Purpose: This phase I study evaluated the safety, tolerability, pharmacokinetics, and preliminary efficacy of the combination of decitabine with vorinostat.

Patients and Methods: Patients with advanced solid tumors or non-Hodgkin's lymphomas were eligible. Sequential and concurrent schedules were studied.

Results: Forty-three patients were studied in 9 different dose levels (6 sequential and 3 concurrent). The maximum tolerated dose (MTD) on the sequential schedule was decitabine 10 mg/m2/day on days 1 to 5 and vorinostat 200 mg three times a day on days 6 to 12. The MTD on the concurrent schedule was decitabine 10 mg/m2/day on days 1 to 5 with vorinostat 200 mg twice a day on days 3 to 9. However, the sequential schedule of decitabine 10 mg/m2/day on days 1 to 5 and vorinostat 200 mg twice a day on days 6 to 12 was more deliverable than both MTDs with fewer delays on repeated dosing and it represents the recommended phase II (RP2D) dose of this combination. Dose-limiting toxicities during the first cycle consisted of myelosuppression, constitutional and gastrointestinal symptoms and occurred in 12 of 42 (29%) patients evaluable for toxicity. The most common grade 3 or higher adverse events were neutropenia (49% of patients), thrombocytopenia (16%), fatigue (16%), lymphopenia (14%), and febrile neutropenia (7%). Disease stabilization for 4 cycles or more was observed in 11 of 38 (29%) evaluable patients.

Conclusion: The combination of decitabine with vorinostat is tolerable on both concurrent and sequential schedules in previously treated patients with advanced solid tumors or non-Hodgkin's lymphomas. The sequential schedule was easier to deliver. The combination showed activity with prolonged disease stabilization in different tumor types. Clin Cancer Res; 17(6); 1582–90. ©2011 AACR.

Translational Relevance

Hypermethylation of cytosines in CpG dinucleotides in the promoter regions of tumor-suppression genes and deacetylation of amino acid residues on the histone tails of nucleosomes, represent two epigenetic mechanisms of gene silencing that can contribute to tumor formation and progression. This phase I–targeted combination trial evaluates the safety, tolerability, pharmacokinetics, and preliminary antitumor activity of the hypomethylating agent decitabine, plus the histone deacetylase inhibitor vorinostat, in patients with advanced solid tumors. Sequential and concurrent administration schedules of these two agents were studied. Dose-limiting toxicities consisted mainly of myelosuppression, constitutional and gastrointestinal symptoms. Disease stabilization for four or more cycles was observed in about 30% of patients. Although the combination of these two types of epigenetics-modulating agents has been examined in hematological malignancies, this study represents one of the first attempts of this strategy in advanced solid tumors.

Hypermethylation of cytosines in CpG dinucleotides in the promoter regions of tumor-suppression genes and deacetylation of amino acid residues on the histone tails of nucleosomes, represent 2 epigenetic mechanisms of gene silencing that can contribute to tumor formation and progression (1, 2). Both events are considered reversible, and agents that inhibit the enzymes responsible for DNA methylation and histone deacetylation have been developed as anticancer agents (3).

Decitabine (5-aza-2′-deoxycytidine), a nucleoside analogue that is incorporated into DNA and acts as an hypomethylating agent by inhibiting DNA methyltransferase, and vorinostat (suberoylanilide hydroxamic acid), a small molecule that binds and directly inhibits histone deacetylase, are 2 agents with epigenetic effects that have shown clinical antitumor activity and are now approved for the treatment of myelodysplastic syndrome and cutaneous T-cell lymphoma, respectively (4–7). The validation of epigenetic treatments as anticancer strategies has supported an increasing number of trials evaluating epigenetic agents alone or in combination with other agents in both hematologic and solid malignancies (8, 9).

The combination of DNA methyltransferase inhibitor (DNMTi) with a histone deacetylase inhibitor (HDACi) represents an area that is gaining attention in the clinical development of epigenetic therapies. This concept is supported by preclinical evidence that DNA methylation and histone deacetylation are functionally linked, leading to transcriptional inactivation of genes critical for tumorigenesis (10, 11). Moreover, the in vitro combination of a DNMTi with an HDACi in hematologic and solid tumor cell lines have shown synergistic effects resulting in increased gene reexpression and superior antitumor activity (12–14).

The optimal schedule of the combination of a DNMTi with an HDACi has not been established yet. Although most of the preclinical studies performed have used a sequential administration of DNMTi followed by HDACi, it remains unclear whether different schedules of administration may have better clinical activity. In the phase I trial reported here, the combination decitabine and vorinostat was studied for the first time in patients with solid tumors and non-Hodgkin's lymphomas (NHL). Two different schedules of administration, sequential and concurrent, were evaluated. The principal objective of this study was to determine the safety and tolerability of the combination. Secondary objectives included the assessments of pharmacokinetics (PK) and preliminary antitumor efficacy.

Patient selection

Patients were eligible if they had a histologically or cytologically documented advanced solid malignancy or NHL, refractory to standard therapy or for which no standard therapy existed. Other key eligibility criteria included: Eastern Cooperative Oncology Group performance status 0 to 2; adequate hematologic, hepatic, and renal functions [white blood cell count ≥ 3 × 109/L, absolute neutrophil count (ANC) ≥ 1.5 × 109/L, platelets ≥ 100 × 109/L, AST (aspartate aminotransferase)/ALT (alanine aminotransferase) ≤ 2.5 times upper limit of normal, bilirubin within normal limits, creatinine ≤ 150 mmol/L and creatinine clearance ≥ 60 mL/minute]; unlimited prior chemotherapy, radiotherapy or targeted agents with at least 3-week interval (6-week interval if prior nitrosoureas or mitomycin C) between study entry and any prior treatment; no valproic acid or other HDACi for at least 2 weeks before study entry; no prior decitabine; and no known brain metastases.

The institutional review board of both participating centers approved the study, which was conducted in accordance with federal and institutional guidelines.

Study design and patient evaluation

This was a 2-center, open-label, phase I study in which intravenous decitabine administered on days 1 to 5 was combined with oral vorinostat either in a sequential (vorinostat starting on day 6) or a concurrent schedule (vorinostat starting on day 3), in 28-day cycles. The study began with dose escalation on the sequential schedule, and once the maximum tolerated dose (MTD) was established, accrual began on the concurrent schedule. On the sequential schedule, the starting dose of decitabine was 20 mg/m2/day on days 1 to 5, given as an intravenous infusion over 1 hour. Vorinostat was given at a starting dose of 100 mg twice a day on days 6 to 21. The starting dose of decitabine was chosen on the basis of published data showing good tolerability and higher response rates in patients with myelodysplastic syndrome treated with 20 mg/m2/day for 5 days (15). The starting dose of vorinostat was based on previous monotherapy studies showing that the maximum tolerated dose was 200 mg orally twice a day continuously in patients with solid tumors and 250 mg orally thrice a day for 14 days every 21 days in patients with hematologic malignancies (16, 17). No dose escalation or modification of the duration of treatment with decitabine was planned, whereas both dose escalation (up to 200 mg orally thrice a day) and evaluation of different durations of treatment (7, 16, or 21 days, starting on day 6) were initially planned for vorinostat. For the concurrent schedule, the starting doses of decitabine and vorinostat were based on the MTD established on the sequential schedule and dose escalation and evaluations of different durations of treatment (7 or 14 days, starting on day 3) were planned for vorinostat only.

Dose escalation in both schedules followed the standard 3 + 3 rule. The RP2D of this study was defined as the dose level at which 1 or less than 1 of 6 patients developed dose-limiting toxicity (DLT) and had the lowest frequency of treatment delays. Toxicity was graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 3.0. DLTs were defined as adverse events occurring during the first cycle of treatment and fulfilling one of the following criteria: ANC < 0.5 × 109/L for 7 days or more, febrile neutropenia, platelets less than 25 × 109/L or grade 3 thrombocytopenia associated with bleeding; any grade 3 or higher or intolerable grade 2 nonhematologic toxicity except alopecia, nausea, and vomiting responsive to antiemetics, diarrhea responsive to medications, and electrolyte abnormalities correctable with supportive therapy; and any toxicity resulting in treatment delay of more than 2 weeks. Response was assessed every 2 cycles using the Response Evaluation Criteria in Solid Tumors (18).

Pretreatment evaluation and safety assessment

Pretreatment evaluation consisted of a complete medical history, physical examination, vital signs, electrocardiogram, complete blood count (CBC), serum chemistries, prothrombin time/INR, and activated partial thromboplastin time, serum or urine pregnancy test and baseline tumor measurements. On days 1, 8, and 15 of each cycle, evaluation consisted of a brief history and physical examination (day 1), vital signs (day 1), CBC, and serum chemistries.

Dose modification

Patients who experienced any DLT had treatment delayed by 1-week intervals until recovery and then may have continued on study with reduction of decitabine and vorinostat by one dose level. If no recovery occurred after a delay of 3 weeks of the next scheduled treatment cycle, patients were discontinued from protocol treatment. Blood counts must be recovered to begin a new treatment cycle.

Duration of study treatment

Patients with an objective response or stable disease were allowed to remain on study until disease progression. Otherwise, study treatment continued until disease progression, unacceptable adverse event, patient's decision to withdraw consent, or changes in the patient's condition that rendered the continuation of study treatment unacceptable.

Pharmacokinetic analysis

Blood samples for evaluation of decitabine were collected on days 1 and 5 of cycle 1, before dosing, 30 minutes after the infusion had started, at the end of infusion and at 5, 20, 35, 45, and 60 minutes from the end of infusion. Blood was collected into a sodium heparin Vacutainer tube and centrifuged at 1,500 × g for 15 minutes. The resulting plasma was transferred into polypropylene tubes and stored at −70°C until analyzed for decitabine concentration, using a validated high performance liquid chromatography with tandem mass spectrometry (LC-MS/MS) (19). On the sequential schedule, blood samples for vorinostat were collected on day 9 of cycle 1 before dosing and at 0.5, 1, 2, 2.5, 3, 4, 6. and 8 hours after dosing. On the concurrent schedule, blood samples were collected on day 4 of cycle 1 at the same time points. Samples were allowed to clot at 4°C for 20 to 30 minutes, and then centrifuged at 2,000 × g for 15 minutes at 4°C. The resulting serum was transferred to polypropylene cryotubes and stored at −70°C until analyzed for vorinostat concentrations with a validated LC-MS/MS assay (20). PK parameters were calculated by noncompartmental methods using WinNonlin (Version 5.2; Pharsight Corp.)

Exploratory analyses were performed for PK parameters and adjustments made for multiple comparisons. t Tests were used for independent group comparisons and paired t tests were used to compare PK parameters of decitabine on day 1 versus day 5 within a dose level.

Patient characteristics

Forty-four patients were enrolled into this study and 43 received treatment for a total of 136 cycles (median 2, range: 1–25; Table 1). One patient with neuroendocrine carcinoma of the pancreas did not receive treatment because baseline CT scans showed no significant growth of disease.

Table 1.

Patient characteristics

CharacteristicPatients
n%
No of patients 44  
Median age, y (Range) 62 (31–77)  
Sex   
 Female 18 40 
 Male 26 60 
ECOG PS   
 0 12 27 
 1 29 66 
 2 
Primary tumor type   
 Colorectal 11 25 
 NHL 
 Breast 
 Melanoma 
 Cholangiocarcinoma 
 NSCLC 
 Duodenal 
 Mesothelioma 
 Appendix 
 Uterine (adenocarcinoma and sarcoma) 
 Stomach 
 Pancreas (mucinous cystic carcinoma and neuroendocrine) 
 Fallopian tube 
 Thymus 
 Liver 
 Ovarian (Sertoli-Leyding cell) 
 Parotid (Non–small-cell and adenoid cystic) 
 Sweat gland adenocarcinoma 
 Lacrimal gland (adenoid cystic) 
 Submandibolar gland (adenoid cystic) 
 Nasal cavity (adenoid cystic) 
 Oral cavity (adenoid cystic) 
No. of prior systemic treatments   
 0 
 1 18 
 2 20 
 3 10 23 
 ≥4 14 32 
CharacteristicPatients
n%
No of patients 44  
Median age, y (Range) 62 (31–77)  
Sex   
 Female 18 40 
 Male 26 60 
ECOG PS   
 0 12 27 
 1 29 66 
 2 
Primary tumor type   
 Colorectal 11 25 
 NHL 
 Breast 
 Melanoma 
 Cholangiocarcinoma 
 NSCLC 
 Duodenal 
 Mesothelioma 
 Appendix 
 Uterine (adenocarcinoma and sarcoma) 
 Stomach 
 Pancreas (mucinous cystic carcinoma and neuroendocrine) 
 Fallopian tube 
 Thymus 
 Liver 
 Ovarian (Sertoli-Leyding cell) 
 Parotid (Non–small-cell and adenoid cystic) 
 Sweat gland adenocarcinoma 
 Lacrimal gland (adenoid cystic) 
 Submandibolar gland (adenoid cystic) 
 Nasal cavity (adenoid cystic) 
 Oral cavity (adenoid cystic) 
No. of prior systemic treatments   
 0 
 1 18 
 2 20 
 3 10 23 
 ≥4 14 32 

All patients had progressed from previous treatments for advanced disease. The median number of prior systemic treatment lines was 3. Three patients were treated previously with radiation therapy only and had not received systemic therapy.

A total of 43 patients received at least 1 cycle of study treatment. At the time of data cut off (March 2010), treatment was discontinued due to radiologically confirmed progression or due to symptomatic deterioration caused by underlying disease in 37 patients; 5 patients discontinued due to adverse events related to study treatment; 1 patient with stable disease after 25 cycles remains on study. There were no treatment-related deaths.

Dose escalation and dose-limiting toxicities

Six dose levels were evaluated on the sequential and 3 on the concurrent schedule (Fig. 1). On the sequential schedule, 8 of 30 (27%) evaluable patients experienced at least 1 DLT. The nature of DLTs was as follows: hematologic in 5 patients, nonhematologic in 2, and both hematologic and nonhematologic in 1 patient. Among hematologic DLTs, grade 4 thrombocytopenia occurred in 4 patients, grade 4 neutropenia lasting 7 days or more occurred in 2 and febrile neutropenia occurred in 2 patients. There was a clear association between hematologic DLTs and higher doses of decitabine. In fact, among 14 patients treated at the 2 dose levels with decitabine given at 10 mg/m2/day, only 1 heavily pretreated patient with NHL who had a prior autotransplant developed a hematologic DLT, consisting of grade 4 thrombocytopenia. Regarding nonhematologic DLTs, 1 patient encountered grade 2 intolerable fatigue, anorexia, and dehydration, 1 had grade 3 fatigue, and 1 had grade 3 constipation (plus febrile neutropenia and grade 4 thrombocytopenia).

Figure 1.

Dose escalation scheme for the sequential and concurrent schedules and specification of DLTs at each dose level.

Figure 1.

Dose escalation scheme for the sequential and concurrent schedules and specification of DLTs at each dose level.

Close modal

Among 12 patients treated on the concurrent schedule, four (33%) developed a DLT. Three patients had nonhematologic DLTs (1 grade 3 fatigue and 2 grade 3 fatigue and grade 3 dehydration), and 1 patient experienced a hematologic DLT (grade 3 febrile neutropenia).

Safety and compliance

Neutropenia and thrombocytopenia were the most frequent adverse events of at least possible attribution to study treatment (Table 2). Fatigue, nausea, diarrhea, and vomiting were the most frequent nonhematologic adverse events (Table 2). Seven patients experienced more than 1 episode of grade 3 fatigue whereas other nonhematologic adverse events were mostly grade 1 or 2.

Table 2.

Selected adverse events at least possibly related to study treatment by schedule and dose level, reported as percentages of patients with at least one occurrence of the event over the total number of patients treated in the respective dose level (frequencies presented refer to all treatment cycles)

Sequential scheduleConcurrent schedule
Adverse events and maximum gradeDL1DL-1DL1aDL1bDL-1bDL-2bDL-2bDL-2cDL-3c
%%%%%%%%%
Neutropenia          
 1/2 33 33 67 25 71 57 57 100 50 
 3/4 100 100 100 25 29 43 29 33 
Thrombocytopenia          
 1/2 66 100 67 25 29 71 57 33 
 3/4 33 33 50 
Fatigue          
 1/2 50 66 67 25 71 57 43 33 
 3/4 17 14 14 14 33 100 
Nausea          
 1/2 33 33 67 100 43 57 67 50 
 3/4 25 
Vomiting          
 1/2 33 33 25 57 29 29 67 50 
 3/4 
Diarrhea          
 1/2 33 67 71 43 14 
 3/4 
Hyponatremia          
 1/2 50 43 29 43 33 50 
 3/4 50 
Dehydration          
 1/2 17 100 
 3/4 17 100 
Febrile neutropenia          
 1/2 
 3/4 50 33 
Myalgia          
 1/2 14 14 
 3/4 14 
Hypotension          
 1/2 14 100 
 3/4 
Sequential scheduleConcurrent schedule
Adverse events and maximum gradeDL1DL-1DL1aDL1bDL-1bDL-2bDL-2bDL-2cDL-3c
%%%%%%%%%
Neutropenia          
 1/2 33 33 67 25 71 57 57 100 50 
 3/4 100 100 100 25 29 43 29 33 
Thrombocytopenia          
 1/2 66 100 67 25 29 71 57 33 
 3/4 33 33 50 
Fatigue          
 1/2 50 66 67 25 71 57 43 33 
 3/4 17 14 14 14 33 100 
Nausea          
 1/2 33 33 67 100 43 57 67 50 
 3/4 25 
Vomiting          
 1/2 33 33 25 57 29 29 67 50 
 3/4 
Diarrhea          
 1/2 33 67 71 43 14 
 3/4 
Hyponatremia          
 1/2 50 43 29 43 33 50 
 3/4 50 
Dehydration          
 1/2 17 100 
 3/4 17 100 
Febrile neutropenia          
 1/2 
 3/4 50 33 
Myalgia          
 1/2 14 14 
 3/4 14 
Hypotension          
 1/2 14 100 
 3/4 

Treatment delay due to related adverse events was calculated in patients who received 2 or more cycles of treatment: on the sequential schedule, a treatment delay for 1 occasion or more occurred in 4 of 4 (100%), 2 of 2 (100%), 2 of 2 (100%), 0 of 2 (0%), 1 of 5 (20%), and 3 of 5 (60%) patients, on dose levels 1, −1, 1a, 1b, −1b, and −2b, respectively. Among patients treated on the concurrent schedule, 1 or more treatment delay occurred in 2 of 6 (33%), 1 of 1 (100%), and 1 of 1 (100%) patients on dose levels −2b, −3b, and −3c respectively.

Dose reductions or omissions occurred as follows: on the sequential schedule 4 of 6 (66%), 1 of 3 (33%), 2 of 4 (50%), 1 of 4 (25%), 2 of 7 (29%), 1 of 7 (14%) patients on dose levels 1, −1, 1a, 1b, −1b, and −2b, respectively, and on the concurrent schedule 0 of 7 (0%), 2 of 3 (66%), and 2 of 2 (100%) patients on dose levels −2b, −3b, and −3c, respectively.

Three patients treated on the sequential schedule (10%) discontinued the study due to adverse events possibly related to the treatment. These consisted of fatigue and nausea in one, nausea and neutropenia in another, and nausea and vomiting in the third. On the concurrent schedule, 2 of 12 (17%) patients discontinued treatment due to treatment related adverse events. One event was fatigue, whereas the other patient developed skin changes that were biopsied and showed a neutrophilic dermatitis consistent with Sweet syndrome, a rare syndrome that has previously been described in patients with myelodysplastic syndrome/acute leukaemia treated with the combination of HDACi and DNMTi (21). After discontinuation of treatment and a course of oral prednisone, the skin changes resolved.

Recommended phase II dose

Based on the occurrence of DLTs reported previously, the MTD for the sequential combination was decitabine 10 mg/m2/day on days 1 to 5 with vorinostat 200 mg 3 times a day on days 6 to 12. The MTD for the concurrent schedule was decitabine 10 mg/m2/day on days 1 to 5 with vorinostat 200 mg 2 times a day on days 3 to 9. Among the sequential schedules, the highest dose with the least treatment delay was achieved in dose level −1b (decitabine 10 mg/m2/day on days 1–5 with vorinostat 200 mg twice daily, days 6–12). This is also the dose level with the highest percentage of patients with stable disease for 4 cycles or more. In fact, 4 of the 11 patients (36%) who remained on study for 4 or more cycles were treated at this dose level. Thus, this dose level represents the recommended phase II dose (RP2D) for the combination of the two drugs.

Antitumor activity

No objective tumor responses were observed. Among 38 patients evaluable for response, 11 (29%) with previously progressive cancers had stable disease for 4 or more cycles of treatment. Three of 11 had stable disease for 8 or more cycles, including 1 patient with colon cancer (8 cycles), 1 patient with thymoma (11 cycles), and 1 patient with mucinous adenocarcinoma of the appendix who remains on study after 25 cycles (Fig. 2).

Figure 2.

Number of cycles of study treatment per tumor type and dose level. DL, dose level; NSC, non small cell; MZL, marginal zone lymphoma; DLBCL, diffuse large B-cell lymphoma; SLL, small lymphocytic lymphoma; NSCLC, non–small-cell lung cancer; ALCL, anaplastic large cell lymphoma.

Figure 2.

Number of cycles of study treatment per tumor type and dose level. DL, dose level; NSC, non small cell; MZL, marginal zone lymphoma; DLBCL, diffuse large B-cell lymphoma; SLL, small lymphocytic lymphoma; NSCLC, non–small-cell lung cancer; ALCL, anaplastic large cell lymphoma.

Close modal

PK analyses

PK results (Tables 3 and 4) of decitabine were similar to those reported in previous studies (22). Decitabine is rapidly eliminated with a half-life of approximately 20 minutes. On the sequential schedule, increasing doses of decitabine did not produce dose proportional increases in maximum plasma concentration (Cmax) and area under the plasma concentration–time curve (AUC). No significant differences were found when comparing PK parameters on day 1 and day 5 within the same dose level in either schedule. In addition, there were no statistically significant differences when comparing PK parameters for the dose of 10 mg/m2/day between the 2 schedules of administration.

Table 3.

Decitabine PK parameters

Sequential schedule
Dose (no.of pts)10 mg/m2 (12)15 mg/m2 (3)20 mg/m2 (4)
PK parameterDay 1Day 5Day 1Day 5Day 1Day 5
Cmax (ng/mL) 57.5 (27.5) 67.5 (24.5) 129.03 (74.25) 142.33 (26.16) 134.88 (31.52) 198.00 (42.86) 
AUCt (ng.h/mL) 47.2 (22.0) 55.8 (21.0) 112.11 (67.59) 113.99 (18.45) 115.38 (18.58) 160.67 (40.40) 
AUCinf (ng.h/mL) 48.4 (22.1) 57.8 (21.4) 115.68 (69.62) 116.33 (19.10) 119.30 (18.24) 164.55 (41.52) 
t1/2 (h) 0.34 (0.11) 0.38 (0.14) 0.38 (0.05) 0.33 (0.03) 0.36 (0.06) 0.42 (0.11) 
CL (l/h) 456 (217) 368 (170) 340 (270) 236 (37) 317 (81) 235 (73) 
   Concurrent schedule    
Dose [no. of pts] 10 mg/m2 [12] 
PK parameter Day 1 Day 5     
Cmax (ng/mL) 88.63 (43.34) 62.91 (18.36)     
AUCt (ng.h/mL) 75.78 (49.59) 56.27 (11.68)     
AUCinf (ng.h/mL) 77.57 (51.12) 57.44 (11.68)     
t1/2 (h) 0.27 (0.06) 0.27 (0.07)     
CL(l/h) 280 (79) 334 (80)     
Sequential schedule
Dose (no.of pts)10 mg/m2 (12)15 mg/m2 (3)20 mg/m2 (4)
PK parameterDay 1Day 5Day 1Day 5Day 1Day 5
Cmax (ng/mL) 57.5 (27.5) 67.5 (24.5) 129.03 (74.25) 142.33 (26.16) 134.88 (31.52) 198.00 (42.86) 
AUCt (ng.h/mL) 47.2 (22.0) 55.8 (21.0) 112.11 (67.59) 113.99 (18.45) 115.38 (18.58) 160.67 (40.40) 
AUCinf (ng.h/mL) 48.4 (22.1) 57.8 (21.4) 115.68 (69.62) 116.33 (19.10) 119.30 (18.24) 164.55 (41.52) 
t1/2 (h) 0.34 (0.11) 0.38 (0.14) 0.38 (0.05) 0.33 (0.03) 0.36 (0.06) 0.42 (0.11) 
CL (l/h) 456 (217) 368 (170) 340 (270) 236 (37) 317 (81) 235 (73) 
   Concurrent schedule    
Dose [no. of pts] 10 mg/m2 [12] 
PK parameter Day 1 Day 5     
Cmax (ng/mL) 88.63 (43.34) 62.91 (18.36)     
AUCt (ng.h/mL) 75.78 (49.59) 56.27 (11.68)     
AUCinf (ng.h/mL) 77.57 (51.12) 57.44 (11.68)     
t1/2 (h) 0.27 (0.06) 0.27 (0.07)     
CL(l/h) 280 (79) 334 (80)     

NOTE: Values reported: Mean (SD)

Abbreviations: t1/2, terminal half-life; AUCt, area under the plasma decitabine concentration versus time curve to the last sampling time; AUCinf, area under the plasma decitabine concentration versus time curve from zero to infinity; CL, clearance.

Table 4.

Vorinostat PK parameters

Sequential schedule
Dose (no. of pts)100 mg BID (13)200 mg BID (10)200 mg TID (7)
PK parameter 
Cmax (ng/mL) 165 (88) 313 (130) 293 (150) 
AUCt (ng.h/mL) 424 (163) 1,021 (251) 1,086 (619) 
AUCinf (ng.h/mL) 446 (158) 1,184 (276) 1,352 (873) 
t1/2 (h) 1.39 (0.48) 2.55 (1.99) 1.71 (0.94) 
CL/F (ml/min) 4.3 (2.3) 3 (0.7) 4 (3.7) 
 Concurrent schedule 
dose (no. of pts) 200 mg BID (10) 200 mg TID (2) 
PK parameter 
Cmax (ng/mL)  214 (114) 227 (378) 
AUCt (ng.h/mL)  573 (216) 883 (1,518) 
AUCinf (ng.h/mL)  708 (235) NA 
t1/2 (h)  2.39 (1.45) NA 
CL/F (mL/min)  5.3 (2.3) NA 
Sequential schedule
Dose (no. of pts)100 mg BID (13)200 mg BID (10)200 mg TID (7)
PK parameter 
Cmax (ng/mL) 165 (88) 313 (130) 293 (150) 
AUCt (ng.h/mL) 424 (163) 1,021 (251) 1,086 (619) 
AUCinf (ng.h/mL) 446 (158) 1,184 (276) 1,352 (873) 
t1/2 (h) 1.39 (0.48) 2.55 (1.99) 1.71 (0.94) 
CL/F (ml/min) 4.3 (2.3) 3 (0.7) 4 (3.7) 
 Concurrent schedule 
dose (no. of pts) 200 mg BID (10) 200 mg TID (2) 
PK parameter 
Cmax (ng/mL)  214 (114) 227 (378) 
AUCt (ng.h/mL)  573 (216) 883 (1,518) 
AUCinf (ng.h/mL)  708 (235) NA 
t1/2 (h)  2.39 (1.45) NA 
CL/F (mL/min)  5.3 (2.3) NA 

NOTE: Values reported: Mean (SD)

Abbreviations: t1/2, terminal half-life; AUCt, area under the plasma vorinostat concentration versus time curve to the last sampling time; AUCinf, area under the plasma vorinostat concentration versus time curve from zero to infinity; CL, clearance.

A comparison of vorinostat PK parameters with increasing doses was possible only for the sequential schedule. AUC and Cmax after a single vorinostat administration increased proportionaly when the dose of vorinostat was increased from 100 mg to 200 mg. Interestingly, Cmax, AUCinf ( = AUC to infinity), and AUCt ( = AUC over dosing interval) were lower in the concurrent schedule than those in the sequential schedule (P = 0.09, 0.004, and 0.0005, respectively) at the 200 mg BID dose level. The difference in the AUCt remains statistically significant after adjustment for multiple comparisons (Supplementary Figure A1).

Aberrant DNA methylation and histone deacetylation are involved in tumor formation and progression and have been evaluated as targets for the development of anticancer agents (1, 2). The possibility of optimally reexpressing methylated genes following treatment with the combination of a DNMTi with an HDACi has been confirmed in preclinical studies and formed the basis for clinical trials using combined epigenetic therapies (23).

Here we report for the first time the results of a phase I trial demonstrating the feasibility of delivering decitabine in combination with vorinostat in patients with advanced solid tumors or NHLs. Decitabine given for 5 days at a dose of 10 mg/m2/day as a 1-hour intravenous infusion can be combined with oral vorinostat either on a sequential (vorinostat 200 mg 3 times a day on days 6–12) or a concurrent schedule (vorinostat 200 mg 2 times a day on days 3–9). The toxicities observed were predictable and manageable at these stated MTD doses. In both schedules, the combination of decitabine and vorinostat appears to have a narrow therapeutic index and both drugs required dose reductions from their single-agent recommended doses used in previous studies of hematologic malignancies and solid tumors. However, the optimal single-agent doses of decitabine and vorinostat in patients with solid tumors remain unknown and there is no clear evidence that higher doses are associated with better outcome.

Among the 2 schedules evaluated in this study, the sequential schedule appears to be easier to deliver and more tolerable. In addition, PK analyses showed that for the same dose of vorinostat, AUC, and Cmax were lower for the concurrent schedule, suggesting a possible unfavorable PK interaction between the 2 drugs. However, the number of patients enrolled in our study was small and the study was not designed to establish if this was due to increased metabolism or reduced absorption of vorinostat. Notably, escalation of vorinostat to 200 mg thrice a day on the concurrent schedule resulted in increased toxicities with 2 of 2 patients developing DLTs. Within the sequential schedule, dose level −1b (decitabine 10 mg/m2/day on days 1–5 and vorinostat 200 mg twice a day on days 6–12) was more favorable in dose delivery without delays, and therefore it represents the dose we recommend for further phase II evaluation. Dose level −1b on the sequential schedule also had the highest percentage of patients achieving stable disease for 4 cycles or more. Although this represents only a small number of patients, rendering it impossible to draw any definitive conclusion regarding superiority of any of the dose levels studied, we believe this dose level warrants further investigation in clinical trials.

Combination therapies employing DNMTi or HDACi with other agents are being pursued clinically (8, 9). A small number of clinical trials have evaluated different combinations of epigenetic agents in patients with hematologic and, more recently, solid malignancies (22, 24–27). There is optimism that combined epigenetic therapy can result in increased antitumor activity in comparison to the use of single-agent DNMTi or HDACi, but this needs validation in randomized studies. In our study we observed stable disease in previously progressing patients with different tumor types, but it is not possible to establish if this is due to the combination of the 2 agents and what the expected outcomes would be if each agent was used alone. Decitabine and vorinostat are active in hematologic malignancies and cutaneous T-cell lymphomas, respectively, but their role in the treatment of solid tumors remains undefined. Among the 2 agents, vorinostat has shown single-agent antitumor activity with reports of stable disease and a few cases of partial responses in patients with different types of solid tumors (16, 28).

This was a small phase I study and it is difficult to speculate based on its results in which tumor types this combination should be further explored. Recently reported studies of vorinostat in patients with solid tumors as single-agent or in combination with chemotherapy have shown preliminary evidence of activity in non–small-cell lung, breast, colorectal, mesothelioma, thyroid, and adenoid cystic carcinoma (16, 28–30). It is therefore reasonable to consider further clinical investigation of this combination in the context of the aforementioned malignancies.

No potential conflicts of interests were disclosed.

Supported by NCI Grants. U01CA132123, U01CA099168, and P30-CA47904

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Esteller
M
. 
Epigenetics in cancer
.
N Engl J Med
2008
;
358
:
1148
59
.
2.
Jones
PA
,
Baylin
SB
. 
The epigenomics of cancer
.
Cell
2007
;
128
:
683
92
.
3.
Piekarz
RL
,
Bates
SE
. 
Epigenetic modifiers: basic understanding and clinical development
.
Clin Cancer Res
2009
;
15
:
3918
26
.
4.
Wilson
VL
,
Jones
PA
,
Momparler
RL
. 
Inhibition of DNA methylation in L1210 leukemic cells by 5-aza-2′-deoxycytidine as a possible mechanism of chemotherapeutic action
.
Cancer Res
1983
;
43
:
3493
6
.
5.
Finnin
MS
,
Donigian
JR
,
Cohen
A
,
Richon
VM
,
Rifkind
RA
,
Marks
PA
, et al
Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors
.
Nature
1999
;
401
:
188
93
.
6.
Kantarjian
H
,
Issa
JP
,
Rosenfeld
CS
,
Bennett
JM
,
Albitar
M
,
DiPersio
J
, et al
Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study
.
Cancer
2006
;
106
:
1794
803
.
7.
Mann
BS
,
Johnson
JR
,
Cohen
MH
,
Justice
R
,
Pazdur
R
. 
FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma
.
Oncologist
2007
;
12
:
1247
52
.
8.
Lane
AA
,
Chabner
BA
. 
Histone deacetylase inhibitors in cancer therapy
.
J Clin Oncol
2009
;
27
:
5459
68
.
9.
Issa
JP
,
Kantarjian
HM
. 
Targeting DNA methylation
.
Clin Cancer Res
2009
;
15
:
3938
46
.
10.
Cameron
EE
,
Bachman
KE
,
Myohanen
S
,
Herman
JG
,
Baylin
SB
. 
Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer
.
Nat Genet
1999
;
21
:
103
7
.
11.
Kikuchi
T
,
Toyota
M
,
Itoh
F
,
Suzuki
H
,
Obata
T
,
Yamamoto
H
, et al
Inactivation of p57KIP2 by regional promoter hypermethylation and histone deacetylation in human tumors
.
Oncogene
2002
;
21
:
2741
9
.
12.
Boivin
AJ
,
Momparler
LF
,
Hurtubise
A
,
Momparler
RL
. 
Antineoplastic action of 5-aza-2′-deoxycytidine and phenylbutyrate on human lung carcinoma cells
.
Anticancer Drugs
2002
;
13
:
869
74
.
13.
Zhu
WG
,
Lakshmanan
RR
,
Beal
MD
,
Otterson
GA
. 
DNA methyltransferase inhibition enhances apoptosis induced by histone deacetylase inhibitors
.
Cancer Res
2001
;
61
:
1327
33
.
14.
Yang
H
,
Hoshino
K
,
Sanchez-Gonzalez
B
,
Kantarjian
H
,
Garcia-Manero
G
. 
Antileukemia activity of the combination of 5-aza-2′-deoxycytidine with valproic acid
.
Leuk Res
2005
;
29
:
739
48
.
15.
Kantarjian
HM
,
O'Brien
S
,
Shan
J
,
Aribi
A
,
Garcia-Manero
G
,
Jabbour
E
, et al
Update of the decitabine experience in higher risk myelodysplastic syndrome and analysis of prognostic factors associated with outcome
.
Cancer
2007
;
109
:
265
73
.
16.
Kelly
WK
,
O'Connor
OA
,
Krug
LM
,
Chiao
JH
,
Heaney
M
,
Curley
T
, 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
.
17.
Garcia-Manero
G
,
Yang
H
,
Bueso-Ramos
C
,
Ferrajoli
A
,
Cortes
J
,
Wierda
WG
, 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
.
18.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
19.
Liu
Z
,
Marcucci
G
,
Byrd
JC
,
Grever
M
,
Xiao
J
,
Chan
KK
. 
Characterization of decomposition products and preclinical and low dose clinical pharmacokinetics of decitabine (5-aza-2′-deoxycytidine) by a new liquid chromatography/tandem mass spectrometry quantification method
.
Rapid Commun Mass Spectrom
2006
;
20
:
1117
26
.
20.
Parise
RA
,
Holleran
JL
,
Beumer
JH
,
Ramalingam
S
,
Egorin
MJ
. 
A liquid chromatography-electrospray ionization tandem mass spectrometric assay for quantitation of the histone deacetylase inhibitor, vorinostat (suberoylanilide hydroxamicacid, SAHA), and its metabolites in human serum
.
J Chromatogr B Analyt Technol Biomed Life Sci
2006
;
840
:
108
15
.
21.
Alencar
C
,
Abramowtiz
M
,
Parekh
S
,
Braunshweig
I
,
Jacobson
M
,
Silverman
L
, et al
Atypical presentations of Sweet's syndrome in patients with MDS/AML receiving combinations of hypomethylating agents with histone deacetylase inhibitors
.
Am J Hematol
2009
;
84
:
688
9
.
22.
Blum
W
,
Klisovic
RB
,
Hackanson
B
,
Liu
Z
,
Liu
S
,
Devine
H
, et al
Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia
.
J Clin Oncol
2007
;
25
:
3884
91
.
23.
Bots
M
,
Johnstone
RW
. 
Rational combinations using HDAC inhibitors
.
Clin Cancer Res
2009
;
15
:
3970
7
.
24.
Garcia-Manero
G
,
Kantarjian
HM
,
Sanchez-Gonzalez
B
,
Yang
H
,
Rosner
G
,
Verstovsek
S
, et al
Phase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemia
.
Blood
2006
;
108
:
3271
9
.
25.
Gore
SD
,
Baylin
S
,
Sugar
E
,
Carraway
H
,
Miller
CB
,
Carducci
M
, et al
Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms
.
Cancer Res
2006
;
66
:
6361
9
.
26.
Braiteh
F
,
Soriano
AO
,
Garcia-Manero
G
,
Hong
D
,
Johnson
MM
,
Silva
Lde P
, et al
Phase I study of epigenetic modulation with 5-azacytidine and valproic acid in patients with advanced cancers
.
Clin Cancer Res
2008
;
14
:
6296
301
.
27.
Lin
J
,
Gilbert
J
,
Rudek
MA
,
Zwiebel
JA
,
Gore
S
,
Jiemjit
A
, et al
A phase I dose-finding study of 5-azacytidine in combination with sodium phenylbutyrate in patients with refractory solid tumors
.
Clin Cancer Res
2009
;
15
:
6241
9
.
28.
Luu
TH
,
Morgan
RJ
,
Leong
L
,
Lim
D
,
McNamara
M
,
Portnow
J
, et al
A phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California Cancer Consortium study
.
Clin Cancer Res
2008
;
14
:
7138
42
.
29.
Ramalingam
SS
,
Maitland
ML
,
Frankel
P
,
Argiris
AE
,
Koczywas
M
,
Gitlitz
B
, et al
Carboplatin and Paclitaxel in combination with either vorinostat or placebo for first-line therapy of advanced non-small-cell lung cancer
.
J Clin Oncol
2010
;
28
:
56
62
.
30.
Fakih
MG
,
Fetterly
G
,
Egorin
MJ
,
Muindi
JR
,
Espinoza-Delgado
I
,
Zwiebel
JA
, et al
A phase I, pharmacokinetic, and pharmacodynamic study of two schedules of vorinostat in combination with 5-fluorouracil and leucovorin in patients with refractory solid tumors
.
Clin Cancer Res
2010
;
16
:
3786
94
.

Supplementary data