 15 months (in 79 patients treated with daratumumab–bortezomib–dexamethasone at a median follow-up time of 25 months) [21].
Compared to the IKEMA trial [8,9], in our cohort, both the mPFS time and ORR were lower (24.4 vs. 35.7 months and 79.3% vs. 86.6%, respectively), despite IKEMA patients having received a median number of two prior lines of treatment. However, all the patients in our study were len-R, whereas only eight len-R patients after one line of therapy were included in the IKEMA study, and specific mPFS data were not reported because of the small sample size. Moreover, in our cohort, there was a higher percentage of patients relapsing with EMD (19.5%), a poor prognosis population significantly less represented in the IsaKd arm of the IKEMA study (6.7%) [25]. The prognostic impact of EMD was confirmed in multivariable analysis. As in the IKEMA trial [26], mOS was not reached in our cohort.
De Novellis et al. [22] also reported a subgroup analysis of 69 real-world RRMM patients treated with IsaKd as a second-line therapy, including 47 len-R ones (68%), whose outcomes were, unfortunately, not specified. With a median follow-up time of 12 months, the ORR was 88%, and at least VGPR was 66%. The PFS and OS were not reached but were negatively influenced by the high cytogenetic risk, prior daratumumab exposure, the achievement of less than the VGPR, and the advanced R-ISS stage. Moreover, progression within 12–24 months after initiating lenalidomide maintenance negatively affected the PFS, while severe renal impairment impacted the OS. In this study, no difference emerged based on prior ASCT, the presence of EMD, or lenalidomide exposure/refractoriness status.
Finally, the IONA-MM is an ongoing multinational observational study, including patients with RRMM treated with IsaKd in a real-world setting after a median of two prior lines of therapy [27]. Among 129 patients previously treated with lenalidomide, 65 received IsaKd as a second-line therapy, with 33 being len-R at the last prior line. In len-R patients, the ORR was 72.1% (49.2% ≥ VGPR and 23% ≥ CR). In those treated at the second line, the ORR was 75% (54.2% had ≥ VGPR and 25% had ≥ CR). However, in the latest interim analysis, the data on the IsaKd outcomes, specifically in len-R patients after the first-line ASCT and lenalidomide maintenance, were not reported.
Our study includes the largest cohort of patients treated with IsaKd after intensive first-line lenalidomide-containing treatment, showing encouraging, though still not completely satisfying, results in this selected subset of patients. Notably, the triplet IsaKd demonstrated clinical benefit also in high-risk MM patients, as the PFS did not differ according to the ISS stage or cytogenetic risk, apparently overcoming known prognostic parameters, although the limited number of patients analyzed could have influenced this result.
An emerging area of interest is the prognostic role of chromosome 1q abnormalities [28,29], which negative impact on prognosis has been widely investigated in newly diagnosed MM and appears to be equally relevant in RRMM [30]. In our cohort, 15 patients showed chromosome 1q abnormalities. As in the IKEMA subgroup analysis [31], IsaKd appeared to mitigate the adverse prognostic impact of this cytogenetic abnormality, supporting its role in this difficult-to-treat subgroup of patients.
Poor survival outcomes are also associated with early relapse (within 12 months) after ASCT, even in the era of novel drugs [32,33]. Again, and consistent with the post hoc subgroup analysis from the IKEMA trial about the outcomes of patients with early versus late relapsed MM [34], our data showed no PFS differences based on the maintenance duration, reinforcing the efficacy of IsaKd, regardless of the relapse timing.
Randomized comparisons of double versus single ASCTs following three-drug induction have suggested a possible benefit of tandem ASCT in patients with high-risk disease [35,36]. In our cohort, no differences in terms of the PFS were observed between patients who underwent single versus tandem ASCTs, indicating that IsaKd can achieve clinical goals, regardless of the number of prior transplant procedures.
Regarding the safety profile, our data are consistent with those of the IKEMA trial, with no new safety signals observed. Notably, twenty-seven patients (32.9%) showed cardiac comorbidities, including hypertension and atrial fibrillation, prior to initiating IsaKd therapy. These findings support the safety of this triplet regimen, even in these patients typically underrepresented in clinical trials.

4. Materials and Methods

4.1. Patients

This retrospective study enrolled 82 patients, at 19 Italian hematology centers, who received at least one cycle of IsaKd, from April 2022 to September 2024, as a second-line treatment (therefore, at the first relapse) after induction therapy, ASCT and lenalidomide maintenance, outside of clinical trials. Data, such as age, gender, date of diagnosis, laboratory parameters, treatment history, and date of the last follow-up or death, were extracted from clinical records at the time of the inclusion and updated on an ongoing basis. The study protocol was reviewed and approved by the involved institutional ethics committees, in accordance with the principles of the Declaration of Helsinki.

4.2. Treatment

All the patients were treated with IsaKd according to marketing approval [8]. Specifically, patients received isatuximab (10 mg/kg) intravenously (on days 1, 8, 15, and 22 of the first 28-day cycle and days 1 and 15 of subsequent cycles), carfilzomib (20 mg/sqm) intravenously (on days 1 and 2 of cycle 1) and then at 56 mg/sqm (on days 8, 9, 15, and 16 of cycle 1 and days 1, 2, 8, 9, 15, and 16 of subsequent cycles), and dexamethasone (20 mg) was administered intravenously or orally (on days 1, 2, 8, 9, 15, 16, 22, and 23) until disease progression, unacceptable toxicity, or clinical decision. Premedication included diphenhydramine (25–50 mg) or its equivalent, ranitidine (50 mg) or its equivalent, and acetaminophen (650–1000 mg) or its equivalent, administered 30 to 90 min before isatuximab infusion. Dexamethasone was administered prior to isatuximab infusion as a part of both premedication and study treatment. All the patients received antibacterial and antiviral prophylaxis during treatment according to each center’s policy.

4.3. Endpoints

The primary endpoints of our analysis were the ORR (at least the PR), PFS, OS, TTNT, and assessment of the safety profile. The treatment response and disease progression were evaluated according to the International Myeloma Working Group (IMWG) criteria [37,38], with the ORR defined as achieving at least a PR and including the VGPR, CR, and sCR. Safety was evaluated using the National Cancer Institute’s Common Terminology Criteria for Adverse Events version 6.0 (CTCAE v6.0).

4.4. Statistical Analysis

Kaplan–Meier analysis was employed to comprehensively evaluate the PFS, OS, and TTNT, using data from datasets after cleaning and preprocessing. The PFS, OS, and TTNT were calculated from the initiation of the IsaKd treatment until disease progression, death, or the last follow-up, and the Kaplan–Meier method was used to estimate survival probabilities, including 1-year probabilities for short-term outcome evaluation. Median values (mPFS, mOS, and mTTNT) and 95% confidence intervals (CIs) were estimated. Log-rank tests were applied to compare survival distributions. Additionally, univariable and multiple Cox regression analyses were performed to assess the prognostic impacts of covariates, such as age, ISS stage, duration of maintenance, number of ASCTs, type of relapse, cytogenetic characteristics, LDH levels, chromosome 1 abnormalities, and EMD presence, providing hazard ratios (HRs) with corresponding 95% CIs. Statistical analyses were conducted using R-4.4.1, utilizing the survival, survminer, and dplyr packages, with a significance threshold set at p ≤ 0.05.

5. Conclusions

The current study has several limitations, mainly because of its retrospective, real-life design, the absence of a control group, a relatively short follow-up period, and the lack of measurable residual disease (MRD) data, which is an emerging issue also in RRMM. Nevertheless, our analysis supports the clinical efficacy and safety of IsaKd as a first-salvage therapy in a specific, and still existing, subset of MM patients who relapse during lenalidomide maintenance following ASCT. This category, which has not been extensively investigated, remains ineligible for more effective therapies, such as CAR-T and bispecific antibodies.

Author Contributions

P.M. and N.S. conceived and led the project; N.S. conducted database building, extraction, and coding; N.S. and E.R. queried and analyzed the data; P.M. and N.S. wrote the main manuscript text and created the tables. All the authors recruited participants, collected data, and reviewed and approved the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the local ethics committee (Comitato Etico Territoriale—Azienda Ospedaliero–Universitaria “Consorziale Policlinico”, 70124, Bari, Italy; study number 7863; date of approval: 15 January 2025).

Informed Consent Statement

Informed consent was obtained from all the subjects involved in this study.

Data Availability Statement

Although these data are not currently publicly available for sharing, requests for sharing can be sent to the corresponding author and will be evaluated on an individual basis. The data will be provided after their deidentification, in compliance with applicable privacy laws, data protection, and requirements for consent and anonymization.

Acknowledgments

Thanks to Gabriella Cangemi (Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Italy), Melania Carlisi (Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy), Valentina Zammit (UO di Oncoematologia e trapianto di midollo, La Maddalena, Palermo, Italy), Santo Neri (Dipartimento di Oncoematologia, Ospedale Papardo, Messina, Italy), Giuseppina Uccello (UOC Ematologia, ARNAS Garibaldi Nesima di Catania, Italy), Vincenzo Leone (UOSD Ematologia Castelvetrano, Italy), and Vito Pier Gagliardi (Hematology and Stem Cell Transplantation Unit, AOUC Policlinico, Bari, Italy). The authors thank Donata Grazia Coladonato for the language revision of this manuscript.

Conflicts of Interest

P.M. has received honoraria from and served on the advisory boards for: Celgene, Janssen, Takeda, Bristol Myers Squibb (BMS), Amgen, Novartis, Gilead, Jazz, Sanofi, AbbVie, and GlaxoSmithKline (GSK). M.T.P. has received honoraria from and served on the advisory boards for: Janssen, BMS, Amgen, Sanofi, GSK, Takeda, Oncopeptides, Pfizer, Menarini, AbbVie. F.D.R. has received onoraria from: GSK, Sanofi, Amgen, Pfizer, and Abbvie. All other authors have declared no conflicts of interest.

References

  1. Merz, M.; Dechow, T.; Scheytt, M.; Schmidt, C.; Hackanson, B.; Knop, S. The clinical management of lenalidomide-based therapy in patients with newly diagnosed multiple myeloma. Ann. Hematol. 2020, 99, 1709–1725. [Google Scholar] [CrossRef] [PubMed]
  2. Moreau, P.; Zamagni, E.; Mateos, M.V. Treatment of patients with multiple myeloma progressing on frontline-therapy with lenalidomide. Blood Cancer J. 2019, 9, 38. [Google Scholar] [CrossRef] [PubMed]
  3. Botta, C.; Martino, E.A.; Conticello, C.; Mendicino, F.; Vigna, E.; Romano, A.; Palumbo, G.A.; Cerchione, C.; Martinelli, G.; Morabito, F.; et al. Treatment of Lenalidomide Exposed or Refractory Multiple Myeloma: Network Meta-Analysis of Lenalidomide-Sparing Regimens. Front. Oncol. 2021, 11, 643490. [Google Scholar] [CrossRef]
  4. Palumbo, A.; Chanan-Khan, A.; Weisel, K.; Nooka, A.K.; Masszi, T.; Beksac, M.; Spicka, I.; Hungria, V.; Munder, M.; Mateos, M.V.; et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2016, 375, 754–766. [Google Scholar] [CrossRef]
  5. Richardson, P.G.; Oriol, A.; Beksac, M.; Liberati, A.M.; Galli, M.; Schjesvold, F.; Lindsay, J.; Weisel, K.; White, D.; Facon, T.; et al. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): A randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 781–794. [Google Scholar] [CrossRef] [PubMed]
  6. Dimopoulos, M.; Weisel, K.; Moreau, P.; Anderson, L.D., Jr.; White, D.; San-Miguel, J.; Sonneveld, P.; Engelhardt, M.; Jenner, M.; Corso, A.; et al. Pomalidomide, bortezomib, and dexamethasone for multiple myeloma previously treated with lenalidomide (OPTIMISMM): Outcomes by prior treatment at first relapse. Leukemia 2021, 35, 1722–1731. [Google Scholar] [CrossRef]
  7. Dimopoulos, M.A.; Terpos, E.; Boccadoro, M.; Delimpasi, S.; Beksac, M.; Katodritou, E.; Moreau, P.; Baldini, L.; Symeonidis, A.; Bila, J.; et al. Daratumumab plus pomalidomide and dexamethasone versus pomalidomide and dexamethasone alone in previously treated multiple myeloma (APOLLO): An open-label, randomised, phase 3 trial. Lancet Oncol. 2021, 22, 801–812. [Google Scholar] [CrossRef]
  8. Moreau, P.; Dimopoulos, M.A.; Mikhael, J.; Yong, K.; Capra, M.; Facon, T.; Hajek, R.; Špička, I.; Baker, R.; Kim, K.; et al. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): A multicentre, open-label, randomised phase 3 trial. Lancet 2021, 397, 2361–2371. [Google Scholar] [CrossRef]
  9. Martin, T.; Dimopoulos, M.A.; Mikhael, J.; Yong, K.; Capra, M.; Facon, T.; Hajek, R.; Špička, I.; Baker, R.; Kim, K.; et al. Isatuximab, carfilzomib, and dexamethasone in patients with relapsed multiple myeloma: Updated results from IKEMA, a randomized Phase 3 study. Blood Cancer J. 2023, 13, 72, Erratum in Blood Cancer J. 2023, 13, 152. [Google Scholar] [CrossRef]
  10. Deckert, J.; Wetzel, M.C.; Bartle, L.M.; Skaletskaya, A.; Goldmacher, V.S.; Vallée, F.; Zhou-Liu, Q.; Ferrari, P.; Pouzieux, S.; Lahoute, C.; et al. SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin. Cancer Res. 2014, 20, 4574–4583. [Google Scholar] [CrossRef]
  11. Feng, X.; Zhang, L.; Acharya, C.; An, G.; Wen, K.; Qiu, L.; Munshi, N.C.; Tai, Y.T.; Anderson, K.C. Targeting CD38 Suppresses Induction and Function of T Regulatory Cells to Mitigate Immunosuppression in Multiple Myeloma. Clin. Cancer Res. 2017, 23, 4290–4300. [Google Scholar] [CrossRef] [PubMed]
  12. Perel, G.; Bliss, J.; Thomas, C.M. Carfilzomib (Kyprolis): A Novel Proteasome Inhibitor for Relapsed and/or Refractory Multiple Myeloma. Pharm. Ther. 2016, 41, 303–307. [Google Scholar]
  13. Kumar, S.K.; Dimopoulos, M.A.; Kastritis, E.; Terpos, E.; Nahi, H.; Goldschmidt, H.; Hillengass, J.; Leleu, X.; Beksac, M.; Alsina, M.; et al. Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: A multicenter IMWG study. Leukemia 2017, 31, 2443–2448. [Google Scholar] [CrossRef]
  14. Gandhi, U.H.; Cornell, R.F.; Lakshman, A.; Gahvari, Z.; McGehee, E.; Jagosky, M.H.; Gupta, R.; Varnado, W.; Fiala, M.A.; Chhabra, S.; et al. Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia 2019, 33, 2266–2275. [Google Scholar] [CrossRef] [PubMed]
  15. Kumar, S.K.; Therneau, T.M.; Gertz, M.A.; Lacy, M.Q.; Dispenzieri, A.; Rajkumar, S.V.; Fonseca, R.; Witzig, T.E.; Lust, J.A.; Larson, D.R.; et al. Clinical course of patients with relapsed multiple myeloma. Mayo Clin. Proc. 2004, 79, 867–874. [Google Scholar] [CrossRef]
  16. Majithia, N.; Rajkumar, S.V.; Lacy, M.Q.; Buadi, F.K.; Dispenzieri, A.; Gertz, M.A.; Hayman, S.R.; Dingli, D.; Kapoor, P.; Hwa, L.; et al. Early relapse following initial therapy for multiple myeloma predicts poor outcomes in the era of novel agents. Leukemia 2016, 30, 2208–2213. [Google Scholar] [CrossRef] [PubMed]
  17. Soekojo, C.Y.; Chung, T.H.; Furqan, M.S.; Chng, W.J. Genomic characterization of functional high-risk multiple myeloma patients. Blood Cancer J. 2022, 12, 24. [Google Scholar] [CrossRef]
  18. 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. Hemasphere 2021, 5, e528, Erratum in Hemasphere 2021, 5, e567; Erratum in Hemasphere 2021, 5, e659. [Google Scholar]
  19. Kastritis, E.; Ntanasis-Stathopoulos, I.; Theodorakakou, F.; Migkou, M.; Roussou, M.; Malandrakis, P.; Kanellias, N.; Eleutherakis-Papaiakovou, E.; Fotiou, D.; Spiliopoulou, V.; et al. Characteristics and Outcomes of Patients with Relapsed/Refractory Multiple Myeloma After Exposure to Lenalidomide in First Line of Therapy: A Single Center Database Review in Greece. Clin. Lymphoma Myeloma Leuk. 2024, 24, 468–477. [Google Scholar] [CrossRef]
  20. Bhutani, M.; Foureau, D.M.; Robinson, M.; Guo, F.; Fesenkova, K.; Atrash, S.; Paul, B.; Varga, C.; Friend, R.; Pineda-Roman, M.; et al. A Clinical and Correlative Study of Elotuzumab, Carfilzomib, Lenalidomide, and Dexamethasone (Elo-KRd) for Lenalidomide Refractory Multiple Myeloma in First Relapse. Clin. Lymphoma Myeloma Leuk. 2023, 23, 535–544. [Google Scholar] [CrossRef]
  21. Rizzello, I.; Sacchetti, I.; Barbato, S.; Solli, V.; Stefanoni, P.; Cani, L.; Pavan, L.; Quaresima, M.; Belotti, A.; Sgherza, N.; et al. P-416 A Multicenter Observational Retrospective Study of Second-Line Treatment with Daratumumab-Bortezomib-Dexamethasone (DaraVd) in Multiple Myeloma (MM) Patients Refractory to Lenalidomide. Clin. Lymphoma Myeloma Leuk. 2024, 24, S275. [Google Scholar] [CrossRef]
  22. De Novellis, D.; Derudas, D.; Vincelli, D.; Fontana, R.; Della Pepa, R.; Palmieri, S.; Accardi, F.; Rotondo, F.; Morelli, E.; Gigliotta, E.; et al. Clinical Efficacy of Isatuximab Plus Carfilzomib and Dexamethasone in Relapsed/Refractory Multiple Myeloma Patients. Eur. J. Haematol. 2025, 114, 105–114. [Google Scholar] [CrossRef] [PubMed]
  23. Mele, G.; Sgherza, N.; Pastore, D.; Musto, P. Strengths and Weaknesses of Different Therapeutic Strategies for the Treatment of Patients with Multiple Myeloma Who Progress After the Frontline Use of Lenalidomide: A Narrative Review. J. Clin. Med. 2024, 13, 6238. [Google Scholar] [CrossRef]
  24. Sonneveld, P.; Zweegman, S.; Cavo, M.; Nasserinejad, K.; Broijl, A.; Troia, R.; Pour, L.; Croockewit, S.; Corradini, P.; Patriarca, F.; et al. Carfilzomib, Pomalidomide, and Dexamethasone as Second-line Therapy for Lenalidomide-refractory Multiple Myeloma. Hemasphere 2022, 6, e786, Erratum in Hemasphere 2023, 7, e856. [Google Scholar] [CrossRef]
  25. Beksac, M.; Spicka, I.; Hajek, R.; Bringhen, S.; Jelínek, T.; Martin, T.; Mikala, G.; Moreau, P.; Symeonidis, A.; Rawlings, A.M.; et al. Evaluation of isatuximab in patients with soft-tissue plasmacytomas: An analysis from ICARIA-MM and IKEMA. Leuk. Res. 2022, 122, 106948. [Google Scholar] [CrossRef] [PubMed]
  26. Yong, K.; Martin, T.; Dimopoulos, M.A.; Mikhael, J.; Capra, M.; Facon, T.; Hajek, R.; Špička, I.; Baker, R.; Kim, K.; et al. Isatuximab plus carfilzomib-dexamethasone versus carfilzomib-dexamethasone in patients with relapsed multiple myeloma (IKEMA): Overall survival analysis of a phase 3, randomised, controlled trial. Lancet Haematol. 2024, 11, e741–e750. [Google Scholar] [CrossRef] [PubMed]
  27. Gaballa, M.R.; Martin, T.G.; Tsukada, N.; Suzuki, K.; Iriuchishima, H.; Chalayer, E.; Camus, V.; Alcala Peña, M.M.; Furlan, A.; Hubmann, M.C.G.; et al. Real-World Experience with Isatuximab in Patients with Relapsed and/or Refractory Multiple Myeloma (RRMM): Iona-MM Second Interim Analysis. Blood 2024, 144 (Suppl. 1), 2411.5. [Google Scholar] [CrossRef]
  28. Neupane, K.; Fortuna, G.G.; Dahal, R.; Schmidt, T.; Fonseca, R.; Chakraborty, R.; Koehn, K.A.; Mohan, M.; Mian, H.; Costa, L.J.; et al. Alterations in chromosome 1q in multiple myeloma randomized clinical trials: A systematic review. Blood Cancer J. 2024, 14, 20. [Google Scholar] [CrossRef]
  29. D’Agostino, M.; Rota-Scalabrini, D.; Belotti, A.; Bertamini, L.; Arigoni, M.; De Sabbata, G.; Pietrantuono, G.; Pascarella, A.; Tosi, P.; Pisani, F.; et al. Additional copies of 1q negatively impact the outcome of multiple myeloma patients and induce transcriptomic deregulation in malignant plasma cells. Blood Cancer J. 2024, 14, 94. [Google Scholar] [CrossRef]
  30. Ziccheddu, B.; Biancon, G.; Bagnoli, F.; De Philippis, C.; Maura, F.; Rustad, E.H.; Dugo, M.; Devecchi, A.; De Cecco, L.; Sensi, M.; et al. Integrative analysis of the genomic and transcriptomic landscape of double-refractory multiple myeloma. Blood Adv. 2020, 4, 830–844. [Google Scholar] [CrossRef]
  31. Martin, T.; Richardson, P.G.; Facon, T.; Moreau, P.; Perrot, A.; Spicka, I.; Bisht, K.; Inchauspé, M.; Casca, F.; Macé, S.; et al. Primary outcomes by 1q21+ status for isatuximab-treated patients with relapsed/refractory multiple myeloma: Subgroup analyses from ICARIA-MM and IKEMA. Haematologica 2022, 107, 2485–2491. [Google Scholar] [CrossRef] [PubMed]
  32. Kumar, S.; Mahmood, S.T.; Lacy, M.Q.; Dispenzieri, A.; Hayman, S.R.; Buadi, F.K.; Dingli, D.; Rajkumar, S.V.; Litzow, M.R.; Gertz, M.A. Impact of early relapse after auto-SCT for multiple myeloma. Bone Marrow Transpl. 2008, 42, 413–420. [Google Scholar] [CrossRef] [PubMed]
  33. Jimenez-Zepeda, V.H.; Reece, D.E.; Trudel, S.; Chen, C.; Tiedemann, R.; Kukreti, V. Early relapse after single auto-SCT for multiple myeloma is a major predictor of survival in the era of novel agents. Bone Marrow Transpl. 2015, 50, 204–208. [Google Scholar] [CrossRef] [PubMed]
  34. Facon, T.; Moreau, P.; Baker, R.; Min, C.K.; Leleu, X.; Mohty, M.; Karlin, L.; Armstrong, N.M.; Tekle, C.; Schwab, S.; et al. Isatuximab plus carfilzomib and dexamethasone in patients with early versus late relapsed multiple myeloma: IKEMA subgroup analysis. Haematologica 2024, 109, 604–616. [Google Scholar] [CrossRef]
  35. Cavo, M.; Gay, F.; Beksac, M.; Pantani, L.; Petrucci, M.T.; Dimopoulos, M.A.; Dozza, L.; van der Holt, B.; Zweegman, S.; Oliva, S.; et al. Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): A multicentre, randomised, open-label, phase 3 study. Lancet Haematol. 2020, 7, e456–e468, Erratum in Lancet Haematol. 2020, 7, e443; Erratum in Lancet Haematol. 2020, 7, e785. [Google Scholar]
  36. Hari, P.N.; Pasquini, M.C.; Stadtmauer, E.A.; Fraser, R.; Fei, M.; Devine, S.M.; Efebera, Y.A.; Geller, N.L.; Horowitz, M.M.; Koreth, J.; et al. Long-term follow-up of BMT CTN 0702 (STaMINA) of postautologous hematopoietic cell transplantation (autoHCT) strategies in the upfront treatment of multiple myeloma (MM). J. Clin. Oncol. 2020, 38, 8506. [Google Scholar] [CrossRef]
  37. Durie, B.G.; Harousseau, J.L.; Miguel, J.S.; Bladé, J.; Barlogie, B.; Anderson, K.; Gertz, M.; Dimopoulos, M.; Westin, J.; Sonneveld, P.; et al. International uniform response criteria for multiple myeloma. Leukemia 2006, 20, 1467–1473. [Google Scholar] [CrossRef]
  38. Rajkumar, S.V.; Harousseau, J.L.; Durie, B.; Anderson, K.C.; Dimopoulos, M.; Kyle, R.; Blade, J.; Richardson, P.; Orlowski, R.; Siegel, D.; et al. Consensus recommendations for the uniform reporting of clinical trials: Report of the International Myeloma Workshop Consensus Panel 1. Blood 2011, 117, 4691–4695. [Google Scholar] [CrossRef]
Figure 1. Treatment efficacy.
Figure 1. Treatment efficacy.
Pharmaceuticals 18 00595 g001
Figure 2. Progression-free survival (A) and overall survival (B).
Figure 2. Progression-free survival (A) and overall survival (B).
Pharmaceuticals 18 00595 g002
Table 1. Main characteristics of the patients at the time of the IsaKd initiation.
Table 1. Main characteristics of the patients at the time of the IsaKd initiation.
Median Age, Years (Range)62 (43–73)
Sex, n (%)
Male42 (51.2)
Female40 (48.8)
M-Protein Type, n (%)
IgG47 (57.3)
IgA19 (23.2)
Light chain only15 (18.3)
Not secernent1 (1.2)
Light Chain Type, n (%)
Kappa44 (53.7)
Lambda37 (45.1)
Not secreting1 (1.2)
Creatinine Clearance, n (%)
<60 mL/min13 (15.8)
≥60 mL/min69 (84.2)
LDH, n (%)
Normal23 (28)
Elevated59 (72)
β2-Microglobulin (mg/L), n (%)
<3.544 (53.7)
≥3.5 <5.517 (20.7)
>5.512 (14.6)
Unknown or missing9 (11)
International Staging System (ISS), n (%)
I38 (46.3)
II23 (28.1)
III12 (14.6)
Not available9 (11.0)
Type of Relapse, n (%)
Laboratory15 (18.3)
Clinical67 (81.7)
FISH Analysis *, n (%; Considering Data Available for 50 Patients)
High risk13 (26)
Standard risk37 (74)
1q21 Abnormalities15 (30)
Gain(1q)9 (60)
Amp(1q)6 (40)
Extramedullary Disease, n (%)16 (19.5)
Induction Treatment Before ASCT, n (%)
VTd71 (86.6)
D-VTd4 (4.9)
PAd3 (3.7)
VRd1 (1.2)
VCd3 (3.6)
Autologous Stem Cell Transplantation, n (%)
Single44 (53.7)
Tandem38 (46.3)
Median Duration of Lenalidomide Maintenance, Months (Range)20 (1–61)
<12 months of lenalidomide maintenance; patients, n (%)25 (30.5)
<24 months of lenalidomide maintenance; patients, n (%)48 (58.5)
Cardiac Comorbidities Before Isa-Kdtreatment, n (%)27 (32.9)
Hypertension, n (%)24 (29.3)
Atrial fibrillation, n (%)3 (3.6)
* High risk was defined as del(17p), t(4;14), or t(14;16); VTd: bortezomib–thalidomide–dexamethasone; D-VTd: daratumumab–bortezomib–thalidomide–dexamethasone; PAd: bortezomib–adriamycin–dexamethasone; VRd: bortezomib–lenalidomide–dexamethasone; VCd: bortezomib–cyclophosphamide–dexamethasone.
Table 2. Safety.
Table 2. Safety.
Hematological Toxicity (Grades III and IV), n (%)
Thrombocytopenia25 (30.5)
Lymphocytopenia16 (19.5)
Neutropenia14 (17.1)
Anemia9 (10.9)
Cardiac Toxicity (Grades I and II), n (%)
Hypertension11 (13.4)
Infectious Events (Any Grade), n (%)
Pneumonia11 (13.4)
Upper airway infection11 (13.4)
SARS-CoV-2 infection4 (4.9)
Sepsis3 (3.6)
CMV * infection2 (2.4)
HZV ** infection1 (1.2)
Conjunctivitis1 (1.2)
Cellulitis1 (1.2)
* Cytomegalovirus; ** herpes zoster virus.
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

Sgherza, N.; Battisti, O.; Curci, P.; Conticello, C.; Palmieri, S.; Derudas, D.; Germano, C.; Martino, E.A.; Mele, G.; Pepa, R.D.; et al. Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study. Pharmaceuticals 2025, 18, 595. https://doi.org/10.3390/ph18040595

AMA Style

Sgherza N, Battisti O, Curci P, Conticello C, Palmieri S, Derudas D, Germano C, Martino EA, Mele G, Pepa RD, et al. Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study. Pharmaceuticals. 2025; 18(4):595. https://doi.org/10.3390/ph18040595

Chicago/Turabian Style

Sgherza, Nicola, Olga Battisti, Paola Curci, Concetta Conticello, Salvatore Palmieri, Daniele Derudas, Candida Germano, Enrica Antonia Martino, Giuseppe Mele, Roberta Della Pepa, and et al. 2025. "Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study" Pharmaceuticals 18, no. 4: 595. https://doi.org/10.3390/ph18040595

APA Style

Sgherza, N., Battisti, O., Curci, P., Conticello, C., Palmieri, S., Derudas, D., Germano, C., Martino, E. A., Mele, G., Pepa, R. D., Fazio, F., Mele, A., Rossini, B., Palazzo, G., Roccotelli, D., Rasola, S., Petrucci, M. T., Pastore, D., Tarantini, G., ... Musto, P. (2025). Efficacy and Safety of Isatuximab, Carfilzomib, and Dexamethasone (IsaKd) in Multiple Myeloma Patients at the First Relapse After Autologous Stem Cell Transplantation and Lenalidomide Maintenance: Results from the Multicenter, Real-Life AENEID Study. Pharmaceuticals, 18(4), 595. https://doi.org/10.3390/ph18040595

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