Skip to main content
Erschienen in: Annals of Hematology 1/2019

Open Access 05.07.2018 | Original Article

Multicenter phase 1/2 study of forodesine in patients with relapsed peripheral T cell lymphoma

verfasst von: Dai Maruyama, Kunihiro Tsukasaki, Toshiki Uchida, Yoshinobu Maeda, Hirohiko Shibayama, Hirokazu Nagai, Mitsutoshi Kurosawa, Yoko Suehiro, Kiyohiko Hatake, Kiyoshi Ando, Isao Yoshida, Michihiro Hidaka, Tohru Murayama, Yoko Okitsu, Norifumi Tsukamoto, Masafumi Taniwaki, Junji Suzumiya, Kazuo Tamura, Takahiro Yamauchi, Ryuzo Ueda, Kensei Tobinai

Erschienen in: Annals of Hematology | Ausgabe 1/2019

Abstract

Peripheral T cell lymphomas are an aggressive group of non-Hodgkin lymphomas with poor outcomes for most subtypes and no accepted standard of care for relapsed patients. This study evaluated the efficacy and safety of forodesine, a novel purine nucleoside phosphorylase inhibitor, in patients with relapsed peripheral T cell lymphomas. Patients with histologically confirmed disease, progression after ≥ 1 prior treatment, and an objective response to last treatment received oral forodesine 300 mg twice-daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Forty-eight patients (median age, 69.5 years; median of 2 prior treatments) received forodesine. In phase 1 (n = 3 evaluable), no dose-limiting toxicity was observed during the first 28 days of forodesine treatment. In phase 2 (n = 41 evaluable), the ORR for the primary and final analyses was 22% (90% CI 12–35%) and 25% (90% CI 14–38%), respectively, including four complete responses (10%). Median PFS and OS were 1.9 and 15.6 months, respectively. The most common grade 3/4 adverse events were lymphopenia (96%), leukopenia (42%), and neutropenia (35%). Dose reduction and discontinuation due to adverse events were uncommon. Secondary B cell lymphoma developed in five patients, of whom four were positive for Epstein-Barr virus. In conclusion, forodesine has single-agent activity within the range of approved therapies in relapsed peripheral T cell lymphomas, with a manageable safety profile, and may represent a viable treatment option for this difficult-to-treat population.

Introduction

Peripheral T cell lymphomas (PTCLs) are an aggressive group of non-Hodgkin lymphomas (NHLs), accounting for 5–10% of all NHLs in Western countries and approximately 20% of NHLs in Japan [1, 2]. The most common PTCL subtypes are PTCL, not otherwise specified (PTCL-NOS); anaplastic large cell lymphoma (ALCL); and angioimmunoblastic T cell lymphoma (AITL) [3, 4]. Newly diagnosed PTCL is most frequently treated with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like chemotherapies [58]. Although most patients respond initially, outcomes remain poor except in patients with anaplastic lymphoma kinase (ALK)-positive ALCL [5]. Median survival in a cohort of 153 North American patients with first-relapsed/refractory PTCLs was 5.5 months, underscoring the need for new treatments in this setting [9].
Prior to forodesine, several agents were approved for treatment of relapsed/refractory PTCL, including the folate antagonist pralatrexate [10] and the histone deacetylase inhibitors romidepsin [11] and belinostat [12] in the USA, and chidamide in China [13]. Additionally, the anti-CC chemokine receptor 4 monoclonal antibody mogamulizumab was approved in Japan for treatment of anti-CC chemokine receptor 4-positive relapsed/refractory PTCL [14], and the anti-CD30 antibody-drug conjugate brentuximab vedotin was approved for treatment of relapsed/refractory ALCL [15, 16]. Objective response rates (ORRs) for these agents in relapsed/refractory PTCL generally ranged from 25 to 35% [1014], with brentuximab vedotin producing an ORR of 86% in ALCL [15] but 21% in non-ALCL PTCL [16].
Children born deficient in purine nucleoside phosphorylase (PNP) have reduced T cell counts, suggesting that PNP may be a target for treatment of T cell-mediated diseases [17]. Forodesine (BCX1777, Immucillin-H) is a novel PNP inhibitor, 100 to 1000 times more potent than other agents of this class [18, 19]. By inhibiting PNP, forodesine augments 2′-deoxyguanosine (dGuo) levels in plasma and T cells. The enzyme 2′-deoxycytidine kinase, which is highly upregulated in malignant T cells, phosphorylates dGuo to form deoxyguanosine monophosphate and then 2′-deoxyguanosine triphosphate. Accumulation of 2′-deoxyguanosine triphosphate causes an imbalance of the deoxyribonucleotide pool leading to T cell apoptosis. Forodesine was tolerated at doses of 200–300 mg once daily and exhibited preliminary evidence of anti-tumor efficacy in patients with PTCLs or cutaneous TCLs [19, 20]. Pharmacokinetic/pharmacodynamic data in healthy volunteers (data on file) suggested that a regimen of 300 mg twice-daily would be tolerable and provide forodesine exposure necessary for improving efficacy. Therefore, we conducted a phase 1/2 study of forodesine 300 mg twice-daily in Japanese patients with relapsed PTCL.

Patients and methods

Study design

This multicenter, open-label study was conducted at 21 sites in Japan from January 2013 to February 2017. The study consisted of two parts: a phase 1 component designed to confirm the safety and tolerability of forodesine 300 mg twice-daily for 28 days in patients with relapsed/refractory PTCL and a phase 2 component designed to evaluate the efficacy and safety of this forodesine regimen in relapsed PTCL. The study was conducted in accordance with ethical principles of the Declaration of Helsinki and in compliance with International Council for Harmonization guidelines for Good Clinical Practice. The Institutional Review Boards of all participating institutions approved the study protocol, and all patients provided written informed consent. The study was registered at ClinicalTrials.​gov (NCT01776411).
In phase 1, forodesine 300 mg (three 100-mg capsules) was given twice-daily after meals in a 28-day cycle. Phase 2 was initiated because none of the first three patients completing the 28-day cycle had dose-limiting toxicity (DLT; defined as treatment-related grade 3/4 non-hematologic toxicity excluding nausea, vomiting, or diarrhea or grade 4 neutropenia or thrombocytopenia lasting ≥ 7 days). During phase 2, patients received forodesine until disease progression (PD), unacceptable toxicity, or withdrawal of consent. Forodesine was stopped temporarily for ≤ 2 weeks in the event of DLT or if needed for management of adverse events (AEs). After recovery, a single-dose reduction to 200 mg twice-daily was allowed.
Patients had study visits on days 1 and 15 of cycles 1–4 and day 1 of subsequent cycles. After 22 patients completed 2 cycles, an interim efficacy analysis was conducted using a Simon minimax 2-step design to assess for futility (≤ 2 patients with objective responses); the study was to be terminated if futility was demonstrated. If futility was not demonstrated, the study was to be continued. Data cutoff was to be conducted when all the patients for efficacy evaluation completed the clinical study procedure by week 24. Based on data obtained until that time point, the primary analysis was to be conducted.

Patients

Japanese patients aged ≥ 20 years with histopathologically confirmed PTCL were eligible if they had PD after ≥ 1 prior treatment and had achieved an objective response on their last treatment. PTCL was defined according to the 2008 WHO Classification [3] and included PTCL-NOS, AITL, ALCL (ALK-positive or ALK-negative), extranodal natural killer cell/TCL (nasal type), enteropathy-associated TCL, hepatosplenic TCL, subcutaneous panniculitis-like TCL, and transformed mycosis fungoides. The PTCL subtype was diagnosed in each institution from lesion biopsy specimens and confirmed by an Independent Pathology Review Committee. Eligible patients had an enlarged lymph node or extranodal mass that was measurable in two perpendicular directions by computed tomography, with the greatest diameter > 1.5 cm; Eastern Cooperative Oncology Group performance status 0 or 1; and adequate hematopoietic, liver, and kidney function.
Patients who had received chemotherapy, radiation therapy, or high-dose corticosteroids (prednisolone ≥ 10 mg/day or equivalent) ≤ 21 days before the first dose of study drug were excluded, as were patients with a history of central nervous system involvement, allogeneic hematopoietic stem cell transplantation, autologous hematopoietic stem cell transplantation ≤ 100 days before study drug, severe cardiovascular or pulmonary disease, uncontrolled diabetes, or positivity for hepatitis B virus surface antigen, anti-hepatitis C virus antibody, or anti-human immunodeficiency virus antibody. Pregnant and lactating women and patients of child-bearing potential unwilling to use adequate contraception were also excluded.

Assessments

Tumor assessments were conducted after every 2 cycles for the first 24 weeks and then every 4 cycles. Efficacy was evaluated by an Independent Efficacy Assessment Committee (IEAC) according to revised International Working Group criteria [21], and classified as complete response (CR), partial response (PR), stable disease, or PD. The primary efficacy endpoint was IEAC-assessed ORR, consisting of the proportion of evaluable patients with CR or PR. Secondary efficacy endpoints included investigator-assessed ORR, duration of response (DoR), time to treatment failure, progression-free survival (PFS), and overall survival (OS). Primary analyses were conducted on data obtained by the time of data cutoff. In addition, final analyses on data from the entire period (including after the data cutoff) were also described for information purposes. For analyses other than the primary endpoint, results of final analyses were described.

Safety assessment

Safety was evaluated throughout the study and was comprised of AE monitoring, laboratory testing, physical examinations, vital-sign measurements, and 12-lead electrocardiograms. Severity of AEs was graded using the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0.

Pharmacokinetics

The first seven patients comprised the full pharmacokinetics set; they received in-patient treatment for the first 4 days and had an additional visit on day 8. Blood samples were collected predose and at 1, 2, 4, 6, 8, and 12 h after the first dose on both days 1 and 15 and also predose on days 2, 3, 4, 8, 29, and 57. In all other patients, blood samples were collected predose for the first dose on days 1, 15, 29, and 57. Plasma forodesine concentrations were measured using a validated liquid chromatography-tandem mass spectrometry (LC/MS/MS) method, with a lower limit of quantitation of 2.5 ng/mL. Key pharmacokinetic parameters, including the observed maximum concentration (Cmax) and the area under the concentration-time curve to the last measurable drug concentration (AUClast), were determined by non-compartmental analysis using WinNonlin. Additional blood samples were collected at the above times for measurement of plasma dGuo concentrations by a validated LC/MS/MS with a lower limit of quantitation of 5.0 ng/mL.

Statistical analysis

Efficacy was evaluated in the full analysis set consisting of patients who received ≥ 1 dose of forodesine and had a postbaseline efficacy assessment. Using the Simon minimax two-step design [22], a sample size of 40 evaluable patients in phase 2 would have 80% statistical power at a one-sided α of 0.05 for showing a threshold ORR of 10%, assuming an expected ORR of 25%. A one-sided binomial test was used to determine if the observed ORR was above the predefined 10% threshold rate. To account for potential non-evaluable patients, the target sample size in phase 2 was set at 43. Time-to-event parameters were evaluated using Kaplan-Meier methods [23]. Safety was assessed in all patients who received ≥ 1 dose of study drug. AEs were coded to the Medical Dictionary for Regulatory Activities–Japanese, version 18.1. Safety parameters were summarized using descriptive statistics.

Results

Patients

Forty-eight patients received forodesine (4 in phase 1, 44 in phase 2) (Fig. 1). Overall, the most common reasons for discontinuation were PD (n = 35) and AEs (n = 8). The study cohort had a median age of 69.5 years (range, 32–79 years) and had received a median of two prior treatment regimens (range, 1–9); most had PTCL-NOS (46%) or AITL (40%) (Table 1).
Table 1
Demographics and baseline characteristics
Characteristic
Phase 1 (n = 4)
Phase 2 (n = 44)
Total (n = 48)
Age, years, median (range)
72.5 (42–76)
69.0 (32–79)
69.5 (32–79)
Sex, n (%)
 Male
1 (25)
30 (68)
31 (65)
 Female
3 (75)
14 (32)
17 (35)
ECOG PS, n (%)
 0
2 (50)
27 (61)
29 (60)
 1
2 (50)
17 (39)
19 (40)
Disease classification, n (%)
 Peripheral T cell lymphoma, NOS
1 (25)
21 (48)
22 (46)
 Angioimmunoblastic T cell lymphoma
2 (50)
17 (39)
19 (40)
 Anaplastic large cell lymphoma
  ALK-positive
1 (25)
0 (0)
1 (2)
  ALK-negative
0 (0)
2 (5)
2 (4)
 Extranodal T cell/NK cell lymphoma, nasal type
0 (0)
1 (2)
1 (2)
 Transformed mycosis fungoides
0 (0)
1 (2)
1 (2)
 Othera
0 (0)
2 (5)
2 (4)
Ann Arbor classification,bn (%)
 Stage I
0 (0)
1 (2)
1 (2)
 Stage II
3 (75)
9 (20)
12 (25)
 Stage III
0 (0)
20 (45)
20 (42)
 Stage IV
1 (25)
13 (30)
14 (29)
LDH (baseline)
 Low/normal
2(50.0)
24 (55)
26 (54)
 High
2(50.0)
20 (45)
22(46)
Prior treatment regimens
 Median (range)
2 (1–9)
2 (1–6)
2 (1–9)
 Chemotherapy, n (%)
4 (100)
44 (100)
48 (100)
 ASCT, n (%)
0 (0)
3 (7)
3 (6)
 Radiation therapy, n (%)
1 (25)
5 (11)
6 (13)
 Monoclonal antibody, n (%)
1 (25)
3 (7)
4 (8)
 Corticosteroid, n (%)
1 (25)
2 (5)
3 (6)
Response to most recent treatment regimen, n (%)
 CR/CRu
3 (75)
20 (45)
23 (48)
 PR
1 (25)
24 (55)
25 (52)
ALK anaplastic lymphoma kinase, ASCT autologous hematopoietic stem cell transplant, CR complete response, CRu unconfirmed complete response, ECOG PS Eastern Cooperative Oncology Group performance status, LDH lactate dehydrogenase, NK cell natural killer cell, NOS not otherwise specified, PR partial response, SD standard deviation
aIncludes two cases judged to be plasmablastic lymphoma and follicular dendritic cell sarcoma, respectively, on the independent central pathology review
bClassification for PTCLs other than transformed mycosis fungiodes. The case of transformed mycosis fungiodes was stage IV by the ISCL-EORTC classification
Patients received forodesine for a median of 2.1 months (range, 0.2–36.0 months). Seventeen patients (35%) had a delay in forodesine dosing because of AEs, but only one patient (2%) had a dose reduction to 200 mg twice-daily (because of pneumonia). The mean daily dose of forodesine was 586.7 mg (standard deviation ± 37.2 mg).

Safety

Lymphopenia occurred in all patients (grade 3/4 in 46 patients [96%]), with all evaluated lymphocyte subsets (CD3+, CD4+, CD8+, CD16+, CD20+, CD56+) showing reductions from baseline (Fig. 2). Other common grade 3/4 hematologic toxicities included leukopenia (42%), neutropenia (35%), and thrombocytopenia (25%; Table 2). Febrile neutropenia occurred in six patients (13%). Grade 3/4 non-hematologic toxicities were uncommon. Adverse events that resulted in discontinuation occurred in 11 patients (23%; only Epstein-Barr virus [EBV]-associated lymphoma (n = 2) led to discontinuation in > 1 patient). One patient in phase 2 died from disseminated intravascular coagulation and multiorgan failure, which was attributed to underlying disease and considered not related to forodesine.
Table 2
Adverse events regardless of causality occurring in ≥ 10% of patients
Adverse event
All grades, n (%)
Grade 3/4, n (%)
Hematologic toxicities
 Lymphopenia
48 (100)
46 (96)
 Leukopenia
35 (73)
20 (42)
 Neutropenia
27 (56)
17 (35)
 Thrombocytopenia
23 (48)
12 (25)
 Anemia
23 (48)
11 (23)
 Febrile neutropenia
6 (13)
6 (13)
Non-hematologic toxicities
 Constipation
13 (27)
1 (2)
 Nasopharyngitis
13 (27)
0 (0)
 Hypoalbuminemia
12 (25)
2 (4)
 Edema
12 (25)
0 (0)
 Pyrexia
11 (23)
1 (2)
 Stomatitis
11 (23)
1 (2)
 Headache
11 (23)
0 (0)
 Insomnia
10 (21)
0 (0)
 Alanine aminotransferase increased
10 (21)
1 (2)
 Nausea
9 (19)
1 (2)
 Decreased appetite
9 (19)
4 (8)
 Malaise
8 (17)
0 (0)
 Aspartate aminotransferase increased
8 (17)
0 (0)
 Rash
8 (17)
0 (0)
 Abnormal hepatic function
7 (15)
3 (6)
 Herpes zoster
7 (15)
1 (2)
 Pruritus
7 (15)
0 (0)
 Protein in urine
7 (15)
0 (0)
 Vomiting
6 (13)
0 (0)
 Cytomegalovirus infection
5 (10)
0 (0)
 Pneumonia
5 (10)
4 (8)
 Hyponatremia
5 (10)
4 (8)
 Decreased weight
5 (10)
1 (2)
Twenty-two patients (46%) experienced serious AEs, most commonly infections (n = 8). Pneumonia (n = 4) was the only serious infection reported in > 1 patient. Secondary B cell lymphoma was reported in five patients; all were women aged 65 to 75 years, and three entered the study with AITL and two with PTCL-NOS. In four of five patients, lymphoma cells were positive for EBV encoded RNA-1 on in situ hybridization. Based on an exploratory analysis, the patients who developed secondary B cell lymphoma had received forodesine for a median of 11.6 months (range, 2.2–16.6 months); the median duration from forodesine initiation to development of secondary B cell lymphoma was 14.3 months (range, 6.7–16.6 months). Median trough lymphocyte counts in patients who did (n = 5) or did not (n = 43) develop secondary B cell lymphoma were 87 per mm3 (range, 51–120) and 71 per mm3 (range, 0–731), respectively. Median trough CD4+ lymphocyte counts in patients who did (n = 5) or did not (n = 42) develop secondary B cell lymphoma were 44 per mm3 (range, 18–162) and 51/mm3 (range, 5–3274), respectively. Outcome for patients who developed secondary B cell lymphoma are shown in Table 3. One patient achieved CR to treatment for secondary B cell lymphoma and survived with PTCL at the time of the final analysis. Three patients died of lymphoma (PTCL and/or secondary B cell lymphoma), and the outcome of one patient is unknown.
Table 3
Outcomes for patients who developed secondary B cell lymphoma
Disease classification
Sex
Agea
Duration of forodesine administration (days)
Duration to development of sBCLb (days)
Treatments for sBCL/response
Outcome
AITL
F
71
447
450
R-DeVIC/CR
Alive with AITL
AITL
F
70
171
203
R-CHOP/PD
Died from lymphoma (AITL, sBCL)
AITL
F
76
67
436
PSL/unknown
Died from lymphoma (AITL, sBCL)
PTCL, NOS
F
72
353
281
PSL/unknown
unknown
PTCL, NOS
F
65
505
506
R-COP/PD R-CHOP/PD DEX/PD
Died from lymphoma (PTCL, NOS, sBCL)
AITL angioimmunoblastic T cell lymphoma; COP rituximab plus cyclophosphamide, vincristine, prednisone; CR complete response; F female; PD progression disease; PSL prednisolone; PTCL peripheral T cell lymphoma; R-CHOP rituximab plus cyclophosphamide, doxorubicin, vincristine, prednisone; R-DeVIC rituximab plus dexamethasone, etoposide, ifosfamide, carboplatin; R-DEX dexamethasone; R-GDP rituximab plus gemcitabine, dexamethasone, cisplatin; sBCL secondary B cell lymphoma
aAge at the time of informed consent
bDuration from initial forodesine administration to development of sBCL

Efficacy

Among the 41 evaluable patients in phase 2, the ORR (IEAC assessment) for the primary analysis was 22% (90% CI 12–35%), and included four with CR (10%) and five with PR (12%) (Table 4), which was significantly greater than the predefined 10% threshold rate (P = 0.018). The investigator-assessed ORR was 22%, and included three patients with CR and six with PR. One of four patients in phase 1 also achieved a PR. Furthermore, the ORR at the final analysis was 25% (90% CI 14–38%), and included four patients with CR (10%) and six with PR (15%) (Table 4). The median time to response was 2.8 months (range, 1.8–12.8 months), and the median time to treatment failure was 2.2 months (95% CI 1.8–5.0 months) (data not shown). Median DoR was 10.4 months (95% CI 5.9–16.0 months) (Fig. 3a), median PFS was 1.9 months (95% CI 1.8–4.6 months) (Fig. 3b), and median OS was 15.6 months (95% CI 10.7–NE months) (Fig. 3c) among evaluable patients. Two-year OS was 39%. Compared with non-responders, the hazard ratios for PFS and OS among responders were 0.21 (95% CI 0.09–0.48) and 0.19 (95% CI 0.04–0.80), respectively (Fig. 4).
Table 4
Objective response rate by IEAC assessment
Response, n (%)
Phase 2 (n = 41)
Phase 1 + 2 (n = 45)
Primary analysis
 DCR (CR + PR+ SD)
16 (39)
17 (38)
 ORR (CR + PR) 90% CI
9 (22)a
12–35
10 (22)
13–35
 CR
4 (10)
4 (9)
 PR
5 (12)
6 (13)
 SD
7 (17)
7 (16)
 PD/RD
24 (59)
26 (58)
 Not evaluable
1 (2)
2 (4)
Final analysis
 DCR (CR + PR+ SD)
16 (39)
17 (38)
 ORR (CR + PR) 90% CI
10 (25)a
14–38
11 (24)
14–37
 CR
4 (10)
4 (9)
 PR
6 (15)
7 (16)
 SD
6 (15)
6 (13)
 PD/RD
24 (59)
26 (58)
 Not evaluable
1 (2)
2 (4)
CR complete response, DCR disease control rate, IEAC Independent Efficacy Assessment Committee, ORR objective response rate, PD progressive disease, PR partial response, RD relapsed disease, SD stable disease
aUniformly minimum variance unbiased estimator (UMVUE)
For the major PTCL subtypes in the phase 1 and 2 cohorts combined, the ORR was 33% (95% CI 13–59%) among 18 evaluable patients with AITL and 23% (95% CI 8–45%) among 22 evaluable patients with PTCL-NOS. ORRs > 30% were observed in several predefined subgroups, including age < 65 years (6/16; 38%), two prior treatment regimens (3/9; 33%), stage III disease (7/19; 37%), and low/normal lactate dehydrogenase (9/25; 36%) (Table 5). In general, patients with CR and PR showed a progressive reduction in target tumor size over time after starting forodesine (Fig. 5).
Table 5
Subgroup analysis of objective response rate, full analysis set
Subgroup
n/Na
ORR (95% CI), %
All evaluable patients in phases 1 + 2
11/45
24 (13–40)
Sex
 Men
8/30
27 (12–46)
 Women
3/15
20 (4–48)
Age group
 < 65 years
6/16
38 (15–65)
 ≥ 65 years
5/29
17 (6–36)
No. of previous anti-tumor regimens
 1
5/19
26 (9–51)
 2
3/9
33 (8–70)
 3
3/10
30 (7–65)
 ≥ 4
0/7
0 (0–41)
Histological classification
 Peripheral T cell lymphoma, NOS
5/22
23 (8–45)
 Angioimmunoblastic T cell lymphoma
6/18
33 (13–59)
Ann Arbor stageb
 Stage I
0/1
0 (0–98)
 Stage II
2/11
18 (2–52)
 Stage III
7/19
37 (16–62)
 Stage IV
2/13
15 (2–45)
ECOG performance status
 0
7/27
26 (11–46)
 1
4/18
22 (6–48)
Target lesion SPD
 < 14 cm2
8/26
31 (14–52)
 ≥ 14 cm2
3/19
16 (3–40)
LDH
 Low/normal
9/25
36 (18–58)
 High
2/20
10 (1–32)
CI confidence interval, ECOG Eastern Cooperative Oncology Group, LDH lactate dehydrogenase, NOS not otherwise specified, ORR objective response rate, SPD sum of the products of the largest diameters of target lesions
aNumber of patients with objective responses divided by the total number of patients in the category
bExcept for transformed mycosis fungoides

Pharmacokinetics

Plasma forodesine concentrations increased over 4 h after the first dose (mean [± standard deviation] Cmax, 435.7 [± 152.9] ng/mL) and then decreased gradually (Fig. 6). On day 15, the mean pretreatment concentration was 509.7 ng/mL (± 180.4) and after dosing, again increased over 4 h to a mean of 683.1 ng/mL (± 162.9) before gradually decreasing. Throughout the treatment period, mean trough plasma forodesine concentrations at each time point remained within a range of 460 to 540 ng/mL. The AUClast values after dosing on days 1 and 15 were 3540 and 6520 ng h/mL, indicating an accumulation ratio of 1.8. Plasma dGuo levels increased over 8 h after forodesine dosing, with mean trough concentrations at each time point within a range of 551 to 840 ng/mL. Plasma forodesine and dGuo concentrations on days 1 and 15 showed a positive relationship according to a maximum drug effect model.

Discussion

This study demonstrated that forodesine has promising single-agent activity and led to its approval in Japan for treatment of relapsed/refractory PTCL. The ORR (22–25%) is comparable with ORRs reported in phase 2 studies of several recently approved agents for PTCL, including pralatrexate, romidepsin, and belinostat [1012]. Differences in patient populations, histopathologic subtype distributions, disease status, and pretreatment characteristics make comparisons across studies difficult. Our study cohort mostly consisted of patients with PTCL-NOS and AITL; the ORR was numerically higher for those with AITL (33%) than for those with PTCL-NOS (23%). In a recent phase 2 study of patients with relapsed/refractory PTCL, lenalidomide demonstrated an ORR of 22%, and also showed a higher ORR in the AITL subset (31%) [24]. Given etiologic differences underlying the various PTCL histologies, it is plausible that specific agents may be used preferentially for specific PTCL subtypes. Indeed, brentuximab vedotin is highly active in patients with relapsed/refractory CD30+ ALCL but has less activity against other PTCL subtypes [15, 16]. Gemcitabine also showed promising activity in a small study of patients with PTCL-NOS and mycosis fungoides [25].
The median time to objective response was 2.8 months, and responses to forodesine appeared durable. In the final data analysis, the ORR was 25% (one patient reached PR after 13 months of administration) and median DoR was 10.4 months (range, 5.9–16.0 months).
The safety profile of forodesine 300 mg twice-daily was acceptable. Although dose delays because of AEs occurred in 35% of patients, dose reduction was only needed in 1 patient (2%), and discontinuation due to AEs occurred in 11 patients (23%). Toxicity consisted mostly of lymphopenia and other hematologic AEs; non-hematologic toxicities were generally mild/moderate in severity. The high rate of lymphopenia is thought to reflect the mechanism of action of forodesine. By inhibiting PNP, forodesine induces lymphocyte apoptosis (mainly T cells), leading to a reduction in lymphocyte counts and causing an immunosuppressive effect that may result in an increased risk of infection and secondary B cell lymphoma.
In this study, five patients developed secondary B cell lymphoma, of whom three had AITL and two had PTCL-NOS, consistent with the general distribution in our study cohort. EBV-driven B cell lymphoproliferation and EBV-related B cell lymphoma secondary to immunosuppression have been reported in patients with AITL and PTCL-NOS [26, 27]. Clonal expansion of EBV-negative B cells has also been described in patients with PTCLs [28, 29]. EBV status was not assessed at enrollment in our study; patients were not treated with anti-viral agents as prophylaxis, and all patients had received prior immunosuppressive chemotherapy. Thus, it cannot be excluded that the secondary lymphomas were already evolving before forodesine initiation. No clear difference was observed between total lymphocyte and CD4+ lymphocyte counts for patients who did or did not develop secondary B cell lymphoma, and risk factors for development of secondary B cell lymphoma were not identified in this study. Future studies should investigate whether EBV status at baseline or during forodesine treatment influences risk of secondary B cell lymphoma. In addition, sufficient attention must be placed on risk of opportunistic infection given that forodesine’s mechanism of action leads to T cell reductions.
In conclusion, forodesine has clinically meaningful single-agent activity, with durable responses, and a manageable safety profile in patients with relapsed PTCL. Compared with PTCL options that require intravenous infusion with frequent or prolonged clinic visits, the oral formulation makes forodesine easier to administer and, in turn, may be more convenient and less burdensome to patients. New therapeutic strategies with forodesine, including combination therapy, are being considered.

Acknowledgements

The authors thank the patients, their families, doctors, nurses, and staff members who participated in this multicenter trial. We thank trial investigator Dr. Yasunobu Abe (Kyushu Cancer Center) who passed away prior to submission of this publication. We also thank Dr. Takashi Terauchi (National Cancer Center Hospital), Dr. Ukihide Tateishi (Yokohama City University Hospital), and Mitsuaki Tatsumi (Osaka University Hospital) who served as members of Image Evaluation Committee; Dr. Yoshihiro Matsuno (Hokkaido University Hospital), Shigeo Nakamura (Nagoya University Hospital), and Koichi Oshima (Kurume University Hospital) who served as members of central pathology review; and Dr. Noboru Horikoshi (Juntendo University) and Dr. Noriko Usui (Jikei University Daisan Hospital) who served as members of the Independent Data Monitoring Committee. Medical writing and editorial assistance was provided by Team 9 Science.

Compliance with ethical standards

Conflict of interest

Dai Maruyama reports research funding from Mundipharma K.K., Chugai Pharma, Kyowa Hakko Kirin, Ono Pharmaceutical, Celgene, Janssen, GlaxoSmithKline, Eisai, Takeda, SERVIER, and Abbvie, MSD, and receiving honoraria from Mundipharma K.K., Takeda, Janssen, Eisai, Biomedis International, Celgene, Sanofi, Kyowa Hakko Kirin, Fujifilm, Mochida Pharmaceutical Co., Ltd., Ono Pharmaceutical, and Chugai Pharma. Toshiki Uchida reports receiving an honorarium from Janssen. Yoshinobu Maeda reports research funding from Chugai Pharma, Kyowa Hakko Kirin, and Ono Pharmaceutical, and an honorarium from Mundipharma K.K. Hirokazu Nagai reports research funding from Chugai Pharma, Mundipharma K.K., Eisai, Sanofi K.K., Janssen, Takeda, Ono Pharmaceutical, and Kyowa Hakko Kirin. Hirohiko Shibayama reports research funding from Takeda, Janssen, Ono Pharmaceutical, Astellas, Teijin Pharma, Shionogi, Taiho, and Mundipharma KK, and honoraria from Takeda, Celgene, Novartis, Janssen, Kyowa Hakko Kirin, and Mundipharma KK. Kiyoshi Ando reports research funding from Mundipharma K.K. Isao Yoshida reports research funding from Kyowa Hakko Kirin and Chugai Pharma, and receiving honoraria from Mundipharma K.K., Celgene, and Kyowa Hakko Kirin. Michihiro Hidaka reports research funding from Mundipharma K.K. and Chugai Pharma. Tohru Murayama reports research funding from Celltrion, and honoraria from Nippon Shinyaku, Taiho Pharma, Janssen, Siemens, Kyowa Hakko Kirin, Novartis, Celgene, Ono Pharmaceutical, Pfizer, and Bristol-Myers Squibb. Junji Suzumiya reports research funding from Kyowa Hakko Kirin, Chugai-Roche, Shinnipponkagaku-PPD, Astellas, Toyama Chemical, Eisai, Takeda, Sumitomo Dainippon, Shionogi, Taiho, Yakult, and SymBio, and receiving honoraria from Kyowa Hakko Kirin, Chugai-Roche, Janssen, Eisai, Takeda, Sumitomo Dainippon, Otsuka, Celgene, Alexion, Novartis, Pfizer, AstraZeneca, Bristol-Myers Squibb, Merck, Ono Pharmaceutical, Eli Lilly, Shire, Gilead, and Zenyaku. Kazuo Tamura reports receiving honoraria from Ono Pharmaceutical, Kyowa Hakko Kirin, and Eli Lilly. Ryuzo Ueda reports research funding and an honorarium from Kyowa Hakko Kirin, serving as a consultant for Mundipharma K.K. and Terumo, and receiving an honorarium from Chugai Pharma. Kensei Tobinai reports receiving research funding from Mundipharma K.K., Chugai Pharma, Kyowa Hakko Kirin, Ono Pharmaceutical, Celgene, Janssen, GlaxoSmithKline, Eisai, Takeda, Servier, and Abbvie, and honoraria from Zenyaku Kogyo, Eisai, Takeda, Mundipharma K.K., Janssen, HUYA Bioscience International, Kyowa Hakko Kirin, Celgene, Chugai Pharma, and Ono Pharmaceutical. All remaining authors declared that they have no conflicts of interest.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declarations and its later amendments or comparable ethical standards.
Informed consent was obtained from all individual participants included in this study.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Anderson JR, Armitage JO, Weisenburger DD, for the Non-Hodgkin’s Lymphoma Classification Project (1998) Epidemiology of the non-Hodgkin’s lymphomas: distributions of the major subtypes differ by geographic locations. Ann Oncol 9:717–720CrossRefPubMed Anderson JR, Armitage JO, Weisenburger DD, for the Non-Hodgkin’s Lymphoma Classification Project (1998) Epidemiology of the non-Hodgkin’s lymphomas: distributions of the major subtypes differ by geographic locations. Ann Oncol 9:717–720CrossRefPubMed
2.
Zurück zum Zitat Chihara D, Ito H, Matsuda T, Shibata A, Katsumi A, Nakamura S, Tomotaka S, Morton LM, Weisenburger DD, Matsuo K (2014) Differences in incidence and trends of haematological malignancies in Japan and the United States. Br J Haematol 164:536–545CrossRefPubMed Chihara D, Ito H, Matsuda T, Shibata A, Katsumi A, Nakamura S, Tomotaka S, Morton LM, Weisenburger DD, Matsuo K (2014) Differences in incidence and trends of haematological malignancies in Japan and the United States. Br J Haematol 164:536–545CrossRefPubMed
3.
Zurück zum Zitat Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW (2008) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue. International Agency for Research on Cancer (IARC), Lyon Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW (2008) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue. International Agency for Research on Cancer (IARC), Lyon
4.
Zurück zum Zitat Campo E, Swerdlow SH, Harris NL, Pileri S, Stein H, Jaffe ES (2011) The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood 117:5019–5032CrossRefPubMedPubMedCentral Campo E, Swerdlow SH, Harris NL, Pileri S, Stein H, Jaffe ES (2011) The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood 117:5019–5032CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat d’Amore F, Gaulard P, Trümper L, Corradini P, Kim WS, Specht L, Bjerregaard Pedersen M, Ladeto M, on behalf of the ESMO Guidelines Committee (2015) Peripheral T-cell lymphomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 26(suppl 5):v108–v115CrossRefPubMed d’Amore F, Gaulard P, Trümper L, Corradini P, Kim WS, Specht L, Bjerregaard Pedersen M, Ladeto M, on behalf of the ESMO Guidelines Committee (2015) Peripheral T-cell lymphomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 26(suppl 5):v108–v115CrossRefPubMed
7.
Zurück zum Zitat Van Obbergh F, Van Hoof A, Verhoef G, Dierickx D, De Wilde V, Offner F, Bron D, Sonet A, André M, Janssens A, Bonnet C, Deprijck B, Zachée P, Kentos A, Schroyens W, Van Den Neste E (2013) Treatment of peripheral T-cell lymphomas: recommendations of the Belgian hematological society (BHS). Belg J Hematol 4:90–101 Van Obbergh F, Van Hoof A, Verhoef G, Dierickx D, De Wilde V, Offner F, Bron D, Sonet A, André M, Janssens A, Bonnet C, Deprijck B, Zachée P, Kentos A, Schroyens W, Van Den Neste E (2013) Treatment of peripheral T-cell lymphomas: recommendations of the Belgian hematological society (BHS). Belg J Hematol 4:90–101
8.
Zurück zum Zitat Kitahara H, Maruyama D, Maeshima AM, Makita S, Miyamoto KI, Fukuhara S, Munakata W, Suzuki T, Kobayashi Y, Tajima K, Terauchi T, Kurihara H, Taiguchi H, Komatsu N, Tobinai K (2017) Prognosis of patients with peripheral T cell lymphoma who achieve complete response after CHOP/CHOP-like chemotherapy without autologous stem cell transplantation as an initial treatment. Ann Hematol 96:411–420CrossRefPubMed Kitahara H, Maruyama D, Maeshima AM, Makita S, Miyamoto KI, Fukuhara S, Munakata W, Suzuki T, Kobayashi Y, Tajima K, Terauchi T, Kurihara H, Taiguchi H, Komatsu N, Tobinai K (2017) Prognosis of patients with peripheral T cell lymphoma who achieve complete response after CHOP/CHOP-like chemotherapy without autologous stem cell transplantation as an initial treatment. Ann Hematol 96:411–420CrossRefPubMed
9.
Zurück zum Zitat Mak V, Hamm J, Chhanabhai M, Shenkier T, Klasa R, Sehn LH, Villa D, Gascoyne RD, Connors JM, Savage KJ (2013) Survival of patients with peripheral T-cell lymphoma after first relapse or progression: spectrum of disease and rare long-term survivors. J Clin Oncol 31:1970–1976CrossRefPubMed Mak V, Hamm J, Chhanabhai M, Shenkier T, Klasa R, Sehn LH, Villa D, Gascoyne RD, Connors JM, Savage KJ (2013) Survival of patients with peripheral T-cell lymphoma after first relapse or progression: spectrum of disease and rare long-term survivors. J Clin Oncol 31:1970–1976CrossRefPubMed
10.
Zurück zum Zitat O’Connor OA, Pro B, Pinter-Brown L, Bartlett N, Popplewell L, Coiffier B, Lechowicz MJ, Savage KJ, Shustov AR, Gisselbrecht C, Jacobsen E, Zinzani PL, Furman R, Goy A, Haioun C, Crump M, Zain JM, His E, Boyd A, Horwitz S (2011) Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol 29:1182–1189CrossRefPubMedPubMedCentral O’Connor OA, Pro B, Pinter-Brown L, Bartlett N, Popplewell L, Coiffier B, Lechowicz MJ, Savage KJ, Shustov AR, Gisselbrecht C, Jacobsen E, Zinzani PL, Furman R, Goy A, Haioun C, Crump M, Zain JM, His E, Boyd A, Horwitz S (2011) Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol 29:1182–1189CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Coiffier B, Pro B, Prince HM, Foss F, Sokol L, Greenwood M, Caballero D, Borchmann P, Morschhauser F, Wilhelm M, Pinter-Brown L, Padmanabhan S, Shustov A, Nichols J, Carroll S, Balser J, Balser B, Horwitz S (2012) Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol 30:631–636CrossRefPubMed Coiffier B, Pro B, Prince HM, Foss F, Sokol L, Greenwood M, Caballero D, Borchmann P, Morschhauser F, Wilhelm M, Pinter-Brown L, Padmanabhan S, Shustov A, Nichols J, Carroll S, Balser J, Balser B, Horwitz S (2012) Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol 30:631–636CrossRefPubMed
12.
Zurück zum Zitat O’Connor OA, Horwitz S, Masszi T, Van Hoof A, Brown P, Doorduijn J, Hess G, Jurczak W, Knoblauch P, Chawla S, Bhat G, Choi MR, Walewski J, Savage K, Foss F, Allen LF, Shustov A (2015) Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the pivotal phase II BELIEF (CLN-19) study. J Clin Oncol 33:2492–2499CrossRefPubMedPubMedCentral O’Connor OA, Horwitz S, Masszi T, Van Hoof A, Brown P, Doorduijn J, Hess G, Jurczak W, Knoblauch P, Chawla S, Bhat G, Choi MR, Walewski J, Savage K, Foss F, Allen LF, Shustov A (2015) Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the pivotal phase II BELIEF (CLN-19) study. J Clin Oncol 33:2492–2499CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Shi Y, Dong M, Hong X, Zhang W, Feng J, Zhu J, Yu L, Ke X, Huang H, Shen Z, Fan Y, Li W, Zhao X, Qi J, Huang H, Zhou D, Ning Z, Lu X (2015) Results from a multicenter, open-label, pivotal phase II study of chidamide in relapsed or refractory peripheral T-cell lymphoma. Ann Oncol 26:1766–1771CrossRefPubMed Shi Y, Dong M, Hong X, Zhang W, Feng J, Zhu J, Yu L, Ke X, Huang H, Shen Z, Fan Y, Li W, Zhao X, Qi J, Huang H, Zhou D, Ning Z, Lu X (2015) Results from a multicenter, open-label, pivotal phase II study of chidamide in relapsed or refractory peripheral T-cell lymphoma. Ann Oncol 26:1766–1771CrossRefPubMed
14.
Zurück zum Zitat Ogura M, Ishida T, Hatake K, Taniwaki M, Ando K, Tobinai K, Fujimoto K, Yamamoto K, Miyamoto T, Uike N, Tanimoto M, Tsukasaki K, Ishizawa K, Suzumiya J, Inagaki H, Tamura K, Akinaga S, Tomonaga M, Ueda R (2014) Multicenter phase II study of mogamulizumab (KW-0761), a defucosylated anti-CC chemokine receptor 4 antibody, in patients with relapsed peripheral T-cell lymphoma and cutaneous T-cell lymphoma. J Clin Oncol 32:1157–1163CrossRefPubMed Ogura M, Ishida T, Hatake K, Taniwaki M, Ando K, Tobinai K, Fujimoto K, Yamamoto K, Miyamoto T, Uike N, Tanimoto M, Tsukasaki K, Ishizawa K, Suzumiya J, Inagaki H, Tamura K, Akinaga S, Tomonaga M, Ueda R (2014) Multicenter phase II study of mogamulizumab (KW-0761), a defucosylated anti-CC chemokine receptor 4 antibody, in patients with relapsed peripheral T-cell lymphoma and cutaneous T-cell lymphoma. J Clin Oncol 32:1157–1163CrossRefPubMed
15.
Zurück zum Zitat Pro B, Advani R, Brice P, Bartlett NL, Rosenblatt JD, Illidge T, Matous J, Ramchandren R, Fanale M, Connors JM, Yang Y, Sievers EL, Kennedy DA, Sustov A (2012) Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J Clin Oncol 30:2190–2196CrossRefPubMed Pro B, Advani R, Brice P, Bartlett NL, Rosenblatt JD, Illidge T, Matous J, Ramchandren R, Fanale M, Connors JM, Yang Y, Sievers EL, Kennedy DA, Sustov A (2012) Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J Clin Oncol 30:2190–2196CrossRefPubMed
16.
Zurück zum Zitat Lamarque M, Bossard C, Contejean A, Brice P, Parrens M, Le Gouill S, Brière J, Bouabdallah R, Canioni D, Tilly H, Bouchindhomme B, Bachy E, Delarue R, Haioun C (2016) Gaulard P (2016) Brentuximab vedotin in refractory or relapsed peripheral T-cell lymphomas: the French named patient program experience in 56 patients. Haematologica 101:e103–e106CrossRefPubMedPubMedCentral Lamarque M, Bossard C, Contejean A, Brice P, Parrens M, Le Gouill S, Brière J, Bouabdallah R, Canioni D, Tilly H, Bouchindhomme B, Bachy E, Delarue R, Haioun C (2016) Gaulard P (2016) Brentuximab vedotin in refractory or relapsed peripheral T-cell lymphomas: the French named patient program experience in 56 patients. Haematologica 101:e103–e106CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Markert ML (1991) Purine nucleoside phosphorylase deficiency. Immunodefic Rev 3:45–81PubMed Markert ML (1991) Purine nucleoside phosphorylase deficiency. Immunodefic Rev 3:45–81PubMed
18.
Zurück zum Zitat Kicska GA, Long L, Hörig H, Fairchild C, Tyler PC, Furneaux RH, Schramm VL, Kaufman HL (2001) Immucillin H, a powerful transition-state analog inhibitor of purine nucleoside phosphorylase, selectively inhibits human T lymphocytes. Proc Natl Acad Sci U S A 98:4593–4598CrossRefPubMedPubMedCentral Kicska GA, Long L, Hörig H, Fairchild C, Tyler PC, Furneaux RH, Schramm VL, Kaufman HL (2001) Immucillin H, a powerful transition-state analog inhibitor of purine nucleoside phosphorylase, selectively inhibits human T lymphocytes. Proc Natl Acad Sci U S A 98:4593–4598CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Ogura M, Tsukasaki K, Nagai H, Uchida T, Oyama T, Suzuki T, Taguchi J, Maruyama D, Hotta T, Tobinai K (2012) Phase I study of BCX1777 (forodesine) in patients with relapsed or refractory peripheral T/natural killer-cell malignancies. Cancer Sci 103:1290–1295CrossRefPubMed Ogura M, Tsukasaki K, Nagai H, Uchida T, Oyama T, Suzuki T, Taguchi J, Maruyama D, Hotta T, Tobinai K (2012) Phase I study of BCX1777 (forodesine) in patients with relapsed or refractory peripheral T/natural killer-cell malignancies. Cancer Sci 103:1290–1295CrossRefPubMed
20.
Zurück zum Zitat Dummer R, Duvic M, Scarisbrick J, Olsen EA, Rozati S, Eggmann N, Goldinger SM, Hutchinson K, Geskin L, Illidge TM, Giuliano E, Kim YH (2014) Final results of a multicenter phase II study of the purine nucleoside phosphorylase (PNP) inhibitor forodesine in patients with advanced cutaneous T-cell lymphomas (CTCL) (mycosis fungoides and Sézary syndrome). Ann Oncol 25:1807–1812CrossRefPubMed Dummer R, Duvic M, Scarisbrick J, Olsen EA, Rozati S, Eggmann N, Goldinger SM, Hutchinson K, Geskin L, Illidge TM, Giuliano E, Kim YH (2014) Final results of a multicenter phase II study of the purine nucleoside phosphorylase (PNP) inhibitor forodesine in patients with advanced cutaneous T-cell lymphomas (CTCL) (mycosis fungoides and Sézary syndrome). Ann Oncol 25:1807–1812CrossRefPubMed
21.
Zurück zum Zitat Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Horning SJ, Coiffier B, Fisher RI, Hagenbeek A, Zucca E, Rosen ST, Stroobants S, Lister TA, Hoppe RT, Dreyling M, Tobinai K, Vose JM, Connors JM, Federico M, Diehl V (2007) Revised response criteria for malignant lymphoma. J Clin Oncol 25:579–586CrossRef Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Horning SJ, Coiffier B, Fisher RI, Hagenbeek A, Zucca E, Rosen ST, Stroobants S, Lister TA, Hoppe RT, Dreyling M, Tobinai K, Vose JM, Connors JM, Federico M, Diehl V (2007) Revised response criteria for malignant lymphoma. J Clin Oncol 25:579–586CrossRef
22.
Zurück zum Zitat Simon R (1989) Optimal two-stage designs for phase II clinical trials. Control Clin Trials 10:1–10CrossRefPubMed Simon R (1989) Optimal two-stage designs for phase II clinical trials. Control Clin Trials 10:1–10CrossRefPubMed
23.
Zurück zum Zitat Kaplan EL, Meier P (1958) Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481CrossRef Kaplan EL, Meier P (1958) Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481CrossRef
24.
Zurück zum Zitat Morschhauser F, Fitoussi O, Haioun C, Thieblemont C, Quach H, Delarue R, Glaisner J, Bosly A, Lister J, Li J, Coiffier B (2013) A phase 2, multicentre, single-arm, open-label study to evaluate the safety and efficacy of single-agent lenalidomide (Revlimid) in subjects with relapsed or refractory peripheral T-cell non-Hodgkin lymphoma: the EXPECT trial. Eur J Cancer 49:2869–2876CrossRefPubMed Morschhauser F, Fitoussi O, Haioun C, Thieblemont C, Quach H, Delarue R, Glaisner J, Bosly A, Lister J, Li J, Coiffier B (2013) A phase 2, multicentre, single-arm, open-label study to evaluate the safety and efficacy of single-agent lenalidomide (Revlimid) in subjects with relapsed or refractory peripheral T-cell non-Hodgkin lymphoma: the EXPECT trial. Eur J Cancer 49:2869–2876CrossRefPubMed
25.
Zurück zum Zitat Zinzani PL, Venturini F, Stefoni V, Fina M, Pellegrini C, Derenzini E, Gandolfi L, Broccoli A, Argnani L, Quirini F, Pileri S, Baccarani M (2010) Gemcitabine as a single agent in pretreated T-cell lymphoma patients: evaluation of the long-term outcomes. Ann Oncol 21:860–863CrossRefPubMed Zinzani PL, Venturini F, Stefoni V, Fina M, Pellegrini C, Derenzini E, Gandolfi L, Broccoli A, Argnani L, Quirini F, Pileri S, Baccarani M (2010) Gemcitabine as a single agent in pretreated T-cell lymphoma patients: evaluation of the long-term outcomes. Ann Oncol 21:860–863CrossRefPubMed
26.
Zurück zum Zitat Zettl A, Lee SS, Rüdger T, Starostik P, Marino M, Kirchner T, Ott M, Müller-Hermelink HK, Ott G (2002) Epstein-Barr virus-associated B-cell lymphoproliferative disorders in angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified. Am J Clin Pathol 117:368–379CrossRefPubMed Zettl A, Lee SS, Rüdger T, Starostik P, Marino M, Kirchner T, Ott M, Müller-Hermelink HK, Ott G (2002) Epstein-Barr virus-associated B-cell lymphoproliferative disorders in angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified. Am J Clin Pathol 117:368–379CrossRefPubMed
27.
Zurück zum Zitat Takahashi T, Maruyama R, Mishima S, Inoue M, Kawakami K, Onishi C, Miyake T, Tanaka J, Nabika T, Ishikura H (2010) Small bowl perforation caused by Epstein-Barr virus-associated B cell lymphoma in a patient with angioimmunoblastic T-cell lymphoma. J Clin Exp Hematopathol 50:59–63CrossRef Takahashi T, Maruyama R, Mishima S, Inoue M, Kawakami K, Onishi C, Miyake T, Tanaka J, Nabika T, Ishikura H (2010) Small bowl perforation caused by Epstein-Barr virus-associated B cell lymphoma in a patient with angioimmunoblastic T-cell lymphoma. J Clin Exp Hematopathol 50:59–63CrossRef
28.
Zurück zum Zitat Balagué O, Martinez A, Colomo L, Roselló E, Garcia A, Martinez-Bernal M, Palacin A, Fu K, Weisenburger D, Colomer D, Burke JS, Warnke RA, Campo E (2007) Epstein-Barr virus negative clonal plasma cell proliferaitons and lymphomas in peripheral T-cell lymphomas: a phenomenon with distinctive clincopathologic features. Am J Surg Pathol 31:1310–1322CrossRefPubMed Balagué O, Martinez A, Colomo L, Roselló E, Garcia A, Martinez-Bernal M, Palacin A, Fu K, Weisenburger D, Colomer D, Burke JS, Warnke RA, Campo E (2007) Epstein-Barr virus negative clonal plasma cell proliferaitons and lymphomas in peripheral T-cell lymphomas: a phenomenon with distinctive clincopathologic features. Am J Surg Pathol 31:1310–1322CrossRefPubMed
29.
Zurück zum Zitat Willenbrock K, Bräuninger A, Hansmann ML (2007) Frequent occurrence of B-cell lymphomas in angioimmunoblastic T-cell lymphoma and proliferation of Epstein-Barr virus-infected cells in early cases. Br J Haematol 138:733–739CrossRefPubMed Willenbrock K, Bräuninger A, Hansmann ML (2007) Frequent occurrence of B-cell lymphomas in angioimmunoblastic T-cell lymphoma and proliferation of Epstein-Barr virus-infected cells in early cases. Br J Haematol 138:733–739CrossRefPubMed
Metadaten
Titel
Multicenter phase 1/2 study of forodesine in patients with relapsed peripheral T cell lymphoma
verfasst von
Dai Maruyama
Kunihiro Tsukasaki
Toshiki Uchida
Yoshinobu Maeda
Hirohiko Shibayama
Hirokazu Nagai
Mitsutoshi Kurosawa
Yoko Suehiro
Kiyohiko Hatake
Kiyoshi Ando
Isao Yoshida
Michihiro Hidaka
Tohru Murayama
Yoko Okitsu
Norifumi Tsukamoto
Masafumi Taniwaki
Junji Suzumiya
Kazuo Tamura
Takahiro Yamauchi
Ryuzo Ueda
Kensei Tobinai
Publikationsdatum
05.07.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Annals of Hematology / Ausgabe 1/2019
Print ISSN: 0939-5555
Elektronische ISSN: 1432-0584
DOI
https://doi.org/10.1007/s00277-018-3418-2

Weitere Artikel der Ausgabe 1/2019

Annals of Hematology 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.