Cancer Letters

Cancer Letters

Volume 284, Issue 2, 1 November 2009, Pages 175-181
Cancer Letters

Fenretinide inhibits myeloma cell growth, osteoclastogenesis and osteoclast viability

https://doi.org/10.1016/j.canlet.2009.04.022Get rights and content

Abstract

Fenretinide (4HPR), a nontoxic analog of ATRA, has been investigated in various malignancies but not in multiple myeloma (MM), a plasma cell malignancy associated with induction of osteolytic bone disease. Here we show that 4HPR induces apoptosis through increased level of ROS and activation of caspase-8, 9 and 3, and inhibits growth of several MM cell lines in a dose-dependent manner. Serum or co-culture with the supportive osteoclasts partially protects MM cells from 4HPR-induced growth inhibition. Sphingosine-1 phosphate (S1P) significantly protects MM cells from 4HPR-induced apoptosis suggesting that as in other malignancies, this drug up-regulates ceramide in MM cells. 4HPR has no toxic effects on non-malignant cells such as blood mononucleated cells, mesenchymal stem cells and osteoblasts, but markedly reduces viability of endothelial cells and mature osteoclasts and inhibits differentiation of osteoclasts and MM-induced tube formation. 4HPR is a potential anti-MM agent, affecting MM cells and MM-induced bone disease and angiogenesis.

Introduction

Multiple myeloma (MM) is characterized by the clonal expansion of monoclonal immunoglobulin-secreting plasma cells within the bone marrow (BM). Myeloma cells are often dependent on the BM microenvironment for growth and their dissemination within the hematopoietic BM are typically associated with induction of severe osteolytic bone disease [1] and angiogenesis [2]. Treatment with high dose therapy and novel agents substantially improved clinical outcome but treatment with these agents often associated with severe adverse effects and development of drug resistance [3]. These clinical observations emphasize the need of additional potent but yet relatively safe, anti-myeloma agents.

Fenretinide (N-(4-hydroxyphenyl) retinamide, or 4HPR), a neoclassical analog of the retinoids all-trans retinoid acid (ATRA), has been successfully tested as a chemopreventive and chemotherapeutic agent on various malignancies, such as prostate, breast and colorectal cancer [4]. It is less toxic and teratogenic than other retinoids [5], thereby making it one of the most promising retinoid anti-tumor compounds. Compared with ATRA, 4HPR exhibits reduced hepatotoxicity and increased efficacy in inhibiting mammary carcinogenesis in animal models [6]. Furthermore, 4HPR lacks the ability to induce point mutations or chromosomal aberrations, and is therefore not genotoxic [7]. 4HPR is currently under clinical evaluation in different cancers but the biological effect and therapeutic value in MM have not been investigated.

The cellular and molecular mechanisms by which 4HPR elicit its cytotoxic action are still not clearly understood. Compared with retinoic acid, a key distinction is that 4HPR induces cell apoptosis rather than differentiation and shows synergistic responses with chemotherapeutic drugs. Induction of apoptosis by 4HPR is mediated through a receptor-independent mechanism and is accompanied by increased cellular levels of ceramide [8], [9] and ROS [10], and activation of caspases [11]. We reasoned that if ceramide is a major mediator of 4HPR-induced apoptosis, the effect of the drug could be counteracted by sphingosine-1-phosphate (S1P), which is another sphingolipid metabolite known to regulate cell growth and survival [12]. Whereas increased ceramide and sphingosine are associated with growth arrest and apoptosis, increased intracellular level of S1P is associated with suppression of apoptosis. Intracellularly, the balance between S1P and ceramide/sphingosine levels, which is regulated by a network of key enzymes, determines the fate of normal and malignant cells, and chemoresistance [12], [13].

In this study, we have tested the effect of 4HPR on growth and survival of MM cells in the absence and presence the protective osteoclasts [14]. We also tested the effect of 4HPR on angiogenesis and osteoclastogenesis, two physiological processes typically stimulated in myelomatous bones.

Section snippets

Reagents and supplies

An antibiotic mixture containing penicillin, streptomycin, and neomycin; α-minimum essential medium (α-MEM); and low-glucose Dulbecco’s modified Eagle’s medium (DMEM-LG) were obtained from Gibco (Grand Island, NY, USA). RPMI 1640 (without l-glutamine), Cellgro COMPLETE™ medium, were purchased from Mediatech, Inc. (Herndon, VA, USA). Fetal bovine serum (FBS) was purchased from Hyclone (Logan, UT, USA). Recombinant human macrophage colony-stimulating factor (M-CSF) and RANKL were purchased from

Effects of 4HPR on growth of peripheral blood mononucleated cells (PBMCs), MSC, HUVEC, osteoblasts and osteoclasts, and growth of MM cells in the absence and presence of serum and osteoclasts

We initially tested the effects of 4HPR on survival and growth of MM cell lines and normal PBMCs and MSCs in the presence of 10% serum. 4HPR uniformly inhibited growth of ARP-1, BN, CAG, and JB MM cells in a dose related manner (0.5–20 μM, 48 h) as determined by MTT assay (Fig. 1A). In contrast, similar concentrations of 4HPR had no significant toxicity on PBMCs or MSCs (Fig. 1A). 4HPR also effectively inhibited survival of mature osteoclasts and HUVEC but had minimal toxic effect on osteoblasts’

Discussion

In the current study we show that 4HPR inhibits survival and growth of several MM cell lines and that these effects are partially attenuated by co-culture with the supportive osteoclasts [14]. In contrast, 4HPR has minimal toxic effect on non-malignant cells (e.g. PBMCs, MSC, osteoblasts). 4HPR has been previously shown to inhibit angiogenesis [22] and differentiation of murine microphage--like cell line [23]. For the first time we showed the ability of 4HPR to suppress MM cell–induced tube

Conflict of interest

Authors have no conflict of interest to disclose.

Acknowledgments

This work was supported by grants from the National Cancer Institute (CA-93897) (S.Y.) and from the Multiple Myeloma Research Foundation (Senior and Translational Research Awards) (S.Y.).

References (30)

  • P.G. Richardson et al.

    New drugs for myeloma

    Oncologist

    (2007)
  • R. Lieberman et al.

    Development of new cancer chemoprevention agents: role of pharmacokinetic/pharmacodynamic and intermediate endpoint biomarker monitoring

    Clin. Chem.

    (1998)
  • F. Rehman et al.

    Fenretinide stimulates redox-sensitive ceramide production in breast cancer cells: potential role in drug-induced cytotoxicity

    Br. J. Cancer.

    (2004)
  • N. Oridate et al.

    Involvement of reactive oxygen species in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells

    J. Natl. Cancer Inst.

    (1997)
  • J.M. Wu et al.

    Mechanism of fenretinide (4-HPR)-induced cell death

    Apoptosis

    (2001)
  • Cited by (16)

    • Fenretinide targets the side population in myeloma cell line NCI-H929 and potentiates the efficacy of antimyeloma with bortezomib and dexamethasone regimen

      2016, Leukemia Research
      Citation Excerpt :

      Previous studies have demonstrated the important role of ROS in regulating CSCs [29]. In addition, fenretinide treatment has been shown to result in higher levels of ROS in MM cells, mediating cellular apoptosis [30]. In our current study, we demonstrated that MM MP cells produced nearly twice the level of ROS compared to MM SP cells.

    • Additive enhancement of apoptosis by TRAIL and fenretinide in metastatic breast cancer cells in vitro

      2014, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      The synthetic retinoid, fenretinide (N-(4-hydroxyphenyl) retinamide, 4-HPR) is one the most studied retinoids due to its low toxicity [3]. Because of this low toxicity profile fenretinide has been studied for its activity on distinct types of cancer cell lines including skin [4], breast [5], lung [6], ovarian [7] and pancreatic [8] cancer cell lines, multiple myeloma [9] and neuroblastoma [10] cell lines. Fenretinide is also widely used in chemoprevention studies in vitro and in vivo and it is an advantageous compound in breast cancer chemoprevention because of its selective uptake in the mammary gland [3].

    • Fenretinide inhibited de novo ceramide synthesis and proinflammatory cytokines induced by Aggregatibacter actinomycetemcomitans

      2013, Journal of Lipid Research
      Citation Excerpt :

      A.a mainly increased long-chain (C16) and very-long-chain (C22, C24, and C24:1) dhCer species (Fig. 2G–J) and ceramide species (Fig. 2K–N). In previous studies, fenretinide has been confirmed as an agonist for SPT (13–15) but also as a potent inhibitor for DEGS (16, 17). Because SPT and DEGS are critical in the de novo pathway for ceramide biosynthesis, we hypothesized that fenretinide would interfere with ceramide synthesis induced by A.a. To test our hypothesis, Raw 264.7 cells were treated with fenretinide (5 μM) or control vehicle ethanol for 4 h. Cells were unstimulated or stimulated with A.a (100 CFUs) for an additional 4 h. Various kinds of sphingolipid levels, including dhSph, dhCer, ceramide, Sph, S1P, and SM, were evaluated in cell extracts by mass spectrometry analysis.

    • Fenretinide prevents lipid-induced insulin resistance by blocking ceramide biosynthesis

      2012, Journal of Biological Chemistry
      Citation Excerpt :

      A quick review of the literature is likely to create confusion about the effect of fenretinide on ceramide metabolism. Early studies erroneously suggested that fenretinide induced a general elevation of ceramides (36–38). Indeed, this increase was thought to trigger the stress responses that halt the growth and promote death of transformed cells.

    View all citing articles on Scopus
    View full text