Vorinostat in solid and hematologic malignancies

Journal of Hematology & Oncology, Jul 2009

Vorinostat (Zolinza®), a histone deacetylase inhibitor, was approved by the US Food and Drug Administration in October 2006 for the treatment of cutaneous manifestations in patients with cutaneous T-cell lymphoma who have progressive, persistent or recurrent disease on or following two systemic therapies. This review summarizes evidence on the use of vorinostat in solid and hematologic malignancies and collated tolerability data from the vorinostat clinical trial program. Pooled vorinostat clinical trial data from 498 patients with solid or hematologic malignancies show that vorinostat was well tolerated as monotherapy or combination therapy. The most commonly reported drug-related adverse events (AEs) associated with monotherapy (n = 341) were fatigue (61.9%), nausea (55.7%), diarrhea (49.3%), anorexia (48.1%), and vomiting (32.8%), and Grade 3/4 drug-related AEs included fatigue (12.0%), thrombocytopenia (10.6%), dehydration (7.3%), and decreased platelet count (5.3%). The most common drug-related AEs observed with vorinostat in combination therapy (n = 157, most of whom received vorinostat 400 mg qd for 14 days) were nausea (48.4%), diarrhea (40.8%), fatigue (34.4%), vomiting (31.2%), and anorexia (20.4%), with the majority of AEs being Grade 2 or less. In Phase I trials, combinations with vorinostat were generally well tolerated and preliminary evidence of anticancer activity as monotherapy or in combination with other systemic therapies has been observed across a range of malignancies. Ongoing and planned studies will further evaluate the potential of vorinostat in combination therapy, including combinations with radiation, in patients with diverse malignancy types, including non-small-cell lung cancer, glioblastoma multiforme, multiple myeloma, and myelodysplastic syndrome.

A PDF file should load here. If you do not see its contents the file may be temporarily unavailable at the journal website or you do not have a PDF plug-in installed and enabled in your browser.

Alternatively, you can download the file locally and open with any standalone PDF reader:

http://www.jhoonline.org/content/pdf/1756-8722-2-31.pdf

Vorinostat in solid and hematologic malignancies

Journal of Hematology & Oncology Vorinostat in solid and hematologic malignancies David Siegel 2 Mohamad Hussein 1 Chandra Belani 0 Francisco Robert 6 Evanthia Galanis 5 Victoria M Richon 4 Jos Garcia-Vargas 4 Cesar Sanz- Rodriguez 3 Syed Rizvi 4 0 Penn State Cancer Institute , Hershey, PA , USA 1 H. Lee Moffitt Cancer Center , Tampa, FL , USA 2 Hackensack University Medical Center , Hackensack, NJ , USA 3 Merck Research Laboratories , Madrid , Spain 4 Merck Research Laboratories , Upper Gwynedd, PA , USA 5 Mayo Clinic College of Medicine , Rochester, MN , USA 6 University of Alabama , Birmingham, AL , USA Vorinostat (Zolinza), a histone deacetylase inhibitor, was approved by the US Food and Drug Administration in October 2006 for the treatment of cutaneous manifestations in patients with cutaneous T-cell lymphoma who have progressive, persistent or recurrent disease on or following two systemic therapies. This review summarizes evidence on the use of vorinostat in solid and hematologic malignancies and collated tolerability data from the vorinostat clinical trial program. Pooled vorinostat clinical trial data from 498 patients with solid or hematologic malignancies show that vorinostat was well tolerated as monotherapy or combination therapy. The most commonly reported drug-related adverse events (AEs) associated with monotherapy (n = 341) were fatigue (61.9%), nausea (55.7%), diarrhea (49.3%), anorexia (48.1%), and vomiting (32.8%), and Grade 3/4 drug-related AEs included fatigue (12.0%), thrombocytopenia (10.6%), dehydration (7.3%), and decreased platelet count (5.3%). The most common drug-related AEs observed with vorinostat in combination therapy (n = 157, most of whom received vorinostat 400 mg qd for 14 days) were nausea (48.4%), diarrhea (40.8%), fatigue (34.4%), vomiting (31.2%), and anorexia (20.4%), with the majority of AEs being Grade 2 or less. In Phase I trials, combinations with vorinostat were generally well tolerated and preliminary evidence of anticancer activity as monotherapy or in combination with other systemic therapies has been observed across a range of malignancies. Ongoing and planned studies will further evaluate the potential of vorinostat in combination therapy, including combinations with radiation, in patients with diverse malignancy types, including non-small-cell lung cancer, glioblastoma multiforme, multiple myeloma, and myelodysplastic syndrome. - Histone Deacetylase Inhibition with Vorinostat as a Target in Oncology Advanced or refractory malignancy remains an area of high unmet medical need as patients often relapse and curative therapy is elusive. The mainstay of treatment is generally cytotoxic chemotherapy which can have limited efficacy and is often associated with significant toxicity; there is a need for novel agents that are not only effective but also well tolerated. In particular, there has been increasing interest in targeted therapies which work at an epigenetic level to influence gene expression and ultimately control tumor growth and proliferation. Histone deacetylase (HDAC) inhibitors represent one such class of new mechanism-based anticancer drugs [1]. Modifications to histones influence chromatin structure, and ultimately gene transcription, including those coding for tumor suppressor proteins. One of the key histone modifications that controls gene transcription is acetylation, which is regulated by two opposing enzymatic activities (histone acetyltransferases [HATs] and HDACs) [1]. Histone acetylation leads to an open chromatin structure, and allows access to transcription binding sites. Although histones are one of the targets of HATs and HDACs, many nonhistone proteins, including transcription factors, tubulin and heat shock protein 90, can also be regulated by acetylation [2,3]. HDACs have been shown to be overexpressed in human cancers, such as gastric, prostate and colon cancer, and are involved in the regulation of transcription with recruitment by oncogenic transcription factors [4]. Therefore, the inhibition of HDACs is a rational target for the development of novel anticancer therapy. To date, 18 HDACs have been identified in mammalian cells, which are categorized into different classes, based on their homology to yeast deacetylases [5]. By inhibiting these enzymes, HDAC inhibitors permit chromatin to assume a more relaxed conformational state, thereby allowing transcription of genes involved in tumor suppression, cell-cycle arrest, cell differentiation, and apoptosis (Figure 1[4]) [6]. A variety of HDAC inhibitors are in clinical development and are being assessed in a number of different cancer indications [7]. There are several chemical families among the HDAC inhibitors, including short-chain fatty acids (butyrate, valproic acid), hydroxamates (vorinostat, tri STAT5 genes p53 genes BCL6 genes Cell cycle arrest and apoptosis aThe sites of action of other antitumor agents are also shown PFrigouporese1d mechanism of acti (...truncated)


This is a preview of a remote PDF: http://www.jhoonline.org/content/pdf/1756-8722-2-31.pdf

David Siegel, Mohamad Hussein, Chandra Belani, Francisco Robert, Evanthia Galanis, Victoria M Richon, José Garcia-Vargas, Cesar Sanz-Rodriguez, Syed Rizvi. Vorinostat in solid and hematologic malignancies, Journal of Hematology & Oncology, 2009, pp. 31, 2, DOI: 10.1186/1756-8722-2-31