Next Article in Journal
Oral Immune-Related Adverse Events Caused by Immune Checkpoint Inhibitors: Salivary Gland Dysfunction and Mucosal Diseases
Previous Article in Journal
Systematic Review of Single-Fraction Stereotactic Body Radiation Therapy for Early Stage Non-Small-Cell Lung Cancer and Lung Oligometastases: How to Stop Worrying and Love One and Done
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Association between Patient Characteristics and the Efficacy and Safety of Selinexor in Diffuse Large B-Cell Lymphoma in the SADAL Study

by
Josée M. Zijlstra
1,*,
George Follows
2,
Rene-Olivier Casasnovas
3,
Joost S. P. Vermaat
4,
Nagesh Kalakonda
5,
Sylvain Choquet
6,
Brian Hill
7,
Catherine Thieblemont
8,
Federica Cavallo
9,
Fatima De la Cruz
10,
John Kuruvilla
11,
Nada Hamad
12,13,14,
Ulrich Jaeger
15,
Paolo Caimi
16,
Ronit Gurion
17,
Krzysztof Warzocha
18,
Sameer Bakhshi
19,
Juan-Manuel Sancho
20,
Michael Schuster
21,
Miklos Egyed
22,
Fritz Offner
23,
Theodoros P. Vassilakopoulos
24,
Priyanka Samal
25,
Matthew Ku
26,
Jenny Xu
27,
Kelly Corona
27,
Kamal Chamoun
27,
Jatin Shah
27,
Miguel Canales
28 and
Marie Maerevoet
29
add Show full author list remove Hide full author list
1
Department of Hematology, Amsterdam UMC, Cancer Center, Vrije Universiteit, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
2
Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
3
Department of Hematology, University Hospital F. Mitterrand and INSERM 1231, 21000 Dijon, France
4
Department of Hematology, Leiden University Medical Center, Albinesdreef 2, 2333 ZA Leiden, The Netherlands
5
Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GE, UK
6
Hematology, Hôpital Pitié Salpêtrière, 47-83 Bd de l'Hôpital, 75013 Paris, France
7
Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
8
Hemato-Oncology, APHP, Saint-Louis Hospital & Paris University, 75010 Paris, France
9
Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino/AOU, Città della Salute e della Scienza di Torino, 10126 Torino, Italy
10
Hospital Universitario Virgen del Rocio, E-41013 Sevilla, Spain
11
Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
12
Department of Hematology, St. Vincent’s Hospital Sydney, Darlinghurst, NSW 2010, Australia
13
St Vincent's Clinical School Sydney, University of New South Wales, Sydney, NSW 2052, Australia
14
School of Medicine, University of Notre Dame Australia, Fremantle, WA 6160, Australia
15
Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
16
Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
17
Hematology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva & Tel-Aviv University, Tel-Aviv 49100, Israel
18
Department of Hematology, Instytut Hematologii I Transfuzjologii, Chocimska 5, 00-791 Warsaw, Poland
19
Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
20
Clinical Hermatology Department, Hospital Germans Trias i Pujol, Institut Català d'Oncologia, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
21
Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY 11794, USA
22
Department of Hematology, Teaching Hospital Mór Kaposi, Tallian Gy. U 20-32, H-7400 Kaposvár, Hungary
23
Department of Hematology, Ghent University Hospital, 9000 Ghent, Belgium
24
Department of Hematology, National and Kapodistrian University of Athens, 15772 Athens, Greece
25
Hematology-Hemato-Oncology, Institute of Medical Sciences & SUM Hospital, Bhubaneswar 751003, India
26
Department of Haematology, St.Vincent’s Hospital, University of Melbourne, Melbourne, VIC 3065, Australia
27
Karyopharm Therapeutics, Newton, MA 02459, USA
28
Department of Hematology, Autonoma University, La Paz University Hospital, 28046 Madrid, Spain
29
Department of Hematology, Institut Jules Bordet, 1070 Brussels, Belgium
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(3), 791; https://doi.org/10.3390/cancers14030791
Submission received: 21 December 2021 / Revised: 24 January 2022 / Accepted: 25 January 2022 / Published: 4 February 2022

Abstract

:

Simple Summary

Diffuse large B-cell lymphoma (DLBCL) is a complex disease. A combination of immunotherapy and chemotherapy is used to treat DLBCL at initial diagnosis. Additional treatments are available when DLBCL does not respond to initial treatment; however, for many patients, DLBCL will stop responding to treatment (relapse) or may not respond at all (refractory). Selinexor is a novel, oral medication that belongs to a class of drugs called selective inhibitors of nuclear export, and it works by killing cancer cells in patients with DLBCL that has relapsed after or is refractory to at least two treatments. When deciding on a course of treatment, it is useful for physicians to know which frequently described clinical characteristics of DLBCL affect the activity and tolerability of selinexor. We found that selinexor showed similar activity and tolerability across most of the frequently described clinical characteristics assessed.

Abstract

Selinexor, an oral selective inhibitor of nuclear export, was evaluated in the Phase 2b SADAL study in patients with diffuse large B-cell lymphoma (DLBCL) who previously received two to five prior systemic regimens. In post hoc analyses, we analyzed several categories of patient characteristics (age, renal function, DLBCL subtype, absolute lymphocyte count, transplant status, number of prior lines of therapy, refractory status, Ann Arbor disease stage, and lactate dehydrogenase) at baseline, i.e., during screening procedures, to determine their potential contributions to the efficacy (overall response rate [ORR], duration of response [DOR], overall survival [OS]) and tolerability of selinexor. Across most categories of characteristics, no significant difference was observed in ORR or DOR. OS was significantly longer for patients < 65 vs. ≥ 65 years, and for those with lymphocyte counts ≥ 1000/µL vs. < 1000/µL or lactate dehydrogenase ≤ ULN vs. > ULN. The most common adverse events (AEs) across the characteristics were thrombocytopenia and nausea, and similar rates of grade 3 AEs and serious AEs were observed. With its oral administration, novel mechanism of action, and consistency in responses in heavily pretreated patients, selinexor may help to address an important unmet clinical need in the treatment of DLBCL.

1. Introduction

Diffuse large B-cell lymphoma (DLBCL) is a complex disease that progresses rapidly and has variable clinical outcomes. It is the most common subtype of non-Hodgkin lymphoma (NHL) accounting for 40% of NHL cases worldwide [1]. Although DLBCL may be diagnosed in people of all age groups, its incidence is highest among people 65–74 years [2].
Clinical characteristics associated with a disease may affect the selection and outcome of treatment regimens. For DLBCL, the immunochemotherapy combination of rituximab with cyclophosphamide, doxorubicin, vincristine, and prednisone is the established frontline treatment, curing up to 60% of patients treated with a full course of therapy [3,4,5]. Potentially curative salvage therapy for patients with relapsing or refractory (R/R) disease includes high-dose chemotherapy followed by autologous stem cell transplantation (ASCT). Chimeric antigen receptor (CAR)-T cell therapy is a more recently approved treatment for R/R DLBCL and, similar to ASCT, CAR-T is limited by availability and patient eligibility [6,7]. Patients who are not candidates for intensive chemotherapy, ASCT, or CAR-T therapy are offered non-curative therapies including chemotherapy combinations with targeted agents or targeted agents alone.
Exportin 1 (XPO1) is the major nuclear exporter of tumor suppressor proteins (TSPs) such as p53, FOXO, IκB, and Rb, and the mRNA cap-binding protein eIF4E [8,9,10]. XPO1 overexpression is observed in many types of cancer including DLBCL, and higher levels of XPO1 are associated with poor prognosis in DLBCL [11] and other cancers [12]. XPO1 overexpression leads to the nuclear export and functional inactivation of TSPs and enhances the levels of eIF4E-associated oncoproteins such as c-Myc [13,14,15]. Selinexor is a first-in-class selective inhibitor of nuclear export (SINE) compound that selectively binds and inactivates XPO1. Inactivation of XPO1 forces the nuclear retention and reactivation of cell cycle regulators and reduces the concentration of the oncoproteins, several of which play critical roles in NHL [8,16,17,18].
Based on results from the Selinexor Against Diffuse Aggressive Lymphoma (SADAL) study [19], the US Food and Drug Administration in 2020 approved the use of single-agent selinexor for the treatment of adult patients with DLBCL that is de novo or transformed from follicular lymphoma after at least two prior lines of systemic therapy [20].
In this report, we describe the results of the post hoc analyses of baseline characteristics for patients with R/R DLBCL from the SADAL study. The objective of the analyses in this report was to determine whether clinically important differences exist in the efficacy and safety of selinexor based on clinical characteristics frequently described in patients with DLBCL.

2. Materials and Methods

2.1. Study Design

SADAL was an open-label, phase 2b study carried out at 59 sites in 19 countries. Details on the design of the study (NCT02227251) have been reported elsewhere [19].

2.2. Endpoints

The efficacy endpoints examined in these post hoc analyses include overall response rate (ORR), the primary endpoint of the SADAL study, duration of response (DOR), and overall survival (OS).

2.3. Baseline Characteristics

In these post hoc analyses, we examined the clinical characteristics recorded at baseline, i.e., during screening procedures for patients enrolled in the SADAL study, to determine whether the characteristics, which are frequently described in patients with DLBCL, are associated with differences in the efficacy and safety of selinexor. The clinical characteristics included in these analyses are: (1) age, <65 years vs. ≥65 years; (2) renal function, creatinine clearance (CrCL) ≤ 60 mL/min vs. > 60 mL/min; (3) DLBCL subtypes, germinal center B-cell (GCB) vs. non-GCB; (4) absolute lymphocyte count (ALC), <1000/µL vs. ≥1000/µL; (5) prior ASCT or transplant ineligible; (6) number of prior lines of therapy, 2 vs. ≥3 prior lines; (7) refractory disease status, progressive disease (PD) < 6 months from first-line therapy (primary refractory) vs. PD ≥ 6 months from first-line (non-primary refractory); (8) Ann Arbor stage 1 or 2 vs. stage 3 or 4; and (9) lactate dehydrogenase (LDH) level, >ULN vs. ≤ULN.

2.4. Statistical Analyses

The primary analysis of the SADAL study was based on a one-sided exact test at an α level of 0·025 to detect a minimum of 25% of patients with a partial response or better against a value of 15% under the null hypothesis with 80% power [19]. These post hoc analyses of the SADAL study included the modified intention-to-treat (mITT) population, i.e., 134 patients who received 60 mg selinexor twice weekly until disease progression or unacceptable toxicity.
At data cutoff (1 August 2019), the primary analysis of ORR was calculated in the mITT population with the exact 2-sided 95% CI [19]. Summary statistics were computed and displayed for each subgroup and according to each assessment timepoint.
Summary statistics for continuous variables minimally included number, mean, standard deviation, minimum, median, and maximum. Frequencies and percentages are presented for categorical variables and a 2-sided 95% exact confidence interval (CI) for ORR. The chi-squared test was used to compare proportions between subgroups. For time-to-event variables, the Kaplan–Meier method was used for descriptive summaries. Log-rank test and Cox proportional hazards model were used to compare survival distributions between subgroups. Statistical analyses were performed using SAS (version 9.4).

3. Results

The SADAL study was initially designed as a randomized trial to evaluate two doses of selinexor, 60 mg and 100 mg, administered twice weekly. A preplanned interim analysis showed that the higher dose of 100 mg had similar levels of efficacy and was associated with greater toxicity compared with the 60-mg dose; consequently, the 100 mg dose was discontinued [19]. The median time from last systemic therapy to the start of treatment with selinexor was 5.4 months.

3.1. Demographics

The demographics of the patients included in this analysis are summarized in Table 1. The median age was 67 years with 44.8% of patients ≥70 years. Most patients were men (59%), median of two prior treatment regimens (range 2–5), 41% of patients received three or more prior treatment regimens, and 29.9% previously underwent an ASCT.

3.2. Duration of Selinexor Treatment

The median duration of selinexor treatment was 9 weeks (range 1–193) for the 134 patients in these post hoc analyses who comprised the mITT population in the SADAL study. Patients with a longer median duration of treatment with selinexor were those who were <65 years (13.5 weeks), prior ASCT (16 weeks), or with LDH ≤ ULN (15 weeks).
The median time on selinexor for responders was 214 days (range 53–1351) compared with 43 days (range 1–288) for those who did not have at least a PR on treatment. Among the patients who received at least two cycles of treatment, ORR was 52%.

3.3. Efficacy

The relationship between the baseline characteristics and the efficacy endpoints of selinexor are summarized in Table 2. Kaplan–Meier analyses of OS based on each characteristic are shown in Figure 1.

3.3.1. Age

ORR was numerically higher in patients <65 years (36.5%) compared with patients ≥65 years (24.4%) (p = 0.19); however, DOR was similar (9.2 months vs. 9.7 months, hazard ratio [HR] 0.95 [95% CI 0.37–2.48], p = 0.94) between the age groups at baseline. Median OS was 7.8 months for older patients compared with the significantly longer median OS of 13.7 months for patients <65 years (HR 1.65 [95% CI 1.03–2.64], p = 0.04).

3.3.2. Renal Function

ORR was similar for patients with baseline CrCl ≤ 60 mL/min or >60 mL/min (29.7% vs. 28.9% [p = 1.00]), and DORs were numerically though not significantly different for patients with CrCl ≤60 mL/min vs. >60 mL/min (23.0 months vs. 9.2 months, HR 0.51 [95%CI 0.16–1.60], p = 0.24). Median OS for patients with CrCl ≤60 mL/min was not significantly different than that for patients with CrCl > 60 mL/min (7.8 months vs. 9.1 months, HR 1.14 [95% CI 0.70–1.42], p = 0.59).

3.3.3. Germinal Center B-Cell versus Non-Germinal Center B-cell

ORR for patients with GCB was numerically (31.7%) but not significantly higher than for non-GCB (24.2%) DLBCL (p = 0.45) and median DOR was 23 months and 9.3 months, respectively (HR 1.58 [95% CI 0.55–4.53], p = 0.39). OS was similar with 9.0 months for patients with the GCB subtype and 8.3 months for patients with non-GCB (HR 0.95 [95% CI 0.61–1.50], p = 0.84).

3.3.4. Absolute Lymphocyte Count

There was no statistically significant difference in ORR between patients with baseline ALC < 1000/µL or ≥1000/µL, with ORR of 25.4% vs. 32.8% (p = 0.45). A trend toward higher DOR was observed in patients with ALC ≥ 1000/µL (4.9 months vs. 23 months, HR 1.83 [95%CI 0.68–4.97], p = 0.23) while median OS was significantly shorter for patients with ALC < 1000/µL (7.6 months vs. 15.5 months, HR 1.79 [95% CI 1.12–2.84], p = 0.01) as previously reported in DLBCL [21].

3.3.5. Prior ASCT vs. Transplant Ineligible

As compared with patients who were transplant ineligible, patients who received a prior ASCT had a significantly better ORR (42.5% vs. 23.4% [p = 0.04]) while median DOR was similar (8.4 months vs. 9.7 months [p = 0.93]). Median OS for patients with prior ASCT was 10.9 months versus 7.8 months for those who were transplant ineligible (HR 0.72; 95% CI 0.44–1.17; p = 0.18). The reasons that patients were ineligible for ASCT may have included one or more of the following: persistent disease (n = 30), failure to collect stem cells (n = 5), age (n = 46), frailty (n = 13), inadequate performance status (n = 10), renal or hepatic dysfunction (n = 3), comorbidities (n = 7), cardiac dysfunction (n = 9), pulmonary dysfunction (n = 3), infection risk (n = 2), patient’s refusal (n = 6), or financial reasons (n = 6).

3.3.6. Prior Therapy

ORR for patients who previously received 2 lines of therapy was 27.8% versus 30.9% for those with ≥3 lines (p = 0.85); median DORs were 10.4 months and 8.4 months (HR 1.51 [95% CI 0.58–3.94], p = 0.40), respectively. Median OS for patients with 2 prior lines of therapy was 9.1 months versus 8.2 months for those with ≥3 lines (HR 0.93; 95% CI 0.60–1.46; p = 0.76).

3.3.7. Disease Refractory Status

The ORR in patients with DLBCL that progressed within 6 months of first-line therapy (defined as primary refractory disease) was numerically lower but not significantly different from that for patients with PD ≥ 6 months after first-line therapy (21.8% vs. 37.1%, p = 0.11). Median DORs were 9.7 months and 9.3 months (HR 0.84 [95% CI 0.28–2.54], p = 0.76), respectively. Median OS for patients with primary refractory disease was 6.6 months versus 11.1 months for those with disease that progressed ≥6 months after first-line therapy (HR 0.84; 95% CI 0.52–1.35; p = 0.46).

3.3.8. Ann Arbor Stage 1 or 2 vs. 3 or 4

Patients with disease stage 1 or 2 versus 3 or 4 at screening had similar ORRs (30.3% vs. 28.7% [p = 1.00], respectively). Median DOR was not reached for patients with stage 1 or 2 disease and was 4.9 months for those with stage 3 or 4 disease. Median OS was similar with 9.8 months for patients with stage 1 or 2 disease and 9.0 months for patients with stage 3 or 4 (HR 0.97 [95% CI 0.57–1.64], p = 0.91). ORR and OS were not statistically different for patients with extranodal disease versus those without it: ORRs were 29.5% vs. 28.2% (p = 1.00), and OS were 8.2 months vs. 11.2 months (HR 1.03 [95% CI 0.62–1.72], p = 0.91).

3.3.9. Lactate Dehydrogenase >ULN vs. ≤ULN

Of the patients with baseline LDH > ULN, 17.4% had a response to treatment while 41.9% of those with baseline LDH ≤ ULN had an ORR (p = 0.004); median DOR was 9.7 months for patients with LDH > ULN and 10.4 months for patients with LDH ≤ ULN. Median OS was significantly shorter for patients with LDH > ULN (5.4 months vs. 20.8 months, HR 2.33 [95% CI 1.45–3.72], p = 0.0003).
Multivariate analysis including age, ALC, and LDH showed that only LDH ≤ ULN was independently associated with higher OS (HR = 2.35 [95% CI 1.45–3.79]) (see Table 2).

3.4. Safety

The safety profile of these post hoc analyses is based on the baseline characteristics and adverse events (AEs) reported by patients during the SADAL study.
Overall, 132 of the 134 patients (98.5%) included in this analysis experienced at least one AE during the study. Across the baseline characteristics, thrombocytopenia (rarely with clinically significant bleeding) and nausea occurred in at least 50% of the patients (Table 3). There were no notable differences in AEs between the categories for each characteristic.
The frequency of grade 3 or higher AEs ranged from 82% to 90% for the categories of each characteristic and was similar for most characteristics including age (<65 years, 82.7%; ≥65 years, 85.4%), DLBCL subtypes (GCB, 81.0%; non-GCB, 86.4%), ALC (<1000/µL, 85.9%; ≥1000/µL 82.0%), disease stage (1 or 2, 81.8%; 3 or 4, 85.1%), and LDH (>ULN, 87.0%; ≤ULN, 82.3%), prior therapies (<2 therapies, 84.8%; ≥3 therapies, 83.6%), and refractory disease (PD < 6 months after first-line therapy, 83.6%; PD ≥ 6 months, 85.5%). Patients with CrCl ≤ 60 mL/min had a slightly higher incidence of grade 3 or higher AEs than patients with CrCl > 60 mL/min (89.2% vs. 82.5%). The incidence of grade 3 or higher AEs was higher for patients with prior ASCT versus transplant ineligible (90.0% vs. 81.9%).
Serious AEs (SAEs) occurred at a similar frequency for most characteristics including age (<65 years, 48.1%; ≥65 years, 46.3%), DLBCL subtype (GCB, 44.4%; non-GCB, 48.5%), ALC (<1000/µL, 49.3%; ≥1000/µL 44.3%), transplant status (prior ASCT, 47.5%; transplant ineligible, 46.8%), number of prior lines of therapy (2 prior lines, 48.1%; ≥3 prior lines, 45.5%), refractory status (PD <6 months after first-line therapy, 45.5%; PD ≥6 months, 53.2%), and LDH (>ULN, 56.5%; ≤ULN, 37.1%). Patients with CrCl >60 mL/min had a slightly higher frequency of SAEs (49.5%) than patients with CrCl of ≤60 mL/min (40.5%). The incidence of SAEs was notably higher for patients with stage 3 or 4 disease (53.5%) compared with those with stage 1 or 2 disease (27.3%) (p = 0.02), consistent with a significant contribution of disease extent to the development of an SAE.
Across baseline characteristics, 17.2% of patients with at least one AE withdrew from treatment with selinexor. Of all patients in this analysis, 48.5% had a dose reduction and a majority (64.2%) had at least one dose that was interrupted or withheld (Table 4). In the SADAL study, the most common dose reduction AEs (3 or more patients [≥2%]) were thrombocytopenia in 30 patients (23.6%), neutropenia in 11 (8.7%), fatigue in 6 (4.7%), and nausea in 4 (3.1%). Common reasons for dose interruption of more than 2 weeks included thrombocytopenia in 18 (14.2%) patients, fatigue in 3 (2.4%), and asthenia in 3 (2.4%).
Of the 134 patients in the mITT, 27 patients (20.1%) died within 30 days of receiving the last dose of selinexor: 22 (16.4%) died due to progressive disease and 5 patients (3.7%) died of an unrelated AE.

4. Discussion

Clinical characteristics associated with DLBCL contribute to outcomes for patients receiving treatment for the disease. Selinexor is a novel, oral nuclear export inhibitor; it is important to determine which clinical parameters of DLBCL are associated with the drug’s activity and tolerability. In these post hoc analyses, we examined multiple frequently described baseline clinical characteristics of patients with R/R DLBCL who were enrolled in the SADAL study. The baseline clinical characteristics examined in these analyses included age, renal function (CrCl), DLBCL subtype, ALC, prior ASCT, number of prior lines of therapy, refractory status, Ann Arbor stage, and LDH level.
It has already been established that age is not a factor in the metabolism of selinexor and has no clinically significant effect on the pharmacokinetics of the drug [22]. In this analysis, we showed that patients with R/R DLBCL who were ≥65 years had a similar clinical benefit when compared with those <65 years and treated with selinexor with comparable ORR, DOR, and perhaps most importantly, overall tolerability. As expected, younger patients (<65 years) had significantly longer OS (p = 0.04) than those ≥65 years, most likely due to comorbid medical conditions in the older population [23]; AE rates were not significantly different in these two populations (Table 3). Doses of treatment were interrupted or withheld for a small proportion of patients <65 years compared with patients ≥65 years (59.6% vs. 67.1%). These results indicate that single-agent oral selinexor can induce durable responses with similar tolerability in younger and older patients with heavily pretreated DLBCL. These observations are particularly important for older patients who may prefer a non-parenteral agent that can be taken at home with proper monitoring.
In addition to age, selinexor metabolism is not affected by renal function with no clinically significant effect on the pharmacokinetics of selinexor [22]. Additionally, renal clearance is a minor route for the elimination of selinexor with most excreted in feces by the hepatobiliary route as unchanged drug or metabolites (unpublished data). In this analysis, patients with reduced renal function (CrCl ≤ 60 mL/min) and those with normal function (CrCl > 60 mL/min) had similar outcomes when treated with selinexor 60 mg twice weekly, unlike other settings in which patients with newly diagnosed DLBCL and lower renal function were associated with lower overall survival [24]. The safety profile in the current analysis was similar between the categories in the proportion of patients who experienced AEs, the types of AEs, and deaths within 30 days of the last dose. This safety profile is similar to that from previous assessments in which patients with multiple myeloma and moderate (CrCl 30–60 mL/min) or severe (CrCl < 30 mL/min) renal impairment had a profile similar to that of selinexor in patients with normal renal function or mild renal impairment (unpublished data), which suggests that treatment with selinexor does not require dose adjustments in patients with renal dysfunction and R/R DLBCL.
Patients who were previously treated for DLBCL had strong and durable responses when treated with single-agent selinexor, regardless of GCB or non-GCB subtype [25].
Low ALC is also a known poor prognostic marker in patients with DLBCL [21,26,27,28]. For patients treated in the SADAL trial, the ORR was similar between patients regardless of baseline ALC; however, significantly longer OS (p = 0.01) was observed in patients with baseline ALC ≥ 1000 µL; responses in these patients tended to last longer. These results are consistent with reports in the literature regarding the poor prognosis of baseline lymphocyte count < 1000/µL, but suggest that the anti-lymphoma activity of selinexor is minimally affected by baseline ALC.
Patients with prior ASCT, compared with those who were transplant ineligible, had a significantly higher ORR; DOR was similar with a trend for a longer OS. These results are not unexpected since patients with prior ASCT were generally fitter and had substantial responses to second-line therapy which permitted the transplant.
Numerous therapies are available to treat R/R DLBCL; however, there is no standard of care after three or more lines. When treated with selinexor, ORR, DOR, and OS were comparable for patients who had 2 versus ≥3 lines of previous treatment, consistent with the novel, non-cross resistant mechanism of action for selinexor. Of note, the population enrolled in the SADAL study represented patients with aggressive disease as reflected in the median time of 5.4 months since last treatment to initiation of selinexor compared with the L-MIND study in which the median time was 9 months for patients treated with tafasitamab plus lenalidomide [29]. Additionally, the analyses in this report showed that ORR was significantly higher for patients who had undergone ASCT than for those without it. ORR was also higher for patients with a response of PR or CR to the last line of therapy than for those without a response. Furthermore, two of the thirty-seven patients who did not have a response of PR or CR with any previous therapy had a response to selinexor (two PR, four CR). In patients with primary refractory DLBCL, ORR was not significantly lower than ORR for patients with non-primary refractory disease, again consistent with selinexor’s novel mechanism of action. These results are in contrast to those from the SCHOLAR-1 analysis, a large patient-level pooled retrospective analysis in refractory disease, in which outcomes were poor for most patients (73%) who did not respond to salvage therapy or were unable to receive ASCT [30]. Because the SCHOLAR-1 analysis was carried out by others, we do not have the data or information on the baseline characteristics needed to compare individual characteristics with those from the SADAL study. However, overall, the outcomes from the SADAL study were better than those published from the SCHOLAR-1 analysis.
These post hoc analyses were limited by their retrospective nature and by the small number of patients in the subgroups. For these reasons some of the comparisons did not have the power to conclude whether differences were statistically significant.
Disease stage and LDH are strong prognostic factors that are part of the IPI and R-IPI prognostic scoring system for newly diagnosed patients. However, the significance of these prognostic scores is still not validated in the relapsed/refractory setting. In the current analysis, we found that, although ORR and OS were statistically similar between patients with different disease stages, ORR was significantly higher for patients with LDH ≤ ULN and OS was significantly longer. These differences should be verified in larger studies, but strongly suggest that single-agent oral selinexor is substantially more active in patients with LDH ≤ ULN and, as a single oral therapy, may be a particularly attractive option for these patients.

5. Conclusions

Patients with R/R DLBCL tend to be clinically complex because of their advanced age and medical history which may include prior treatments, use of concomitant medications, comorbidities, and other medical concerns. As a result, these patients usually are unable to tolerate multiple cycles of standard combination therapies for DLBCL creating an unmet medical need, especially for patients with R/R DLBCL previously treated with multiple lines of therapy. Selinexor showed similar activity and tolerability across most of the frequently described clinical characteristics assessed here (age, renal function, DLBCL subtype, lymphocyte counts, prior ASCT, number of prior lines of therapy, refractory status), but appeared to be less active in patients with LDH levels >ULN. Notably, SAE rates were about twice as high in patients with stage 3/4 disease as compared with stage 1/2 DLBCL, consistent with a significant contribution to AEs from the tumor itself. Selinexor, with its novel mechanism of action, ease of oral administration, and ability to produce rapid and durable responses in patients with heavily pretreated disease, may help to fill this important unmet clinical need. Combination therapy studies with selinexor (NCT04442022, NCT04607772) are ongoing to determine optimal dosing and response rates/durability; these regimens are highly likely to be substantially more active than single-agent selinexor.

Author Contributions

M.M. and J.S. contributed to the study design; J.M.Z., G.F., R.-O.C., J.S.P.V., N.K., S.C., B.H., C.T., F.C., F.D.l.C., J.K., N.H., U.J., P.C., R.G., K.W., S.B., J.-M.S., M.S., M.E., F.O., T.P.V., P.S., M.K., J.S., M.C. and M.M. collected the data; J.X. analyzed the data; K.C. (Kelly Corona) and K.C. (Kamal Chamoun). played an important role in interpreting the results. All authors have read and agreed to the published version of the manuscript.

Funding

Karyopharm Therapeutics sponsored the SADAL study. Karyopharm Therapeutics paid for the post-hoc analyses described here and for the medical writing services provided to prepare this report.

Institutional Review Board Statement

The institutional review board approved the protocol (Protocol Number:KCP-330-009), and the study was done in accordance with the International Conference on Harmonisation Good Clinical Practice guidelines and the principles of the Declaration of Helsinki.

Informed Consent Statement

The SADAL study was approved and performed in accordance with the International Conference on Harmonization, the Guidelines for Good Clinical Practice, appropriate regulatory requirements, and with approval of institutional review boards at individual enrolling institutions. All patients provided written informed consent before study start.

Data Availability Statement

The data presented in this study are available on request.

Acknowledgments

Medical writing services were provided by Christine Kuepfer.

Conflicts of Interest

G.F. reports advisory boards/lecturing for Karyopharm, Roche, Abbvie, Janssen, and AstraZeneca. R-O.C. reports advisory boards for Roche, Takeda, Bristol-Myers Squibb, Amgen, Gilead, Merck, Abbvie, and research grants from Roche and Gilead outside the submitted work. C.T. reports advisory boards for Novartis, Kite Pharma, Roche, Bristol-Myers Squibb, Amgen, Gilead, Merck, Abbvie and Incyte. J.K. reports research funding from Janssen, Roche, and AstraZeneca; honoraria from Amgen, Antengene, AstraZeneca, Bristol-Myers Squibb, Gilead, Incyte, Janssen, Karyopharm, Merck, Novartis, Pfizer, Roche, Seattle Genetics, and TG Therapeutics; consulting for Abbvie, Bristol-Myers Squibb, Gilead, Karyopharm, Merck, Roche, and Seattle Genetics; and serving on a data safety and monitoring board for Karyopharm. N.H. reports advisory boards for Novartis, Gilead, Roche, Janssen, CSL, and Antegen. U.J. reports honoraria for advisory boards for Karyopharm. P.F.C. reports research funding from ADC Therapeutics; grants from Genentech; consulting from ADC Therapeutics, Kite Pharma, Verastem Oncology, Seattle Genetics, Amgen and TG Therapeutics; and speaker’s bureau from Celgene. R.G. reports consulting for Roche, Jansen, Takeda, Gilead, Medison, Novartis and Neopharm. M.S. reports personal fees from Karyopharm during the conduct of the study, and personal fees from Amgen, Abbvie, Gilead, Takeda, Celgene, Pharmacyclics, Astellas, Verastem, Merck, Novartis, Genentech, and Seattle Genetics, outside the submitted work. T.P.V. reports research support from Merck, Takeda, Amgen, Pfizer, and Dr. Reddy’s; honoraria from Takeda, Roche, Genesis Pharma, Merck, Novartis, Amgen, Glaxo, Abbvie, Integris, and Astra Zeneca; and serves on the scientific advisory board of Takeda, Roche, Genesis Pharma, and Novartis. M.C. reports speaking for Amgen, Janssen, Novartis, Roche, Sandoz, and Takeda; advisory boards/consulting for Bristol-Myers Squibb/Celgene, Gilead/Kite Pharma, iQone, Janssen, Karyopharm, Novartis, Roche, and Sandoz; CME support, travel and accommodation from Gilead/Kite Pharma, Janssen, Novartis, Roche, and Sandoz. J.X., K.C. (Kelly Corona), K.C. (Kamal Chamoun), and J.S. are employees and stockholders of Karyopharm Therapeutics. All other authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Stewart, B.W.; Wild, C.P. (Eds.) World Cancer Report 2014; World Health Organization, International Agency for Research on Cancer, WHO Press: Geneva, Switzerland, 2015; Available online: http://publications.iarc.fr/Non-Series-Publications/World-Cancer-Reports/World-Cancer-Report-2014 (accessed on 22 October 2020).
  2. Howlader, N.; Noone, A.M.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. (Eds.) SEER Cancer Statistics Review, 1975–2017; National Cancer Institute: Bethesda, MD, USA, 2020. Available online: https://seer.cancer.gov/statfacts/html/dlbcl.html (accessed on 22 October 2020).
  3. Pfreundschuh, M.; Trümper, L.; Österborg, A.; Pettengell, R.; Trneny, M.; Imrie, K.; Ma, D.; Gill, D.; Walewski, J.; Zinzani, P.L.; et al. CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: A randomised controlled trial by the MabThera International Trial (MInT) Group. Lancet Oncol. 2006, 7, 379–391. [Google Scholar] [CrossRef]
  4. Coiffier, B.; Lepage, E.; Brière, J.; Herbrecht, R.; Tilly, H.; Bouabdallah, R.; Morel, P.; Van Den Neste, E.; Salles, G.; Gaulard, P.; et al. CHOP Chemotherapy plus Rituximab Compared with CHOP Alone in Elderly Patients with Diffuse Large-B-Cell Lymphoma. N. Engl. J. Med. 2002, 346, 235–242. [Google Scholar] [CrossRef] [PubMed]
  5. Feugier, P.; Van Hoof, A.; Sebban, C.; Solal-Celigny, P.; Bouabdallah, R.; Fermé, C.; Christian, B.; Lepage, E.; Tilly, H.; Morschhauser, F.; et al. Long-Term Results of the R-CHOP Study in the Treatment of Elderly Patients with Diffuse Large B-Cell Lymphoma: A Study by the Groupe d’Etude des Lymphomes de l’Adulte. J. Clin. Oncol. 2005, 23, 4117–4126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. KYMRIAH (Tisagenlecleucel) Prescribing Information; Novartis Pharmaceuticals Corporation: East Hanover, NJ, USA, 2018.
  7. YESCARTA (Axicabtagene Ciloleucel) Prescribing Information; Kite Pharma: Santa Monica, CA, USA, 2019.
  8. Conforti, F.; Wang, Y.; Rodriguez, J.A.; Alberobello, A.T.; Zhang, Y.W.; Giaccone, G. Molecular pathways: Anticancer activity by inhibition of nucleocytoplasmic shuttling. Clin. Cancer Res. 2015, 21, 4508–4513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Tan, D.S.; Bedard, P.L.; Kuruvilla, J.; Siu, L.L.; Razak, A.R. Promising SINEs for embargoing nuclear-cytoplasmic export as an anticancer strategy. Cancer Discov. 2014, 4, 527–537. [Google Scholar] [CrossRef] [Green Version]
  10. Okamura, M.; Inose, H.; Masuda, S. RNA export through the NPC in eukaryotes. Genes 2015, 6, 124–149. [Google Scholar] [CrossRef]
  11. Luo, B.; Huang, L.; Gu, Y.; Li, C.; Lu, H.; Chen, G.; Peng, Z.; Feng, Z. Expression of exportin-1 in diffuse large B-cell lymphoma: Immunohistochemistry and TCGA analyses. Int. J. Clin. Exp. Pathol. 2018, 11, 5547–5560, Erratum in Int. J. Clin. Exp. Pathol. 2019, 12, 2817–2818. [Google Scholar]
  12. Sendino, M.; Omaetxebarria, M.J.; Rodriguez, J.A. Hitting a moving target: Inhibition of the nuclear export receptor XPO1/CRM1 as a therapeutic approach in cancer. Cancer Drug. Resist. 2018, 1, 139–163. [Google Scholar] [CrossRef] [Green Version]
  13. Gandhi, U.H.; Senapedis, W.; Baloglu, E.; Unger, T.J.; Chari, A.; Vogl, D.; Cornell, R.F. Clinical Implications of Targeting XPO1-mediated Nuclear Export in Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2018, 18, 335–345. [Google Scholar] [CrossRef] [Green Version]
  14. Han, X.; Wang, J.; Shen, Y.; Zhang, N.; Wang, S.; Yao, J.; Shi, Y. CRM1 as a new therapeutic target for non-Hodgkin lymphoma. Leuk. Res. 2015, 39, 38–46. [Google Scholar] [CrossRef]
  15. Culjkovic-Kraljacic, B.; Baguet, A.; Volpon, L.; Amri, A.; Borden, K.L. The oncogene eIF4E reprograms the nuclear pore complex to promote mRNA export and oncogenic transformation. Cell Rep. 2012, 2, 207–215. [Google Scholar] [CrossRef] [PubMed]
  16. Parikh, K.; Cang, S.; Sekhri, A.; Liu, D. Selective inhibitors of nuclear export (SINE)—A novel class of anti-cancer agents. J. Hematol. Oncol. 2014, 7, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Camus, V.; Miloudi, H.; Taly, A.; Sola, B.; Jardin, F. XPO1 in B cell hematological malignancies: From recurrent somatic mutations to targeted therapy. J. Hematol. Oncol. 2017, 10, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Hill, R.; Cautain, B.; de Pedro, N.; Link, W. Targeting nucleocytoplasmic transport in cancer therapy. Oncotarget 2014, 5, 11–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Kalakonda, N.; Maerevoet, M.; Cavallo, F.; Follows, G.; Goy, A.; Vermaat, J.S.P.; Casasnovas, O.; Hamad, N.; Zijlstra, J.M.; Bakhshi, S.; et al. Selinexor in patients with relapsed or refractory diffuse large B-cell lymphoma (SADAL): A single-arm, multinational, multicentre, open-label, phase 2 trial. Lancet Haematol. 2020, 7, e511–e522. [Google Scholar] [CrossRef]
  20. U.S. Food and Drug Administration. Food and Drug Administration 2020. FDA Approves Selinexor for Relapsed/Refractory Diffuse Large B-Cell Lymphoma. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-selinexor-relapsedrefractory-diffuse-large-b-cell-lymphoma (accessed on 15 January 2021).
  21. Porrata, L.F.; Ristow, K.; Haberman, T.M.; Witzig, T.E.; Inwards, D.J.; Ansell, S.M.; Johnston, P.B.; Micallef, I.N.; Colgan, J.P.; White, W.L.; et al. Absolute Lymphocyte Count at the Time of Relapse Predicts Survival in Patients with Diffuse Large B-Cell Lymphoma. Blood 2008, 112, 1763. [Google Scholar] [CrossRef]
  22. Karyopharm Therapeutics Inc. XPOVIO (Selinexor) Prescribing Information; Karyopharm Therapeutics Inc.: Newton, MA, USA, 2020. [Google Scholar]
  23. Kobayashi, Y.; Miura, K.; Hojo, A.; Hatta, Y.; Tanaka, T.; Kurita, D.; Iriyama, N.; Kobayashi, S.; Takeuchi, J. Charlson Comorbidity Index is an independent prognostic factor among elderly patients with diffuse large B-cell lymphoma. J. Cancer Res. Clin. Oncol. 2011, 137, 1079–1084. [Google Scholar] [CrossRef]
  24. Nishimura, N.; Fukuta, T.; Nishihara, A.; Shirouchi, Y.; Uryu, H.; Inoue, N.; Kusano, Y.; Tsuyama, N.; Takeuchi, K.; Mishima, Y.; et al. Impact of creatinine clearance in patients with diffuse large B-cell lymphoma treated with R-CHOP: A real-world long-term observation analysis at a single institute. Blood 2018, 132 (Suppl. S1), 5399. [Google Scholar] [CrossRef]
  25. Casasnovas, R.O.; Follows, G.; Zijlstra, J.M.; Vermaat, J.S.; Kalakonda, N.; Choquet, S.; Van Den Neste, E.; Hill, B.; Thieblemont, C.; Cavallo, F.; et al. Comparison of the effectiveness and safety of the oral selective inhibitor of nuclear export, selinexor, in diffuse large B cell lymphoma subtypes. Clin. Lymphoma Myeloma Leuk. 2021, 22, 24–33. [Google Scholar] [CrossRef]
  26. Oki, Y.; Yamamoto, K.; Kato, H.; Kuwatsuka, Y.; Taji, H.; Kagami, Y.; Morishima, Y. Low absolute lymphocyte count is a poor prognostic marker in patients with diffuse large B-cell lymphoma and suggests patients’ survival benefit from rituximab. Eur. J. Haematol. 2008, 81, 448–453. [Google Scholar] [CrossRef]
  27. Prochazka, V.; Trneny, M.; Salek, D.; Belada, D.; Kozak, T.; Papajik, T.; Pytlik, R.; Vasova, I.; Sykorova, A.; Jankovska, M.; et al. Median absolute lymphocyte count independently predicts survival of elderly patients with diffuse large B-cell lymphoma treated with R-chemotherapy: Analysis of 651 patients included in the Czech Lymphoma Project. Blood 2010, 116, 2882. [Google Scholar] [CrossRef]
  28. Panizo, C.; Rodríguez, A.J.; Gutiérrez, G.; Díaz, F.J.; González-Barca, E.; De Oña, R.; Grande, C.; Sancho, J.M.; García-Álvarez, M.F.; Sánchez-González, B.; et al. Evaluation of clinical and biological prognostic factors in relapsed or refractory diffuse large B-cell lymphoma patients after previous treatment with rituximab and chemotherapy: Results of the PRO-R-IPI study. Clin. Lymphoma Myeloma Leuk. 2015, 15, 398–403. [Google Scholar] [CrossRef] [PubMed]
  29. U.S. Food and Drug Administration. Center for Drug Evaluation and Research. Application No. 761163Orig1s000 Monjuvi. 24 July 2019. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/761163Orig1s000MultidisciplineR.pdf (accessed on 6 October 2021).
  30. Crump, M.; Neelapu, S.S.; Farooq, U.; Neste, E.V.D.; Kuruvilla, J.; Westin, J.; Link, B.K.; Hay, A.; Cerhan, J.R.; Zhu, L.; et al. Outcomes in refractory diffuse large B-cell lymphoma: Results from the international SCHOLAR-1 study. Blood 2017, 130, 1800–1808. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Kaplan–Meier estimated overall survival based on baseline characteristics: (a) age, <65 years vs. ≥65 years; (b) renal function, creatinine clearance (CrCL) ≤ 60 mL/min vs. >60 mL/min; (c) DLBCL subtypes, germinal center (GCB) vs. non-GCB; (d) absolute lymphocyte count (ALC), <1000/µL vs. ≥1000/µL; (e) prior ASCT or transplant ineligible; (f) number of prior lines of therapy, 2 vs. ≥3 prior lines; (g) refractory disease status, progressive disease (PD) < 6 months from first line therapy (primary refractory) vs. PD ≥ 6 months from first line (non-primary refractory); (h) Ann Arbor stage 1 or 2 vs. stage 3 or 4; (i) lactate dehydrogenase (LDH) level, >ULN vs. ≤ULN. Abbreviations: ALC = absolute lymphocyte count, ASCT = autologous stem cell transplant, CrCl = creatinine clearance, DLBCL = diffuse large B-cell lymphoma, GCB = germinal center B-cell, LDH = lactate dehydrogenase, ULN = upper limit of normal.
Figure 1. Kaplan–Meier estimated overall survival based on baseline characteristics: (a) age, <65 years vs. ≥65 years; (b) renal function, creatinine clearance (CrCL) ≤ 60 mL/min vs. >60 mL/min; (c) DLBCL subtypes, germinal center (GCB) vs. non-GCB; (d) absolute lymphocyte count (ALC), <1000/µL vs. ≥1000/µL; (e) prior ASCT or transplant ineligible; (f) number of prior lines of therapy, 2 vs. ≥3 prior lines; (g) refractory disease status, progressive disease (PD) < 6 months from first line therapy (primary refractory) vs. PD ≥ 6 months from first line (non-primary refractory); (h) Ann Arbor stage 1 or 2 vs. stage 3 or 4; (i) lactate dehydrogenase (LDH) level, >ULN vs. ≤ULN. Abbreviations: ALC = absolute lymphocyte count, ASCT = autologous stem cell transplant, CrCl = creatinine clearance, DLBCL = diffuse large B-cell lymphoma, GCB = germinal center B-cell, LDH = lactate dehydrogenase, ULN = upper limit of normal.
Cancers 14 00791 g001
Table 1. Demographics.
Table 1. Demographics.
CharacteristicAgeCrClALC aTransplantStage of DiseaseLDH bTotal
<65 years (n = 52)≥65 years (n = 82)≤60 mL/min
(n = 37)
>60 mL/min
(n = 97)
<1000/µL (n = 71)≥1000/µL
(n = 61)
Prior ASCT (n = 40)Transplant Ineligible
(n = 94)
1 or 2
(n = 33)
3 or 4
(n = 101)
>ULN
(n = 69)
≤ULN
(n = 62)
(N = 134)
Median age, years (min, max)57
(35, 64)
73
(65, 91)
74
(52, 91)
65
(35, 83)
67
(35, 91)
67
(44, 87)
64
(41, 77)
69.5
(35, 91)
70
(35, 87)
67
(41, 91)
65
(35, 86)
69
(41, 91)
67
(35, 91)
≥70, n (%)060
(73.2)
26
(70.3)
34
(35.1)
33
(46.5)
26
(42.6)
13
(32.5)
47
(50.0)
18
(54.5)
42
(41.6)
30
(43.5)
30
(48.4)
60
(44.8)
Male, n (%)32
(61.5)
47
(57.3)
14 (37.8)65
(67.0)
43
(60.6)
35
(57.4)
27
(67.5)
52
(55.3)
15
(45.5)
64
(63.4)
41
(59.4)
37
(59.7)
79
(59.0)
DLBCL subtype,
n (%)
GCB28
(53.8)
35
(42.7)
15 (40.5)48
(49.5)
31
(43.7)
31
(50.8)
25
(62.5)
38
(40.4)
15
(45.5)
48
(47.5)
33
(47.8)
28
(45.2)
63
(47.0)
Non-GCB21
(40.4)
45
(54.9)
21 (56.8)45
(46.4)
39
(54.9)
26
(42.6)
13
(32.5)
53
(56.4)
17
(51.5)
49
(48.5)
35
(50.7)
30
(48.4)
66
(49.3)
Non-classified3
(5.8)
2
(2.4)
1
(2.7)
4
(4.1)
1
(1.4)
4
(6.6)
2
(5.0)
3
(3.2)
1
(3.0)
4
(4.0)
1
(1.4)
4
(6.5)
5
(3.7)
No. prior regimens
Median
(min, max)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2
(2, 5)
2, n (%)30 (57.7)49 (59.8)21 (56.8)58
(59.8)
44
(62.0)
34
(55.7)
21 (52.5)58
(61.7)
19
(57.6)
60
(59.4)
40
(58.0)
37
(59.7)
79
(59.0)
3, n (%)13
(25.0)
20
(24.4)
12 (32.4)21
(21.6)
15
(21.1)
17
(27.9)
14
(35.0)
19 (20.2)8
(24.2)
25
(24.8)
19
(27.5)
13
(21.0)
33
(24.6)
4, n (%)6
(11.5)
10
(12.2)
3
(8.1)
13
(13.4)
9
(12.7)
7
(11.5)
3
(7.5)
13 (13.8)4
(12.1)
12
(11.9)
7
(10.1)
9
(14.5)
16
(11.9)
5, n (%)3
(5.8)
3
(3.7)
1
(2.7)
5
(5.2)
3
(4.2)
3
(4.9)
2
(5.0)
4
(4.3)
2
(6.1)
4
(4.0)
3
(4.3)
3
(4.8)
6
(4.5)
Prior ASCT,
n (%)
32
(61.5)
31
(37.8)
13 (35.1)50
(51.5)
24
(33.8)
37
(60.7)
40
(100)
09
(27.3)
31
(30.7)
17
(24.6)
23
(37.1)
40
(29.9)
Refractory Status c
Primary,
n (%)
22
(42.3)
33
(40.2)
15
(40.5)
40
(41.2)
27
(38.0)
28
(45.9)
11
(27.5)
44
(46.8)
11
(33.3)
44
(43.6)
27
(43.6)
26
(37.7)
55
(41.0)
Non-primary refractory,
n (%)
23
(44.2)
39
(47.6)
16
(43.2)
46
(47.4)
34
(47.9)
26
(42.6)
23
(57.5)
39
(41.5)
17
(51.5)
45
(44.6)
26
(41.9)
36
(52.2)
62
(46.3)
Abbreviations: ALC = absolute lymphocyte count, ASCT = autologous stem cell transplant, CrCl = creatinine clearance, DLBCL = diffuse large B-cell lymphoma, GCB = germinal center B-cell, LDH = lactate dehydrogenase, max = maximum, min = minimum, ULN = upper limit of normal. a Data missing for 2 patients. b Data missing for 3 patients. c Primary refractory is defined as disease progression within 6 months of first-line therapy. Non-primary refractory disease is defined as disease progression ≥6 months after first-line therapy.
Table 2. Efficacy of selinexor based on baseline characteristics in the mITT population.
Table 2. Efficacy of selinexor based on baseline characteristics in the mITT population.
ORR aDOR bPFSOS b
VariableNo. Patientsn (%)p-ValueMedian Monthsp-ValueMedian Monthsp-ValueMedian Monthsp-ValueMultivariate
Analysis
p-Value/HR (95%CI)
Overall13439 (29.1)--9.3--2.6 9.0
Age --
<65 years5219 (36.5)0.199.70.943.60.9113.70.040.03/1.7 (1.05,2.78)
≥65 years8220 (24.4) 9.22.37.8
CrCl
≤60 mL/min3711 (29.7)1.0023.00.243.50.667.80.59
>60 mL/min9728 (28.9) 9.22.39.1
DLBCL Subtype
GCB6320 (31.7)0.45230.393.60.1059.00.836
Non-GCB6616 (24.2)9.32.18.3
Lymphocyte c
<1000/µL7118 (25.4)0.454.90.232.10.137.60.010.07/1.56 (0.97,251)
≥1000/µL6120 (32.8) 233.615.5
Transplant
Prior ASCT4017 (42.5)0.048.40.934.60.1710.90.18
Transplant ineligible9422 (23.4) 9.7 2.17.8
No. Prior Therapies
27922 (27.8)0.8510.40.403.70.359.10.76
≥35517 (30.9)8.42.18.2
Refractory Status d
Primary5512 (21.8)0.1110.40.751.90.026.60.46
Non-primary refractory6223 (37.1)4.93.811.1
Ann Arbor Stage
1 or 23310 (30.3)1.00NR0.0034.00.049.80.91
3 or 410129 (28.7) 4.92.39.0
LDH e
≤ULN6226 (41.9)0.00410.40.983.80.00420.8<0.001<0.001/2.35 (1.45,3.79)
>ULN6912 (17.4) 9.71.95.4
Abbreviations: ASCT = autologous stem cell transplant, CrCl = creatinine clearance, DOR = duration of response, LDH = lactate dehydrogenase, No. = number, NR = Not reached, ORR = overall response rate, OS = overall survival, PFS = progression-free survival, ULN = upper limit of normal. a Comparison was calculated using 2-sided p-value (chi-squared test). b Comparison was calculated using 2-sided p-value (log-rank test). c Data from 2 patients missing. d Primary refractory is defined as disease progression within 6 months of first-line therapy. Non-primary refractory disease is defined as disease progression ≥6 months after first-line therapy. e Data from 3 patients missing. Definitions: Overall response rate is the proportion of patients who achieve a partial response or complete response.
Table 3. Adverse events (≥10%) based on age and transplant status.
Table 3. Adverse events (≥10%) based on age and transplant status.
Adverse EventAge (Years)TransplantTotal
<65≥65Prior ASCTTransplant Ineligible(N = 134)
n (%)n (%)n (%)
Patients with ≥1 AE51 (98.1)81 (98.8)40 (100.0)92 (97.9)132 (98.5)
Thrombocytopenia36 (69.2)46 (56.1)35 (87.5)47 (50.0)82 (61.2)
Nausea28 (53.8)48 (58.5)25 (62.5)51 (54.3)76 (56.7)
Fatigue24 (46.2)39 (47.6)22 (55.0)41 (43.6)63 (47.0)
Anaemia23 (44.2)34 (41.5)19 (47.5)38 (40.4)57 (42.5)
Decreased appetite18 (34.6)31 (37.8)15 (37.5)34 (36.2)49 (36.6)
Diarrhoea15 (28.8)31 (37.8)19 (47.5)27 (28.7)46 (34.3)
Neutropenia18 (34.6)24 (29.3)16 (40.0)26 (27.7)42 (31.3)
Constipation17 (32.7)23 (28.0)16 (40.0)24 (25.5)40 (29.9)
Weight decreased13 (25.0)27 (32.9)12 (30.0)28 (29.8)40 (29.9)
Vomiting11 (21.2)27 (32.9)11 (27.5)27 (28.7)38 (28.4)
Pyrexia12 (23.1)17 (20.7)9 (22.5)20 (21.3)29 (21.6)
Asthenia8 (15.4)20 (24.4)8 (20.0)20 (21.3)28 (20.9)
Cough10 (19.2)14 (17.1)8 (20.0)16 (17.0)24 (17.9)
Dizziness7 (13.5)12 (14.6)5 (12.5)14 (14.9)19 (14.2)
Upper respiratory tract infection10 (19.2)9 (11.0)6 (15.0)13 (13.8)19 (14.2)
Hypotension7 (13.5)10 (12.2)7 (17.5)10 (10.6)17 (12.7)
Oedema peripheral3 (5.8)13 (15.9)5 (12.5)11 (11.7)16 (11.9)
Hyponatraemia5 (9.6)10 (12.2)2 (5.0)13 (13.8)15 (11.2)
Dyspnoea5 (9.6)9 (11.0)3 (7.5)11 (11.7)14 (10.4)
Abbreviations: AE = adverse event, ASCT = autologous stem cell transplant.
Table 4. Summary of selinexor reduction, interruption, and duration.
Table 4. Summary of selinexor reduction, interruption, and duration.
VariableNo. of PatientsDose Reduction
n (%)
Dose Interruption/Withheld
n (%)
Duration of Selinexor Treatment Median Weeks
(min, max)
Overall13465 (48.5%)86 (64.2%)9 (1, 193)
Age
<65 years5226 (50.0%)31 (59.6%)13.5 (1, 193)
≥65 years8239 (47.6%)55 (67.1%)8 (1, 124)
CrCl
≤60 mL/min3717 (45.9)25 (67.6)8 (1, 124)
>60 mL/min9748 (49.5)61 (62.9)9 (1, 193)
DLBCL Subtype
GCB6328 (44.4)40 (63.5)10 (1, 193)
non-GCB6633 (50.0)43 (65.2)8 (1, 183)
Lymphocyte
<1000/µL71239 (40.8)50 (70.4)9 (1, 193)
≥1000/µL6134 (55.7)34 (55.7)9 (1, 183)
Transplant
Prior ASCT4026 (65.0)30 (75.0)16 (1, 183)
Transplant ineligible9439 (41.5)56 (59.6)9 (1, 193)
No. Prior Therapies
27938 (48.1)53 (67.1)9 (1, 193)
≥35527 (49.1)33 (60.0)8 (2, 183)
Refractory Status a
Primary refractory5525 (45.5)35 (63.6)9 (1, 183)
Non-primary refractory6232 (51.6)45 (72.6)11 (1, 124)
Ann Arbor Stage
1 or 23320 (60.6)22 (66.7)9 (1, 183)
3 or 410145 (44.6)64 (63.4)9 (1, 193)
LDH b
≤ULN6237 (59.7)45 (72.6)15 (1, 193)
>ULN6927 (39.1)41 (59.4)6 (1, 95)
Abbreviations: ASCT = autologous stem cell transplant, CrCl = creatinine clearance, GCB = germinal center B-cell; LDH = lactate dehydrogenase, max = maximum, min = minimum, No. = number, ULN = upper limit of normal. a Primary refractory is defined as disease progression within 6 months of first-line therapy. Non-primary refractory disease is defined as disease progression ≥6 months after first-line therapy. b Data missing for 3 patients.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zijlstra, J.M.; Follows, G.; Casasnovas, R.-O.; Vermaat, J.S.P.; Kalakonda, N.; Choquet, S.; Hill, B.; Thieblemont, C.; Cavallo, F.; Cruz, F.D.l.; et al. The Association between Patient Characteristics and the Efficacy and Safety of Selinexor in Diffuse Large B-Cell Lymphoma in the SADAL Study. Cancers 2022, 14, 791. https://doi.org/10.3390/cancers14030791

AMA Style

Zijlstra JM, Follows G, Casasnovas R-O, Vermaat JSP, Kalakonda N, Choquet S, Hill B, Thieblemont C, Cavallo F, Cruz FDl, et al. The Association between Patient Characteristics and the Efficacy and Safety of Selinexor in Diffuse Large B-Cell Lymphoma in the SADAL Study. Cancers. 2022; 14(3):791. https://doi.org/10.3390/cancers14030791

Chicago/Turabian Style

Zijlstra, Josée M., George Follows, Rene-Olivier Casasnovas, Joost S. P. Vermaat, Nagesh Kalakonda, Sylvain Choquet, Brian Hill, Catherine Thieblemont, Federica Cavallo, Fatima De la Cruz, and et al. 2022. "The Association between Patient Characteristics and the Efficacy and Safety of Selinexor in Diffuse Large B-Cell Lymphoma in the SADAL Study" Cancers 14, no. 3: 791. https://doi.org/10.3390/cancers14030791

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