Skip to main content
Cancer Science logoLink to Cancer Science
. 2009 Sep 10;101(1):196–200. doi: 10.1111/j.1349-7006.2009.01360.x

Potential efficacy of the oral histone deacetylase inhibitor vorinostat in a phase I trial in follicular and mantle cell lymphoma

Takashi Watanabe 1,, Harumi Kato 2, Yukio Kobayashi 1, Satoshi Yamasaki 1, Yuriko Morita‐Hoshi 1, Hiroki Yokoyama 1, Yasuo Morishima 2, Justin L Ricker 3, Tetsuya Otsuki 4, Akiko Miyagi‐Maesima 5, Yoshihiro Matsuno 5, Kensei Tobinai 1
PMCID: PMC11159849  PMID: 19817748

Abstract

Vorinostat (suberoylanilide hydroxamic acid, SAHA, Zolinza) is a histone deacetylase inhibitor with clinical activity in cutaneous T‐cell lymphoma (CTCL). A phase I trial of oral vorinostat was conducted in Japanese patients with malignant lymphoma. Vorinostat 100 or 200 mg was administered twice daily for 14 consecutive days followed by a 1‐week rest interval. Of 10 patients enrolled, four had follicular lymphoma (FL), two mantle cell lymphoma (MCL), two diffuse large B‐cell lymphoma, and two CTCL (median age, 60 years; median number of prior regimens, 3). Vorinostat was well tolerated up to 200 mg with only one of six patients developing a dose‐limiting toxicity (DLT; Grade 3 anorexia/hypokalemia). Common Grade 3 events were reversible neutropenia (30%), thrombocytopenia, and hypermagnesemia (20% each). The median number of treatment cycles was five (range, 1–36); two patients were continuing treatment. The overall response rate was 40%, with two complete responses/unconfirmed (CRu) and one partial response among FL patients and one CRu among MCL patients. One FL patient maintained CRu for 18.0 months. The median time to achieve CRu among the three patients was 8 months. These data suggest that further investigations of vorinostat in non‐Hodgkin lymphoma, focusing on FL and MCL, are warranted. (Cancer Sci 2009)


In 2002, the worldwide incidence of non‐Hodgkin lymphoma (NHL) was estimated to be 300 571, corresponding to 171 820 deaths.( 1 ) Common types of NHL include diffuse large B‐cell lymphoma (DLBCL) and follicular lymphoma (FL). Because relapse is common in NHL, especially for indolent B‐cell lymphoma even after the advent of rituximab, novel effective treatments are clearly needed.

Vorinostat (Zolinza; Merck, Whitehouse Station, NJ, USA) is an inhibitor of class I and II histone deacetylase (HDAC) enzymes that regulate transcriptional and post‐transcriptional processes by removing acetyl groups from proteins.( 2 ) In preclinical studies, vorinostat exposure resulted in cell cycle arrest and antiproliferative and antitumor activities in lymphoma models.( 3 , 4 , 5 ) In October 2006, vorinostat was approved in the United States for the treatment of cutaneous manifestations in patients with cutaneous T‐cell lymphoma (CTCL) who have progressive, persistent, or recurrent disease on or following two systemic therapies.( 6 , 7 ) Clinical activity of vorinostat has also been observed in patients with transformed DLBCL,( 8 , 9 ) but it was limited against DLBCL.( 10 )

Recently, the results of a phase I study of oral vorinostat in American patients with advanced leukemias and myelodysplastic syndromes were reported.( 11 ) The present study was conducted to evaluate vorinostat in Japanese patients with malignant lymphoma (ML). The primary objectives were to determine the maximum tolerated dose (MTD) and evaluate dose‐limiting toxicities (DLTs) and the overall safety profile of vorinostat in the first cycle. Secondary objectives were to assess the pharmacokinetics and the overall safety profiles in subsequent cycles and to determine the recommended phase II dose in patients with ML. Exploratory objectives were to evaluate response according to the International Workshop Response Criteria ( 12 ) and to evaluate the pharmacodynamic effects on normal cells via assessment of histone acetylation.

Materials and Methods

Patient eligibility.  Patients with ML who had received one to four prior regimens and had relapsed after complete response or partial response or were refractory to previous chemotherapies were eligible. Patients must have been ≥20 years, but ≤75 years in age, with Eastern Cooperative Oncology Group performance status of 0–1 and adequate hepatic (total bilirubin ≤1.5× the upper limit of normal [ULN]; aspartate aminotransferase and alanine aminotransferase ≤2.5 × ULN), hematologic (neutrophil count ≥1500/μL; platelet count ≥100 000/μL; hemoglobin ≥9.0 g/dL, prothrombin and activated partial thromboplastin time ≤1.5 × ULN), and renal function (creatinine ≤1.5 × ULN and creatinine clearance ≥50 mL/min). Exclusion criteria included: prior HDAC inhibitor treatment or allogeneic stem cell transplantation, prior therapy within 4 weeks of study, central nervous system involvement, uncontrolled concomitant illness, pregnancy, lactation, or concurrent malignancies. This open‐label, single‐arm, non‐randomized phase I trial (Protocol 030) was approved by the institutional review boards of the National Cancer Center Hospital, Tokyo, Japan, and Aichi Cancer Center Hospital, Nagoya, Japan. Prior to enrollment, all patients provided written informed consent in accordance with the Declaration of Helsinki.

Study design, treatment, and evaluation.  Oral vorinostat 100 or 200 mg was administered twice daily (bd) for 14 consecutive days followed by a 7‐day rest in a 3‐week cycle. The study used a ‘3 + 3’ dose escalation design as described elsewhere.( 11 ) Dose‐limiting toxicities were: (i) Grade 4 neutropenia persisting for ≥5 days; (ii) Grade ≥3 infection with Grade 3 or 4 neutropenia; (iii) Grade ≥3 thrombocytopenia requiring blood transfusion; (iv) Grade 4 thrombocytopenia or anemia; (v) Grade ≥3 non‐hematologic toxicities other than anorexia, nausea/vomiting, and fatigue; or (vi) Grade ≥3 anorexia, nausea/vomiting, and fatigue refractory to treatment during the first cycle. The MTD was defined as the dose level at which all of the three patients initially enrolled or three or more of the six enrolled patients developed any DLT(s) in the first cycle. For patients on dose level 2 (200 mg), the dose was reduced to 100 mg if a DLT occurred during the first or subsequent cycles, or if a drug‐related adverse event (AE) resolved to Grade 1 or less within 1 week after cycle completion. Once a patient underwent a dose reduction, an increase to the previous dose level was not permitted even after resolution of the toxicity. Patients were to be discontinued for disease progression, unacceptable toxicity, withdrawal of consent, lost to follow‐up, or at the investigator’s discretion. Safety was assessed using National Cancer Institute Common Terminology Criteria for Adverse Events, version 3.0.( 12 ) Adverse events that were definitely, probably, or possibly related to vorinostat were categorized as drug‐related. Efficacy was assessed according to the International Workshop Response Criteria.( 13 )

Pharmacokinetic and pharmacodynamic analyses.  The pharmacokinetics of vorinostat were investigated in Cycle 1. Patients received vorinostat once daily on Days 1 and 17, and bd on Days 3 to 16 using 100 mg on dose level 1 and 200 mg on dose level 2 as a schedule in Cycle 1. Intensive blood sampling was performed at pre‐dose and 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10, 12, and 24 h post‐dose on Days 1 and 17. Based on measured serum concentrations, area under the concentration‐time curves from time 0 to 12 h (AUC0‐12hr), AUC from time zero to infinity (AUC0‐∞), the maximum concentration (C max), time to maximum concentration (T max), and terminal half life (t 1/2) were determined on Days 1 and 17. The geometric mean ratio for AUC0‐∞ or AUC0‐12h on Day 1 and AUC0‐12h on Day 17 were calculated. Trough values of vorinostat on Days 4 and 10 were also determined. Urine samples were collected at pre‐dose and during 0–6, 6–12, and 12–24 h post‐dose on Days 1 and 17. Based on measured urine concentrations, urinary recovery (% of dose) was calculated.

Blood samples (5 mL) were collected on Day 1 of Cycle 1 at pre‐dose and at 2, 4, 8, and 24 h. Peripheral blood mononuclear cells (PBMCs) were lysed and utilized to assess histone H3 acetylation by enzyme‐linked immunosorbent assay as previously described.( 11 )

Results

Patient population.  The baseline patient characteristics of the 10 enrolled patients are shown in Table 1. The median number of prior regimens was three (range, 1–4). The median study duration was 3.1 months (range, 0.4–26.1+).

Table 1.

 Baseline patient characteristics (n =10)

Characteristic Number of patients
Median age, years (range) 60 (52–74)
Male 4
Female 6
ECOG performance status
 0 8
 1 2
Disease type
 FL 4
 MCL 2
 DLBCL 2
 CTCL 2
Stage at enrollment
 II 4
 III 3
 IV 3
Number of prior regimens
 1 1
 2 2
 3 4
 4 3
 Median 3

CTCL, cutaneous T‐cell lymphoma; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; FL, follicular lymphoma; MCL, mantle cell lymphoma.

Safety and toxicity.  Nine patients completed the first treatment cycle and were evaluated for DLTs; one patient was excluded due to disease progression during the first cycle. None of the three evaluable patients who received vorinostat 100 mg bd developed a DLT. One of six patients treated at the 200 mg bd dose level developed a DLT (Grade 3 anorexia and hypokalemia). In an ex‐Japanese trial, vorinostat administered at a dose of 300 mg bd for 14 consecutive days every 3 weeks exceeded the MTD, and 200 mg was defined as the MTD on this schedule.( 11 ) Therefore, the dosing regimen of this trial was changed so that the maximum dose level was 200 mg bd Although the MTD was not reached, vorinostat 200 mg bd, which was the maximum dose in this study, for 14 consecutive days every 3 weeks is the recommended phase II dose and schedule of vorinostat in Japanese patients with NHL.

The most common drug‐related AEs for the first and subsequent cycles are shown in Table 2. All 10 patients received at least one dose of vorinostat and were evaluable for safety. Hematologic drug‐related adverse events included neutropenia, thrombocytopenia, anemia (70% each), and leukopenia (60%). Non‐hematologic drug‐related adverse events included anorexia, hyperglycemia, proteinuria (70% each); and fatigue, hypocalcemia, nausea, and diarrhea (60% each). Most of these events were Grade ≤2, and all were reversible and rapidly resolved during the rest period. Grade 3 drug‐related adverse events were neutropenia (30%), thrombocytopenia, hypermagnesemia (20% each), anorexia, diarrhea, hypokalemia, and hyponatremia (10% each).

Table 2.

 Most common drug‐related adverse events in all cycles (n =10)

Events Grade 1 or 2 Grade 3
n n
Hematologic
 Neutropenia 4 3
 Thrombocytopenia 5 2
 Anemia 7 0
 Leukopenia 6 0
Non‐hematologic
 Anorexia 6 1
 Hyperglycemia 7 0
 Proteinuria 7 0
 Fatigue 6 0
 Hypocalcemia 6 0
 Nausea 6 0
 Diarrhea 5 1
 Increased alanine aminotransferase 5 0
 Dysgeusia 5 0
 Weight loss 5 0
 Hypermagnesemia 3 2
 Hypokalemia 4 1
 Hyponatremia 3 1
 Vomiting 4 0
 Headache 4 0
 Nail disorder 4 0
 Rash 4 0
 Decreased albumin 4 0
 Increased creatinine 4 0
 Increased lactate dehydrogenase 4 0
 Decreased total protein 4 0

Drug‐related adverse events for all grades reported in 40% or above of patients are listed.

Four patients had dose modifications in the second cycle or later due to AEs (anorexia, Cycle 1; leukopenia and neutropenia, Cycle 1; neutropenia, Cycle 3; hypermagnesemia, Cycle 2). Six patients discontinued vorinostat in Cycles 1–5 due to disease progression, and two patients were continuing on vorinostat treatment as of March 2008. There were no discontinuations due to AEs and no sudden deaths. However, two patients (patients #6 and #7 in Table 3) who responded to vorinostat and therefore received it for several months developed Grade 1 nail brittleness (Fig. 1).

Table 3.

 Clinical outcomes of patients

Patient number Disease type Dose (mg) Number of prior regimens Best overall response† Duration of best response‡ (month) On‐study duration (month)
 1 DLBCL 100 bd (14/21 days) 3 PD 0.8
 2 CTCL 3 PD 0.4
 3 MCL 4 SD 2.7
 4 CTCL 3 SD 3.6
 5 FL 200 bd (14/21 days) 3 SD 2.4
 6 FL 4 CRu 18.0+ 26.1+
 7 FL 1 PR 6.2 25.9+
 8 MCL 2 CRu 1.7 23.1
 9 FL 2 CRu 1.3 12.7
10 DLBCL 4 PD 1.4

†Responses were determined according to the International Workshop Response Criteria( 13 ). ‡For patients who achieved CRu, only the duration of CRu is shown (duration of PR is not shown). −, not applicable; +, ongoing; bd, twice daily; CRu, complete response/unconfirmed; CTCL, cutaneous T‐cell lymphoma; DLBCL, diffuse large B‐cell lymphoma; FL, follicular lymphoma; MCL, mantle cell lymphoma; PD, progressive disease; PR, Partial response; SD, stable disease.

Figure 1.

Figure 1

 Chips and streaks of the nails observed several months after oral intake of vorinostat in the two patients (patients #6 and #7 in Table 3) who responded to vorinostat.

Efficacy.  The clinical outcomes of the patients are shown in Table 3. The overall response rate was 40%. Of the four patients with FL assigned to 200 mg bd, two achieved complete responses/unconfirmed (CRu), and one had a partial response (PR). One of the two patients with mantle cell lymphoma (MCL) received 200 mg bd and achieved a CRu. In addition, three patients (MCL, CTCL, and FL) had stable disease. The median time to achieve CRu among the three patients was 8 months.

Pharmacokinetics and pharmacodynamics.  Maximum plasma concentrations were observed in the range of 1.50–3.44 h after oral intake in both single and multiple doses of vorinostat at both dose levels. Vorinostat was then rapidly eliminated with an apparent t1/2 of 1.25 to 1.40 h (Table 4, Fig. 2). The accumulation ratios of vorinostat following 14 days of multiple dosing at dose levels 1 and 2 were 1.35 and 1.34, respectively (Table 4). Two inactive metabolites (the O‐glucuronide of vorinostat and 4‐anilino‐4‐oxobutanoic acid) were measured in serum. AUCs of the O‐glucuronide and 4‐anilino‐4‐oxobutanoic acid metabolites following administration of vorinostat in the fed state were 2‐ to 4‐fold and 8‐ to 10‐fold higher, respectively, than that of vorinostat (data not shown). The O‐glucuronide and 4‐anilino‐4‐oxobutanoic acid metabolites were the main compounds recovered in the urine following administration of vorinostat. Only a small amount of vorinostat (less than 1% of the dose) was observed in the urine (Fig. 3).

Table 4.

 Summary of pharmacokinetic parameters of vorinostat

Parameter n Day 1 n Day 17
SD, fed MD, fed
Geometric mean ± SD Geometric mean ± SD
Dose level 1 (100 mg bd)
 AUC0–∞ (μM h) 4 1.03 ± 0.45 3 1.42 ± 0.61
 AUC0–12h (μM h) 4 1.01 ± 0.44 3 1.41 ± 0.59
C max (μM) 4 0.28 ± 0.10 3 0.34 ± 0.12
T max (h), median 4 3.44 3 1.50
t 1/2 (h), harmonic mean 4 1.27 3 1.34
 Accumulation ratio§ 3 1.35
Dose level 2 (200 mg bd)
 AUC0–∞ (μM h) 6 2.67 ± 0.88 5 3.77 ± 1.29
 AUC0–12h (μM h) 6 2.65 ± 0.87 5 3.30 ± 0.27
C max (μM) 6 0.66 ± 0.23 5 0.83 ± 0.28
T max (h), median 6 2.98 5 3.05
t 1/2 (h), harmonic mean 6 1.25 5 1.40
 Accumulation ratio§ 5 1.34

†Patients received vorinostat once daily on Days 1 and 17, and twice daily on Days 3 to 16 in the 1st cycle for pharmakokinetic data analysis. ‡One patient received 100 mg MD bd was excluded due to incompletion of the 1st cycle, and one patient received 200 mg MD bd was excluded due to occurrence of dose‐limiting toxicity. §AUC0–12h, Day 17/AUC0–12h, Day 1. AUC0 – ∞ , area under the concentration–time curve from time zero to infinity; AUC0–12h, AUC from time zero to 12 h; bd, twice daily; C max, maximum concentration; T max, time to maximum concentration; MD, multiple doses; SD, single dose; t 1/2, terminal half life.

Figure 2.

Figure 2

 Mean serum concentration‐time profiles following administration of single/multiple oral doses of vorinostat. In each dose level (Lv), vorinostat was administered with single dose on Day 1, and then with multiple doses of twice daily (bd) on Days 3–16 and once daily (qd) Day 17. All of the doses were performed in the fed state. In Lv 1, pharmacokinetic data of vorinostat were obtained from four patients on Days 1, 4 and 10, and from three patients on Day 17 (one subject discontinued on Day 11 because of impossibility of dose‐limiting toxicity [DLT] assessment). In Lv 2, pharmacokinetic data of vorinostat were obtained from six patients on Days 1, 4, and 10, and from five patients on Day 17 (one subject discontinued on Day 11 because of DLT).

Figure 3.

Figure 3

 Mean urinary excretion following administration of single/ multiple oral doses of vorinostat. In each dose level (Lv), vorinostat was administered with a single dose on Day 1, and then with multiple doses twice daily (bd) on Days 3–16 and once daily (qd) on Day 17. All of the doses were performed in the fed state. β‐Oxidative form and glucuronide indicate 4‐anilino‐4‐oxobutanoic acid and O‐glucuronide of vorinostat, respectively.

PBMCs from all 10 patients enrolled were assessed for pharmacodynamics. Histone H3 acetylation was induced 1.8‐fold relative to baseline by 8 h after treatment and returned to baseline by 24 h in PBMCs of patients receiving the 200 mg, but not the 100 mg, vorinostat dose level (Fig. 4).

Figure 4.

Figure 4

 Average histone H3 acetylation on Day 1 by dose group. Peripheral blood mononuclear cells from all 10 patients were assessed.

Discussion

In an ex‐Japanese trial, vorinostat administered at a dose of 300 mg bd for 14 consecutive days every 3 weeks exceeded the MTD, and 200 mg was defined as the MTD for that schedule.( 11 ) Therefore, the dosing regimen of the present trial was changed so that the maximum dose level was 200 mg bd for 14 consecutive days every 3 weeks. Although two DLTs occurred in one of 10 patients (Grade 3 anorexia and Grade 3 hypokalemia), the MTD was not reached in this trial. Therefore, the maximum acceptable dose was judged as 200 mg bd for 14 consecutive days every 3 weeks.

The safety profile of vorinostat in Japanese patients was similar to that in non‐Japanese patients.( 6 , 7 , 8 , 9 , 10 , 11 , 14 , 15 ) Also, the pharmacokinetic results were consistent with those in non‐Japanese patients.( 8 , 14 ) Vorinostat was rapidly eliminated from the systemic circulation with slight accumulation following multiple doses. The hematologic AEs with vorinostat were reversible, as the patients quickly recovered from these events during the rest period. Therefore, it may be appropriate to study this agent in combination with traditional chemotherapy drugs, which often cause hematologic toxicities in patients with hematological malignancies. Another HDAC inhibitor, depsipeptide, has been associated with cardiotoxicity, such as ventricular tachycardia and QT interval corrected for heart rate (QTc) interval prolongation.( 16 , 17 ) However, no patients in this study had a clinically significant cardiac arrhythmia or QTc interval prolongation. An interesting finding worth mentioning is that patients who responded to treatment and received vorinostat for a longer duration developed Grade 1 nail brittleness. To our knowledge, this finding has not been previously reported in trials with vorinostat.

The overall response rate was 40%, including three CRus and one PR. One of the CRus occurred in a heavily pretreated patient who had received four prior regimens. Two CRus and one PR occurred in FL patients, and one CRu occurred in an MCL patient. It should be noted that activity has been achieved at doses up to 200 mg on this schedule, as was reported previously in other solid and hematologic malignancies.( 11 , 18 ) The median time to achieve CRu among the three patients was 8 months. Therefore, it may be important that the establishment of the optimal dose and manner without either severe toxicities or disease progression enables patients to take this kind of agent with mild activity for a longer duration and to achieve favorable outcomes. Previous pre‐clinical studies showed that vorinostat suppressed translation of cyclin D1 in MCL cells.( 19 ) In the present study, histone acetylation was induced 1.8‐fold in PBMCs from patients on the 200 mg bd dose level. Although the MTD was not reached, favorable responses were achieved with vorinostat as monotherapy in patients with FL and MCL. The vorinostat dose was not increased above 200 mg bd for 14 consecutive days every 3 weeks because of known toxicity data from ex‐Japanese studies.

In conclusion, oral vorinostat was well tolerated up to 200 mg bd for 14 consecutive days every 3 weeks in Japanese patients with NHL. Although evaluation of response was an exploratory objective in this phase I trial, the findings of this study highlight the potential single‐agent activity of vorinostat in FL or MCL. This study provides support for further investigation of vorinostat as monotherapy or in combination with other active agents in NHL. Thus, these data suggest that phase II studies, focusing on FL and MCL, are warranted and a phase II study for indolent lymphoma including FL and MCL has been started in Asia.

Disclosure Statement

Justin L. Ricker was an employee of Merck & Co., Inc., at the time this manuscript was written. Tetsuya Otsuki is an employee of Banyu Pharmaceutical Co., Ltd., and holds stock and stock options of Merck & Co., Inc. The remaining authors report no potential conflicts of interest.

Acknowledgments

We thank all of the investigators including Drs Takashi Terauchi (Diagnostic Division, Research Center for Cancer Prevention and Screening, National Cancer Center) and Teruhisa Azuma and Masakazu Mori (Hematology Division, National Cancer Center Hospital) for their response assessment by reviews of serial computed tomography performed every 3 months; and a clinical research coordinator, Ms Tamami Yamano, at National Cancer Center Hospital, for her data collection. We also thank Dr Shinichi Kanazu (Banyu Pharmaceutical) and Dr James S. Hardwick (Merck Research Laboratories) for their support in preparing this manuscript and thank Ms Mary Flynn (Merck Research Laboratories) for her assistance with the histone acetylation assays. This trial was supported by research funding from Merck Research Laboratories, Rahway, NJ, USA, and Banyu Pharmaceutical, Tokyo, Japan.

ClinicalTrials.gov Identifier: NCT00127140

Present addresses: S.Y.: Division of Basic and Translational Research, Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Y.M.‐H.: Department of Laboratory Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo‐ku, Tokyo, Japan; H.Y.: Department of Oncology and Hematology, Jikei University School of Medicine, Komae‐shi, Tokyo, Japan; J.L.R.: Department of Clinical Oncology, Abbott Laboratories, Abbott Park, IL, USA; Y.M.: Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.

References

  • 1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin 2005; 55: 74–108. [DOI] [PubMed] [Google Scholar]
  • 2. Kelly WK, O’Connor OA, Marks PA. Histone deacetylase inhibitors: from target to clinical trials. Expert Opin Investig Drugs 2002; 11: 1695–713. [DOI] [PubMed] [Google Scholar]
  • 3. Sakajiri S, Kumagai T, Kawamata N, Saitoh T, Said JW, Koeffler HP. Histone deacetylase inhibitors profoundly decrease proliferation of human lymphoid cancer cell lines. Exp Hematol 2005; 33: 53–61. [DOI] [PubMed] [Google Scholar]
  • 4. Zhang C, Richon V, Ni X, Talpur R, Duvic M. Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T‐cell lymphoma cells: relevance to mechanism of therapeutic action. J Invest Dermatol 2005; 125: 1045–52. [DOI] [PubMed] [Google Scholar]
  • 5. Heider U, Kaiser M, Sterz J et al. Histone deacetylase inhibitors reduce VEGF production and induce growth suppression and apoptosis in human mantle cell lymphoma. Eur J Haematol 2006; 76: 42–50. [DOI] [PubMed] [Google Scholar]
  • 6. Duvic M, Talpur R, Ni X et al. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T‐cell lymphoma (CTCL). Blood 2007; 109: 31–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Olsen EA, Kim YH, Kuzel TM et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T‐cell lymphoma. J Clin Oncol 2007; 25: 3109–15. [DOI] [PubMed] [Google Scholar]
  • 8. Kelly WK, O’Connor OA, Krug LM et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 2005; 23: 3923–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. O’Connor OA, Heaney ML, Schwartz L et al. Clinical experience with intravenous and oral formulations of the novel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. J Clin Oncol 2006; 24: 166–73. [DOI] [PubMed] [Google Scholar]
  • 10. Crump M, Coiffer B, Jacobsen ED et al. Phase II trial of oral vorinostat (suberoylanilide hydroxamic acid) in relapsed diffuse large‐B‐cell lymphoma. Ann Oncol 2008; 19: 964–9. [DOI] [PubMed] [Google Scholar]
  • 11. Garcia‐Manero G, Yang H, Bueso‐Ramos C et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood 2008; 111: 1060–6. [DOI] [PubMed] [Google Scholar]
  • 12. National Cancer Institute common terminology criteria for adverse events, version 3.0. [Cited 16 Jan 2007.] Available from URL: http://ctep.cancer.gov/forms/CTCAEv3.pdf. [Google Scholar]
  • 13. Cheson BD, Horning SJ, Coiffier B et al. Report of an international workshop to standardize response criteria for non‐Hodgkin’s lymphomas. NCI Sponsored International Working Group. J Clin Oncol 1999; 17: 1244–53. [DOI] [PubMed] [Google Scholar]
  • 14. Rubin EH, Agrawal NG, Friedman EJ et al. A study to determine the effects of food and multiple dosing on the pharmacokinetics of vorinostat given orally to patients with advanced cancer. Clin Cancer Res 2006; 12: 7039–45. [DOI] [PubMed] [Google Scholar]
  • 15. Blumenschein GR Jr, Kies MS, Papadimitrakopoulou VA et al. Phase II trial of the histone deacetylase inhibitor vorinostat (Zolinza®, suberoylanilide hydroxamic acid, SAHA) in patients with recurrent and/or metastatic head and neck cancer. Invest New Drugs 2008; 26: 81–7. [DOI] [PubMed] [Google Scholar]
  • 16. Piekarz RL, Frye AR, Wright JJ et al. Cardiac studies in patients treated with depsipeptide, FK228, in a phase II trial for T‐cell lymphoma. Clin Cancer Res 2006; 12: 3762–73. [DOI] [PubMed] [Google Scholar]
  • 17. Shah MH, Binkley P, Chan K et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin Cancer Res 2006; 12: 3997–4003. [DOI] [PubMed] [Google Scholar]
  • 18. Fujiwara Y, Yamamoto N, Yamada Y et al. Phase I and pharmacokinetic study of vorinostat (suberoylanilide hydroxamic acid) in Japanese patients with solid tumors. Cancer Sci 2009; 100: 1728–34. [DOI] [PubMed] [Google Scholar]
  • 19. Kawamata N, Chen J, Koeffler PH. Suberoylanilide hydroxamic acid (SAHA;vorinostat) suppresses translation of cyclin D1 in mantle cell lymphoma cells. Blood 2007; 110: 2667–73. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancer Science are provided here courtesy of Wiley

RESOURCES