Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleClinical Studies
Open Access

Measurable Residual Disease Assessment Using Next-Generation Flow in Patients With Relapsed and Refractory Multiple Myeloma Treated With a Combination of Carfilzomib, Lenalidomide, and Dexamethasone

TAKESHI YOROIDAKA, TAKESHI YAMASHITA, RYOICHI MURATA, KYOKO YOSHIHARA, SATOSHI YOSHIHARA, MIKIO UEDA, SHINJI NAKAO, KOSEI MATSUE and HIROYUKI TAKAMATSU
Anticancer Research January 2023, 43 (1) 157-165; DOI: https://doi.org/10.21873/anticanres.16145
TAKESHI YOROIDAKA
1Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan;
2Department of Hematology, Ishikawa Prefectural Central Hospital, Ishikawa, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TAKESHI YAMASHITA
3Department of Internal Medicine, Keiju Kanazawa Hospital, Ishikawa, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RYOICHI MURATA
3Department of Internal Medicine, Keiju Kanazawa Hospital, Ishikawa, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KYOKO YOSHIHARA
4Department of Hematology, Hyogo Medical University, Hyogo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SATOSHI YOSHIHARA
4Department of Hematology, Hyogo Medical University, Hyogo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MIKIO UEDA
3Department of Internal Medicine, Keiju Kanazawa Hospital, Ishikawa, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHINJI NAKAO
1Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KOSEI MATSUE
5Department of Hematology/Oncology, Kameda Medical Center, Chiba, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROYUKI TAKAMATSU
1Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan;
6Faculty of Transdisciplinary Sciences for Innovation, Institute of Transdisciplinary Sciences for Innovation, Kanazawa University, Ishikawa, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: takamaz@staff.kanazawa-u.ac.jp
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Carfilzomib, lenalidomide, and dexamethasone (KRD) therapy is widely used for patients with relapse/refractory multiple myeloma (RRMM). However, the response in patients who underwent assessment for measurable residual disease (MRD) has not been elucidated in a prospective study. We aimed to clarify the response rate and outcome of KRD therapy in patients in RRMM, including those with MRD. Patients and Methods: Twenty-one consecutive RRMM patients treated with KRD at 4 Japanese Centers between September 2016 and October 2018 were enrolled and assessed for MRD in the bone marrow (cut-off: 1×10−5) using the EuroFlow-next-generation flow (NGF) method. Results: The median number of therapy lines before KRD was 3 (range=1-6), and the median number of KRD cycles was 4 (range=1-22). As the best overall response post-KRD therapy, 52% (11/21) of patients achieved a MRD negative complete response, 71% (15/21) achieved stringent complete response/complete response, and 14% (3/21) achieved a very good partial response. MRD negativity was achieved in 12 of 16 (75%) and 14 of 21 (67%) patients during and after KRD treatment, respectively. The 2-year progression-free survival and overall survival from the start of KRD therapy were 100% and 100%, respectively, in MRD-positive cases and 88% and 100%, respectively, in MRD-negative cases (median follow-up=1.8 years). Grade 3/4 toxicities were reported in 15 patients (71%), with thrombocytopenia being the most frequent toxicity (6 patients, 29%). Conclusion: This is the first study that prospectively assessed MRD of patients with RRMM after KRD therapy. KRD treatment achieved a high MRD negativity rate and good outcomes with manageable toxicities.

Key Words:
  • Multiple myeloma
  • next-generation flow
  • measurable residual disease
  • relapsed and refractory
  • carfilzomib
  • lenalidomide
  • dexamethasone (KRD)

Multiple myeloma (MM) is an incurable hematological malignancy, and obtaining a deep response is essential as it leads to prolonged progression-free survival (PFS) and overall survival (OS) (1). Methods for measuring measurable residual disease (MRD), such as next-generation flow (NGF) and next-generation sequencing (NGS), make it possible to stratify patients with complete response (CR) based on MRD levels, and novel agents increase the rate of CR and deeper responses (2-7).

Recently, a meta-analysis showed that MRD negativity was associated with significantly improved survival outcomes regardless of disease setting, MRD sensitivity thresholds, cytogenetic risk, method of MRD assessment, and depth of clinical response at the time of MRD measurement (1, 7).

Carfilzomib is a next-generation proteasome inhibitor (PI) that functions as an irreversible inhibitor of the β5 chymotryptic subunit of the 20S proteasome. This new PI showed superior outcomes in relapse/refractory multiple myeloma (RRMM) patients in a head-to-head comparison between carfilzomib plus dexamethasone and bortezomib plus dexamethasone (8). Several phase 3 studies showed the clinical efficacy of three-drug regimens containing carfilzomib, such as carfilzomib plus lenalidomide plus dexamethasone (KRD), carfilzomib plus dexamethasone plus daratumumab, isatuximab plus pomalidomide plus dexamethasone, and carfilzomib plus pomalidomide plus dexamethasone for RRMM patients (5, 9-11). Though several studies assessed MRD after KRD treatment in newly diagnosed multiple myeloma (NDMM), to our knowledge, no study assessing MRD after KRD therapy in patients with RRMM has been conducted. Reports from patients with NDMM showed that approximately 60% achieved complete response (CR) or stringent CR and 56-62% achieved MRD negativity by multiparameter flow cytometry (12-14).

We hypothesized that deep response could predict better prognosis and, thus, we prospectively assessed the response of RRMM treated with KRD by NGF. This is the first study that prospectively assessed MRD of the patients with RRMM after KRD therapy.

Patients and Methods

Study design and subjects. This multicenter, open-label, prospective study was conducted at 4 Japanese Centers: the Kanazawa University, the Keiju Kanazawa Hospital, the Hyogo Medical University, and the Kameda Medical Center. Studies were conducted between September 2016 and October 2018. Both transplant-eligible and -ineligible patients with RRMM were enrolled to assess the response. All patients received the physician’s choice of induction therapy and were treated with KRD after the diagnosis of RRMM. Patients with RRMM received KRD in 28-day cycles until disease progression or unacceptable toxicity. MM diagnosis was made according to the International Myeloma Working Group (IMWG) criteria (15) and response to therapy was assessed using the International Uniform Response Criteria (16). Stringent CR was defined as CR with normal free light chain ratio and absence of clonal cells in bone marrow (BM) biopsy by immunohistochemistry. Flow MRD-negative was defined as an MRD-negative status in a patient with CR. BM cells from 21 patients were analyzed using G-banding and FISH at diagnosis, and t(4;14), t(14;16), and/or del(17p13) were defined as high-risk chromosomal abnormalities by FISH. Adverse events (AEs) were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0 (17). The study protocol was approved by the local ethics committees of Kanazawa University (No. 2016-125) and at each institute, and registered in the public clinical trial database (UMIN 000027259).

KRD treatment. Carfilzomib was administered intravenously on days 1, 2, 8, 9, 15, and 16. The dose was 20 mg/m2 for days 1 and 2 of cycle 1, and 27 mg/m2 thereafter. Lenalidomide was administered orally at 25 mg on days 1 to 21 of each cycle, and dexamethasone was administered orally or intravenously at 40 mg on days 1, 8, 15, and 22 of each cycle. Dose modification was permitted to manage toxicity.

Assessment of MRD. MRD was prospectively assessed in all patients using the EuroFlow-NGF method after KRD treatment. As part of routine clinical care, 2 ml of BM aspirate were collected and analyzed at the Kanazawa University (Kanazawa, Japan). The details of EuroFlow-NGF are described elsewhere (18). Briefly, the EuroFlow method uses ammonium chloride-based bulk lysis, followed by surface staining with antibodies against CD138-BV421, CD27-BV510, CD38 multi-epitope (ME)-FITC, CD56-PE, CD45-PerCP Cy5.5, CD19-PECy7, CD117-APC, and CD81-APC-C750 in tube 1, and surface/intracellular staining using antibodies against CD138-BV421, CD27-BV510, CD38 ME-FITC, CD56-PE, CD45-PerCP Cy5.5, CD19-PECy7, cytoplasmic (cy)IgEmbedded Image-APC, and cyIgλ-APC-C750 after permeabilization in tube 2. An anti-CD38ME antibody was used to prevent the interference of anti-CD38 monoclonal antibodies, such as daratumumab. The FACSCanto™ II (BD Biosciences, Franklin Lakes, NJ, USA) flow cytometer was used to measure all samples, and gating and identification of clonal abnormal plasma cells were manually performed by experts using the Infinicyt software (Cytognos, Salamanca, Spain). The lower limit of MRD detection was set at 1×10−5. Flow MRD-negativity was determined using the IMWG criteria (patients with CR or better and with at least 10−5 MRD negativity).

Statistical analyses. Baseline characteristics were reported descriptively, with continuous variables summarized as median and range. Response to therapy was defined in “Study design and subjects”. The estimated OS and PFS were reported using the Kaplan-Meier method. All analyses were performed using GraphPad Prism version 9.3.1 (San Diego, CA, USA).

Results

Patient characteristics. A total of 21 patients were enrolled, and their responses were assessed between September 9, 2016, and October 16, 2018. The median patient age was 66 (range=30-83) years at the start of KRD treatment, the M-protein type was most commonly IgG in patients (71.4%), and 81.0% (17/21) of patients had International Staging System stage I or II disease. A total of 19% (4/21) of patients showed high-risk chromosomal abnormalities [del17p (n=3), t(14;16) (n=1)]. The median number of therapy lines before KRD was 3 (range=1-6), and the median number of KRD cycles was 4 (range=1-22). The median number of therapy lines after KRD was one (range=0-5) (Table I and Table II). The dose of KRD was not reduced in any patient due to adverse effect, but one patient (case 9) received an escalated dose of carfilzomib (36 mg/m2) from cycle 2 because of insufficient response (19) after the approval of the Department of Hematology/Oncology at the Kameda Medical Center. The MRD of 15 patients after KRD treatment (median, 4 cycles) and 6 patients (cases 5, 7, 9, 10, 16 and 21) after KRD therapy (median, 3 cycles) plus other regimens were assessed using NGF (Figure 1). After assessing the response to treatment, three patients (cases 2, 6, and 20) did not receive any subsequent therapy, 4 patients (cases 9, 14, 19, and 21) received autologous stem cell transplantation (ASCT), and 14 patients continued various treatments without ASCT (Table II).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Baseline characteristics.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Cycles of KRD therapy before NGF.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Disease response and treatment status timelines. Each color bar shows the response after KRD treatment. Patients are grouped by their MRD status. The lower limit of MRD detection was set at 1×10−5. KRD, Carfilzomib plus lenalidomide plus dexamethasone; MRD, measurable residual disease; CA, chromosomal abnormality; CR, complete response; PD, progressive disease; PR, partial response; sCR, stringent complete response; Tx, therapy; VGPR, very good partial response.

KRD response. The rate of CR or better before KRD treatment was 9.5%, and the rate of a very good partial response (VGPR) or better was 42.8% (Figure 2). Two cases (case 12 and case 13) who had stringent CR before KRD therapy subsequently received KRD therapy because both cases remained MRD positive. Progressive disease (PD) and stable disease (SD) were reported in 14.3% (3/21) and 14.3% (3/21) of patients, respectively. The rate of CR or better after KRD therapy was 71.4% (15/21) and the rate of VGPR or better was 85.7% (18/21). Notably, the overall response rate was 100% and there was no PD or SD. The response was upgraded in 19 (90%) patients and maintained in two partial response (PR) cases (10%) after KRD treatment. After KRD therapy, MRD negativity was achieved in 67% (14/21) of patients (Figure 1). As the best overall response post-KRD treatment, 52% (11/21) of patients achieved Flow MRD-negative. Three of the four patients (cases 2, 4, and 11) with high-risk cytogenetics achieved Flow MRD-negative results after KRD treatment. Although one patient (case 9) remained PR status after KRD therapy, they achieved MRD negativity with subsequent therapy.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Response rates before and after KRD therapy. The response rate was assessed in patients with RRMM before and after KRD treatment. Fifteen patients were assessed for response after KRD therapy (median 4 cycles), and 6 patients (cases 5, 7, 9, 10, 16, and 21) were assessed for response after KRD treatment (median 3 cycles) and other regimens. KRD, Carfilzomib plus lenalidomide plus dexamethasone; ORR, overall response rates; RRMM, relapse/refractory multiple myeloma.

Outcomes. After a median follow-up of 21.0 months (range=5.0-30.0) post-KRD treatment, the estimated 2-year PFS rate from the start of KRD treatment was 95% and the 2-year OS rate was 100% (Figure 3). Among the 7 patients who were MRD-positive, the estimated 2-year PFS rate was 100%, and the 2-year OS rate was 100%. In contrast, the estimated 2-year PFS rate was 93%, and the 2-year OS rate was 100% among the 14 patients who were MRD-negative. Of note, case 6, who achieved MRD negativity and received no therapy, relapsed with bone-related extramedullary disease (EMD) adjacent to the thoracic spine (Figure 1).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Survival after KRD therapy. (A) Progression-free survival and (B) overall survival. Median follow-up: 21 months. KRD, Carfilzomib plus lenalidomide plus dexamethasone.

Safety and tolerability. Table III summarizes the incidence of grade 3/4 AEs that occurred during KRD treatment. Fifteen (71%) patients reported grade 3/4 AEs. Grade 3/4 non-hematologic AEs included hypertension (14%), elevated liver function test results (14%), heart failure (5%), pneumonia (5%), sepsis (5%), fatigue (5%), and peripheral neuropathy (5%). Hematological grade 3/4 toxicities included thrombocytopenia (29%), lymphopenia (14%), and neutropenia (14%). Four grade 4 AEs were reported in two patients, including neutropenia and thrombocytopenia in one patient, and pneumonia and sepsis in the second patient. No grade 5 AEs or treatment-related deaths were reported throughout the treatment duration. Overall, KRD therapy was well tolerated, and all patients received at least one cycle of KRD with supportive measures.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table III.

Grade 3/4 adverse events.

Discussion

To the best of our knowledge, this is the first study to evaluate the response, including MRD, after KRD treatment in patients with RRMM. In this study, we demonstrated that KRD therapy can induce deep responses in patients with RRMM. Ninety percent of patients upgraded their response after KRD treatment, and 11/21 (52%) of patients achieved CR and MRD negativity (Flow MRD-negative). All patients, except one who relapsed with extramedullary disease, survived without progression during the follow-up period. The 2-year PFS and OS rates were 95% and 100%, respectively. Induction therapy with KRD and ASCT could induce 58-89% MRD negativity in NDMM patients (14, 20, 21), and KRD treatment for NDMM without intent for immediate ASCT achieved 10% MRD negativity (4). Although no studies assessing MRD after KRD treatment in the refractory and relapse settings have been reported, several studies have reported that the rates of MRD negativity after treatment were 13-46% in patients with RRMM (22-24). Compared to these studies, the rate of MRD negativity was higher in our cohort. This could be partially due to the differences in the population and timing of KRD treatment. The median number of prior regimens was three, and 57% (12 of 21) of patients received ASCT before KRD therapy in this study. In patients who received ASCT before KRD therapy, the response assessment reflected the efficacy of both ASCT and KRD therapies, which could lead to overestimation of the better responses of KRD therapy. Most MRD-negative patients (93%, 13 of 14) showed no progression, but one patient relapsed with bone-related EMD after acquiring MRD-negativity and no treatment. The patient had skin lesions before KRD treatment. Paiva et al. reported that an analysis of patients with NDMM-registered in the PETHEMA/GEM2012MENOS65 trial showed that 7% of patients experienced disease progression despite undetectable MRD assessed by NGF. Most patients relapsed with extraosseous plasmacytomas, which were already observed at diagnosis (25). Because MRD assessment by NGF carries a risk of false-negative results in the presence of EMD, imaging studies, such as positron-emission tomography/computerized tomography and whole-body magnetic resonance imaging should be performed (25). All four patients who had high-risk cytogenetic abnormalities obtained MRD negativity after KRD treatment. Previous studies showed that the rate of MRD negativity was lower in patients with high-risk cytogenetics than in those with standard-risk cytogenetics; however, there was no difference in PFS and OS after acquiring MRD negativity (20, 26-28). However, whether KRD therapy could overcome the risk of high-risk cytogenetics is still controversial (14, 20). As shown for the daratumumab-combination regimens in patients with RRMM, the rates of patients who sustained MRD negativity for more than 12 months were lower in the high-risk cytogenetic group than in the standard-risk cytogenetic group (29), and this study suggested that a short follow-up period could lead to overestimation of the regimen when comparing the effectiveness of the cytogenetic risks.

In this study, 15 patients (71%) reported grade 3/4 AEs during KRD treatment. Major grade 3 non-hematological toxicities were hypertension (14%) and elevated liver function test (14%), and grade 3/4 thrombocytopenia (29%) was the most frequently observed hematological toxicity. The types of AEs were consistent with those previously reported and the rates of AEs were less frequent (10, 19). The reason for the low rates of grade 3/4 AEs could be the small number of KRD cycles in this study.

Our study has several limitations. First, the MRD of six patients (cases 5, 7, 9, 10, 16, and 21) was assessed after KRD and other therapy regimens, which could lead to the overestimation of KRD response. In fact, two patients (cases 9 and 10) received a daratumumab-containing regimen, and one patient (case 21) received ASCT, both of which could induce a high rate of undetectable MRD. Second, the median follow-up period (21 months) was relatively short, and we could not show a significant difference in outcomes between MRD-positive and MRD-negative patients.

Novel drugs and combinations have made it possible to achieve good responses and MRD negativity even in RRMM (5, 11, 30, 31). In the CANDOR trial (Dara-KD vs. KD), 12.5% of patients with RRMM achieved Flow MRD-negative status, and no patients progressed to disease during the follow-up period. Similarly, 20% of patients with RRMM who received isatuximab-KD achieved Flow MRD-negative results in the IKEMA trial. This suggests that novel drugs and their combinations have a great impact on MRD status and outcomes in patients with RRMM.

In conclusion, these data suggest that KRD therapy has the potential to induce deeper response and lead to good outcomes, even in patients with RRMM. KRD treatment also demonstrated favorable tolerability and safety profiles. Further studies are needed to confirm the long-term efficacy and safety of KRD therapy in RRMM patients.

Acknowledgements

The Authors thank the patients and their physicians for contributing to this study.

Footnotes

  • Authors’ Contributions

    T. Yoroidaka, T. Yamashita, R.M., K.Y., S.Y., and H.T. collected clinical data and blood samples. T.Y. and H.T. performed flow cytometry experiments and analyzed data. H.T. designed the research. T.Y. and H.T. wrote the manuscript. All Authors critically reviewed the manuscript and checked the final version.

  • Conflicts of Interest

    This work was supported by Ono Pharmaceutical Co., Ltd. HT received honoraria from Bristol-Myers Squibb company and Ono Pharmaceutical Co., Ltd. SY received honoraria from Bristol-Myers Squibb company and Ono Pharmaceutical Co., Ltd. The rest of Authors do not have any relationships to disclose.

  • Received October 21, 2022.
  • Revision received November 8, 2022.
  • Accepted November 21, 2022.
  • Copyright © 2023 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Munshi NC,
    2. Avet-Loiseau H,
    3. Anderson KC,
    4. Neri P,
    5. Paiva B,
    6. Samur M,
    7. Dimopoulos M,
    8. Kulakova M,
    9. Lam A,
    10. Hashim M,
    11. He J,
    12. Heeg B,
    13. Ukropec J,
    14. Vermeulen J,
    15. Cote S and
    16. Bahlis N
    : A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma. Blood Adv 4(23): 5988-5999, 2020. PMID: 33284948. DOI: 10.1182/bloodadvances.2020002827
    OpenUrlCrossRefPubMed
  2. ↵
    1. Facon T,
    2. Kumar SK,
    3. Plesner T,
    4. Orlowski RZ,
    5. Moreau P,
    6. Bahlis N,
    7. Basu S,
    8. Nahi H,
    9. Hulin C,
    10. Quach H,
    11. Goldschmidt H,
    12. O’Dwyer M,
    13. Perrot A,
    14. Venner CP,
    15. Weisel K,
    16. Mace JR,
    17. Raje N,
    18. Tiab M,
    19. Macro M,
    20. Frenzel L,
    21. Leleu X,
    22. Ahmadi T,
    23. Wang J,
    24. Van Rampelbergh R,
    25. Uhlar CM,
    26. Tromp B,
    27. Delioukina M,
    28. Vermeulen J and
    29. Usmani SZ
    : 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 22(11): 1582-1596, 2021. PMID: 34655533. DOI: 10.1016/S1470-2045(21)00466-6
    OpenUrlCrossRefPubMed
    1. Mateos MV,
    2. Cavo M,
    3. Blade J,
    4. Dimopoulos MA,
    5. Suzuki K,
    6. Jakubowiak A,
    7. Knop S,
    8. Doyen C,
    9. Lucio P,
    10. Nagy Z,
    11. Pour L,
    12. Cook M,
    13. Grosicki S,
    14. Crepaldi A,
    15. Liberati AM,
    16. Campbell P,
    17. Shelekhova T,
    18. Yoon SS,
    19. Iosava G,
    20. Fujisaki T,
    21. Garg M,
    22. Krevvata M,
    23. Chen Y,
    24. Wang J,
    25. Kudva A,
    26. Ukropec J,
    27. Wroblewski S,
    28. Qi M,
    29. Kobos R and
    30. San-Miguel J
    : Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial. Lancet 395(10218): 132-141, 2020. PMID: 31836199. DOI: 10.1016/S0140-6736(19)32956-3
    OpenUrlCrossRefPubMed
  3. ↵
    1. Kumar SK,
    2. Jacobus SJ,
    3. Cohen AD,
    4. Weiss M,
    5. Callander N,
    6. Singh AK,
    7. Parker TL,
    8. Menter A,
    9. Yang X,
    10. Parsons B,
    11. Kumar P,
    12. Kapoor P,
    13. Rosenberg A,
    14. Zonder JA,
    15. Faber E Jr.,
    16. Lonial S,
    17. Anderson KC,
    18. Richardson PG,
    19. Orlowski RZ,
    20. Wagner LI and
    21. Rajkumar SV
    : 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 21(10): 1317-1330, 2020. PMID: 32866432. DOI: 10.1016/S1470-2045(20)30452-6
    OpenUrlCrossRefPubMed
  4. ↵
    1. Dimopoulos M,
    2. Quach H,
    3. Mateos MV,
    4. Landgren O,
    5. Leleu X,
    6. Siegel D,
    7. Weisel K,
    8. Yang H,
    9. Klippel Z,
    10. Zahlten-Kumeli A and
    11. Usmani SZ
    : Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. Lancet 396(10245): 186-197, 2020. PMID: 32682484. DOI: 10.1016/S0140-6736(20)30734-0
    OpenUrlCrossRefPubMed
    1. Lahuerta JJ,
    2. Paiva B,
    3. Vidriales MB,
    4. Cordón L,
    5. Cedena MT,
    6. Puig N,
    7. Martinez-Lopez J,
    8. Rosiñol L,
    9. Gutierrez NC,
    10. Martín-Ramos ML,
    11. Oriol A,
    12. Teruel AI,
    13. Echeveste MA,
    14. de Paz R,
    15. de Arriba F,
    16. Hernandez MT,
    17. Palomera L,
    18. Martinez R,
    19. Martin A,
    20. Alegre A,
    21. De la Rubia J,
    22. Orfao A,
    23. Mateos MV,
    24. Blade J,
    25. San-Miguel JF and GEM (Grupo Español de Mieloma)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group
    : Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol 35(25): 2900-2910, 2017. PMID: 28498784. DOI: 10.1200/JCO.2016.69.2517
    OpenUrlCrossRefPubMed
  5. ↵
    1. Landgren O,
    2. Devlin S,
    3. Boulad M and
    4. Mailankody S
    : Role of MRD status in relation to clinical outcomes in newly diagnosed multiple myeloma patients: a meta-analysis. Bone Marrow Transplant 51(12): 1565-1568, 2016. PMID: 27595280. DOI: 10.1038/bmt.2016.222
    OpenUrlCrossRefPubMed
  6. ↵
    1. Dimopoulos MA,
    2. Moreau P,
    3. Palumbo A,
    4. Joshua D,
    5. Pour L,
    6. Hájek R,
    7. Facon T,
    8. Ludwig H,
    9. Oriol A,
    10. Goldschmidt H,
    11. Rosiñol L,
    12. Straub J,
    13. Suvorov A,
    14. Araujo C,
    15. Rimashevskaya E,
    16. Pika T,
    17. Gaidano G,
    18. Weisel K,
    19. Goranova-Marinova V,
    20. Schwarer A,
    21. Minuk L,
    22. Masszi T,
    23. Karamanesht I,
    24. Offidani M,
    25. Hungria V,
    26. Spencer A,
    27. Orlowski RZ,
    28. Gillenwater HH,
    29. Mohamed N,
    30. Feng S,
    31. Chng WJ and ENDEAVOR Investigators
    : Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol 17(1): 27-38, 2016. PMID: 26671818. DOI: 10.1016/S1470-2045(15)00464-7
    OpenUrlCrossRefPubMed
  7. ↵
    1. Mehta P,
    2. Yadav N,
    3. Bhaarat M,
    4. Mirgh S,
    5. Khushoo V,
    6. Thekuddan S,
    7. Agrawal N,
    8. Ahmed R and
    9. Bhurani D
    : Carfilzomib-Pomalidomide-Dexamethasone (KPD) in relapsed/refractory multiple myeloma patients - an institutional retrospective analysis. Blood 134(Suppl_1): 5572-5572, 2019. DOI: 10.1182/blood-2019-131674
    OpenUrlCrossRef
  8. ↵
    1. Stewart AK,
    2. Rajkumar SV,
    3. Dimopoulos MA,
    4. Masszi T,
    5. Špička I,
    6. Oriol A,
    7. Hájek R,
    8. Rosiñol L,
    9. Siegel DS,
    10. Mihaylov GG,
    11. Goranova-Marinova V,
    12. Rajnics P,
    13. Suvorov A,
    14. Niesvizky R,
    15. Jakubowiak AJ,
    16. San-Miguel JF,
    17. Ludwig H,
    18. Wang M,
    19. Maisnar V,
    20. Minarik J,
    21. Bensinger WI,
    22. Mateos MV,
    23. Ben-Yehuda D,
    24. Kukreti V,
    25. Zojwalla N,
    26. Tonda ME,
    27. Yang X,
    28. Xing B,
    29. Moreau P,
    30. Palumbo A and ASPIRE Investigators
    : Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med 372(2): 142-152, 2015. PMID: 25482145. DOI: 10.1056/NEJMoa1411321
    OpenUrlCrossRefPubMed
  9. ↵
    1. Moreau P,
    2. Dimopoulos MA,
    3. Mikhael J,
    4. Yong K,
    5. Capra M,
    6. Facon T,
    7. Hajek R,
    8. Špička I,
    9. Baker R,
    10. Kim K,
    11. Martinez G,
    12. Min CK,
    13. Pour L,
    14. Leleu X,
    15. Oriol A,
    16. Koh Y,
    17. Suzuki K,
    18. Risse ML,
    19. Asset G,
    20. Macé S,
    21. Martin T and IKEMA study group
    : Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): a multicentre, open-label, randomised phase 3 trial. Lancet 397(10292): 2361-2371, 2021. PMID: 34097854. DOI: 10.1016/S0140-6736(21)00592-4
    OpenUrlCrossRefPubMed
  10. ↵
    1. Gay F,
    2. Musto P,
    3. Rota-Scalabrini D,
    4. Bertamini L,
    5. Belotti A,
    6. Galli M,
    7. Offidani M,
    8. Zamagni E,
    9. Ledda A,
    10. Grasso M,
    11. Ballanti S,
    12. Spadano A,
    13. Cea M,
    14. Patriarca F,
    15. D’Agostino M,
    16. Capra A,
    17. Giuliani N,
    18. de Fabritiis P,
    19. Aquino S,
    20. Palmas A,
    21. Gamberi B,
    22. Zambello R,
    23. Petrucci MT,
    24. Corradini P,
    25. Cavo M and
    26. Boccadoro M
    : Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): a randomised, open-label, phase 2 trial. Lancet Oncol 22(12): 1705-1720, 2021. PMID: 34774221. DOI: 10.1016/S1470-2045(21)00535-0
    OpenUrlCrossRefPubMed
    1. Korde N,
    2. Roschewski M,
    3. Zingone A,
    4. Kwok M,
    5. Manasanch EE,
    6. Bhutani M,
    7. Tageja N,
    8. Kazandjian D,
    9. Mailankody S,
    10. Wu P,
    11. Morrison C,
    12. Costello R,
    13. Zhang Y,
    14. Burton D,
    15. Mulquin M,
    16. Zuchlinski D,
    17. Lamping L,
    18. Carpenter A,
    19. Wall Y,
    20. Carter G,
    21. Cunningham SC,
    22. Gounden V,
    23. Sissung TM,
    24. Peer C,
    25. Maric I,
    26. Calvo KR,
    27. Braylan R,
    28. Yuan C,
    29. Stetler-Stevenson M,
    30. Arthur DC,
    31. Kong KA,
    32. Weng L,
    33. Faham M,
    34. Lindenberg L,
    35. Kurdziel K,
    36. Choyke P,
    37. Steinberg SM,
    38. Figg W and
    39. Landgren O
    : Treatment with carfilzomib-lenalidomide-dexamethasone with lenalidomide extension in patients with smoldering or newly diagnosed multiple myeloma. JAMA Oncol 1(6): 746-754, 2015. PMID: 26181891. DOI: 10.1001/jamaoncol.2015.2010
    OpenUrlCrossRefPubMed
  11. ↵
    1. Kazandjian D,
    2. Korde N,
    3. Mailankody S,
    4. Hill E,
    5. Figg WD,
    6. Roschewski M and
    7. Landgren O
    : Remission and progression-free survival in patients with newly diagnosed multiple myeloma treated with carfilzomib, lenalidomide, and dexamethasone: five-year follow-up of a phase 2 clinical trial. JAMA Oncol 4(12): 1781-1783, 2018. PMID: 30477009. DOI: 10.1001/jamaoncol.2018.5457
    OpenUrlCrossRefPubMed
  12. ↵
    1. Rajkumar SV,
    2. Dimopoulos MA,
    3. Palumbo A,
    4. Blade J,
    5. Merlini G,
    6. Mateos MV,
    7. Kumar S,
    8. Hillengass J,
    9. Kastritis E,
    10. Richardson P,
    11. Landgren O,
    12. Paiva B,
    13. Dispenzieri A,
    14. Weiss B,
    15. LeLeu X,
    16. Zweegman S,
    17. Lonial S,
    18. Rosinol L,
    19. Zamagni E,
    20. Jagannath S,
    21. Sezer O,
    22. Kristinsson SY,
    23. Caers J,
    24. Usmani SZ,
    25. Lahuerta JJ,
    26. Johnsen HE,
    27. Beksac M,
    28. Cavo M,
    29. Goldschmidt H,
    30. Terpos E,
    31. Kyle RA,
    32. Anderson KC,
    33. Durie BG and
    34. Miguel JF
    : International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol 15(12): e538-e548, 2014. PMID: 25439696. DOI: 10.1016/S1470-2045(14)70442-5
    OpenUrlCrossRefPubMed
  13. ↵
    1. Kumar S,
    2. Paiva B,
    3. Anderson KC,
    4. Durie B,
    5. Landgren O,
    6. Moreau P,
    7. Munshi N,
    8. Lonial S,
    9. Bladé J,
    10. Mateos MV,
    11. Dimopoulos M,
    12. Kastritis E,
    13. Boccadoro M,
    14. Orlowski R,
    15. Goldschmidt H,
    16. Spencer A,
    17. Hou J,
    18. Chng WJ,
    19. Usmani SZ,
    20. Zamagni E,
    21. Shimizu K,
    22. Jagannath S,
    23. Johnsen HE,
    24. Terpos E,
    25. Reiman A,
    26. Kyle RA,
    27. Sonneveld P,
    28. Richardson PG,
    29. McCarthy P,
    30. Ludwig H,
    31. Chen W,
    32. Cavo M,
    33. Harousseau JL,
    34. Lentzsch S,
    35. Hillengass J,
    36. Palumbo A,
    37. Orfao A,
    38. Rajkumar SV,
    39. Miguel JS and
    40. Avet-Loiseau H
    : International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol 17(8): e328-e346, 2016. PMID: 27511158. DOI: 10.1016/S1470-2045(16)30206-6
    OpenUrlCrossRefPubMed
  14. ↵
    1. US Department of Health and Human Services NIoH, National Cancer Institute
    . Common Terminology Criteria for Adverse Events (CTCAE). Version 5.0, 2017. Available at: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf [Last accessed on November 21, 2022]
  15. ↵
    1. Flores-Montero J,
    2. Sanoja-Flores L,
    3. Paiva B,
    4. Puig N,
    5. García-Sánchez O,
    6. Böttcher S,
    7. van der Velden VHJ,
    8. Pérez-Morán JJ,
    9. Vidriales MB,
    10. García-Sanz R,
    11. Jimenez C,
    12. González M,
    13. Martínez-López J,
    14. Corral-Mateos A,
    15. Grigore GE,
    16. Fluxá R,
    17. Pontes R,
    18. Caetano J,
    19. Sedek L,
    20. Del Cañizo MC,
    21. Bladé J,
    22. Lahuerta JJ,
    23. Aguilar C,
    24. Bárez A,
    25. García-Mateo A,
    26. Labrador J,
    27. Leoz P,
    28. Aguilera-Sanz C,
    29. San-Miguel J,
    30. Mateos MV,
    31. Durie B,
    32. van Dongen JJM and
    33. Orfao A
    : Next Generation Flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia 31(10): 2094-2103, 2017. PMID: 28104919. DOI: 10.1038/leu.2017.29
    OpenUrlCrossRefPubMed
  16. ↵
    1. Jakubowiak AJ,
    2. Dytfeld D,
    3. Griffith KA,
    4. Lebovic D,
    5. Vesole DH,
    6. Jagannath S,
    7. Al-Zoubi A,
    8. Anderson T,
    9. Nordgren B,
    10. Detweiler-Short K,
    11. Stockerl-Goldstein K,
    12. Ahmed A,
    13. Jobkar T,
    14. Durecki DE,
    15. McDonnell K,
    16. Mietzel M,
    17. Couriel D,
    18. Kaminski M and
    19. Vij R
    : A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma. Blood 120(9): 1801-1809, 2012. PMID: 22665938. DOI: 10.1182/blood-2012-04-422683
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Jasielec JK,
    2. Kubicki T,
    3. Raje N,
    4. Vij R,
    5. Reece D,
    6. Berdeja J,
    7. Derman BA,
    8. Rosenbaum CA,
    9. Richardson P,
    10. Gurbuxani S,
    11. Major S,
    12. Wolfe B,
    13. Stefka AT,
    14. Stephens L,
    15. Tinari KM,
    16. Hycner T,
    17. Rojek AE,
    18. Dytfeld D,
    19. Griffith KA,
    20. Zimmerman TM and
    21. Jakubowiak AJ
    : Carfilzomib, lenalidomide, and dexamethasone plus transplant in newly diagnosed multiple myeloma. Blood 136(22): 2513-2523, 2020. PMID: 32735641. DOI: 10.1182/blood.2020007522
    OpenUrlCrossRefPubMed
  18. ↵
    1. Gay F,
    2. Cerrato C,
    3. Petrucci M,
    4. Zambello R,
    5. Gamberi B,
    6. Ballanti S,
    7. Omedè P,
    8. Palmieri S,
    9. Troia R,
    10. Spada S,
    11. Gozzetti A,
    12. Caravita T,
    13. Spadano A,
    14. Palumbo A,
    15. Montefusco V,
    16. Musto P,
    17. Cavo M and
    18. Boccadoro M
    : Efficacy of carfilzomib lenalidomide dexamethasone (KRd) with or without transplantation in newly diagnosed myeloma according to risk status: Results from the FORTE trial. Journal of Clinical Oncology 37(15_suppl): 8002-8002, 2020. DOI: 10.1200/JCO.2019.37.15_suppl.8002
    OpenUrlCrossRef
  19. ↵
    1. Dimopoulos MA,
    2. San-Miguel J,
    3. Belch A,
    4. White D,
    5. Benboubker L,
    6. Cook G,
    7. Leiba M,
    8. Morton J,
    9. Ho PJ,
    10. Kim K,
    11. Takezako N,
    12. Moreau P,
    13. Kaufman JL,
    14. Sutherland HJ,
    15. Lalancette M,
    16. Magen H,
    17. Iida S,
    18. Kim JS,
    19. Prince HM,
    20. Cochrane T,
    21. Oriol A,
    22. Bahlis NJ,
    23. Chari A,
    24. O’Rourke L,
    25. Wu K,
    26. Schecter JM,
    27. Casneuf T,
    28. Chiu C,
    29. Soong D,
    30. Sasser AK,
    31. Khokhar NZ,
    32. Avet-Loiseau H and
    33. Usmani SZ
    : Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POLLUX. Haematologica 103(12): 2088-2096, 2018. PMID: 30237262. DOI: 10.3324/haematol.2018.194282
    OpenUrlAbstract/FREE Full Text
    1. Paiva B,
    2. Chandia M,
    3. Puig N,
    4. Vidriales MB,
    5. Perez JJ,
    6. Lopez-Corral L,
    7. Ocio EM,
    8. Garcia-Sanz R,
    9. Gutierrez NC,
    10. Jimenez-Ubieto A,
    11. Lahuerta JJ,
    12. Mateos MV and
    13. San Miguel JF
    : The prognostic value of multiparameter flow cytometry minimal residual disease assessment in relapsed multiple myeloma. Haematologica 100(2): e53-e55, 2015. PMID: 25381128. DOI: 10.3324/haematol.2014.115162
    OpenUrlFREE Full Text
  20. ↵
    1. Spencer A,
    2. Lentzsch S,
    3. Weisel K,
    4. Avet-Loiseau H,
    5. Mark TM,
    6. Spicka I,
    7. Masszi T,
    8. Lauri B,
    9. Levin MD,
    10. Bosi A,
    11. Hungria V,
    12. Cavo M,
    13. Lee JJ,
    14. Nooka AK,
    15. Quach H,
    16. Lee C,
    17. Barreto W,
    18. Corradini P,
    19. Min CK,
    20. Scott EC,
    21. Chanan-Khan AA,
    22. Horvath N,
    23. Capra M,
    24. Beksac M,
    25. Ovilla R,
    26. Jo JC,
    27. Shin HJ,
    28. Sonneveld P,
    29. Soong D,
    30. Casneuf T,
    31. Chiu C,
    32. Amin H,
    33. Qi M,
    34. Thiyagarajah P,
    35. Sasser AK,
    36. Schecter JM and
    37. Mateos MV
    : Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of CASTOR. Haematologica 103(12): 2079-2087, 2018. PMID: 30237264. DOI: 10.3324/haematol.2018.194118
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Paiva B,
    2. Puig N,
    3. Cedena MT,
    4. Rosiñol L,
    5. Cordón L,
    6. Vidriales MB,
    7. Burgos L,
    8. Flores-Montero J,
    9. Sanoja-Flores L,
    10. Lopez-Anglada L,
    11. Maldonado R,
    12. de la Cruz J,
    13. Gutierrez NC,
    14. Calasanz MJ,
    15. Martin-Ramos ML,
    16. Garcia-Sanz R,
    17. Martinez-Lopez J,
    18. Oriol A,
    19. Blanchard MJ,
    20. Rios R,
    21. Martin J,
    22. Martinez-Martinez R,
    23. Sureda A,
    24. Hernandez MT,
    25. de la Rubia J,
    26. Krsnik I,
    27. Moraleda JM,
    28. Palomera L,
    29. Bargay J,
    30. Van Dongen JJM,
    31. Orfao A,
    32. Mateos MV,
    33. Blade J,
    34. San-Miguel JF,
    35. Lahuerta JJ and GEM (Grupo Español de Mieloma)/PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group
    : Measurable residual disease by next-generation flow cytometry in multiple myeloma. J Clin Oncol 38(8): 784-792, 2020. PMID: 31770060. DOI: 10.1200/JCO.19.01231
    OpenUrlCrossRefPubMed
  22. ↵
    1. Perrot A,
    2. Lauwers-Cances V,
    3. Corre J,
    4. Robillard N,
    5. Hulin C,
    6. Chretien ML,
    7. Dejoie T,
    8. Maheo S,
    9. Stoppa AM,
    10. Pegourie B,
    11. Karlin L,
    12. Garderet L,
    13. Arnulf B,
    14. Doyen C,
    15. Meuleman N,
    16. Royer B,
    17. Eveillard JR,
    18. Benboubker L,
    19. Dib M,
    20. Decaux O,
    21. Jaccard A,
    22. Belhadj K,
    23. Brechignac S,
    24. Kolb B,
    25. Fohrer C,
    26. Mohty M,
    27. Macro M,
    28. Richardson PG,
    29. Carlton V,
    30. Moorhead M,
    31. Willis T,
    32. Faham M,
    33. Anderson KC,
    34. Harousseau JL,
    35. Leleu X,
    36. Facon T,
    37. Moreau P,
    38. Attal M,
    39. Avet-Loiseau H and
    40. Munshi N
    : Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma. Blood 132(23): 2456-2464, 2018. PMID: 30249784. DOI: 10.1182/blood-2018-06-858613
    OpenUrlAbstract/FREE Full Text
    1. Goicoechea I,
    2. Puig N,
    3. Cedena MT,
    4. Burgos L,
    5. Cordón L,
    6. Vidriales MB,
    7. Flores-Montero J,
    8. Gutierrez NC,
    9. Calasanz MJ,
    10. Ramos MM,
    11. Lara-Astiaso D,
    12. Vilas-Zornoza A,
    13. Alignani D,
    14. Rodriguez I,
    15. Sarvide S,
    16. Alameda D,
    17. Garcés JJ,
    18. Rodriguez S,
    19. Fresquet V,
    20. Celay J,
    21. Garcia-Sanz R,
    22. Martinez-Lopez J,
    23. Oriol A,
    24. Rios R,
    25. Martin-Sanchez J,
    26. Martinez-Martinez R,
    27. Sarra J,
    28. Hernandez MT,
    29. de la Rubia J,
    30. Krsnik I,
    31. Moraleda JM,
    32. Palomera L,
    33. Bargay J,
    34. Martinez-Climent JA,
    35. Orfao A,
    36. Rosiñol L,
    37. Mateos MV,
    38. Lahuerta JJ,
    39. Blade J,
    40. San Miguel J and
    41. Paiva B
    : Deep MRD profiling defines outcome and unveils different modes of treatment resistance in standard- and high-risk myeloma. Blood 137(1): 49-60, 2021. PMID: 32693406. DOI: 10.1182/blood.2020006731
    OpenUrlCrossRefPubMed
  23. ↵
    1. San-Miguel J,
    2. Avet-Loiseau H,
    3. Paiva B,
    4. Kumar S,
    5. Dimopoulos MA,
    6. Facon T,
    7. Mateos MV,
    8. Touzeau C,
    9. Jakubowiak A,
    10. Usmani SZ,
    11. Cook G,
    12. Cavo M,
    13. Quach H,
    14. Ukropec J,
    15. Ramaswami P,
    16. Pei H,
    17. Qi M,
    18. Sun S,
    19. Wang J,
    20. Krevvata M,
    21. DeAngelis N,
    22. Heuck C,
    23. Van Rampelbergh R,
    24. Kudva A,
    25. Kobos R,
    26. Qi M and
    27. Bahlis NJ
    : Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE. Blood 139(4): 492-501, 2022. PMID: 34269818. DOI: 10.1182/blood.2020010439
    OpenUrlCrossRefPubMed
  24. ↵
    1. Avet-Loiseau H,
    2. San-Miguel J,
    3. Casneuf T,
    4. Iida S,
    5. Lonial S,
    6. Usmani SZ,
    7. Spencer A,
    8. Moreau P,
    9. Plesner T,
    10. Weisel K,
    11. Ukropec J,
    12. Chiu C,
    13. Trivedi S,
    14. Amin H,
    15. Krevvata M,
    16. Ramaswami P,
    17. Qin X,
    18. Qi M,
    19. Sun S,
    20. Qi M,
    21. Kobos R and
    22. Bahlis NJ
    : Evaluation of sustained minimal residual disease negativity with daratumumab-combination regimens in relapsed and/or refractory multiple myeloma: analysis of POLLUX and CASTOR. J Clin Oncol 39(10): 1139-1149, 2021. PMID: 33513030. DOI: 10.1200/JCO.20.01814
    OpenUrlCrossRefPubMed
  25. ↵
    1. Siegel DS,
    2. Schiller GJ,
    3. Samaras C,
    4. Sebag M,
    5. Berdeja J,
    6. Ganguly S,
    7. Matous J,
    8. Song K,
    9. Seet CS,
    10. Talamo G,
    11. Acosta-Rivera M,
    12. Bar M,
    13. Quick D,
    14. Anz B,
    15. Fonseca G,
    16. Reece D,
    17. Pierceall WE,
    18. Chung W,
    19. Zafar F,
    20. Agarwal A and
    21. Bahlis NJ
    : Pomalidomide, dexamethasone, and daratumumab in relapsed refractory multiple myeloma after lenalidomide treatment. Leukemia 34(12): 3286-3297, 2020. PMID: 32376855. DOI: 10.1038/s41375-020-0813-1
    OpenUrlCrossRefPubMed
  26. ↵
    1. Attal M,
    2. Richardson PG,
    3. Rajkumar SV,
    4. San-Miguel J,
    5. Beksac M,
    6. Spicka I,
    7. Leleu X,
    8. Schjesvold F,
    9. Moreau P,
    10. Dimopoulos MA,
    11. Huang JS,
    12. Minarik J,
    13. Cavo M,
    14. Prince HM,
    15. Macé S,
    16. Corzo KP,
    17. Campana F,
    18. Le-Guennec S,
    19. Dubin F,
    20. Anderson KC and ICARIA-MM study group
    : Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet 394(10214): 2096-2107, 2019. PMID: 31735560. DOI: 10.1016/S0140-6736(19)32556-5
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 43 (1)
Anticancer Research
Vol. 43, Issue 1
January 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Measurable Residual Disease Assessment Using Next-Generation Flow in Patients With Relapsed and Refractory Multiple Myeloma Treated With a Combination of Carfilzomib, Lenalidomide, and Dexamethasone
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
14 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Measurable Residual Disease Assessment Using Next-Generation Flow in Patients With Relapsed and Refractory Multiple Myeloma Treated With a Combination of Carfilzomib, Lenalidomide, and Dexamethasone
TAKESHI YOROIDAKA, TAKESHI YAMASHITA, RYOICHI MURATA, KYOKO YOSHIHARA, SATOSHI YOSHIHARA, MIKIO UEDA, SHINJI NAKAO, KOSEI MATSUE, HIROYUKI TAKAMATSU
Anticancer Research Jan 2023, 43 (1) 157-165; DOI: 10.21873/anticanres.16145

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Measurable Residual Disease Assessment Using Next-Generation Flow in Patients With Relapsed and Refractory Multiple Myeloma Treated With a Combination of Carfilzomib, Lenalidomide, and Dexamethasone
TAKESHI YOROIDAKA, TAKESHI YAMASHITA, RYOICHI MURATA, KYOKO YOSHIHARA, SATOSHI YOSHIHARA, MIKIO UEDA, SHINJI NAKAO, KOSEI MATSUE, HIROYUKI TAKAMATSU
Anticancer Research Jan 2023, 43 (1) 157-165; DOI: 10.21873/anticanres.16145
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Pelvic Recurrence After Curative Resection for Rectal Adenocarcinoma: Impact of Surgery on Survival
  • Glasgow Prognostic Score Predicts Survival and Recurrence Pattern in Patients With Hepatocellular Carcinoma After Hepatectomy
  • Small Bowel Lipomatosis: An Unusual Radiological Finding in Patients With Renal Cell Cancer on Pazopanib
Show more Clinical Studies

Similar Articles

Keywords

  • Multiple myeloma
  • next-generation flow
  • measurable residual disease
  • relapsed and refractory
  • carfilzomib
  • lenalidomide
  • dexamethasone (KRD)
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire