Japanese Dental Science Review (2010) 46, 173—187
a v a i l a b l e a t w w w. s c i e n c e d i r e c t . c o m
journal homepage: www.elsevier.com/locate/jdsr
Review article
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma Hiroshi Sakagami * Division of Pharmacology, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan Received 4 August 2009; received in revised form 2 November 2009; accepted 13 January 2010
KEYWORDS Oral squamous cell carcinoma; Apoptosis; Autophagy; Structure
Summary This review searched previous works of apoptosis induction in human oral squamous cell carcinoma (OSCC) cells. Many chemotherapeutic drugs, natural products, metabolic inhibitors, hormone receptor ligands and gene manipulations induced the apoptosis directly or indirectly by reverting the anti-apoptotic pathways. However, these antitumor agents, sometimes induced incomplete apoptosis or other types of cell death characterized by lack of caspase activation and internucleosomal DNA fragmentation, depending on the target cells or the chemical structure of inducers. There were very few investigations that have compared the cytotoxicity of these antitumor substances against OSCC cells with that of normal oral cells. Furthermore, their apoptosis-inducing activity has not been correlated well with tumor-specificity (higher cytotoxicity against tumor cells versus normal cells). These accumulated evidences provide the cautionary note against the apoptosis-oriented research. # 2010 Japanese Association for Dental Science. Published by Elsevier Ireland. All rights reserved.
Contents 1. 2.
3.
Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . Classification of three major types of cell death 2.1. Apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Autophagy . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Necrosis . . . . . . . . . . . . . . . . . . . . . . . . . . Apoptosis inducers . . . . . . . . . . . . . . . . . . . . . . . 3.1. Apoptosis inducers against OSCC (Table 1) 3.1.1. Chemotherapeutic drugs. . . . . . . . 3.1.2. Natural products . . . . . . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
. . . . . . . . .
* Tel.: +81 49 279 2758; fax: +81 49 285 5171. E-mail address:
[email protected]. 1882-7616/$ — see front matter # 2010 Japanese Association for Dental Science. Published by Elsevier Ireland. All rights reserved. doi:10.1016/j.jdsr.2010.01.004
. . . . . . . . .
174 174 174 174 175 175 175 175 175
174
4. 5.
6. 7.
8.
H. Sakagami 3.1.3. Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1.4. Hormone receptor ligands . . . . . . . . . . . . . . . . . 3.1.5. Gene manipulations . . . . . . . . . . . . . . . . . . . . . . 3.1.6. Others . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Head and neck squamous cell carcinoma (HNSCC) (Table 3.3. Salivary gland carcinoma . . . . . . . . . . . . . . . . . . . . . . . . Anti-apoptotic mechanism in OSCC (Table 3) . . . . . . . . . . . . . . Potentiators of apoptosis (Table 4). . . . . . . . . . . . . . . . . . . . . . 5.1. Chemotherapeutic drugs. . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.3. Others . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Induction of non-apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . Tumor-specificity (Table 5). . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.1. Flavonoid-related polyphenols . . . . . . . . . . . . . . . . . . . . 7.2. Coumarins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.3. Tannins-related compounds . . . . . . . . . . . . . . . . . . . . . . 7.4. Terpenoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.5. Ketones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.6. Synthetic compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.7. Antitumor antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . 7.8. Plant extracts. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1. Introduction Squamous cell carcinoma (SCC) is a form of cancer of the carcinoma type that may occur in many different organs, including the skin, lips, mouth, esophagus, urinary bladder, prostate, lungs, vagina, and cervix. It is a malignant tumor of squamous epithelium, and invasive cancers are able to spread to other organs and cause metastasis. Most cases of head and neck cancer are due to SCC, and caused by tobacco and alcohol, and human papilloma virus. Oral cancer is one of the most disfiguring types of cancer, since the surgical removal of the tumor may result in facial distortion. Oral squamous cell carcinoma (OSCC) is the fifth most common cancer worldwide, with the number of cases consistently increasing in developing countries. Despite focused efforts to improve therapy, 5-year survival rates for persons with advanced-stage OSCC remain discouragingly low. OSCC, like other types of cancer, is a genetic disease, resulting in the loss of differentiation, and possibly generated by the decline of apoptotic potential and immunity [1]. Aggressive OSCC with a high score of malignancy showed reduced expression of the tumor suppressor gene phosphatase and tensin (PTEN) homologue deleted on chromosome 10 [2], p53 positivity and low apoptotic index [3], and increased expression of anti-apoptotic proteins such as survivin [4] and Bcl-2 [5]. Down-regulation of heat shock protein 27 enhanced the transformation of oral epithelial dysplasia into OSCC, possibly by impairing the protective mechanism against mutagenesis induced by environmental factors [6]. Carcinogenesis of OSCC is related to the overexpression of prolyl isomerase Pin 1 [7], a stronghold for the therapy of Alzheimer’s disease. These data suggest that OSCC may be produced by an imbalance of the regulation between cell survival and apoptosis. Early detection combined with strategies for local intervention, such as chemoprevention prior to SCC development,
.. .. .. .. 2) .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
176 176 176 176 177 177 177 178 178 179 179 180 180 180 180 182 182 182 182 183 183 183 184 184
could dramatically improve clinical outcomes. Microarray analysis of small noncoding microRNAs (miRNAs) in the patient can be a useful prognostic markers [8]. Recently, the targeted elimination of OSCC cells by inducing apoptosis has emerged as a valued strategy to combat oral cancer. Studies utilizing a variety of chemical or biological interventions demonstrated promising results for induction of apoptosis in oral malignant cells [9]. There are at least three types of cell death: apoptosis, autophagy and necrosis [10,11]. The present review surveys recent studies of the type of cell death (either apoptosis or non-apoptosis) induced by various antitumor agents in OSCC and the relationship to their tumor-specificity.
2. Classification of three major types of cell death 2.1. Apoptosis Apoptosis is characterized by morphological changes such as cell shrinkage, pyknosis, densed cytoplasm, tightly packed organelles, chromatin condensation, and loss of cell surface microvilli [12]. Markers of apoptosis include DNA fragmentation (assessed by TUNEL method on fixed cell slide preparations, proportion of sub-G1 cells on FACS analysis, ladder pattern on agarose gel electrophoresis), decline of mitochondrial membrane potential, cytochrome c release from mitochondria, activation of caspase-3 using specific synthetic substrate or poly(ADP-ribose)polymerase (PARP).
2.2. Autophagy Autophagic cell death is characterized by the sequestration of cytoplasm and organelles in double or multimembrane vesicles and delivery to the cell’s own lysosomes for subse-
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma quent degradation [13]. The process of autophagy depends on both continuous protein synthesis and the continuous presence of ATP [14]. Markers of autophagy is formation of phagophore and autophagosome detected by electron microscopy, Atg8/LC3 western blotting and ubiquitin-like protein conjugation systems (increase in the amount of LC3-II, and Atg12—Atg5 conjugation) or fluorescent microscopy (increase in punctuate LC3 (or Atg18), TOR and Atg1 kinase activity) [11].
2.3. Necrosis Necrosis is an uncontrolled and passive process that usually affects large fields of cells. Necrotic cell injury is mediated by two main mechanisms, interference with the energy supply of the cell and direct damage to cell membranes [10]. Necrosis is characterized by cell swelling, formation of cytoplasmic vacuoles, distended endoplasmic reticulum, formation of cytoplasmic blebs, condensed swollen or ruptured mitochondria, disaggregation and detachment of ribosomes, disrupted organelle membranes, swollen and ruptured lysosomes and eventually disruption of the cell membrane [15].
3. Apoptosis inducers 3.1. Apoptosis inducers against OSCC (Table 1) 3.1.1. Chemotherapeutic drugs Chemotherapeutic drugs are the most frequently used apoptosis inducers of OSCC. 5-FU, peplomycin, cisplatin (CDDP) and g-ray, NO generator (ONOO, SIN-1 and SNP) induced apoptosis in OSCC cells (OSC-4) [16]. These treatments slightly increased the intracellular concentration of NO and O2 levels, and induced the nitration of cytochrome c and caspase-3. CDDP induced the rapid decline of surviving gene expression [17] and augmentation of apoptosome molecules (cytochrome c and Apaf-1) and activation of caspase-9 and capase-3, but no change in NF-kB nor the expression of anti-apoptotic proteins (TRAF-1, TRAF-2, c-FLIP). This suggests that cisplatin exerts its apoptotic action by the mitochondria-mediated activation of caspases but not by the activation of caspases due to the inhibition of NF-kB activity that follows the suppression of anti-apoptotic proteins [18]. Low-dose of CDDP enhanced the Fas/CD95) expression and increased susceptibility to apoptosis in oral cancer cell line (UT-SCC-20A) [19]. Carboplatin (CBDCA) induced apoptosis in human welldifferentiated tongue squamous carcinoma cell lines (SCC-9 and SCC-25). The carboplatin-induced apoptosis was obliterated by neutralizing anti-Fas (APO-1/CD95) and anti-Fas ligand (FasL). Anti-Fas antibody itself was inactive, but in the presence of carboplatin, markedly enhanced the apoptosis. There was large enhancement of FADD expression, but no alterations in Fas or FasL expression upon carboplatin treatment. Suppression of FADD expression using the specific antisense oligonucleotide resulted in a failure of carboplatin induction of cell death. These data suggest that carboplatin may switch nonfunctional Fas to functional Fas by up-regulation of FADD expression, resulting in activation of Fas-sensitive pathway leading to apoptosis [20]. Carboplatin induced apoptosis in OSCC (MIT7) probably through the cleavage of Bax-alpha and Bcl-x(L) [21].
175
Paclitaxel, an antimitotic chemotherapeutic agent, induced apoptosis in human OSCC cell line (NB-1) by downregulation of Bcl-2 and up-regulation of Bax [22]. Docetaxel induced Bax expression at both protein and mRNA levels in not all OSCC cell lines and clinical cases, but the enhanced expression of Bax has been suggested to be contributed to the antitumor effects of chemotherapy [23]. Peplomycin and bleomycin induced apoptosis in OSCC (SSKCN) [24]. Pingyangmycin (bleomycin A) induced growth arrest at G2-M phase of the cell cycle and apoptosis in OSCC (KB cells) [25]. 137 Cs irradiation, 5-fluorouracil (5-FU) or CDDP enhanced the apoptosis of OSCC induced by interleukin-2-activated lymphocytes [26]. Low-dose (0.5-Gy) of radiation caused enhanced apoptosis in human OSCC cells (T-167, T-409) without induction of transmembrane protein P-glycoprotein (MDR1), while higher dose (2-Gy) of radiation increased expression of MDR1 by NF-kB, NF-Y. Low-dose fractionated radiation can be used as an adjuvant for chemotherapy [27]. Heat stress induced 100-fold increase in the production of heat shock proteins, and prevented the NF-kB activation (nuclear translocation), may ultimately promote apoptosis in OSCC Ca9-22 cell line [28]. Hyperthermia induced apoptosis in OSCC cells (wild-type p53), accompanied by decrease of the gene expression of IL-12. On the other hand, IL-12Rb1 increased in the mutated p53 cells. p53 status is a useful candidate for a predictive indicator of the effectiveness in hyperthermic therapy [29]. Molecular inhibition of epidermal growth factor receptor (EGFR) signaling is a promising cancer treatment strategy. Treatment of OSCC cells with an anti-EGFR monoclonal antibody, Cetuximab (C225, Erbitux) and an EGFR tyrosine kinase inhibitor (TKI), AG1478, which target the extracellular and intracellular domains of the receptor, respectively, inhibited phosphorylation of EGFR and its downstream effector molecule Akt and amplified the induction of Fas-mediated apoptosis. The pro-apoptotic activity of EGFR inhibitors in OSCC cells depends on the extrinsic pathway of the caspase cascade accompanied by the down-regulation of cellular FLICEinhibitory protein (c-FLIP) [30]. 3.1.2. Natural products Essential oil of a perennial small herbal plant induced apoptosis via MAPK activation, based on the inhibition of MAPK kinase inhibitor [31]. Soft coral extract induced apoptosis in OSCC (SCC4, SCC9, SCC25) [32]. Ethanol extract of freezedried black raspberries suppressed the proliferation of OSCC cell lines without affecting viability, inhibited the production of vascular endothelial growth factor (VEGF), and induced both apoptosis and terminal differentiation. This extract may be a promising candidate for use as a chemopreventive agent in persons with oral epithelial dysplasia [33]. Theaflavin-3,30 -digallate, a polyphenol in black tea, induced apoptosis in HSC-2 cell by its prooxidant action (elevation of reactive oxygen species (ROS) production) whereas normal human gingival fibroblast (GN) was more resistant [34]. Green tea polyphenol induced apoptosis in OSCC cell line accompanied by the gradual decline of mitochondrial function. The cells deleted of caspase-3 gene did not undergo apoptosis. This suggests that green tea polyphenol-induced apoptosis is a mitochondria-targeted, caspase-3-executed mechanism [35]. Green tea polyphenols (catechins) induced
176 apoptosis in OSCC cells, and p58/KIP2 is a determinant prosurvival factor for cell protection from green tea polyphenolinduced apoptosis [36]. Curcumin alone or in combination with tea polyphenols inhibited the oral carcinogenesis in hamsters, possibly by suppressing the cell proliferation, induction of apoptosis and angiogenesis [37]. Stilbenes (resveratrol, piceatannol, rhaponticin), stilbene trimers (sophorastilbene A, ()-e-viniferin), a stilbene dimer ((+)-a-viniferin) and flavonoids (kaempferol, fisetin, quercetin, isoliquiritigenin, butein) induced activation of caspase-3, 8 and -9, and internucleosomal DNA fragmentation in human promyelocytic leukemia HL-60 and OSCC (HSC-2) cells [38]. Ovatodiolide, a diterpenoid from a Chinese herb, induced G2/M arrest and apoptosis in OSCC cell line Ca9-22 by Nacetyl-L-cysteine (NAC)-inhibitable ROS generation [39]. Shikonin, a naphthoquinone pigment isolated from the Chinese herbal therapeutic, Zicao, has been shown to exhibit antioxidant and anticancer effects. Shikonin induced apoptosis in human OSCC (Tca8113) cells via inactivation of NF-kB pathway [40]. Paradol, a pungent phenolic compound found in ginger and other Zingiberaceae plants, induced apoptosis in OSCC (KB) cells through a caspase-3-dependent mechanism [41]. 3.1.3. Inhibitors Cyclooxygenase-2 (COX-2), an enzyme that catalyzes the synthesis of prostaglandins (PGs), is made inducible by various stimuli such as inflammation. COX-2 is commonly overexpressed in a variety of premalignant and malignant conditions including oral leukoplakia and SCC. Overexpression of COX-2 plays an important role in the development and progression of different cancers. Immunohistochemical analysis demonstrated that expression of COX-2 and E2 promoterbinding factor-1 (E2F-1) proteins was increased in both oral dysplasia and SCC compared to the normal epithelium. A selective COX-2 inhibitor, NS398, suppressed PGE2 level, and expressions of COX-2, pRb and E2F-1 proteins, and induced G1 arrest and apoptosis [42]. Apoptosis-inducing activity of four selective COX-2 inhibitors (celecoxib, NS-398, nimesulide, meloxicam) was compared using OSCC (KB) cells. Celecoxib and NS-398 strongly suppressed the proliferation of KB cells at 10—100 mM, whereas nimesulide and meloxicam are less potent. All COX-2 inhibitors increased COX-2 protein expression but suppressed PGE2 production. Only celecoxib induced apoptosis. Celecoxib is a good therapeutic candidate for treating OSCC through the suppression of cell proliferation and the induction of apoptosis in a COX-2 independent manner [43]. Sulindac, a non-steroidal anti-inflammatory drug (NSAID), induced growth inhibition and apoptosis in OSCC (SCCa), and caused up-regulation of the protein and mRNA expression levels of COX-2 and PPARg. Treatment of antisense PPARg oligonuleotides abolished sulindac’s growth inhibitory effect, suggesting that up-regulation of PPARg expression and activation may be, at least partially, responsible for sulindac’s antiproliferative effect [44]. Protein kinase C (PKC) inhibitor safingol induced apoptosis in OSCC cells, accompanied by release of endonuclease G from mitochondria and translocated to nucleus [45]. Safingol induced the apoptosis in OSCC by mechanism involved with increase of Bmi and decrease of Bcl-xL and phosphorylated focal adhesion kinase (FAK) [46]. UCN-01 (7-hydroxystaurosporine), a PKC inhibitor, also induced apoptosis and G1 arrest
H. Sakagami in OSCC cell lines. HSC-3 (primary-type OSCC) were less sensitive than LMF4 cells (metastatic-type OSCC)[47]. Protein phosphatase inhibitors, okadaic acid (OA) and calyculin A (CA), induced apoptosis in three OSCC cell lines (SCC-25, SCCKN, SCCTF) [48]. Okadaic acid induced apoptosis in OSCC (SCC-25) cells by enhancing the Fas and FasL expression [49]. Flavopiridol, a synthetic flavone, inhibited the growth (by reducing the expressions of cyclin A, cyclin B and cyclin D1, and cyclin-dependent kinases (CDK) activation kinase and CDC25C) and induced apoptosis (by activating the Bcl-x pathway) in OSCC cells [50]. Proteasome inhibitors, carbobenzoxy-L-leucyl-L-leucyl-Lnorvalinal, or lactacystin, induced apoptosis in OSCC cells, but this was inhibited by antisense p27Kip1 oligonucleotide. The accumulation of p27Kip1 may play an important role in the apoptosis induced by proteasome inhibitor [51]. 3.1.4. Hormone receptor ligands 15-Deoxy-D12, D14-prostaglandin (PG) J2, a proliferator-activated receptor g (PPARg) ligand, induced growth inhibition and apoptosis in OSCC cells, whereas rosiglitazone and ciglitazone, thiazolidinedione family of PPARg activators, did not exert a growth inhibitory effect. 15-PGJ2, but not other PPARg activators, induced significant reduction of both phosphorylated and unphosphorylated Stat3 (signal transducer and activator of transcription). This suggests that 15-PG induces apoptosis in OSCC by PPARg-independent pathways [52]. Cyclopentenone 15-Deoxy-D12, D14 PGJ2 induced apoptosis in OSCC (B88) cell lines by down-regulation of the constitutive and IL-6-mediated JAK (Janus kinase) phosphorylation as well as Stat3 phosphorylation. The apoptosis-inducing activity of this compound required the a,b-unsaturated ketone structure within the cyclopentenone ring [52]. All-trans retinoic acid (ATRA) and 1a,25(OH)2vitamin D3 (calcitriol) suppressed the cell proliferation and induced apoptosis in OSCC (KB) cells, upregulated sensitivity of the chemotherapeutics drugs and down-regulated several angiogenesis factors and survivin [53]. Retinoic acid induced apoptosis in OSCC (Tca83) cells by enhancement of Fas gene transcription and translation, without change in FasL expression [54]. 3.1.5. Gene manipulations RNAi targeting urokinase-type plasminogen activator receptor (u-PAR) induced apoptosis as well as oral cancer invasion and metastasis [55]. Stable transfection of intracellular fragment of Notch induced G0—G1 cell cycle arrest and apoptosis in human tongue cancer cell line (Tca8113), accompanied by down-regulation of Wnt-b-catenin, increase of p21 and p53 expression, and decrease in Skp2 (S-phase kinase-associated protein 2) and Bcl-2 (B-cell lymphocytic-leukaemia protooncogene 2) expression [56]. 3.1.6. Others Nitric oxide (NO) donor, nitroprusside (SNP), induced keratinocyte differentiation markers at lower concentrations, while it induced apoptosis at higher concentrations in human immortalized keratinocytes (IHOK) and primary oral cancer cells (HK4) [57]. NO generator (ONOO, SIN-1 and SNP) induced apoptosis in OSCC (OSC-4) cells [16].
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma Selenium (Se) compounds induced apoptosis in OSCC (HSC3) cells, accompanied by the loss of mitochondrial membrane potential and glutathione (GSH), without a concurrent increase in ROS. The apoptosis-inducing activity of Se was diminished by GSH or compounds that elevated the intracellular GSH level, but augmented by buthionine sulfoximine that reduced GSH level. These data suggest possible role of GSH in the mitochondrial apoptosis of OSCC caused by Se [58]. Arsenic trioxide induced toxic damage and apoptosis characterized by caspase-3 activation, mitochondrial transmembrane potential collapse and G2/M arrest, but no change in p16, p53 and Bcl-2, in OSCC cell line. Tubulins and mitochondria may be the chief action position of arsenic trioxide [59]. Taurolidine, a derivative of the amino acid taurine, induced apoptosis in two OSCC cell lines (SCC4, SCC15), in contrast to the reference group treated with povidone iodine or the untreated control group [60].
177
reduced. Intraperitoneal administration of vitamin E succinate slowed the growth of JHU-022 solid tumor xenograft in immunodeficient mice [65]. An active component of OK-432, a streptococcal preparation, induced apoptosis in human HNSCC cell line by the activation of caspases through p53-independent pathway via TLR4 signaling [66]. The expression of E1A induced down-regulation of the expression of EGFR that was overexpressed in four HNSCC cell lines (Table 2). Overexpression of an exogenously introduced EGFR blocked E1A induction of apoptosis in these cells. These data suggest that E1A induces apoptosis by novel pathway involving EGFR [67]. Oral administration of vesnarinone, a cardiotonic, to a patient with histopathological recurrent oral cancer with well-differentiated squamous cell carcinoma, resulted in complete remission of the tumour [68].
3.3. Salivary gland carcinoma 3.2. Head and neck squamous cell carcinoma (HNSCC) (Table 2) Cetuximab is a chimeric mouse/human monoclonal antibody used to treat metastatic colorectal cancer and head and neck cancer. The antibody binds to the EGFR, a signalling protein that normally controls cell division. In some cancers, this receptor is altered to cause uncontrolled cell division. Cetuximab blocks EGFR and stops the uncontrolled cell division. Cetuximab induced apoptosis in HNSSC by inhibiting the MAPK and Janus kinase (JAK)/STAT-3 pathways [61]. Combination treatment with 13-retinoic acid, interferona2a and a-tocopherol apparently further inhibited the growth of five squamous cell carcinoma of the head and neck (SCCHN) in comparison to any single agent and two-drug combinations. Three-drug combination induced significant accumulation of the cells at S-phase and induced apoptosis. These data supported the promising outcomes of the phase III trial with a combination of these three drugs in patients with advanced head and neck cancer in the adjuvant settings [62]. Galanin is a neuropeptide present in humans and other mammals. It is a peptide consisting of a chain of 29 amino acids (or 30 amino acids in humans).Galanin is formed by the cleavage of a prepropeptide encoded by a gene known as GAL. Galanin induced apoptosis in galanin receptor-transfected HNSCC cell line with mutant p53 [63]. Alterations in histone acetylation status have been implicated in carcinogenesis. Histone deacetylase inhibitors, such as suberoylanilide hydroxamic acid, can potentially reactivate aberrantly silenced genes by restoring histone acetylation and allowing gene transcription. Suberoylanilide hydroxamic acid induced both mitochondrial pathway of apoptosis (cytochrome c release, caspase-3, caspase-9 activation) and extrinsic apoptosis pathway (increased Fas and Fas ligand (FasL) expression, activation of caspase-8, cleavage of Bid), having comparatively little activity against precancerous and normal oral cells [64]. Vitamin E succinate (a-TOS) induced apoptosis in five HNSCC cell lines (JHU-011, JHU-013, JHU-019, JHU-022, JHU-029) accompanied by elevation of ceramide, sphingomyelinase activity and p53. On the other hand, the amounts of nuclear factor kB, Bcl-2, and Bcl-X(L) proteins were
Slindac, a non-steroidal anti-inflammatory drug (NSAID), induced a significant decrease in cell proliferation and an increase in apoptosis in two salivary gland adenocarcinoma cell lines (HSY, HSG) [69].
4. Anti-apoptotic mechanism in OSCC (Table 3) OSCC (Ho-1-N-1 SP) cells survived even under treatment with various agents (5-FU, carboplatin), possibly due to high level expression of ABC transporters (ABCB1, ABCG2) and antiapoptotic protein (CFLAR, BCL2, BCL2A1) [70]. Hyperthermia-resistant human squamous cell carcinoma (SAS) cell: higher expression of Bcl-2, Bcl-xL, NF-kB, COX-2, STAT3, IL-6, IKKa/1) [71]. The imbalance between expression of anti-apoptotic and pro-apoptotic Bcl-2 family genes may promote survival in the oral cell lines [72]. The frequency of bcl-2 expression was associated with tumor histologic grade and decrease in apoptotic index in tongue SCC [73]. ()-Epigallocatechin-3-gallate (EGCG), a major component of green tea, induced p57, a cyclin-dependent kinase and apoptosis inhibitor, in a dose- and time-dependent manner in normal keratinocytes, but not in OSCC (SCC25, OSC2) cells. The chemopreventive effect of green tea polyphenols may involve p57-mediated cell cycle regulation in normal epithelial cell [74]. Paired box gene 9 (Pax9) and c-myb are transcription factors that regulate the expression of the genes involved in mediating cell proliferation, resistance to apoptosis, and migration. Pax9 and c-myb expressions in KB cells seem to be essential for cell growth, and survival was enhanced by c-myb. Disruption of the function of c-myb and Pax9 induced G0 arrest [75]. The mouse double minute 2 (MDM2) plays a pivotal role in radiotherapy by down regulating p53. A functional T-to-G polymorphism at nucleotide 309 in MDM promoter intron 1 (SNP309) influences transcription activity. A G-to-C SNP at p53 codon 72 results in an Arg/Pro polymorphism, which is associated with apoptosis induction potential and p53 mutation status. The MDM2 SNP309 G/G polymorphism was associated with poor overall survival in advanced OSCC and the overall survival and disease-free survival of irradiated patients. The
178 Table 1
H. Sakagami Substances that induce apoptosis in OSCC cells.
Inducers Antitumor agents 5-FU Cisplatin (CDDP) Carboplatin (CBDCA) Paclitaxel Docetaxel Peplomycin, bleomycin Radiation, g-ray, Cs Heat shock, hyperthermia Anti-EGFR mAb, EGFR TKI Natural products Essential oil Soft coral extracts Black raspberry extract Theaflavin-3,30 -digallate Green tea polyphenols Curcumin Stilbenes, flavonoids Diterpenoid Shikonin, a naphthoquinone pigment Paradol Inhibitors Selective COX-2 inhibitor, NS398, celecoxib PKC inhibitor safingol, 7-hydroxystaurosporine Phosphatase inhibitors: Okadaic acid calyculin A CDK inhibitor, flavopiridol Proteasome inhibitors Ligands to hormone receptors Cyclopentenone 15-deoxy-D12, D14- PGJ2 All-trans retinoic acid 1a,25(OH)2vitamin D3
Possible mechanism
Survivin #, FADD ", no change in NF-kB CDKIs " Cleavage of Bax-alpha and Bcl-x(L) BAX " G2-M block IL-12 # c-FLIP #
p57/KIP2 #
NF-kB #
Fas receptor ", FasL " CDKs # p21Kip1 accumulation JAK signalling #, Stat3 # Survivin #, VEGF #
Gene manipulation siRNA targeting u-PAR Notch overexpression Others NO generator, ONOO, SIN-1 and SNP Selenium compounds Arsenic trioxide Taurolidine
Reference [16,26,97] [16,17,18,26,80] [20,21] [22] [23] [16,24,25] [16,26,27] [28,29] [30] [31] [32] [33] [34] [35,36,74] [37] [38] [39] [40] [41] [42,43,44] [45,46,47] [48,49] [50] [51] [52] [53] [53] [55] [56] [16,57] [58] [59] [60]
GSH #
combination of MDM2 SNP309 G/G and p53 codon 72 Arg/Arg polymorphism is associated with the worst overall survival and disease-free survival. Both MDM2 SNP309 and p53 codon 72 SNP could be useful factors for evaluating the outcome of advanced OSCC treated with adjuvant radiation [76]. p58/KIP2 is a determinant pro-survival factor for cell protection from green tea polyphenol-induced apoptosis [36]. The nu/nu mice inoculated with Mn-SOD antisense-transfected SCC cells showed prolonged survival time, as compared with mice inoculated with control vector-transfected SCC cells after treatment with antitumor agents such as 5-FU, peplomycin, CDDP or g-ray. SCC cells transfected with MnSOD antisense group showed higher incidence of apoptosis than that transfected with empty vector. Mn-SOD may act as a negative regulator of apoptosis [77]. OSCC cell lines with lower Mn-SOD activity were easily committed to apoptosis
upon treatment with 5-FU, peplomycin or g-rays, than those with higher Mn-SOD. This suggests that Mn-SOD negatively regulates the apoptosis by these inducers [78].
5. Potentiators of apoptosis (Table 4) Apoptosis induction may be potentiated by reversing the anti-apoptotic mechanism.
5.1. Chemotherapeutic drugs CDDP administered for 24 h before 5-FU treatment for 48 h induced apoptosis in human oral cancer cells (B88) more efficiently than simultaneous administration of CDDP and 5FU or the sequential treatment of 5-FU followed by CDDP [79].
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma Table 2
Apoptosis induction in HNSCC cells.
Inducers
Possible mechanism
Cetuximb Retinoic acid + IFN-a + vitamin E Gelanin HDA inhibitor, suberoylanilide hydroxamic acid Vitamin E succinate OK-432, a streptococcal preparation Expression of E1A Vesnarinone, cardiotonic (clinical trial)
Table 3
Reference
Ceramide ", p53 ", NF-kB #, Bcl-2 #, Bcl-xl # p53-independent pathway via TLR4 signaling EGFR #
Anti-apoptotic mechanism in OSCC cells.
Anti-apoptotic mechanism
Reference
ABC transporters: ABCB1, ABCG2 Bim ", Bcl-xL #, phosphorylated FAK # Bcl-2, Bcl-xL, NF-kB, COX-2, STAT3, IL-6, IKKa/1 Pax9, c-myb SNP 309 and p53 codon 72 polymorphisms Cyclin-dependent kinase inhibitor p57/KIP2 Mn-SOD
[70] [46] [71,73] [75] [76] [36,74] [77,78]
Tamoxifen alone induced a transient G1 arrest by upregulation of cyclin-dependent kinase inhibitors (CDKIs) such as p21/Waf-1, p27/Kip1 and p15/INK4a in HNSCC cell lines (HN5, HN6), and sensitized the cells to apoptosis induced by CDDP. Tamoxifen stimulated the secretion of TGF-b1, and anti-TGF-b1 blocking antibody prevented both the blockade of cellular proliferation and increased expression of CDKIs. This suggests the role of TGF-b1 for the tamoxifen stimulation of cisplatin-induced apoptosis [80]. Hyperthermia is used as one of the treatment modalities for various types of cancers, but the acquisition of thermotolerance in cancer, through the induction of heat shock proteins (Hsps), renders hyperthermia less effective. PreTable 4
179
[61] [62] [63] [64] [65] [66] [67] [68]
treatment of HSC-2 cells with IFN-g suppressed Hsp27 transcription and promoter activity, and enhanced the induction of cell death by hyperthermia and cisplatin treatment [81].
5.2. Inhibitors Etodolac, a selective COX-2 inhibitor, enhanced the carboplatin (CBDCA)-induced apoptosis of OSCC (SCC-25) cell line through the suppression of FAP-1 (anti-apoptotic tyrosine phosphatase) expression, although etodolac alone did not induce the apoptosis [82]. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid, enhanced CDDP-induced apoptosis in the OSCC cell line (HSC-3), by rapidly up-regulating the endoplasmic reticulum (ER) stress-associated events such as the sustained phosphorylation of eukaryotic transition initiation factor-2 (eIF2), activation of protein phosphatase 1 and Akt dephosphorylation [83]. Co-administration of low-dose cisplatin (4 mg/ml) and suberoylanilide hydroxamic acid (2 mM) synergistically induced cytotoxicity and apoptosis in OSCC (Tca8113, KB) cell lines [84].
5.3. Others Resveratrol (trans-resveratrol) is a phytoalexin produced naturally by several plants when under attack by pathogens
Potentiators of apoptosis induction.
Potentiator Antitumor agents Cisplatin Cs, 5-FU, CDDP Tamoxifen IFN-g Inhibitors Selective COX-2 inhibitor, etodolac HDA inhibitor, suberoylanilide hydroxamic acid Others Resveratrol Glycerol Infection of wild-type p53-encoding adenovirus
Inducer
Possible mechanism
Reference
Agonistic Fas antibody (CH11), 5-FU LAK Cisplatin Hyperthermia, cisplatin, ATLA
Soluble Fas", Bcl-2#
[19,79]
CDKIs", TGF-b1 secretion " Hsp27 transcription and promoter activity #
[26] [80] [81,88]
Carboplatin (CBDCA) CDDP
FAP-1 # Protein phosphatase ", Akt dephosphorylation
[82] [83,84]
Vincristin, adriamycin, paclitaxel X-irradiation X-irradiation
Bcl-2 #, MDR1 #
[85] [86] [87]
180 such as bacteria or fungi, and found in the skin of red grapes and is a constituent of red wine. Resveratrol down-regulated the expression of Bcl-2 and MDR1, and enhanced the sensitivity of human epidermoid carcinoma KBv200 cells [85]. Pretreatment with glycerol enhanced the effectiveness of X-irradiation in Ca9-22 cells bearing a mutant p53. Glycerol restored the DNA-binding activity of mutant p53 for a p53consensus sequence to levels similar to that of wild-type p53 [86]. Infection with wild-type p53-encoding adenovirus alone, or X-irradiation alone, significantly inhibited the growth of OSCC (HSC-4 and SAS) cells, but combined treatment was most effective, even in mutant p53-accumulated HSC-4 cells [87]. ATLA and interferon-g synergistically stimulated the apoptosis in OSCC cell lines [88].
6. Induction of non-apoptosis As compared with the studies of apoptosis induction in OSCC, those of non-apoptosis induction are limited. Morin (3,5,7,20 ,40 -pentahydroxyflavone) induced G2/M arrest, without induction of apoptosis in human OSCC cells, via inhibition of AKT activation [89]. Intratumoral laser illumination of OSCC transplated nude mice induced necrosis, but not apoptosis, in OSCC cells [90]. We have recently reported that a,b-unsaturated ketones such as 1-trichloroacetyl-3-bromo-2-methoxyazulene and 1trichloroacetyl-3-chloro-2-ethoxyazulene induced autophagic cell death characterized by the vacuolization detected by transmission electron microscopy, and the granular distribution of acridine orange and translocation of LC3-GFP into the autophagosome of OSCC (HSC-4) cells. Although HL-60 cells are easily committed to apoptosis by many inducers, this cell line did not express apoptosis markers, such as internucleosomal DNA fragmentation and caspase activation, upon treatment with 4,4-dimethyl-2-cyclopenten-1-one, a-methyleneg-butyrolactone, 5,6-dihydro-2H-pyran-2-one, 3,3,3-trifluoro-2-hydroxy-1-phenyl-1-propanone, codeinone (an oxidative product of codeine) or morphinone (an oxidative metabolite of morphine) [91]. We found that 3,3,3-trifluoro-2-hydroxy-1-phenyl-1-propanone induced autophagic cell death (characterized by acridine and LC3-GFP accumulation in the autophagosome) in human OSCC cell lines (HSC-2, HSC-4) [92]. Furthermore, morphinone induced the formation of autophagosome engulfing organelles in HL-60 cells. The addition of N-acetyl-L-cysteine (NAC) reduced the cytotoxic activity of codeinone by 30-folds, whereas other antioxidants (cysteine, ascorbate, catalase) and metals (FeCl3, CoCl2, CuCl2) were almost inactive. Similarly, the cytotoxic activity of 4,4-dimethyl-2-cyclopenten-1-one, a-methyleneg-butyrolactone, 5,6-dihydro-2H-pyran-2-one was almost completely eliminated by NAC. This suggests that the cytotoxicity of a,b-unsaturated ketones is generated by the interaction between the b-position of the a,b-unsaturated carbonyl moiety and SH group of any targeted molecules (the so-called ‘‘non-sterically hindered Michael acceptor’’). We also found that morphinone induced a significant reduction of mitochondrial size. It is not clear at present whether this resulted from the perturbation of mitochondrial morphogenesis or the fragmentation of mitochondria as recently reported in Bax/Bak double knockout mouse embryonic
H. Sakagami fibroblast [93]. It has been recently reported that autophagy selectively degrades mitochondria, blocking the vicious cycle between defective mitochondria and ROS. These data suggest the mitochondrial target of autophagy.
7. Tumor-specificity (Table 5) There are very few reports available that have dealt with the tumor-specificity of natural and synthetic compounds. Theaflavin-3,30 -digallate showed a concentration and time-dependent inhibition, with the OSCC cells more sensitive than the normal GN46 fibroblasts [34]. Morin showed higher cytotoxicity against OSCC than normal oral mucosa cells (NOMC) (TS = 1.6) whereas quercetin, kaempferol, datiscetin and galangin showed little or no tumor-specificity (TS = 1.03, 1.06, 1.00 and 1.08, respectively) [89]. Chemopreventive effects of green tea polyphenols may involve p57-mediated cell cycle regulation in normal epithelial cells [74]. Histone deacetylase inhibits suberoylanilide hydroxamic acid induced growth inhibition and apoptosis induction in HNSCC cell lines, but had limited effects on premalignant and normal cells [64]. Based on these backgrounds, we have surveyed a total of 1000 compounds for their tumor-specificity. The tumor-specific cytotoxicity index (TS) was determined by the ratio of the mean 50% cytotoxic concentration (CC50) against normal human oral cells (gingival fibroblast (HGF), pulp cell (HPC), periodontal ligament fibroblast (HPLF)) to that against human OSCC cell lines (HSC-2, HSC-3, HSC-4, NA, Ca9-22), submandibular gland carcinoma (HSG) [38,94,95] (Table 5). Preparations of normal cells from the extracted teeth and periodontal tissues have been approved by the intramural ethic committee after obtaining the informed consent from the donors. The compounds analyzed were mostly isolated and identified by our groups.
7.1. Flavonoid-related polyphenols Flavonoids, generally, showed weak tumor-specificity (TS = 1.2—4.0). Licochalcone B, a chalcone derivative without an isoprenoid group, showed the highest TS value of 31.7. Isoprenoid-substituted chalcone, prenylated isoflavone and genistein had higher cytotoxicity, but without high tumorspecificity, suggesting that the prenylation itself is not necessary for the tumor-specificity. Among the benzophenones, compounds with two isoprenoid groups had higher cytotoxicity than the monoprenylated compound, but with minor TS values. Anthraquinones exhibited relatively higher TS values. Among them, emodin and aloe-emodin, without glycosylation, were the most potent (TS = 8.5 and >18.6, respectively). Stilbenes (resveratrol, piceatannol, rhaponticin), stilbene trimers (sophorastilbene A, ()-e-viniferin), a stilbene dimer ((+)-a-viniferin) (TS = 1.4—3.6) and flavonoids (kaempferol, fisetin, quercetin, isoliquiritigenin, butein) (TS = 1.4—4.7) gave lower TS values.
7.2. Coumarins Coumarin itself and its 7-hydroxy-, 6-methoxy-7-hydroxy and 5,6-dimethoxy-derivatives were relatively non-toxic to all cell lines. Its 6,7-dihydroxy derivatives (esculetin) revealed a tumor cell line-specific cytotoxicity (TS > 5.1). Higher tumor-specificity of 4-methyl (TS > 8.3), 3,4-dimethyl
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma Table 5
181
Tumor-specificity of various groups of compounds against OSCC cells.
Compounds
TS (range) (no. of compounds)
Flavonoids Flavones, flavonols, isoprenylated flavonoids Flavonoids Isoprenylated flavonoids 2-Arylbenzofurans Benzophenones Xanthones Anthraquinones Phenylbutanone glucoside Stilbene glucoside Naphthalene glucosides Stilbenes
1.2 0.6 (0.3—3.2) (n = 36) 3.3 4.0 (0.8—31.7) (n = 27) 2.1 0.4 (1.6—3.0) (n = 22) 1.2 0.2 (1.0—1.5) (n = 6) 1.7 0.4 (1.2—2.3) (n = 5) 1.3 0.4 (n = 9) 3.8 4.9 (1.0—18.6) (n = 13) 2.4 (1.5—3.3) (n = 2) 1.8 0.8 (1.0—2.6) (n = 3) 1.3 (1.1—1.4) (n = 2) 3.0 1.2 (1.4—4.7) (n = 6)
Coumarins
2.4 3.0 (1.0—11.0) (n = 23)
Tannin-related compounds Procyanidins Flavonoids Monohydrolyzable tannins Oligomeric hydrolysable tannins Macrocyclic ellagitannins
4.8 2.3 1.1 0.1 1.5 0.5 1.4 0.2 4.4 2.7
(1.0—7.4) (1.0—1.2) (1.0—2.5) (1.2—1.5) (2.3—8.2)
Terpens Triterpenes Triterpene glycosides Triterpenes, triterpene glycosides, chromones Cycloartane glycosides Furostaol glycosides
1.5 0.7 1.4 0.5 1.0 0.1 1.1 0.2 2.5 4.1
(0.7—2.8) (n = 8) (1.0—2.4) (n = 21) (0.8—1.3) (n = 20) (0.9—1.4) (n = 7) (0.4—17.0) (n = 17)
Ketones a,b-Unsaturated ketones Cyclice a,b-unsaturated ketones a-Hydroxyketones b-Diketones Trifluoromethylketones Azulenequinones
1.2 0.3 (0.6—1.9) (n = 26) >229.0 (n = 1) 5.7 6.0 (1.0—17.6) (n = 8) 1.8 1.4 (0.3—6.3) (n = 22) 2.6 1.6 (n = 6) 2.6 2.3 (1.0—10.2) (n = 27)
Bacterial products Anthracyclines Mitomycin C Bleomycin, peplomycin Nocobactines
>167 89 (n = 4) >29 (n = 1) >3.8 0.2 (n = 2) 62.0 (43.9—80.0) (n = 2)
Others Vitamin K2 derivatives Prenylalcohols Azulenes Trihaloacetylazulenes Tropolones Berberines 3,5-Dibenzoyl-1,4-dihydropyridines Styrylchromones Isoxazole derivatives
1.9 0.2 (1.7-2.0) (n = 3) 1.3 0.3 (1.0-1.8) (n = 5) 1.7 1.0 (0.8—5.7) (n = 27) 6.5 10.7 (n = 26) 2.6 1.8 (1.0—9.9) (n = 27) 3.8 (3.6—4.0) (n = 2) >43.0 (>33 to >53) (n = 2) 7.3 6.1 (1.1—17.4) (n = 6) 1.2 0.2 (0.9—1.6) (n = 24)
Antioxidants Epigallocatechin gallate (EGCG) Catechin Quercetin Isoliquiritigenin Ascorbic acid (vitamin C) Gallic acid Chlorogenic acid
4.1 (n = 1) 1.0 (n = 1) 3.3 (n = 1) 4 (n = 1) 2.5 (n = 1) 1.1 (n = 1) 1.7 (n = 1)
(n = 6) (n = 4) (n = 7) (n = 3) (n = 4)
182
H. Sakagami
Table 5 (Continued )
Compounds Curcumin Morin Quercetin Kaemferol Datiscetin Galangin
TS (range) (no. of compounds) 1.7 (n = 1) 1.64 (n = 1) 1.06 (n = 1) 1.06 (n = 1) 1.00 (n = 1) 1.08 (n = 1)
Data derived from [38,94,95].
(TS > 9.3) and 3,4-cycloalkyl derivatives (TS > 11.0) than 6,7-dihydroxycoumarin may be due to their higher lipophilicity.
7.3. Tannins-related compounds Procyanidin B-2 (MW 578) (TS = 5.8), procyanidin C-1 (MW 866) (TS = >6.7) and procyanidin oligomers (MW 3170) (TS > 7.4) had higher cytotoxicity and tumor-specific cytotoxicity against OSCC cells than did catechin (MW 290) (TS = 1.0), ()-epicatechin (MW 290) (TS = 4.0) and ()-epigallocatechin gallate (EGCG) (MW 458) (TS = 4.1). Among hydrolyzable tannins (which contain glucose in the core of the molecule), three oligomeric hydrolyzable tannins (MW 1854—1873) had an approximately two-fold higher cytotoxicity than seven monomeric hydrolyzable tannins. Macrocyclic hydrolyzable tannins (oenothein B, woodfordin C, camelliin B, woodfordin D) (MW 1568—2506) exhibited one order higher cytotoxicity than monomeric hydrolyzable tannins. The tumor-specific cytotoxicity of macrocylic hydrolyzable tannins (TS = 2.7—8.2) was two- or three-fold higher than monomeric (TS = 1.0—2.5) or oligomeric hydrolyzable (TS = 1.2—1.5) tannins. On the other hand, gallic acid (MW 170) (TS = 1.1), methylgallate (MW 184) (TS = 1.3), ellagic acid (MW 302) (TS = 1.0) and chlorogenic acid (MW 354) (TS = 1.7) had much lower tumor-specific cytotoxicity. These data suggest that a more condensed structure, with (hydrolyzable tannins) or without glucose in the molecule (condensed tannins such as procyanidin oligomers), increases the tumor-specificity.
7.4. Terpenoids Generally, terpenoids had low tumor-specificity (TS = 0.5— 2.4), except for oleanolic acid (TS > 2.8) and 22a-methoxyfurostanol monodesmosides (TS = 7.4, >17,4).
7.5. Ketones The cytotoxic activity of a,b-unsaturated ketones declined with the introduction of methyl group at C-3 (b) or the addition of N-acetyl-L-cysteine (NAC), suggesting that their cytotoxicity is generated by the interaction between the C-3 and SH group of targeted molecules. Codeinone and morphinone (oxidative metabolites of codeine or morphine, respectively) containing a a,b-unsaturated ketone backbone showed lower tumor-specificity (TS = 3.8 and 3.1, respectively). Cyclic a,b-unsaturated ketones, such as 3-arylidene1-(4-nitrophenylmethylene)-3,4-dihydro-1H-naphthalen-2ones exhibited unusually high tumor-specificity (TS > 229).
Among 8 hydroxyketones, deferiprone (TS > 17.6), mimosine (TS > 7.9), tropolone (TS > 4.1) and hinokitiol (TS = 10.7) had the highest tumor-specific cytotoxicity. On the other hand, maltol (TS = 1.0), kojic acid (TS = 1.4), 3methyl-1,2-cyclopentanedione (TS = 1.3), 1,2-cyclohexanedione (TS = 1.2) had lower tumor-specificity. Among 23 b-diketones, 3-formylchromone was the most tumor-specific (TS = 6.3), followed by (+)- and ()-3-(trifluoroacetyl)camphor (TS = 4.4), 4,4,4-trifluoro-1-phenyl1,3-butanedione (TS = 3.4) and ()-3-(trifluoroacetyl)camphor (TS = 3.3); others including curcumin were much less active (TS = 0.9—2.2). Among 27 azulenequinone derivatives, 3-phenoxy-1,5azulenequinone (TS > 8.5) and 7-isopropyl-3-(4-methylanilino)-2-methyl-1,5-azulenequinone (TS = 10.2) had highest tumor-specificity and apoptosis-inducing activity (caspase activation). Among 27 tropolone derivatives, 5-aminotropolone was the most tumor-specific (TS = 9.9) and had the highest apoptosis-inducing activity.
7.6. Synthetic compounds Among 27 azulene derivatives, 2-acetylaminoazulene (TS > 3.6), diethyl 2-chloroazulene-1,3-dicarboxylate (TS > 5.7) and methyl 7-isopropyl-2-methoxyazulene-1-carboxylate (TS = 2.4) had the highest tumor-specificity. Chlorination of azulene resulted in the elevation of both cytotoxicity and tumor-specificity, while fluorination of the same compound was not so effective. There was no apparent difference between the cytotoxic activity of 2-methoxyazulenes and 2-ethoxyazulenes. Trichloroacetylazulenes generally gave higher cytotoxicity and tumor-specificity as compared with the corresponding trifluoroacetylazulenes. Substitution of chlorine, bromine or iodine at the C-3 position further enhanced their cytotoxicity to four tumor cell lines. Among 20 trihaloacetylazulene derivatives, 1-trichloroacetyl-3-bromo-2-methoxyazulene and 1-trichloroacetyl-3-chloro-2-ethoxyazulene had the highest tumor-specificity (TS = >3.5 and >2.5, respectively). Berberines exhibited some tumor-specificity (TS = 3.6— 4.0). Two 3,5-dibenzoyl-1,4-dihydropyridines had higher tumor-specificity, but only weakly induced apoptosis markers (DNA fragmentation, caspase activation). All six styrylchromones had higher cytotoxic activity against tumor cell lines than against normal cells. Styrylchromones, with one to three methoxy groups, had higher tumor-specificity and water solubility (TS = 5—17) and induced DNA fragmentation and caspase-3, -8 and -9 activation. Twenty-four 3-acetyland 3-benzoylisoxazole derivatives exhibited much lower tumor-specificity (TS = 0.9—1.6).
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma Flavopiridol, a synthetic flavone, induced apoptosis (subG1 DNA content, DNA fragmentation, PARP cleavage) via activation of Bcl-x in OSCC cell lines [50].
7.7. Antitumor antibiotics Doxorubicin, an anthracycline antibiotic isolated from Streptomyces peucetius var. caesius has been used for the treatment of cancer of the bladder, breast (in combination with other anticancer agents) and prostate, but is, however, suspected to be a human carcinogen. Doxorubicin, used as a positive control in our screening system, exhibited the highest tumor-specific cytotoxic activity (TS = 255.0). It activated caspase-3, -8 and -9 in both OSCC (HSC-2) and promyelocytic leukemia HL-60 cells, but only induced internucleosomal DNA fragmentation in HL-60 cells. Western blot analysis showed that doxorubicin did not significantly change the intracellular concentration of Bcl-2, Bax or Bad in HL-60 cells. Real time PCR analysis showed that HPC cells (normal) expressed the highest amount of mdr1 mRNA, followed by HSC-2 (tumor) > HGF (normal) > HSC-3 (tumor) > HPLF (normal) > HSG (tumor) > HL-60 (tumor). These data suggest that mdr1 expression in the tumor cells seems to be unrelated to the tumor-specificity of doxorubicin. Other anthracyclines such as mitoxantrone (TS > 259) and daunorubicin (TS > 164) produced comparable tumor-specificity with doxorubicin. Idarubicin exhibited slightly lower cytotoxicity and tumorspecificity (TS = 47). On the other hand, the tumor-specificity of other groups of antitumor antibiotics such as mitomycin C (TS > 29), bleomycin (TS > 3.7) and peplomycin (TS > 4.0) was much lower. This confirms the antitumor potential of anthracylines for the treatment of OSCC. Nocobactins NA-a (NBNAa) and NA-b (NBNAb) are mycobactin-like siderophores, which may play a role in the uptake of iron from the proteins of the host by chelation of ferric ion (Fe3+). These compounds exhibited high tumor-specificity index (TS = 80.0 and 43.9, respectively).
7.8. Plant extracts Poly-herbal extracts of Himalaya (HD-12, DLH-3073) exhibited highly tumor-specific cytotoxicity to tumor cell lines (TS = >1070 and >106, respectively) [96]. These extracts produced radicals under alkaline condition and scavenged O2. The tumor-specificity and antioxidant properties suggest their medicinal efficacy. The identification of the active principle is essential.
8. Conclusion The present review article demonstrated that OSCC cells can be committed to either apoptosis or non-apoptosis, depending on the type or sensitivity of target cells. OSCC showed different susceptibility of apoptosis, upon treatment with 5FU [97]. There was a considerable variation (approximately 10—20-fold) in drug-sensitivity of five OSCC cell lines. The sensitivity to mitomycin C is in the following order (from sensitive to resistant): HSC-2 (CC50 = 3.5 mM) > HSC-3 (9.7 mM) > Ca9-22 (16.4 mM) > HSC-4 (18.0 mM) > NA (37.8 mM). The sensitivity to bleomycin is in the following order: HSC-2 (CC50 = 4.6 mM) > HSC-3 (6.3 mM) > HSC-4
183
(77.4 mM) > NA (91.6 mM) > Ca9-22 (111.6 mM). The sensitivity to peplomycin is in the following order: HSC-2 (CC50 = 9.9 mM) > HSC-3 (25.2 mM) > NA (143.2 mM) > HSC4 (175.8 mM) > Ca9-22 (216.9 mM) [94]. In general, OSCC cells are relatively resistant to Fas-mediated apoptosis; this may be due to a lower expression of FAS [5] or the cellular FLICE-inhibitory protein (c-FLIP) [98]. A cyclooxygenase (COX)-2 inhibitor (NS398) induced G0/G1 arrest, but no apoptosis in OSCC cells [99]. On the other hand, human glioblastoma cell lines (M059J, M059K, U373-MG and T98G) has been reported to more exclusively commit to autophagy (characterized by autophagosome formation, the accumulation of Agp8p/Aut7p and LC3 (Atg8 homolog) in autophagosome, and the inhibition of cell death by 3-methyladenine, an autophagic inhibitor), upon exposure to radiation (137Cs)[100], arsenic trioxide [101], ceramide [102] or temozolomide (a new alkylating agent) [103] or doxorubicin [104]. Anthracyclines, with the highest tumor-specificity, also induced non-apoptotic cell death in acute myeloblastic leukemia [105], cardiomyocytes [106] and breast cancer cells [107]. Another factor that determines the type of cell death is the category of compounds that is used. a,b-Unsaturated ketones (2-cyclohexen-1-one, 2-cyclopenten-1-one, 4,4dimethyl-2-cyclopenten-1-one, 2-cyclohepten-1-one, amethylene-g-butyrolactone, 6-dihydro-2H-pyran-2-one, methyl 2-oxo-2H-pyran-3-carboxylate, codeinone, morphinone) and 3,5-dibenzoyl-1,4-dihydropyridines activated caspase only marginally in HL-60 cells. Morphinone, an oxidative metabolite of morphine, similarly induced non-apoptotic cell death, accompanied by a significant reduction of mitochondrial size. There is evidence that modulation of one form of cell death may lead to another. Various stimuli (such as etoposide, TNF, hyperthermia, UV irradiation, ascorbic acid, hydrogen peroxide) induced apoptosis (internucleosomal DNA fragmentation) in various human leukemia cell lines at relatively lower concentrations, whereas it induced necrotic cell death (smear pattern of DNA fragmentation) at higher concentrations [108]. Recently, cross-talk between apoptotic and autophagic pathways has been suggested [109,110]. This review reveals that tumor-specificity and apoptosis induction do not always correlate with each other. This means that having an apoptosis-inducing activity does not guarantee its antitumor activity. Similar is true for autophagy-inducing compounds, since a,b-unsaturated ketones which exclusively induce non-apoptosis, have shown broad range of tumor-specificity (TS = 1—200). The tumor-specific cytotoxicity may be affected by various factors such as the co-existence of both hydrophilic and hydrophobic groups in the same molecule, the presence of an isoprenyl group, a halogen and/or a polycyclic structure, a highly condensed structure, and lipophilicity. The expression of multi-drug resistant proteins and drug metabolizing enzymes may also play an important role. External factors that may affect the tumor-specificity include the type of serum, oxygen concentration, metallic ion presence/concentration and external pressure. The cytotoxic activity of curcumin and nobobactins was significantly inhibited by the addition of FeCl3 due to the chelate formation. Systematization of the relationship between various factors mentioned above and tumor-specificity may contribute to the quest for more active compounds.
184 Both the resistance of tumor cells to anticancer drugs and the dose-related toxicity remain the most important problems in the chemotherapy of clinical OSCC. Researchers have been seeking a combinative treatment regimen to improve the effect of chemotherapy. 13-Retinoic acid, interferon-a2a and a-tocopherol synergistically induced apoptosis in squamous cell carcinoma of the head and neck (SCCHN) may be a basis of the promising outcomes of the phase III trial with a combination of these three drugs in patients with advanced head and neck cancer in the adjuvant settings [62].
Conflict of interest There is no financial and personal relationships with other people or organizations that could inappropriately influence this review article.
Acknowledgement This article is supported in part by a Grant-in-Aid from the Ministry of Education, Science, Sports and Culture of Japan (Grant no: Sakagami, No. 19592156).
References [1] Vogelstein B, Kinzler K. Cancer genes and the pathways they control. Nat Med 2004;10:789—99. [2] Squarize CH, Castilho RM, Santos Jr PD. Immunohistochemical evidence of PTEN in oral squamous cell carcinoma and its correlation with the histological malignancy grading system. J Oral Pathol Med 2002;31:379—84. [3] Tokman B, Gultekin SE, Sezer C, Alpar R. The expression of p53, p16 proteins and prevalence of apoptosis in oral squamous cell carcinoma. Correlation with mode of invasion grading system. Saudi Med J 2004;25:1922—30. [4] Jane C, Nerurkar AV, Shirsat NV, Deshpande RB, Amrapurkar AD, Karjodkar FR. Increased survivin expression in high-grade oral squamous cell carcinoma: a study in Indian tobacco chewers. J Oral Pathol Med 2006;35:595—601. [5] Wang X, Sun SZ, Zhang XY, Ma BL. The expression of Fas and bcl2 in hamster buccal carcinogenesis. Shanghai Kou Qiang Yi Xue 2005;14:155—8. [6] Leonardi R, Pannone G, Margo G, Kudo Y, Takata T, Lo Muzio L. Differential expression of heat shock protein 27 in normal oral mucosa, oral epithelial dysplasia and squamous cell carcinoma. Oncol Rep 2002;9:261—6. [7] Miyashita H, Mori S, Motegi K, Fukumoto M, Uchida T. Pin 1 is overexpressed in oral squamous cell carcinoma and its levels correlate with cyclin D1 overexpression. Oncol Rep 2003; 10:455—61. [8] Childs G, Fazzari M, Kung G, Kawachi N, Brabdwein-Gensler M, McLemore M, et al. Low-level expression of microRNAs let-7d and miR-205 are prognostic markers of head and neck squamous cell carcinoma. Am J Pathol 2009;174:736—45. [9] Hsu S, Singh B, Schuster G. Induction of apoptosis in oral cancer cells: agents and mechanisms for potential therapy and prevention. Oral Oncol 2004;40:461—73. [10] Elmore S, Apoptosis:. A review of programmed cell death. Toxicol Pathol 2007;35:495—516. [11] Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 2008;4:151—75. [12] Hacker G. The morphology of apoptosis. Cell Tissue Res 2000; 301:5—17.
H. Sakagami [13] Noda T, Suzuki K, Ohsumi Y. Yeast autophagosomes: de novo formation of a membrane structure. Trends Cell Biol 2002; 12:231—5. [14] Ohsumi Y. Molecular dissection of autophagy: two ubiquitinlike systems. Nat Rev Mol Cell Biol 2001;2:211—6. [15] Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol 1995;146:3—15. [16] Ueta E, Kamatani T, Yamamoto T, Osaki T. Tyrosine-nitration of caspase 3 and cytochrome c does not suppress apoptosis induction in squamous cell carcinoma cells. Int J Cancer 2003;103:717—22. [17] Xu JH, Huang HZ, Pan CB, Zhang B, Zhang LT. Role of surviving gene on the apoptosis of Tca8113 cells induced by cisplatin. Hua Xi Kou Qiang Yi Xue Za Zhi 2007;25:173—6 [in Chinese]. [18] Azuma M, Tamatani T, Ashida Y, Takashima R, Harada K, Sato M. Cisplatin induces apoptosis in oral squamous carcinoma cells by the mitochondria-mediated but not the NF-kB-suppressed pathway. Oral Oncol 2003;39:282—9. [19] Sundelin K, Roberg K, Gre ˚kansson L. Effects of ´nman R, Ha cisplatin, alpha-interferon, and 13-cis retinoic acid on the expression of Fas (CD95), intercellular adhesion molecule-1 (ICAM-1), and epidermal growth factor receptor (EGFR) in oral cancer cell lines. J Oral Pathol Med 2007;36:177—83. [20] Mishima K, Nariai Y, Yoshimura Y. Carboplatin induces Fas (APO1/CD95)-dependent apoptosis of human tongue carcinoma cells: sensitization for apoptosis by upregulation of FADD expression. Int J Cancer 2003;105:593—600. [21] Itoh M, Chiba H, Noutomi T, Takada E, Mizuguchi J. Cleavage of Bax-alpha and Bcl-x(L) during carboplatin-mediated apoptosis in squamous cell carcinoma cell lines. Oral Oncol 2000; 36:277—85. [22] Nonaka M, Ikeda H, Fujisawa A, Uehara M, Inokuchi T. Induction of apoptosis by paclitaxel in human oral carcinoma cells. Int J Oral Maxillofac Surg 2006;35:649—52. [23] Takemura K, Noguchi M, Ogi K, Tokino T, Kubota H, Miyazaki A, et al. Enhanced Bax in oral SCC in relation to antitumor effects of chemotherapy. J Oral Pathol Med 2005;34:93—9. [24] Okamura H, Morimoto H, Haneji T. Peplomycin-induced apoptosis in oral squamous carcinoma cells depends on bleomycin sensitivity. Oral Oncol 2001;37:379—85. [25] Tai KW, Chou MY, Hu CC, Yang JJ, Chang YC. Induction of apoptosis in KB cells by pingyangmycin. Oral Oncol 2000; 36:242—7. [26] Yamamoto T, Yoneda K, Ueta E, Doi S, Osaki T. Enhanced apoptosis of squamous cell carcinoma cells by interleukin-2activated cytotoxic lymphocytes combined with radiation and anticancer drugs. Eur J Cancer 2000;36:2007—17. [27] Shareef MM, Brown B, Shajahan S, Sathishkumar S, Arnold SM, Mohiuddin M, et al. Lack of P-glycoprotein expression by lowdose fractionated radiation results from loss of nuclear factorkappaB and NF-Y activation in oral carcinoma cells. Mol Cancer Res 2008;6:89—98. [28] King TA, Ghazaleh RA, Juhn SK, Adams GL, Ondrey FG. Induction of heat shock protein 70 inhibits NF-kappa-B in squamous cell carcinoma. Otolaryngol Head Neck 2005;13:70—9. [29] Yasumoto J, Kirita T, Takahashi A, Ohnishi K, Imai Y, Yuki K, et al. Apoptosis-related gene expression after hyperthermia in human tongue squamous cell carcinoma cells harboring wild-type or mutated-type p53. Cancer Lett 2004;10: 41—51. [30] Iwase M, Takaoka S, Uchida M, Yoshiba S, Kondo G, Watanabe H, et al. Epidernal growth factor receptor inhibitors enhance susceptibility to Fas-mediated apoptosis in oral squamous cell carcinoma cells. Oral Oncol 2008;44:361—8. [31] Cha JD, Jeong MR, Kim HY, Lee JC, Lee KY. MAPK activation is necessary to the apoptotic death of KB cells induced by the essential oil isolated from Artemisia iwayomogi. J Ethnopharmacol 2009;123:308—14.
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma [32] Liang CH, Wang GH, Liaw CC, Lee MF, Wang SH, Cheng DL, et al. Extracts from Cladiella australis, Clavularia viridis and Klyxum simplex (soft corals) are capable of inhibiting the growth of human oral squamous cell carcinoma cells. Mar Drugs 2008; 6:595—606. [33] Rodrigo KA, Rawal Y, Renner RJ, Schwartz SJ, Tian Q, Larsen PE, et al. Suppression of the tumorigenic phenotype in human oral squamous cell carcinoma cells by an ethanol extract derived from freeze-dried black raspberries. Nutr Cancer 2006;54:58—68. [34] Schuck AG, Ausubel MB, Zuckerbraun HL, Babich H. Theaflavin3,30 -digallate, a component of black tea: an inducer of oxidative stress and apoptosis. Toxicol In Vitro 2008;22:598—609. [35] Hsu S, Lewis J, Singh B, Schoenlein P, Osaki T, Athar M, et al. Green tea polyphenol targets the mitochondrial in tumor cells inducing caspase 3-dependent apoptosis. Anticancer Res 2003;23:1533—9. [36] Hsu S, Yu FS, Lewis J, Singh B, Borke J, Osaki T, et al. Induction of p57 is required for cell survival when exposed to green tea polyphenols. Anticancer Res 2002;22:4115—20. [37] Li N, Chen X, Han C, Chen J. Chemopreventive effect of tea and curcumin on DMBA-induced oral carcinogenesis in hamsters. Wei Sheng Yan Jiu 2002;31:354—7 [in Chinese]. [38] Sakagami H, Kobayashi M, Chien C-H, Kanegae H, Kawase M. Selective toxicity and type of cell death induced by various natural and synthetic compounds in oral squamous cell carcinoma. In Vivo 2007;21:311—20. [39] Hou YY, Wu ML, Hwang YC, Chang FR, Wu YC, Wu CC. The natural diterpenoid ovatodiolide induces cell cycle arrest and apoptosis in human oral squamous cell carcinoma Ca9-22 cells. Life Sci 2009;85:26—32. [40] Min R, Tong J, Wenjun Y, Wenhu D, Xiaojian Z, Jiacai H, et al. Growth inhibition and induction of apoptosis in human oral squamous cell carcinoma Tca-8113 cell lines by Shikonin was partly through the inactivation of NF-kappa B pathway. Phytother Res 2008;22:407—15. [41] Keum YS, Kim J, Lee KH, Park KK, Surh YJ, Lee JM, et al. Induction of apoptosis and capase-3 activity by chemopreventive [6]-paradol and structurally related compounds in KB cells. Cancer Lett 2002;177:41—4. [42] Du Y, Zhang S, Wang Z, Zhou W, Luan M, Yang X, et al. Induction of apoptosis and cell cycle arrest by NS398 in oral squamous cell carcinoma cells via downregulation of E2 promoter-binding factor-1. Oncol Rep 2008;20:605—11. [43] Ko SH, Choi GJ, Lee JH, Han YA, Lim SJ, Kim SH. Differential effects of selective cyclooxygenase-2 inhibitors in inhibiting proliferation and induction of apoptosis in oral squamous cell carcinoma. Oncol Rep 2008;19:425—33. [44] Nikitakis NG, Hebert C, Lopes MA, Reynolds MA, Sauk JJ. PPARgamma-mediated antineoplastic effect of NSAID sulindac on human oral squamous carcinoma cells. Int J Cancer 2002;98:817—23. [45] Hamada M, Sumi T, Iwai S, Nakazawa M, Yura Y. Induction of endonuclease G-mediated apoptosis in human oral squamous cell carcinoma cells by protein kinase C inhibitor safingol. Apoptosis 2006;11:47—56. [46] Noda T, Iwai S, Hamada M, Fujita Y, Yura Y. Induction of apoptosis of detached oral squamous cell carcinoma cells by safingol. Possible role of Bim, focal adhesion kinase and endonuclease G. Apoptosis 2009;14:287—97. [47] Otsubo A, Bhawal UK, Nomura Y, Mitani Y, Ozawa K, Kuniyasu H, et al. UCN-01 (7-hydroxystaurosporine) induces apoptosis and G1 arrest of both primary and metastatic oral cancer cell lines in vitro. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2007;103:391—7. [48] Fujita M, Seta C, Fukuda J, Kobayashi S, Haneji T. Induction of apoptosis in human oral squamous carcinoma cell lines by protein phosphatase inhibitors. Oral Oncol 1999;35:401—8.
185
[49] Goto K, Fukuda J, Haneji T. Okadaic acid stimulates apoptosis through expression of Fas receptor and Fas ligand in human oral squamous carcinoma cells. Oral Oncol 2002;38:16—22. [50] Mihara M, Shintani S, Nakashiro K, Hamakawa H. Flavopiridol, a cyclin dependent kinase (CDK) inhibitor, induces apoptosis by regulating Bcl-x in oral cancer cells. Oral Oncol 2003;39:49—55. [51] Kudo Y, Takata T, Ogawa I, Kaneda T, Sato S, Takekoshi T, et al. p27Kip1 accumulation by inhibition of proteasome function induces apoptosis in oral squamous cell carcinoma cells. Clin Cancer Res 2000;6:916—23. [52] Siavash H, Nikitakis NG, Sauk JJ. Abrogation of IL-6-mediated JAK signalling by the cyclopentenone prostaglandin 15dPGJ(2) in oral squamous carcinoma cells. Br J Cancer 2004; 91:1074—80. [53] Sakate K, Takagi E, Ishii A, Kato Y, Imagawa Y, Kimura Y, et al. Anti-tumor effect of vitamin A an D on head and neck squamous cell carcinoma. Auris Nasus Larynx 2003;30:403—12. [54] Gaun W, Yu S, Gao Y. Alternation on Fas/FasL gene expression in oral squamous cell carcinoma cell line Tca83 by retinoid acid. Zhonghua Kou Qiang Yi Xue Za Zhi 2001;36:215—8. [55] Zhou H, Tang Y, Liang X, Yang X, Yang J, Zhu G, et al. RNAi targeting urokinase-type plasminogen activator receptor inhibits metastasis and progression of oral squamous cell carcinoma in vivo. Int J Cancer 2009;125:453—62. [56] Duan L, Yao J, Wu X, Fan M. Growth suppression induced by Notch 1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells. Biol Cell 2006;98:479—90. [57] Lee SK, Kim HS, Lee HJ, Lee J, Jeon BH, Jun CD, et al. Dual effect of nitric oxide in immortalized and malignant human oral keratinocytes: induction of apoptosis and differentiation. J Oral Pathol Med 2006;35:352—60. [58] Takahashi M, Sato T, Shinohara F, Echigo S, Rikiishi H. Possible role of glutathione in mitochondrial apoptosis human oral squamous cell carcinoma caused by inorganic selenium compounds. Int J Oncol 2005;27:489—95. [59] Guo J, Zhang ZY. Investigation of apoptosis mechanism of arsenic trioxide on oral squamous cell carcinoma. Zhoughua Kou Qiang Yi Xue Za Zhi 2003;38:20—3 [in Chinese]. [60] Petrovic L, Schlegel KA, Ries J, Park J, Diebel E, Schultze-Mosgau S, et al. In vitro effect of taurolidine on squamous cell carcinoma in the oral cavity. Mund Kiefer Gesichtschir 2003;7:102—7. [61] Bonner JA, Raisch KP, Trummell HQ, Robert F, Meredith RF, Spencer SA, et al. Enhanced apoptosis with combination C225/ radiation treatment serves as the impetus for clinical investigation in head and neck cancer. J Clin Oncol 2000;18(21 Suppl.):47S—53S. [62] Zhang X, Chen ZG, Khuri FR, Shin DM. Induction of cell cycle arrest and apoptosis by a combined treatment with 13-cisretinoic acid, interferon-alpha2a, and alpha-tocopherol in squanous cell carcinoma of the head and neck. Head Neck 2007;29:351—61. [63] Kanazawa T, Kommareddi PK, Iwashita T, Kumar B, Misawa K, Misawa Y, et al. Galanin receptor subtype 2 suppresses cell proliferation and induces apoptosis in p53 mutant head and neck cancer cells. Clin Cancer Res 2009;15:2222—30. [64] Gillenwater AM, Zhong M, Lotan R. Histone deacetylase inhibitor suberoylanilide hydroxamic acid induces apoptosis through both mitochondrial and Fas (Cd95) signaling in head and neck squamous carcinoma cells. Mol Cancer Ther 2007;6:2967—75. [65] Gu X, Song X, Dong Y, Cai H, Walters E, Zhang R, et al. Vitamin E succinate induces ceramide-mediated apoptosis in head and neck squamous cell carcinoma in vitro and in vivo. Clin Cancer Res 2008;15:1840—8. [66] Tano T, Okamoto M, Oshikawa T, Ahmed SU, Sasai A, Sato M. Induction of apoptosis in human head and neck cancer cell lines by an active component of OK-432 through p53-independent pathway via toll-like receptor (TLR)4 signalling. Gan To Kagaku Ryoho 2005;32:1562—4 [in Japanese].
186 [67] Flinterman M, Ga ¨ken J, Farzaneh F, Tavassoli M. E1A-mediated suppression of EGFR expression and induction of apoptosis in head and neck squamous carcinoma cell lines. Oncogene 2003;22:1965—77. [68] Sato M, Harada K, Yura Y, Azuma M, Kawamata H, Iga H, et al. The treatment with differentiation- and apoptosis-inducing agent, vesnarinone, of a patient with oral squamous cell carcinoma. Apoptosis 1997;2:313—8. [69] Nikitakis NG, Scheper MA, Papanikolaou VS, Sauk JJ. The oncogenic effects of constitutive Stat3 signaling in salivary gland cancer cells are mediated by survivin and modulated by the NSAID sulindac. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2009;107:826—36. [70] Yajima T, Ochiai H, Uchiyama T, Takano N, Shibahara T, Azuma T. Resistance to cytotoxic chemotherapy-induced apoptosis in side population cells of human oral squamous cell carcinoma cell line Ho-1-N-1. Int J Oncol 2009;35:273—80. [71] Kajihara A, Takahashi A, Ohnishi K, Imai Y, Yamakawa N, Yasumoto J, et al. Protein microarray analysis of apoptosisrelated protein expression following heat shock in human tongue squamous cell carcinomas containing different p53 phenotypes. Int J Hyperthermia 2008;24:605—12. [72] Mallick S, Patil R, Gyanchandani R, Pawar S, Palve V, Kannan S, et al. Human oral cancers have altered expression of Bcl-2 family members and increased expression of the anti-apoptotic splice variant of Mcl-1. J Pathol 2009;217:398—407. [73] Yao L, Iwai M, Furuta I. Correlations of bcl-2 and p53 expression with the clinicopathological features in tongue squamous cell carcinomas. Oral Oncol 1999;35:56—62. [74] Hus S, Lewis JB, Borke JL, Singh B, Dickinson DP, Caughman GB, et al. Chemopreventive effects of green tea polyphenols correlate with reversible induction of p57 expression. Anticancer Res 2001;3743—8. [75] Lee JC, Sharma M, Lee YH, Lee NH, Kim SY, Yun JS, et al. Pax9 mediated cell survival in oral squamous carcinoma cell enhanced by c-myb. Cell Biochem Funct 2008;26:892—9. [76] Tu HF, Chen HW, Kao SY, Lin SC, Liu CJ, Chang KW. MDM2 SNP 309 and p53 codon 72 polymorphisms are associated with the outcome of oral carcinoma patients receiving postoperative irradiation. Radiother Oncol 2008;87:243—52. [77] Ueta E, Yoneda K, Kimura T, Tatemoto Y, Doi S, Yamamoto T, et al. Mn-SOD antisense upregulates in vivo apoptosis of squamous cell carcinoma cells by anticancer drugs and gamma-rays regulating expression of the BCL-2 family proteins, COX-2 and p21. Int J Cancer 2001;94:545—50. [78] Ueta E, Yoneda K, Yamamoto T, Osaki T. Manganese superoxide dismutase negatively regulates the induction of apoptosis by 5fluorouracil, peplomycin and gamma-rays in squamous cell carcinoma cells. Jpn J Cancer Res 1999;90:555—64. [79] Azuma M, Harada K, Supriatno, Tamatani T, Motegi K, Ashida Y, et al. Potentiation of induction of apoptosis by sequential treatment with cisplatin followed by 5-fluorouracil in human oral cancer cells. Int J Oncol 2004;24:1449—55. [80] Tavassoli M, Soltaninia J, Rudnicka J, Mashanyare D, Johnson N, Ga ¨ken J. Tamoxifen inhibits the growth of head and neck cancer cells and sensitizes these cells to cisplatin-induced apoptosis: role of TGF-beta 1. Carcinogenesis 2002;23: 1569—75. [81] Oba M, Yano S, Shuto T, Suico MA, Eguma A, Kai H. IFN-gamma down-regulates Hsp27 and enhances hyperthermia-induced tumor cell death in vitro and tumor suppression in vivo. Int J Oncol 2008;32:1317—24. [82] Mishima K, Nariai Y, Yoshimura Y. Etodolac, a selective cyclooxygenase-2 inhibitor, enhances carboplatin-induced apoptosis of human tongue carcinoma cells by down-regulation of FAP-1 expression. Oral Oncol 2005;41:77—81. [83] Suzuki M, Endo M, Shinohara F, Echigo S, Rikiishi H. Enhancement of cisplatin cytotoxicity by SAHA involves endoplasmic reticulum
H. Sakagami
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97] [98]
[99]
stress-mediated apoptosis in oral squamous cell carcinoma cells. Cancer Chemother Pharmacol 2009;64:1115—22. Shen J, Huang C, Jiang L, Gao F, Wang Z, Zhang Y, et al. Enhancement of cisplatin induced apoptosis by suberoylanilide hydroxamic acid in human oral squamous cell carcinoma cell lines. Biochem Pharmacol 2007;73:1901—9. Quan F, Pan CE, Zhang SQ, Yan LY, Yu L. Reversal effect of resveratrol on chemotherapy resistance in KBv200 cell line and underlying mechanisms. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2009;44:63—8 [in Chinese]. Imai Y, Ohnishi K, Yasumoto J, Kajiwara A, Yamakawa N, Takahashi A, et al. Glycerol enhances radiosensitivity in a human oral squamous cell carcinoma cell line (Ca9-22) bearing a mutant p53 gene via Bax-mediated induction of apoptosis. Oral Oncol 2005;41:631—6. Wakasa T, Inoue T, Kawai N, Murakami J, Kishi K, Fukui K. The combination of ionizing radiation and expression of a wild type p53 gene via recombinant adenovirus induced a prominent tumour suppressing effect in human oral squamous cell carcinoma. Br J Radiol 2002;75:657—62. Chen W, He R, Zhang Z, Lin G, Zhou X. Enhanced efficacy of the combination of all-trans-retinoic acid and gamma-interferon in inhibiting proliferation of Tca8113 cells. Chin J Dent Res 1999;2:54—60. Brown J, O’Prey J, Harrison PR. Enhanced sensitivity of human oral tumours to the flavonol, morin, during cancer progression: involvement of the Akt and stress kinase pathways. Carcinogenesis 2003;24:171—7. Schleier P, Berndt A, Berndt A, Dahse R, Zenk W, Hyckel P, et al. Experimental 5-aminolevulinic acid-induced photodynamic therapy (ALA-PDT) of oral carcinomas. Procedure in treatment of solid tumors and elucidation of cell death. Mund Kiefer Gesichtschir 2001;5:98—104 [in German]. Sakagami H, Kawase M, Wakabayashi H, Kurihara T. Factors that affect the type of cell death induced by chemicals. Autophagy 2007;3:493—5. Ideo A, Hashimoto K, Shimada J, Kawase M, Sakagami H. Type of cell death induced by a-trifluoromethyl acyloins in oral squamous cell carcinoma. Anticancer Res 2009;29:175—82. Karbowski M, Norris KL, Cleland MM, Jeong S-Y, Youle RJ. Role of Bax and Bak in mitochondrial morphogenesis. Nature 2006;443:658—62. Sakagami H, Chowdhury SA, Suzuki F, Hashimoto K, Hatano H, Takekawa F, et al. Tumor-specific cytotoxic activity of polyphenols, terpenoids, ketones and other synthetic compounds. In: Motohashi N, editor. Functional polyphenols and carotenes with antioxidative action. Kerala, India: Research Signpost; 2005. p. 133—76. Sakagami H, Kobayashi M, Ishihara M, Kikukchi H, Nakamura Y, Kawase M, et al. Tumor-specificity and type of cell death induced by heterocycles. In: Motohashi N, editor. Topics in heterocyclic chemistry, vol. 15. Berlin Heidelberg: Springer-Verlag; 2008. p. 173—99. Miyamoto M, Sakagami H, Minagawa K, Kikuchi H, Nishikawa H, Satoh K, et al. Tumor-specificity and radical scavenging activity of poly-herbal formula. Anticancer Res 2002;22: 1217—23. Tong D, Poot M, Hu D, Oda D. 5-Fluorouracil-induced apoptosis in cultured oral cancer cells. Oral Oncol 2000;36:236—41. Kondo G, Iwase M, Watanabe H, Uchida M, Takaoka S, Ohashi M, et al. Enhancement of susceptibility to Fas-mediated apoptosis in oral squamous cell carcinoma cells by phosphatidylinositol 3-kinase inhibitor. Oral Oncol 2006;42:745—52. Minter HA, Eveson JW, Huntley S, Elder DJ, Hague A. The cyclooxygenase 2-selective inhibitor NS398 inhibits proliferation of oral carcinoma cell lines by mechanism dependent and independent of reduced prostaglandin E2 synthesis. Clin Cancer Res 2003;9:1885—97.
Apoptosis-inducing activity and tumor-specificity of antitumor agents against oral squamous cell carcinoma [100] Daido S, Yamamoto A, Fujiwara K, Sawaya R, Kondo S, Kondo Y. Inhibition of the DNA-dependent protein kinase catalytic subunit radiosensitizes malignant glioma cells by inducing autophagy. Cancer Res 2005;65:4368—75. [101] Kanzawa T, Zhang L, Xiao L, Germano IM, Kondo Y, Kondo S. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene 2005;24:980—91. [102] Daido S, Kanzawa T, Yamamoto A, Takeuchi H, Kondo Y, Kondo S. Pivotal role of the cell death factor BNIP3 in ceramideinduced autophagic cell death in malignant glioma cells. Cancer Res 2004;64:4286—93. [103] Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ 2004;11: 448—57. [104] Hopkins-Donaldson S, Yan P, Bourloud KB, Muhlethaler A, Bodmer JL, Gross N. Doxorubicin-induced death in neuroblastoma does not involve death receptors in S-type cells and is caspaseindependent in N-type cells. Oncogene 2002;21:6132—7. [105] Carter BZ, Kornblau SM, Tsao T, Wang RY, Schober WD, Milella M, et al. Caspase-independent cell death in AML: caspase
[106]
[107]
[108]
[109]
[110]
187
inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis. Blood 2003;102:4179—86. Lim CC, Zuppinger C, Guo X, Kuster GM, Helmes M, Eppenberger HM, et al. Anthracyclines induce calpain-dependent titin proteolysis and necrosis in cardiomyocytes. J Biol Chem 2004;279:8290—9. Bilir A, Altinoz MA, Erkan M, Ozmen V, Aydiner A. Autophagy and nuclear changes in FM3A breast tumor cells after epirubicin, medroxyprogesterone and tamoxifen treatment in vitro. Pathobiology 2001;69:120—6. Yanagisawa-Shiota F, Sakagami H, Kuribayashi N, Iida M, Sakagami T, Takeda M. Endonuclease activity and induction of DNA fragmentation in human myelogenous leukemic cell lines. Anticancer Res 1995;15:259—66. Cui Q, Tashiro S, Onodera S, Ikejima T. Augmentation of oridonin-induced apoptosis observed with reduced autophagy. J Pharmacol Sci 2006;101:230—9. Li D, Cui Q, Chen SG, Wu LJ, Tashiro S, Onodera S, et al. Inactivation of ras and changes of mitochondrial membrane potential contribute to oridonine-induced autophagy in a 431 cells. J Pharmacol Sci 2007;105:22—3.