Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1891 word(s) 1891 2021-10-09 04:19:39 |
2 update references and layout Meta information modification 1891 2021-10-11 04:26:17 | |
3 The format is correct Meta information modification 1891 2021-11-12 09:18:49 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Yadav, D. B Cell Lymphoma 2 and Cancer Therapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/14922 (accessed on 29 March 2024).
Yadav D. B Cell Lymphoma 2 and Cancer Therapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/14922. Accessed March 29, 2024.
Yadav, Dharmendra. "B Cell Lymphoma 2 and Cancer Therapy" Encyclopedia, https://encyclopedia.pub/entry/14922 (accessed March 29, 2024).
Yadav, D. (2021, October 10). B Cell Lymphoma 2 and Cancer Therapy. In Encyclopedia. https://encyclopedia.pub/entry/14922
Yadav, Dharmendra. "B Cell Lymphoma 2 and Cancer Therapy." Encyclopedia. Web. 10 October, 2021.
B Cell Lymphoma 2 and Cancer Therapy
Edit

Bcl-2 is an anti-apoptotic protein that is associated with several cancer progression. Bcl-2 was the first protein to be documented among the Bcl-2 family proteins. It was the first gene exhibited to promote prolonged cell survival and growth rather than enhanced proliferation, which revealed that inhibition of cell death is vital in tumorigenesis.

B cell lymphoma 2 cancers apoptosis inhibitors clinical trials targeted therapy

1. Introduction

Bcl-2 is an anti-apoptotic protein that is associated with several cancer progression [1][2]. Bcl-2 was the first protein to be documented among the Bcl-2 family proteins [3]. It was the first gene exhibited to promote prolonged cell survival and growth rather than enhanced proliferation, which revealed that inhibition of cell death is vital in tumorigenesis [4]. Bcl-2 represses apoptosis by inhibiting cytochrome c ( cyt- c ) release from the mitochondria, preventing activation of caspases responsible for apoptosis [5]. Elevated expression of Bcl-2 is involved in many cancer progressions [6]. The increased expression of Bcl-2 is linked with advanced stage neoplasms and poor differentiation [7]. Bcl-2 plays a vital role in angiogenesis and cancer growth [8]. Apoptosis is the most important mechanism of cell death in response to tumor therapies [9]. Targeting the Bcl-2 protein may improve apoptosis by chemotherapeutic agents [10][11]. Bcl-2 inhibitors have exhibited promising effects in several malignancies with a single drug and combination with other drugs since Bcl-2 is a potential therapeutic target for malignancies [12].

Bcl-2 is the most significant protein, its overexpression and phosphorylation may be linked to regulation of cell growth, cell cycle, proliferation, DNA repair, and tumorigenesis. Elevated expression of Bcl-2 protein has been found in several human cancers [13][14]. Bcl-2 participates in an oncogenic function via survival signaling pathways and its role on the mitochondrial membrane. Several signaling molecules regulate Bcl-2. The phosphorylation of Bcl-2 on Ser70 through growth factor-activated protein kinases triggers the anti-apoptotic function of Bcl-2 [15][16]. p53 regulates Bcl-2 family proteins and hence controls mitochondria-mediated apoptosis [17]. p53 is a controller of the expression of the Bcl-2 gene. It increases Bcl-2 protein expression [18][19][20]. Overexpression of Bcl-2 showed more activation of signaling molecules in pancreatic cancer cells [21]. Signaling pathways play a major role in controlling the Bcl-2 family of proteins that initiates cell growth/survival via activation of Bcl-2 [22][23].

Here, this review article highlights the present findings on the role of Bcl-2 in the progression of various cancers and its significance as a therapeutic target. We also discussed a comprehensive study of Bcl-2 inhibitors as a therapeutic target for cancer therapy. This investigation summarizes the potential of Bcl-2-inhibitors in the treatment of cancer and novel plans to utilize these inhibitors in pre-clinical and clinical trial applications.

2. Bcl-2 Family Proteins Mediated Apoptosis

Apoptosis is the complex and fine-regulated appearance of programmed cell death. It takes part in vital functions such as embryogenesis, immunity, tissue development, and maintenance of homeostasis. Hence, excessive or insufficient cell death may lead to many pathological conditions like cancer [24][25]. Apoptosis is an important program of a living cell [24][26]. Two pathways initiate apoptosis, the extrinsic pathway and the intrinsic pathway ( Figure 1 ). The extrinsic pathway starts with the activation of particular death receptors and ligand receptors. These ligands contain Apo2L/TRAIL and CD95L/FasL, which bind with DR4/DR5 (TRAIL-R1/R2) and CD95/Fas [27]. The intrinsic pathway of apoptosis is primarily triggered via the Bcl-2 family proteins [28]. Bcl-2 family proteins participate in p53-mediated apoptosis, which interacts with the Bcl-2 family proteins, reduces the mitochondrial membrane permeabilization (MMP), and releases cyt-c that activates caspase-9 [29][30][31][32]. The cyt- c and Apaf-1 binding activate pro-caspase-9, forming an “apoptosome” [33]. Both pathways activate caspase-3 and follow the apoptosis pathway ( Figure 1 ) [34].

Figure 1. The intrinsic and extrinsic pathways of apoptosis. The two pathways that initiate apoptosis are the extrinsic (or death receptor initiated by ligand binding and subsequent activation of caspase-8) pathway and intrinsic pathway (driven by Bcl-2 family proteins, release cyt-c and activation of caspase-9) of apoptosis. Both pathways lead to a common apoptosis pathway by activating caspases-3, -6, and -7, triggering apoptosis. Bcl-2 family proteins are primarily localized to mitochondria and present on the ER and the perinuclear membrane in hematopoietic cells.

Bcl-2 family proteins include both pro-apoptotic (Bax, Bak, Bid, Bim, Bad, Bik, Puma, Noxa, etc.) and anti-apoptotic (Bcl-2, Bcl-xL , Bcl-w, Mcl-1, etc.) proteins [35][36]. Bcl-2 family proteins contribute to a common genetic region, the Bcl-2 homology (BH) domain, containing four conserved BH domains [32][37]. The Bcl-2 family proteins are categorized into anti-apoptotic proteins, pro-apoptotic multi-domain Bcl-2 proteins, and BH3-only proteins. The anti-apoptotic compose BH1-4 [38], while Bax and Bak compose BH1-3 and so-called ‘multi-domain’ pro-apoptotic proteins [38]. Bcl-2 family proteins were demonstrated to contribute to cancer progression [39][40]. All anti-apoptotic Bcl-2 proteins seem to play the role of oncoproteins, and pro-apoptotic proteins may have a role as tumor suppressors. These molecules perform important functions in cancer progression [41][42]. Further, the BH3 domain combines with the anti-apoptotic proteins, neutralizing them and initiating apoptosis [43]. The down-expression of pro-apoptotic proteins is linked with cancer progression [44]. Pro-apoptotic Bax was associated with oral cancer progression and drug resistance [45]. Anti-apoptotic Bcl-2 proteins play a major role in the progression of various neoplasms [46] and fuel rapid cell cycle regulation [47], angiogenesis, and regulation of gene expression [48]. Overexpression of anti-apoptotic proteins may drive carcinogenesis [49][50]. Anti-apoptotic Bcl-xL was involved in oral cancer progression as well as cisplatin resistance [51].

3. Discovery of Bcl-2

The oncogenic perspective on Bcl-2 was primarily proposed in 1988 by Reed et al. utilizing gene transfer technology [52]. Bcl-2 gene rearrangements were explained to be linked with reduced prognosis in large-cell non-Hodgkin’s lymphomas (NHL) [53]. The study of chromosomal rearrangements, which occur in human cancers, has been the most potent and exciting approach for analyzing the molecular systems that trigger tumor cell growth. The examination of the t(14:18) translocation breakpoint and the finding of the Bcl-2 gene had a unique impact on our thoughts on fundamental cell biology in malignant and normal cells [54][55][56]. Moreover, t(14:18) chromosomal translocations were observed via PCR in several normal individuals [57][58]. The Bcl-2 proto-oncogene was initially cloned from the t(14;18) translocation breakpoint in human follicular B cell lymphoma [3]. It is encoded by the Bcl-2 gene in the case of the human genome, and it is specified as an oncogene [56]. It was identified due to its involvement in t(14;18) chromosomal translocations examined in non-Hodgkin’s lymphomas [56]. In translocations t(14;18), the Bcl-2 gene is present on chromosome 18, which becomes combined with the immunoglobulin heavy-chain (IgH) locus present at chromosome 14.

Effects in transcriptional activation of Bcl-2 gene and the over-production of its protein occur in B-cells. Hence, Bcl-2 was expected to present another growth-inducing oncogene while it appeared to start the translocation breakpoint sequence. Bcl-2 was established to increase cell growth and obstruct cell death [3][59]. A study of experimental retrovirus-induced Bcl-2 overexpression in the mouse bone marrow cells showed that Bcl-2 over-expression alone could not be sufficient for tumorigenesis [3][59]. Increased Bcl-2 protein expression can certify the perseverance of a cell clone through the more active Bcl-2 gene, awaiting promotion of tumorigenic alterations or mutations appearing in the result. The introduction of the cDNA of Bcl-2 in IL-3-dependent lymphoid and myeloid cells endorsed cell growth in the absence of the cytokine by the cells being sustained in a G0 position [3][59]. Experiments among transgenic mice, which overproduced Bcl-2 in B-cells, have powerfully confirmed the theory that inhibition of apoptosis characterizes one of the crucial steps in the process of tumorigenesis credibly [60][61]. The results achieved with mouse models are similar to the detected impact of the expression of Bcl-2 in cancers. Chromosomal Bcl-2 translocations affecting overexpression of protein are observed in follicular center B-cell lymphoma and CLL. These cancers are benign and consist of primarily quiet, non-cycling cancer cells [62][63].

4. Bcl-2 Inhibitors

We summarize here the critical Bcl-2 inhibitors from published patents and literature ( Table 1 ). Bcl-2 inhibitors may inhibit cancer cell growth and survival ( Figure 2 ). These inhibitors were reported in the patents as well as literature databases.

Figure 2. Bcl-2-mediated apoptosis cascade and the mechanism of action of inhibitors. Over-expression of Bcl-2 might be blocked and inhibited through Bcl-2 inhibitors. Inhibitors block cancer progression by targeting signaling molecules or protein kinases that abrogate Bcl-2 expression and decrease the expression of Bcl-2 by inhibiting cell growth and proliferation and initiating apoptosis.
Table 1. Bcl-2 inhibitors are used for the therapeutic targeting of several cancers.
Agents Chemical Structure IC50 for Bcl-2 (μM) IC50 for Bcl-xL (μM) Used for Treatment Clinical Status References
G3139 Ijms 22 10442 i001 NA NA Solid Tumor (ST), SCLC, Melanoma
Leukemia, etc.
Phase 1 [64][65]
EGCG Ijms 22 10442 i002 0.45 0.59 HNSCC, OSCC, other cancers Phase 1/2 [66][67][68]
ABT-737 Ijms 22 10442 i003 0.12 0.064 HNSCC, ST, PC, Leukemia, etc. Phase 1/2 [66][69][70]
ABT-263 Ijms 22 10442 i004 NA NA ST, Haemato
logical malig-
nancies, SCLC
Phase 1/2 [71][72]
ABT-199 Ijms 22 10442 i005 0.1 NA ST, Breast cancer Approved for use in CLL [73][74]
TW-37 Ijms 22 10442 i006 NA NA HNSCC, Prostrate Cancer, PC, Phase 1/2 [75][76]
Gossypol Ijms 22 10442 i007 0.28–10 0.4–3.03 HNSCC, ST, PC Phase 1/2 [66][77]
GX15-070 (Obatoclax) Ijms 22 10442 i008 NA NA HNSCC, PC, ST,
NSCLC
Phase 1 [78][79]
HA14-1 Ijms 22 10442 i009 ~9 NA HNSCC, leukemia, lymphoma, colon cancer, etc. Pre-clinical [78][80][81]
Chelerythrine Ijms 22 10442 i010 ~10 ~10 HNSCC, ST, etc.   [66][82]
S55746 Ijms 22 10442 i011 NA NA Hematological tumor Phase 1 [83]

Epigallocatechin-3-gallate (EGCG) is the most abundant and closely examined polyphenol in green tea [84][85]. The chemotherapeutic effects of EGCG have been documented against various cancers [86][87][88]. EGCG alters and inhibits the Bcl-2 family protein ratio and activates caspases in cancer cells [89][90][91]. It has anticancer effects that increase Bax and Bak and decrease Bcl-xL and Bcl-2, initiating activation of caspases-9 and inducing cell death in SCC cancer cells [92][93]. EGCG leads to down-regulation of Bcl-2 and Bcl-xL [94]. The interaction of EGCG with p53 disrupts p53 with its regulatory E3 ligase MDM2. It reduces the ubiquitination of p53 through MDM2, since EGCG interrupts the binding of p53 for its regulator MDM2, stabilizing p53 through blocking p53 ubiquitination as well as degradation [95]. EGCG was documented from a library of some 2295 phytochemicals as an inhibitor of p53 with MDM2 interaction [96].

ABT-737 is a potent inhibitor of Bcl-2 and Bcl-xL [97], and its application in cancer therapeutics has been observed [69]. ABT-737 has its effect on activating caspase-3, leading to cell death. ABT-737 up-regulates pro-apoptotic Noxa expression via synergistic combination with chemotherapy [69]. ABT-737 binds with a very high affinity to Bcl-xL/Bcl-2 because of their similar structure [70]. Hence, Bcl-2 is inhibited due to the binding of ABT-737 in its hydrophobic groove, which relocates any bound pro-apoptotic BH3-containing proteins [70]. ABT-737 stimulated the caspase-3 activation as well as cleavage of PARP, which induced cell death. ABT-737 decreased the activation of Akt expression, which is involved in this signaling pathway in the inhibition of gastric cancer cell growth. p53 was observed to be linked to the effect of ABT-737 as well as naringenin in gastric cells [98]. Data showed ABT-737 and epothilone B caused a blockade of the signaling pathways that could synergistically stimulate apoptosis and anti-proliferation in human cancer cells [99].

ABT-199 (Venetoclax) is a promising and selective inhibitor of Bcl-2 protein, which has exhibited clinical efficacy in multiple hematological cancers [100]. ABT-199 is an inhibitor that exclusively maintains binding for Bcl-2 [101]. ABT-199 efficiently stimulates cell death in Bcl-2-dependent cancers without inducing thrombocytopenia. A single dose of ABT-199 stimulated cancer lysis syndrome in leukemia, showing potent anticancer action in vivo in humans. It is presently one of the most stimulating drugs for hematological cancers in clinical progress. Due to the important function of Bcl-2 in B-cells, clinical studies with ABT-199 presently concentrate exclusively on hematological cancers. A selective inhibitor of Bcl-2 can induce apoptosis. SCLC exhibits an elevated expression of Bcl-2 and can be liable to single-drug treatment by ABT-199. Through overexpression of Bcl-xL, ABT-199 showed pre-clinical trial activity in breast cancer cells [74] by investigation of the promise of ABT-199 as a Bcl-2 inhibitor.

References

  1. Kiyoshima, T.; Yoshida, H.; Wada, H.; Nagata, K.; Fujiwara, H.; Kihara, M.; Hasegawa, K.; Someya, H.; Sakai, H. Chemoresistance to concanamycin a1 in human oral squamous cell carcinoma is attenuated by an hdac inhibitor partly via suppression of bcl-2 expression. PLoS ONE 2013, 8, e80998.
  2. Gilormini, M.; Malesys, C.; Armandy, E.; Manas, P.; Guy, J.B.; Magné, N.; Rodriguez-Lafrasse, C.; Ardail, D. Preferential targeting of cancer stem cells in the radiosensitizing effect of abt-737 on hnscc. Oncotarget 2016, 7, 16731.
  3. Hockenbery, D.; Nuñez, G.; Milliman, C.; Schreiber, R.D.; Korsmeyer, S.J. Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 1990, 348, 334–336.
  4. Cory, S.; Adams, J.M. The bcl2 family: Regulators of the cellular life-or-death switch. Nat. Rev. Cancer 2002, 2, 647–656.
  5. Yin, X.M.; Oltvai, Z.N.; Korsmeyer, S.J. Bh1 and bh2 domains of bcl-2 are required for inhibition of apoptosis and heterodimerization with bax. Nature 1994, 369, 321–323.
  6. Nix, P.; Cawkwell, L.; Patmore, H.; Greenman, J.; Stafford, N. Bcl-2 expression predicts radiotherapy failure in laryngeal cancer. Br. J. Cancer 2005, 92, 2185–2189.
  7. Jackel, M.C.; Dorudian, M.A.; Marx, D.; Brinck, U.; Schauer, A.; Steiner, W. Spontaneous apoptosis in laryngeal squamous cell carcinoma is independent of bcl-2 and bax protein expression. Cancer 1999, 85, 591–599.
  8. Nor, J.E.; Christensen, J.; Liu, J.; Peters, M.; Mooney, D.J.; Strieter, R.M.; Polverini, P.J. Up-regulation of bcl-2 in microvascular endothelial cells enhances intratumoral angiogenesis and accelerates tumor growth. Cancer Res. 2001, 61, 2183–2188.
  9. Cho, H.Y.; Park, H.S.; Lin, Z.; Kim, I.; Joo, K.J.; Cheon, J. BCL6 gene mutations in transitional cell carcinomas. J. Int. Med. Res. 2007, 35, 224–230.
  10. Hong, J.; Kim, A.J.; Park, J.S.; Lee, S.H.; Lee, K.C.; Park, J.; Sym, S.J.; Cho, E.K.; Shin, D.B.; Lee, J.H. Additional rituximab-CHOP (R-CHOP) versus involved-field radiotherapy after a brief course of R-CHOP in limited, non-bulky diffuse large B-cell lymphoma: A retrospective analysis. Korean. J. Hematol. 2010, 45, 253–259.
  11. Yoshino, T.; Shiina, H.; Urakami, S.; Kikuno, N.; Yoneda, T.; Shigeno, K.; Igawa, M. BCL-2 expression as a predictive marker of hormone-refractory prostate cancer treated with taxane-based chemotherapy. Clin. Cancer Res. 2006, 12, 6116–6124.
  12. Perini, G.F.; Ribeiro, G.N.; Neto, J.V.P.; Campos, L.T.; Hamerschlak, N. BCL-2 as therapeutic target for hematological malignancies. J. Hematol. Oncol. 2018, 11, 1–15.
  13. Tsujimoto, Y. BCL-2 family of proteins: Life-or-death switch in mitochondria. Biosci. Rep. 2002, 22, 47–58.
  14. Hong, J.; Lee, Y.; Park, Y.; Kim, S.G.; Hwang, K.H.; Park, S.H.; Jeong, J.; Kim, K.H.; Ahn, J.Y.; Park, S.; et al. Role of FDG-PET/CT in detecting lymphomatous bone marrow involvement in patients with newly diagnosed diffuse large B-cell lymphoma. Ann. Hematol. 2012, 91, 687–695.
  15. Deng, X.; Ruvolo, P.; Carr, B.; May, W.S., Jr. Survival function of erk1/2 as il-3-activated, staurosporine-resistant bcl2 kinases. Proc. Natl. Acad. Sci. USA 2000, 97, 1578–1583.
  16. Mai, H.; May, W.S.; Gao, F.; Jin, Z.; Deng, X. A functional role for nicotine in BCL2 phosphorylation and suppression of apoptosis. J. Biol. Chem. 2003, 278, 1886–1891.
  17. Hong, J.; Park, S.; Park, J.; Jang, S.J.; Ahn, H.K.; Sym, S.J.; Cho, E.K.; Shin, D.B.; Lee, J.H. CD99 expression and newly diagnosed diffuse large B-cell lymphoma treated with rituximab-CHOP immunochemotherapy. Ann. Hematol. 2012, 91, 1897–1906.
  18. Miyashita, T.; Krajewski, S.; Krajewska, M.; Wang, H.G.; Lin, H.K.; Liebermann, D.A.; Hoffman, B.; Reed, J.C. Tumor suppressor p53 is a regulator of BCL-2 and bax gene expression in vitro and in vivo. Oncogene 1994, 9, 1799–1805.
  19. Miyashita, T.; Reed, J.C. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 1995, 80, 293–299.
  20. Sakuragi, N.; Salah-eldin, A.E.; Watari, H.; Itoh, T.; Inoue, S.; Moriuchi, T.; Fujimoto, S. Bax, BCL-2, and p53 expression in endometrial cancer. Gynecol. Oncol. 2002, 86, 288–296.
  21. Mortenson, M.M.; Schlieman, M.G.; Virudalchalam, S.; Bold, R.J. Overexpression of BCL-2 results in activation of the akt/nf-kb cell survival pathway. J. Surg. Res. 2003, 114, 302.
  22. Hong, J.; Park, S.; Park, J.; Kim, H.S.; Kim, K.H.; Ahn, J.Y.; Rim, M.Y.; Jung, M.; Sym, S.J.; Cho, E.K.; et al. Evaluation of prognostic values of clinical and histopathologic characteristics in diffuse large B-cell lymphoma treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone therapy. Leuk. Lymphoma 2011, 52, 1904–1912.
  23. Cook, S.J.; Stuart, K.; Gilley, R.; Sale, M.J. Control of cell death and mitochondrial fission by erk1/2 map kinase signalling. FEBS J. 2017, 284, 4177–4195.
  24. Lee, H.G.; Kim, S.Y.; Kim, I.; Kim, Y.K.; Kim, J.A.; Kim, Y.S.; Lee, H.S.; Park, J.; Kim, S.J.; Shim, H.; et al. Prediction of survival by applying current prognostic models in diffuse large B-cell lymphoma treated with R-CHOP followed by autologous transplantation. Blood. Res. 2015, 50, 160–166.
  25. Lee, J.; Kang, Y.J.; Ahn, J.; Song, S.H. Indolent B-Cell Lymphoid Malignancy in the Spleen of a Man Who Handled Benzene: Splenic Marginal Zone Lymphoma. Saf. Health Work 2017, 8, 315–317.
  26. Lee, S.H.; Kim, H.C.; Kim, Y.J. B-Cell Lymphoma in a Patient With a History of Foreign Body Injection. J. Craniofac. Surg. 2017, 28, 504–505.
  27. Lee, K.C.; Lee, S.H.; Sung, K.; Ahn, S.H.; Choi, J.; Lee, S.H.; Lee, J.H.; Hong, J.; Park, S.H. A Case of Primary Breast Diffuse Large B-Cell Lymphoma Treated with Chemotherapy Followed by Elective Field Radiation Therapy: A Brief Treatment Pattern Review from a Radiation Oncologist’s Point of View. Case. Rep. Oncol. Med. 2015, 2015, 907978.
  28. Lee, S.P.; Park, S.; Park, J.; Hong, J.; Ko, Y.H. Clinicopathologic characteristics of CD99-positive diffuse large B-cell lymphoma. Acta Haematol. 2011, 125, 167–174.
  29. Subedi, L.; Gaire, B.P.; Do, M.H.; Lee, T.H.; Kim, S.Y. Anti-neuroinflammatory and neuroprotective effects of the Lindera neesiana fruit in vitro. Phytomedicine 2016, 23, 872–881.
  30. Lai, D.; Visser-Grieve, S.; Yang, X. Tumour suppressor genes in chemotherapeutic drug response. Biosci. Rep. 2012, 32, 361–374.
  31. Oda, E.; Ohki, R.; Murasawa, H.; Nemoto, J.; Shibue, T.; Yamashita, T.; Tokino, T.; Taniguchi, T.; Tanaka, N. Noxa, a BH3-only member of the BCL-2 family and candidate mediator of p53-induced apoptosis. Science 2000, 288, 1053–1058.
  32. Lee, Y.; Hwang, K.H.; Hong, J.; Park, J.; Lee, J.H.; Ahn, J.Y.; Kim, J.H.; Lee, H.; Kim, S.G.; Shin, J.Y. Usefulness of (18)F-FDG PET/CT for the Evaluation of Bone Marrow Involvement in Patients with High-Grade Non-Hodgkin’s Lymphoma. Nucl. Med. Mol. Imaging 2012, 46, 269–277.
  33. Hong, J.; Yoon, H.H.; Ahn, H.K.; Sym, S.J.; Park, J.; Park, P.W.; Ahn, J.Y.; Park, S.; Cho, E.K.; Shin, D.B.; et al. Prognostic role of serum lactate dehydrogenase beyond initial diagnosis: A retrospective analysis of patients with diffuse large B cell lymphoma. Acta Haematol. 2013, 130, 305–311.
  34. Jang, H.R.; Song, M.K.; Chung, J.S.; Yang, D.H.; Lee, J.O.; Hong, J.; Cho, S.H.; Kim, S.J.; Shin, D.H.; Park, Y.J.; et al. Maximum standardized uptake value on positron emission tomography/computed tomography predicts clinical outcome in patients with relapsed or refractory diffuse large B-cell lymphoma. Blood Res. 2015, 50, 97–102.
  35. Lee, Y.S.; Jun, H.S. Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta-cells. Metabolism 2014, 63, 9–19.
  36. Jeong, E.K.; Jang, H.J.; Kim, S.S.; Oh, M.Y.; Lee, D.H.; Eom, D.W.; Kang, K.S.; Kwan, H.C.; Ham, J.Y.; Park, C.S.; et al. Protective effect of eupatilin against renal ischemia-reperfusion injury in mice. Transplant. Proc. 2015, 47, 757–762.
  37. Jung, M.Y.; Seo, C.S.; Baek, S.E.; Lee, J.; Shin, M.S.; Kang, K.S.; Lee, S.; Yoo, J.E. Analysis and Identification of Active Compounds from Gami-Soyosan Toxic to MCF-7 Human Breast Adenocarcinoma Cells. Biomolecules 2019, 9, 272.
  38. Reed, J.C. Pro-apoptotic multi-domain BCL-2/bax-family proteins: Mechanisms, physiological roles, and therapeutic opportunities. Cell Death Differ. 2006, 13, 1378–1386.
  39. Camisasca, D.R.; Honorato, J.; Bernardo, V.; da Silva, L.E.; da Fonseca, E.C.; de Faria, P.A.; Dias, F.L.; Lourenco Sde, Q. Expression of BCL-2 family proteins and associated clinicopathologic factors predict survival outcome in patients with oral squamous cell carcinoma. Oral Oncol. 2009, 45, 225–233.
  40. Strasser, A.; Huang, D.C.; Vaux, D.L. The role of the BCL-2/ced-9 gene family in cancer and general implications of defects in cell death control for tumourigenesis and resistance to chemotherapy. Biochim. Biophys. Acta 1997, 1333, F151–F178.
  41. Jordan, R.C.; Catzavelos, G.C.; Barrett, A.W.; Speight, P.M. Differential expression of BCL-2 and bax in squamous cell carcinomas of the oral cavity. Eur. J. Cancer Part B Oral Oncol. 1996, 32, 394–400.
  42. Xie, X.; Clausen, O.P.; de Angelis, P.; Boysen, M. The prognostic value of spontaneous apoptosis, bax, BCL-2, and p53 in oral squamous cell carcinoma of the tongue. Cancer 1999, 86, 913–920.
  43. Huang, D.C.; Strasser, A. BH3-only proteins-essential initiators of apoptotic cell death. Cell 2000, 103, 839–842.
  44. Lavieille, J.P.; Gazzeri, S.; Riva, C.; Reyt, E.; Brambilla, C.; Brambilla, E. P53 mutations and p53, waf-1, bax and BCL-2 expression in field cancerization of the head and neck. Anticancer Res. 1998, 18, 4741–4749.
  45. Kim, J.; Hong, J.; Kim, S.G.; Hwang, K.H.; Kim, M.; Ahn, H.K.; Sym, S.J.; Park, J.; Cho, E.K.; Shin, D.B.; et al. Prognostic Value of Metabolic Tumor Volume Estimated by (18) F-FDG Positron Emission Tomography/Computed Tomography in Patients with Diffuse Large B-Cell Lymphoma of Stage II or III Disease. Nucl. Med. Mol. Imaging 2014, 48, 187–195.
  46. Quinn, B.A.; Dash, R.; Azab, B.; Sarkar, S.; Das, S.K.; Kumar, S.; Oyesanya, R.A.; Dasgupta, S.; Dent, P.; Grant, S.; et al. Targeting mcl-1 for the therapy of cancer. Expert Opin. Investig. Drugs 2011, 20, 1397–1411.
  47. Quinn, L.M.; Richardson, H. Bcl-2 in cell cycle regulation. Cell Cycle 2004, 3, 7–9.
  48. Ma, C.; Zhang, J.; Durrin, L.K.; Lv, J.; Zhu, D.; Han, X.; Sun, Y. The BCL2 major breakpoint region (mbr) regulates gene expression. Oncogene 2007, 26, 2649–2657.
  49. Chipuk, J.E.; Moldoveanu, T.; Llambi, F.; Parsons, M.J.; Green, D.R. The BCL-2 family reunion. Mol. Cell 2010, 37, 299–310.
  50. Youle, R.J.; Strasser, A. The BCL-2 protein family: Opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008, 9, 47–59.
  51. Kim, S.S.; Jang, H.J.; Oh, M.Y.; Lee, J.H.; Kang, K.S. Tetrahydrocurcumin Enhances Islet Cell Function and Attenuates Apoptosis in Mouse Islets. Transplant. Proc. 2018, 50, 2847–2853.
  52. Reed, J.C.; Cuddy, M.; Slabiak, T.; Croce, C.M.; Nowell, P.C. Oncogenic potential of BCL-2 demonstrated by gene transfer. Nature 1988, 336, 259–261.
  53. Yunis, J.J.; Mayer, M.G.; Arnesen, M.A.; Aeppli, D.P.; Oken, M.M.; Frizzera, G. BCL-2 and other genomic alterations in the prognosis of large-cell lymphoma. N. Engl. J. Med. 1989, 320, 1047–1054.
  54. Bakhshi, A.; Jensen, J.P.; Goldman, P.; Wright, J.J.; McBride, O.W.; Epstein, A.L.; Korsmeyer, S.J. Cloning the chromosomal breakpoint of t(14;18) human lymphomas: Clustering around jh on chromosome 14 and near a transcriptional unit on 18. Cell 1985, 41, 899–906.
  55. Cleary, M.L.; Smith, S.D.; Sklar, J. Cloning and structural analysis of cdnas for BCL-2 and a hybrid BCL-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell 1986, 47, 19–28.
  56. Tsujimoto, Y.; Finger, L.R.; Yunis, J.; Nowell, P.C.; Croce, C.M. Cloning of the chromosome breakpoint of neoplastic b cells with the t(14;18) chromosome translocation. Science 1984, 226, 1097–1099.
  57. Limpens, J.; de Jong, D.; van Krieken, J.H.; Price, C.G.; Young, B.D.; van Ommen, G.J.; Kluin, P.M. BCL-2/jh rearrangements in benign lymphoid tissues with follicular hyperplasia. Oncogene 1991, 6, 2271–2276.
  58. Limpens, J.; Stad, R.; Vos, C.; de Vlaam, C.; de Jong, D.; van Ommen, G.J.; Schuuring, E.; Kluin, P.M. Lymphoma-associated translocation t(14;18) in blood b cells of normal individuals. Blood 1995, 85, 2528–2536.
  59. Vaux, D.L.; Cory, S.; Adams, J.M. BCL-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-b cells. Nature 1988, 335, 440–442.
  60. McDonnell, T.J.; Deane, N.; Platt, F.M.; Nunez, G.; Jaeger, U.; McKearn, J.P.; Korsmeyer, S.J. BCL-2-immunoglobulin transgenic mice demonstrate extended b cell survival and follicular lymphoproliferation. Cell 1989, 57, 79–88.
  61. McDonnell, T.J.; Korsmeyer, S.J. Progression from lymphoid hyperplasia to high-grade malignant lymphoma in mice transgenic for the t(14; 18). Nature 1991, 349, 254–256.
  62. Cleary, M.; Rosenberg, S.A. The BCL-2 gene, follicular lymphoma, and hodgkin’s disease. J. Natl. Cancer Inst. 1990, 82, 808–809.
  63. Horning, S.J.; Rosenberg, S.A. The natural history of initially untreated low-grade non-hodgkin’s lymphomas. N. Engl. J. Med. 1984, 311, 1471–1475.
  64. O’Brien, S.M.; Cunningham, C.C.; Golenkov, A.K.; Turkina, A.G.; Novick, S.C.; Rai, K.R. Phase I to II multicenter study of oblimersen sodium, a BCL-2 antisense oligonucleotide, in patients with advanced chronic lymphocytic leukemia. J. Clin. Oncol. 2005, 23, 7697–7702.
  65. Rudin, C.M.; Salgia, R.; Wang, X.; Hodgson, L.D.; Masters, G.A.; Green, M.; Vokes, E.E. Randomized phase II study of carboplatin and etoposide with or without the BCL-2 antisense oligonucleotide oblimersen for extensive-stage small-cell lung cancer: Calgb 30103. J. Clin. Oncol. 2008, 26, 870–876.
  66. Zhai, D.; Jin, C.; Satterthwait, A.C.; Reed, J.C. Comparison of chemical inhibitors of anti-apoptotic BCL-2-family proteins. Cell Death Differ. 2006, 13, 1419–1421.
  67. Masuda, M.; Suzui, M.; Weinstein, I.B. Effects of epigallocatechin-3-gallate on growth, epidermal growth factor receptor signaling pathways, gene expression, and chemosensitivity in human head and neck squamous cell carcinoma cell lines. Clin. Cancer Res. 2001, 7, 4220–4229.
  68. Leone, M.; Zhai, D.; Sareth, S.; Kitada, S.; Reed, J.C.; Pellecchia, M. Cancer prevention by tea polyphenols is linked to their direct inhibition of anti-apoptotic BCL-2-family proteins. Cancer Res. 2003, 63, 8118–8121.
  69. Li, R.; Zang, Y.; Li, C.; Patel, N.S.; Grandis, J.R.; Johnson, D.E. Abt-737 synergizes with chemotherapy to kill head and neck squamous cell carcinoma cells via a noxa-mediated pathway. Mol. Pharmacol. 2009, 75, 1231–1239.
  70. Vogler, M. Targeting BCL2-proteins for the treatment of solid tumours. Adv. Med. 2014, 2014, 943648.
  71. Roberts, A.W.; Seymour, J.F.; Brown, J.R.; Wierda, W.G.; Kipps, T.J.; Khaw, S.L.; Carney, D.A.; He, S.Z.; Huang, D.C.; Xiong, H.; et al. Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: Results of a phase i study of navitoclax in patients with relapsed or refractory disease. J. Clin. Oncol. 2012, 30, 488–496.
  72. Tse, C.; Shoemaker, A.R.; Adickes, J.; Anderson, M.G.; Chen, J.; Jin, S.; Johnson, E.F.; Marsh, K.C.; Mitten, M.J.; Nimmer, P.; et al. Abt-263: A potent and orally bioavailable BCL-2 family inhibitor. Cancer Res. 2008, 68, 3421–3428.
  73. Pan, R.; Hogdal, L.J.; Benito, J.M.; Bucci, D.; Han, L.; Borthakur, G.; Cortes, J.; DeAngelo, D.J.; Debose, L.; Mu, H. Selective Bcl-2 inhibition by abt-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov. 2014, 4, 362–375.
  74. Vaillant, F.; Merino, D.; Lee, L.; Breslin, K.; Pal, B.; Ritchie, M.E.; Smyth, G.K.; Christie, M.; Phillipson, L.J.; Burns, C.J.; et al. Targeting BCL-2 with the BH3 mimetic abt-199 in estrogen receptor-positive breast cancer. Cancer Cell 2013, 24, 120–129.
  75. Ashimori, N.; Zeitlin, B.D.; Zhang, Z.; Warner, K.; Turkienicz, I.M.; Spalding, A.C.; Teknos, T.N.; Wang, S.; Nor, J.E. Tw-37, a small-molecule inhibitor of BCL-2, mediates s-phase cell cycle arrest and suppresses head and neck tumor angiogenesis. Mol. Cancer Ther. 2009, 8, 893–903.
  76. Wang, Z.; Song, W.; Aboukameel, A.; Mohammad, M.; Wang, G.; Banerjee, S.; Kong, D.; Wang, S.; Sarkar, F.H.; Mohammad, R.M. Tw-37, a small-molecule inhibitor of BCL-2, inhibits cell growth and invasion in pancreatic cancer. Int. J. Cancer 2008, 123, 958–966.
  77. Oliver, C.L.; Bauer, J.A.; Wolter, K.G.; Ubell, M.L.; Narayan, A.; O’Connell, K.M.; Fisher, S.G.; Wang, S.; Wu, X.; Ji, M.; et al. In vitro effects of the BH3 mimetic, (−)-gossypol, on head and neck squamous cell carcinoma cells. Clin. Cancer Res. 2004, 10, 7757–7763.
  78. Chen, J.; Freeman, A.; Liu, J.; Dai, Q.; Lee, R.M. The apoptotic effect of ha14-1, a BCL-2-interacting small molecular compound, requires bax translocation and is enhanced by pk11195. Mol. Cancer Ther. 2002, 1, 961–967.
  79. Huang, S.; Okumura, K.; Sinicrope, F.A. BH3 mimetic obatoclax enhances trail-mediated apoptosis in human pancreatic cancer cells. Clin. Cancer Res. 2009, 15, 150–159.
  80. Wang, J.L.; Liu, D.; Zhang, Z.-J.; Shan, S.; Han, X.; Srinivasula, S.M.; Croce, C.M.; Alnemri, E.S.; Huang, Z. Structure-based discovery of an organic compound that binds BCL-2 protein and induces apoptosis of tumor cells. Proc. Natl. Acad. Sci. USA 2000, 97, 7124–7129.
  81. Sinicrope, F.A.; Penington, R.C.; Tang, X.M. Tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis is inhibited by BCL-2 but restored by the small molecule BCL-2 inhibitor, ha 14-1, in human colon cancer cells. Clin. Cancer Res. 2004, 10, 8284–8292.
  82. Hoffmann, T.K.; Leenen, K.; Hafner, D.; Balz, V.; Gerharz, C.D.; Grund, A.; Ballo, H.; Hauser, U.; Bier, H. Anti-tumor activity of protein kinase c inhibitors and cisplatin in human head and neck squamous cell carcinoma lines. Anticancer Drugs 2002, 13, 93–100.
  83. Casara, P.; Davidson, J.; Claperon, A.; Le Toumelin-Braizat, G.; Vogler, M.; Bruno, A.; Chanrion, M.; Lysiak-Auvity, G.; Le Diguarher, T.; Starck, J.B.; et al. S55746 is a novel orally active BCL-2 selective and potent inhibitor that impairs hematological tumor growth. Oncotarget 2018, 9, 20075–20088.
  84. Lambert, J.D.; Yang, C.S. Cancer chemopreventive activity and bioavailability of tea and tea polyphenols. Mutat. Res. 2003, 523, 201–208.
  85. Yang, C.S.; Lambert, J.D.; Ju, J.; Lu, G.; Sang, S. Tea and cancer prevention: Molecular mechanisms and human relevance. Toxicol. Appl. Pharmacol. 2007, 224, 265–273.
  86. Mukhtar, H.; Ahmad, N. Tea polyphenols: Prevention of cancer and optimizing health. Am. J. Clin. Nutr. 2000, 71, 1698S–1702S.
  87. Yang, C.S.; Maliakal, P.; Meng, X. Inhibition of carcinogenesis by tea. Annu. Rev. Pharmacol. Toxicol. 2002, 42, 25–54.
  88. Tang, S.N.; Singh, C.; Nall, D.; Meeker, D.; Shankar, S.; Srivastava, R.K. The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (egcg) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J. Mol. Signal. 2010, 5, 14.
  89. Adhami, V.M.; Ahmad, N.; Mukhtar, H. Molecular targets for green tea in prostate cancer prevention. J. Nutr. 2003, 133, 2417S–2424S.
  90. Chung, L.Y.; Cheung, T.C.; Kong, S.K.; Fung, K.P.; Choy, Y.M.; Chan, Z.Y.; Kwok, T.T. Induction of apoptosis by green tea catechins in human prostate cancer du145 cells. Life Sci. 2001, 68, 1207–1214.
  91. Amin, A.R.; Khuri, F.R.; Chen, Z.G.; Shin, D.M. Synergistic growth inhibition of squamous cell carcinoma of the head and neck by erlotinib and epigallocatechin-3-gallate: The role of p53-dependent inhibition of nuclear factor-kappab. Cancer Prev. Res. 2009, 2, 538–545.
  92. Shankar, S.; Ganapathy, S.; Hingorani, S.R.; Srivastava, R.K. Egcg inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer. Front. Biosci. 2008, 13, 440–452.
  93. Shankar, S.; Ganapathy, S.; Srivastava, R.K. Green tea polyphenols: Biology and therapeutic implications in cancer. Front. Biosci. 2007, 12, 4881–4899.
  94. Lin, H.Y.; Hou, S.C.; Chen, S.C.; Kao, M.C.; Yu, C.C.; Funayama, S.; Ho, C.T.; Way, T.D. (−)-Epigallocatechin gallate induces fas/cd95-mediated apoptosis through inhibiting constitutive and il-6-induced jak/stat3 signaling in head and neck squamous cell carcinoma cells. J. Agric. Food Chem. 2012, 60, 2480–2489.
  95. Zhao, J.; Blayney, A.; Liu, X.; Gandy, L.; Jin, W.; Yan, L.; Ha, J.H.; Canning, A.J.; Connelly, M.; Yang, C.; et al. Egcg binds intrinsically disordered n-terminal domain of p53 and disrupts p53-mdm2 interaction. Nat. Commun. 2021, 12, 986.
  96. Lee, H.G.; Choi, Y.; Kim, S.Y.; Kim, I.; Kim, Y.K.; Kim, Y.S.; Lee, H.S.; Kim, S.J.; Kim, J.A.; Park, B.B.; et al. R-CHOP chemoimmunotherapy followed by autologous transplantation for the treatment of diffuse large B-cell lymphoma. Blood. Res. 2014, 49, 107–114.
  97. Oltersdorf, T.; Elmore, S.W.; Shoemaker, A.R.; Armstrong, R.C.; Augeri, D.J.; Belli, B.A.; Bruncko, M.; Deckwerth, T.L.; Dinges, J.; Hajduk, P.J.; et al. An inhibitor of BCL-2 family proteins induces regression of solid tumours. Nature 2005, 435, 677–681.
  98. Zhang, H.; Zhong, X.; Zhang, X.; Shang, D.; Zhou, Y.I.; Zhang, C. Enhanced anticancer effect of abt-737 in combination with naringenin on gastric cancer cells. Exp. Ther. Med. 2016, 11, 669–673.
  99. Li, Y.L.; Sun, J.; Hu, X.; Pan, Y.N.; Yan, W.; Li, Q.Y.; Wang, F.; Lin, N.M.; Zhang, C. Epothilone b induces apoptosis and enhances apoptotic effects of abt-737 on human cancer cells via pi3k/akt/mtor pathway. J. Cancer Res. Clin. Oncol. 2016, 142, 2281–2289.
  100. Cang, S.; Iragavarapu, C.; Savooji, J.; Song, Y.; Liu, D. Abt-199 (venetoclax) and BCL-2 inhibitors in clinical development. J. Hematol. Oncol. 2015, 8, 129.
  101. Souers, A.J.; Leverson, J.D.; Boghaert, E.R.; Ackler, S.L.; Catron, N.D.; Chen, J.; Dayton, B.D.; Ding, H.; Enschede, S.H.; Fairbrother, W.J.; et al. Abt-199, a potent and selective BCL-2 inhibitor, achieves anti-tumor activity while sparing platelets. Nat. Med. 2013, 19, 202–208.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 718
Revisions: 3 times (View History)
Update Date: 12 Nov 2021
1000/1000