Next Article in Journal
Targeted Next-Generation Sequencing of 117 Routine Clinical Samples Provides Further Insights into the Molecular Landscape of Uveal Melanoma
Previous Article in Journal
Cancer-Selective Treatment of Cancerous and Non-Cancerous Human Cervical Cell Models by a Non-Thermally Operated Electrosurgical Argon Plasma Device
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Carfilzomib Based Treatment Strategies in the Management of Relapsed/Refractory Multiple Myeloma with Extramedullary Disease

1
Department of Internal Medicine II, University Hospital of Würzburg, D-97080 Würzburg, Germany
2
Mildred Scheel Early Career Center, University Hospital of Würzburg, D-97080 Würzburg, Germany
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(4), 1035; https://doi.org/10.3390/cancers12041035
Submission received: 13 March 2020 / Revised: 18 April 2020 / Accepted: 20 April 2020 / Published: 23 April 2020

Abstract

:
Published experience with carfilzomib in patients with relapsed/refractory multiple myeloma (RRMM) and extramedullary disease (EMD) is still limited. The current study aimed to assess the efficacy and safety of carfilzomib containing therapy regimens in EMD. We retrospectively analyzed 45 patients with extramedullary RRMM treated with carfilzomib from June 2013 to September 2019. The median age at the start of carfilzomib was 64 (range 40–80) years. Twenty (44%) and 25 (56%) patients had paraosseous manifestation and EMD without adjacency to bone, respectively. The serological overall response rate (ORR) was 59%. Extramedullary response was evaluable in 33 patients, nine (27%) of them achieved partial remission (PR) (ORR = 27%). In 15 (33%) patients, we observed no extramedullary response despite serological response. The median progression-free survival (PFS) and overall survival (OS) were five (95% CI, 3.5–6.5) and ten (95% CI, 7.5–12.5) months, respectively. EMD without adjacency to bone was associated with a significantly inferior PFS (p = 0.004) and OS (p = 0.04) compared to paraosseous lesions. Carfilzomib based treatment strategies showed some efficacy in heavily pretreated patients with extramedullary RRMM but could not overcome the negative prognostic value of EMD. Due to the discrepancy between serological and extramedullary response, evaluation of extramedullary response using imaging is mandatory in these patients.

1. Introduction

In multiple myeloma (MM), tumor growth is usually strongly dependent on the bone marrow microenvironment [1]. However, extramedullary disease (EMD) has been reported to occur in 6%–37% of MM patients, and the frequency increases during the course of the disease. The incidence can be as high as 70% in autopsy studies [2,3,4]. In addition, the pathogenesis of EMD remains largely unclear. Upregulation of angiogenesis and adhesion related genes might be a mechanism in EMD development [5]. C-X-C-motif chemokine receptor 4 (CXCR4) expression and hypoxia have also been reported to play a potential role in EMD growth [6,7]. Moreover, high-risk cytogenetics are significantly enriched in MM with EMD [8,9,10,11,12], and studies consistently showed EMD to be associated with poor outcome [12,13,14,15,16]. To date, there is no established approach for treatment of relapsed/refractory multiple myeloma (RRMM) with EMD.
Earlier, we and others reported on intensive multi-agent therapies, such as VDT-PACE (bortezomib, dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, and etoposide) and Dexa-BEAM (dexamethasone, carmustine, etoposide, cytarabine, and melphalan), which have shown some efficacy [17,18]. Furthermore, allogeneic stem cell transplantation has been reported to be an option for EMD [19]. In the phase 2 SIRIUS trial, the overall response rate (ORR) of daratumumab monotherapy was only 21.4% (3/14) in patients with EMD vs. 30.4% (28/92) of patients without EMD [20]. Notably, none of the above-mentioned strategies led to long-term disease-free survival. Therefore, identifying novel treatment strategies for patients suffering from EMD still represents an urgent medical need. Unfortunately, patients with extramedullary RRMM have only limited access to treatments within clinical trials due to the common exclusion criteria, such as non-secretory disease, central nervous system (CNS) involvement, and poor performance status.
Carfilzomib is a new generation proteasome inhibitor (PI), which in contrast to bortezomib, irreversibly binds to the 20S subunit of the proteasome and has been approved by the US Food and Drug Administration (FDA) for the treatment of RRMM [21]. In the phase 3 ENDEAVOR trial, superior efficacy of carfilzomib compared to bortezomib was observed in patients with RRMM [22], and patients with high-risk cytogenetics benefited from the replacement of bortezomib with carfilzomib [23]. Moreover, the combination of carfilzomib with immunomodulatory drugs (IMiD) lenalidomide and pomalidomide or the monoclonal antibody daratumumab is highly effective in RRMM [24,25,26]. However, published experience with carfilzomib in patients with extramedullary RRMM is still limited. The current study aimed to analyze carfilzomib containing therapy regimens in EMD.

2. Methods

2.1. Patients

We performed a single-center retrospective analysis. Utilizing our electronic database, we identified patients with extramedullary RRMM treated with carfilzomib-based regimens from June 2013 to September 2019 at our institution. RRMM was defined according to the current consensus recommendations [27]. EMD included paraosseous lesions originating from bone and extramedullary plasmocytomas without direct bone contact. The diagnosis of EMD was based on the histology of tumor bulk or, if biopsy was not possible, on imaging using computed tomography (CT), diffusion weighted magnetic resonance imaging (MRI), or positron emission tomography (PET). We did not include plasma cell leukemia in this analysis. Patients with at least one of the following aberrations were considered as high-risk cytogenetics: del(17p), t(4;14), t(14;16), and t(14;20) [28,29,30]. We retrieved and investigated patients’ demographic characteristics at diagnosis of MM and at the start of carfilzomib-containing treatment, MM related data (time point of diagnosis, subtype, cytogenetics, prior therapy lines, response and survival outcome), EMD related data (localization, adjacency to bones and secretory activity), treatment and adverse events (AEs) during therapy.

2.2. Treatment, Response, and Outcome

Carfilzomib was given on day 1, 2, 8, 9, 15, and 16 as intravenous (IV) short infusion in a 28-day cycle. We started carfilzomib at a dose of 20 mg/m2 on day 1 and 2 of the first cycle, and the dose was increased to 27 mg/m2 on day 8 if tolerated. Doses of carfilzomib were escalated or reduced according to the treating physician’s discretion. In our study, carfilzomib was administered in combination with at least one additional drug, including dexamethasone, IMiD, alkylating agent, and monoclonal antibody, i.e., daratumumab and elotuzumab.
We analyzed ORR, clinical benefit rate (CBR), overall survival (OS) and progression-free survival (PFS) following the current criteria of the International Myeloma Working Group (IMWG) [31,32]. Adverse events (AE) during carfilzomib containing treatment were characterized according to the Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0.

2.3. Statistical Analysis

Using descriptive statistics, we summarized patients’ characteristics as absolute number and percentage, and if not otherwise stated as median and range. The survival analysis was performed with Kaplan–Meier method, and log-rank test was used to compare the survival outcome between subgroups. We used Fisher’s exact test to evaluate the difference in response rate (ORR and CBR) between the subgroups. These analyses were performed with GraphPad Prism 5.0. A p-value less than 0.05 was considered as statistically significant.

3. Results

3.1. Patients’ Characteristics

In total, we identified 45 patients with extramedullary RRMM that were treated with carfilzomib containing regimens. The majority of the patients (n = 33, 73%) were male, and the median age at the start of carfilzomib for EMD was 64 (range 40–80) years. At the initial diagnosis of MM, primary EMD with and without adjacency to bone was already present in 16 (35%) and 1 (2%) patients, respectively. At presentation, 22 patients (49%) had high-risk cytogenetics from bone marrow biopsy. At start of carfilzomib as salvage therapy for secondary EMD, 20 (44%) and 25 (56%) patients suffered from EMD with and without adjacency to bone, respectively. Monoclonal protein in serum was detectable in the majority of the patients (n = 42, 93%). Muscle, skin, and soft tissue manifestation was the most frequent EMD presentation in our cohort (n = 38, 84%). Spinal cord and paravertebral lesions were seen in 25 patients (56%). Twenty (44%), 13 (29%), 11 (24%), and 2 (4%) patients had lymph node manifestation, malignant pleural effusion, parenchymal organ involvement, and gastrointestinal tract lesions, respectively. Lactate dehydrogenase (LDH) was elevated in 22 (49%) patients at start of carfilzomib for EMD.
In our cohort, patients had been treated with a median of four (range 1–9) prior lines of therapy. Forty-three (96%) patients were exposed to bortezomib, and eight (18%) patients had received carfilzomib prior to secondary EMD. Lenalidomide, pomalidomide, and thalidomide had been given in 35 (78%), 22 (49%), and 10 (22%) patients, respectively. Eighteen (40%) and three (7%) were treated with monoclonal antibodies daratumumab and elotuzumab, respectively. Forty-four (98%) and seven (15%) patients underwent autologous and allogeneic stem cell transplant (SCT), respectively. Seventy-three percent of the patients (n = 33) were refractory to the last line of therapy. Patients’ characteristics are summarized in Table 1.

3.2. Treatment and Response to Therapy

Overall, carfilzomib was administered twice weekly, and patients received a median of three (range 1–18) cycles of carfilzomib. The maximal dose of carfilzomib ranged from 15 to 56 mg/m2, and the majority of patients (n = 25, 56%) received a maximal carfilzomib dose of 27 mg/m2 twice weekly. All the patients were treated with dexamethasone 20–40 mg qw. The treatment regimens are summarized in Table 2. Regimens and doses were modified according to the treating physician’s choice.
We first analyzed the best serological response in the 42 patients with M protein and measurable disease. Overall, we observed a serological ORR of 59% with 19% (n = 8) very good partial remission (VGPR) and 40% (n = 17) partial remission (PR). Ten (24%) patients achieved stable disease (SD), resulting in a serological CBR of 83%. Seven (17%) patients experienced serological progression while being treated with carfilzomib. Imaging follow-up data were available in 33 patients to determine the best response of EMD. No patient with isolated skin lesions, which could have been evaluated without imaging, was included in our study. We observed an extramedullary ORR of 27% (n = 9) with 27% (n = 9) PR. Nine (27%) patients had SD at the EMD sites, and, therefore, the CBR in this group was 54%. In addition, extramedullary progression was observed in nine (27%) patients. In six (18%) patients, a mixed response of EMD lesions was observed, with one EMD lesion progressing under therapy but another lesion responding. Data of response to treatment are summarized in Table 2. Similarly, a high proportion (n = 15, 33%) of differential response between serological parameters and EMD lesions was observed. Taken together, our data demonstrated an acceptable ORR in this heavily pretreated group of patients, but, notably, no complete remission (CR) could be achieved in our cohort.
KRd (carfilzomib, lenalidomide, and dexamethasone) was the most frequently administered regimen in our cohort (n = 17). In this subgroup, we observed a serological ORR of 76% with 47% (n = 8) VGPR and 29% (n = 5) PR, and a serological CBR of 88% with 12% (n = 2) SD. Extramedullary response was available in 11 patients of which 45% (n = 5) and 9% (n = 1) patients achieved PR and SD, respectively. Two (18%) patients had PD on treatment, and a mixed response was observed in three (27%) patients.

3.3. Survival Analyses

In our cohort, median PFS and median OS were five (95% CI, 3.5–6.5) and ten (95% CI, 7.5–12.5) months, respectively (Figure 1a,b). In univariate analysis, patients suffering from extramedullary plasmacytomas without direct bone contact had a significantly inferior PFS (p = 0.004) and OS (p = 0.04) compared to those with paraosseous lesions only (Figure 2a,b). Furthermore, elevated LDH indicated a significantly inferior PFS (p = 0.0008), and a trend towards inferior OS (p = 0.06) (Figure 2c,d). Another negative prognostic factor was refractory disease to the last line of therapy with poorer PFS (p = 0.0005) and OS (p < 0.0001) compared to those with a progression from remission (Figure 2e,f). Furthermore, the PFS of patients who were treated with ≥ 4 prior lines of therapy was significantly shorter when compared with those who received less than four prior therapy lines (p = 0.02, Figure 2g). We also observed that patients who received ≥ 4 prior therapy lines had a trend towards an inferior OS compared to those treated with less than four prior lines of therapy (p = 0.08, Figure 2h). However, high-risk cytogenetics showed no significant negative prognostic value in our cohort (not shown in Figure 2). Due to the limited number of patients in our study, we did not perform a multivariate analysis.

3.4. Adverse Events (AEs)

Hematological AEs were the most common AEs during the treatment. We observed anemia, leukopenia, neutropenia, and thrombocytopenia grade ≥ 3 in 25 (56%), 24 (53%), 13 (29%), and 22 (49%) patients, respectively. When necessary, granulocyte colony-stimulating factor (G-CSF) and red cell or platelet concentrates were administered according to the current guidelines [33,34]. Among the non-hematological AEs, pneumonia (n = 6, 13%) was the most common AE grade ≥ 3. Additionally, five (11%) patients developed heart failure grade ≥ 3, possibly related to carfilzomib. In one of the five patients, the treatment had to be withdrawn after one cycle, and in the other four patients, treatment was continued after recovery of cardiac function. In total, two (4%) patients died due to pneumonia during the second and fourth cycles of treatment, respectively. One of both patients had a non-secretory MM with EMD and achieved an extramedullary mixed response after three cycles of Dara-KPd (daratumumab, carfilzomib, pomalidomide, and dexamethasone). In the other patient, PD was observed after one cycle of KRd. AEs are summarized in Table 3.

4. Discussion

The optimal management of extramedullary RRMM remains largely unclear. In the current study, we retrospectively analyzed the role of carfilzomib in the management of EMD. In total, we observed a serological ORR of 59% and a serological CBR of 83%. With regard to extramedullary response, ORR and CBR were 27% and 54%, respectively. To the best of our knowledge, there is no prospective study specifically investigating carfilzomib based treatment in EMD. Carfilzomib based treatments had shown EMD efficacy in case reports [35,36,37]. In a recent retrospective observational study, Muchtar et al. reported on a subgroup of patients with extramedullary RRMM who achieved a lower ORR (40%) and CBR (43.3%) when compared with patients without EMD (ORR = 49%, CBR = 63.5%) [38].
Of note, serological response differed from the extramedullary response in 33% of patients of our cohort, and six patients achieved an extramedullary mixed response. Indeed, intra-tumor genomic heterogeneity with advanced clones located to EMD might explain the lower response rates of these lesions compared to intramedullary disease [39,40]. Our study highlights the need for medical imaging in the follow-up of patients with EMD [41]. In our current study, the median PFS was five (95% CI, 3.5–6.5) months, and the median OS was ten (95% CI, 7.5–12.5) months. Comparably, Muchtar et al. reported a median duration of response of 3.9 months in patients with extramedullary RRMM, who were treated with carfilzomib containing regimens [38]. In a previous study of our institution investigating patients with EMD prior to the carfilzomib era, patients were treated with intensive chemotherapy, such as VDT-PACE, VRD-ICE, RAD, VCDT, and autologous or allogeneic SCT, and we observed a median PFS and OS of two (95% CI, 0.08–3.92) and seven (95% CI, 3.56–10.43) months, respectively [11]. Additionally, Rasche et al. reported a median PFS of four months in patients with extramedullary RRMM treated with DexaBEAM [17]. At our institution, the novel CXCR4-directed endoradiotherapy (ERT) has shown promising efficacy (ORR 75%) but no favorable survival outcome (median PFS 54 days, range 13–175 days and median OS 223 days, range 13–313 days) in extramedullary relapsed MM [42].
Our results suggest that patients with extramedullary plasmacytomas not adjacent to bone had a significantly inferior PFS and OS compared with those with paraosseous EMD. Similarly, Beksac et al. also reported that paraosseous lesions indicated a significantly superior PFS and OS when compared to extramedullary plasmacytomas without bone contact [43]. Furthermore, we observed that patients who were refractory to the last line of therapy represented a negative prognostic factor for PFS and OS. Our findings suggested that carfilzomib-containing treatment had only limited efficacy in this patient group.
Overall, five (11%) patients developed acute heart failure grade ≥ 3, and two (4%) patients died due to pneumonia during treatment. Hematological AEs including anemia, leukopenia, neutropenia, and thrombocytopenia grade ≥ 3 were observed in 25 (56%), 24 (53%), 13 (29%), and 22 (49%) patients, respectively. Due to the regular use of G-SCF and transfusion of blood products, these hematological AEs were well manageable. Generally, the rates of AEs grade ≥ 3 and the mortality during treatment were comparable to previous clinical studies investigating carfilzomib based therapy in RRMM [44,45,46].
There were several limitations to our current study. First, this is a retrospective analysis based on a relatively small number of patients who received heterogeneous carfilzomib containing treatment regimens. Second, the missing values might be a further limitation of our study.

5. Conclusions

In conclusion, compared to previous studies prior to the carfilzomib era, carfilzomib-based treatment strategies have shown similar ORR but improved survival outcome (PFS and OS) in patients with extramedullary RRMM. However, EMD still heralds poor prognosis, especially in patients being refractory to the last therapy line. In addition, due to the high proportion of discrepant response between serological parameters and EMD lesion, evaluation of extramedullary response using imaging should be performed in these patients.

Author Contributions

Conceptualization, X.Z., H.E., K.M.K., and L.R.; Data curation, X.Z.; Formal analysis, X.Z.; Funding acquisition, H.E., K.M.K., and L.R.; Investigation, X.Z., H.E., K.M.K., and L.R.; Methodology, X.Z., H.E., K.M.K., and L.R.; Project administration, X.Z. and L.R.; Resources, X.Z., P.F., H.E., K.M.K., and L.R.; Software, X.Z.; Supervision, X.Z., H.E., and L.R.; Validation, X.Z. and H.E.; Visualization, X.Z.; Writing—original draft, X.Z.; Writing—review and editing, P.F., K.N., K.M., J.M., D.B., S.K., M.J.S., F.K., S.D., H.E., K.M.K., and L.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Leo Rasche was supported by the IZKF Würzburg (Z-3R/2) and by the German Cancer Aid via the MSNZ program. Martin Kortüm was supported by the the German Cancer Aid via the MSNZ program and the Bayerisch–Tschechische Hochschulagentur (BTHA).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bladé, J.; De Larrea, C.F.; Rosiñol, L.; Cibeira, M.T.; Jiménez, R.; Powles, R. Soft-Tissue Plasmacytomas in Multiple Myeloma: Incidence, Mechanisms of Extramedullary Spread, and Treatment Approach. J. Clin. Oncol. 2011, 29, 3805–3812. [Google Scholar] [CrossRef]
  2. Sevcikova, S.; Minarik, J.; Stork, M.; Jelinek, T.; Pour, L.; Hajek, R. Extramedullary disease in multiple myeloma—Controversies and future directions. Blood Rev. 2019, 36, 32–39. [Google Scholar] [CrossRef] [PubMed]
  3. Weinstock, M.; Ghobrial, I.M. Extramedullary multiple myeloma. Leuk. Lymphoma 2012, 54, 1135–1141. [Google Scholar] [CrossRef]
  4. Weinstock, M.; Aljawai, Y.; Morgan, E.A.; Laubach, J.; Gannon, M.; Roccaro, A.; Varga, C.; Mitsiades, C.S.; Paba-Prada, C.; Schlossman, R.; et al. Incidence and clinical features of extramedullary multiple myeloma in patients who underwent stem cell transplantation. Br. J. Haematol. 2015, 169, 851–858. [Google Scholar] [CrossRef] [Green Version]
  5. Hedvat, C.; Comenzo, R.L.; Teruya-Feldstein, J.; Olshen, A.B.; Ely, S.A.; Osman, K.; Zhang, Y.; Kalakonda, N.; Nimer, S.D. Insights into extramedullary tumour cell growth revealed by expression profiling of human plasmacytomas and multiple myeloma. Br. J. Haematol. 2003, 122, 728–744. [Google Scholar] [CrossRef] [PubMed]
  6. Azab, A.K.; Hu, J.; Quang, P.; Azab, F.; Pitsillides, C.; Awwad, R.; Thompson, B.; Maiso, P.; Sun, J.D.; Hart, C.; et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood 2012, 119, 5782–5794. [Google Scholar] [CrossRef] [Green Version]
  7. Roccaro, A.; Mishima, Y.; Sacco, A.; Moschetta, M.; Tai, Y.-T.; Shi, J.; Zhang, Y.; Reagan, M.R.; Huynh, D.; Kawano, Y.; et al. CXCR4 regulates extra-medullary myeloma through epithelial-mesenchymal transition-like transcriptional activation. Cell Rep. 2015, 12, 622–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Bešše, L.; Sedlaříková, L.; Greslikova, H.; Kupska, R.; Almáši, M.; Penka, M.; Jelinek, T.; Pour, L.; Adam, Z.; Kuglik, P.; et al. Cytogenetics in multiple myeloma patients progressing into extramedullary disease. Eur. J. Haematol. 2015, 97, 93–100. [Google Scholar] [CrossRef]
  9. Deng, S.; Xu, Y.; An, G.; Sui, W.; Zou, D.; Zhao, Y.; Qi, J.; Li, F.; Hao, M.; Qiu, L. Features of Extramedullary Disease of Multiple Myeloma: High Frequency of P53 Deletion and Poor Survival: A Retrospective Single-Center Study of 834 Cases. Clin. Lymphoma Myeloma Leuk. 2015, 15, 286–291. [Google Scholar] [CrossRef] [PubMed]
  10. Billecke, L.; Penas, E.M.M.; May, A.M.; Engelhardt, M.; Nagler, A.; Leiba, M.; Schiby, G.; Kröger, N.; Zustin, J.; Marx, A.H.; et al. Cytogenetics of extramedullary manifestations in multiple myeloma. Br. J. Haematol. 2013, 161, 87–94. [Google Scholar] [CrossRef] [Green Version]
  11. Rasche, L.; Bernard, C.; Topp, M.S.; Kapp, M.; Duell, J.; Wesemeier, C.; Haralambieva, E.; Maeder, U.; Einsele, H.; Knop, S. Features of extramedullary myeloma relapse: High proliferation, minimal marrow involvement, adverse cytogenetics: A retrospective single-center study of 24 cases. Ann. Hematol. 2012, 91, 1031–1037. [Google Scholar] [CrossRef] [PubMed]
  12. Usmani, S.Z.; Heuck, C.; Mitchell, A.; Szymonifka, J.; Nair, B.; Hoering, A.; Alsayed, Y.; Waheed, S.; Haider, S.; Restrepo, A.; et al. Extramedullary disease portends poor prognosis in multiple myeloma and is over-represented in high-risk disease even in the era of novel agents. Haematologica 2012, 97, 1761–1767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Varettoni, M.; Corso, A.; Pica, G.; Mangiacavalli, S.; Pascutto, C.; Lazzarino, M. Incidence, presenting features and outcome of extramedullary disease in multiple myeloma: A longitudinal study on 1003 consecutive patients. Ann. Oncol. 2010, 21, 325–330. [Google Scholar] [CrossRef] [PubMed]
  14. Pour, L.; Ševčíková, S.; Greslikova, H.; Kupska, R.; Majkova, P.; Zahradova, L.; Sandecka, V.; Adam, Z.; Krejci, M.; Kuglik, P.; et al. Soft-tissue extramedullary multiple myeloma prognosis is significantly worse in comparison to bone-related extramedullary relapse. Haematol. 2013, 99, 360–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Short, K.D.; Rajkumar, S.V.; Larson, D.; Buadi, F.; Hayman, S.; Dispenzieri, A.; Gertz, M.; Kumar, S.; Mikhael, J.; Roy, V.; et al. Incidence of extramedullary disease in patients with multiple myeloma in the era of novel therapy, and the activity of pomalidomide on extramedullary myeloma. Leukemia 2011, 25, 906–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Mangiacavalli, S.; Pompa, A.; Ferretti, V.; Klersy, C.; Cocito, F.; Varettoni, M.; Cartia, C.S.; Cazzola, M.; Corso, A. The possible role of burden of therapy on the risk of myeloma extramedullary spread. Ann. Hematol. 2016, 96, 73–80. [Google Scholar] [CrossRef]
  17. Rasche, L.; Strifler, S.; Duell, J.; Rosenwald, A.; Buck, A.; Maeder, U.; Einsele, H.; Knop, S. The lymphoma-like polychemotherapy regimen “Dexa-BEAM” in advanced and extramedullary multiple myeloma. Ann. Hematol. 2014, 93, 1207–1214. [Google Scholar] [CrossRef]
  18. Lakshman, A.; Singh, P.P.; Rajkumar, S.V.; Dispenzieri, A.; Lacy, M.Q.; Gertz, M.A.; Buadi, F.K.; Dingli, D.; Hwa, Y.L.; Fonder, A.L.; et al. Efficacy of VDT PACE-like regimens in treatment of relapsed/refractory multiple myeloma. Am. J. Hematol. 2017, 93, 179–186. [Google Scholar] [CrossRef]
  19. Rasche, L.; Röllig, C.; Stuhler, G.; Danhof, S.; Mielke, S.; Grigoleit, G.U.; Dissen, L.; Schemmel, L.; Middeke, J.M.; Rücker, V.; et al. Allogeneic Hematopoietic Cell Transplantation in Multiple Myeloma: Focus on Longitudinal Assessment of Donor Chimerism, Extramedullary Disease, and High-Risk Cytogenetic Features. Boil. Blood Marrow Transplant. 2016, 22, 1988–1996. [Google Scholar] [CrossRef] [Green Version]
  20. Lonial, S.; Weiss, B.M.; Usmani, S.Z.; Singhal, S.; Chari, A.; Bahlis, N.J.; Belch, A.; Krishnan, A.; Vescio, R.A.; Mateos, M.-V.; et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): An open-label, randomised, phase 2 trial. Lancet 2016, 387, 1551–1560. [Google Scholar] [CrossRef]
  21. Groen, K.; Van De Donk, N.W.C.J.; Stege, C.; Zweegman, S.; Nijhof, I. Carfilzomib for relapsed and refractory multiple myeloma. Cancer Manag. Res. 2019, 11, 2663–2675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Dimopoulos, M.A.; Goldschmidt, H.; Niesvizky, R.; Joshua, D.; Chng, W.-J.; Oriol, A.; Orlowski, R.Z.; Ludwig, H.; Facon, T.; Hajek, R.; et al. Carfilzomib or bortezomib in relapsed or refractory multiple myeloma (ENDEAVOR): An interim overall survival analysis of an open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 1327–1337. [Google Scholar] [CrossRef]
  23. Chng, W.-J.; Goldschmidt, H.; Dimopoulos, M.A.; Moreau, P.; Joshua, D.; Palumbo, A.; Facon, T.; Ludwig, H.; Pour, L.; Niesvizky, R.; et al. Carfilzomib–dexamethasone vs bortezomib–dexamethasone in relapsed or refractory multiple myeloma by cytogenetic risk in the phase 3 study ENDEAVOR. Leukemia 2017, 31, 1368–1374. [Google Scholar] [CrossRef]
  24. Stewart, A.K.; Rajkumar, S.V.; Dimopoulos, M.A.; Masszi, T.; Spicka, I.; Oriol, A.; Hajek, R.; Rosiñol, L.; Siegel, D.S.; Mihaylov, G.G.; et al. Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. N. Engl. J. Med. 2015, 372, 142–152. [Google Scholar] [CrossRef] [PubMed]
  25. Chari, A.; Lopez, J.M.; Mateos, M.-V.; Bladé, J.; Benboubker, L.; Oriol, A.; Arnulf, B.; Rodriguez-Otero, P.; Pineiro, L.; Jakubowiak, A.; et al. Daratumumab plus carfilzomib and dexamethasone in patients with relapsed or refractory multiple myeloma. Blood 2019, 134, 421–431. [Google Scholar] [CrossRef] [Green Version]
  26. Shah, J.J.; Stadtmauer, E.A.; Abonour, R.; Cohen, A.D.; Bensinger, W.I.; Gasparetto, C.; Kaufman, J.L.; Lentzsch, S.; Vogl, D.T.; Gomes, C.L.; et al. Carfilzomib, pomalidomide, and dexamethasone for relapsed or refractory myeloma. Blood 2015, 126, 2284–2290. [Google Scholar] [CrossRef]
  27. 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] [Green Version]
  28. Fonseca, R.; Blood, E.; Rué, M.; Harrington, D.; Oken, M.; Kyle, R.A.; Dewald, G.W.; Van Ness, B.; Van Wier, S.A.; Henderson, K.J.; et al. Clinical and biologic implications of recurrent genomic aberrations in myeloma. Blood 2003, 101, 4569–4575. [Google Scholar] [CrossRef] [Green Version]
  29. Mikhael, J.; Dingli, D.; Roy, V.; Reeder, C.B.; Buadi, F.K.; Hayman, S.R.; Dispenzieri, A.; Fonseca, R.; Sher, T.; Kyle, R.A.; et al. Management of Newly Diagnosed Symptomatic Multiple Myeloma: Updated Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) Consensus Guidelines 2013. Mayo Clin. Proc. 2013, 88, 360–376. [Google Scholar] [CrossRef]
  30. Chng, W.J.; Dispenzieri, A.; Chim, C.-S.; Fonseca, R.; Goldschmidt, H.; Lentzsch, S.; Munshi, N.; Palumbo, A.; San-Miguel, J.F.; Sonneveld, P.; et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia 2013, 28, 269–277. [Google Scholar] [CrossRef]
  31. Durie, B.G.M.; Harousseau, J.-L.; Miguel, J.S.; Blade, 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] [PubMed] [Green Version]
  32. Kumar, S.K.; Paiva, B.D.L.; Anderson, K.C.; Durie, B.; Landgren, O.; Moreau, P.; Munshi, N.C.; Lonial, S.; Bladé, J.; Mateos, M.-V.; et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016, 17, e328–e346. [Google Scholar] [CrossRef]
  33. Smith, T.; Bohlke, K.; Lyman, G.H.; Carson, K.R.; Crawford, J.; Cross, S.J.; Goldberg, J.M.; Khatcheressian, J.L.; Leighl, N.B.; Perkins, C.L.; et al. Recommendations for the Use of WBC Growth Factors: American Society of Clinical Oncology Clinical Practice Guideline Update. J. Clin. Oncol. 2015, 33, 3199–3212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lancet Haematol. Updates on blood transfusion guidelines. Lancet Haematol. 2016, 3, e547. [Google Scholar] [CrossRef]
  35. Mele, G.; Pastore, D. Efficacy of Carfilzomib, Lenalidomide, and Dexamethasone for Extramedullary Intracranial Localization of Multiple Myeloma. Case Rep. Hematol. 2018, 2018, 1–3. [Google Scholar] [CrossRef] [Green Version]
  36. Español, I.; Romera, M.; Gutiérrez-Meca, M.D.; García, M.D.C.; Tejedor, A.; Martínez, A.; Ibáñez, J.; De Arriba, F.; Minguela, A.; Iturbe, T.; et al. Carfilzomib and dexamethasone for extramedullary myeloma with pleuropericardial involvement. Clin. Case Rep. 2017, 5, 1258–1260. [Google Scholar] [CrossRef] [Green Version]
  37. Parnell, K.; Ahmed, M.; Smalligan, R.D.; Nadesan, S. Extramedullary plasmacytoma mimicking colon carcinoma: An unusual presentation and review of the literature. BMJ Case Rep. 2015, 2015. [Google Scholar] [CrossRef]
  38. Muchtar, E.; Gatt, M.E.; Rouvio, O.; Ganzel, C.; Chubar, E.; Suriu, C.; Tadmor, T.; Shevetz, O.; Lavi, N.; Shochat, T.; et al. Efficacy and safety of salvage therapy using Carfilzomib for relapsed or refractory multiple myeloma patients: A multicentre retrospective observational study. Br. J. Haematol. 2015, 172, 89–96. [Google Scholar] [CrossRef]
  39. Rasche, L.; Chavan, S.S.; Stephens, O.W.; Patel, P.H.; Tytarenko, R.; Ashby, C.; Bauer, M.A.; Stein, C.; Deshpande, S.; Wardell, C.; et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat. Commun. 2017, 8, 268. [Google Scholar] [CrossRef]
  40. López-Anglada, L.; Gutiérrez, N.C.; García, J.L.; Mateos, M.V.; Flores, T.; San-Miguel, J.F. P53deletion may drive the clinical evolution and treatment response in multiple myeloma. Eur. J. Haematol. 2010, 84, 359–361. [Google Scholar] [CrossRef] [Green Version]
  41. Hillengass, J.; Usmani, S.; Rajkumar, S.V.; Durie, B.G.M.; Mateos, M.-V.; Lonial, S.; Joao, C.; Anderson, K.C.; García-Sanz, R.; Riva, E.; et al. International myeloma working group consensus recommendations on imaging in monoclonal plasma cell disorders. Lancet Oncol. 2019, 20, e302–e312. [Google Scholar] [CrossRef]
  42. Lapa, C.; Herrmann, K.; Schirbel, A.; Hänscheid, H.; Lückerath, K.; Schottelius, M.; Kircher, M.; Werner, R.A.; Schreder, M.; Samnick, S.; et al. CXCR4-directed endoradiotherapy induces high response rates in extramedullary relapsed Multiple Myeloma. Theranostics 2017, 7, 1589–1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Beksac, M.; Seval, G.C.; Kanellias, N.; Coriu, D.; Rosiñol, L.; Ozet, G.; Goranova-Marinova, V.; Unal, A.; Bila, J.; Ozsan, H.; et al. A real world multicenter retrospective study on extramedullary disease from Balkan Myeloma Study Group and Barcelona University: Analysis of parameters that improve outcome. Haematologica 2019, 105, 201–208. [Google Scholar] [CrossRef]
  44. Dimopoulos, M.A.; Moreau, P.; Iida, S.; Huang, S.-Y.; Takezako, N.; Chng, W.J.; Zahlten-Kumeli, A.; Sersch, M.A.; Li, J.; Huang, M.; et al. Outcomes for Asian patients with multiple myeloma receiving once- or twice-weekly carfilzomib-based therapy: A subgroup analysis of the randomized phase 3 ENDEAVOR and A.R.R.O.W. Trials. Int. J. Hematol. 2019, 110, 466–473. [Google Scholar] [CrossRef] [PubMed]
  45. Moreau, P.; Mateos, M.-V.; Berenson, J.R.; Weisel, K.; Lazzaro, A.; Song, K.; Dimopoulos, M.A.; Huang, M.; Zahlten-Kumeli, A.; Stewart, A.K. Once weekly versus twice weekly carfilzomib dosing in patients with relapsed and refractory multiple myeloma (A.R.R.O.W.): Interim analysis results of a randomised, phase 3 study. Lancet Oncol. 2018, 19, 953–964. [Google Scholar] [CrossRef]
  46. Mateos, M.-V.; Goldschmidt, H.; San-Miguel, J.F.; Mikhael, J.; DeCosta, L.; Zhou, L.; Obreja, M.; Blaedel, J.; Szabo, Z.; Leleu, X. Carfilzomib in relapsed or refractory multiple myeloma patients with early or late relapse following prior therapy: A subgroup analysis of the randomized phase 3 ASPIRE and ENDEAVOR trials. Hematol. Oncol. 2018, 36, 463–470. [Google Scholar] [CrossRef]
Figure 1. Progression-free survival (PFS) (a) (n = 45) and overall survival (OS) (b) (n = 45) of patients.
Figure 1. Progression-free survival (PFS) (a) (n = 45) and overall survival (OS) (b) (n = 45) of patients.
Cancers 12 01035 g001
Figure 2. Progression-free survival (PFS) (a) and overall survival (OS) (b) in patients with extramedullary disease (EMD) adjacent to bone or not (EMD with adjacency to bone, n = 20; EMD without adjacency to bone, n = 25). PFS (c) and OS (d) in patients with normal vs. elevated lactate dehydrogenase (LDH) level (LDH < 250 IU/l, n = 23; LDH ≥ 250 IU/l, n = 22). PFS (e) and OS (f) in patients who were refractory to the last therapy line (n = 33) vs. who were not (n = 12). PFS (g) and OS (h) of patients with ≥ 4 prior therapy lines (n = 25) and < 4 prior therapy lines (n = 20).
Figure 2. Progression-free survival (PFS) (a) and overall survival (OS) (b) in patients with extramedullary disease (EMD) adjacent to bone or not (EMD with adjacency to bone, n = 20; EMD without adjacency to bone, n = 25). PFS (c) and OS (d) in patients with normal vs. elevated lactate dehydrogenase (LDH) level (LDH < 250 IU/l, n = 23; LDH ≥ 250 IU/l, n = 22). PFS (e) and OS (f) in patients who were refractory to the last therapy line (n = 33) vs. who were not (n = 12). PFS (g) and OS (h) of patients with ≥ 4 prior therapy lines (n = 25) and < 4 prior therapy lines (n = 20).
Cancers 12 01035 g002
Table 1. Patients’ characteristics.
Table 1. Patients’ characteristics.
Patients, n45
Gender, n (%)
Male33 (73)
Female12 (27)
Subtype, n (%)
IgG 26 (58)
IgA14 (31)
LC5 (11)
ISS Stage, n (%)
I22 (49)
II6 (13)
III8 (18)
NA9 (20)
Cytogenetics, n (%)
High-risk22 (49)
Standard-risk18 (40)
NA5 (11)
Age at Start of Carfilzomib due to EMD Relapse, Median, Years (Range)64 (40–80)
Bone Marrow Involvement at Start of Carfilzomib due to EMD Relapse
Yes21 (47)
No4 (9)
NA20 (44)
Serological MM Activity at Start of Carfilzomib due to EMD Relapse, n (%)
With Secretory Activity42 (93)
Non-Secretory3 (7)
Serum LDH at Start of Carfilzomib due to EMD Relapse, n (%)
Elevated22 (49)
Normal23 (51)
Prior Lines of Therapy, n (%)
1–215 (33)
3–516 (36)
≥ 614 (31)
Response Status to The Last Therapy Line, n (%)
Refractory to The Last Line of Therapy33 (73)
Progression from Remission12 (27)
Characteristics of EMD at Start of Carfilzomib, n (%)
EMD Adjacent to Bone20 (44)
EMD without Adjacency to Bone25 (56)
Presentation/Localization of EMD, n (%)
Muscle, Skin, and Soft Tissue38 (84)
Spinal Cord and Paravertebral Lesion25 (56)
Lymph Node20 (44)
Pleural Effusion13 (29)
Parenchymal Organ11 (24)
Gastrointestinal Tract2 (4)
Prior Treatment, n (%)
PIs
Bortezomib43 (96)
Carfilzomib8 (18)
IMiDs
Lenalidomide35 (78)
Pomalidomide22 (49)
Thalidomide10 (22)
Monoclonal Antibodies
Daratumumab18 (40)
Elotuzumab3 (7)
SCT
Prior Autologous SCT44 (98)
Prior Allogenic SCT7 (15)
EMD—extramedullary disease; IMiDs—immunomodulatory drugs; ISS—The Multiple Myeloma International Staging System; LC—light chain; LDH—lactate dehydrogenase; MM—multiple myeloma; NA–not available; PIs—proteasome inhibitors; SCT—stem cell transplant.
Table 2. Treatment and response.
Table 2. Treatment and response.
PatRegimenNumber of CyclesMaximal Dose of CarfDosing of IMiD, Alkylating Agents and Monoclonal AntibodiesBest Response
SerologyEMD
1Kd1156 mg/m2N/APRPR
2Kd115 mg/m2N/ASDn.a.
3Kd156 mg/m2N/APRSD
4KBd127 mg/m2Benda 70 mg/m2 qwN/APD
5KBd127 mg/m2Benda 70 mg/m2 qwPDPD
6KCyd527 mg/m2Cyclo 200 mg qwPRSD
7KCyd256 mg/m2Cyclo 300 mg qwPRn.a.
8KCyd127 mg/m2Cyclo 750 mg qwPRPR
9KCyd120 mg/m2Cyclo 300 mg qwPRmixed response
10KCyd327 mg/m2Cyclo 300 mg qwSDSD
11KRd1127 mg/m2Rev 25 mg qdPRPR
12KRd720 mg/m2Rev 15 mg qodVGPRPR
13KRd636 mg/m2Rev 5 mg qdVGPRn.a.
14KRd220 mg/m2Rev 5 mg qodSDn.a.
15KRd627 mg/m2Rev 10 mg qdVGPRmixed response
16KRd327 mg/m2Rev 10 mg qdSDPD
17KRd227 mg/m2Rev 25 mg qdPDPD
18KRd227 mg/m2Rev 25 mg qdVGPRn.a.
19KRd1827 mg/m2Rev 25 mg qdVGPRPR
20KRd627 mg/m2Rev 25 mg qdPRmixed response
21KRd327 mg/m2Rev 25 mg qdPRPR
22KRd527 mg/m2Rev 25 mg qdVGPRPR
23KRd327 mg/m2Rev 20 mg qdPRmixed response
24KRd927 mg/m2Rev 25 mg qdVGPRn.a.
25KRd627 mg/m2Rev 15 mg qdPRn.a.
26KRd327 mg/m2Rev 10 mg qdVGPRSD
27KRd127 mg/m2Rev 10 mg qdPDn.a.
28KPd327 mg/m2Pom 4 mg qdSDmixed response
29KTd456 mg/m2Thal 50 mg qdSDPD
30KRCyd120 mg/m2Cyclo 300 mg qw, Rev 15 mg qdPDPD
31KRCyd356 mg/m2Cyclo 300 mg qw, Rev 10 mg qdPDPD
32KRCyd327 mg/m2Cyclo 300 mg qw, Rev 10 mg qdSDn.a.
33KTCyd436 mg/m2Cyclo 300 mg qw, Thal 100 mg qdPRSD
34Dara-Kd256 mg/m2Dara 16 mg/kg qwPRSD
35Dara-Kd120 mg/m2Dara 16 mg/kg qwPRSD
36Dara-KCyd227 mg/m2Dara 16 mg/kg qw, Cyclo 300 mg qwSDSD
37Dara-KCyd227 mg/m2Dara 16 mg/kg qw, Cyclo 200 mg qwN/An.a.
38Dara-KCyd315 mg/m2Dara 16 mg/kg qw, Cyclo 200 mg qwPRPR
39Dara-KCyd556 mg/m2Dara 16 mg/kg qw, Cyclo 300 mg qwPRSD
40Dara-KPd327 mg/m2Dara 16 mg/kg qw, Pom 3 mg qdN/Amixed response
41Dara-KPd220 mg/m2Dara 16 mg/kg qw, Pom 2 mg qodPDPD
42Dara-KPCyd120 mg/m2Dara 16 mg/kg qw, Pom 2 mg qd, Cyclo 250 mg qw PDn.a.
43Dara-KPCyd427 mg/m2Dara 16 mg/kg qw, Pom 2 mg qd, Cyclo 200 mg qw SDPD
44Dara-KPCyd115 mg/m2Dara 16 mg/kg qw, Pom 2 mg qd, Cyclo 200 mg qw SDn.a.
45Elo-KPd136 mg/m2Elo 10 mg/kg q2w, Pom 2 mg qdPRPR
Benda—bendamustine; Carf—carfilzomib; Dara—daratumumab; Dara-Kcyd—daratumumab, carfilzomib, cyclophosphamide, dexamethasone; Dara-Kd—daratumumab, carfilzomib, dexamethasone; Dara-KPCyd–daratumumab, carfilzomib, pomalidomide, cyclophosphamide, dexamethasone; Dara-KPd—daratumumab, carfilzomib, pomalidomide, dexamethasone; Elo—elotuzumab; Elo-KPd—elotuzumab, carfilzomib, pomalidomide, dexamethasone; EMDextramedullary disease; ImiD—immunomodulatory drugs; KBd—carfilzomib, bendamustine, dexamethasone; KcyD—carfilzomib, cyclophosphamide, dexamethasone; Kd—carfilzomib, dexamethasone; KPd—carfilzomib, pomalidomide, dexamethasone; KRCyd - carfilzomib, lenalidomide, cyclophosphamide, dexamethasone; KRd—carfilzomib, lenalidomide, dexamethasone; KTCyd—carfilzomib, thalidomide, cyclophosphamide, dexamethasone; KTd—carfilzomib, thalidomide, dexamethasone; N/A—not applicable; n.a.—not available; Pat—Patient; PD—progressive disease; Pom—pomalidomide; PR–partial remission; Rev—lenalidomide; SD–stable disease; Thal—thalidomide; VGPR—very good partial remission.
Table 3. Adverse events during therapy.
Table 3. Adverse events during therapy.
Adverse EventsAny Grade ≥ 2Grade 3Grade 4
Hematological Events, n (%)
Anemia37 (82)25 (56)
White Blood Cell Decreased32 (71)19 (42)5 (11)
Neutrophil Count Decreased30 (67)7 (16)6 (13)
Platelet Count Decreased29 (64)9 (20)13 (29)
Febrile Neutropenia1 (2) 1 (2)
Non-Hematological Events, n (%)
Pneumonia6 (13)4 (9)
Heart Failure6 (13)5 (11)2 (4)
Influenza4 (9)4 (9)
Upper Respiratory Infection3 (7)3 (7)
Liver Enzyme Increased2 (4)1 (2)
Urinary Tract Infection2 (4)2 (4)
Cytokine Release Syndrome 1 (2)1 (2)
Gastrointestinal Infection1 (2)1 (2)
Catheter Related Infection1 (2)1 (2)
Peripheral Polyneuropathy1 (2)1 (2)
Convulsion1 (2)1 (2)
Renal Failure1 (2)1 (2)
Oral Hemorrhage 1 (2)1 (2)
Bacterial Meningitis1 (2)1 (2)
Skin Infection1 (2)1 (2)
Sinusitis1 (2)1 (2)
Atrial Fibrillation1 (2)
Thromboembolic Events2 (4)
Death2 (4)

Share and Cite

MDPI and ACS Style

Zhou, X.; Flüchter, P.; Nickel, K.; Meckel, K.; Messerschmidt, J.; Böckle, D.; Knorz, S.; Steinhardt, M.J.; Krummenast, F.; Danhof, S.; et al. Carfilzomib Based Treatment Strategies in the Management of Relapsed/Refractory Multiple Myeloma with Extramedullary Disease. Cancers 2020, 12, 1035. https://doi.org/10.3390/cancers12041035

AMA Style

Zhou X, Flüchter P, Nickel K, Meckel K, Messerschmidt J, Böckle D, Knorz S, Steinhardt MJ, Krummenast F, Danhof S, et al. Carfilzomib Based Treatment Strategies in the Management of Relapsed/Refractory Multiple Myeloma with Extramedullary Disease. Cancers. 2020; 12(4):1035. https://doi.org/10.3390/cancers12041035

Chicago/Turabian Style

Zhou, Xiang, Patricia Flüchter, Katharina Nickel, Katharina Meckel, Janin Messerschmidt, David Böckle, Sebastian Knorz, Maximilian Johannes Steinhardt, Franziska Krummenast, Sophia Danhof, and et al. 2020. "Carfilzomib Based Treatment Strategies in the Management of Relapsed/Refractory Multiple Myeloma with Extramedullary Disease" Cancers 12, no. 4: 1035. https://doi.org/10.3390/cancers12041035

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