Anti-cancer peptides from bacteria

  • Tomasz M. KarpiÅ„ski Department of Medical Microbiology, PoznaÅ„ University of Medical Sciences, Poland;
  • Anna K. Szkaradkiewicz Department of Conservative Dentistry and Periodontology, PoznaÅ„ University of Medical Sciences, Poland
Keywords: anticancer peptides, bacteria, apoptosis, toxins, azurin, Entap
DOI: 10.3329/bjp.v8i3.15704

Abstract

Cancer is a leading cause of death in the world. The rapid development of medicine and pharmacology allows to create new and effective anticancer drugs. Among modern anticancer drugs are bacterial proteins. Until now has been shown anticancer activity among others azurin and exotoxin A from Pseudomonas aeruginosa, Pep27anal2 from Streptococcus pneumoniae, diphtheria toxin from Corynebacterium diphtheriae, and recently discovered Entap from Enterococcus sp. The study presents the current data regarding the properties, action and anticancer activity of listed peptides.

Introduction

Attempts to use bacteria or their products for the treatment of cancer dates back to turn of the XIX and XX century. William Coley (Coley, 1909) in the treatment of patients with unresectable tumors applied the treatment with bacterial culture supernatants of Streptococcus pyogenes and Serratia marcescens. This preparation called Coley's toxins was used in approximately 1200 patients with malignancy, often yielding regression of the tumor and in 30 patients, supposedly - a complete cure. Currently, it is assumed that the main factor responsible for therapeutic effect of Coley's toxins was induction of enhanced tumor necrosis factor-α (TNF-α) secretion in the body of the patient. The antitumor efficacy of TNF-α was confirmed in an animal model, observing the inhibition of growth and even complete regression of the tumor (Gratia and Linz, 1931; Carswell et al., 1975). Some studies have shown that the pathogenic microorganisms may proliferate inside hypoxic cancer lesions, simultaneously stimulating the immune system of the host during infection, leading to inhibition of cancer progression. An example would be a vaccine strain BCG (Mycobacterium bovis Calmette-Guerin), used in the treatment of superficial bladder cancer (Alexandroff et al., 1999) or attenuated Salmonella typhimurium (Pawelek et al., 1997), and anaerobic bacteria of the Clostridium genus (Dang et al., 2001) leading to enhanced tumor regression in mice. Currently, it is considered that the selected microbial infections can lead to activation of macrophages and lymphocytes, and result in the production of cytotoxic agents, especially TNF-α, having anticancer activity (Patyar et al., 2010). Currently, hope in the treatment of cancer relies, among other, on low molecular weight proteins, including those produced by microorganisms. In this paper, are presented basic anticancer peptides of bacterial origin. Selected characteristics of the below described peptides are presented in Tables I and II.

Table I: Origin, molecular weights and structure of described anticancer peptides. For the visualization of the peptides structures were used modelling servers: I-TASSER http://zhanglab.ccmb.med.umich.edu/I-TASSER/ and SWISS-MODEL http://swissmodel.expasy.org/ (Kiefer et al., 2009)

Peptide Origin Molecular weight Structure
Azurin Pseudomonas aeruginosa strains 14 kDa

128 amino acids

Exotoxin A Pseudomonas aeruginosa strains 66 kDa

638 amino acids

Diphtheria toxin Corynebacterium diphtheria strains 60 kDa

538 amino acids

Pep27anal2 Streptococcus pneumoniae strains 3.3 kDa

27 amino acids:

Entap Enterococus sp. strains 6.2 kDa 58-62 amino acids (the sequence remains to be studied)

Table II: Target tumor cells and mode of cytotoxic action of described anticancer peptides

Peptide Target tumor cells Mode of cytotoxic action
Azurin Breast cancer, melanoma, squamous carcinoma, reticulum cell sarcoma Deregulation of proliferation and induction of caspase-dependent apoptosis
Exotoxin A Pancreatic cancer, melanoma, head and neck squamous carcinoma, lung carcinoma, breast carcinoma, multiple myeloma Inhibition of protein synthesis (ADP-ribosylation of cytoplasmic elongation factor 2)
Diphtheria toxin T cell lymphoma, glioblastoma, malignant brain tumors, adrenocortical carcinoma Inhibition of protein synthesis (ADP-ribosylation of cytoplasmic elongation factor 2)
Pep27anal2 Leukemia, gastric cancer, breast cancer Cellular permeabilization, deregulation of proliferation and induction of caspase-independent apoptosis
Entap Gastric adenocarcinoma, uterine cervix adenocarcinoma, mammary gland adenocarcinoma, prostate carcinoma, colorectal adenocarcinoma Deregulation of proliferation and induction of autophagous apoptosis

Materials and Methods

Azurin

Azurin (14 kDa, 128 amino acids) is a copper-containing single-domain protein with a rigid β-barrel structure, produced by Pseudomonas aeruginosa. Azurin contains a hydrophobic patch (van de Kamp et al., 1990). Azurin is a member of the cupredoxin family of redox proteins (Adman, 1991). The copper in azurin can be eliminated, creating apo-azurin (Nar et al., 1991). The apo-azurin has a very low redox activity (0.02 and 0.01% of the holoenzyme wt azurin) but demonstrates significant cytotoxicity (Goto et al., 2003). Ability of azurin to destroy tumor cells appears to rely on the stability of the p53 protein which inhibits the development of the cancer. In nucleus azurin enhances the intracellular levels of p53 and Bax, what triggers the release of mitochondrial cytochrome c into the cytosol. This process activates the caspase cascade (including caspase-7 and caspase-9) initiating the apoptotic process (Punj et al., 2004). Apoptosis is an essential process in cellular development and in maintaining cellular homeostasis. Azurine much more effectively penetrates into the tumor cells than into the healthy. By this process is probably responsible domain Azu 50-70 (p28), consisting of 28 amino acids, and has molecular weight 2.8 kDa (Bernardes et al., 2013). p28 preferentially enter the human breast cancer cell lines MCF-7, ZR-75-1, and T47D through a caveolin-mediated pathway. p28 and similar peptides that reduce degradation of p53 may provide a unique series of cytostatic and cytotoxic chemotherapeutic agents (Yamada et al., 2009). Studies have shown direct antiangiogenic effect of p28 on endothelial cells. p28 preferentially penetrated human endothelial cells and inhibited VEGF- and bFGF-induced migration, capillary tube formation and neoangiogenesis (Mehta et al., 2011). This suggests that p28 is a unique agent possessing both antiangiogenic and cytostatic properties. Azurin binds to the NH2-terminal domain and DNA-binding domain (DBD) of p53, but not the COOH-terminal region of p53 (Punj et al., 2004; Apiyo and Wittung-Stafshede, 2005). The azurin:p53 complex is stable (Taranta et al., 2008). Vasu et al. showed that azurine acts strongly cytotoxic, with respect to the breast cancer MCF-7 cell line, causing more than 50% increase in intensity of apoptosis. However, in the breast cancer MDA-MB-157, MDD2 and MDA-MB-231 cells, the increase in intensity of apoptosis was 15-18% (Vasu et al., 2007). It has been shown that in p53 wild-type MCF-7 breast cancer cells, treatment with azurin caused an increase in p53 levels in both nuclear and cytoplasmic fractions (Punj et al., 2004; Yamada et al., 2004; Yamada et al., 2009). Yamada et al. conducted a study in which over 22 days in immunocompromised mice and transplanted human melanoma UISO-Mel-2 cells was azurine administered. It was found that in mice treated with azurine there was a reduction in tumor size compared to mice not treated and there have been no fatal cases (Yamada et al., 2002a; Yamada et al., 2002b). In other studies, the azurin-treated oral squamous carcinoma cells showed decreased viability, morphological changes, DNA breakage, and increase in p53 and cyclin B1 protein levels. Increase of oral squamous carcinoma cells sensitivity to anticancer drugs was found in combination treatment of azurin with other anticancer agents (Choi et al., 2011). Azurin induces a p53-mediated apoptosis also in mammalian J774 macrophage-like cells from reticulum cell sarcoma. A high level of reactive oxygen species, generated during treatment of macrophages with azurin, correlates with its cytotoxicity (Yamada et al., 2002b). The studies showed that intravenous administration of azurin p28 to mice showed its t(1/2 beta) 0.23 h, clearance 1.7 l/kg/h, and volume of distribution at steady state 4.1 l/kg (Gorman et al., 2010). p28 does not exhibit preclinical immunogenicity or toxicity. The no observed adverse effect level was 120 mg/kg iv in female mice. In non-human primates no observed adverse effect level of p28 was defined as the highest studied dose (120 mg/kg/dose; 1,440 mg/m2/dose) (Jia et al., 2011). p28 has undergone phase I clinical trial (http://www.cdgti.com; IND 77754).

Exotoxin A

Exotoxin A (66 kDa, 638 amino acids) is the main toxin secreted by Pseudomonas aeruginosa. Its operation is identical with diphtheria toxin. It has activity of ADP-ribosyl transferase. It inactivates elongation factor-2 (EF-2) resulting in inhibition of protein synthesis. Exotoxin A production is dependent on the presence of iron. Exotoxin A contains four functional domains: The receptor binding domain (Ia; aa 1–252), the translocation domain (II; aa 253–364), the domain Ib (aa 365–404), and the cytotoxic domain (aa 405–613) (Wolf and Elsässer-Beile, 2010). Exotoxin A is usually used as an immunotoxin, in combination with various ligands. Deimmunized Pseudomonas exotoxin cloned with human epidermal growth factor (EGF) and interleukin 4 had anti-cancer effect on orthotopic MIA PaCa-2 pancreatic cancer and selectively prevented metastasis (Oh et al., 2012). Exotoxin A-based immunotoxin, the 9.2.27PE, and the BH-3 mimetic compound ABT-737 resulted in synergistic cytotoxicity, and the melanoma cell lines death associated with apoptosis (Risberg et al., 2011). Exotoxin A cloned with an anti-CD133 scFv reactive (dCD133KDEL) specifically kills CD133+ tumor initiating cells and can arrest the proliferation of head and neck squamous carcinoma cells in vitro and in vivo (Waldron et al., 2011). A fusion protein composed of circularly permuted IL-4 and a truncated form of exotoxin (PE) [IL-4(38-37)-PE38KDEL] effectively kills most multidrug-resistant tumor cells including lung carcinoma, breast carcinoma, and multiple myeloma (de Jong et al., 2003).

Result and Discussion

Diphtheria toxin

Diphtheria toxin (60 kDa, 538 amino acids) is exotoxin secreted by Corynebacterium diphtheria strains, which have the capacity for its production as a result of infection with bacteriophage B. The gene that encodes diphtheria toxin (tox) is present in some corynephages, and diphtheria toxin is only produced by C. diphtheriae isolates that harbor tox+ phages (Holmes, 2000). The toxin is composed of subunits A and B. The A subunit terminates host cell protein synthesis by inactivating elongation factor-2 (EF-2) preventing cell protein synthesis, thus leading to cell death. The B subunit contains receptor-binding region and a translocation region (Murphy, 2011). Diphtheria toxin (DT) has shown anticancer activity in both experimental models and humans. However DT has also adverse effects, therefore, DT is used in anticancer therapy in conjunction with other substances. Denileukin diftitox (DD; Ontak) is a recombinant DNA-derived cytotoxic protein composed of the enzymatically active domain (fragment A and a portion of fragment B) of the diphtheria toxin, followed by sequences of human interleukin (IL)-2 (Bacha et al., 1998). This molecule is used in treatment of chronic T cell lymphoma (CTCL) expressing CD25. Currently denileukin diftitox is in phase III studies (Duvic et al., 2013). DTAT is DT-based immunotoxin targeting tumor vascular endothelium. In vitro, DTAT has shown a potent antitumor activity against uPAR-expressing glioblastoma cells (U118MG, U373MG, and U87MG) and human umbilical vein endothelial cells. In vivo, DTAT caused a significant regression of small U118MG cell-induced tumors in mice (Vallera et al., 2002). Tf-CRM107 is a conjugate of human transferrin (Tf) and a genetic mutant of diphtheria toxin (CRM107) that lacks native toxin binding. Tf-CRM107 is targeting malignant brain tumors. The studies showed potent antitumor activity in a phase I clinical trial, in which 60% of patients showed a >50% reduction in tumor volume (Laske et al., 1997). Cross-reacting material 197 (CRM197) is the nontoxic mutant of diphtheria toxin. CRM197 commonly acts as an immunological adjuvant, or as an inhibitor of heparin-binding epidermal growth factor. The studies showed, that CRM197 blocked growth, reduced angiogenesis and induced apoptosis in human adrenocortical carcinoma (Martarelli et al., 2009). The combination of CRM197 plus doxorubicin enhanced cytotoxicity in a T-cell acute lymphoblastic leukemia cell line (Kunami et al., 2011) and that the combination with cisplatin inhibited growth and led to apoptosis of glioma cells (Wang et al., 2012).

Pep27anal2

Pep27anal2, is an analogue of Pep27 (3.3-3.6 kDa, 27-30 amino acids), the signal peptide produced by Streptococcus pneumoniae. It initiates death program of S. pneumoniae, and also has antimicrobial properties (Sung et al., 2007). Lee et al. synthesized an analog of Pep27 peptide, called Pep27anal2, composed of 27 amino acids. Obtained analog Pep27anal2, proved to be more hydrophobic molecule compared to the parent peptide. Lee et al. using the MTT assay, demonstrated a reduction of proliferation of leukemia cells (AML-2, HL-60, Jurkat), gastric cancer cells (SNU-601) and breast cancer cells (MCF-7). At the same time it was found that Pep27anal2 penetrates the cell membrane, and then induces caspase-independent and cytochrome c-independent apoptosis. Authors suggest that an important role in membrane permeabilization and in antitumor activity plays a hydrophobicity of peptide (Lee et al., 2005). Huang et al. believe that the hydrophobicity of the peptides plays a crucial role in the mechanism of action against cancer cells, which should be taken into account in the design of potential anticancer peptides (Huang et al., 2011).

Entap

A new, recently discovered anticancer peptide is Entap - Enterococcal antiproliferative peptide (6.2 kDa), produced by clinical strains of Enterococcus genus bacteria (Szkaradkiewicz et al., 2012; Karpiński, 2012). In in vitro experiments strong antiproliferative activity of Entap was demonstrated on human neoplastic cells, including gastric adenocarcinoma cells (AGS), uterine cervix adenocarcinoma cells (HeLa), mammary gland adenocarcinoma (MDA-MB-231), prostate carcinoma (22Rv1) and colorectal adenocarcinoma (HT-29). Antiproliferative activity of Entap in cancer cells is manifested by inhibition of their cell cycle at the phase of G1 and by induction of autophagous apoptosis. In ultrastructural studies of cancer cells exposed to Entap traits of degradation and autophagous apoptosis were detected: cytoplasm contained numerous vacuoles and structures corresponsing to autophagosomes and autolysosomes. Entap manifests thermal stability and high content of hydrophobic amino acids, which determines unique character of its molecule (Karpiński, 2012; Karpiński et al., 2013).

Conclusion

A lot of antitumor peptides, including some of bacterial peptides are characterized by low molecular weight and hydrophobicity. These features appear to be important for the penetration stage of these peptides into tumor cells, the surface of which differs in some of the characteristics from normal cells. Bacterial peptides are a specific group of anticancer drugs, now widely studied. Some of these are in clinical development what gives us hope for their pharmacological use in cancers treatment.

References

Adman ET. Copper protein structures. Adv Protein Chem. 1991; 42: 145-97.

Alexandroff AB, Jackson AM, O’Donnell MA, James K. BCG immunotherapy of bladder cancer: 20 years on. Lancet 1999; 353: 1689-94.

Apiyo D, Wittung-Stafshede P. Unique complex between bacterial azurin and tumor-suppressor protein p53. Biochem Biophys Res Commun. 2005; 332: 965-68.

Bacha P, Williams DP, Waters C, Williams JM, Murphy JR, Strom TB. Interleukin 2 receptor-targeted cytotoxicity. Interleukin 2 receptor-mediated action of a diphtheria toxin-related interleukin 2 fusion protein. J Exp Med. 1988; 167: 612-22.

Bernardes N, Chakrabarty AM, Fialho AM. Engineering of bacterial strains and their products for cancer therapy. Appl Microbiol Biotechnol. 2013; 97: 5189-99.

Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 1975; 72: 3666-70.

Choi JH, Lee MH, Cho YJ, Park BS, Kim S, Kim GC. The bacterial protein Azurin enhances sensitivity of oral squamous carcinoma cells to anticancer drugs. Yonsei Med J. 2011; 52: 773-78.

Coley WB. The treatment of inoperable sarcoma by bacterial toxins (the mixed toxins of the Streptococcus erysipelatis and Bacillus prodigiosus). Proc R Soc Med Surg. 1909; 3: 1-48.

Dang LH, Bettegowda C, Huso DL, Kinzler KW, Vogelstein B. Combination bacteriolytic therapy for the treatment of experimental tumors. Proc Natl Acad Sci USA. 2001; 98: 15155-60.

De Jong MC, Scheffer GL, Broxterman HJ, Hooijberg JH, Slootstra JW, Meloen RH, Kreitman RJ, Husain SR, Joshi BH, Puri RK, Scheper RJ. Multidrug-resistant tumor cells remain sensitive to a recombinant interleukin-4-Pseudomonas exotoxin, except when overexpressing the multidrug resistance protein MRP1. Clin Cancer Res. 2003; 9: 5009-17.

Duvic M, Geskin L, Prince HM. Duration of response in cutaneous T-cell lymphoma patients treated with denileukin diftitox: Results from 3 Phase III studies. Clin Lymphoma Myeloma Leuk. 2013; 13: 377-84.

Gorman GS, Coward LU, Freeman L, Noker PE, Beattie CW, Jia L. A novel and rapid LC/MS/MS assay for bioanalysis of Azurin p28 in serum and its pharmacokinetics in mice. J Pharm Biomed Anal. 2010; 53: 991-96.

Goto M, Yamada T, Kimbara K, Horner J, Newcomb M, Gupta TK, Chakrabarty AM. Induction of apoptosis in macrophages by Pseudomonas aeruginosa azurin: Tumour-suppressor protein p53 and reactive oxygen species, but not redox activity, as critical elements in cytotoxicity. Mol Microbiol. 2003; 47: 549-59.

Gratia A, Linz R. Le phénoméne de Schwartzmann dans le sarcome du Cobaye. CR Séances Soc Biol Paris. 1931; 108: 427-28.

Holmes RK. Biology and molecular epidemiology of diphtheria toxin and the tox gene. J Infect Dis. 2000; 181: S156-67.

Huang YB, Wang XF, Wang HY, Liu Y, Chen Y. Studies on mechanism of action of anticancer peptides by modulation of hydrophobicity within a defined structural framework. Mol Cancer Ther. 2011; 10: 416-26.

Jia L, Gorman GS, Coward LU, Noker PE, McCormick D, Horn TL, Harder JB, Muzzio M, Prabhakar B, Ganesh B, Das Gupta TKD, Beattie CW. Preclinical pharmacokinetics, metabolism, and toxicity of azurin-p28 (NSC-745104) a peptide inhibitor of p53 ubiquitination. Cancer Chemother Pharmacol. 2011; 68: 513-24.

Karpiński TM, Szkaradkiewicz AK, Gamian A. New enterococcal anticancer peptide. 23rd European Congress of Clinical Microbiology and Infectious Diseases. Berlin, Germany, 27-30 April 2013. Program and Abstracts. 2013, p1661.

Karpiński TM. New peptide (Entap) with antiproliferative activity produced by bacteria of Enterococcus genus (in Polish). Habilitation thesis. Scientific Publisher of Poznań University of Medical Sciences, 2012.

Kiefer F, Arnold K, Künzli M, Bordoli L, Schwede T. The SWISS-MODEL Repository and associated resources. Nucleic Acids Res. 2009; 37: D387-92.

Kunami N, Yotsumoto F, Ishitsuka K, Fukami T, Odawara T, Manabe S, Ishikawa T, Tamura K, Kuroki M, Miyamoto S. Antitumor effects of CRM197, a specific inhibitor of HB-EGF, in T-cell acute lymphoblastic leukemia. Anticancer Res. 2011; 31: 2483-88.

Laske DW, Youle RJ, Oldfield EH. Tumor regression with regional distribution of the targeted toxin TF-CRM107 in patients with malignant brain tumors. Nat Med. 1997; 3: 1362-68.

Lee DG, Hahm KS, Park Y, Kim HY, Lee W, Lim SC, Seo YK, Choi CH. Functional and structural characteristics of anticancer peptide Pep27 analogues. Cancer Cell Int. 2005; 5: 21.

Martarelli D, Pompei P, Mazzoni G. Inhibition of adrenocortical carcinoma by diphtheria toxin mutant CRM197. Chemotherapy 2009; 55: 425-32.

Mehta RR, Yamada T, Taylor BN, Christov K, King ML, Majumdar D, Lekmine F, Tiruppathi C, Shilkaitis A, Bratescu L, Green A, Beattie CW, Das Gupta TK. A cell penetrating peptide derived from azurin inhibits angiogenesis and tumor growth by inhibiting phosphorylation of VEGFR-2, FAK and Akt. Angiogenesis 2011; 14: 355-69.

Murphy JR. Mechanism of diphtheria toxin catalytic domain delivery to the eukaryotic cell cytosol and the cellular factors that directly participate in the process. Toxins (Basel). 2011; 3: 294-308.

Nar H1, Messerschmidt A, Huber R, van de Kamp M, Canters GW. Crystal structure of Pseudomonas aeruginosa apo-azurin at 1.85 Ã… resolution. FEBS Lett. 1992; 306: 119-24.

Oh S, Todhunter DA, Panoskaltsis-Mortari A, Buchsbaum DJ, Toma S, Vallera DA. A deimmunized bispecific ligand-directed toxin that shows an impressive anti-pancreatic cancer effect in a systemic nude mouse orthotopic model. Pancreas 2012; 41: 789-96.

Patyar S, Joshi R, Byrav DS, Prakash A, Medhi B, Das BK. Bacteria in cancer therapy: A novel experimental strategy. J Biomed Sci. 2010; 17: 21.

Pawelek JM, Low KB, Bermudes D. Tumor-targeted Salmonella as a novel anticancer vector. Cancer Res. 1997; 57: 4537-44.

Punj V, Bhattacharyya S, Saint-Dic D, Vasu C, Cunningham EA, Graves J, Yamada T, Constantinou AI, Christov K, White B, Li G, Majumdar D, Chakrabarty AM, Das Gupta TK. Bacterial cupredoxin azurin as an inducer of apoptosis and regression in human breast cancer. Oncogene 2004; 23: 2367-78.

Punj V, Bhattacharyya S, Saint-Dic D, Vasu C, Cunningham EA, Graves J, Yamada T, Constantinou AI, Christov K, White B, Li G, Majumdar D, Chakrabarty AM, Das Gupta TK. Bacterial cupredoxin azurin as an inducer of apoptosis and regression in human breast cancer. Oncogene 2004; 23: 2367-78.

Risberg K, Fodstad O, Andersson Y. Synergistic anticancer effects of the 9.2.27PE immunotoxin and ABT-737 in melanoma. PLoS One. 2011; 6: e24012.

Sung WS, Park Y, Choi CH, Hahm KS, Lee DG. Mode of antibacterial action of a signal peptide, Pep27 from Streptococcus pneumoniae. Biochem Biophys Res Commun. 2007; 363: 806-10.

Szkaradkiewicz A, Karpiński TM, Gamian A, Brzozowska E, Zeidler A. Identification of carcinoma suppressive factor produced by bacteria of Enterococcus genus. Clin Microbiol Infect. 2012; 18: 1366.

Taranta M, Bizzarri AR, Cannistraro S. Probing the interaction between p53 and the bacterial protein azurin by single molecule force spectroscopy. J Mol Recognit. 2008; 21: 63-70.

Vallera DA, Li C, Jin N, Panoskaltsis-Mortari A, Hall WA. Targeting urokinase-type plasminogen activator receptor on human glioblastoma tumors with diphtheria toxin fusion protein DTAT. J Natl Cancer Inst. 2002; 94: 597-606.

van de Kamp M, Silvestrini MC, Brunori M, Van Beeumen J, Hali FC, Canters GW. Involvement of the hydrophobic patch of azurin in the electron-transfer reactions with cytochrome c551 and nitrite reductase. Eur J Biochem. 1990; 194: 109-18.

Waldron NN, Kaufman DS, Oh S, Inde Z, Hexum MK, Ohlfest JR, Vallera DA. Targeting tumor-initiating cancer cells with dCD133KDEL shows impressive tumor reductions in a xenotransplant model of human head and neck cancer. Mol Cancer Ther. 2011; 10: 1829-38.

Wang L, Wang P, Liu Y, Xue Y. Regulation of cellular growth, apoptosis, and Akt activity in human U251 glioma cells by a combination of cisplatin with CRM197. Anticancer Drugs. 2012; 23: 81-89.

Wolf P, Elsasser-Beile U. Pseudomonas exotoxin A-based immunotoxins for targeted cancer therapy. In: Emerging cancer therapy: Microbial approaches and biotechnological tools. Fialho AM, Chakrabarty AM (eds). John Wiley and Sons, 2010.

Yamada T, Goto M, Punj V, Zaborina O, Chen ML, Kimbara K, Majumdar D, Cunningham E, Das Gupta TK, Chakrabarty AM. Bacterial redox protein azurin, tumor suppressor protein p53, and regression of cancer. Proc Natl Acad Sci U S A. 2002a; 99: 14098-103.

Yamada T, Goto M, Punj V, Zaborina O, Kimbara K, Das Gupta TK, Chakrabarty AM. The bacterial redox protein azurin induces apoptosis in J774 macrophages through complex formation and stabilization of the tumor suppressor protein p53. Infect Immun. 2002b; 70: 7054-62.

Yamada T, Hiraoka Y, Ikehata M, Kimbara K, Avner BS, Das Gupta TK, Chakrabarty AM. Apoptosis or growth arrest: Modulation of tumor suppressor p53's specificity by bacterial redox protein azurin. Proc Natl Acad Sci USA. 2004; 101: 4770-75.

Yamada T, Mehta RR, Lekmine F, Christov K, King ML, Majumdar D, Shilkaitis A, Green A, Bratescu L, Beattie CW, Das Gupta TK. A peptide fragment of azurin induces a p53-mediated cell cycle arrest in human breast cancer cells. Mol Cancer Ther. 2009; 8: 2947-58.

Published
2013-08-17

Apply citation style format of Bangladesh Journal of Pharmacology

Section
Mini Review
Financial Support
Self-funded
Conflict of Interest
Authors declare no conflict of interest