Review

Caspase-9 as a therapeutic target for treating cancer Bonglee Kim, Sanjay K Srivastava & Sung-Hoon Kim† †

1.

Introduction

2.

Regulators of caspase-9

3.

Caspase-9 activation by natural

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

compounds 4.

Clinical trials for caspase-9

5.

Conclusion

6.

Expert opinion

Kyunghee University, College of Korean Medicine, Cancer Preventive Material Development Research Center, Seoul, South Korea

Introduction: Caspase-9 is the apoptotic initiator protease of the intrinsic or mitochondrial apoptotic pathway, which is activated at multi-protein activation platforms. Its activation is believed to involve homo-dimerization of the monomeric zymogens. It binds to the apoptosome to retain substantial catalytic activity. Variety of apoptotic stimuli can regulate caspase-9. However, the mechanism of action of various regulators of caspase-9 has not been summarized and compared yet. In this article, we elucidate the regulators of caspase-9 including microRNAs, natural compounds that are related to caspase-9 and ongoing clinical trials with caspase-9 to better understand the caspase-9 in suppressing cancer. Areas covered: In this study, the basic mechanism of apoptosis pathways, regulators of caspase-9 and the development of drugs to regulate caspase-9 are reviewed. Also, ongoing clinical trials for caspase-9 are discussed. Expert opinion: Apoptosis has crucial role in cancer, brain disease, aging and heart disease to name a few. Since caspase-9 is an initiator caspase of apoptosis, it is an important therapeutic target of various diseases related to apoptosis. Therefore, a deep understanding on the roles as well as regulators of caspase-9 is required to find more effective ways to conquer apoptosis-related diseases especially cancer. Keywords: apoptosis, apoptosis-activating factor-1, apoptosome, cancer, caspase-3, caspase-9 Expert Opin. Ther. Targets (2015) 19(1):113-127

1.

Introduction

Apoptosis is a programmed cell death, which plays critical roles in development, immune system and homeostasis of a multicellular organism [1]. Failure of this process can cause serious diseases such as cancer, Alzheimer’s disease, stroke, viral infection or autoimmune diseases [2-6]. The proteins responsible for inducing apoptosis could be targeted therapeutically to overcome resistance to apoptosis. Therefore, understanding the mechanism of apoptosis is critical for a successful therapy. Caspases are cysteine-dependent aspartate-specific proteases, involved in the process of apoptosis. Caspases can be divided into two groups; initiator caspases, such as caspase-2, -8, -9 and -10, and effector caspases, such as caspase-3 and -7 [1,7]. Caspase-9 activity is enhanced by phosphorylation at Tyr153 [8] and inhibited by phosphorylation at Thr125 [9-12], Ser196 [13], Ser144 [14] and Ser348 [15]. Unlike effector caspases, caspase-9 does not need to be cleaved, but just dimerized to be activated. Two different pathways exist in apoptosis process, extrinsic and intrinsic pathway. The binding of death receptors with their respective ligands results in the sequential activation of caspase-8 and -3 in extrinsic apoptosis. This pathway is downregulated by cFLIP and human X-chromosome-linked inhibitor of apoptosis protein (XIAP). DNA-damaging agents activate mitochondrial or intrinsic pathway by inducing the release of the cytochrome c. Cytochrome c, a peripheral protein of 10.1517/14728222.2014.961425 © 2015 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 All rights reserved: reproduction in whole or in part not permitted

113

B. Kim et al.

Article highlights. . .

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

.

.

. .

Caspase-9 participates in apoptosis process as initiator caspase. In the process of intrinsic apoptosis pathway, cytochrome c is released in cytosol from mitochondria, forms apoptosome with apoptosis-activating factor-1 and caspase-9, activating caspase-9. Caspase-9 is regulated by several signal pathways such as hepatocellular carcinoma antigen 66, histone H1.2, putative HLA-DR-associated protein, nucleotide-binding domain and caspase recruitment domain, death effector filament-forming Ced-4-like apoptosis protein, Nucling, Akt, Apaf-1-interacting protein, c-Abl (an ubiquitous nonreceptor tyrosine kinase), CDK1/cyclin B1, casein kinase 2, dual-specificity tyrosine-phosphorylationregulated protein kinase, MAPK, hepatitis B X-interacting protein, heat shock protein 70, protein kinase A, PKC, tumor-upregulated caspase-associated recruitment domain (CARD)-containing antagonist of caspase nine, human X-chromosome-linked inhibitor of apoptosis protein and microRNAs. A large number of natural compounds were reported to enhance caspase-9 and suppress cancer by inducing apoptosis. Two clinical trials with caspase-9 as a suicide gene for T cells against leukemia are undergoing. Future research should focus on finding safer and more effective drugs, based on a better understanding of caspase-9.

This box summarizes key points contained in the article.

recruitment domain (NAC), death effector filament-forming Ced-4-like apoptosis protein (DEFCAP), Nucling and inhibited by Akt, Apaf-1-interacting protein (APIP), c-Abl (an ubiquitous nonreceptor tyrosine kinase), cyclin-dependent kinases 1(CDK1)-cyclin B1, casein kinase 2 (CK2), dualspecificity tyrosine-phosphorylation-regulated protein kinase (DYRK1A), extracellular signal-regulated kinase (ERK)2, hepatitis B X-interacting protein (HBXIP), heat shock protein 70 (Hsp70), protein kinase A (PKA), protein kinase C (PKC), p38a, tumor-upregulated caspase-associated recruitment domain (CARD)-containing antagonist of caspase nine (TUCAN), XIAP and miRNA-24a, -133, -585, -23a (Table 1), (Figure 1). HCA66 Microdeletions of HCA66 around NF1 gene are frequently associated with a severe form of neurofibromatosis type I microdeletion syndrome in patients. The patients usually show a more severe clinical phenotype, including frequent tumor incidence than other NF1 patients [34]. HCA66 mRNA is expressed in most human tissues but expressed highly in heart, liver and placenta [35]. Piddubnyak et al. reported that HCA66 significantly increased the interaction of caspase-9 with Apaf-1 in 293 T cells. On the other hand, depletion of HCA66 inhibited recruitment of caspase-9 by Apaf-1.The N-terminus of HCA66 showed inhibitory effect on caspase9-related cell death [35]. 2.1

Histone H1.2 Mass spectrometry analysis showed that histone H1, H2A and H2b have regulatory effects on caspases. Ruiz-Vela et al. developed a novel nonhypotonic cell-free system based on the zwitterionic detergent CHAPS to further study its interaction. As a result, pro-apoptotic histone H1.2 but not H1.0, H1.3, H1.4 or H1.5 induced activation of caspase-3,-7 and -9. Also, histone H1.2 interacts with Apaf-1, caspase-9, caspase-3 and cytochrome c after UV irradiation [36]. 2.2

the mitochondrial inner membrane, functions as an electron shuttle [16]. Cytochrome c once released in the cytosol forms apoptosome with apoptosis-activating factor-1 (Apaf-1) and procaspase-9 [17]. Procaspase-9 is activated by apoptosome, which in turn activates procaspase-3 to caspase-3 leading to apoptosis. Caspase-9 is a key caspase in intrinsic apoptosis pathway. Mutation in caspase-9 gene results in embryonic lethality and impaired brain development [18]. Like other initiator caspases, procaspase-9 is an inactive monomer (zymogen) at physiological conditions [19]. Apoptosome binds to multiple procaspase-9 and promotes the dimerization of caspase-9 resulting in its activation [20]. In other words, apoptosome serves as a recruitment platform that promotes procaspase-9 activation [7]. A number of epidemiological and experimental studies have reported caspase-9-mediated apoptosis by natural compounds, including isothiocyanates [21-23], emodin [24,25], tanshinone IIA [26-29], ursolic acid [30] and decursin [31-33].

Putative HLA-DR-associated protein A small molecule, PETCM (a-(trichloromethyl)-4-pyridineethanol) is known as apoptosome formation enhancer and caspase-3 activator. Jiang et al. showed that there are Q-ft, Q30 and Q100 proteins in HeLa cell extracts. These three proteins were identified by chromatography from Q100, PHAPI, PHAPI2a and PHAPIII. PHAP did not affect the efficiency of apoptosome formation but enhance caspase-9 activation after apoptosome formation in mammalian cells [37]. 2.3

Nucleotide-binding domain and caspase recruitment domain

2.4

2.

Regulators of caspase-9

Caspase-9 is regulated by many factors. It has been reported that caspase-9 is activated by hepatocellular carcinoma antigen 66 (HCA66), histone H1.2, putative HLA-DR-associated protein (PHAP), nucleotide-binding domain and caspase 114

CED4 family proteins play role in programmed cell death [38]. CED3 binds to oligomerized CED4, exerting protease activation [39]. According to Chu et al., NAC is expressed in kidney, brain and epidermis. Overexpressed NAC showed synergistic effect with Apaf-1 and pro-caspase-9 in the activation of

Expert Opin. Ther. Targets (2015) 19(1)

Role of caspase-9 in apoptosis

Table 1. Regulators of caspase-9.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

Regulator

Efficacy

Mechanism

System

HCA66

Activation or inhibition

In vitro

HEK293T, HeLa, MCF-7

[35]

Histone H1.2

Activation

In vitro

MEFs

[36]

PHAP

Activation

In vitro

HeLa

[37]

NAC

Activation

In vitro

HEK293T, human tissue

[40]

DEFCAP Akt

Activation Inhibition

In vitro In vitro

HEK293, K562, MCF-7 HEK293T, 267B, DLD-1

[41] [13]

APIP c-Abl

Inhibition Inhibition

In vitro In vitro

C2C12, HEK293 U-937

[51] [8]

CDK1/ Cyclin B1 CK2

Inhibition

In vitro

HeLa

[9]

In vitro

MEFs, FL5.12

[15]

DYRK1A

Inhibition

In vitro

HeLa, U2OS

[12,64]

ERK

Inhibition Inhibition

In vitro, In vivo In vitro

HeLa, U2OS, Xenopus egg extract Jurkat

[10,11,154]

HBXIP Hsp70

Inhibition

In vitro

Jurkat, HEK293T

[71,72]

PKA

Inhibition

In vitro

U2OS, HeLa, HEK293

[76]

PKC

Inhibition

In vitro

U2OS, HeLa, HEK293

[14]

TUCAN

Inhibition

In vitro

Jurkat, HEK293T, MCF7

[79]

XIAP

Inhibition

Increases casp-9 recruitment to the apoptosome or blocks casp-9 recruitment Enhances association of casp-9 and apoptosome Enhances casp-9 activity after apoptosome formation Increases casp-9 recruitment to the apoptosome Enhances casp-9 activity Inhibits casp-9 by phosphorylation at Ser196 Inhibits casp-9 binding to Apaf-1 Inhibits casp-9 by phosphorylation at Tyr153 Inhibits casp-9 by phosphorylation at Thr125 Inhibits casp-9 by phosphorylation at Ser348 Inhibits casp-9 by phosphorylation at Thr125 Inhibits casp-9 by phosphorylation at Thr125 Binds to survivin, inhibiting recruitment of casp-9 to Apaf-1 Prevents oligomerization of Apaf-1 or recruitment of casp-9 to Apaf-1 Prevents recruitment of casp-9 to Apaf-1 Inhibits casp-9 by phosphorylation at Ser144 Binds to procasp-9 and suppresses its activation Inhibits casp-9 activity by blocking homo-dimerization

In vitro

Escherichia coli Bl21, HEK293, Jurkat

[83] [84]

Inhibition

Cell lines

Ref.

[69]

DEFCAP: Death effector filament-forming Ced-4-like apoptosis protein; ERK: Extracellular signal-regulated kinase; HBXIP: Hepatitis B X-interacting protein; Hsp70: Heat shock protein 70; NAC: Nucleotide-binding domain and caspase recruitment domain; PHAP: Putative HLA-DR associated protein-1; PKA: Protein kinase A; PKC: Protein kinase C; TUCAN: Tumor-up-regulated CARD-containing antagonist of caspase nine; XIAP: Human X-chromosome-linked inhibitor of apoptosis protein.

caspases and induction of apoptosis. Association of NAC and Apaf-1increased recruitment and proteolysis process of pro-caspase-9 after cytochrome c stimulation [40]. Death effector filament-forming Ced-4-like apoptosis protein

2.5

DEFCAP is a member of the mammalian Ced-4 family of apoptotic proteins. The mammalian Ced-4 proteins (Apaf-1, Nod1 and DEFCAP) contain CARD. Human DEFCAP mRNA is expressed in liver, spleen and K562, a chronic myelogenous leukemia cell line. DEFCAP has two isoforms, DEFCAP-L and DEFCAP-S. Overexpression of DEFCAPL, but not DEFCAPS-S, induced apoptosis in MCF-7, a breast cancer cell line. Hlaing et al. indicated that DEFCAP may play a role in targeting caspase-9 but the mechanism remains unclear [41].

Nucling Nucling is a highly expressed protein during cardiac muscle differentiation and isolated from murine embryonal carcinoma cells [42]. Nucling has inhibitory effects on galectin-3, which blocks apoptosis process [43]. A bovine homolog of Nucling, b CAP73 is b-actin-specific binding protein, regulates b-actin assembly [44]. Sakai et al. reported that Nucling was expressed in an Apaf-1/pro-caspase-9 complex after UV irradiation. Nucling has role in apoptosis, specifically, stabilizes apoptosome and is required for the translocation of Apaf-1 to the nucleus [45]. 2.6

Akt Akt, a serine threonine protein kinase B, plays a critical role in malignant transformation and subsequent processes of growth, proliferation and metastases [46]. It is reported that 2.7

Expert Opin. Ther. Targets (2015) 19(1)

115

B. Kim et al.

P Ser196

P Tyr153 P Thr125 P Ser348 P Thr125 P Thr125

HCA66 Histone H1.2

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

PHAP NAC DEFCAP

Akt APIP

HBXIP

C-Abl

Hsp70

CDK1/Cyclin B1

PKA

CK2

PKC

DYRK1A

TUCAN

ERK

XIAP

P Ser144 miR-133 miR-24a miR-582-5p miR-23a

Caspase-9

Cytochrome c Apaf-1

Caspase-3

Apoptosis

Figure 1. Regulation of the caspase-9. APIP: Apaf-1-interacting protein; DEFCAP: Death effector filament-forming Ced-4-like apoptosis protein; ERK: Extracellular signal-regulated kinase; HBXIP: Hepatitis B X-interacting protein; Hsp70: Heat shock protein 70; NAC: Nucleotide-binding domain and caspase recruitment domain; PKA: Protein kinase A; PKC: Protein kinase C; PHAP: Putative HLA-DR-associated protein; TUCAN: Tumor-upregulated caspase-associated recruitment domain (CARD)-containing antagonist of caspase nine; XIAP: Human X-chromosome-linked inhibitor of apoptosis protein.

activation of Akt especially if PTEN expression is lost, is associated with a poor 5-year survival in several cancers [47]. Akt inhibits cytochrome c induced cleavage of pro-caspase-9. Akt phosphorylates pro-caspase-9 at Ser196 and inactivates it in vitro and in vivo but not any other caspases such as caspase-3 or caspase-8 [13].

phosphorylates caspase-9 at Tyr-153, attenuating the activity of caspase-9-induced apoptosis. Conversely, when c-Abl is inhibited by STI571, the process of caspase-9 phosphorylation is attenuated, indicating that caspase-9 is downregulated by c-Abl. [8]. CDK1/cyclin B1 The cell cycle is one of the most significant mechanism in growth and development, and its deregulation in many human disorders. Researchers have greatly expanded knowledge on the cell cycle and have contributed to the universally accepted view of how the basic cell cycle machinery is regulated [56]. CDK1 is one of the family of enzymes essential for the progression of the cells. Moreover, checkpoint control and DNA repair, requires the phosphorylation of a wide variety of target substrates by CDK [57]. Studies have shown that caspase-9 is regulated by CDK1/cyclin B1 by phosphorylation at Thr125, a single major inhibitory site during mitosis [9]. 2.10

2.8

Apaf-1-interacting protein

APIP has been reported to inhibit two main types of programmed cell death, caspase-1-dependent pyroptosis and caspase-9-dependent apoptosis [48,49]. Also, APIP induces the activation of Akt and ERK1/2 [50]. Cho et al. revealed that APIP acts as a negative regulator of ischemic injury. APIP binds competitively to CARD of Apaf-1 with caspase-9. It also inhibits apoptosis process by suppressing the downstream signal of cytochrome c [51]. c-Abl c-Abl plays role in cell proliferation, differentiation, apoptosis and cell adhesion [52,53]. In response to DNA damage, c-Abl is activated and phosphorylates caspase-9 at Tyr-153 [54,55]. According to Raina et al., c-Abl binds directly to caspase-9, 2.9

116

Casein kinase 2 Casein kinase 2 (protein kinase, CK2) is a eukaryotic serine/ threonine kinase with multiple substrates and affects several 2.11

Expert Opin. Ther. Targets (2015) 19(1)

Role of caspase-9 in apoptosis

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

signaling pathways in differentiation, proliferation, stress response, DNA damage, circadian rhythm and apoptosis [58,59]. It is overexpressed in many cancer cells and protects cancer cells from apoptosis [60]. Unlike other protein kinases, constitutively active CK2 has two catalytic subunits, a and a¢, and two regulatory b units [61]. McDonnell et al. revealed that CK2 phosphorylates murine caspase-9 at Ser348 in vitro and interferes with its cleavage at Asp353 by caspase-8 during intrinsic apoptosis [15]. 2.12 Dual-specificity tyrosine-phosphorylationregulated protein kinase 1A

The dual-specificity tyrosine-phosphorylation-regulated protein kinase (DYRK) family comprises at least seven mammalian isoforms (DYRK1A, DYRK1B, DYRK1C, DYRK2, DYRK3, DYRK4A and DYRK4B), the yeast homolog Yak1p and the Drosophila kinase minibreak [62]. The human gene for DYRK1A is a candidate gene for mental retardation in Down’s syndrome [63]. Seifert et al. revealed that DYRKS1A plays role in inhibitory phosphorylation of caspase-9 at Thr125 and involves its co-localization in the nucleus [12,64]. MAPK All eukaryotic cells have multiple MAPK pathways, which coordinately regulate diverse cellular activities running the gamut from gene expression, mitosis and metabolism to motility, survival, apoptosis and differentiation. It is reported that five distinct groups of MAPKs have been characterized in mammals: extracellular signal-regulated kinases (ERKs) 1 and 2 (ERK1/2), JNKs-1, 2 and 3, p38 isoforms a, b, g, and d, ERKs 3 and 4, and ERK5 [65]. The ERKs are most commonly activated by mitogenes, whereas JNK and p38MAPKs are responsive to stress and inflammatory signals [66]. Allan et al. and Martin et al. showed that inhibition of caspase-9 activity due to phosphorylation at Thr125 was mediated by ERK1/2 and not JNK or p38a/b [10,11]. Also, activated purified wildtype ERK protein showed inhibition of post-cytochrome c induced apoptosis in meiotic X. Laevis egg extracts. 2.13

2.14

Hepatitis B X-interacting protein

HBXIP was found as interactor of HBX protein of hepatitis B virus [67]. HBX is a candidate of oncogenic protein implicated in apoptosis [68]. Survivin, anti-apoptotic protein, is highly expressed in many cancers [3]. Survivin and HBXIP make a complex, bind to pro-caspase-9 and inhibit its recruitment to Apaf-1. Hence HBXIP acts as cofactor of survivin in suppression of apoptosis process [69]. Heat shock protein 70 The high expression of inducible Hsp70 is known to correlate with poor prognosis in many cancers. Hsp70 plays role in survival as well as resistance to chemotherapeutic agents and promotes tumor cell invasion [70]. According to Saleh et al., Hsp70 interacts with Apaf-1, preventing its oligomerization

and association of Apaf-1 with procaspase-9 [71]. On the other hand, Beere et al. showed that Hsp70 does not block Apaf-1 oligomerization but prevents caspase activation mediated by cytochrome c/dATP, binding to Apaf-1 and blocking the recruitment of apoptosome [72]. Protein kinase A It has been reported that cAMP inhibits apoptosis [73,74]. cAMP binds to PKA, dissociates the holoenzyme and releases the free catalytic subunits. In other words, cAMP regulates apoptosis by the activation of PKA [75]. Martin et al. showed that PKA phosphorylates caspase-9 at Ser99, 183 and 195. But this event is not necessary for inhibition of caspase-9. Instead, PKA inhibits caspase-9/3 activation through prevention of apoptosome assembly [76]. 2.16

Protein kinase C Another protein kinase, PKC is also a modulator of apoptosis. The PKC family, which consists of at least 10 isoforms with distinct methods of regulation, has been shown to have both inhibitory and activating effect on apoptosis. PKC a, b, " and atypical isoforms are anti-apoptotic, whereas the d and q isoforms are involved in the stimulation of apoptosis [77]. Interestingly, caspase-9 is phosphorylated at Ser144 by PKC z and inhibits apoptosis [14]. 2.17

2.18 Tumor-upregulated CARD-containing antagonist of caspase nine

The CARD protein interaction motif is found in apoptosis regulatory proteins [78]. CARD-containing pro-caspases are -1, -2, -4, -5, -9, -11, -12 and -13. Also, there are several noncaspase CARD-containing proteins such as Apaf-1, NAC, cIAP1, cIAP2 etc. According to Pathan et al., CARD-carrying protein, TUCAN, selectively binds to procaspase-9, inhibiting its apoptotic activity [79]. 2.19 Human X-chromosome-linked inhibitor of apoptosis protein

Inhibitor of apoptosis protein (IAP) family proteins are characterized by the baculoviral IAP repeat (BIR), the name of which was derived from the original discovery of these apoptosis suppressors in the genomes of baculoviruses by Birnbaum et al. [80]. XIAP has been reported to suppress apoptosis by inhibition of active caspase-9 through its BIR3 domain [81,82]. XIAP binds to the amino terminus of the linker peptide on the procaspase-9 on Asp315, suppressing the activity of caspase-9 [83]. Shiozaki et al. revealed that XIAP blocks caspase-9 homo-dimerization thus inhibiting its activity [84].

2.15

microRNAs Several studies have shown that microRNAs, small noncoding RNA, play critical role in cancer development, invasion and metastasis [85-91]. Thousands of microRNAs were discovered up to date. These control about one-third of all human 2.20

Expert Opin. Ther. Targets (2015) 19(1)

117

B. Kim et al.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

Table 2. miRNAs regulating caspase-9. Regulator

Efficacy

Mechanism

System

miRNA-133

Inhibition

In vitro

H9C2, HEK293

[94]

miRNA-24a miRNA-582-5p

Inhibition Inhibition

Inhibits casp-9 expression at both protein and mRNA levels Inhibits casp-9 and Apaf-1 activities Inhibits casp-9 expression at both protein and mRNA levels

In vivo In vitro

[95] [99]

miRNA-23a

Inhibition

X. Laevis eggs GBM stem cell lines (0308, 0822, 1288), U373, A172 HCT116, HT29

Inhibits casp-9 activity

mRNAs. Single microRNAs targets whole complex of mRNAs, which have same signal pathways and same target [92,93]. microRNA-133, preferentially expressed in cardiac and skeletal muscles, is known as regulator of differentiation and proliferation. microRNA-133 repressed caspase-9 expression at both the protein and mRNA levels [94]. microRNA-24a is also believed to have inhibitory effects on caspase-9. microRNA-24a is expressed in the neural retina and is required for correct eye morphogenesis in Xenopus. Primary microRNA RT-PCR from stages 7 to 31 showed that microRNA-24a expression began at stage 19 and was expressed throughout maturation. The controlling of eye size in xenopus was regulated by microRNA-24a overexpression as shown in loss-of-function experiments. microRNA-24a inhibited caspase-9 activity [95]. Glioblastoma multiforme is a brain cancer, its morbidity in the US is around 17,600 [96]. After surgical resection, glioblastoma multiforme recur shortly, indicating possible existence of glioblastoma stem cells, which may have resisted chemotherapy [97,98]. According to Floyd et al., microRNA-582-5p was predicted to target caspase-9 and that was confirmed by Western blotting, qPCR. microRNA-582-5p effectively inhibited caspase-9 protein and mRNA expressions [99]. Shang et al., showed that microRNA-23a (miR-23a)-regulated apoptosis in response to the 5-fluorouracil-induced intrinsic apoptotic pathway. 5-fluorouracil treatment increased the expression of miR-23a and decreased the level of Apaf-1 in colon cancer cells. Apaf-1, as a target gene of miR-23a, was identified and miR-23a antisense activated caspase-9 (Table 2) [100].

In vitro

Caspase-9 activation by natural compounds Accumulating literature suggests that natural compounds play significant inhibitory role in cancer development, invasion and drug resistance. Natural compounds often modify all phases of cancer and several of these targets at the same time [101]. It is no wonder that around 75% of the approved chemotherapeutic agents are of natural origin [102]. During the past decades, to reduce the side effects and enhance the therapeutic effects, botanical compounds play important role in the development of drugs. To suppress cancer growth, most of the natural compounds induce apoptosis via activation of caspase-9. Some of the natural compounds, such as emodin, phenethyl isothiocyanate and 118

Ref.

[100]

fucosterol, are reported to regulate caspase-9 and enhance the apoptosis process in several cancers (Table 3) [24,101,103-139]. 4.

Clinical trials for caspase-9

Two clinical trials targeting caspase-9 were recently initiated by Brenner et al. [140]. The Phase I trial, ‘CASPALLO: Allodepleted T cells transduced with inducible casepase-9 suicide gene’ is being currently conducted against acute lymphoblastic leukemia, non-Hodgkin’s lymphoma, myelodysplastic syndrome and chronic myeloid leukemia. This trial is currently active but not recruiting any patients. Patients will be receiving a stem cell transplant and will be given chemotherapy, to supposedly kill their stem cells. It is hard to find a perfect match for the patients, so find a close relative whose stem cells can be used, so-called ‘allogenic.’ To avoid graft-versushost disease (GvHD), allogenic cells will be grafted in the patients. Specialized cells are made to carry inducible caspase-9 (iCasp9). These cells can be killed when they encounter a specific drug called AP1903. This drug is being tested for GvHD [140]. Another ongoing clinical trial on “Administration of donor T cells with the caspase-9 suicide gene” is in recruiting stage currently. The base concept is the same as former one, but to find a safe and effective dose of iCasp9 and those specialized T cells, so can recover patient’s immune system faster [141]. 5.

3.

Cell lines

Conclusion

Caspase-9, initiator caspase, induces death signals by activating other caspases, and its expression triggers the apoptosis. Caspase-9 has a pivotal role in apoptosis in multiple cancer cells types. The positive regulators such as HCA66, histone H1.2, PHAP and NAC and negative regulators such as Akt, APIP, c-Abl and microRNAs have been reported. The regulation of caspase-9 could be phosphorylation at different sites, recruitment of caspase-9 to apoptosome or inhibition of caspase-9 expression. Activation of caspase-9 by natural compounds has been studied exhaustively. Development of new drugs against cancer from natural resources is important because resistance to conventional drugs is a big issue in cancer treatment. There are ongoing clinical trials in USA using caspase-9 as a suicide gene in T cells. These efforts are

Expert Opin. Ther. Targets (2015) 19(1)

Expert Opin. Ther. Targets (2015) 19(1)

Green tea

Ellagitannins

Rheum palmatum L.

Polytrichum commune L.exHedw Algae

Glycine max

EGCG

Ellagic acid

Emodin

Ethyl acetate

Genistein

Efficacy

Apoptosis

Apoptosis, cell cycle arrest

Apoptosis

Apoptosis

Apoptosis

Apoptosis

Apoptosis

Apoptosis, cell cycle arrest, sensitization of drug resistance Apoptosis, anti-angiogenesis, chemo-sensitization, anti-metastasis

Apoptosis

Apoptosis

ERK: Extracellular signal-regulated kinase; N/A: Not available.

Fucosterol

Clematis ganpiniana

D Rhamnose b-Hederin

Curcumin

Curcuma longa

Berberis amurensis

Berbamine

Cudraxanthone I

Erythrina suberosa

Origin

Alpinumisoflavone

Name

Table 3. Natural compounds regulating caspase-9.

Cytochrome c, casp-3, -8, -9, Fas, FasL, FADD " MMP # FasL, cytochrome c, Bid, caspase-3, -9, miR-574-3p " PI3K, Akt, MMP, MDM2, EGFR #

Bax, casp-3, -8, -9, PARP, p53, cytochrome c, Apaf-1, FasL, HSP " Bcl-2, Bcl-xl, Mcl-1, b-catenin, MMP-2, -9, VEGF-1, HDAC, NF-kB # ERK, cytochrome c release, casp-3, -9 " PI3K/Akt # Casp-3, -4, -9, p38, Bax, cytochrome c, ROS " MMP # Casp-3, -9 " MMP # Casp-3, -8, -9, Apaf-1, Fas, FasL, FADD " Pro-casp-3, -8, -9, PCNA, cFLIP # ROS, Casp-9 " MMP #

Cytochrome c, Bax, casp-3, -9 and PARP " Casp-3, -9, PARP, JNK, c-Jun " Cyclin D1, D2 # Casp-3/7, -8, -9 " MMP #

Mechanism

54

N/A

299.7

518

0.17 ± 0.02

39.6 ± 14.2

N/A

484 ± 74

N/A

N/A

N/A

Bioavailability (mg·h/l)

In vitro, in vivo

In vitro

In vitro

In vitro

In vitro

In vitro, in vivo

In vitro

In vitro, in vivo

In vitro

In vitro

In vitro

System

Cell lines

MCF-7, MCF-10a, HeLa, PC3, DU145, RWPE-1, xenograft mouse

HL-60

L1210

HeLa HBZY-1

MCF-7, MDA-MB-231, BT474, SUM1315, MCF-10A TSGH-8301, T98G, U87MG, SMMC7721, xenograft mouse SY5Y

A549, HepG2, A2780CP, MCF-7, mice, C57 BL/6EAE mice

CCRF-CEM, HepG2, MDA-MB-231, HCT116, U87MG, AML12

G292, KHOS, MG-63

HL-60

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

[136-139]

[109]

[107]

[24] [156]

[104]

[133-135]

[106]

[129] [155] [130-132] [127,128]

[111]

[123]

[101]

Ref.

Role of caspase-9 in apoptosis

119

120

Juglans mandshurica

Chromohalobacter salexigens Olea europaea Sophora family

Juglanthraquinone C

K30

Expert Opin. Ther. Targets (2015) 19(1)

Sargassum fusiforme

Citrus peel oil

Tannins

SFPS-B2

Tangeretin

Tannic acid

Apoptosis

Apoptosis

Apoptosis, cell cycle arrest Apoptosis

Apoptosis

Apoptosis Apoptosis, cell cycle arrest Apoptosis

Apoptosis

Apoptosis, cell cycle arrest

Apoptosis, cell cycle arrest Apoptosis

Apoptosis, cell cycle arrest

ROS, casp-3, -9, p53, p21, Bax " MMP # Casp-3, -9, Bax/Bcl-2 ratio " Ki67, cyclin A, E, CDK2, Cip1/p21 # Casp-3/7, -9, PARP-1 " MMP # JNK, casp-9 " p21, p53, Casp-9 " Bcl-2/BID ratio # Casp-3, -9, sub-G1 " MMP, SOD, GSH, Bcl-2/Bax ratio # Bax, casp-3, -9 " MMP, Bcl-xl # Bax/Bcl-2 ratio, Casp-3, -9, PARP, ROS, P53 " MPTP, Casp-3, -9, PARP, Bax, cytochrome c " MMP, Bcl-2 # Casp-3, -9, PARP, Bax, Bid, tBid, p53, p-21/cip1, Fas " Casp-3/7, -9 "

DR4, FasL, Bax/Bcl-2 ratio, casp-3, -8, -9, Bid " Bax, P21, casp-3, -9 " Bcl-2, cycline A, E, CDK2 # ROS, casp-3, -9 "

Apoptosis

N/A

N/A

N/A

N/A

88.7 ± 2.2

N/A

N/A N/A

N/A

N/A

N/A

N/A

N/A

N/A

40.0 ± 15.7

Casp-3, -8, -9 "

Apoptosis

Bioavailability (mg·h/l) N/A

Mechanism Sub-G1, casp-9 "

Efficacy Apoptosis

ERK: Extracellular signal-regulated kinase; N/A: Not available.

Phenethyl isothiocyanate PAP-3

Entada phaseoloides MERR. Cruciferous vegetables P. abalonus

Phaseoloideside E

Masilinic acid Matrine

Isodon rubescens

Thujopsis dolabrata Siebold and Zucc. Var. hondai Makino Crataegus Laevigata

Goniothalamus macrophyllus Panax quinquefolius Allium nigrum

Origin

Jaridonin

Hawthorn fruit

Ginsenoside 20(s)-Rg3 Hexane extract of aged black garlic Hinokitiol

Goniothalamin

Name

Table 3. Natural compounds regulating caspase-9 (continued).

vitro vitro, vivo vitro

In vitro

In vitro

In vitro

In vitro

In vitro

In In in In

In vitro

In vitro

In vitro

In vitro

In vitro, in vivo

In vitro

In vitro

In vitro

System

Cell lines

MCF10A, MCF-7, MDA-MB-231

AGS, A549, H460, and H1299

SGC-7901

MCF-7

CCA, KKU-100

HeLa, MCF-7, DU125 Caco-2, HT-29 Eca-109, Xenograft model Ec-109, L-O2, 3T3-L1

HepG2

EC9706

HCG-116, SW-620, Xenograft model MCF-7

U937

AGS

HeLa

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

[122]

[108] [157]

[109]

[120]

[121]

[21,115]

[118] [122]

[119]

[124]

[114]

[113]

[112]

[116]

[117]

[103]

Ref.

B. Kim et al.

necessary to better understand caspase-9-related apoptosis to lead in the identification of novel therapeutic targets.

[101]

[126]

[30]

[30]

[105]

Ref.

Role of caspase-9 in apoptosis

HepG2, Kunming mice HL-60 In vitro, in vivo In vitro N/A

1007.1

N/A

K562, Xenograft model PC-3, LNCaP, DU145 In vitro, in vivo In vivo N/A

EC-1, ECa-109 In vitro 346.98 ± 32.748

Apoptosis, cell cycle arrest Apoptosis Juglans cathayensis

Erythrina suberosa

2-Methoxyjuglone

4’-Methoxy licoflavanone

ERK: Extracellular signal-regulated kinase; N/A: Not available.

Apoptosis Ursolic acid

Marsdenia tenacissima Tenacissoside C

Apoptosis

Bax/Bcl-2 ratio, caspase-3, -9, p53, p21 " Akt1, cyclin B1, CDC2 # Bax, Bak, casp-3, -9 " Bcl-2, Bcl-xl # Caspase-3,-9, Bax, PARP, GSK3b " Bcl-XL, Bcl-2, Mcl-1, Wnt5a/b and b-catenin # Cytochrome c, casp-3, -9, PARP " Cytochrome c release, BAX, activation of casp-3, -9 and PARP cleavage Apoptosis Salviae miltiorrhizaew Tanshinone IIA

System Bioavailability (mg·h/l) Mechanism Efficacy Origin Name

Table 3. Natural compounds regulating caspase-9 (continued).

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

Cell lines

6.

Expert opinion

Apoptosis is a program of cell suicide and plays role in embryonic development and homeostasis of adult tissues [142,143]. The basic concept of apoptosis was established in 1972 by Kerr et al. [144]. Apoptosis is characterized by condensation and fragmentation of nuclear chromatin, compaction of cytoplasmic organelles, dilatation of the endoplasmic reticulum and phagocytosis of apoptotic cells [145]. Inappropriate or resistance to apoptosis is related to many human diseases such as neurodegenerative diseases like Alzheimer’s disease, autoimmune diseases and cancer [146]. Researchers have reported hundreds of genes that control the initiation, execution and regulation of apoptosis [142]. Caspases play pivotal role in the apoptosis process. Caspase-9 is involved in apoptosis pathways as an initiator caspase, activated by apoptosome that consist of Apaf-1, cytochrome c and dATP. Caspase-9 bears high similarity to caspase-3, the major difference between caspase-9 and other CED-3 subfamily is the active-site pentapeptide QACGG, Gly takes place instead of the usual Arg [147]. In this pathway, series of steps are tightly regulated, for example, cytochrome c is released from mitochondria, binding Apaf-1, forming apoptosome by oligomerization of Apaf-1, eventually activating caspase-9 [148]. Activated caspase-9 cleaves and activates the effector caspase-3. Much of the fundamental process of caspase-9 activation has been studied and now known at atomic level, but there are remaining challenges in viewing the entire process. For example, exact arrangement of Apaf1 with caspase-9 is not clear. There are significant difference in the predicted size of apoptosome and the experimentally observed size of apoptosome [149]. Also, it is not clear why affinity of caspase-9 is reduced during apoptosis process. Several studies reported that caspase-9 is directly targeted for activation or inhibition by several molecules, including protein kinases, heat shock protein and microRNAs (Tables 1 and 2). Similarly, several agents whether natural or synthetic have been shown to activate caspase-9 leading to apoptosis (Table 3). These molecules/agents can be candidate mediators for development of new drugs, regulating apoptosis in cancer cells. The benefits of targeting caspase-9 in cancer treatment are that cancer cells are more susceptible to certain treatment than normal cells. According to Warburg effect, cancer cells rely on glycolysis to meet high metabolic demands, producing energy at much higher rate through glycolysis followed by lactic acid fermentation in the cytosol, rather than by a relatively lower rate of glycolysis followed by oxidation of pyruvate in the mitochondria of normal cells. Even in the presence of sufficient oxygen, glucose metabolism is upregulated in cancer cells. There are increasing number of studies that have recently shown that caspases play a role in multiple cellular processes

Expert Opin. Ther. Targets (2015) 19(1)

121

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

B. Kim et al.

including immunity, cell survival, cell proliferation, cell cycle regulation and cell differentiation other than apoptosis purpose [150-152]. How and why this process takes place is not clear. Also, few caspases like caspase-1 and -5 are involved in inflammation and are called inflammatory caspases. Like apoptosome formation during apoptosis, inflammasome is formed during inflammation and is composed of caspase-1, NALP3 and ASC [153]. In-depth studies are required to show the activation of caspase-9 and its role in inducing apoptosis in numerous cancer cells. Also, more clinical trials are demanded to establish the role of caspase-9 in suppressing cancer. As an apoptosis-initiator, caspase-9 remains important in the field of apoptosis. Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

2.

3.

4.

5.

Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 1997;3(8):917-21

SK Srivastava was supported by in part by R01 grant CA129038 awarded by National Cancer Institute, NIH, and MRC grant2007--0054931 to Kim. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

10.

11.

12.

Allan LA, Morrice N, Brady S, et al. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol 2003;5(7):647-54 Martin MC, Allan LA, Mancini EJ, et al. The docking interaction of caspase-9 with ERK2 provides a mechanism for the selective inhibitory phosphorylation of caspase-9 at threonine 125. J Biol Chem 2008;283(7):3854-65 Seifert A, Allan LA, Clarke PR. DYRK1A phosphorylates caspase 9 at an inhibitory site and is potently inhibited in human cells by harmine. FEBS J 2008;275(24):6268-80

lead compounds isolated from marine organisms of potential relevance in cancer treatment. Mutat Res 2014. [Epub ahead of print] 18.

Hakem R, Hakem A, Duncan GS, et al. Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell 1998;94(3):339-52

19.

Renatus M, Stennicke HR, Scott FL, et al. Dimer formation drives the activation of the cell death protease caspase 9. Proc Natl Acad Sci USA 2001;98(25):14250-5

20.

Pop C, Timmer J, Sperandio S, et al. The apoptosome activates caspase-9 by dimerization. Mol Cell 2006;22(2):269-75

13.

21.

Logue SE, Gustafsson AB, Samali A, et al. Ischemia/reperfusion injury at the intersection with cell death. J Mol Cell Cardiol 2005;38(1):21-33

Cardone MH, Roy N, Stennicke HR, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998;282(5392):1318-21

Gupta P, Kim B, Kim SH, et al. Molecular targets of isothiocyanates in cancer: recent advances. Mol Nutr Food Res 2014;58(8):1685-707

14.

Brady SC, Allan LA, Clarke PR. Regulation of caspase 9 through phosphorylation by protein kinase C zeta in response to hyperosmotic stress. Mol Cell Biol 2005;25(23):10543-55

22.

15.

McDonnell MA, Abedin MJ, Melendez M, et al. Phosphorylation of murine caspase-9 by the protein kinase casein kinase 2 regulates its cleavage by caspase-8. J Biol Chem 2008;283(29):20149-58

Cang S, Ma Y, Chiao JW, et al. Phenethyl isothiocyanate and paclitaxel synergistically enhanced apoptosis and alpha-tubulin hyperacetylation in breast cancer cells. Exp Hematol Oncol 2014;3(1):5

23.

Cuddihy SL, Brown KK, Thomson SJ, et al. Induction of apoptosis by phenethyl isothiocyanate in cells overexpressing Bcl-XL. Cancer Lett 2008;271(2):215-21

Garrido C, Galluzzi L, Brunet M, et al. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ 2006;13(9):1423-33 Interesting review on mechanisms of apoptosis relatedcytochrome c release.

24.

Yaoxian W, Hui Y, Yunyan Z, et al. Emodin induces apoptosis of human cervical cancer hela cells via intrinsic mitochondrial and extrinsic death receptor pathway. Cancer Cell Int 2013;13(1):71

Beesoo R, Neergheen-Bhujun V, Bhagooli R, et al. Apoptosis inducing

25.

Ha MK, Song YH, Jeong SJ, et al. Emodin inhibits proinflammatory

Oliveira JB, Gupta S. Disorders of apoptosis: mechanisms for autoimmunity in primary immunodeficiency diseases. J Clin Immunol 2008;28(Suppl 1):S20-8

7.

Wurstle ML, Laussmann MA, Rehm M. The central role of initiator caspase-9 in apoptosis signal transduction and the regulation of its activation and activity on the apoptosome. Exp Cell Res 2012;318(11):1213-20

8.

Raina D, Pandey P, Ahmad R, et al. c-Abl tyrosine kinase regulates caspase-9 autocleavage in the apoptotic response to DNA damage. J Biol Chem 2005;280(12):11147-51

122

Declaration of interest

Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer. Nature 2001;411(6835):342-8

6.

9.

SK Srivastava is currently an International Scholar at Kyunghee University, Seoul, South Korea.

mitotic cells against apoptosis. Mol Cell 2007;26(2):301-10

Park HH. Structural features of caspaseactivating complexes. Int J Mol Sci 2012;13(4):4807-18 Pawlowski K, Pio F, Chu Z, et al. PAAD - a new protein domain associated with apoptosis, cancer and autoimmune diseases. Trends Biochem Sci 2001;26(2):85-7

Acknowledgements

Allan LA, Clarke PR. Phosphorylation of caspase-9 by CDK1/cyclin B1 protects

16.

..

17.

Expert Opin. Ther. Targets (2015) 19(1)

Role of caspase-9 in apoptosis

responses and inactivates histone deacetylase 1 in hypoxic rheumatoid synoviocytes. Biol Pharm Bull 2011;34(9):1432-7 26.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

27.

28.

29.

Jiang FL, Leo S, Wang XG, et al. Effect of tanshinone IIA on cardiomyocyte hypertrophy and apoptosis in spontaneously hypertensive rats. Exp Ther Med 2013;6(6):1517-21 Jiao JW, Wen F. Tanshinone IIA acts via p38 MAPK to induce apoptosis and the down-regulation of ERCC1 and lungresistance protein in cisplatin-resistant ovarian cancer cells. Oncol Rep 2010;25(3):781-8 Jung JH, Kwon TR, Jeong SJ, et al. Apoptosis induced by tanshinone IIA and cryptotanshinone is mediated by distinct JAK/STAT3/5 and SHP1/2 signaling in chronic myeloid leukemia K562 cells. Evid Based Complement Alternat Med 2013;2013:805639 Yun SM, Jung JH, Jeong SJ, et al. Tanshinone IIA induces autophagic cell death via activation of AMPK and ERK and inhibition of mTOR and p70 S6K in KBM-5 leukemia cells. Phytother Res 2013;28(3):458-64

35.

36.

..

37.

Piddubnyak V, Rigou P, Michel L, et al. Positive regulation of apoptosis by HCA66, a new Apaf-1 interacting protein, and its putative role in the physiopathology of NF1 microdeletion syndrome patients. Cell Death Differ 2007;14(6):1222-33 Ruiz-Vela A, Korsmeyer SJ. Proapoptotic histone H1.2 induces CASP-3 and -7 activation by forming a protein complex with CYT c, APAF-1 and CASP-9. FEBS Lett 2007;581(18):3422-8 Interesting paper on apoptosome and caspase-3, -7 activation. Jiang X, Kim HE, Shu H, et al. Distinctive roles of PHAP proteins and prothymosin-alpha in a death regulatory pathway. Science 2003;299(5604):223-6

38.

Yuan JY, Horvitz HR. The Caenorhabditis elegans genes ced-3 and ced-4 act cell autonomously to cause programmed cell death. Dev Biol 1990;138(1):33-41

39.

Yang X, Chang HY, Baltimore D. Essential role of CED-4 oligomerization in CED-3 activation and apoptosis. Science 1998;281(5381):1355-7

the induction of stress-induced apoptosis. J Biol Chem 2004;279(39):41131-40 46.

Cho DC. Targeting the PI3K/Akt/ mTOR pathway in malignancy: rationale and clinical outlook. BioDrugs 2014;8(4):373-81

47.

Ocana A, Vera-Badillo F, Al-Mubarak M, et al. Activation of the PI3K/mTOR/Akt Pathway and survival in solid tumors: systematic review and meta-analysis. PLoS One 2014;9(4):e95219

48.

Kang W, Hong SH, Lee HM, et al. Structural and biochemical basis for the inhibition of cell death by APIP, a methionine salvage enzyme. Proc Natl Acad Sci USA 2013;111(1):E54-61

49.

Kang W, Yang JK. Crystallization and preliminary X-ray crystallographic analysis of human Apaf-1-interacting protein. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012;68(Pt 12):1518-20

50.

Cho DH, Lee HJ, Kim HJ, et al. Suppression of hypoxic cell death by APIP-induced sustained activation of Akt and ERK1/2. Oncogene 2007;26(19):2809-14

30.

Park JH, Kwon HY, Sohn EJ, et al. Inhibition of Wnt/beta-catenin signaling mediates ursolic acid-induced apoptosis in PC-3 prostate cancer cells. Pharmacol Rep 2013;65(5):1366-74

40.

Chu ZL, Pio F, Xie Z, et al. A novel enhancer of the Apaf1 apoptosome involved in cytochrome c-dependent caspase activation and apoptosis. J Biol Chem 2001;276(12):9239-45

51.

Cho DH, Hong YM, Lee HJ, et al. Induced inhibition of ischemic/hypoxic injury by APIP, a novel Apaf-1-interacting protein. J Biol Chem 2004;279(38):39942-50

31.

Jang J, Jeong SJ, Kwon HY, et al. Decursin and doxorubicin are in synergy for the induction of apoptosis via STAT3 and/or mTOR pathways in human multiple myeloma cells. Evid Based Complement Alternat Med 2013;2013:506324

41.

Hlaing T, Guo RF, Dilley KA, et al. Molecular cloning and characterization of DEFCAP-L and -S, two isoforms of a novel member of the mammalian Ced-4 family of apoptosis proteins. J Biol Chem 2001;276(12):9230-8

52.

Chen L, Wang Z, Tang B, et al. Altered expression of c-Abl in patients with epilepsy and in a rat model. Synapse 2014;68(7):306-16

53.

Arora S, Saini S, Fukuhara S, et al. MicroRNA-4723 inhibits prostate cancer growth through inactivation of the Abelson family of nonreceptor protein tyrosine kinases. PLoS One 2013;8(11):e78023

54.

Yu F, Finley RL Jr, Raz A, et al. Galectin-3 translocates to the perinuclear membranes and inhibits cytochrome c release from the mitochondria. A role for synexin in galectin-3 translocation. J Biol Chem 2002;277(18):15819-27

Kharbanda S, Ren R, Pandey P, et al. Activation of the c-Abl tyrosine kinase in the stress response to DNA-damaging agents. Nature 1995;376(6543):785-8

55.

Welch AY, Herman IM. Cloning and characterization of betaCAP73, a novel regulator of beta-actin assembly. Int J Biochem Cell Biol 2002;34(7):864-81

Yuan ZM, Huang Y, Fan MM, et al. Genotoxic drugs induce interaction of the c-Abl tyrosine kinase and the tumor suppressor protein p53. J Biol Chem 1996;271(43):26457-60

56.

Raynaud C, Mallory AC, Latrasse D, et al. Chromatin meets the cell cycle. J Exp Bot 2014;65(10):2677-89

Sakai T, Liu L, Teng X, et al. Nucling recruits Apaf-1/pro-caspase-9 complex for

57.

Koltovaya NA. [Involvement of cyclindependent kinase CDK1/CDC28 in

32.

Choi SR, Lee JH, Kim JY, et al. Decursin from Angelicagigas Nakai induces apoptosis in RC-58T/h/ SA#4 primary human prostate cancer cells via a mitochondria-related caspase pathway. Food Chem Toxicol 2011;49(10):2517-23

33.

Yim D, Singh RP, Agarwal C, et al. A novel anticancer agent, decursin, induces G1 arrest and apoptosis in human prostate carcinoma cells. Cancer Res 2005;65(3):1035-44

34.

De Raedt T, Brems H, Wolkenstein P, et al. Elevated risk for MPNST in NF1 microdeletion patients. Am J Hum Genet 2003;72(5):1288-92

42.

43.

44.

45.

Sakai T, Liu L, Shishido Y, et al. Identification of a novel, embryonal carcinoma cell-associated molecule, nucling, that is up-regulated during cardiac muscle differentiation. J Biochem 2003;133(4):429-36

Expert Opin. Ther. Targets (2015) 19(1)

123

B. Kim et al.

58.

59.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

124

Meggio F, Pinna LA. One-thousand-andone substrates of protein kinase CK2? FASEB J 2003;17(3):349-68 Graciotti M, Alam M, Solyakov L, et al. Malaria protein kinase CK2 (PfCK2) shows novel mechanisms of regulation. PLoS One 2014;9(3):e85391 Intemann J, Saidu NE, Schwind L, et al. ER stress signaling in ARPE-19 cells after inhibition of protein kinase CK2 by CX-4945. Cell Signal 2014;26(7):1567-75 Litchfield DW. Protein kinase CK2: structure, regulation and role in cellular decisions of life and death. Biochem J 2003;369(Pt 1):1-15 Becker W, Weber Y, Wetzel K, et al. Sequence characteristics, subcellular localization, and substrate specificity of DYRK-related kinases, a novel family of dual specificity protein kinases. J Biol Chem 1998;273(40):25893-902 Becker W, Joost HG. Structural and functional characteristics of Dyrk, a novel subfamily of protein kinases with dual specificity. Prog Nucleic Acid Res Mol Biol 1999;62:1-17 Seifert A, Clarke PR. p38alpha- and DYRK1A-dependent phosphorylation of caspase-9 at an inhibitory site in response to hyperosmotic stress. Cell Signal 2009;21(11):1626-33 Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 2004;68(2):320-44 Pramanik R, Qi X, Borowicz S, et al. p38 isoforms have opposite effects on AP-1-dependent transcription through regulation of c-Jun. The determinant roles of the isoforms in the p38 MAPK signal specificity. J Biol Chem 2003;278(7):4831-9 Melegari M, Scaglioni PP, Wands JR. Cloning and characterization of a novel hepatitis B virus x binding protein that inhibits viral replication. J Virol 1998;72(3):1737-43

70.

71.

72.

73.

74.

75.

Juhasz K, Lipp AM, Nimmervoll B, et al. The complex function of hsp70 in metastatic cancer. Cancers (Basel) 2013;6(1):42-66 Saleh A, Srinivasula SM, Balkir L, et al. Negative regulation of the Apaf-1 apoptosome by Hsp70. Nat Cell Biol 2000;2(8):476-83 Beere HM, Wolf BB, Cain K, et al. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase9 to the Apaf-1 apoptosome. Nat Cell Biol 2000;2(8):469-75 Li X, Tao H, Xie K, et al. cAMP signaling prevents podocyte apoptosis via activation of protein kinase A and mitochondrial fusion. PLoS One 2014;9(3):e92003 Naderi EH, Findley HW, Ruud E, et al. Activation of cAMP signaling inhibits DNA damage-induced apoptosis in BCP-ALL cells through abrogation of p53 accumulation. Blood 2009;114(3):608-18 Chen TC, Hinton DR, Zidovetzki R, et al. Up-regulation of the cAMP/ PKA pathway inhibits proliferation, induces differentiation, and leads to apoptosis in malignant gliomas. Lab Invest 1998;78(2):165-74

76.

Martin MC, Allan LA, Lickrish M, et al. Protein kinase A regulates caspase-9 activation by Apaf-1 downstream of cytochrome c. J Biol Chem 2005;280(15):15449-55

77.

Gutcher I, Webb PR, Anderson NG. The isoform-specific regulation of apoptosis by protein kinase C. Cell Mol Life Sci 2003;60(6):1061-70

78.

Hofmann K, Bucher P, Tschopp J. The CARD domain: a new apoptotic signalling motif. Trends Biochem Sci 1997;22(5):155-6

79.

Pathan N, Marusawa H, Krajewska M, et al. TUCAN, an antiapoptotic caspaseassociated recruitment domain family protein overexpressed in cancer. J Biol Chem 2001;276(34):32220-9

80.

Birnbaum MJ, Clem RJ, Miller LK. An apoptosis-inhibiting gene from a nuclear polyhedrosis virus encoding a polypeptide with Cys/His sequence motifs. J Virol 1994;68(4):2521-8

Murakami S. Hepatitis B virus X protein: a multifunctional viral regulator. J Gastroenterol 2001;36(10):651-60 Marusawa H, Matsuzawa S, Welsh K, et al. HBXIP functions as a cofactor of

XIAP: differential roles of the linker versus the BIR domain. Cell 2001;104(5):781-90

survivin in apoptosis suppression. EMBO J 2003;22(11):2729-40

regulation of cell cycle]. Genetika 2014;49(7):797-813

81.

Huang Y, Park YC, Rich RL, et al. Structural basis of caspase inhibition by Expert Opin. Ther. Targets (2015) 19(1)

82.

Sun M, Meares G, Song L, et al. XIAP associates with GSK3 and inhibits the promotion of intrinsic apoptotic signaling by GSK3. Cell Signal 2009;21(12):1857-65

83.

Srinivasula SM, Hegde R, Saleh A, et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 2001;410(6824):112-16

84.

Shiozaki EN, Chai J, Rigotti DJ, et al. Mechanism of XIAP-mediated inhibition of caspase-9. Mol Cell 2003;11(2):519-27

85.

Hsieh WJ, Lin FM, Huang HD, et al. Investigating microRNA-target interaction-supported tissues in human cancer tissues based on miRNA and target gene expression profiling. PLoS One 2014;9(4):e95697

86.

Humeau M, Torrisani J, Cordelier P. miRNA in clinical practice: pancreatic cancer. Clin Biochem 2013;46(10-11):933-6

87.

Kim CH, Kim HK, Rettig RL, et al. miRNA signature associated with outcome of gastric cancer patients following chemotherapy. BMC Med Genomics 2011;4:79

88.

Li J, Kong X, Zhang J, et al. MiRNA-26b inhibits proliferation by targeting PTGS2 in breast cancer. Cancer Cell Int 2013;13(1):7

89.

Tchatchou S, Jung A, Hemminki K, et al. A variant affecting a putative miRNA target site in estrogen receptor (ESR) 1 is associated with breast cancer risk in premenopausal women. Carcinogenesis 2009;30(1):59-64

90.

Tie Y, Liu B, Fu H, et al. Circulating miRNA and cancer diagnosis. Sci China C Life Sci 2009;52(12):1117-22

91.

Zhu Z, Gao W, Qian Z, et al. Genetic variation of miRNA sequence in pancreatic cancer. Acta Biochim Biophys Sin (Shanghai) 2009;41(5):407-13

92.

Liu J. Control of protein synthesis and mRNA degradation by microRNAs. Curr Opin Cell Biol 2008;20(2):214-21

93.

Lima RT, Busacca S, Almeida GM, et al. MicroRNA regulation of core apoptosis pathways in cancer. Eur J Cancer 2011;47(2):163-74

Role of caspase-9 in apoptosis

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

94.

dependent depolarization of mitochondria and activation of caspase-9 and -3 in human neuroblastoma cells. Biomed Pharmacother 2014;68(1):129-35

Xu C, Lu Y, Pan Z, et al. The musclespecific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. J Cell Sci 2007;120(Pt 17):3045-52

105.

95.

Walker JC, Harland RM. microRNA-24a is required to repress apoptosis in the developing neural retina. Genes Dev 2009;23(9):1046-51

Booth BW, Inskeep BD, Shah H, et al. Tannic Acid preferentially targets estrogen receptor-positive breast cancer. Int J Breast Cancer 2013;2013:369609

106.

96.

Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro Oncol 2010;15(Suppl 2):ii1-56

Cheng L, Xia TS, Wang YF, et al. The apoptotic effect of D Rhamnose betaHederin, a novel oleanane-type triterpenoid saponin on breast cancer cells. PLoS One 2014;9(3):e90848

97.

Singh S, Dirks PB. Brain tumor stem cells: identification and concepts. Neurosurg Clin N Am 2007;18(1):31-8; viii

98.

Vescovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nat Rev Cancer 2006;6(6):425-36

99.

100.

101.

Floyd DH, Zhang Y, Dey BK, et al. Novel Anti-Apoptotic MicroRNAs 582-5p and 363 Promote Human Glioblastoma Stem Cell Survival via Direct Inhibition of Caspase 3, Caspase 9, and Bim. PLoS One 2014;9(5):e96239 Shang J, Yang F, Wang Y, et al. MicroRNA-23a antisense enhances 5-fluorouracil chemosensitivity through APAF-1/caspase-9 apoptotic pathway in colorectal cancer cells. J Cell Biochem 2014;115(4):772-84 Kumar S, Pathania AS, Saxena AK, et al. The anticancer potential of flavonoids isolated from the stem bark of Erythrina suberosa through induction of apoptosis and inhibition of STAT signaling pathway in human leukemia HL-60 cells. Chem Biol Interact 2013;205(2):128-37

102.

Newman DJ, Cragg GM. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J Nat Prod 2012;75(3):311-35

103.

Alabsi AM, Ali R, Ali AM, et al. Induction of caspase-9, biochemical assessment and morphological changes caused by apoptosis in cancer cells treated with goniothalamin extracted from Goniothalamus macrophyllus. Asian Pac J Cancer Prev 2013;14(11):6273-80

104.

Alfredsson CF, Ding M, Liang QL, et al. Ellagic acid induces a dose- and time-

107.

108.

109.

110.

111.

112.

113.

114.

Cheng X, Xiao Y, Wang P, et al. The ethyl acetate fraction of Polytrichum commune L.ex Hedw induced cell apoptosis via reactive oxygen species in L1210 cells. J Ethnopharmacol 2013;148(3):926-33 Dong Y, Cao A, Shi J, et al. Tangeretin, a citrus polymethoxyflavonoid, induces apoptosis of human gastric cancer AGS cells through extrinsic and intrinsic signaling pathways. Oncol Rep 2014;31(4):1788-94 Ji YB, Ji CF, Yue L. Study on human promyelocytic leukemia HL-60 cells apoptosis induced by fucosterol. Biomed Mater Eng 2014;24(1):845-51 Ji YB, Ji CF, Yue L. Human gastric cancer cell line SGC-7901 apoptosis induced by SFPS-B2 via a mitochondrial-mediated pathway. Biomed Mater Eng 2014;24(1):1141-7 Kuete V, Sandjo LP, Ouete JL, et al. Cytotoxicity and modes of action of three naturally occurring xanthones (8hydroxycudraxanthone G, morusignin I and cudraxanthone I) against sensitive and multidrug-resistant cancer cell lines. Phytomedicine 2014;21(3):315-22 Lee YS, Choi KM, Kim W, et al. Hinokitiol inhibits cell growth through induction of S-phase arrest and apoptosis in human colon cancer cells and suppresses tumor growth in a mouse xenograft experiment. J Nat Prod 2013;76(12):2195-202 Li T, Zhu J, Guo L, et al. Differential effects of polyphenols-enriched extracts from hawthorn fruit peels and fleshes on cell cycle and apoptosis in human MCF7 breast carcinoma cells. Food Chem 2013;141(2):1008-18 Ma YC, Ke Y, Zi X, et al. Jaridonin, a novel ent-kaurene diterpenoid from

Expert Opin. Ther. Targets (2015) 19(1)

Isodon rubescens, inducing apoptosis via production of reactive oxygen species in esophageal cancer cells. Curr Cancer Drug Targets 2013;13(6):611-24 115. Mo S, Xiong H, Shu G, et al. Phaseoloideside E, a novel natural triterpenoid saponin identified from Entada phaseoloides, induces apoptosis in Ec-109 esophageal cancer cells through reactive oxygen species generation. J Pharmacol Sci 2013;122(3):163-75 116. Park C, Park S, Chung YH, et al. Induction of apoptosis by a hexane extract of aged black garlic in the human leukemic U937 cells. Nutr Res Pract 2014;8(2):132-7 117. Park EH, Kim YJ, Yamabe N, et al. Stereospecific anticancer effects of ginsenoside Rg3 epimers isolated from heat-processed American ginseng on human gastric cancer cell. J Ginseng Res 2014;38(1):22-7 118. Reyes-Zurita FJ, Rufino-Palomares EE, Medina PP, et al. Antitumour activity on extrinsic apoptotic targets of the triterpenoid maslinic acid in p53deficient Caco-2 adenocarcinoma cells. Biochimie 2013;95(11):2157-67 119. Sagar S, Esau L, Holtermann K, et al. Induction of apoptosis in cancer cell lines by the Red Sea brine pool bacterial extracts. BMC Complement Altern Med 2013;13:344 120. Shi X, Zhao Y, Jiao Y, et al. ROS-dependent mitochondria molecular mechanisms underlying antitumor activity of Pleurotus abalonus acidic polysaccharides in human breast cancer MCF-7 cells. PLoS One 2013;8(5):e64266 121. Tusskorn O, Prawan A, Senggunprai L, et al. Phenethyl isothiocyanate induces apoptosis of cholangiocarcinoma cells through interruption of glutathione and mitochondrial pathway. Naunyn Schmiedebergs Arch Pharmacol 2013;386(11):1009-16 122. Wang Q, Du H, Geng G, et al. Matrine inhibits proliferation and induces apoptosis via BID-mediated mitochondrial pathway in esophageal cancer cells. Mol Biol Rep 2014;41(5):3009-20 123. Yang F, Nam S, Zhao R, et al. A novel synthetic derivative of the natural product berbamine inhibits cell viability and induces apoptosis of human osteosarcoma cells, associated with

125

B. Kim et al.

activation of JNK/AP-1 signaling. Cancer Biol Ther 2013;14(11):1024-31

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

124. Yao Y, Zhang YW, Sun LG, et al. Juglanthraquinone C, a novel natural compound derived from Juglans mandshurica Maxim, induces S phase arrest and apoptosis in HepG2 cells. Apoptosis 2012;17(8):832-41 125. Ye B, Yang J, Li J, et al. In vitro and in vivo antitumor activities of tenacissoside C from Marsdenia tenacissima. Planta Med 2013;80(1):29-38 126. Yu HY, Zhang XQ, Li X, et al. 2-methoxyjuglone induces apoptosis in HepG2 human hepatocellular carcinoma cells and exhibits in vivo antitumor activity in a H22 mouse hepatocellular carcinoma model. J Nat Prod 2013;76(5):889-95 127. Feng J, Tao T, Yan W, et al. Curcumin inhibits mitochondrial injury and apoptosis from the early stage in EAE mice. Oxid Med Cell Longev 2014;2014:728751 128. Malhotra A, Nair P, Dhawan DK. Study to evaluate molecular mechanics behind synergistic chemo-preventive effects of curcumin and resveratrol during lung carcinogenesis. PLoS One 2014;9(4):e93820 129. Li Y, Zhang S, Geng JX, et al. Curcumin inhibits human non-small cell lung cancer A549 cell proliferation through regulation of Bcl-2/Bax and cytochrome C. Asian Pac J Cancer Prev 2013;14(8):4599-602 130. Mohankumar K, Pajaniradje S, Sridharan S, et al. Mechanism of apoptotic induction in human breast cancer cell, MCF-7, by an analog of curcumin in comparison with curcumin--an in vitro and in silico approach. Chem Biol Interact 2014;210:51-63 131. Yallapu MM, Maher DM, Sundram V, et al. Curcumin induces chemo/radiosensitization in ovarian cancer cells and curcumin nanoparticles inhibit ovarian cancer cell growth. J Ovarian Res 2010;3:11 132. Cao Y, Liu Q. Therapeutic targets of multiple angiogenic factors for the treatment of cancer and metastasis. Adv Cancer Res 2007;97:203-24 133. Li W, Nie S, Yu Q, et al. (-)Epigallocatechin-3-gallate induces

126

apoptosis of human hepatoma cells by mitochondrial pathways related to reactive oxygen species. J Agric Food Chem 2009;57(15):6685-91 134. Chen NG, Lu CC, Lin YH, et al. Proteomic approaches to study epigallocatechin gallate-provoked apoptosis of TSGH-8301 human urinary bladder carcinoma cells: roles of Akt and heat shock protein 27-modulated intrinsic apoptotic pathways. Oncol Rep 2011;26(4):939-47 135. Das A, Banik NL, Ray SK. Flavonoids activated caspases for apoptosis in human glioblastoma T98G and U87MG cells but not in human normal astrocytes. Cancer 2010;116(1):164-76 136. Chen FP, Chien MH. Phytoestrogens induce differential effects on both normal and malignant human breast cells in vitro. Climacteric 2014;1-26 137. Dhandayuthapani S, Marimuthu P, Hormann V, et al. Induction of apoptosis in HeLa cells via caspase activation by resveratrol and genistein. J Med Food 2013;16(2):139-46 138. Chiyomaru T, Yamamura S, Fukuhara S, et al. Genistein up-regulates tumor suppressor microRNA-574-3p in prostate cancer. PLoS One 2013;8(3):e58929 139. Pham J, Brownlow B, Elbayoumi T. Mitochondria-specific pro-apoptotic activity of genistein lipidic nanocarriers. Mol Pharm 2013;10(10):3789-800 140. ClinicalTrials.gov. CASPALLO: allodepleted T Cells Transduced With Inducible Caspase 9 Suicide Gene. 2008.Available from: https://clinicaltrials. gov/ct2/show/NCT00710892? term=caspase-9&rank=2 141. ClinicalTrials.gov. Administration of Donor T Cells With the Caspase-9 Suicide Gene (DOTTI). 2011.Available from: https://clinicaltrials. gov/ct2/show/NCT01494103? term=caspase-9&rank=1 142. Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell 2004;116(2):205-19 143. Schafer ZT, Kornbluth S. The apoptosome: physiological, developmental, and pathological modes of regulation. Dev Cell 2006;10(5):549-61 . Excellent review on structural, functional analyses of apoptosome.

Expert Opin. Ther. Targets (2015) 19(1)

144. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972;26(4):239-57 145. Arends MJ, Wyllie AH. Apoptosis: mechanisms and roles in pathology. Int Rev Exp Pathol 1991;32:223-54 146. Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol 2004;5(11):897-907 147. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997;326(Pt 1):1-16 148. Allan LA, Clarke PR. Apoptosis and autophagy: regulation of caspase-9 by phosphorylation. FEBS J 2009;276(21):6063-73 . Excellent review on regulation of caspase-9 by phosphorylation. 149. Cain K, Bratton SB, Langlais C, et al. Apaf-1 oligomerizes into biologically active approximately 700-kDa and inactive approximately 1.4-MDa apoptosome complexes. J Biol Chem 2000;275(9):6067-70 150. Yu X, Acehan D, Menetret JF, et al. A structure of the human apoptosome at 12.8 A resolution provides insights into this cell death platform. Structure 2005;13(11):1725-35 151. Yi CH, Yuan J. The Jekyll and Hyde functions of caspases. Dev Cell 2009;16(1):21-34 . Excellent paper onfunctions of caspases. 152. Bratton SB, Salvesen GS. Regulation of the Apaf-1-caspase-9 apoptosome. J Cell Sci 2010;123(Pt 19):3209-14 . Excellent review on regulation of apoptosome. 153. Bae JY, Park HH. Crystal structure of NALP3 protein pyrin domain (PYD) and its implications in inflammasome assembly. J Biol Chem 2011;286(45):39528-36 .. Interesting paper on inflammasome and caspases. 154. Tashker JS, Olson M, Kornbluth S. Post-cytochrome C protection from apoptosis conferred by a MAPK pathway in xenopus egg extracts. Mol Biol Cell 2002;13(2):393-401 155. Dai XZ, Yin HT, Sun LF, et al. Potential therapeutic efficacy of curcumin in liver cancer. Asian Pac J Cancer Prev 2013;14(6):3855-9

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by JHU John Hopkins University on 01/07/15 For personal use only.

Role of caspase-9 in apoptosis

156.

Gao J, Wang F, Wang W, et al. Emodin suppresses hyperglycemia-induced proliferation and fibronectin expression in mesangial cells via inhibiting cFLIP. PLoS One 2014;9(4):e93588

157.

Charoensinphon N, Qiu P, Dong P, et al. 5-Demethyltangeretin inhibits human nonsmall cell lung cancer cell growth by inducing G2/M cell cycle arrest and apoptosis. Mol Nutr Food Res 2013;57(12):2103-11

Affiliation Bonglee Kim1,2 MD (KMD) PhD, Sanjay K Srivastava1,3 PhD & Sung-Hoon Kim†4 MD (KMD) PhD † Author for correspondence 1 Kyunghee University, College of Korean Medicine, Cancer Preventive Material Development Research Center, 1 Hoegi-dong, Dongdaemun-ku, Seoul 131-701, South Korea 2 Postdoctoral Research Fellow, Texas Tech University Health Sciences Center, Department of Biomedical Sciences and Cancer Biology Center, Amarillo, TX 79106, USA 3 Professor, Texas Tech University Health Sciences Center, Department of Biomedical Sciences and Cancer Biology Center, Amarillo, TX 79106, USA 4 Professor, Kyunghee University, College of Korean Medicine, Cancer Preventive Material Development Research Center, 1 Hoegi-dong, Dongdaemun-ku, Seoul 131-701, South Korea Tel: +82 2 964 1074; Fax: +82 2 964 1064; E-mail: [email protected]

Expert Opin. Ther. Targets (2015) 19(1)

127

Caspase-9 as a therapeutic target for treating cancer.

Caspase-9 is the apoptotic initiator protease of the intrinsic or mitochondrial apoptotic pathway, which is activated at multi-protein activation plat...
668KB Sizes 4 Downloads 11 Views