Skip to main content
The Oncologist logoLink to The Oncologist
. 2016 Jun 3;21(7):785–786g. doi: 10.1634/theoncologist.2016-0060

A Phase II Trial of a Histone Deacetylase Inhibitor Panobinostat in Patients With Low-Grade Neuroendocrine Tumors

Ning Jin a,, Sam J Lubner a,b, Daniel L Mulkerin a,b, Saurabh Rajguru a, Lakeesha Carmichael b,c, Herb Chenv b,d, Kyle D Holen a,b, Noelle K LoConte a,b
PMCID: PMC4943400  PMID: 27261467

Abstract

Lessons Learned

  • Pancreatic neuroendocrine tumors versus carcinoid tumors should be examined separately in clinical trials.

  • Progression-free survival is more clinically relevant as the primary endpoint (rather than response rate) in phase II trials for low-grade neuroendocrine tumors.

Background.

The most common subtypes of neuroendocrine tumors (NETs) are pancreatic islet cell tumors and carcinoids, which represent only 2% of all gastrointestinal malignancies. Histone deacetylase (HDAC) inhibitors have already been shown to suppress tumor growth and induce apoptosis in various malignancies. In NET cells, HDAC inhibitors have resulted in increased Notch1 expression and subsequent inhibition of growth. We present here a phase II study of the novel HDAC inhibitor panobinostat in patients with low-grade NET.

Methods.

Adult patients with histologically confirmed, metastatic, low-grade NETs and an Eastern Cooperative Oncology Group (ECOG) performance status of ≤2 were treated with oral panobinostat 20 mg once daily three times per week. Treatment was continued until patients experienced unacceptable toxicities or disease progression. The study was stopped at planned interim analysis based on a Simon two-stage design.

Results.

Fifteen patients were accrued, and 13 were evaluable for response. No responses were seen, but the stable disease rate was 100%. The median progression-free survival (PFS) was 9.9 months, and the median overall survival was 47.3 months. Fatigue (27%), thrombocytopenia (20%), diarrhea (13%), and nausea (13%) were the most common related grade 3 toxicities. There was one grade 4 thrombocytopenia (7%). These results did not meet the prespecified criteria to open the study to full accrual.

Conclusion.

The HDAC inhibitor panobinostat has a high stable disease rate and reasonable PFS in low-grade NET, but has a low response rate.

Discussion

Therapeutic options for advanced low-grade NET are limited. First-line therapy involves a somatostatin analog, such as octreotide long-acting repeatable. Biologic therapies targeting mTOR pathway and vascular endothelial growth factor have been approved for the treatment of patients with well-differentiated pancreatic NET: sunitinib and everolimus, respectively. Both agents demonstrated the benefit of prolongation of PFS.

Panobinostat is a potent, orally active, pan-HDAC inhibitor that can affect multiple cancer-related pathways, including cell-cycle regulation, differentiation, and apoptosis. Several preclinical studies indicated that HDAC inhibitors, valproic acid and suberoyl bishydroxamic acid, can activate Notch1 signaling, suppress NET tumor markers, and inhibit NET cell growth. Based on the evidence that Notch1 activation can lead to NET differentiation and suppression of tumor growth, we opened a phase II clinical trial of panobinostat in patients with metastatic low-grade NETs.

The total number of subjects initially planned in this study was 33. An interim analysis was planned when 13 evaluable patients had been accrued. All patients had stable disease as best response (Fig. 1). Because of lack of objective response, the study was closed to accrual. The median progression-free survival was 9.9 months (90% confidence interval [CI], 4.1–16.9), and the median overall survival was 47.27 months (90% CI, 17.87 to not reached), with the total follow-up time of 5 years. Panobinostat was tolerated relatively well in patients in our study. Fatigue, thrombocytopenia, anorexia, diarrhea, and nausea were the most common grade 3 treatment related toxicities (Adverse Events Table). Patients with pancreatic NETs (4 of 5 patients, 80%) underwent more than 10 cycles of panobinostat in this study. This would seem to be a clinically meaningful delay in time to progression of the cancer. It is increasingly clear that pancreatic NETs and carcinoid subtypes have different biology, respond differently to therapeutic agents, and should be evaluated as separate entities in clinical trials.

Figure 1.

Figure 1.

Kaplan-Meier curve for median progression-free survival, which is 9.90 months with 90% confidence interval (4.10–16.9 months).

Our study was terminated early because of the use of objective response rate as the primary outcome measure, which is the shortcoming of this study. We would use progression-free survival (PFS) as the primary endpoint if the study should be repeated. Overall survival is not a practical endpoint for advanced NET studies.

Panobinostat showed favorable clinical activity in different hematologic malignancies, such as in relapsed Hodgkin lymphoma, myelofibrosis, refractory cutaneous T-cell lymphoma, and multiple myeloma, as either single-agent or combination treatment. However, HDAC inhibitors have not demonstrated effectiveness in clinical trials involving solid tumors. Their limitations in solid tumors could be related to drug instability because of short protein kinase half-life, tissue impermeability in tumor microenvironment, drug resistance due to activation of signal transducers and activators of transcription signaling pathway, antiapoptotic effect of nuclear transcription nuclear factor κB, and lack of the specific target in solid tumors. The question regarding the role of Notch1 in well-differentiated NET remains unanswered.

Trial Information

Disease

Neuroendocrine – pancreatic

Disease

Neuroendocrine – other

Stage of disease / treatment

Metastatic / Advanced

Prior Therapy

No designated number of regimens

Type of study - 1

Phase II

Type of study - 2

Single Arm

Primary Endpoint

Overall Response Rate

Secondary Endpoint

Progression-Free Survival

Secondary Endpoint

Overall Survival

Secondary Endpoint

Toxicity

Secondary Endpoint

Tolerability

Additional Details of Endpoints or Study Design

The primary endpoint was the tumor response rate (complete or partial response) using Response Evaluation Criteria in Solid Tumors (RECIST) criteria. A Simon optimal two-stage design was used to test the null hypothesis that the true response rate was 6% versus the alternative hypothesis that it was 20%. With a significance level of 10% and a power of 85%, at least one response was required among the first 13 evaluable patients to proceed to the second stage, where additional patients would be enrolled for a total of 30 evaluable patients.

Investigator's Analysis

Level of activity did not meet planned clinical trial endpoint.

Drug Information

Drug 1
Generic/Working name

Panobinostat

Trade name

Farydak

Company name

Novartis

Drug type

Small molecule

Drug class

HDAC

Dose

20 mg per flat dose

Route

Oral (po)

Schedule of Administration

Once daily, three times per week

Patient Characteristics

Number of patients, male

10

Number of patients, female

5

Stage

Metastatic low-grade neuroendocrine tumors

Age

Median (range): 57 (40–80)

Number of prior systemic therapies

Median (range): Not collected

Performance Status: ECOG

0 — 10

1 — 5

2 — 0

3 — 0

Unknown —

Primary Site:

Lung and bronchus, 2

Pancreas, 5

Rectum, 1

Small Intestine, 5

Unknown, 2

Cancer Types or Histologic Subtypes

Low,* 6

Well Differentiated,* 5

Grade unknown, not stated, or not applicable, 4

Primary Assessment Method

Control Arm: Total Patient Population

Number of patients screened

15

Number of patients enrolled

15

Number of patients evaluable for toxicity

15

Number of patients evaluated for efficacy

13

Response assessment CR

n = 0

Response assessment PR

n = 0

Response assessment SD

n = 13

Response assessment PD

n = 0

(Median) duration assessments PFS

9.9 months, CI: 90

(Median) duration assessments OS

47.3 months

Kaplan-Meier time units

Months

graphic file with name theoncologist_1660CTRt1.jpg

Adverse Events

graphic file with name theoncologist_1660CTRt2.jpg

Assessment, Analysis, and Discussion

Completion

Study completed

Terminated reason

Did not fully accrue

Pharmacokinetics / Pharmacodynamics

Not Collected

Investigator's Assessment

Level of activity did not meet planned clinical trial endpoint

NETs are uncommon tumors arising from the neuroendocrine system. The gastrointestinal (GI) tract, pancreas, and lung are the most common primary tumor sites in patients with NETs [1], and gastroenteropancreatic (GEP) NETs represent approximately 2% of all gastrointestinal malignant neoplasms. The clinical evaluation for NETs should incorporate several key factors, such as anatomic site, histology, grade, differentiation, and hormone secretion. In particular, the anatomic site of origin is now recognized as a key determinant of treatment selection [2]. According to the 2010 World Health Organization grading system for GEP NETs, there are three grades (G1, G2, and G3) for differentiation on pathology report, based on Ki-67 and mitotic counts [3]. Well-differentiated NETs include G1 and G2; poorly differentiated NETs are G3.

For patients with well-differentiated and functional tumors, somatostatin analogs such as octreotide long-acting repeatable are the mainstay of treatment for control tumor growth as well as symptomatic control, with improved PFS and quality of life [4, 5]. Recent randomized studies for biologic therapies targeting mTOR pathway and vascular endothelial growth factor demonstrated prolongation of PFS compared with placebo for pancreatic NET; for example, everolimus (11 vs. 4.6 months) [6] or sunitinib (11.4 vs. 5.5 months) [7].

Early studies suggested that activation of the Notch pathway can lead to neuroendocrine differentiation in gastrointestinal carcinoid tumor [8] and inhibit NET cell growth [9]. Of note, it is well recognized that Notch can function as either an oncogene or a tumor suppressor, depending on the cell type [1012]. Both valproic acid (VPA) and suberoyl bishydroxamic acid, two histone deacetylase (HDAC) inhibitors, can activate Notch1 signaling, suppress neuroendocrine tumor markers, and inhibit NET cell growth both in vitro and in vivo [13, 14]. Furthermore, our group previously conducted a pilot clinical trial of VPA for patients with advanced carcinoid cancer. Five of the six patients (62.5%) assessable for radiographic response were noted to have stable disease by RECIST, and one patient with an unconfirmed partial response was noted to have a 40-fold increase in Notch1 mRNA levels [15].

Panobinostat (LBH589) is a potent, orally active, pan-HDAC inhibitor that can affect multiple cancer-related pathways via nonhistone protein targets, including cell-cycle regulation, differentiation, and apoptosis [16, 17]. Based on the role of Notch1 activation in suppression of NET tumor markers and tumor growth, we opened a phase II clinical trial of HDAC inhibitor panobinostat in patients with metastatic low-grade NETs.

The total number of subjects initially planned for this study was 33. An interim analysis was planned when 13 evaluable patients had been accrued. At least one response was required among the first 13 evaluable patients to proceed to the second stage, where additional patients would be enrolled. Among 15 patients who were accrued, 66.6% were male, age range 40–80 years old, 54% carcinoid, and 33% pancreatic NET (Table 1). Because of the lack of objective response in the interim analysis, the study was closed. Panobinostat was tolerated relatively well in patients in our study. The most common toxicities of all grades were thrombocytopenia, fatigue, diarrhea, nausea, hyperglycemia, hypertriglyceridemia, and thyroid dysfunction. Fatigue (27%), thrombocytopenia (20%), anorexia (20%), diarrhea (13%), and nausea (13%) were the most common treatment-related grade 3 toxicities. There was one case (7%) of grade 4 toxicity of thrombocytopenia (Adverse Events Table). Eight patients needed dose modifications because of adverse events, such as thrombocytopenia, neutropenia, and fatigue, during their treatment courses (data not shown). All patients had stable disease as best response (Fig. 1). Three of 13 patients underwent only 2 cycles of treatment, whereas 7 patients underwent more than 10 cycles of treatment (Table 2). Median progression-free survival was 9.9 months (90% CI, 4.1–16.9) (Fig. 2), and median overall survival was 47.27 months (90% CI, 17.87 to not reached), with the total follow-up time of 5 years (Fig. 3).

Table 1.

Patient characteristics

graphic file with name theoncologist_1660CTRt3.jpg

Table 2.

Patient tumor characteristics and treatment outcome

graphic file with name theoncologist_1660CTRt4.jpg

Figure 2.

Figure 2.

Percentage change in tumor size based on Response Evaluation Criteria in Solid Tumors (RECIST) criteria. Waterfall plot of radiographic changes from baseline to best response of 13 evaluable patients, revealing that every patient (100%) has stable disease. Stable disease is defined as neither sufficient shrinkage to qualify for partial response (less than 30% decrease in the sum of the longest diameters of target lesions) nor sufficient increase to qualify for progressive disease (at least 20% increase in the sum of the longest diameters of target lesions).

Figure 3.

Figure 3.

Kaplan-Meier curve for overall survival, which is 47.27 months with 90% confidence interval, with a follow-up time of 5 years.

Abbreviation: CI, confidence interval.

graphic file with name theoncologist_1660CTR_f4.jpg

Kaplan-Meier curve for median progression-free survival, which is 9.90 months with 90% confidence interval (4.10–16.9 months).

Patients with pancreatic NETs (four of five patients; one patient withdrew early) underwent more than 10 cycles of panobinostat in this study, although our sample size is too small to draw any conclusion. It is increasingly clear that pancreatic NETs (PNETs) and carcinoid subtypes have different biology, respond differently to therapeutic agents, and should be evaluated as separate entities in clinical trials [18]. Examination of the PNETs and GI carcinoid demonstrated only a few areas of overlap in the accumulation of genetic aberrations, using comparative genomic hybridization, microsatellite analysis, and sequencing techniques [19]. As to treatment, PNETs are more sensitive to cytotoxic chemotherapy than carcinoid tumors, such as streptozocin, capecitabine, and temozolomide, as shown in early clinical trials as well as recent retrospective clinical studies [2022].

Our study was terminated early because of the use of objective response rate as the primary outcome measure, which is the shortcoming of this study. We would use progression-free survival as the primary endpoint if the study should be repeated. The challenge in phase II studies in NETs is the small patient population available and the often long survival postprogression [23]. Overall survival is not a practical endpoint for advanced NET studies, because of the nature of indolent disease and the availability of multiple sequential therapies [2].

Panobinostat showed favorable clinical activity in different hematologic malignancies, such as in relapsed Hodgkin lymphoma [24], myelofibrosis [25], refractory cutaneous T-cell lymphoma [26], and multiple myeloma [27], as either single-agent or combination treatment. However, the results of recent clinical trials of panobinostat in solid tumors are disappointing, including castration-resistant prostate cancer [28], metastatic renal cell cancer [29], and pancreatic cancer [30]. Generally, HDAC inhibitors have not demonstrated effectiveness in clinical trials involving solid tumors. Their limitations in solid tumors could be related to drug instability due to short protein kinase half-life [31, 32], tissue impermeability in tumor microenvironment, drug resistance due to activation of the signal transducers and activators of transcription signaling pathway [33, 34], antiapoptotic effect of nuclear transcription nuclear factor κB [35], and lack of the specific target in solid tumors [36]. There was no study participant in our trial who underwent pretreatment and posttreatment biopsy for Notch1 activity, which is the limitation of this study.

In conclusion, panobinostat has a high stable disease rate and reasonable PFS in low-grade NET. Oral panobinostat at a dose of 20 mg three times weekly was relatively well tolerated in patients in this study. Four of five patients with PNETs had durable stable disease on panobinostat, which is encouraging. Further studies of panobinostat in combination with other agents are indicated in patients with NETs. The questions regarding off-target effects of panobinostat and the role of Notch1 in well-differentiated NET remain unanswered. In future clinical trials, it is important to develop pharmacodynamics biomarkers to predict treatment response in patients.

Supplementary Material

Data Set

Acknowledgments

This work was supported by University of Wisconsin Carbone Cancer Center Support Grant P30 CA014520 and the Novartis Pharmaceutics Corporation.

Footnotes

ClinicalTrials.gov Identifier: NCT00985946

Sponsor: Novartis

Principal Investigator: Noelle K. LoConte

IRB Approved: Yes

Click here to access other published clinical trials.

Disclosures

Kyle D. Holen: AbbVie (OI). The other authors indicated no financial relationships.

(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board

References

  • 1.Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: Epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072. doi: 10.1200/JCO.2007.15.4377. [DOI] [PubMed] [Google Scholar]
  • 2.Kunz PL. Carcinoid and neuroendocrine tumors: Building on success. J Clin Oncol. 2015;33:1855–1863. doi: 10.1200/JCO.2014.60.2532. [DOI] [PubMed] [Google Scholar]
  • 3.Bosman FT, Carneiro F, Hruban RH, Theise ND, editors. WHO Classification of Tumours of the Digestive System. 4th ed. Geneva, Switzerland: WHO Press; 2010. [Google Scholar]
  • 4.Rinke A, Müller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: A report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–4663. doi: 10.1200/JCO.2009.22.8510. [DOI] [PubMed] [Google Scholar]
  • 5.Toumpanakis C, Caplin ME. Update on the role of somatostatin analogs for the treatment of patients with gastroenteropancreatic neuroendocrine tumors. Semin Oncol. 2013;40:56–68. doi: 10.1053/j.seminoncol.2012.11.006. [DOI] [PubMed] [Google Scholar]
  • 6.Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–513. doi: 10.1056/NEJMoa1003825. [DOI] [PubMed] [Google Scholar]
  • 8.Nakakura EK, Sriuranpong VR, Kunnimalaiyaan M, et al. Regulation of neuroendocrine differentiation in gastrointestinal carcinoid tumor cells by notch signaling. J Clin Endocrinol Metab. 2005;90:4350–4356. doi: 10.1210/jc.2005-0540. [DOI] [PubMed] [Google Scholar]
  • 9.Kunnimalaiyaan M, Yan S, Wong F, et al. Hairy Enhancer of Split-1 (HES-1), a Notch1 effector, inhibits the growth of carcinoid tumor cells. Surgery. 2005;138:1137–1142; discussion 1142. doi: 10.1016/j.surg.2005.05.027. [DOI] [PubMed] [Google Scholar]
  • 10.Sriuranpong V, Borges MW, Ravi RK, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res. 2001;61:3200–3205. [PubMed] [Google Scholar]
  • 11.Radtke F, Raj K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756–767. doi: 10.1038/nrc1186. [DOI] [PubMed] [Google Scholar]
  • 12.Miyamoto Y, Maitra A, Ghosh B, et al. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565–576. doi: 10.1016/s1535-6108(03)00140-5. [DOI] [PubMed] [Google Scholar]
  • 13.Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, et al. Valproic acid activates notch-1 signaling and regulates the neuroendocrine phenotype in carcinoid cancer cells. The Oncologist. 2007;12:942–951. doi: 10.1634/theoncologist.12-8-942. [DOI] [PubMed] [Google Scholar]
  • 14.Greenblatt DY, Cayo M, Ning L, et al. Suberoyl bishydroxamic acid inhibits cellular proliferation by inducing cell cycle arrest in carcinoid cancer cells. J Gastrointest Surg. 2007;11:1515–1520; discussion 1520. doi: 10.1007/s11605-007-0249-1. [DOI] [PubMed] [Google Scholar]
  • 15.Mohammed TA, Holen KD, Jaskula-Sztul R, et al. A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. The Oncologist. 2011;16:835–843. doi: 10.1634/theoncologist.2011-0031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Atadja P. Development of the pan-DAC inhibitor panobinostat (LBH589): Successes and challenges. Cancer Lett. 2009;280:233–241. doi: 10.1016/j.canlet.2009.02.019. [DOI] [PubMed] [Google Scholar]
  • 17.Wagner JM, Hackanson B, Lübbert M, et al. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics. 2010;1:117–136. doi: 10.1007/s13148-010-0012-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine tumors: Consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol. 2011;29:934–943. doi: 10.1200/JCO.2010.33.2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zikusoka MN, Kidd M, Eick G, et al. The molecular genetics of gastroenteropancreatic neuroendocrine tumors. Cancer. 2005;104:2292–2309. doi: 10.1002/cncr.21451. [DOI] [PubMed] [Google Scholar]
  • 20.Murray-Lyon IM, Eddleston AL, Williams R, et al. Treatment of multiple-hormone-producing malignant islet-cell tumour with streptozotocin. Lancet. 1968;2:895–898. doi: 10.1016/s0140-6736(68)91058-1. [DOI] [PubMed] [Google Scholar]
  • 21.Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas. Cancer. 2011;117:268–275. doi: 10.1002/cncr.25425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fine RL, Gulati AP, Krantz BA, et al. Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience. Cancer Chemother Pharmacol. 2013;71:663–670. doi: 10.1007/s00280-012-2055-z. [DOI] [PubMed] [Google Scholar]
  • 23.Yao JC, Lagunes DR, Kulke MH. Targeted therapies in neuroendocrine tumors (NET): Clinical trial challenges and lessons learned. The Oncologist. 2013;18:525–532. doi: 10.1634/theoncologist.2012-0434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Younes A, Sureda A, Ben-Yehuda D, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: Results of a phase II study. J Clin Oncol. 2012;30:2197–2203. doi: 10.1200/JCO.2011.38.1350. [DOI] [PubMed] [Google Scholar]
  • 25.DeAngelo DJ, Mesa RA, Fiskus W, et al. Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis. Br J Haematol. 2013;162:326–335. doi: 10.1111/bjh.12384. [DOI] [PubMed] [Google Scholar]
  • 26.Duvic M, Dummer R, Becker JC, et al. Panobinostat activity in both bexarotene-exposed and -naïve patients with refractory cutaneous T-cell lymphoma: Results of a phase II trial. Eur J Cancer. 2013;49:386–394. doi: 10.1016/j.ejca.2012.08.017. [DOI] [PubMed] [Google Scholar]
  • 27.Richardson PG, Schlossman RL, Alsina M, et al. PANORAMA 2: Panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood. 2013;122:2331–2337. doi: 10.1182/blood-2013-01-481325. [DOI] [PubMed] [Google Scholar]
  • 28.Rathkopf D, Wong BY, Ross RW, et al. A phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancer. Cancer Chemother Pharmacol. 2010;66:181–189. doi: 10.1007/s00280-010-1289-x. [DOI] [PubMed] [Google Scholar]
  • 29.Hainsworth JD, Infante JR, Spigel DR, et al. A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma. Cancer Invest. 2011;29:451–455. doi: 10.3109/07357907.2011.590568. [DOI] [PubMed] [Google Scholar]
  • 30.Wang H, Cao Q, Dudek AZ. Phase II study of panobinostat and bortezomib in patients with pancreatic cancer progressing on gemcitabine-based therapy. Anticancer Res. 2012;32:1027–1031. [PubMed] [Google Scholar]
  • 31.Morita S, Oizumi S, Minami H, et al. Phase I dose-escalating study of panobinostat (LBH589) administered intravenously to Japanese patients with advanced solid tumors. Invest New Drugs. 2012;30:1950–1957. doi: 10.1007/s10637-011-9751-0. [DOI] [PubMed] [Google Scholar]
  • 32.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;23:3923–3931. doi: 10.1200/JCO.2005.14.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bromberg J. Stat proteins and oncogenesis. J Clin Invest. 2002;109:1139–1142. doi: 10.1172/JCI15617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fantin VR, Loboda A, Paweletz CP, et al. Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res. 2008;68:3785–3794. doi: 10.1158/0008-5472.CAN-07-6091. [DOI] [PubMed] [Google Scholar]
  • 35.Fantin VR, Richon VM. Mechanisms of resistance to histone deacetylase inhibitors and their therapeutic implications. Clin Cancer Res. 2007;13:7237–7242. doi: 10.1158/1078-0432.CCR-07-2114. [DOI] [PubMed] [Google Scholar]
  • 36.Slingerland M, Guchelaar HJ, Gelderblom H. Histone deacetylase inhibitors: An overview of the clinical studies in solid tumors. Anticancer Drugs. 2014;25:140–149. doi: 10.1097/CAD.0000000000000040. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Set

Articles from The Oncologist are provided here courtesy of Oxford University Press

RESOURCES