Topics in Anti-Cancer Research: Volume 10 -  - E-Book

Topics in Anti-Cancer Research: Volume 10 E-Book

0,0
72,24 €

-100%
Sammeln Sie Punkte in unserem Gutscheinprogramm und kaufen Sie E-Books und Hörbücher mit bis zu 100% Rabatt.
Mehr erfahren.
Beschreibung

Topics in Anti-Cancer Research covers new developments in the field of cancer diagnosis and drug therapy. Novel drugs as anticancer agents include natural and synthetic phenazirines and other anti-cancer compounds. The series also covers information on the current understanding of the pathology and molecular biology of specific neoplasms.
The diversity of research topics published in this book series give broad and valuable perspectives for cancer researchers, clinicians, cancer professionals aiming to develop novel anti-cancer targets and patents for the treatment of various cancers.
The topics covered in this volume are:
- Peptides can play a major role in combating cancer diseases
- Studying of the CLL after treatment using fractal parameter of neoplastic lymphocytes detection (ΛNLD)
- Mechanistic insight of rhenium-based compounds as anti- cancer agents
- Targeting cancer-specific inflammatory components in cancer therapeutics
- Marine natural products as a source of novel anticancer agents: a treasure from the ocean
- PDX clinical trial design in anti-cancer research

Das E-Book können Sie in Legimi-Apps oder einer beliebigen App lesen, die das folgende Format unterstützen:

EPUB

Seitenzahl: 296

Bewertungen
0,0
0
0
0
0
0
Mehr Informationen
Mehr Informationen
Legimi prüft nicht, ob Rezensionen von Nutzern stammen, die den betreffenden Titel tatsächlich gekauft oder gelesen/gehört haben. Wir entfernen aber gefälschte Rezensionen.



Table of Contents
BENTHAM SCIENCE PUBLISHERS LTD.
End User License Agreement (for non-institutional, personal use)
Usage Rules:
Disclaimer:
Limitation of Liability:
General:
PREFACE
List of Contributors
Peptides Can Play a Major Role in Combating Cancer Diseases
Abstract
INTRODUCTION
Cancer Cells versus Normal Cells
Cell Membrane and Non-Membrane Involved Anti-Cancer Mechanisms of Peptides
Cell Membrane Involved Mechanism of Anticancer Peptides
Non-Membrane Involved Mechanisms of Anticancer Peptides
Anticancer Peptides Categories
Antimicrobial Peptides
Anti-inflammatory Peptides
Antioxidant Peptides
Peptide Conjugates
Peptides as Targeting Probes (Homing Peptides) for Cancer Detection
Summary
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Studying of the CLL After Treatment Using Fractal Parameter of Neoplastic Lymphocytes Detection (λnld)
Abstract
INTRODUCTION
Leukemia Types
Diagnosis
Blood Tests for Diagnosis
Image Processing for Diagnoses
Image Acquisition
Image Segmentation
Nucleus Segmentation
Features
Classification
Treatment
METHODS AND MATERIALS
The Fractal Dimension of Blood Lymphocytes
RESULT
Fractal Parameters of Neoplastic Lymphocytes Detection (λnld)
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Mechanistic Insight of Rhenium-Based Compounds as Anti-Cancer Agents
Abstract
INTRODUCTION
RHENIUM BASED DRUGS IN CANCER IMAGING AND CANCER THERAPY
MOLECULAR INSIGHTS OF ANTICANCER POTENTIAL OF RHENIUM-BASED COMPOUNDS
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEGEMENTS
REFERENCE
Targeting Cancer-Specific Inflammatory Components In Cancer Therapeutics
Abstract
INTRODUCTION
HISTORICAL PROSPECTIVE OF CANCER TREATMENT
Molecular basis of cancer and mechanism of cancer resistance
Deregulation of NF-κB Pathway
Classical/Canonical Pathway
Alternative/non-canonical Pathway
Deregulation of PI3K/Akt Pathway
Cancer Related Inflammation (CRI)
Deciphering the molecular targets of CRI for targeted cancer therapy
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Marine Natural Products as a Source of Novel Anticancer Agents: A Treasure from the Ocean
Abstract
INTRODUCTION
MARINE-DERIVED NATURAL PRODUCTS AND THEIR CLASSIFICATION
MARINE NATURAL PRODUCTS AS AN ANTICANCER AGENT
Marine Herbs
Marine Algae
Marine Sponges
Marine Fungi
Marine Bacteria
Marine Coral Reefs
Marine Carrageenan Polysaccharides
Marine Seaweeds
Marine Ascidiaceans
Marine Diatoms
Marine Actinomycetes
Marine-Derived Drugs in Clinical Trials
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
PDX Clinical Trial Design in Anti-Cancer Research
Abstract
INTRODUCTION
RATIONALE FOR PRECLINICAL PDX-BASED STUDIES
METHODS FOR DEVELOPMENT OF PDX MODELS
Choice of Proper Animal for PDX Development
Single-Gene Mutation Mouse Strains
Nude mice (Athymic mice)
Beige Mice
Beta2-microglobulin (β2m)-deficient Strain
Non-Obese Diabetic (NOD) Mice
Severe Combined Immunodeficient (SCID) Mice
Rag-deficient Strains
Perforin-deficient Strain
IL2Rγ-deficient Strain
JAK3-deficient Strain
Multi-Gene Mutation Mouse Strains
SCID-Beige Strain
NOD-SCID (NOD.Cg-Prkdcscid) Strain
NOD-Rag1null Strain
NOD-Rag2null Strain
NOD-SCID β2mnull Strain
NOD-Rag1null Pfpnull Strain
NOG (NOD/Shi-scid IL2Rgnull) Strain
NSG (NOD/LtSz-scid IL2Rgnull) Strain
NOJ (NOD-SCID Jak3null) Strain
NRG (NOD-Rag1null IL2Rgnull) Strain
NSG-β2mnull Strain
NSS (NOD.Cg-PrkdcscidTg (CMV-IL3,CSF2,KITLG)) Strain
NSGS (NOD/LtSz-scid IL2Rgnull-3/GM/SF) strain
NOG-EXL (NOD/Shi-scid IL2Rgnull Tg(SV40/HTLV-IL3,CSF2))
BRJ (BALB/c-Rag2null Jak3null) Strain
Nude-RJ (Nude-Rag2null Jak3null) Strain
General Procedure of PDX Development
Step I: Tissue Procurement and Shipping
Tumor Tissue Collection in Hematologic Malignancies
Tumor Tissue Collection from Solid Tumors
(A). Core Needle Biopsy
(B). Resected Tumor Sampling
(C). Neoplastic Effusions Sampling
Step II: Tissue Preparation (Processing)
Graft Preparation of Hematologic Malignancies
Graft Preparation of Solid Tumors
Tumor Fragment Preparation
Single-cell Suspension Preparation
Step III: Tumor Implantation
Implantation of Hematologic Tumor Grafts
Implantation of Solid Tumor Grafts
Heterotopic Implantation
Orthotopic Implantation
Breast Cancer
Lung Cancer
Liver Tumors
Pancreatic Cancer
Colon Cancer
Ovarian Cancer
Cervical Cancer
Renal Tumors
Prostate Cancer
Melanoma
Brain Tumors
Hints on Some PDX Models
CHARACTERIZATION AND VALIDATION OF PDX MODELS
Clinicodemographic Features of the Submitter Patient
Histopathologic Background of the Primary Tumor
Quality Assurance of the PDX Tumor
Pathogen Contamination Assessments
Propagability or Tumor Regrowth Profile
Establishment of a Well-Characterized PDX Biorepository/Biobank
PDX-BASED TRIAL DESIGN
Methodologic Considerations of PDX Clinical Trials
Sample Size Estimation
Model Establishment
Treatment plan
Staging Day
Route of Administration
Treatment Dose
Dosing Interval
Treatment Duration
Outcome Measures
Ethical Considerations
LIMITATIONS OF PDX-BASED STUDIES
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Patents Book Series
“Topics in Anti-Cancer Research”
(Volume 10)
Edited by
Atta-ur-Rahman, FRS
Kings College,
University of Cambridge,
Cambridge,
UK

BENTHAM SCIENCE PUBLISHERS LTD.

End User License Agreement (for non-institutional, personal use)

This is an agreement between you and Bentham Science Publishers Ltd. Please read this License Agreement carefully before using the ebook/echapter/ejournal (“Work”). Your use of the Work constitutes your agreement to the terms and conditions set forth in this License Agreement. If you do not agree to these terms and conditions then you should not use the Work.

Bentham Science Publishers agrees to grant you a non-exclusive, non-transferable limited license to use the Work subject to and in accordance with the following terms and conditions. This License Agreement is for non-library, personal use only. For a library / institutional / multi user license in respect of the Work, please contact: [email protected].

Usage Rules:

All rights reserved: The Work is the subject of copyright and Bentham Science Publishers either owns the Work (and the copyright in it) or is licensed to distribute the Work. You shall not copy, reproduce, modify, remove, delete, augment, add to, publish, transmit, sell, resell, create derivative works from, or in any way exploit the Work or make the Work available for others to do any of the same, in any form or by any means, in whole or in part, in each case without the prior written permission of Bentham Science Publishers, unless stated otherwise in this License Agreement.You may download a copy of the Work on one occasion to one personal computer (including tablet, laptop, desktop, or other such devices). You may make one back-up copy of the Work to avoid losing it.The unauthorised use or distribution of copyrighted or other proprietary content is illegal and could subject you to liability for substantial money damages. You will be liable for any damage resulting from your misuse of the Work or any violation of this License Agreement, including any infringement by you of copyrights or proprietary rights.

Disclaimer:

Bentham Science Publishers does not guarantee that the information in the Work is error-free, or warrant that it will meet your requirements or that access to the Work will be uninterrupted or error-free. The Work is provided "as is" without warranty of any kind, either express or implied or statutory, including, without limitation, implied warranties of merchantability and fitness for a particular purpose. The entire risk as to the results and performance of the Work is assumed by you. No responsibility is assumed by Bentham Science Publishers, its staff, editors and/or authors for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products instruction, advertisements or ideas contained in the Work.

Limitation of Liability:

In no event will Bentham Science Publishers, its staff, editors and/or authors, be liable for any damages, including, without limitation, special, incidental and/or consequential damages and/or damages for lost data and/or profits arising out of (whether directly or indirectly) the use or inability to use the Work. The entire liability of Bentham Science Publishers shall be limited to the amount actually paid by you for the Work.

General:

Any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims) will be governed by and construed in accordance with the laws of Singapore. Each party agrees that the courts of the state of Singapore shall have exclusive jurisdiction to settle any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims).Your rights under this License Agreement will automatically terminate without notice and without the need for a court order if at any point you breach any terms of this License Agreement. In no event will any delay or failure by Bentham Science Publishers in enforcing your compliance with this License Agreement constitute a waiver of any of its rights.You acknowledge that you have read this License Agreement, and agree to be bound by its terms and conditions. To the extent that any other terms and conditions presented on any website of Bentham Science Publishers conflict with, or are inconsistent with, the terms and conditions set out in this License Agreement, you acknowledge that the terms and conditions set out in this License Agreement shall prevail.

Bentham Science Publishers Pte. Ltd. 80 Robinson Road #02-00 Singapore 068898 Singapore Email: [email protected]

PREFACE

The tenth volume of Topics in Anti-Cancer Research covers new developments in the field of cancer. It comprises six comprehensive chapters covering exciting contributions in frontier areas of anti-cancer research.

Tehrani, in chapter 1, discusses the mechanism of anticancer activity of the peptides that can play a major role in combating cancer diseases. Rezaei and Rostami in chapter 2 of the book, present an appropriate model for investigating the possibility of chronic lymphocytic leukemia (CLL) control using fractal parameter. Veena et al. discuss the mechanistic insight of rhenium-based compounds as anti-cancer agents in the next chapter of the book. Veena et al., in the fourth chapter of the book, provide insights on targeting cancer-specific inflammatory components in cancer therapeutics. Mandlik and Mandlik in the fifth chapter, discuss the anticancer potential of marine natural products in a diversity of flora and fauna, as well as their probable mechanisms of action. In the last chapter, Kajbafzadeh et al. address patient-derived xenograft (PDX) clinical trial designs in anti-cancer research.

I am thankful to the authors for their excellent contributions and to the reviewers for their in-depth and comprehensive comments for the improvement of the chapters. I am also grateful to Mr. Mahmood Alam (Editorial Director), Mr. Obaid Sadiq (Incharge Books Department), Ms. Asma Ahmed (Senior Manager Publications) and other colleagues for their support and assistance in the finalization of this volume.

Atta-ur-Rahman, FRS Kings College University of Cambridge Cambridge UK

List of Contributors

A. HarikrishnanDepartment of Chemistry, School of Arts and Sciences, Vinayaka Mission Research Foundation-Aarupadai Veedu (VMRF-AV) Campus, Paiyanoor, Chennai-603104, Tamil Nadu, IndiaAmir Arsalan KhorsandPDX Core Laboratory, Gene Therapy Research Center, Digestive Diseases Research Institute Tehran University of Medical Sciences, Tehran, Iran PDX Platform, Biomarker Evaluation and Supervision Team for Personalized Medicine, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, IranAbdol-Mohammad KajbafzadehPDX Platform, Biomarker Evaluation and Supervision Team for Personalized Medicine, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, IranDeepa S. MandlikBharati Vidyapeeth (Deemed to be) University, Poona College of Pharmacy, Erandawane, Pune Maharashtra, IndiaMohammad Hassan Houshdar TehraniSchool of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, IranMohammad Reza RezaeiDepartment of Nuclear Engineering, Faculty of Sciences and Modern Technologies, Graduate University of Advanced Technology, Kerman, IranPravallika MandlipalliDepartment of Biotechnology, School of Applied Sciences, REVA University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka Post, Bengaluru – 560 064, Karnataka State, IndiaSatish K. MandlikBharati Vidyapeeth (Deemed to be) University, Poona College of Pharmacy, Erandawane, Pune Maharashtra, IndiaSamad MuhammadnejadPDX Core Laboratory, Gene Therapy Research Center, Digestive Diseases Research Institute Tehran University of Medical Sciences, Tehran, Iran PDX Platform, Biomarker Evaluation and Supervision Team for Personalized Medicine, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, IranSeyed Mostafa MonzaviPDX Core Laboratory, Gene Therapy Research Center, Digestive Diseases Research Institute Tehran University of Medical Sciences, Tehran, Iran Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, IranSupreetha B.S.Department of Biotechnology, School of Applied Sciences, REVA University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka Post, Bengaluru – 560 064, Karnataka State, IndiaVeena V.Department of Biotechnology, School of Applied Sciences, REVA University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka Post, Bengaluru – 560 064, Karnataka State, IndiaZahra RostamiShahid Beheshti University of Medical Science, Tehran, Iran

Peptides Can Play a Major Role in Combating Cancer Diseases

Mohammad Hassan Houshdar Tehrani1,*
1 School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran

Abstract

Cancer diseases affecting many organs of human body have caused a major concern among the people all over the world. The conventional anticancer drugs, although have given some relief in the patient conditions, still cannot provide reliable treatment. Moreover, these drugs produce side effects in patients and in the worse cases, the problem of rising resistance phenomena against such drugs gradually put the patients’ lives even in more serious situation. Therefore, identifying and introducing compounds with new identities to produce effective treatment with low side effects are highly demanded. Small peptides with anticancer activity have been shown to fulfill this demand. Peptides, with naturally or synthetic origin, have several advantages over common drug molecules such as low toxicity, low immunogenicity, amenable to several changes in their sequences and thus giving various homologues or analogues. Moreover, peptides in conjugation with heterocyclic active compounds and/or known anticancer drugs may result in molecules with new identities which show both benefits of individual components within their unit structures. In this regard, peptide conjugates may play a role, not only as anticancer agents but also as cell-membrane penetrating and/ or cell targeting agents to help direct cancerous tissue internalization of the known anticancer agents, and so, preventing or lowering the incidence of side effects of the anticancer drugs on healthy tissues. In this chapter on the basis of several experiments, information about various peptide categories, their analogues and conjugation with other bioactive compounds is given. The discussion is focused on the anticancer activity of peptides, those primarily known for other biological activities. Understanding the cause of these activities may help to find out and make clearer the mechanism of anticancer activity of the peptides.

Keywords: Anticancer, Bioactive compounds, Cell-membrane penetrating, Cell targeting agents, Peptides, Peptide conjugation.
*Corresponding author Mohammad Hassan Houshdar Tehrani: School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; E-mail: [email protected]

INTRODUCTION

Cancer is defined as an uncontrollable growth and division of cells involving nearly every part ofthe body. The cancerous cells can even migrate and invade

other organ tissues, which cause a phenomenon called metastasis [1]. Cancer metastasis involving many organs eventually leads patients to face death.

According to the World Health Organization (WHO) reports, it was estimated that 9.6 million deaths due to cancer occurred in 2018 and the incidence of cancer involved 18.1 million new cases worldwide and the cases will rise up to 29.5 million by 2040 [2]. In the world, 1 in 6 deaths occurs due to cancer [3]. Cancer can also cause enormous economic pressure along with immense social and emotional stress on patients and their families, especially in underdeveloped or developing countries with low-income people [4]. Cancer treatment with the conventional chemotherapy although shows effectiveness but presents side effects in healthy tissues of the body. The other important drawback of the chemotherapy regimen is the occurrence of drug resistance among cancer variants [5]. Peptides nowadays have been considered for application in many diseases and abnormal conditions including, infection, pain, inflammation, immune problems, diabetes and hypertension [6, 7]. Application of peptides has also been suggested in various cancers [8]. Compared with the conventional drugs, peptides have many advantages such as easily preparation, low toxicity and immunogenicity content, easily amenable to several changes in their sequences, good biocompatibility, high tissue penetration and low probability of raising resistance [8, 9]. The main disadvantage of peptides is their low stability against enzymatic lysis in gastrointestinal (GI) tract when they are administered for oral application. However, this unfavorable property of the peptides can be improved by employing several ways including peptide cyclization [10, 11], rearrangement of amino acid residues in the peptide chains [12], L-amino acids exchange by D- congeners [13, 14] and N- or C- terminal capping of the peptides in order to be stable against aminopeptidase or carboxypeptidase enzymes [10]. The N- or C- terminal modification of the peptides by different bioactive molecules may also be employed for several purposes or diseases, where such peptides are considered as conjugated peptides [15, 16] or hybrid peptides [10]. Peptides can be used not only as bioactive molecules by their own, but also as carrier of other bioactive agents for enhancing entrance into the target cells (thus, such peptides are so-called cell penetrating peptides, CPP) or targeting specific body organs with the aim of recognizing or even treating infected organs/tissues (so, the peptide are named tumor targeting peptides, TPP, or Radionuclide-Labeled Peptides) [17].

The main focus of this subject is on the use of peptides as anticancer agents in cancer research. Meanwhile, considering other activities of these peptides, the attempt is made to correlate such activities with anticancer properties of the peptides through which the kind of mechanism of anticancer activities involved may be deduced for the peptides. To organize the discussion, at first the characteristics of cancer cells will be overviewed. Several mechanisms of action suggested for anticancer molecules will then be discussed. Different classes of anticancer peptides already designed and used for other biological activities, will make the other parts of this subject followed by summary and concluding remarks which come at the end.

Cancer Cells versus Normal Cells

The main characteristic of cancer cells is fast growing and dividing in an earlier and unusual time compared with normal cells, so that they make tumor (the mass of abnormal cells) which may often migrate from the initial place to the other parts of body and invade healthy tissues (metastasis) [9, 18]. On the other hand, normal cells grow and divide in time and remain wherever the body needs them. Normal cells need feeding for proliferation and therefore, new blood vessels are produced to afford this demand accordingly (angiogenesis phenomenon). Normal cells die whenever they are old or damaged in a programming manner (apoptosis phenomenon) or may be repaired when needed. Cancer cells, by capturing and employing angiogenesis mechanism, do not die and often survive in an unlimited time [19]. Accordingly, some functions of cancer cells become different from those of normal cells. These functions as related to the unblocked apoptosis are down regulation of apoptotic-induced proteins Bax and tumor suppressor protein p53, overexpression of matrix metalloproteinase 2 (MMP2), upregulation of the anti-apoptotic proteins Bcl-2, Bcl-XL, Bcl-Xs, and XIAP [20]. Apart from these different phenomena, size and shape of cancer cells are different from those of normal cells. Moreover, cell membrane in cancerous cells is characterized by phosphatidylserine (PS) exposed mainly outside of the membrane (outer leaflet), while in normal cells PS is buried inside the cell membrane along with phosphatidylethanolamine (inner leaflet) [4]. Since PS is a lipid with negative charge, it causes the cancerous cell surface becomes anionic, while zwitterionic phosphatidylcholine (PC) and sphingomyelin (SM) make an overall neutral charge in the normal cell surface (outer leaflet) [21]. Moreover, negative charge of cancerous cell membrane is potentiated by sialic acid attached- glycoproteins like mucins overexpressed in cancer cells [20]. In addition, proteoglycans containing glycosaminoglycan as side chains, bearing high negative charge (because of presenting many sulfate groups) are expressed differently in cancerous cells compared with normal cells [20]. It is also reported that the glucose metabolism ends with the higher secretion of lactate ions in cancer cells. Lactate ions, by neutralizing positive ions environmentally distributed, stabilize the negative charge of cancer cell membrane [22]. Also, interestingly, a greater number of microvilli structures are presented in cancerous rather than normal cell surface area [20]. This latter property increases the membrane surface of the cancer cells in favor of attracting higher concentration of cationic amphipathic molecules like peptides, comparatively.

Cell Membrane and Non-Membrane Involved Anti-Cancer Mechanisms of Peptides

Generally, anticancer peptides can affect their toxicity on cancer cells by membrane- involved or non-membrane involved mechanisms [4]. However, considering the fast killing of the cancer cells by peptides makes this logical thinking that membrane lysis with no receptor involvement is the most probable mechanism of action implemented by anticancer peptides [20].

Cell Membrane Involved Mechanism of Anticancer Peptides

By the first mechanism, peptides trigger cancer cell membrane which contains higher surface anionic charge, compared with normal cells, due to the presence of increased level of Phosphatidylserine and other molecules such as proteoglycans as mentioned earlier. This mechanism of action is the same kind of mechanism assumed for the action of anti-microbial peptides (AMPs). Cationic as well as lipophilic characteristics are the two factors considered for the anticancer activities of peptides, thus the reason why these agents are named as cationic amphipathic peptides (CAPs) [23]. It is to be mentioned that the negative charge density on bacterial cell membrane is lower than that of human cancer cell membrane and, therefore, anti-microbial peptides with positive charge (+4) may show weaker action on human cancer cell membrane which effectively responds to anticancer peptides with higher positive charge (+7) [20]. However, negative charge of cancer cell membrane does not always determine the level of anti-cancer activity of the peptides and other characteristics of cancer cell membranes may also enhance the action of CAPs [24]. It has been assumed that anti-cancer peptides with membrane- involved mechanism, similar to AMPs, make cell membrane disruption through several models such as barrel-stave, toroidal, carpet [4] and Soft Membranes Adapt and Respond, also Transiently’ (SMART) models [25].

Non-Membrane Involved Mechanisms of Anticancer Peptides

Apart from interaction with cell membrane and disruption of cytoplasmic membrane, some peptides may enter the cancerous cells and interact with intracellular components such as mitochondria [4] and induce a programmed cell death, apoptosis, by a so-called intrinsic pathway [8]. Induced-apoptosis by peptides through interaction with mitochondria is assumed to involve cardiolipin, an anionic phospholipid occurred with a high amount in the mitochondrial membrane [20]. If cytoplasmic membrane disruption occurs with or without mitochondrial involvement, necrotic mechanism of peptide action results [8, 20]. Intrinsic pathway starts by the apoptotic proteins, e.g. cytochrome c, released from mitochondria and regulated by the Bcl-2 proteins. Through interaction of peptides with mitochondria, membrane potential rapidly decreases which indicates that the mitochondrial pores are open. This results in the intrinsic pathway of apoptosis by the increased release of pro-apoptotic Bax protein and the decreased release of anti-apoptotic proteins such as Bcls into cytosol [20, 26].

Anticancer Peptides Categories

Anticancer peptides can be subdivided into several classes from the point of their primary activities found at the time of discovery or designing. However, the present survey of anticancer peptide classes is limited to and focused on those previously employed for other applications in our lab, so far. These peptide categories include antimicrobial, anti-inflammatory and antioxidant peptides. Also carrier and homing peptides for cancer diagnostic agents will be discussed in the following sections. Through review of the peptides action, the probable relevant mechanism of anticancer activity will be put forward.

Antimicrobial Peptides

Antimicrobial peptides (AMPs) have been known as a part of defense immune system distributed in all prokaryotes and eukaryotes [27]. Although naturally occurred from the time of discovery, many AMPs have been modified and developed towards to enhance their activity as well as to reduce their instability against protease enzymes [18, 28, 29]. Due to negatively charged cell membranes of the microorganisms, the common mechanism of action of AMPs are assumed to be an electrostatic interaction between AMPs and the cell membrane of the microorganisms [18, 24]. This binding can be potentiated by a hydrophobic interaction of AMPs, with cell membrane as the most cell membranes contain lipophilic layers in their structures [28]. That is the reason why AMPs are classified as molecules with charged and lipophilic characteristic structures [27]. After this initial interaction, pore formation within cell membrane can be induced by AMPs which allows them to proceed cell membrane disruption [27]. This may be followed by further interactions of AMPs with components inside the cells [29]. As mentioned earlier, cancer cells are characterized by higher surface anionic charge, compared with normal cells due to several anionic molecules present in the outer leaflet of cancer cell membrane. Therefore, considering similarity in the mechanism of action, AMPs can be employed for cancer treatment, as well. Of course, not all AMPs are good candidates for such kind of application because some AMPs may show toxicity against not only cancerous but also normal cells, as this subject has been considered by some authors for classifying AMPs [27]. Plants are among the main resources of AMPs. Moreover, many AMPs with cyclic structures (e.g., cyclotides) identified and isolated from plants have been synthesized and optimized. Also, AMPs have been made by de novo designing, as reported in literature [30-34]. The plants of Caryophyllaceae family are composed of 81 genera from which Dianthus genus covers more than 300 species [35, 36]. Several cyclic peptides, generally called dianthins, isolated from the species of Dianthus, have shown various biological activities [36-39]. Moreover, the synthesis of some dianthins and their analogues has been attempted and the relevant biological activities have been examined according to several reports [40-42]. Among the various activities mentioned for dianthins are the antimicrobial and anthelmintic activities reported for Dianthin A [40] and Longicalycinin A [41]. Longicalycinin A, a cyclic peptide with the sequence of (cyclo-(Phe-Tyr-Pro-Phe-Gly)), was previously isolated from Dianthus superbus var. longicalycinus [43]. It was also synthesized by a solution phase method in 2007 [41]. It has been shown that Longicalycinin A possesses a high cytotoxic activity against several cancerous cell lines [41, 43]. The cytotoxicity of the solid-phase synthesized Longicalycinin A was also reported in 2013 [42]. Being interested for further studies, the synthesis of this cyclopeptide and its several linear and cyclic analogues was carried out in our lab [44, 45]. Using different assay experiments including MTT, flow cytometry and lysosomal membrane integrity methods, it was found that the most of the designed and synthesized Longicalycinin A analogues were cytotoxic against colon HT-29 and heptic HepG2 cancer cells. Among the analogues, two cyclic peptides with the structures of cyclo-(Thr-Val-Pro-Phe-Ala) and cyclo-(Phe-Ser-Pro-Phe-Ala) demonstrated better anticancer activities and their cytotoxic activities against normal fibroblast cells were negligible [45]. The mechanism of anticancer action of these cyclopeptides was assumed to be through apoptosis induction. In the other experiment a designed and synthesized heptapeptide analogue of Longicalycinin A with the linear sequence of Cys-Phe-Tyr-Pro-Phe-Gly-Cys and cyclized by the disulfide bond, also showed good anticancer activity against HepG2 and HT-29 cell lines while presented a safety profile against fibroblast cells [45].

Anti-inflammatory Peptides

Inhibitors of cyclooxygenase enzymes (COX enzymes) have been long time known as non-steroidal anti-inflammatory drugs (NSAIDs) [46]. Since these drugs generally inhibit non-specifically the action of the both COX-1 and COX-2 enzymes, some adverse effects, most importantly peptic ulcer and kidney damage, occur in patients who use these drugs [47, 48]. It has been shown that these side effects caused by NSAIDs are due to the inhibition of COX-1 enzyme activity [49]. COX-1 and COX-2 catalyze the production of prostaglandins from arachidonic acid which is a fatty acid [46]. Prostaglandins such as PGG2 and PGH2 mediate pain and inflammatory responses against some internal or external stimuli and so, COX inhibitors are used to make a relief from such unpleasant conditions in patients [50]. On the other hand, prostaglandins are necessary for protection of gastric lining, kidney tissue, etc [51]. Some prostaglandins such as PGE2 and PGI2 are responsible for the inhibition of gastric acid secretion caused by the release of gastrin or histamine [50]. COX-1, a constitutional isoenzyme, acts more specifically on arachidonic acid to produce prostaglandins, while COX-2, mainly an inducible isoenzyme produced by cytokines, generally catalyzes all the fatty acids including arachidonic acid [50]. Therefore, inhibition of COX-1, in addition to anti-inflammatory effect, may result in the increase of gastric acid secretion, decrease of gastric mucosa protection and induction of renal side effects. Whereas, it is assumed that inhibition of COX-2 selectively reduces prostaglandins in the inflammatory tissues [49]. As a result, to obtain more specific action, many COX-2 inhibitors have been introduced in market [52]. Unfortunately, COX-2 inhibitors have been shown to cause some adverse effects mainly on heart and kidney, as well [53]. On the other hand, some COX-2 inhibitors (i.e., celecoxib) have been shown to reduce polyp growth in colon and produce anticancer activity specifically against colon tumors, prostate and breast cancers [54]. The cause of this activity was described as the overexpression of COX-2 found in such tissues. To reduce side effects and meanwhile potentiate anticancer activity of COX-2 inhibitors, researchers have been encouraged to search for new compounds. Peptides, currently used in many pharmacological fields as antihypertensive agents, opioids, immune system modulators, antioxidative and anti-infective agents [55], have attracted scientists to find new anti-inflammatory agents based on peptide scaffolds [56, 57]. Moreover, anticancer activities of such compounds, if found, would be quite favorable. Considering the structure-activity relationship (SAR) studies of COX-2 inhibitors, some di, tri, and tetrapeptide analogues were designed and synthesized in our lab as COX-2 inhibitors. Of course, anticancer activities of these peptides were looked for, as well. The peptides were designed to contain the compulsory pharmacophoric moieties needed for COX-2 inhibitory action [58, 59]. These pharmacophoric parts were a phenyl ring containing N3 or SO2Me group connected at the para position, one or two amino acids bearing a ring structure and one amino acid with free carboxyl group to represent the C-terminal moiety of the molecules. For the simplest molecules, p-N3 (or p-SO2Me) Benzoyl moiety, as one amino acid analogue, was coupled with an amino acid though an amide (peptide) bond to produce some dipeptide analogues. Using a COX-1/COX-2 activity assay kit [60], the results showed that the both selectivity and potency could be affected depending on the kind of amino acid connected to the benzoyl moiety bearing N3 or SO2Me group at the para position. In the series of dipeptide analogues [61], the best result of COX-2 inhibitory activity was found for compound p-N3Bz-His with selectivity index SI (COX-1 IC50/COX-2 IC50) 351.2 which was comparatively less than that of celecoxib (SI; 405). Considering the tripeptide analogues, the COX-2 inhibitory results were more optimistic [62]. In these series of tripeptide analogues, compound p-MeSO2Bz-Tyr-Glu gave the best selectivity index (SI; >500). Compounds p-N3Bz-Pro-Glu and p-N3Bz-Phe-Glu gave SI equal to 439.2 and 463.6, respectively. All these three compounds showed higher SI compared to celecoxib. Following COX-2 inhibitory experiments, in vitro antiproliferative activity of the three peptide analogues using MTT test [63] showed that the best cytotoxic activity belongs to p-MeSO2Bz-Phe-Glu against Hep-G2, A549 and HELA, but not on MCF-7. Peptide p-MeSO2Bz-Tyr-Asp showed comparably a good cytotoxic activity on MCF-7, HepG2 and A549 cancer cells. Hela cells were not affected by this peptide. Compound p-N3Bz-Pro-Glu gave an inhibitory action against proliferation of MCF-7 and Hela cells better than HepG2 and A549 cells, in comparison with celecoxib [62]. Tetrapeptide analogues with the general formula p-MeSO4Bz-Y-X-Asp synthesized in our lab, demonstrated toxic activity (the growth inhibitory effect) against four different cancer cells; MCF-7, HepG2, HT-29 and A549, employing MTT assay [64]. Among these peptides, four compounds showed anticancer activity even better than celecoxib. Moreover, these compounds demonstrated weak or no toxicity on human skin fibroblast giving a desirable safety profile result. However, COX-2 inhibition by these peptides was not noticeable (the data are not published). Table 1 shows the di, tri and tetrapeptides formula which demonstrated noticeable COX-2 inhibitory and/or anticancer activity among the peptides synthesized in our lab. As a conclusion from these studies, tripeptide analogues designed as general formula p-N3 (or p-MeSO2)Bz-Amino Acid1(with a ring structure)-Amino Acid2 (acidic), may be good candidates as COX-2 inhibitors with peptide scaffolds which possess anticancer activity, as well.

Table 1The designed di, tri and tetrapeptides which showed noticeable activity as COX-2 inhibitor and /or anticancer agents.Dipeptide Azide Analogues, COX-2 InhibitorTripeptide Azide Analogues, COX-2 Inhibitors/anticancer AgentsTripeptide Methylsulfone Analogues, COX-2 Inhibitors/anticancer AgentsTetrapeptide Methylsulfone Analogues Anticancer Agentsp-N3Bz-Hisp-N3Bz-Phe-Glup-MeSO2Bz-Tyr-Glup-MeSO4Bz-Ser-Tyr-Asp-p-N3Bz-Pro-Glup-MeSO2Bz-Tyr-Aspp-MeSO4Bz-Ser-Phe-Asp--p-MeSO2Bz-Phe-Glup-MeSO4Bz-Gly-His-Asp---p-MeSO4Bz-Ile-His-Asp

Antioxidant Peptides

Internal and external oxidants may cause oxidative damage on body tissues leading to occur many diseases including cancer [55, 65]. In fact, cancerous condition can be produced by oxidative stress or by the action of oxidizing agents such as reactive oxygen species (ROS) which induce DNA damage or mutation [66]. By contrast, many biological molecules including glutathione (an endogen tripeptide), melatonin, carnosine and some other related dipeptides produced in biological system can inhibit such cellular damage and give protective shields against the harmful oxidizing agents. Carnosine (β- Alanine-L-histidine), discovered by Gulvich and Amiradzibi [67], is an unusual dipeptide biosynthesized and stored in high concentration inside animal organs, e.g., heart, brain and muscles. Many interesting chemical properties of carnosine such as zwitterionic characteristic, divalent metal ion chelating and good buffering activity at physiological pH, have made carnosine to play various favorable physiological roles in animal bodies. Carnosine is a natural antioxidant which acts as a scavenger to reduce ROS production and aldehyde formation resulted by the fatty acids oxidation in oxidative stress situations. Carnosine can also inhibit protein glycosylation. Both Protein glycation and ROS production are presumably involved in many degenerative diseases such as diabetes, atherosclerosis, austin and Alzheimer. Glycoproteins also promote the process of aging [68-71]. There are few reports considering the inhibitory action of carnosine against neoplastic cells [72-74]. The mode of inhibitory action of carnosine on tumor growth is still not clear, although some suggestions have been put forwards, so far [75, 76]. To investigate carnosine as anticancer agent, this peptide along with several relevant linear and cyclic dimer analogues were designed and synthesized in our lab and their cytotoxic activity on cancer cells HepG2 and HT-29 were evaluated using MTT assay, flow cytometry analysis and lysosomal membrane integrity determination method [77]. Among the synthesized peptides, one linear peptide, Pro-β-Ala-His-β-Ala-His, as well as two cyclopeptides, cyclo(β-Ala-His-Pro-β- Ala-His) and cyclo(Pro-β-Ala-His-β-Ala-His), were found to be cytotoxic on HepG2 and HT-29 cancer cells, compared with 5- Fluorouracil as control drug (Table 1). These peptides also showed safety profile results on fibroblast cells [77]. In the other experiment, carnosine and inversed sequence related peptide analogues were also synthesized and their biologic activities were examined [78]. Among the inversed peptide analogues, a linear peptide and its cyclic congener with the formula β-Ala-His-Pro-His-β-Ala showed considerable cytotoxicity on the hepatic and colon cancer cell lines. The other cyclic peptide with the structure cyclo(His-β-Ala-Pro-β-Ala-His) also showed promising as a cytotoxic agent (Table 2) [78]. In general, our results showed that carnosine and related designed peptides contain cytotoxicity activity on cancerous cells and produce their cytotoxicity, at least partly, by cell apoptosis induction.

Table 2Viability percentage of the cell lines, HepG2 and HT-29, exposed to the linear and cyclic Carnosine analogues by MTT and Flow cytometry assays.CompoundsViability Percentage (HepG2)MTT (Flow Cytometry Assay)Viability Percentage (HT-29)MTT (Flow Cytometry Assay)Carnosine8.01 (7.86)27.79 (0.42)Pro-β-Ala-His-β-Ala-His3.28 (0.33)13.97 (32.43)cyclo(β-Ala-His-Pro-β-Ala-His)9.2 (0.55)11.76 (0.18)Cyclo(Pro-β-Ala-His-β-Ala-His)19.31 (0.45)12.44 (0.26)β-Ala-His-Pro-His-β-Ala7.03 (0.09)11.49 (9.19)Cyclo(β-Ala-His-Pro-His-β-Ala)7.01 (0.9)21.3 (0.29)Cyclo(His-β-Ala-Pro-β-Ala-His)9.36 (0.72)10.75 (0.39)5-FU3.229.28

Peptide Conjugates

Some of the synthetic quinolone antibiotics including ciprofloxacin have shown anticancer activity in addition to their antimicrobial activity [79-82