latest treatment strategies aiming for a cure in ... · latest treatment strategies aiming for a...

7
Vol:.(1234567890) International Journal of Hematology (2020) 111:512–518 https://doi.org/10.1007/s12185-020-02841-w 1 3 PROGRESS IN HEMATOLOGY Latest treatment strategies aiming for a cure in transplant‑eligible multiple myeloma patients: how I cure younger MM patients with lower cost Kenshi Suzuki 1 Received: 6 February 2020 / Accepted: 6 February 2020 / Published online: 3 March 2020 © Japanese Society of Hematology 2020 Abstract This article presents a practical overview of the treatment of younger, newly diagnosed multiple myeloma patients, focus- ing on novel treatment strategies. With the introduction of effective new agents, multiple myeloma is one of the most active and changing fields in clinical oncology. In addition, monitoring technology has become reliable and practical. Achieving and sustaining minimal residual disease negativity (MRD– ), such as multiparameter flow cytometry (MFC) < 10 −5 , is one of the goals of therapy. MRD– is significantly associated with prolonged progression-free survival, whereas MRD persis- tence (MRD +) is an independent factor for poor progression-free survival. Evidence from several recent studies evaluating modern therapy has further supported the positive correlation between depth of response and outcomes. Multiple myeloma can become a chronic illness with sustained MRD– in a significant number of patients. Our ultimate hope is to leverage tumoricidal-immunomodulatory sequential therapies and to cure a subset of our patients. Keywords Multiple myeloma · Autologous stem cell transplantation · Minimal residual disease negativity · Cure of myeloma · Healthcare costs Introduction Multiple myeloma (MM) is an incurable disease, and responsiveness to treatment varies among patients due to the high heterogeneity of the disease [1, 2]. The decision on which treatment is best for MM has been a difficult issue. However, treatment of MM has advanced remarkably in the past 10 years (Fig. 1). MM patients can achieve long- term remissions and survival [39]. According to an IMWG Research Project, sustaining a complete response (CR) at 1 year was associated with better progression-free survival (PFS) (median 3.3 years vs. 2.6 years, P < 0.0001), as well as overall survival (OS) (median 8.5 years vs. 6.3 years, P < 0.0001) [7]. This article reviews the latest trends and future developments in the treatment of MM. Induction regimen Bortezomib‑lenalidomide‑dexamethasone (VRd/ VRd‑lite)‑autologous stem cell transplantation (ASCT) For newly diagnosed, physically fit patients younger than 70 years, ASCT after induction therapy remains stand- ard [10, 11]. Incorporation of novel agents into induction regimens further improves survival outcomes. A three- drug combination that includes immunomodulatory drugs (IMiDs) such as lenalidomide (Len) or pomalidomide (Pom), antibodies, and proteasome inhibitors (PIs) may be optimal. Len/dexamethasone (Dex) (Rd) is the backbone of many triplet regimens (Fig. 2). Bortezomib (Bor)-containing triplet regimens, such as Bor/thalidomide (Thal)/Dex (VTd), Bor/Len/Dex (VRd), and Bor/cyclophosphamide (CPA)/Dex (VCd), are highly active in patients with newly diagnosed MM. Recent studies show that triplet regimens contain- ing IMiDs and PIs offer better response rates, as well as improved PFS compared with doublets. More importantly, a Recent Advances in Biology and Treatment of Multiple Myeloma * Kenshi Suzuki [email protected] 1 Director of Myeloma and Amyloidosis Center, Japanese Red Cross Medical Center, Shibuya-ku, Hiroo 4-1-22, Tokyo, Japan

Upload: others

Post on 08-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

Vol:.(1234567890)

International Journal of Hematology (2020) 111:512–518https://doi.org/10.1007/s12185-020-02841-w

1 3

PROGRESS IN HEMATOLOGY

Latest treatment strategies aiming for a cure in transplant‑eligible multiple myeloma patients: how I cure younger MM patients with lower cost

Kenshi Suzuki1

Received: 6 February 2020 / Accepted: 6 February 2020 / Published online: 3 March 2020 © Japanese Society of Hematology 2020

AbstractThis article presents a practical overview of the treatment of younger, newly diagnosed multiple myeloma patients, focus-ing on novel treatment strategies. With the introduction of effective new agents, multiple myeloma is one of the most active and changing fields in clinical oncology. In addition, monitoring technology has become reliable and practical. Achieving and sustaining minimal residual disease negativity (MRD– ), such as multiparameter flow cytometry (MFC) < 10−5, is one of the goals of therapy. MRD– is significantly associated with prolonged progression-free survival, whereas MRD persis-tence (MRD +) is an independent factor for poor progression-free survival. Evidence from several recent studies evaluating modern therapy has further supported the positive correlation between depth of response and outcomes. Multiple myeloma can become a chronic illness with sustained MRD– in a significant number of patients. Our ultimate hope is to leverage tumoricidal-immunomodulatory sequential therapies and to cure a subset of our patients.

Keywords Multiple myeloma · Autologous stem cell transplantation · Minimal residual disease negativity · Cure of myeloma · Healthcare costs

Introduction

Multiple myeloma (MM) is an incurable disease, and responsiveness to treatment varies among patients due to the high heterogeneity of the disease [1, 2]. The decision on which treatment is best for MM has been a difficult issue. However, treatment of MM has advanced remarkably in the past 10 years (Fig. 1). MM patients can achieve long-term remissions and survival [3–9]. According to an IMWG Research Project, sustaining a complete response (CR) at 1 year was associated with better progression-free survival (PFS) (median 3.3 years vs. 2.6 years, P < 0.0001), as well as overall survival (OS) (median 8.5 years vs. 6.3 years, P < 0.0001) [7]. This article reviews the latest trends and future developments in the treatment of MM.

Induction regimen

Bortezomib‑lenalidomide‑dexamethasone (VRd/VRd‑lite)‑autologous stem cell transplantation (ASCT)

For newly diagnosed, physically fit patients younger than 70 years, ASCT after induction therapy remains stand-ard [10, 11]. Incorporation of novel agents into induction regimens further improves survival outcomes. A three-drug combination that includes immunomodulatory drugs (IMiDs) such as lenalidomide (Len) or pomalidomide (Pom), antibodies, and proteasome inhibitors (PIs) may be optimal. Len/dexamethasone (Dex) (Rd) is the backbone of many triplet regimens (Fig. 2). Bortezomib (Bor)-containing triplet regimens, such as Bor/thalidomide (Thal)/Dex (VTd), Bor/Len/Dex (VRd), and Bor/cyclophosphamide (CPA)/Dex (VCd), are highly active in patients with newly diagnosed MM. Recent studies show that triplet regimens contain-ing IMiDs and PIs offer better response rates, as well as improved PFS compared with doublets. More importantly, a

Recent Advances in Biology and Treatment of Multiple Myeloma

* Kenshi Suzuki [email protected]

1 Director of Myeloma and Amyloidosis Center, Japanese Red Cross Medical Center, Shibuya-ku, Hiroo 4-1-22, Tokyo, Japan

Page 2: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

513Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:…

1 3

recent phase 3 trial has shown that OS is superior with VRd as initial therapy compared with Rd. In patients over the age of 65 and those who have received more than four cycles of Rd, stem cells must be mobilized with granulocyte colony-stimulating factor (G-CSF) together with plerixafor [12]. In the IFM/DFCI trial, the CR rate was higher in the ASCT group than in the VRd-alone group (59% vs. 48%, P = 0.03), as was the minimal residual disease negativity (MRD–) rate (79% vs. 65%, P < 0.001) [10]. However, OS at 4 years did not differ significantly between the two groups (81% and 82%, respectively). An increasing range of treatment options is available for newly diagnosed and relapsed or refractory MM (RRMM); however, there remains no global standard of care. Despite clinical trials showing benefits of continuous

versus fixed duration therapy, shorter treatment durations are seen in the real world [13].

Autologous stem cell transplantation

High-dose melphalan followed by ASCT improves PFS and OS in MM, with treatment-related mortality (TRM) less than 1%. Hence this treatment should be considered in all eligible patients. Although early ASCT after four cycles of initial therapy is preferred, some randomized trials show similar OS whether ASCT is early or delayed. Recent stud-ies comparing up-front ASCT with a protracted course of novel agents with deferred ASCT at first relapse found that the former approach resulted in superior CR, MRD− rates, and PFS. Furthermore, ASCT can be considered for salvage in fit patients if the interval between the first ASCT and relapse is 18 months or more. The time to progression of disease (PD) (19 vs. 11 months) and OS (67 vs. 52 months) were significantly better with salvage ASCT. ASCT remains important in the era of novel agents [14].

Post transplantation consolidation/maintenance therapy

In general, the term “consolidation” refers to a short course of therapy following definitive initial therapy, while “main-tenance” refers to a more prolonged course of treatment with a lower-intensity regimen. The IMWG Research Project found a statistical cure fraction of 14.3% [15]. This result identifies CR as an important predictor of long-term sur-vival for ASCT-eligible MM patients, and clinical variables reflective of higher disease burden as poor prognostic mark-ers. The study identified achieving CR in the first year of diagnosis as an important landmark associated with superior PFS and OS. After achieving MRD–, sustaining it by various combination strategies including immunological effects may be more important for the cure (Fig. 3).

Multi consolidation/maintenance until sustained MRD‑negativity for 2 years by continuous DRd‑Kd‑EPd‑IRd/PCd

Despite improvements in PFS and OS, relapses are almost inevitable for most MM patients, primarily due to incom-plete eradication of MRD. To improve the outcome of ASCT, it is important to eradicate MM tumor cells through the synergistic immunological effects of ASCT and peri- and post-ASCT treatment (Fig. 4). In retrospective observational studies, prolonged therapy appeared to result in better out-comes. There is a great debate concerning the role of achiev-ing MRD– with multiparameter flow cytometry (MFC) and sustained MRD– as a goal of therapy. Although data show

Fig. 1 Historical perspective: Progress in multiple myeloma treat-ment options. Overall survival has improved from 3 to 7–10 years and beyond. Many powerful new drugs are further increasing the depth and duration of remission

Fig. 2 Many options for initial therapy of multiple myeloma (NCCN Guideline 2020). Choice of initial treatment is complicated by the novel agents. Triplet regimens increase response and improve out-come

Page 3: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

514 K. Suzuki

1 3

that MRD– has favorable prognostic value, additional trials are needed to determine if changes in treatment need to be made based on MRD status (Fig. 5).

Anti‑CD38 antibodies

A synergistic effect of anti-CD38 antibodies with IMiDs is supposed because the CD38 expression level in MM cell lines is substantially up-regulated by Pom and Len [16–19]. A global, ongoing phase 3 clinical trial (the MAIA trial) is comparing the clinical benefit of daratumumab (Dara)-Rd versus Rd in patients with newly diagnosed MM. In MAIA, Dara-Rd treatment significantly reduced the risk of PD or death by 44%.

Isatuximab (Isa) is another antibody that targets CD38. Single-agent Isa had an overall response rate (ORR) of 24.3%, with a median PFS and OS of 3.7 and 18.6 months, respectively, in patients with RRMM [20]. Several stud-ies are currently ongoing, including one of Pd with or without Isa in RRMM (ORR 62%, with a median PFS of 17.6 months) and another of VRd with or without Isa in patients with newly diagnosed MM [21].

Fig. 3 Imaging for cure by combined tumoricidal and immunomodu-latory sequential therapies. After achieving minimal residual disease negativity (MRD–, sustaining it by various combination strategies including immunological effects may be more important for the cure

Fig. 4 Strategy of treatment of multiple myeloma in transplant-eligi-ble patients in our institute (plan). For the induction and consolida-tion phases, agents with deep and quick response should be selected; however, for the maintenance phase, agents with high safety and immunological power should be considered. We divided treatment into four phases. The initial therapy consisted of four courses of VRd-lite followed by PBSC-harvest. In cases of under partial response, an additional two cycles of KRd therapy may be performed with the use of plerixafor (a phase). In our clinical practice, up-front ASCT is per-formed, followed by continuous multi consolidation/maintenance reg-imens until minimal residual disease negativity (MRD–) is sustained for 2 years with DRd-Kd-EPd-Rd every 6 months (b phase). Patients with confirmed MRD– at 2 years will be actively monitored for resur-gence of MRD or clinical relapse. After that, the treatment is stopped, with careful watching. Progression of disease (PD) is defined as (1) > 25% elevation in M-protein, (2) hypercalcemia, (3) progres-sion of anemia, (4) bone pain, (5) β2-MG elevation, (6) additional chromosomal abnormalities, or (7) elevation of bone marrow (BM) myeloma cells. After PD, problem-oriented ASCT may be performed after two cycles of VTD-PACE therapy (c phase). In patients under 60  years of age whose families agree to allo-SCT, we choose allo-SCT with ERd-R maintenance (d phase). BM aspiration for evalua-tion of depth of response and MRD before ASCT, after DRd therapy, and at 6 months between regimens

Fig. 5 Minimal residual disease-driven decision making at consoli-dation/maintenance periods. Monitoring of minimal residual disease (MRD) kinetics not only can predict prognosis more precisely, but also can be helpful to guide treatment decisions, particularly when deciding when to start retreatment of relapsed patients. MRD nega-tivity (MRD–) guides treatment modification. MFC is currently con-sidered a sensitive co-adjuvant test, not only for diagnostic screening but also as a surrogate marker of multiple myeloma. Bone marrow aspiration is performed for evaluation of depth of response and MRD before and after ASCT with 6-month intervals between each regi-men. Our optimal responses are VGPR at harvest periods, CR at after ASCT, and MRD– at the end of DRd. MRD-driven decision-making should be done at consolidation/maintenance periods

Page 4: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

515Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:…

1 3

KRd/Kd

In Japan, KRd was approved for RRMM by the phase 3 ASPIRE trial and Kd by the phase 3 ENDEAVOR trial [22–24]. In addition, once-weekly administration of Car was submitted for RRMM by the phase 3 ARROW trial. Once-weekly KRd (carfilzomib 56 mg/m2) showed prom-ising activity with an acceptable safety profile in patients with newly diagnosed MM and reduce the burden on elderly patients and those living far from hospitals. These results merit additional evaluation of a convenient wKRd regimen in patients with newly diagnosed MM.

Anti‑SLAMF7 antibody

Signaling lymphocytic activation molecule family member 7 (SLAMF7) is expressed on myeloma cells [25]. Elotu-mumab (Elo) in combination with Rd (ERd) was approved for RRMM according to the results of the ELOQUENT-2 study. The ELOQUENT-3 study showed promising efficacy of Elo in combination with Pd (EPd). EPd had good ORR (53%) compared with Pd (26%). The most remarkable fea-ture of Elo is its high safety. Interestingly, adding Elo to Pom-Dex improved Pom-induced neutropenia [26–29].

IRd

Ixazomib (Ixa) was approved in Japan as the first oral PI in combination with Rd (IRd) for RRMM (TOURMALINE-MM1 [30–33]). In the TOURMALINE-MM3 trial, patients after ASCT were randomly assigned to receive Ixa main-tenance therapy or placebo. A 28% reduction in the risk of progression or death with Ixa versus placebo (median PFS, 26.5 vs. 21.3 months) was reported. No increase in second malignancies was noted with Ixa therapy compared with placebo.

PVd/PCd

Pd is approved in Japan for patients with RRMM [34]. In the phase 3 OPTIMISMM trial, Bor + Pd (PVd) prolonged PFS compared with Vd in RRMM patients who had received one to three prior regimens including Len (median 11.2 vs. 7.1 months, P < 0.0001) [35–37]. An oral regimen of Pom/CPA/Dex (PCd) in the first relapse after exposure to Len and Bor is efficacious and safe. PCd maintenance may be useful for sustained MRD–. The importance of an IMiD-free interval until the start of Pd was reported. OS, PFS, and PFS2 were improved in patients who had an inter-val of ≥ 18 months from the last dose of Len to the first dose of Pom compared with those who had an interval of < 18 months (PFS, 10.5 vs. 4 months, P = 0.002). This result suggests that the Len-free interval may prolong PFS

and shows the possibility of the emergence of IMiD-sensi-tive clones after an IMiD-free interval.

Ongoing clinical trials of four‑drug combinations (quad induction)

I believe that we are moving toward using quadruplet treat-ment regimens, based on data from the CASSIOPEIA trial and others. In CASSIOPEIA, at 100 days after ASCT, 39% of patients in the Dara-VTd group versus 26% in the VTd group achieved CR or better, and 64% versus 44% achieved MRD– (both P < 0.0001) [38]. In a trial of Dara + VRd (GRIFFIN), 16 patients were enrolled and completed nine or more cycles of Dara-VRd [39]. By the end of consoli-dation, all patients had reached VGPR or better, and 63% achieved CR (including 25% sCR and 50% MRD–). Twelve patients developed infections. No deaths due to serious adverse events were reported, and no patients discontinued treatment due to adverse events. Based on this study, a phase 3 study of Dara-VRd was initiated. In this study, patients in the Dara-VRd group who achieve sustained MRD– for 12 months after ≥ 24 months of maintenance will stop Dara but continue Len maintenance until PD; upon loss of CR or MRD–, patients will restart Dara treatment. The results of this study will address how MRD– guides treatment modi-fication. A phase 2 trial of the combination of Dara with KRd is ongoing.

Experimental maintenance strategy

Some studies are ongoing to aim to improve response. The duration of maintenance and the stopping rule differ among the protocols. It is too early to estimate how much the MRD– rate would increase during maintenance. In the Dara-VRd safety-run in cohort (n = 16), the response contin-ued to deepen during maintenance (sCR, 25% at the end of consolidation, 63% during maintenance). It is anticipated the more patients with MRD– will be seen after longer follow-up times.

Anti‑B‑cell maturation antigen (BCMA) chimeric antigen receptor‑T (CAR‑T)

BCMA has been targeted for CAR-T cell approaches against MM, and preclinical studies showed regression of BCMA-expressing MM cells. The first phase 1 study of CAR-T cell therapy targeting BCMA in 33 patients with a median of 7 prior regimens was reported [40]. The data cutoff date was 6.2 months after the last infusion date. Twenty-five patients had cytokine release syndrome, which was grade 3 in two patients. Neurologic toxic effects occurred in 14 patients and

Page 5: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

516 K. Suzuki

1 3

were grade 1 or 2 in 13 patients. One patient had a revers-ible grade 4 neurologic toxic effect. The ORR was 85%, including 45% CR. Six of the 15 patients who had a CR have had a relapse. CAR-T cell expansion was associated with responses, and CAR-T cells persisted up to 1 year after infusion. In the future, we should consider early introduction of CAR-T therapy for high-risk patients, such as those who relapse within a year after ASCT.

VTD‑PACE

In addition to the regimens discussed above, another option is multiagent combination chemotherapy, such as VTD-PACE (Bor/Thal/Dex, cisplatin, doxorubicin, CPA, and etoposide). VTD-PACE is particularly useful in patients with aggressive disease, such as plasma cell leukemia or multiple extramedullary plasmacytomas [41]. The efficacy of VTD-PACE-like regimens in the treatment of RRMM may be based on the combination of PACE for 4 days of continuous drip infusion with an interval of 4–6 weeks.

Allo‑SCT

Previous studies suggested that allogeneic stem cell trans-plantation (allo-SCT) can overcome high-risk cytogenet-ics associated with poor outcome in MM. Despite having the potentially curative therapy for MM, the use of allo-SCT remains controversial due to the unacceptable high non-relapse mortality (10–15%), extensive morbid chronic graft-versus-host disease (50–80%), and high relapse rates (50–60%) [42]. Allo-SCT is not recommended as part of initial therapy. There are conflicting data about its clinical benefit, and the TRM rate (10–20%) remains a concern. For patients who survive the first year, the high GVHD rate and the likelihood of relapse remain formidable issues. It would be reasonable to consider allo-SCT in selected young patients with high-risk disease who are willing to accept high TRM for a chance of better long-term survival [43].

Supportive care

Although our previous consensus for adjunctive manage-ment of vascular thrombosis, skeletal complications, infec-tions, and peripheral neuropathy remains valid, emerging concerns pertaining to specific new drugs will need to be addressed. Hepatitis B virus infection is highly endemic in Asia, and a major concern is the risk of its reactivation after the use of anti-CD38 therapy. Hence, it is mandatory to screen for both HBsAg and anti-hepatitis B virus core antibody.

Financial impact

With all myeloma therapies given as combinations, three major considerations for the practicing hematologist are combined toxicities across agents, decision making for ther-apeutic strategies offered to patients, and total costs, includ-ing traffic fees. Several other factors should be considered when planning treatment strategy, such as logistics, drug availability, social considerations, comorbidities, and patient preference, as each may influence treatment choice. There-fore, biomarker-driven or other decision support tools for precision medicine would be of significant utility in guiding myeloma practice to match each patient (Fig. 6). The thera-peutic progress in MM has come with a significant financial impact on both patients and society. Most of the new drugs approved for MM in recent years cost more than $100,000 per year of therapy. With a shifting paradigm toward con-tinuous therapy, the cumulative cost of treatment can be as high as $500,000 for initial therapy, including induction, consolidation, and maintenance. Since most patients who achieve remission with initial therapy eventually relapse, the cost of newer agents creates an unsustainable financial burden for patients and the society. A study in the US state of Washington reported that the rate of financial bankruptcy was 2.65 times higher in cancer patients than in those with-out cancer. We present data to argue that these medications and associated expenses are of direct benefit to patients and society. Long-term studies have shown that, even with trans-plantation, a small minority of patients can be cured now.

Fig. 6 Future treatment strategy. Evidence of future treatment strat-egy will come from Japan. Future treatment will be like Total Ther-apy X, which consists of a triplet (or quadruplet treatment regimens) and two to four regimens every 6 months. New myeloma treatments are evaluated for efficacy, cost, and safety. Admission periods maybe 2–3  months (including ASCH + ASCT, Dara-induction, and any infections). Total healthcare costs should be 0.5 million dollars, over 10 years survival with good QOL

Page 6: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

517Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:…

1 3

Perhaps an optimal strong induction (e.g., Dara-KRd) fol-lowed by consolidation with ASCT and CAR-T will cure more patients. If so, the future will be not only brighter, but also more economical.

Conclusion

As mentioned above, the therapy and treatment strategy of MM have largely changed in recent years. Ongoing efforts to improve the treatment paradigm even further include using oncogenomics to better characterize molecular pathogenesis and to develop refined patient stratification and personalized treatment of MM using immune-based therapies, including antibodies, cytokines, and novel immunocytic strategies. Understanding the profile of oncogene and tumor suppres-sor gene mutations with their interactions and impact on the prognosis of MM can improve the definition of disease subsets and identify pathways important in disease patho-biology. At the same time, it is becoming more important to control the disease over the long term while maintaining the quality of life of the patient. The increased number of treatment options enables us to select personalized medicine according to the systemic condition of the patient. Moreover, understanding the characteristics of novel agents and using them in combination with existing drugs appropriately for the individual patient is critical. Finally, we should consider how we can help patients to live long, active lives throughout the treatment.

Compliance with ethical standards

Conflict of interest K. Suzuki received honoraria from Celgene, Take-da, ONO, Amgen, Novartis, Sanofi, Bristol-Myers Squibb, AbbVie and Janssen, and consulted for Amgen, Janssen, Takeda, and Celgene; received research funding from Bristol-Myers Squibb, Celgene and Amgen.

References

1. Suzuki K. Current therapeutic strategy for Multiple myeloma. J Clin Oncol. 2013;43:116–24.

2. Kim K, Lee JH, Kim JS, Min CK, Yoon SS, Shimizu K, et al. Clinical profiles of multiple myeloma in Asia–an Asian Myeloma Network study. Am J Hematol. 2014;89:751–6.

3. Barlogie B. Toward a cure for multiple myeloma? N Engl J Med. 1991;325:1304–6.

4. Barlogie B, Mitchell A, van Rhee F, Epstein J, Morgan GJ, Crow-ley J. Curing myeloma at last: defining criteria and providing the evidence. Blood. 2014;124:3043–51.

5. Katodritou E, Papadaki S, Konstantinidou P, Terpos E. Is it possi-ble to cure myeloma without allogeneic transplantation? Transfus Apheres Sci. 2016;54:63–70.

6. Davies FE. Is molecular remission the goal of multiple myeloma therapy? ASH Educ Book. 2017;1:205–11.

7. Usmani SZ, Hoering A, Cavo M, Miguel JS, Goldschimdt H, Hajek R, et al. Clinical predictors of long-term survival in newly diagnosed transplant eligible multiple myeloma—an IMWG Research Project. Blood Cancer J. 2018;8:123–30.

8. Ravi P, Shaji K, Kumar SK, Cerhan JR, Maurer MJ, Dingli D, et al. Defining cure in multiple myeloma: a comparative study of outcomes of young individuals with myeloma and curable hematologic malignancies. Blood Cancer J. 2018;8:26–33.

9. Choudhry P, Galligan D, Wiita AP. Seeking convergence and cure with new myeloma therapies. Trends Cancer. 2018;4:567–82.

10. Durie BGM, Hoering A, Abidi MH, Rajkumar SV, Epstein J, Kahanic SP, et al. Bortezomib with lenalidomide and dexameth-asone 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;389:519–27.

11. Attal M, Lauwers-Cances V, Hulin C, Leleu X, Caillot D, Escoffre M, et al. IFM 2009 Study: Lenalidomide, Bortezomib, and Dex-amethasone with transplantation for Myeloma. N Engl J Med. 2017;376:1311–20.

12. Miyazaki K, Suzuki K. Poor mobilizer and its countermeasures. Transfus Apher Sci. 2018;57:623–7.

13. Dhakal B, Szabo A, Chhabra S, Hamadani M, D’Souza A, Usmani SZ, et al. Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: a systematic review and meta-analysis. JAMA Oncol. 2018;4:343–50.

14. Cook G, Royle KL, O’Connor S, Cairns DA, Ashcroft AJ, Wil-liams CD, et al. National Cancer Research Institute Haemato-oncology Clinical Studies Group: the effect of salvage autolo-gous stem-cell transplantation on overall survival in patients with relapsed multiple myeloma (final results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a randomised, open-label, phase 3 trial. Lancet Haematol. 2016;3:340–51.

15. Botta et al. Best of ASH. Monday, December 3, 2018 San Diego, Blood Cancer in press 2018.

16. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hans-son M, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373:1207–19.

17. Palumbo A, Chanan-Khan A, Weisel K, Nooka AK, Masszi T, Beksac M, et al. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375:754–66.

18. Dimopoulos MA, Oriol A, Nahi H, San-Miguel J, Bahlis NJ, Usm-ani SZ, et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375:1319–31.

19. Suzuki K, Dimopoulos MA, Takezako N, Okamoto S, Shinagawa A, Matsumoto M, et al. Daratumumab, lenalidomide, and dexa-methasone in East Asian patients with relapsed or refractory mul-tiple myeloma: subgroup analyses of the phase 3 POLLUX study. Blood Cancer J. 2018;8:41.

20. Martin T, Baz R, Benson DM, et al. A phase 1b study of isatuxi-mab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood. 2017;129:3294–303.

21. Mikhael J, Richardson PG, Usmani SZ, Raje N, Bensinger W, Karanes C, et al. A phase 1b study of Isatuximab plus pomalido-mide/dexamethasone in relapsed/refractory multiple myeloma. Blood. 2019;134:123–33.

22. Siegel DS, Dimopoulos MA, Ludwig H, Facon T, Goldschmidt H, Jakubowiak A, et al. Improvement in overall survival with carfil-zomib, lenalidomide, and dexamethasone in patients with relapsed or refractory multiple myeloma. J Clin Oncol. 2018;36:728–34.

23. Moreau P, Mateos MV, Berenson JR, Weisel K, Lazzaro A, Song K, et al. Once weekly versus twice weekly carfilzomib dos-ing in patients with relapsed and refractory multiple myeloma (A.R.R.O.W): interim analysis results of a randomized, phase 3 study. Lancet Oncol. 2018;19:953–64.

Page 7: Latest treatment strategies aiming for a cure in ... · Latest treatment strategies aiming for a cure in transplant-eligible multiple myeloma patients:… 513 13 recentphase3trialhasshownthatOSissuperiorwithVRd

518 K. Suzuki

1 3

24. Suzuki K, Ri M, Chou T, Sugiura I, Takezako N, Sunami K, et al. Carfilzomib, lenalidomide and dexamethasone in patients with heavily pretreated multiple myeloma: a phase 1 study in Japan. Cancer Sci. 2017;108:461–8.

25. Kikuchi J, Hori M, Iha H, Toyama-Sorimachi N, Hagiwara S, Kuroda Y, et al. Soluble SLAMF7 promotes the growth of mye-loma cells via homophilic interaction with surface SLAMF7. Leukemia. 2020;34:180–95.

26. Lonial S, Dimopoulos M, Palumbo A, White D, Grosicki S, Spicka I, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med. 2015;373:621–31.

27. Suzuki K, Sunami K, Ohashi K, Iida S, Mori T, Handa H, et al. Randomized phase 3 study of elotuzumab for relapsed or refrac-tory multiple myeloma: ELOQUENT-2 Japanese patient suba-nalysis. Blood Cancer J. 2017;7:e540.

28. Dimopoulos MA, Lonial S, Betts KA, Chen C, Zichlin ML, Brun A, et al. Elotuzumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended 4-year follow-up and analysis of relative progression-free survival from the rand-omized ELOQUENT-2 trial. Cancer. 2018;124:4032–43.

29. Dimopoulos MA, Dytfeld D, Grosicki S, Moreau P, Takezako N, Hori M, et al. Elotuzumab plus Pomalidomide and Dexametha-sone for Multiple Myeloma. N Engl J Med. 2018;379:1811–22.

30. Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, et al. Oral ixazomib, lenalidomide, and dexamethasone for multi-ple myeloma. N Engl J Med. 2017;374:1621–34.

31. Suzuki K, Handa H, Chou T, Ishizawa K, Takubo T, Kase Y, et al. Phase 1 study of ixazomib alone or combined with lenalidomide-dexamethasone in Japanese patients with relapsed/refractory mul-tiple myeloma. Int J Hematol. 2017;105:445–52.

32. Dimopoulos MA, Gay F, Schjesvold F, Beksac M, Hajek R, Wei-sel KC, et al. Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): a double-blind, randomized, placebo-controlled phase 3 trial. Lancet. 2019;393:253–64.

33. Avet-Loiseau H, Bahlis NJ, Chng WJ, Masszi T, Viterbo L, Pour L, et al. Ixazomib significantly prolongs progression-free sur-vival in high-risk relapsed/refractory myeloma patients. Blood. 2017;130:2610–8.

34. Suzuki K, Matsue K, Sunami K. Pomalidomide + Bortezomib + Low-Dose Dexamethasone (PVd) in Japanese Patients Enrolled in OPTIMISMM. In: The 81th annual meeting of the JSH. 2019; OS1-70-4.

35. Garderet L, Kuhnowski F, Berge B, Roussel M, Escoffre-Barbe M, Lafon I, et al. Pomalidomide, cyclophosphamide, and dexametha-sone for relapsed multiple myeloma. Blood. 2018;132:2555–633.

36. Richardson PG, Siegel DS, Vij R, Hofmeister CC, Baz R, Jagan-nath S, et al. Pomalidomide alone or in combination with low-dose dexamethasone in relapsed and refractory multiple myeloma: a randomized phase 2 study. Blood. 2014;123:1826–32.

37. Miguel JS, Weisel K, Moreau P, Lacy M, Song K, Delforge M, et al. Pomalidomide plus low-dose dexamethasone versus high dose dexamethasone alone for patients with relapsed and refrac-tory multiple myeloma (MM-003): a randomized, open-label, phase 3 trial. Lancet Oncol. 2013;14:1055–66.

38. Moreau P, Attal M, Hulin C, Arnulf B, Belhadj K, Benboubker L, et  al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma

(CASSIOPEIA): a randomized, open-label, phase 3 study. Lancet. 2019;10192:29–38.

39. Peter MV, Cesar R, Brandi R, Nitya N, Luciano JC, Yana L, et  al. Efficacy and updated safety analysis of a safety run-in cohort from griffin, a phase 2 randomized study of Daratumumab (Dara), Bortezomib (V), Lenalidomide (R), and Dexamethasone (D; Dara‐Vrd) Vs. Vrd in patients (Pts) with Newly Diagnosed (ND) Multiple Myeloma (MM) Eligible for High-Dose Therapy (HDT) and Autologous Stem Cell Transplantation (ASCT). Blood. 2018;132:151.

40. Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 2019;380:1726–37.

41. Lakshman A, Singh PP, Rajkumar SV, Dispenzieri A, Lacy MQ, Gertz MA, et al. Efficacy of VDT PACE-like regimens in treat-ment of relapsed/refractory multiple myeloma. Am J Hematol. 2018;93:179–86.

42. Dhakal B, Vesole DH, Hari PN. Allogeneic stem cell transplanta-tion for multiple myeloma: is there a future? Bone Marrow Trans-plant. 2016;51:492–500.

43. Kawamura K, Tsukada N, Kanda Y, Ishida T, Suzuki K, Ikeda T, et al. The role of allogeneic transplantation for multiple myeloma in the era of novel agents: a study from the Japanese Society of Myeloma. Biol Blood Marrow Transplant. 2018;24:1392–8.

44. Evens AM, Jovanovic BD, Su YC, Raisch DW, Ganger D, Belknap SM, et al. Rituximab associated hepatitis B virus (HBV) reactiva-tion in lymphoproliferative diseases: meta-analysis and examina-tion of FDA safety reports. Ann Oncol. 2011;22:1170–80.

45. Johnsrud AJ, Johnsrud JJ, Susanibar SA, Kamimoto JJ, Kothari A, Burgess M, et al. Infectious and immunological sequelae of dara-tumumab in multiple myeloma. Br J Haematol. 2019;185:187–9.

46. Siddiqui M, Rajkumar SV. The high cost of cancer drugs and what we can do about it. Mayo Clin Proc. 2012;87:935–43.

47. Ramsey S, Blough D, Kirchhoff A, Kreizenbeck K, Fedorenko C, Snell K, et al. Washington State cancer patients found to be at greater risk for bankruptcy than people without a cancer diagno-sis. Health Affairs (Project Hope). 2013;32:1143–52.

48. Usmani SZ, Cavenagh JD, Belch AR, Hulin C, Basu S, White D, et al. Cost-effectiveness of lenalidomide plus dexamethasone versus bortezomib plus melphalan and prednisone in transplant-ineligible US patients with newly diagnosed multiple myeloma. J Med Econ. 2016;19:243–58.

49. MacEwan JP, Batt K, Yin W, Peneva D, Sison S, Vine S, et al. Economic burden of multiple myeloma among patients in suc-cessive lines of therapy in the United States. Leuk Lymphoma. 2018;59:941–9.

50. Fonseca R, Abouzaid S, Bonafede M, Cai Q, Parikh K, Cosler L, et al. Trends in overall survival and costs of multiple myeloma, 2000–2014. Leukemia. 2017;31:1915–21.

51. Walker BA, Mavrommatis K, Wardell CP, Ashby TC, Bauer M, Davies FE, et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood. 2018;132:587–97.

Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.