oncoapoptotic markers in oral cancer: prognostics and therapeutic perspective

12
REVIEW ARTICLE Oncoapoptotic Markers in Oral Cancer: Prognostics and Therapeutic Perspective Anubhav Jain Saurabh Bundela Ram P. Tiwari Prakash S. Bisen Ó Springer International Publishing Switzerland 2014 Abstract Oral cancer is one of the most commonly found cancers in many South-Asian underdeveloped countries, especially among men in comparison to women. When considering the mortality rate among all types of existing cancers, in India oral cancer is the primary reason for death in men. Some of the major reasons contributing to the high mortality rate are late diagnosis, lack of treatment options and high prevalence of tobacco consumption. Oral cancer is often diagnosed at a stage when cancer cells have already become aggressive and become resistant to standard ther- apeutic options. Progression, apoptosis, angiogenesis, metastasis and invasion behold great capability to treat and detect cancer at the molecular level. Dysregulation of apoptosis is one of the most common molecular events known to be associated with the development of oral cancer. In this review, we discuss key apoptotic markers which can be used as prognostic and/or therapeutic targets in oral cancer. Key Points Apoptosis, tightly regulated by various molecular factors, is an evolutionary conserved process which maintains subtle balance between progression and cell death. The regulation of apoptosis involves tumor suppressor genes, oncogenes, the Bcl-2 gene family, receptor and mitochondrial apoptotic factors and caspases. Dysregulation of apoptosis is one of the most common molecular events known to be associated with the development of oral cancer and can be used as a prognostic marker for oral cancer. Therapeutic strategies, by leveraging apoptotic machinery, are focused around promoting apoptosis through activation of the death receptors, pro- apoptotic factors and inhibition of anti-apoptotic factors such as inhibitors of apoptosis (IAPs) and Bcl-2. 1 Introduction Oral cancer is an umbrella term which includes cancer of the lip, tongue, mouth, oropharynx, piriform sinus, hypo- pharynx and other ill-defined sites of the lip, oral cavity and pharynx. Oral cancer has emerged as the top killer among men in India; more than 20 % of cancer-related A. Jain Diagnostic R&D Division, RFCL Limited, New Delhi, India S. Bundela Department of Postgraduate Studies and Research in Biological Sciences, Rani Durgawati Vishwavidyalaya, Jabalpur, India R. P. Tiwari Rapid Diagnostic Group of Companies, New Delhi, India P. S. Bisen (&) School of Studies in Biotechnology, Jiwaji University, Gwalior 474 011, India e-mail: [email protected] P. S. Bisen Defense Research & Development Establishment, DRDO, Ministry of Defense, Government of India, Jhansi Road, Gwalior 474 002, India Mol Diagn Ther DOI 10.1007/s40291-014-0104-5

Upload: puchd

Post on 20-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

REVIEW ARTICLE

Oncoapoptotic Markers in Oral Cancer:Prognostics and Therapeutic Perspective

Anubhav Jain • Saurabh Bundela •

Ram P. Tiwari • Prakash S. Bisen

� Springer International Publishing Switzerland 2014

Abstract Oral cancer is one of the most commonly found

cancers in many South-Asian underdeveloped countries,

especially among men in comparison to women. When

considering the mortality rate among all types of existing

cancers, in India oral cancer is the primary reason for death

in men. Some of the major reasons contributing to the high

mortality rate are late diagnosis, lack of treatment options

and high prevalence of tobacco consumption. Oral cancer

is often diagnosed at a stage when cancer cells have already

become aggressive and become resistant to standard ther-

apeutic options. Progression, apoptosis, angiogenesis,

metastasis and invasion behold great capability to treat and

detect cancer at the molecular level. Dysregulation of

apoptosis is one of the most common molecular events

known to be associated with the development of oral

cancer. In this review, we discuss key apoptotic markers

which can be used as prognostic and/or therapeutic targets

in oral cancer.

Key Points

Apoptosis, tightly regulated by various molecular

factors, is an evolutionary conserved process which

maintains subtle balance between progression and

cell death.

The regulation of apoptosis involves tumor

suppressor genes, oncogenes, the Bcl-2 gene family,

receptor and mitochondrial apoptotic factors and

caspases.

Dysregulation of apoptosis is one of the most

common molecular events known to be associated

with the development of oral cancer and can be used

as a prognostic marker for oral cancer.

Therapeutic strategies, by leveraging apoptotic

machinery, are focused around promoting apoptosis

through activation of the death receptors, pro-

apoptotic factors and inhibition of anti-apoptotic

factors such as inhibitors of apoptosis (IAPs) and

Bcl-2.

1 Introduction

Oral cancer is an umbrella term which includes cancer of

the lip, tongue, mouth, oropharynx, piriform sinus, hypo-

pharynx and other ill-defined sites of the lip, oral cavity

and pharynx. Oral cancer has emerged as the top killer

among men in India; more than 20 % of cancer-related

A. Jain

Diagnostic R&D Division, RFCL Limited, New Delhi, India

S. Bundela

Department of Postgraduate Studies and Research in Biological

Sciences, Rani Durgawati Vishwavidyalaya, Jabalpur,

India

R. P. Tiwari

Rapid Diagnostic Group of Companies, New Delhi, India

P. S. Bisen (&)

School of Studies in Biotechnology, Jiwaji University,

Gwalior 474 011, India

e-mail: [email protected]

P. S. Bisen

Defense Research & Development Establishment, DRDO,

Ministry of Defense, Government of India, Jhansi Road,

Gwalior 474 002, India

Mol Diagn Ther

DOI 10.1007/s40291-014-0104-5

deaths in India were due to oral cancer [1]. Oral cancer is

also the seventh most common form of cancer worldwide

and is estimated to increase by over 60 % in next two

decades [2]. Technological constraints do not allow cancers

of the oral cavity to be detected at an early and clinically

manageable stage. The understanding of risk factor asso-

ciated with oral cancer and unambiguous diagnosis of oral

cancer at an early stage is highly desirable to improve

survival rate. Several key genes have been implicated in

oral carcinogenesis. These implicated genes are involved in

processes such as cell proliferation, differentiation, and cell

survival. The study of signal transduction pathways for

mechanisms of apoptosis has significantly advanced our

understanding of human cancer, subsequently leading to

more effective treatments. Apoptosis is a critical process

responsible for maintaining the subtle balance between

progression and cell death. It is an evolutionary conserved

process which is tightly regulated by various molecular

factors in normal cells, and its dysregulation is associated

with diseases such as cancer, heart attack, liver failure,

stroke, etc. [3]. Cancer cells adapt various strategies to

survive and grow in the cellular microenvironment; eva-

sion of apoptosis is one such strategies. Chemotherapeutic

drugs act by killing cancerous cells which invariably

involves the apoptotic pathway. Dysregulation of factors

involved in the apoptotic pathway leads to the development

of chemoresistance [3, 4].

Factors involved in the initiation of apoptosis mainly act

through two pathways—extrinsic and intrinsic apoptotic

pathways [5] (Fig. 1). The extrinsic apoptotic pathway is

initiated by interaction between death ligands with extra-

cellular death receptors. Activated death receptors, along

with an adaptor protein such as FADD, and procaspase 8

form the death-inducible signaling complex (DISC), which

in turn leads to the activation of apoptosis effector protein

caspase 3 through caspase 8. The intrinsic apoptotic path-

way, which is also known as the mitochondrial pathway, is

initiated in response to DNA damage or cytotoxic stress.

The Bcl-2 protein family is the main regulator of the

intrinsic apoptotic pathway; it consists of both anti-apop-

totic and pro-apoptotic factors. The pro-apoptotic factors

(e.g. Bcl-2-associated protein X [Bax], Bak, Noxa, Puma)

are responsible for increasing mitochondrial membrane

permeability, whereas anti-apoptotic factors (e.g. Bcl-xL,

Bcl-2 and Mcl-1) are responsible for maintaining mito-

chondrial membrane integrity. The induction of the

intrinsic apoptotic pathway requires the presence of Bak or

Bax [6]. Activation of Bak and Bax is achieved either by

direct interaction with t-Bid or through neutralization of

Bcl-2 family proteins which keep a check on Bax and Bak.

BH3-only proteins such as Puma, Noxa, Bid and Bad, are

some of the important factors that are involved in the

neutralization of anti-apoptotic Bcl-2 proteins [7].

Mitochondria releases the inhibitor of inhibitors of apop-

tosis (IAPs), such as DIABLO/Smac and Omi/HtrA2,

apoptosis-inducing factor (AIF) and endonuclease G.

These proteins can contribute to apoptosis via caspase-

independent pathways to cell death. p53 is another

important regulator of apoptosis which mainly acts by

transcriptional regulation of apoptosis effector genes.

Among the IAP gene family protein, survivin is distinctly

identified as a critical protein with a dual role in the inhi-

bition of apoptosis and regulation of mitosis. These key

players of apoptosis can be exploited for therapeutic and/or

prognostic application in oral cancer. Activation of the

death receptors, pro-apoptotic factors (Caspases, p53) and

inhibition of anti-apoptotic factors such as IAPs and Bcl-2,

are some therapeutic strategies for controlling cancer.

Dysregulation of factors of apoptosis pathways can be used

as prognostic and/or prognostic markers for oral cancer

(Table 1).

2 Apoptotic Markers of Relevance to Prognosis

and Prediction of Outcome in Oral Cancer

2.1 Bcl-2 and Bcl-2-associated protein X (Bax)

Bcl-2 is a human proto-oncogene located on chromosome

18q21; it codes for Bcl-2 protein which is an integral

membrane protein located in the membranes of the endo-

plasmic reticulum (ER), nuclear envelope, and in the outer

membranes of mitochondria [8]. Bax is the most charac-

teristic death-promoting member of the Bcl-2 family. The

activation of the caspase cascade is triggered by the

translocation of Bax protein from the cytosol to the mito-

chondria, and the activated caspase cascade leads to cell

death [9]. The Bcl-2 family comprises cell death antago-

nists, such as Bcl-2 and Bcl-XL, and death agonists, such

as Bax and Bad. The Bcl-2 family proteins decide whether

a cell continues to survive or instead commits to death

through the mitochondrial apoptotic pathway, and carries

high prognostic significance in oral cancer. Bcl-2, Bax and

Bcl-2/Bax ratio can be used as effective biomarkers to

predict the prognosis of oral cancer. The 5-year survival

rate was significantly higher in patients with a ratio of Bcl-

2/Bax B1 than in those with Bcl-2/Bax [1

(76.79 ± 6.69 % vs. 59.26 ± 6.69 %; p = 0.0489) [10,

11]. The expression of Bcl-2 and Bax is reported to be a

significant predictor of invasiveness in oral cancer [12].

Bcl-2 and Bax expression were also found to be indepen-

dent prognostic factors for overall survival and cancer-

specific 5-year survival (CSS). Bcl-2 expression emerged

as an independent marker of favorable CSS, and expression

of Bcl-2 family members was found to be an important

marker of a favorable prognosis in oral squamous cell

A. Jain et al.

carcinoma (OSCC) [13]. In oral cancer, Bcl-2 is observed

from the initial stages of carcinogenesis up to the appear-

ance of metastasis [14–16]. The association between Bcl-2

expression and oral cancer has also been refuted in some

other studies. Yuen et al. [17] observed no association

between Bcl-2 expression and survival in OSCC. Another

study reported associations between increased Bcl-2

expression, neck metastasis and poor survival, but did not

detect any prognostic significance for Bax expression [18].

The lack of consensus in these studies may be attributed to

the lack of robust and reliable techniques for measuring

biomarker expression. Recently, automated quantitative

immunohistochemistry-based techniques, such as AQUA,

were used to eliminate observer bias in the measurement of

protein expression, and found Bax expression as an inde-

pendent prognostic marker in OSCC [19].

2.2 Survivin

Survivin is an important member of the IAP protein family

and has become the focus of various cancer research studies,

mainly due to its highly specific expression in most solid and

hematological malignancies [20]. Survivin negatively reg-

ulates apoptosis by inhibiting caspase activation. The over-

expression of survivin imparts survival benefit to cancer cells

and is therefore consistently associated with tumor aggres-

siveness and poor prognosis. For example, normal oral

mucosa and skin, including adnexal structures, are negative

for survivin. In contrast, one study found that 56 % and 64%

of oral and cutaneous squamous cell carcinomas,

respectively, were strongly positive for survivin [21]. The

expression of survivin is associated with larger, more poorly

differentiated tumors and lymph node metastasis.

The overexpression of survivin has been reported in

various human cancers, including oral cancer [22–24]. In a

study on OSCC, high survivin messenger RNA (mRNA)

expression was found to be correlated with poorer tumor

differentiation, higher clinical stage, and the presence of

lymph node metastasis (p\ 0.05). Multivariate analysis

showed that the status of survivin mRNA could be an

independent prognostic factor for OSCC patients (hazard

ratio [HR] 2.71; 95 % confidence interval 1.46–5.10;

p = 0.012), and upregulation of survivin is associated with

a poor prognosis and chemoresistance [25]. Clinicopatho-

logical analysis revealed a significant correlation between

survivin expression and lymph node metastasis (p = 0.006)

and proliferation (p\ 0.001) [26]. Survivin is found to be

prominently expressed in more than 80 % of OSCC, and its

expression often increased in poorly differentiated tumors

and it’s high expression correlated with poor survival rates

[27]. The difference between cytoplasmic and nuclear sur-

vivin is an indicator for survivin activity in tumor cells in

oral cancer patients and this difference may serve as a

predictive marker of outcome in oral cancer patients

undergoing multi-modality therapy [28]. The high expres-

sion of survivin in the oral cavity is an early predictor of

malignant transformation in pre-cancerous and cancerous

lesions [29]. In their study of OSCC in Taiwan, Lin et al.

found no significant correlation between survivin expres-

sion and patient age, sex, oral habits, cancer location, or

Fig. 1 Extrinsic and intrinsic

apoptotic pathway. Apart from

the intrinsic apoptotic pathway,

the extrinsic pathway is another

way through which cells

undergo apoptotic death. The

extrinsic pathway is triggered

by factors (death ligands)

located outside the cell, which

activates specific death

receptors on the cell surface.

The pro-apoptotic ligands

include Apo2L/TRAIL and

CD95L/FasL, which bind to

their cognate receptors, DR4/

DR5 and CD95/Fas,

respectively. The intrinsic

pathway is triggered in response

to stimuli generated due to DNA

damage or cytotoxic stress. Bcl-

2 family proteins and factors

such cytochrome-c, SMAC,

OMI, etc. located inside the

mitochondrion play an

important role in the intrinsic

pathway

Prognostic and Therapeutic Potential of Apoptotic Markers in Oral Cancer

TNM status, but patients with high survivin expression, an

advanced stage, a larger tumour size, or positive lymph

node metastases had a significantly shorter overall survival

than the others [30]. The expression of survivin is a strong

predictor of aggressive and invasive phenotype in oral

cancer patients, and therefore could be used for tailoring

therapeutic procedure [31].

2.3 p53

p53 holds a crucial position in the network of signaling

pathways that are essential for the regulation of cell

growth, and extrinsic and intrinsic apoptotic pathways [32,

33]. Transcriptional regulation of apoptosis effector genes

is the main route by which p53 regulates apoptosis. p53

activates transcription of various pro-apoptotic genes such

as Noxa, PUMA, Bax, Bad, and Bim. It represses tran-

scription of various anti-apoptotic factors such as survivin,

Bcl-2, and Bcl-xL. p53 activates apoptotic protease acti-

vating factor 1 (APAF1) and thereby contributes toward the

formation of apoptosome. p53 promotes the extrinsic

apoptotic pathway by activation of death receptors such as

tumor necrosis factor (TNF)-a, Fas, and DR5.

Deregulation of the p53 tumor suppression network is

observed in many tumor types, including oral cancer, and is

associated with poor prognosis [22, 34]. Mutation in p53 is

Table 1 Role of apoptotic markers in cancer diagnosis and therapy

Apoptotic markers Cancer prognosis Cancer treatment

Survivin Prognosis Transcription factors blockers: YM155, STA3 and SOX2 inhibitors

Lymph node metastasis Antisense oligonucleotides: LY2181308, EZN-3042

Invasiveness siRNA

miRNA (miRNA-34)

Tumor grading Oligopeptides (Shepherdin)

Targeting HSP90 to interfere with protein stabilityChemoresistance

Immunotherapy

p53 Prognosis Gene addition therapy (Ad-p53)

Oncolytic viruses (ONYX-015)

Diagnosis p53 activators: RITA, Nutin-3, MI-219, BDA

Mutant-type p53 reactivator: PRIMA-1, WR-1065, MIRA-1Treatment response

MDM2/p53 interaction blockers: benzodiazepinedione antagonistics

Mcl-1 Treatment response CDK inhibitors: flavopiridol, SNS-032, Sorafenib

Deubiquitinase inhibitors: WP1130

Radioresistance BH3 mimetics: obatoclax, gossypol, sabutoclax, BH3-M6

siRNA

Bcl-2 Prognosis Ribozyme

Interference with protein stabilitySurvival

Invasiveness

Cancer-specific 5-year survival

Bax Prognosis –

Survival

Invasiveness

Cancer-specific 5-year

survival

Fas/FasL Prognosis –

Tumor differentiation

Invasiveness

Tumor grading

Apoptotic marker

TRAIL – Esculetin

Monoclonal antibody

Bcl-2/Bax Prognosis –

Survival

A. Jain et al.

frequently reported in various cancers, including oral

cancer. The loss of function of p53 mutant proteins may

predict a significantly low pathological complete response

rate and suboptimal response to cisplatin-based neoadju-

vant chemotherapy in patients with oral cavity SCC [35].

In a study, 100 % of the oral cancers and 66 % (19/29) of

the pre-malignant lesions examined from Yemen and India

overexpressed p53 [36]. Also, co-expression and correla-

tion between p53 and Ki-67 have been demonstrated in oral

cancer, suggesting that alterations in the p53 protein might

lead to increased cell proliferation. Furthermore, overex-

pression of p53 has been suggested to be a reliable indi-

cator for OC development [37]. The credibility of p53 as an

independent prognostic factor is still not fully established

[38]; however, in several studies it has been reported to be

a significant clinical factor when used in combination with

other markers, such as cyclin D1 and epidermal growth

factor receptor (EGFR) [39]. Mutation in p53 has been

reported to be the most frequent genetic alterations in

human cancer [40]. p53 gene mutations may be better

predictors of recurrence than the expression of the protein,

and serum p53 levels may be more efficient prognosticators

than its tissue immunodetection [41]. The expression of

p53 in the dysplastic epithelium, in association with p16

and Ki-67, can be used as a marker to identify the pre-

cancerous stage of oral cancer [42]. Cox proportional

hazards model showed the presence of p53 antibodies to be

an independent prognostic factor (p = 0.020; HR 3.509)

[43, 44]. The mutations in p53 have been used in prediction

and/or screening of neoplastic samples in OSCC [40, 45].

There are many point mutations reported in p53 by various

research groups and it is difficult to identify a particular

point mutation that is specific for oral cancer, therefore

limiting its application as a cost-effective marker in the

early detection of oral cancer.

2.4 Fas/Fas Ligand

Fas ligand (FasL) is a death ligand that belongs to the TNF

family. It is predominantly expressed in activated T cells.

Activation of Fas with FasL induces the extrinsic apoptotic

pathway in normal and tumor cells. FasL is overexpressed,

and Fas receptor is under expressed, in OSCC, and this

expression pattern correlates with poorly differentiated

tumors [46–48]. High expression of FasL was found to be

associated with greater tumor size and advanced stages of

oral cancer [47]. The downregulation of Fas expression in

keratinocytes and lymphocytes of oral lichen planus (OLP)

specimens, together with upregulation of FasL, may serve

as an initial prognostic biomarker in oral cancer develop-

ment [49]. A significant positive correlation was found

between the argyrophilic nucleolar organizer region

(AgNOR) parameters and Fas/FasL expression as apoptotic

markers in the tumoral cells of oral and oropharyngeal SCC

(p\ 0.05) [50]. AgNOR count was found to be a strong

proliferation marker in patients with OSCC, and Fas and

FasL staining was useful in tumor grading [50].

2.5 Myeloid Cell Leukemia-1

Myeloid cell leukemia-1 (Mcl-1) is a protein encoded by

the MCL1 gene, which is an anti-apoptotic member of the

Bcl-2 family protein. It contains three BH domains and

differs from other anti-apoptotic members of the Bcl-2

family which contain four BH domains. Mcl-1 was found

to mediate resistance to apoptosis in gastric cancer cells by

blocking the mitochondrial pathway of cell death [51].

Mcl-1 was demonstrated to predict outcome in oral cancer

patients treated with definitive radiotherapy [52]. Expres-

sion of the Mcl-1L isoform was found to be significantly

associated with the radioresistance of OSCC [53]. Treat-

ment with Mcl-1L small interfering RNAs (siRNAs) alone

or in combination with ionizing radiation significantly

increased apoptosis in radioresistant oral squamous carci-

noma cells. The Bak/Mcl-1 ratio was reported to be a

useful biomarker to predict clinical outcomes of oral ver-

rucous hyperplasia (OVH) and oral leukoplakia (OL)

lesions treated with photodynamic therapy [54].

2.6 Other Apoptotic Markers

There are other factors from the apoptotic pathway which

can be used as diagnostic and/or prognostic markers for

oral cancer. Growth differentiation factor 15 (GDF15) is a

protein from the transforming growth factor (TGF)-bsuperfamily that has a regulatory role in inflammatory and

apoptotic pathways. A significant high serum level of

GDF15 was reported to be a potential diagnostic and

prognostic marker for oral cancer [55]. Caspases such as

caspase 8 and 9 are significantly expressed in OSCC.

Moreover the expression of caspase 7 was reported to be a

predictor of locoregional recurrence of OSCC [56].

X-linked IAP (XIAP) is reported to be prognostic, and a

treatment response marker [47, 57], and was also found to

be related to histological differentiation [58]. Loss of p21

expression was found to be associated with clinicopatho-

logical factors related to tumor progression, invasiveness,

aggressiveness, and malignancy [59, 60].

3 Apoptotic Markers in Oral Cancer Therapeutics

3.1 Gene Therapy

Gene therapy involves the manipulation of the existing

genetic material by transfer of genetic material into

Prognostic and Therapeutic Potential of Apoptotic Markers in Oral Cancer

targeted cells. Gene therapy is based on the premise that

‘the normal physiological state can be restored when the

gene exerts its normal function’. The comprehensive

understanding of molecular events involved in the devel-

opment of disease, along with the accessibility of target

organ/tissue for delivering therapeutic material, and in vivo

stability of therapeutic material to produce the desired

effect, are some of the major factors responsible for the

success of gene therapy. The easy accessibility of the oral

cavity makes oral cancer an ideal candidate for gene

therapy [61]. The gene therapy procedure typically

involves the following steps: (a) identification of the target

gene, which is involved in disease and qualifies to be a

candidate for gene therapy; (b) isolation and amplification

of the candidate gene; (c) cloning of the candidate gene on

a specific vector; and (d) delivery of candidate genes

through ex vivo or in vivo [4].

3.1.1 Gene Therapy Strategies to Induce Apoptosis in Oral

Cancer Cells

3.1.1.1 Gene Addition Therapy Cancer formation is

typically facilitated by inactivation of tumor suppressor

genes such as p53; gene addition therapy intents to coun-

teract this phenomenon by introduction of the tumor sup-

pressor gene to cancer cells with the help of the appropriate

delivery system. Alteration of p53 is known to be one of

the earliest events in the progressive development of oral

cancer from pre-cancerous lesions [62]. The feasibility and

efficacy of adenoviral-mediated p53 (Ad-p53) gene transfer

were found to be promising in the treatment of advanced

head and neck cancer [63, 64]. These results formed the

basis of human trials for designing clinical therapeutic

regimen of Ad-p53 gene therapy (ClincalTrials.gov num-

ber: NCT00064103).

3.1.1.2 Oncolytic Virus-Mediated Gene Therapy The

application of viruses in cancer therapy is based on the

observation that a cancer regression is seen in cancer

patients when infected with viruses. Advancements in

genetic manipulation techniques have made it possible to

design viruses that can selectively target and kill tumor

cells. The replication of the virus is critically dependent on

the E1B viral protein, and in the absence of a functional

p53 protein. Viruses without the E1B protein are unable to

replicate in the presence of the functional p53 pathway in

the host cell. The p53 pathway becomes dysfunctional in

oral cancer cells; this fact is leveraged in the treatment

strategy of utilizing the oncolytic virus ONYX-015, which

lacks the viral E1B protein [65]. ONYX-015 replicates in

tumor cells and thereby selectively lyses tumor cells

because of the absence of the functional p53 pathway in

these tumor cells. ONYX-015 is currently being evaluated

as a preventive treatment for pre-cancerous oral tissue,

since even in pre-cancerous cells p53 pathway-inactivating

mutations will allow ONYX-015 to destroy and eliminate

the pre-cancerous cells before a tumor develops. The effi-

cacy of ONYX-015 in cancer treatment can be further

enhanced in combination with chemotherapy [66, 67].

3.1.1.3 Antisense RNA This technology is based on post-

transcriptional silencing of the target mRNA with the help

of the introduction of sequence-specific antisense RNA,

also known as siRNA. Apoptosis is evaded in cancer cells

by involvement of survival factors such as Bcl-2, XIAP,

and survivin. These genes involved in the survival pathway

can be selectively inhibited by sequence-specific antisense

RNA. The effective inhibition of oral cancer growth

mediated via induction of apoptosis was observed after the

introduction of siRNA targeting survivin [68, 69]. The

selective silencing of Mcl-1 by siRNA resulted in inhibi-

tion of growth of OSCC accompanied with apoptosis [70].

The silencing of other survival genes such as Bcl-x or Bcl-

xL by siRNA has been reported to induce apoptosis in

various tumor cells and sensitize tumor cells to chemo-

therapy [71, 72]. Induction of apoptosis by an anti-Bcl-2

ribozyme, delivered by an adenovirus vector, and HSP70

oligonucleotide treatment to decrease expression of Bcl-2

has been reported in oral cancer cells [73, 74].

3.2 Targeted Therapy

Chemotherapy is a standard treatment option for various

cancers, including those of the oral cavity. The main

drawback of chemotherapy is indiscriminately killing

normal proliferating cells along with intended cancer cells,

which causes unavoidable toxicities. Owing to scientific

advancements made in the last couple of decades, we are

now in a better position as far as molecular understanding

of oral carcinogenesis is concerned. The detailed study and

understanding of molecular events of oral carcinogenesis

has gifted us with molecular targets which could form the

base for therapeutic intervention. However, this informa-

tion is not as comprehensive as in the case of other well-

studied cancers of breast, lung and colorectum.

Cetuximab is the only approved targeted therapy avail-

able for oral cancer. It targets EGFR, which is involved in

cell progression [75]. Although KRAS, NRAS and BRAF

mutations have been established as potential predictive

biomarkers for cetuximabin in colorectal cancer, little is

known about predictive markers for cetuximabin in oral

cancer. Other targeted therapy for oral cancer in clinical

trial phase targets molecules such as CD44 [GSK1120212]

[76], PPARgamma [Pioglitazone] (ClinicalTrials.gov

number NCT00951379), and EpCAM [Proxinium] (Clini-

calTrials.gov number NCT00272181). There are a host of

A. Jain et al.

potential targets from the apoptotic pathway which can be

exploited for therapeutic intervention in oral cancer; some

of the important targets are discussed here.

3.2.1 Targets of the Extrinsic Pathway

Death ligands (TNF-a, CD95L, TRAIL) and their receptors(DR-4 or TRAIL-R1 and DR-5 or TRAIL-R2) are important

targets from the extrinsic apoptotic pathway. Esculetin was

reported to enhance TRAIL-mediated apoptosis, in the oral

cancer cell line [77]. Monoclonal antibodies (HGS-ETR1,

HGS-ETR2, HGS-TR2J, TRA-8) with agonist function at

death receptors have also been used to induce apoptosis;

however, they have not been tested for oral cancer treat-

ment. Results from preclinical and clinical studies have

shown the potential utility of TRAIL-targeted therapies in

advanced cancers, including oral cancer [78–81].

3.2.2 Targets of Intrinsic Pathway

The intrinsic apoptotic pathway or mitochondrial pathway

is mainly regulated by Bcl-2 family proteins (Fig. 1). The

Bcl-2 protein family consists of anti-apoptotic (e.g. Bcl-xL,

Bcl-2 and Mcl-1) and pro-apoptotic (e.g. Bax, Bak, Noxa,

Puma) factors. The strategies used for inducing apoptosis

through the Bcl-2 family proteins are: (a) silencing or

downregulation of anti-apoptotic proteins by antisense

techniques; (b) release of pro-apoptotic protein from the

Bcl-2 complex, through the application of the BH3 domain

peptide; and (c) release of pro-apoptotic protein from the

Bcl-2 complex, through the application of synthetic small-

molecule drugs such as tetrocarcin A (TC-A) [82, 83],

antimycin A3 [84], and chelerythrine [85]. Small molecules

such as Gossypol, ABT-737, ABT-263, GX15-070 and

HA14-1 have been developed that directly interact with

anti-apoptotic Bcl-2 proteins. These agents mimic the

action of BH3 proteins and interact with anti-apoptotic Bcl-

2 proteins at their BH3-binding groove [86].

The execution of apoptosis involves initiator caspases

(caspase 8, 9, 10) and executioner caspases as caspase 3, 4

and 7. Activation of these caspases by external compound is

an alternative pathway for killing cancer cells through

apoptosis. Many research groups have attempted to find the

caspase activator based on analysis of compound screening

data from high-throughput screening (HTS) studies. In one

such screening study, dichlorobenzyl carbamates and in-

dolones were detected as strong caspase activators [87].

Maxim Pharmaceuticals’ MX-2060 series of caspase-acti-

vating compounds is reported to have caspase-activating

attributes [88]. Caspase activity is inhibited by a class of

regulatory proteins known as IAPs. Survivin and XIAP are

prominent members of the IAP family of proteins which are

currently been pursued by various research groups as

potential cancer therapeutic targets. Cisplatin induces

apoptosis in oral cancer cells through inhibition of XIAP

[89]. Caspase 9 contains the IAP-binding motif (IBM)

through which it binds with the BIR-3 domain of XIAP. A

domain homologous to IBM is found in mitochondrial

proteins SMAC and Omi/HtrA2. SMAC interacts with

XIAP and releases caspase 9 for inducing apoptosis. SMAC

mimetic drugs hold potential in cancer treatment [4].

3.2.3 Targeting p53

p53 is the key regulator of various critical pathways, including

apoptosis; it becomes dysfunctional in the cancerous condi-

tion, thereby imparting survival benefit to the cancer cell.

Restoration of wild-type p53 (WT p53) is a potential strategy

in anticancer therapy.MDM2acts as an inhibitor of p53 and is

found to be overexpressed in various cancers. Blocking of

p53/MDM2 interaction by a variety of agents, such as small-

molecule inhibitors, synthetic peptides and benzodiazepin-

edione antagonists [90–92], has led to p53-dependent apop-

tosis in cancer cells. Activation of mutant-type p53 (MT p53)

by small-molecule activators offers an opportunity to inhibit

tumor growth through p53-mediated pathways. Some of the

WTp53 activators are RITA [93], Nutlin-3 [94],MI-219 [95],

BDA [92], HLI98C [96], Tenovin-1 [97] and JJ78:12 [98].

Mutant-type p53 is unable to perform its normal function

because of the defect in its folding caused by various point

mutations associatedwith cancer. The function ofMTp53 can

be restored with the help of peptides and small-molecule

compounds which act as a stabilizer. They include synthetic

peptides derived from the C-terminus of p53 [99], as well as

peptides such as CDB3 [100, 101], and compounds isolated

fromchemical library screenings suchasPRIMA-1 [102] (p53

reactivation and induction of massive apoptosis) and CP-

31398 [103–106]. Some of the other small-molecule reacti-

vators ofMTp53 areMIRA-1 [107], ellipiticine [108], P53R3

[109] and WR-1065 [110].

3.2.4 Targeting Survivin

Survivin is an attractive target for oral cancer because of its

highly specific expression in cancer tissues [4]. Survivin

can be inhibited at multiple levels, such as (a) interference

with survivin gene (BIRC5) expression by inhibiting its

transcription factors; (b) post-transcriptional regulation by

antisense technologies; (c) regulating upstream regulators

of survivin; (d) disturbing protein stability by modulating

interacting partners; (e) modulating protein stability; and

(f) activation of the immune response. Sepantronium bro-

mide (YM155) can inhibit tumor growth by inhibiting

survivin promoter-reporter gene construct; it was found to

reverse cisplatin resistance in head and neck cancer cells

[111, 112]. The expression of survivin is also controlled by

Prognostic and Therapeutic Potential of Apoptotic Markers in Oral Cancer

transcriptional factors of research interest such as STAT3

and SOX2, and, therefore, targeting these transcription

factors offers an attractive strategy to inhibit survivin [113–

115]. Caspase 2 is shown to repress survivin by interfering

with its transactivation by nuclear factor kappa B (NF-jB)[116]. The post-transcriptional regulation of survivin can

be achieved with the help of antisense oligonucleotides

(ASOs), siRNAs, and micro RNAs (miRNAs). LY2181308

is survivin-directed ASO which was found to effectively

inhibit survivin and enhanced effect of gemcitabine, pac-

litaxel, and docetaxel [117]. EZN-3042 is an ASO that was

to effectively inhibit the expression of survivin [118].

siRNAs directed against survivin have also shown to

effectively inhibit survivin expression [119–121]. miRNA-

34a was found to negatively regulate survivin in gastric

cancer cells, and its modulation in cancer can be consid-

ered as an alternative strategy to inhibit survivin [122]. The

stability of survivin protein can be compromised by tar-

geting its chaperon proteins such as HSP90. Shepherdin is

an oligopeptide that inhibits the formation of survivin

HSP90 complex because of the presence of survivin

sequence from lysine 79 to glycine 83 [123]. Survivin is a

tumor-associated antigen (TAA) which is corroborated by

the presence of survivin-specific CD8? T cells and IgG

antibodies in cancer patients [124]. Survivin-derived epi-

topes have been used in vaccination, and have been shown

to elicit immune response against tumors [125, 126].

Application of peptide ligands is yet another exploratory

strategy to inhibit survivin [127].

3.2.5 Targeting Myeloid Cell Leukemia-1 (Mcl-1)

Although there is no drug that has been designed to directly

target Mcl-1, some drugs such as cyclin-dependent kinase

(CDK) inhibitors (flavopiridol [128], SNS-032 [129], so-

rafenib [130–132]) and deubiquitinase inhibitors (WP1130

[133, 134]) have been reported to elicit anti-cancer activity

by targeting Mcl-1 along with its intended targets. The

CDK inhibitor roscovitine significantly increased ABT-737

(BH3 mimetic drug) lethality in human leukemia cells

through a mechanism involving downregulation of Mcl-1

[135]. BH3 mimetic drugs targeting Mcl-1 are obatoclax

(GX15) [136], gossypol [137], sabutoclax (BI-97C1) [138]

and BH3-M6 [139]. The inhibition of survivin expression

by sequence-specific siRNA was found to induce apoptosis

and growth inhibition in OSCC [70].

4 Conclusions

The molecular events associated with apoptosis in cancer

cells are strategically tuned to support survival and pro-

liferation necessary for tumorous growth. These molecular

events are significantly different in cancer cells when

compared with normal cells, and therefore offer great

potential to be utilized as diagnostic/prognostic/therapeutic

markers. Barring a few gaps, our understanding of the role

of the apoptotic pathway in oral carcinogenesis is almost

complete, which has given us key apoptotic factors such as

p53, survivin, Bcl-2/Bax, Mcl-1 and Fas. It would be

interesting to see how many of these factors will be able to

cross the translational gap between bench and bedside.

Until now, there have been very few markers that have

been able to successfully cross this translational gap

because of inherent challenges in the path of biomolecules

to qualify as definitive biomarkers. Some of the challenges

are related to its specificity and ability to detect oral cancer

much before the clinical manifestation of cancer. More-

over, biomarkers should be available in body fluids such as

blood and saliva for easy detection through non-invasive

tests. Apoptotic markers such as p53, survivin, Bcl-2/Bax

and Mcl-1 have emerged as potential biomarkers for the

diagnosis and prognosis of oral cancer. These biomarkers

should also be studied in various combinations to explore

any hidden pattern that can form the basis of designing the

diagnostic application. With the accumulation of knowl-

edge about the role of apoptosis in oral carcinogenesis, we

should be able to, unambiguously, assign the role of the

biomarker as a single entity and its role in the group

towards contributing towards cancer development; such

knowledge would help the researcher to design precise

analytical tests with high specificity and sensitivity.

Oral cancer is currently managed by the integration of

traditional methods such as surgery, radiation therapy and

chemotherapy. The current treatment options have failed to

improve survival rate, which is why target-based therapies

need to be brought into clinical practice instead of tradi-

tional therapies. The targeted elimination of oral squamous

cell carcinoma cells by inducing apoptosis has emerged as

a valued strategy to combat oral cancer. Targeted therapies

require a detailed understanding of the underlying disease

process. Gene therapies such as ONYX-015, Ad-p53 and

antisense techniques should be treated as a turning point for

oral cancer treatment. The BH3 mimetic class of drugs

have the potential to inhibit multiple anti-apoptotic Bcl-2

family proteins, and could therefore prove to be attractive

treatment options, especially in cases when cancer cells

develop resistance to the drug targeting a single apoptotic

target. With a better understanding of the role of the

apoptotic pathway in oral cancer, we should expect tar-

geted therapies with better efficacy and minimal toxicity.

The success of therapies focused on apoptotic markers

would depend on the skillful combination of diverse drug

classes (such as gene therapy, BH3 mimetic, small-mole-

cule inhibitors, etc.) to effectively kill cancer cells without

developing resistance to therapy.

A. Jain et al.

Acknowledgements and Disclosures The authors are thankful to

the Council of Scientific and Industrial Research (CSIR), New Delhi,

for the award of Emeritus Scientist to Professor P.S. Bisen. No

sources of funding were used to prepare this article. The authors have

no conflicts of interest that are directly relevant to the content of this

article.

References

1. Dikshit R, Gupta PC, Ramasundarahettige C, et al. Million

Death Study Collaborators. Cancer mortality in India: a

nationally representative survey. Lancet. 2012;379:1807–16.

2. Mathers CD, Loncar D. Projections of global mortality and

burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442.

3. Bisen PS, Khan Z, Bundela S. Biology of oral cancer - key

apoptotic regulators. 1st ed. Boca Raton: CRC Press; 2013.

4. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell.

2000;100:57–70.

5. Green DR. Apoptotic pathways: ten minutes to dead. Cell.

2005;121:671–4.

6. Wei MC, Zong WX, Cheng EH, et al. Proapoptotic BAX and

BAK: a requisite gateway to mitochondrial dysfunction and

death. Science. 2001;292:727–30.

7. TaylorRC,CullenSP,MartinSJ.Apoptosis: controlleddemolition

at the cellular level. Nat Rev Mol Cell Biol. 2008;9:231–41.

8. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM.

Cloning of the chromosome breakpoint of neoplastic B cells

with the t(14;18) chromosome translocation. Science.

1984;226:1097–9.

9. Antonsson B. Bax and other pro-apoptotic Bcl-2 family ‘‘killer-

proteins’’ and their victim, the mitochondrion. Cell Tissue Res.

2001;306:347–61.

10. Xie X, Clausen OP, Sudbo J, Boysen M. Diagnostic and prog-

nostic value of nucleolar organizer regions in normal epithelium,

dysplasia and squamous cell carcinoma of the oral cavity.

Cancer. 1999;79:2200–8.

11. Zhang M, Zhang P, Zhang C, et al. Prognostic significance of

Bcl-2 and Bax protein expression in the patients with oral

squamous cell carcinoma. J Oral Pathol Med. 2009;38:307–13.

12. Kato K, Kawashiri S, Yoshizawa K, Kitahara H, Yamamoto E.

Apoptosis-associated markers and clinical outcome in human oral

squamous cell carcinomas. J Oral Pathol Med. 2008;37:364–71.

13. Camisasca DR, Honorato J, Bernardo V, et al. Expression of

Bcl-2 family proteins and associated clinicopathologic factors

predict survival outcome in patients with oral squamous cell

carcinoma. Oral Oncol. 2009;45:225–33.

14. Gomes CC, Bernardes VF, Diniz MG, De Marco L, Gomez RC.

Anti-apoptotic gene transcription signature of salivary gland

neoplasms. BMC Cancer. 2012;12:61.

15. Sulkowska M, Famulski W, Chyczewski L, Sulkowski S.

Evaluation of p53 and bcl-2 oncoprotein expression in precan-

cerous lesions of the oral cavity. Neoplasma. 2001;48(2):94–8.

16. Kuropkat C, Venkatesan TK, Caldarelli DD, et al. Abnormalities

of molecular regulators of proliferation and apoptosis in carci-

noma of the oral cavity and oropharynx. Auris Nasus Larynx.

2002;29:165–74.

17. Yuen AP, Lam KY, Choy JT, Ho WK, Wong LY, Wei WI.

Clinicopathologic significance of bcl-2 expression in the surgi-

cal treatment of oral tongue carcinoma. Eur J Surg Oncol.

2002;28:667–72.

18. de Vicente JC, Olay S, Lequerica-Fernandez P, Sanchez-May-

oral J, Junquera LM, Fresno MF. Expression of Bcl-2 but not

Bax has a prognostic significance in tongue carcinoma. J Oral

Pathol Med. 2006;35:140–5.

19. Bose P, Klimowicz AC, Kornaga E, et al. Bax expression

measured by AQUAnalysis is an independent prognostic marker

in oral squamous cell carcinoma. BMC Cancer. 2012;12:332.

20. Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene,

survivin, expressed in cancer and lymphoma. Nat Med.

1997;3:917–21.

21. Muzio L, Staibano S, Pannone G, et al. Expression of the

apoptosis inhibitor survivin in aggressive squamous cell carci-

noma. Exp Mol Pathol. 2001;70:249–54.

22. De Maria S, Pannone G, Bufo P, Santoro A, Serpico R, Metafora

S, et al. Survivin gene-expression and splicing isoforms in oral

squamous cell carcinoma. J Cancer Res Clin Oncol.

2009;135:107–16.

23. Khan Z, Tiwari RP, Mulherkar R, Shah NK, Bisen PS. Detection

of survivin and p53 in human oral cancer: correlation with

clinicopathologic findings. Head Neck. 2009;31:1039–48.

24. Khan Z, Khan N, Tiwari RP, Patro IK, Prasad GB, Bisen PS.

Down-regulation of survivin by oxaliplatin diminishes radiore-

sistance of head and neck squamous carcinoma cells. Radiother

Oncol. 2010;96:267–73.

25. Su L, Wang Y, Xiao M, Lin Y, Yu L. Up-regulation of survivin

in oral squamous cell carcinoma correlates with poor prognosis

and chemoresistance. Oral Surg Oral Med Oral Pathol Oral

Radiol Endod. 2010;110:484–91.

26. Kim YH, Kim SM, Kim YK, Hong SP, Kim MJ, Myoung H.

Evaluation of survivin as a prognostic marker in oral squamous

cell carcinoma. J Oral Pathol Med. 2010;39:368–75.

27. Muzio LL, Pannone G, Staibano S, et al. Survivin expression in

oral squamous cell carcinoma. Br J Cancer. 2003;89:2244–8.

28. Engels K, Knauer SK, Metzler D, et al. Dynamic intracellular

survivin in oral squamous cell carcinoma: underlying molecular

mechanism and potential as an early prognostic marker.

J Pathol. 2007;211:532–40.

29. Lo Muzio L, Pannone G, Leonardi R, et al. Survivin, a potential

early predictor of tumor progression in the oral mucosa. J Dent

Res 2003;82:923–8.

30. Lin CY, Hung HC, Kuo RC, Chiang CP, Kuo MYP. Survivin

expression predicts poorer prognosis in patients with areca quid

chewing-related oral squamous cell carcinoma in Taiwan. Oral

Oncol. 2005;41:645–54.

31. Lo Muzio L, Farina A, Rubini C, et al. Survivin as prognostic

factor in squamous cell carcinoma of the oral cavity. Cancer

Lett. 2005;225:27–33.

32. Mineta H, Borg A, Dictor M, et al. p53 mutation, but not p53

overexpression, correlates with survival in head and neck

squamous cell carcinoma. Br J Cancer. 1998;78:1084–90.

33. Melino G, De Laurenzi V, Vousden KH. p73: friend or foe in

tumorigenesis. Nat Rev Cancer. 2002;2:605–15.

34. Khan Z, Bisen PS. Oncoapoptotic signaling and deregulated

target genes in cancers: Special reference to oral cancer. BBA

Rev Can. 2013;1836:123–45.

35. Perrone F, Bossi P, Cortelazzi B, et al. TP53 mutations and

pathologic complete response to neoadjuvant cisplatin and flu-

orouracil chemotherapy in resected oral cavity squamous cell

carcinoma. J Clin Oncol. 2010;28:761–6.

36. Raju B, Mehrotra R, Oijordsbakken G, Al-Sharabi AK, Vasstrand

EN, Ibrahim SO. Expression of p53, cyclin D1 and Ki-67 in pre-

malignant and malignant oral lesions: association with clinico-

pathological parameters. Anticancer Res. 2005;25:4699–706.

37. Smeets SJ, van der Plas M, Schaaij-Visser TB, et al. Immor-

talization of oral keratinocytes by functional inactivation of the

p53 and pRb pathways. Int J Cancer. 2011;128:1596–605.

38. Iamaroon A, Khemaleelakul U, Pongsiriwet S, Pintong J.

Coexpression of p53 and Ki67 and lack of EBV expression inoral squamous cell carcinoma. J Oral Pathol Med.

2004;33:30–6.

Prognostic and Therapeutic Potential of Apoptotic Markers in Oral Cancer

39. Shiraki M, Odajima T, Ikeda T, et al. Combined expression of

p53, cyclin D1 and epidermal growth factor receptor improves

estimation of prognosis in curatively resected oral cancer. Mod

Pathol. 2005;18:1482–9.

40. Boyle JO, Mao L, Brennan JA, et al. Gene mutations in saliva as

molecular markers for head and neck squamous cell carcinomas.

Am J Surg. 1994;168:429–32.

41. Scully C, Field JK, Tanzawa H. Genetic aberrations in oral or

head and neck squamous cell carcinoma 3: clinico-pathological

applications. Oral Oncol. 2000;36:404–13.

42. Angiero F, Berenzi A, Benetti A, et al. Expression of p16, p53

and Ki-67 proteins in the progression of epithelial dysplasia of

the oral cavity. Anticancer Res. 2008;28:2535–9.

43. Ralhan R, Nath N, Agarwal S, Mathur M, Wasylyk B, Shukla

NK. Circulating p53 antibodies as early markers of oral cancer:

correlation with p53 alterations. Clin Cancer Res.

1998;4:2147–52.

44. Yamazaki Y, Chiba I, Ishikawa M, et al. Serum p53 antibodies

as a prognostic indicator in oral squamous cell carcinoma.

Odontology. 2008;96:32–7.

45. Lopez M, Aguirre JM, Cuevas N, Anzola M, Videgain J, Agu-

irregaviria J. Use of cytological specimens for p53 gene alter-

ations detection in oral squamous cell carcinoma risk patients.

Clin Oncol. 2004;16:366–70.

46. Chen Q, Samaranayake LP, Zhen X, Luo G, Nie M, Li B. Up-

regulation of Fas ligand and down regulation of Fas expression

in oral carcinogenesis. Oral Oncol. 1999;35:548–53.

47. Das SN, Khare P, Singh MK, Sharma SC. Fas receptor (CD95)

& Fas ligand (CD178) expression in patients with tobacco-

related intraoral squamous cell carcinoma. Ind J Med Res.

2011;134:54–60.

48. Lee YB, Kyung Kim E, Park HJ, et al. Expression of Fas and

Fas ligand in primary cutaneous squamous cell carcinoma in

association with grade of tumor differentiation. Int J Dermatol.

2013;52:1092–7.

49. Hadzi-Mihailovic M, Raybaud H, Monteil R, Jankovic L.

Expression of Fas/FasL in patients with oral lichen planus.

J BUON. 2009;14:487–93.

50. Guler N, Uckan S, Celik I, Oznurlu Y, Uckan D. Expression of

Fas and Fas-ligand and analysis of argyrophilic nucleolar

organizer regions in squamous cell carcinoma: relationships

with tumor stage and grade, and apoptosis. Int J Oral Maxillofac

Surg. 2005;34:900–6.

51. Akagi H, Higuchi H, Sumimoto H, et al. Suppression of myeloid

cell leukemia-1 (Mcl-1) enhances chemotherapy-associated

apoptosis in gastric cancer cells. Gastric Cancer.

2013;16:100–10.

52. Mallick S, Agarwal J, Kannan S, Pawar S, Kane S, Teni T.

PCNA and anti-apoptotic Mcl-1 proteins predict disease-free

survival in oral cancer patients treated with definitive radio-

therapy. Oral Oncol. 2010;46:688–93.

53. Palve VC, Teni TR. Association of anti-apoptotic Mcl-1L iso-

form expression with radioresistance of oral squamous carci-

noma cells. Radiat Oncol. 2012;7:135.

54. Yu CH, Chen HM, Lin HP, Chiang CP. Expression of Bak and

Bak/Mcl-1 ratio can predict photodynamic therapy outcome for

oral verrucous hyperplasia and leukoplakia. J Oral Pathol Med.

2013;42:257–62.

55. Schiegnitz E, Kammerer PW, Koch FP, Kruger M, Berres M,

Al-Nawas B. GDF 15 as an anti-apoptotic, diagnostic and

prognostic marker in oral squamous cell carcinoma. Oral Oncol.

2012;48:608–14.

56. Coutinho-Camillo CM, Lourenco SV, Nishimoto IN, Kowalski

LP, Soares FA. Caspase expression in oral squamous cell car-

cinoma. Head Neck. 2011;33:1191–8.

57. Yang XH, Feng ZE, YanM, et al. XIAP is a predictor of cisplatin-

based chemotherapy response and prognosis for patients with

advanced head and neck cancer. PLoS One. 2012;7:e31601.

58. Tamatani T, Takamaru N, Uchida D, Nagai H, Fujisawa K,

Miyamoto Y. The expression of X-linked inhibitor of apoptosis

in human oral squamous cell carcinoma and its relationship with

clinical factors [abstract no. 4954]. In: Proceedings of the 103rd

Annual Meeting of the American Association for Cancer

Research; 2012 Mar 31–Apr 4; Chicago, IL. Philadelphia (PA):

AACR; Cancer Res 2012;72(8 Suppl 1). doi:10.1158/1538-

7445.AM2012-4954

59. Goto M, Tsukamoto T, Inada K, et al. Loss of p21WAF1/CIP1

expression in invasive fronts of oral tongue squamous cell car-

cinomas is correlated with tumor progression and poor prog-

nosis. Oncol Rep. 2005;14:837–46.

60. Nemes JA, Nemes Z, Marton IJ. p21WAF1/CIP1 expression is a

marker of poor prognosis in oral squamous cell carcinoma.

J Oral Pathol Med. 2005;34:274–9.

61. Xi S, Grandis JR. Gene therapy for the treatment of oral squa-

mous cell carcinoma. J Dent Res. 2003;82:11–6.

62. Lopez-Martinez M, Anzola M, Cuevas N, Aguirre JM, De-

Pancorbo M. Clinical applications of the diagnosis of p53

alterations in squamous cell carcinoma of the head and neck.

Med Oral. 2002;7:108–20.

63. Liu TJ, Zhang WW, Taylor DL, Roth JA, Goepfert H, Clayman

GL. Growth suppression of human head and neck cancer cells

by the introduction of a wild-type p53 gene via a recombinant

adenovirus. Cancer Res. 1994;54:3662–7.

64. Clayman GL, el-Naggar AK, Lippman SM, et al. Adenovirus-

mediated p53 gene transfer in patients with advanced recurrent

head and neck squamous cell carcinoma. J Clin Oncol.

1998;16:2221–32.

65. Rudin CM, Cohen EE, Papadimitrakopoulou VA, et al. An

attenuated adenovirus, ONYX-015, as mouthwash therapy for

premalignant oral dysplasia. J Clin Oncol. 2003;21:4546–52.

66. Heise C, Kirn DH. Replication-selective adenoviruses as onco-

lytic agents. J Clin Invest. 2000;105:847–51.

67. Nemunaitis J, Cunningham C, Tong AW, et al. Pilot trial of

intravenous infusion of a replication-selective adenovirus

(ONYX-015) in combination with chemotherapy or IL-2 treat-

ment in refractory cancer patients. Cancer Gene Ther.

2003;10:341–52.

68. Li G, Tan X-H. Inhibitory effect on growth of oral cancer cell

lines KB induced by survivin RNAi. China Res Prev Treat.

2011;38:257–60.

69. Khan Z, Tiwari RP, Khan N, Prasad GB, Bisen PS. Induction of

apoptosis and sensitization of head and neck squamous carci-

noma cells to cisplatin by targeting survivin gene expression.

Curr Gene Ther. 2012;12:444–53.

70. Nagata M, Wada K, Nakajima A, et al. Role of myeloid cell

leukemia-1 in cell growth of squamous cell carcinoma. J Phar-

macol Sci. 2009;110:344–53.

71. Leech SH, Olie RA, Gautschi O, et al. Induction of apoptosis in

lung-cancer cells following BCL-XL antisense treatment. Int J

Cancer. 2000;86:570–6.

72. Simoes-Wust AP, Olie RA, Gautschi O, et al. BCL-XL antisense

treatment induces apoptosis in breast carcinoma cells. Int J

Cancer. 2000;87:582–90.

73. Gibson SA, Pellenz C, Hutchison RE, Davey FR, Shillitoe EJ.

Induction of apoptosis in oral cancer cells by an anti-bcl-2

ribozyme delivered by an adenovirus vector. Clin Cancer Res.

2000;6:213–22.

74. Kaur J, Kaur J, Ralhan R. Induction of apoptosis by abrogation

of HSP70 expression in human oral cancer cells. Int J Cancer.

2000;85:1–5.

A. Jain et al.

75. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus ce-

tuximab for locoregionally advanced head and neck cancer:

5-year survival data from a phase 3 randomised trial, and rela-

tion between cetuximab-induced rash and survival. Lancet

Oncol. 2010;11:21–8.

76. Judd NP, Winkler AE, Murillo-Sauca O, et al. ERK1/2 regula-

tion of CD44 modulates oral cancer aggressiveness. Cancer Res.

2012;72:365–74.

77. Kok SH, Yeh CC, Chen ML, Kuo MY. Esculetin enhances

TRAIL-induced apoptosis through DR5 upregulation in human

oral cancer SAS cells. Oral Oncol. 2009;45:1067–72.

78. Finnberg N, El-Deiry WS. Selective TRAIL-induced apoptosis

in dysplastic neoplasia of the colon may lead to new neoadju-

vant or adjuvant therapies. Clin Cancer Res. 2006;12:4132–6.

79. Rowinsky EK. Targeted induction of apoptosis in cancer man-

agement: the emerging role of tumor necrosis factor-related

apoptosis-inducing ligand receptor activating agents. J Clin

Oncol. 2005;23:9394–407.

80. Fukuda M, Hamao A, Tanaka A, et al. Tumor necrosis factor-

related apoptosis-inducing ligand (TRAIL/APO2L) and its

receptors expression in human squamous cell carcinoma of the

oral cavity. Oncol Rep. 2003;10:1113–9.

81. Teng MS, Brandwein-Gensler MS, Teixeira MS, Martignetti JA,

Duffey DC. A study of TRAIL receptors in squamous cell

carcinoma of the head and neck. Arch Otolaryngol Head Neck

Surg. 2005;131:407–12.

82. Nakashima T, Miura M, Hara M. Tetrocarcin A inhibits mito-

chondrial functions of Bcl-2 and suppresses its anti-apoptotic

activity. Cancer Res. 2000;60:1229–35.

83. Tinhofer I, Anether G, Senfter M, et al. Stressful death of

T-ALL tumor cells after treatment with the anti-tumor agent

Tetrocarcin-A. FASEB J. 2002;16:1295–7.

84. Tzung SP, Kim KM, Basanez G, et al. Antimycin A mimics a

cell-death-inducing Bcl-2 homology domain 3. Nat Cell Biol.

2001;3:183–91.

85. Chan SL, LeeMC, TanKO, et al. Identification of chelerythrine as

an inhibitor of BclXL function. J Biol Chem. 2003;278:20453–6.

86. Kang MH, Reynolds CP. Bcl-2 inhibitors: targeting mitochon-

drial apoptotic pathways in cancer therapy. Clin Cancer Res.

2009;15:1126–32.

87. Nguyen JT, Wells JA. Direct activation of the apoptosis

machinery as a mechanism to target cancer cells. Proc Natl Acad

Sci USA. 2003;100:7533–8.

88. Zhang HZ, Kasibhatla S, Wang Y, et al. Discovery, character-

ization and SAR of gambogic acid as a potent apoptosis inducer

by a HTS assay. Bioorg Med Chem. 2004;12:309–17.

89. Matsumiya T, Imaizumi T, Yoshida H, Kimura H, Satoh K.

Cisplatin inhibits the expression of X-chromosome-linked

inhibitor of apoptosis protein in an oral carcinoma cell line. Oral

Oncol. 2001;37:296–300.

90. Midgley CA, Desterro JM, Saville MK, et al. An N-terminal

p14ARF peptide blocks Mdm2-dependent ubiquitination in vitro

and can activate p53 in vivo. Oncogene. 2000;19:2312–23.

91. Chene P, Fuchs J, Bohn J, et al. A small synthetic peptide, which

inhibits the p53-hdm2 interaction, stimulates the p53 pathway in

tumour cell lines. J Mol Biol. 2000;299:245–53.

92. Grasberger BL, Lu T, Schubert C, et al. Discovery and cocrystal

structure of benzodiazepinedione HDM2 antagonists that acti-

vate p53 in cells. J Med Chem. 2005;48:909–12.

93. Issaeva N, Bozko P, Enge M, et al. Small molecule RITA binds

to p53, blocks p53-HDM-2 interaction and activates p53 func-

tion in tumors. Nat Med. 2004;10:1321–8.

94. Vassilev LT, Vu BT, Graves B, et al. In vivo activation of the

p53 pathway by small-molecule antagonists of MDM2. Science.

2004;303:844–8.

95. Shangary S, Qin D, McEachern D, et al. Temporal activation of

p53 by a specific MDM2 inhibitor is selectively toxic to tumors

and leads to complete tumor growth inhibition. Proc Natl Acad

Sci USA. 2008;105:3933–8.

96. Yang Y, Ludwig RL, Jensen JP, et al. Small molecule inhibitors

of HDM2 ubiquitin ligase activity stabilize and activate p53 in

cells. Cancer Cell. 2005;7:547–59.

97. Lain S, Hollick JJ, Campbell J, et al. Discovery, in vivo activity,

and mechanism of action of a small-molecule p53 activator.

Cancer Cell. 2008;13:454–63.

98. Staples OD, Hollick JJ, Campbell J, et al. Characterization,

chemical optimization and anti-tumour activity of a tubulin

poison identified by a p53-based phenotypic screen. Cell Cycle.

2008;7:3417–27.

99. Selivanova G, Iotsova V, Okan I, et al. Restoration of the growth

suppression function of mutant p53 by a synthetic peptide

derived from the p53 C-terminal domain. Nat Med.

1997;3:632–8.

100. Friedler A, Hansson LO, Veprintsev DB, et al. A peptide that

binds and stabilizes p53 core domain: chaperone strategy for

rescue of oncogenic mutants. Proc Natl Acad Sci USA.

2002;99:937–42.

101. Issaeva N, Friedler A, Bozko P, et al. Rescue of mutants of the

tumor suppressor p53 in cancer cells by a designed peptide. Proc

Natl Acad Sci USA. 2003;100:13303–7.

102. Bykov VJ, Issaeva N, Shilov A, et al. Restoration of the tumor

suppressor function to mutant p53 by a low-molecular weight

compound. Nat Med. 2002;8:282–8.

103. Foster BA, Coffey HA, Morin MJ, Rastinejad F. Pharmaco-

logical rescue of mutant p53 conformation and function. Sci-

ence. 1999;286:2507–10.

104. Wang W, Takimoto R, Rastinejad F, El-Deiry WS. Stabilization

of p53 by CP-31398 inhibits ubiquitination without altering

phosphorylation at serine 15 or 20 or MDM2 binding. Mol Cell

Biol. 2003;23:2171–81.

105. Luu Y, Bush J, Cheung KJ Jr, Li G. The p53 stabilizing com-

pound CP-31398 induces apoptosis by activating the intrinsic

Bax/mitochondrial/caspase-9 pathway. Exp Cell Res.

2002;276:214–22.

106. Takimoto R, Wang W, Dicker DT, et al. The mutant p53-con-

formation modifying drug, CP-31398, can induce apoptosis of

human cancer cells and can stabilize wild-type p53 protein.

Cancer Biol Ther. 2002;1:47–55.

107. Bykov VJ, Issaeva N, Zache N, et al. Reactivation of mutant p53

and induction of apoptosis in human tumor cells by maleimide

analogs. J Biol Chem. 2005;280:30384–91.

108. Xu GW, Mawji IA, Macrae CJ, et al. A high-content chemical

screen identifies ellipticine as a modulator of p53 nuclear

localization. Apoptosis. 2008;13:413–22.

109. Weinmann L, Wischhusen J, Demma MJ, et al. A novel p53

rescue compound induces p53-dependent growth arrest and

sensitises glioma cells to Apo2L/TRAIL-induced apoptosis. Cell

Death Differ. 2008;15:718–29.

110. North S, Pluquet O, Maurici D, El-Ghissassi F, Hainaut P.

Restoration of wild-type conformation and activity of a tem-

perature-sensitive mutant of p53 (p53(V272M)) by the cyto-

protective aminothiol WR1065 in the esophageal cancer cell line

TE-1. Mol Carcinog. 2002;33:181–8.

111. Nakahara T, Takeuchi M, Kinoyama I, et al. YM155, a novel

small-molecule survivin suppressant, induces regression of

established human hormone-refractory prostate tumor xeno-

grafts. Cancer Res. 2007;67:8014–21.

112. Kumar B, Yadav A, Lang JC, et al. YM155 reverses cisplatin

resistance in head and neck cancer by decreasing cytoplasmic

survivin levels. Mol Cancer Ther. 2012;11:1988–98.

Prognostic and Therapeutic Potential of Apoptotic Markers in Oral Cancer

113. Zhang M, Zhu W, Ding N, Zhang W, Li Y. Identification and

characterization of small molecule inhibitors of signal trans-

ducer and activator of transcription 3 (STAT3) signaling path-

way by virtual screening. Bioorg Med Chem Lett.

2013;23:2225–9.

114. Zhang X, Sun Y, Pireddu R, et al. A novel inhibitor of STAT3

homodimerization selectively suppresses STAT3 activity and

malignant transformation. Cancer Res. 2013;73:1922–33.

115. Feng R, Zhou S, Liu Y, et al. Sox2 protects neural stem cells

from apoptosis via up-regulating survivin expression. Biochem

J. 2013;450:459–68.

116. Guha M, Xia F, Raskett CM, Altieri DC. Caspase 2-mediated

tumor suppression involves survivin gene silencing. Oncogene.

2010;29:1280–92.

117. Carrasco RA, Stamm NB, Marcusson E, Sandusky G, Iversen P,

Patel BK. Antisense inhibition of survivin expression as a cancer

therapeutic. Mol Cancer Ther. 2011;10:221–32.

118. Sapra P, Wang M, Bandaru R, Zhao H, Greenberger LM, Horak

ID. Down-modulation of survivin expression and inhibition of

tumor growth in vivo by EZN-3042, a locked nucleic acid

antisense oligonucleotide. Nucleosides Nucleotides Nucleic

Acids. 2010;29:97–112.

119. Hendruschk S, Wiedemuth R, Aigner A, et al. RNA interference

targeting survivin exerts antitumoral effects in vitro and in

established glioma xenografts in vivo. Neuro Oncol.

2011;13:1074–89.

120. Liu X, Gao R, Dong Y, et al. Survivin gene silencing sensitizes

prostate cancer cells to selenium growth inhibition. BMC Can-

cer. 2010;10:418.

121. Montazeri Aliabadi H, Landry B, Mahdipoor P, Uludag H.

Induction of apoptosis by survivin silencing through siRNA

delivery in a human breast cancer cell line. Mol Pharm.

2011;8:1821–30.

122. Cao W, Fan R, Wang L, et al. Expression and regulatory

function of miRNA-34a in targeting survivin in gastric cancer

cells. Tumour Biol. 2013;34:963–71.

123. Kang BH, Plescia J, Dohi T, Rosa J, Doxsey SJ, Altieri DC.

Regulation of tumor cell mitochondrial homeostasis by an

organelle-specific Hsp90 chaperone network. Cell.

2007;131:257–70.

124. Reuschenbach M, von Knebel Doeberitz M, Wentzensen N. A

systematic review of humoral immune responses against tumor

antigens. Cancer Immunol Immunother. 2009;58:1535–44.

125. Widenmeyer M, Griesemann H, Stevanovic S, et al. Promiscu-

ous survivin peptide induces robust CD4?T-cell responses in

the majority of vaccinated cancer patients. Int J Cancer.

2012;131:140–9.

126. Becker JC, Andersen MH, Hofmeister-Muller V, et al. Survivin-

specific T-cell reactivity correlates with tumor response and

patient survival: a phase-II peptide vaccination trial in metastatic

melanoma. Cancer Immunol Immunother. 2012;61:2091–103.

127. Weiss A, Brill B, Borghouts C, Delis N, Mack L, Groner B.

Survivin inhibition by an interacting recombinant peptide,

derived from the human ferritin heavy chain, impedes tumor cell

growth. J Cancer Res Clin Oncol. 2012;138:1205–20.

128. Rosato R, Almenara J, Kolla S, et al. Mechanism and functional

role of XIAP and Mcl-1 down-regulation in flavopiridol/vori-

nostat antileukemic interactions. Mol Cancer Ther. 2007;6:692.

129. Chen R, Wierda W, Chubb S, et al. Mechanism of action of

SNS-032, a novel cyclin-dependent kinase inhibitor, in chronic

lymphocytic leukemia. Blood. 2009;113:4637.

130. Rahmani M, Davis E, Bauer C, Dent P, Grant S. Apoptosis

induced by the kinase inhibitor BAY 43-9006 in human leuke-

mia cells involves down-regulation of Mcl-1 through inhibition

of translation. J Biol Chem. 2005;280:35217.

131. Yu C, Bruzek L, Meng X, et al. The role of Mcl-1 downregu-

lation in the proapoptotic activity of the multikinase inhibitor

BAY 43–9006. Oncogene. 2005;24:6861.

132. Rahmani M, Davis E, Crabtree T, et al. The kinase inhibitor

sorafenib induces cell death through a process involving

induction of endoplasmic reticulum stress. Mol Cell Biol.

2007;27:5499.

133. Schwickart M, Huang X, Lill J, et al. Deubiquitinase USP9X

stabilizes MCL1 and promotes tumour cell survival. Nature.

2010;463:103.

134. Sun H, Kapuria V, Peterson L, et al. Bcr-abl ubiquitination and

Usp9x inhibition block kinase signaling and promote CML cell

apoptosis. Blood. 2011;117:3151.

135. Chen S, Dai Y, Harada H, Dent P, Grant S. Mcl-1 down-regu-

lation potentiates ABT-737 lethality by cooperatively inducing

Bak activation and Bax translocation. Cancer Res. 2007;67:782.

136. Nguyen M, Marcellus R, Roulston A, et al. Small molecule

obatoclax (GX15-070) antagonizes MCL-1 and overcomes

MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci

USA. 2007;104:19512.

137. Wei J, Kitada S, Rega M, et al. Apogossypol derivatives as

antagonists of antiapoptotic Bcl-2 family proteins. Mol Cancer

Ther. 2009;8:904.

138. Wei J, Stebbins J, Kitada S, et al. BI-97C1, an optically pure

apogossypol derivative as pan-active inhibitor of antiapoptotic

B-cell lymphoma/leukemia-2 (Bcl-2) family proteins. J Med

Chem. 2010;53:4166.

139. Kazi A, Sun J, Doi K, et al. The BH3 alpha-helical mimic BH3-

M6 disrupts bcl-X(L), bcl-2, and MCL-1 protein-protein inter-

actions with Bax, Bak, Bad, or Bim and induces apoptosis in a

Bax-and Bim-dependent manner. J Biol Chem. 2011;286:

9382–92.

A. Jain et al.