Advances in Cancer Drug Targets: Volume 1 -  - E-Book

Advances in Cancer Drug Targets: Volume 1 E-Book

0,0
26,52 €

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

Advances in Cancer Drug Targets is an e-book series that brings together recent expert reviews published on the subject with a focus on strategies for synthesizing and isolating organic compounds and elucidating the structure and nature of DNA. The reference work serves to give readers a brief yet comprehensive glance at current theory and practice behind employing chemical compounds for tackling tumor suppression, DNA site specific drug targeting and the inhibition of enzymes involved in growth control pathways. The reviews presented in this series are written by experts in pharmaceutical sciences and molecular biology. These reviews have been carefully selected to present development of new approaches to anti-cancer therapy and anti-cancer drug development. This e-book volume will be of special interest to molecular biologists and pharmaceutical scientists.

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

EPUB

Seitenzahl: 473

Veröffentlichungsjahr: 2013

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
Welcome
Table of Contents
Title
BENTHAM SCIENCE PUBLISHERS LTD.
End User License Agreement (for non-institutional, personal use)
Usage Rules:
Disclaimer:
Limitation of Liability:
General:
FOREWORD
LIST OF CONTRIBUTORS
The PIK3CA Gene as a Mutated Target for Cancer Therapy
Abstract
INTRODUCTION
TARGETING PIK3CA MUTATIONS
PI3K Inhibitors
AKT Inhibitors
mTOR Inhibitors
Possibilities of Novel Dual Inhibition
CONCLUSIONS
CONFLICT OF INTEREST
ACKNOWLEGMENTS
DISCLOSURE
Abbreviations
REFERENCES
AKT Signaling in Regulating Angiogenesis
Abstract
Introduction
Angiogenesis and Cancer
AKT signaling is activated by VEGF and ANGiopoietins
MIRNAS AND PI3K/AKT SIGNALING PATHWAY
Role of AKT in tumor angiogenesis
Role of AKT in normal vascularization
AKT regulates VEGF expression
AKT regulates HIF-1α expression
Heavy metals regulate VEGF and HIF-1α expression via AKT pathway
AKT is the common target inhibited by different natural PRODUCTS with potential chemoprevention effects
Therapeutic perspectives for targeting AKT and mTOR
ACKNOWLEDGEMENTS
CONFLICT OF INTEREST
DISCLOSURE
ABBREVIATIONS
References
Inhibitors of Cyclin Dependent Kinases: Useful Targets for Cancer Treatment (An Update)
Abstract
INTRODUCTION
CDK INHIBITORS
CDK Inhibitory Proteins or CKIs
Small Molecule Inhibitors
Cip/Kip Family
INK4 Family
Small Molecules as CDK Inhibitors
Staurosporine and Analogues
Flavonoid Derivatives
Purine Derivatives
Indole Derivatives
Pyridine Derivatives
Pyrimidines Derivatives
Indirubin Derivatives
Pyrazole Derivatives
Thiazole Derivatives
Paullones and Derivatives
Hymenialdisine and Analogues
Miscellaneous Compounds
CONCLUSION
ACKNOWLEDGEMENT
CONFLICT OF INTEREST
DISCLOSURE
REFERENCES
Cellular FLICE-Like Inhibitory Protein (C-FILP ): A Key Anti-Apoptotic Factor and a Target for Cancer Therapy
Abstract
INTRODUCTION
APPOPTOSIS SIGNALING PATHWAYS
C-FILP AND THE DEATH RECEPTOR APOPTOSIS PATHWAY
STRUCTURE OF C-FILP
C-FILP FUNCTION
TRANSCRIPTION AND TRANSLATION REGULATION OF C-FILP
C-FILP DEGRADATION
C-FILP VARIANTS AS TARGETS FOR CANCER THERAPY
AGENTS KNOWN TO DOWNREGULATE C-FILP
DISCLOSURE
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Pin1: A Promising Novel Diagnostic and Therapeutic Target that Acts on Numerous Cancer-Driving Pathways
Abstract
Introduction
Overview & Structural Basis for PIN1 Mode of Action
DISCOVERY AND CELL CYCLE FUNCTION
PIN1 AND CANCER
Regulation of PIN1
PIN1: A UNIQUE CANCER TARGET ACTING ON NUMEROUS CANCER-DRIVING PATHWAYS
Closing Remarks
ACKNOWLEDGEMENTS
CONFLICT OF INTEREST
DISCLOSURE
Abbreviations
REFERENCES
Anticancer Immunotherapy in Combination with Proapoptotic Therapy - Possible Therapeutic Strategies for Enhancement of Anticancer Immune Reactivity in Autologous Immunocompetent Cells and After Allogeneic Stem Cell Transplantation
Abstract
INTRODUCTION
IMMUNOGENIC CANCER CELL DEATH
INDUCTION OF ANTICANCER IMMUNE REACTIVITY: IMMUNOREGULATORY DENDRITIC CELLS ORCHESTRATE THE DEVELOPMENT OF ANTI-CANCER T CELL REACTIVITY
INDUCTION OF ANTI-CANCER T CELL REACTIVITY BY DENDRITIC CELL VACCINES
THE EFFECTOR PHASE OF ANTICANCER IMMUNITY
The Intrinsic Apoptosis-Inducing Pathway is Essential for Induction of Cancer Cell Apoptosis by Cytotoxic T Cells
The External Death Receptor Pathways of Apoptotis Induction
Pharmacological Targeting of the External Death Receptor Pathway: A Possible Therapeutic Strategy to Increase Cancer Cell Susceptibility to T cell Cytotoxicity
Pharmacological Targeting of Survival Pathways: An Alternative Strategy to Increase Cancer Cell Susceptibility to T cell Cytotoxicity
IMMUNOSUPPRESSIVE EFFECTS OF CHEMOTHERAPY
REGULATORY T CELLS, TH17 T CELLS AND MESENCHYMAL STROMAL/STEM CELLS - KEY REGULATORS OF ANTICANCER IMMUNE REACTIVITY
Treg Cells
Th17 Cells - A Dynamic Proinflammatory Phenotype
The Th17: Treg Ratio After Intensive Chemotherapy
Mesenchymal Stromal/Stem Cells - Therapeutic Use Versus Therapeutic Targeting
ALLOGENEIC STEM CELL TRANSPLANTATION - AN ESTABLISHED COMBINATION OF PROAPOPTOTIC CHEMOTHERAPY AND IMMUNOTHERAPY
Allotransplantation in Leukemia - is Immunogenic Apoptosis Possible?
Reduction of the Cancer Cell Burden by Pretransplant Conditioning Therapy - Effects on Host Dendritic Cells and the Induction of Antileukemic Immune Reactivity
The Posttransplant Immune System - Pharmacological Agents with Immunosuppressive and Antileukemic Effects
Treg and Th17 Cells in Patients Receiving Allogeneic Stem Cell Transplantation
What can we Learn from the Allotransplantation Experience About Combination of Chemotherapy and Immunotherapy?
CONCLUDING REMARKS
ACKNOWLEDGEMENTS
CONFLICT OF INTEREST
DISCLOSURE
ABBREVATIONS
REFERENCES
Melatonin and Breast Cancer: Selective Estrogen Enzyme Modulator Actions
Abstract
INTRODUCTION
THE ROLE OF ESTROGENS IN BREAST CANCER
INTRACRINOLOGY OF ESTROGEN DEPENDENT HUMAN BREAST CARCINOMA
HORMONAL THERAPY IN BREAST CANCER
MELATONIN, THE MAIN PINEAL HORMONE
MELATONIN AND ESTROGENS IN BREAST CANCER
MELATONIN AS A SELECTIVE ESTROGEN ENZYME MODULATOR
Anti-Aromatase Actions of Melatonin
Anti-Sulfatase Actions of Melatonin
17β-Hydroxysteroid Dehydrogenase Inhibitory Actions of Melatonin
Estrogen Sulfotranferase Stimulatory Actions of Melatonin
Regulation of the Enzymes Related to the Local Production of Estrogens in Human Normal Breast Tissue and Breast Carcinoma Tissue by Melatonin
CONCLUSIONS
ACKNOWLEDGEMENTS
CONFLICT OF INTEREST
DISCLOSURE
REFERENCES
Targeting Cancer and Neuropathy with Histone Deacetylase Inhibitors
Abstract
Epigenetic regulation of gene expression
Targeting histone acetylation with HDACI
VPA and cancer treatment
Peripheral neuropathy as a critical limiting factor in anticancer treatment
Cisplatin-induced PN as a paradigm of CIPN
VPA in CDDP-induced PN: two birds with one stone?
Other histone hyper-acetylating strategies: Acetyl-L-carnitine
Conclusions
DISCLOSURE
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
List of Abbreviations
References
Advances in Cancer Drug Targets
(Volume 1)
Editor
Atta-ur-Rahman
Honorary Life Fellow
Kings College
University of 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 following DRM (Digital Rights Management) policy may also be applicable to the Work at Bentham Science Publishers’ election, acting in its sole discretion:25 ‘copy’ commands can be executed every 7 days in respect of the Work. The text selected for copying cannot extend to more than a single page. Each time a text ‘copy’ command is executed, irrespective of whether the text selection is made from within one page or from separate pages, it will be considered as a separate / individual ‘copy’ command.25 pages only from the Work can be printed every 7 days.

3. 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 the U.A.E. as applied in the Emirate of Dubai. Each party agrees that the courts of the Emirate of Dubai 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 Ltd. Executive Suite Y - 2 PO Box 7917, Saif Zone Sharjah, U.A.E. Email: [email protected]

FOREWORD

Cancer drug therapy has evolved significantly in the last two decades. Progress in the synthesis and isolation of organic compounds as well as our understanding of the structure and nature of DNA have also contributed to advances in anti-cancer drug development. Contemporary biochemical strategies to combat cancers range from cytotoxic and DNA damaging mechanisms to regulating programmed cell death (apoptosis). This eBook is the first volume of the series Advances in Cancer Drug Targets and is a compilation of recent expert reviews published in Current Cancer Drug Targets on the subject with a focus on these strategies. The reference work serves to give readers a brief yet comprehensive overview of current theory and practice behind employing chemical compounds for tackling tumor suppression, DNA site specific drug targeting and the inhibition of enzymes involved in growth control pathways. The reviews, written by experts in pharmaceutical sciences and molecular biology, have been carefully selected and represent an attempt to collect and current research on anti-cancer drug development for interested researchers and pharmaceutical scientists. Advances in Cancer Drug Targets should also provide insights into development of new approaches to anti-cancer therapy. I hope that the articles contained in this volume will be greatly enjoyed by the readers.

I would like to thank Mr. Mahmood Alam, Mr. Taimur Khan, Ms. Nazia Kamran and Ms. Humaira Hashmi for their help and support.

LIST OF CONTRIBUTORS

Atta-ur-RahmanHonorary Life Fellow Kings College, University of Cambridge, Cambridge, UKCarolina Alonso-GonzálezDepartment of Physiology and Pharmacology, School of Medicine, University of Cantabria, SpainVirginia Alvarez-GarcíaDepartment of Physiology and Pharmacology, School of Medicine, University of Cantabria, SpainØystein BruserudInstitute of Medicine, University of Bergen, Haukeland University Hospital, NorwayGuido CavalettiDepartment of Neuroscience and Biomedical Technologies, University of Milano-Bicocca, ItalyJustin CidadoThe Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, USASamuel CosDepartment of Physiology and Pharmacology, School of Medicine, University of Cantabria, SpainDavid CosgroveDivision of Medical Oncology, The Johns Hopkins University, School of Medicine, USASarah CroessmannThe Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, USAElisabeth ErsværInstitute of Medicine, University of Bergen, NorwayGreg FinnCancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, USABjørn Tore GjertsenSection for Hematology, Department of Medicine, Haukeland University Hospital, NorwayAlicia GonzálezDepartment of Physiology and Pharmacology, School of Medicine, University of Cantabria, SpainJohn P. GustinThe Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, USABing-Hua JiangDepartment of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, USAAstrid Olsnes KittangSection for Hematology, Department of Medicine, Haukeland University Hospital, NorwayLing-Zhi LiuDepartment of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, USAKun Ping LuCancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, USAMan-Li LuoCancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, USACarlos Martínez-CampaDepartment of Physiology and Pharmacology, School of Medicine, University of Cantabria, SpainGuro Kristinv MelveDepartment of Immunology and Transfusion Medicine, Haukeland University Hospital, NorwayBen Ho ParkDepartment of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, USAHåkon ReikvamInstitute of Medicine, University of Bergen, Haukeland University Hospital, NorwayVirginia Rodriguez-MenendezDepartment of Neuroscience and Biomedical Technologies, University of Milano-Bicocca, ItalyAhmad R. SafaDepartment of Pharmacology and Toxicology, Indiana University Simon Cancer Center, Indiana University School of Medicine, USAPooja S. SharmaDepartment of Chemistry, C.S.S.S. (P.G.) College, IndiaRajan SharmaDepartment of Chemistry, C.S.S.S. (P.G.) College, IndiaLucio TremolizzoDepartment of Neuroscience and Biomedical Technologies, University of Milano-Bicocca, ItalyXiao Zhen ZhouCancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, USA

The PIK3CA Gene as a Mutated Target for Cancer Therapy

Sarah Croessmann,Justin Cidado,John P. Gustin,David Cosgrove,Ben Ho Park1,2,*
1The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA and 2The Johns Hopkins University, Department of Chemical and Biomolecular Engineering, Baltimore, MD21218, USA

Abstract

The development of targeted therapies with true specificity for cancer relies upon exploiting differences between cancerous and normal cells. Genetic and genomic alterations including somatic mutations, translocations, and amplifications have served as recent examples of how such differences can be exploited as effective drug targets. Small molecule inhibitors and monoclonal antibodies directed against the protein products of these genetic anomalies have led to cancer therapies with high specificity and relatively low toxicity. Our group and others have demonstrated that somatic mutations in the PIK3CA gene occur at high frequency in breast and other cancers. Moreover, the majority of mutations occur at three hotspots, making these ideal targets for therapeutic development. Here we review the literature on PIK3CA mutations in cancer, as well as existing data on p110α inhibitors and inhibitors of downstream effectors for potential use as targeted cancer therapeutics.

Keywords::PIK3CA, mutation, oncogene, PI3 kinase, AKT, mTOR.
*Address correspondence to Ben Ho Park: Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, 1650 Orleans Street, Room 151, Baltimore, MD, 21287, USA; Tel: 410-502-7399; Fax: 410-614-8397; E-mail: [email protected]

INTRODUCTION

Insults to the genome are a hallmark of cancer and a driving force in carcinogenesis. These genomic alterations are an obvious difference between normal and cancerous cells, and provide an opportunity for exploitation as potential targets for therapeutics. Over the past decade, successful clinical trials of imatinib, gefitinib, erlotinib, and trastuzumab, which are specific for BCR-ABL translocations [1], epidermal growth factor receptor (EGFR) mutations [2, 3], and HER2/neu amplifications [4-6] respectively, have illustrated the ability to develop drugs that target genetic abnormalities in human malignancies. This has opened

the possibility for future streamlined therapies based on the genomic landscape of an individual’s cancer.

Phosphatidylinositol 3-kinases (PI3K)s are heterodimeric proteins consisting of a regulatory subunit (p85) and a catalytic component (p110), and there are several isoforms and classes of known PI3Ks. The p110α catalytic subunit of the Class I PI3Kα, encoded by the gene PIK3CA, is one of the most highly mutated oncogenes in human cancers. High mutational frequencies of PIK3CA have been reported in colorectal [7], breast [8] and liver cancers [9] while lower rates of mutation have been described in many other human malignancies including ovarian [10, 11], lung [7, 9], gastric [7, 9, 12, 13], and brain cancers [7, 9, 14-21]. In breast cancer, PIK3CA has a reported overall mutational rate of 25%, with more than 80% being attributed to “hot spot” regions within exon 9 of the helical domain and exon 20 of the catalytic domain [8]. These three mutations, E542K and E545K in exon 9 and H1047R in exon 20 lead to increased PI3K catalytic activity. This increased activity results in cellular transformation through growth factor- and anchorage-independent cellular proliferation [22-24]. Studying the effects of these mutations in colorectal cells [25-27], breast epithelial cells [28, 29], and chicken embryo fibroblasts [30, 31] have illustrated a direct connection between these mutations and carcinogenesis. Through crystallographic and biochemical methods, it has been determined that the probable oncogenic mechanism of the E545K mutation is the disruption of an inhibitory charge-charge interaction between p110α and the N-terminal SH2 domain of the p85 regulatory subunit [32] (Fig. 1). It has been previously proposed that the oncogenic mechanism of the E542K mutation is similar to E545K, exhibiting a change in the interaction of p110α with the p85 regulatory subunit. The proposed oncogenic mechanism of the H1047R mutation differs from the exon 9 mutations. The H1047R mutation increases the binding affinity of p110α for the negatively charged phosphatidylinositol substrate leading to increased activity and transforming potential [33]. Furthermore, aberrant PI3K signaling has also been linked to resistance of cells in preclinical models to a number of targeted and cytotoxic cancer therapies, including trastuzumab and paclitaxel resistance in human breast epithelial cells harboring PIK3CA mutations [28, 34]. Clinically, the presence of PIK3CA mutations has been linked to both favorable [35, 36] and unfavorable [37, 38] patient prognosis, and it has also been reported that exon 9 mutations have a less favorable prognosis than exon 20 mutations in breast cancer [39]. The reasons for these conflicting data are not clear, but likely reflect limited sample sizes and difference in treatment regimens between the various studies.

Figure 1)

A representation of the Domains of the PI3K Subunits p110α and p85α. The p110α catalytic subunit has 5 domains including adaptor-binding domain (ABD), the Ras-binding domain (RBD), a calcium binding domain (C2), a helical domain and a kinase domain. The p85α regulatory subunit contains 5 domains as well, which include a Src homology 3 domain (SH3), a GTPase activating protein domain (GAP), an N-terminal Src homology 2 domain (nSH2), an inter- Src homology 2 domain (iSH2), and a C-terminal Src homology 2 domain (cSH2). The exon 9 hotspot mutations, E542K and E545K, occur in the helical domain of the catalytic subunit p110α, and the charge reversal caused by these mutations inhibits electrostatic interactions between those amino acids on the p110α helical domain and R340 and K379 on the nSH2 domain of p85α. The exon 20 hotspot mutation, H1047R, is in the kinase domain of p110α, and this mutation has been proposed to form a hydrogen bond with L956 of p110α, which in turn leads to catalytic activity of p110α.

TARGETING PIK3CA MUTATIONS

With the recent success of biologics and small molecular inhibitors for anti-cancer therapy, the identification and validation of additional therapeutic targets is of great importance and interest. PIK3CA somatic mutations would be ideal for targeting due to their high rate of occurrence and the fact that 80% to 90% of these mutations are in one of the three previously discussed hotspots. Below, we review several classes of targeted compounds that may have clinical utility for the treatment of cancers harboring PIK3CA mutations.

PI3K Inhibitors

The most direct method of targeting cancers that have PIK3CA mutations would be to develop inhibitors that have high specificity for mutant p110α but not its wild type counterpart. The ability to create relatively mutation-specific small molecule inhibitors is exemplified by erlotinib and gefitinib, which were initially developed as EGFR inhibitors but were found to be most effective in patients whose tumors contained specific EGFR mutations [40, 41]. This finding is attributed to oncogene addiction [40], which is the phenomenon whereby cancer cells become dependent on particular alterations for survival as well as preservation of the malignant state. Thus, specific genetic changes within cancer cells, such as the PIK3CA hotspot mutations, result in increased growth signals from aberrantly activated pathways, and therefore removal of these signals leads to decreased cellular growth and apoptosis [42]. Recent structural data demonstrate that rational design of mutation specific inhibitors is feasible [43], and therefore the emergence of targeted mutant PIK3CA therapies is likely to be imminent.

Prior to the discovery of somatic PIK3CA mutations in human cancers, the PI3K enzyme was already recognized as being an important molecule in mediating carcinogenesis. As such, inhibitors of PI3K were developed with the hope that a therapeutic window could be achieved. The earliest and best characterized PI3K inhibitors are wortmannin and LY294002. Both of these compounds have been shown to be effective anti-tumor agents in in vitro cell culture models, as well as in in vivo animal models [44-46]. LY294002 has been shown to inhibit both in vitro PI3K activity and phosphorylation of downstream effectors of PIK3CA in breast epithelial [28] and colorectal cancer cells [27]. However, due to their poor pharmacological properties and marked cytotoxicity, LY294002 and wortmannin do not have clinical utility as reviewed by Workman [47]. Additionally, they are not ideal for specifically targeting PIK3CA mutants because they can inhibit other kinases of the PI-3 kinase-like kinase (PIKK) family, and several other kinases such as the mammalian target of rapamycin (mTOR) [48-52]. Despite having limited preclinical studies due to poor solubility, instability and high toxicity, both the structures of LY294002 and wortmannin have served as templates for derivatives and prodrugs with better pharmacological properties [53, 54]. The prodrug, SF-1126, is a vasculature-targeting Arg-Gly-Asp peptide conjugated to LY294002 that exhibits increased stability and tolerance in murine models [55]. Similarly, wortmannin has served as an analog template for a pan-PI3K inhibitor, PX-866, which demonstrated increased cell permeability and a prolonged serum half-life [56]. Recently, a number of groups have reported developing p110α selective inhibitors [57-63], and some have demonstrated efficacy in vitro and in vivo [57-59, 61, 62, 64]. The development of subsequent generations of PI3K pathway inhibitors and combined therapies have aided in circumventing the limitations of first generation inhibitors [65].

Class-specific and isoform-specific PI3K inhibitors are attractive target options, providing the opportunity for global inhibition of downstream components. It is important to note, however, that PI3K inhibitors have the potential for greater toxicity due to their importance for normal cellular processes. Class I PI3Ks inhibitors, such as GDC-0941[66], XL-147 [67], BKM120 [68], GSK1059615, CAL-101[69], and PX-866[70], are currently in clinical development. Despite significant homology between the different p110 catalytic subunits (α, β, δ, γ), recent structural data supports the potential for success of current Class I PI3K p110 isoform-specific inhibitors [43], with a particular interest in p110α specific inhibitors due to the possibility of oncogenic PIK3CA mutations being predictors of response [71]. BYL719, GDC-0032, and INK-1117, are three p110α-specific inhibitors currently in early clinical development [72]. Initial studies have identified other p110 isoform-specific inhibitors TGX-221[73], CAL-101, and AS-252424 [74] for p110β, p110δ, and p110γ, respectively. Isoform-specific inhibitors demonstrate the ability to target particular alterations in the PI3K pathway with theoretically less off target effects. In addition to isoform specific inhibitors, Class specific inhibitors in pre-clinical studies have shown the possibility of preferential responses to tumors harboring PIK3CA mutations. For example the novel Class I PI3K inhibitor, CH5132799, is particularly selective for cancer cells harboring PIK3CA mutations [75]. In addition, other PI3K isoforms may also prove to be attractive targets for breast cancer therapy. For example, studies have demonstrated that expression of PIK3CB in conjunction with HER2 amplification, can lead to a worse overall prognosis [76]. The future development of isoform specific PI3K inhibitors may allow for novel therapeutic development and targeting of breast cancers based upon the genetic alterations that drive the PI3K pathway.

Buttressed against these exciting developments, specific targeting of PIK3CA may be problematic, because as previously mentioned, PIK3CA is involved in a number of signaling pathways associated with normal cellular function, such as insulin signaling [63, 77]. This may result in PIK3CA inhibitors that are prohibitively cytotoxic, thus limiting their clinical benefit. To illustrate this point, some PIK3CA inhibitors have been shown to abrogate the effects of insulin in mice [63]. Similarly, PIK3CA deficient mice have recently been shown to have an increased rate of heart failure in response to cardiac stress [78]. Thus, the possibility of a significant side effect profile has led to the development of compounds designed to target downstream effectors within the PI3K pathway with the hope that this may be a more effective strategy to target cancers containing PIK3CA mutations.

AKT Inhibitors

AKT, also known as protein kinase B, is a serine threonine kinase directly downstream of PI3K, and due to its central role in signal transduction, the dysregulation of AKT is commonly associated with many different cancers [79]. More specifically, constitutive activation of AKT has been associated with PIK3CA mutations in several in vitro cell models [26-30]. Therefore, AKT inhibitors may prove to be useful in targeting cancers with PIK3CA mutations.

AKT was identified as an oncogene over two decades ago [80], and multiple AKT inhibitors have been developed and used successfully to inhibit AKT activation and cellular growth in in vitro and in vivo tumor models [81-108]. Unlike PI3K, development of isoform-specific AKT inhibitors has proven difficult due to the high degree of homology among the three AKT isoforms. Perifosine is the most studied AKT inhibitor, proving efficacious at inhibiting the growth of various cancer cell types [105]. As a single agent, Perifosine has not shown therapeutic benefit in several phase II trials, however, Perifosine may have some therapeutic potential when used in combination with radiation or other standard cytotoxic agents, as evidenced by in vitro studies [109-111]. Another AKT inhibitor, Miltefosine, has shown efficacy in clinical trials for the topical treatment of cutaneous lymphoma and breast cancer skin metastases [112-115]. API-2, another early AKT inhibitor [102], had shown some efficacy in early phase trials, but was abandoned due to excessive toxicity [116, 117]. These early AKT specific compounds aided in directing the investigation of more specific AKT inhibitors at lower doses than those used previously, thus potentially avoiding detrimental side effects. Recent strategies for developing AKT inhibitors have included ATP-competitive inhibitors, phosphotidylinositol analogs, and allosteric inhibitors. GSK690693 and A-443654, two pan-AKT ATP-competitive agents, have demonstrated anti-tumor activity in preclinical models and are currently in early Phase trials [118, 119]. Allosteric inhibitors, such as MK-2205, have demonstrated relatively high selectivity and shown marked suppression of breast cancer growth, and are currently in late Phase II clinical trials [120]. These compounds interact with the pleckstrin homology (PH) domain or hinge region, promoting an inactive form of the enzyme by preventing localization to the membrane or access to the PDK-1-dependent phosphorylation site.

Additional studies are needed to further elucidate the relationship between PIK3CA mutations and AKT as there are three distinct isoforms (AKT1, AKT2, and AKT3) and each may have different effects on tumor growth and/or cytotoxicity. To date, PIK3CA mutations have been most closely linked to AKT1 in in vitro cell models [26, 28-30], with a single study examining all three isoforms revealing that AKT1 may be most affected by PIK3CA mutations [27]. However, isoform specific functions have been delineated through various laboratory studies. For example, RNA interference (RNAi) mediated gene knockdown of AKT1 in breast cancer cell lines has been shown to increase cell motility [121], and AKT1 knockout mice are small in size and infertile [122]. In addition, AKT2 knockout mice develop diabetes mellitus [123], while AKT3 knockout mice exhibit abnormal brain development [124]. Although AKT isoforms are highly homologous, selective AKT1 and 2 inhibitors have been developed and have shown promise in vitro [97]. Further study of the relative effects of inhibiting different AKT isoforms in cancers harboring PIK3CA mutations will be required, as the potential for significant toxicities remains obstacle challenge for using AKT inhibitors as effective targeted therapies for these cancers.

mTOR Inhibitors

mTOR is a downstream effector of PIK3CA and is very important for many cellular processes, including cell proliferation [125] and angiogenesis [126]. The mTOR pathway has been shown to be activated by PIK3CA mutations in both chicken embryo fibroblasts [30] and colorectal cancer cells [26]. Therefore, blocking the mTOR pathway may prove to be an effective strategy for targeting aberrant growth signaling in cancers with PIK3CA mutations.

mTOR inhibitors are the most mature in terms of their development and clinical use of potential compounds targeting cancers harboring PIK3CA mutations. Rapamycin, the prototypical mTOR inhibitor, was initially developed in the early 1970’s as an antifungal agent [127] and was later FDA approved as an immunosuppressive therapy [128]. As seen with most first generation inhibitors, rapamycin exhibited undesirable pharmacological properties. More reliable rapamycin analogs, or “rapalogs”, were developed and have shown cytostatic activity in preclinical models and anti-tumor activity when used in combination with chemotherapies [71]. RAD001 (everolimus), CCI-779 (temsirolimus), and AP-23573 (deferolimus), are three rapalogs that have exhibited variable and moderate success. Previously, the best evidence of response was shown in the treatment of advanced renal cancers with temsirolimus [129]. More recently, the BOLERO-2 trial demonstrated that in metastatic breast cancer patients refractory to hormone therapies, the addition of everolimus to exemestane lead to improved progression free survival compared to women taking exemestane alone [130].

The proposed mechanism of action of rapamycin and its analogs is the formation of a complex with the FK506-binding protein (FKBP12). This complex can then bind the C-terminal region of mTOR, interfering with the kinase activity of the multimeric mTORC1 complex but not with mTORC2 [131]. Furthermore, identifying biomarkers that predict for response to mTOR inhibitors is paramount for the development of these agents, as response to mTOR inhibitors has been highly variable. Theoretically, PIK3CA mutations may lend themselves as predictive biomarkers for response to mTOR inhibitors. For example, rapamycin and its analogs prevent the transformation of chicken embryo fibroblasts expressing PIK3CA mutations [30], inhibit PIK3CA induced tumor growth in chicken embryos [31], and reduce the formation of abnormal human breast epithelial cell acini induced by mutant PIK3CA overexpression [28].

Despite the clinical potential of mTOR inhibitors, significant hurdles for their further development still exist. mTOR forms two complexes within cells, mTORC1 and mTORC2 [132]. mTORC1 is known to mediate a negative feedback loop with PI3K/AKT signaling, and therefore inhibiting mTOR pharmacologically causes a paradoxical upregulation of PI3K/AKT growth promoting signaling [133]. Additionally, mTORC2 directly phosphorylates AKT [134] but is only rarely inhibited by rapamycin and its analogs in a cell/tissue type dependent manner [135]. Despite the initial promise of the previously mentioned mTORC1 allosteric inhibitors, feedback activation of PI3K and AKT persists via mTORC2. This suggests a potential shift in the focus of compound development to ATP-competitive compounds that can inactivate both complexes and completely abrogate mTOR signaling. PP242, its derivative INK128, as well as several other pan-mTOR inhibitors such as AZD-8055 and OSI-027, are ATP-competitive compounds in early phase clinical trials for solid malignancies including breast cancers [136, 137]. Other effective strategies for targeting this complex signaling pathway in the future may include combining current mTOR inhibitors with either newer PI3K/AKT inhibitors or more traditional therapies, such as chemotherapy and endocrine therapy. In addition, it is possible that the current development of new inhibitors capable of blocking mTORC2, or alternatively both mTORC1 and mTORC2, could provide a more effective therapeutic regimen.

Possibilities of Novel Dual Inhibition

Due to the genetic instability of most human cancers, it can be expected that any targeted therapy when used as a single agent will ultimately succumb to drug resistance. However, the development of targeted therapies with minimal side effects would hopefully enable the combinatorial use of multiple drugs with non-overlapping toxicities, to effectively treat and eradicate the disease. Recently, it was demonstrated that the combination of p110α and mTOR inhibitors could prevent the increase in AKT signaling caused by mTOR inhibition alone [138]. A single molecule, PI-103, was found to effectively inhibit both p110α and mTOR, and its efficacy has been shown in gliomas, ovarian cancer, and breast cancer in in vitro and in vivo models [138, 139]. This class of molecules, capable of inhibiting multiple targets within the PI3K pathway, may prove to be an effective strategy for targeting cancer cells containing PIK3CA mutations.

In addition, combination therapies with existing anti-neoplastic drugs are also being explored. For example, PIK3CA mutations have been positively correlated with increased expression of both estrogen receptor alpha (ERα) and HER2 in the NCI 60 panel of cancer cell lines [140] as well breast tumor samples [141]. Hormonal therapies and trastuzumab are currently used to target ERα and HER2 respectively. However, it has been shown that increased AKT signaling is associated with resistance to both of these therapies [142-145], and it is therefore possible that PIK3CA mutations may actually confer resistance to these drugs. Recently, a study demonstrated that estrogen deprivation in ER (+) breast tumors increased the apoptotic effects of the PI3K and dual-PI3K/mTOR inhibitors, BKM120 and BGT226 respectively. Increased drug efficacy was observed when used in combination with fulvestrant, a selective estrogen receptor down-regulator (SERD) [146]. Recently, correlations have been made between the presence of mutant PIK3CA and activation of the MAPK pathway [147, 148]. While the development of combination therapies is still in its infancy, recent studies suggest that dual inhibition of both the PI3K and MAPK pathways presents the best strategy for targeting cancer with PIK3CA mutations, a testable hypothesis currently the focus of clinical trials.

CONCLUSIONS

Due to their high frequency in many cancers, PIK3CA mutations are a prime candidate for targeted therapeutics. However given the fact that these mutations have only recently been accurately characterized, much work lies ahead to fully elucidate their biological and clinical significance. The process of discovery for developing targeted PIK3CA therapies remains in its infancy. However, the wealth of potential targets along the PI3K pathway is certainly enticing from a developmental therapeutics viewpoint, and the opportunity exists to significantly impact cancer care with future success in this arena.

CONFLICT OF INTEREST

S.C., J.C., J.P.G. and D.P.C. declare no conflict of interests. B.H.P. is a member of the scientific advisory board of Horizon Discovery, LTD and receives royalties and payments for services provided and licensed reagents. B.H.P. is also a paid consultant for GlaxoSmithKline. The terms of these agreements are regulated by The Johns Hopkins University policies on conflicts of interest.

ACKNOWLEGMENTS

This work was supported by The Flight Attendant’s Medical Research Institute (FAMRI), NIH/NCI CA109274, the Summer Running Fund, The Susan G. Komen for the Cure Foundation, Mary Kay Ash Charitable Foundation and the Avon Foundation. J.P.G. is a past recipient of a Department of Defense Breast Cancer Research Program Predoctoral Fellowship Award W81XWH-06-1-0325. S.C., J.C. and D.P.C. were previously supported on NIH Institutional Training Grants CA067751, GM008752, CA09071 and an American Cancer Society Young Investigator Award (D.P.C.).

DISCLOSURE

The chapter submitted for the eBook Series entitled “Advances in Cancer Drug Targets, Volume 1” is an update of our article in Current Cancer Drug Targets, Volume 8, Issue 8, 2008, with additional text and references.

Abbreviations

EGFRepidermal growth factor receptorPI3Kphosphatidylinositol 3-kinasemTORmammalian target of rapamycinPIKKPI-3 kinase-like kinaseRNAiRNA interferenceERαestrogen receptor alpha

REFERENCES

[1]O’Brien S.G., et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia.N. Engl. J. Med.2003348119941004[2]Paez J.G., et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.Science2004304567614971500[3]Lynch T.J., et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.N. Engl. J. Med.20043502121292139[4]Romond E.H., et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer.N. Engl. J. Med.20053531616731684[5]Piccart-Gebhart M.J., et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer.N. Engl. J. Med.20053531616591672[6]Mass R.D., et al. Evaluation of clinical outcomes according to HER2 detection by fluorescence in situ hybridization in women with metastatic breast cancer treated with trastuzumab.Clin. Breast Cancer200563240246[7]Samuels Y., et al. High frequency of mutations of the PIK3CA gene in human cancers.Science20043045670554554[8]Bachman K.E., et al. The PIK3CA gene is mutated with high frequency in human breast cancers.Cancer Biol. Ther.200438772775[9]Lee J.W., et al. PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas.Oncogene200524814771480[10]Levine D.A., et al. Frequent mutation of the PIK3CA gene in ovarian and breast cancers.Clin. Cancer Res.200511828752878[11]Campbell I.G., et al. Mutation of the PIK3CA gene in ovarian and breast cancer.Cancer Res.2004642176787681[12]Velho S., et al. The prevalence of PIK3CA mutations in gastric and colon cancer.Eur. J. Cancer2005411116491654[13]Li V.S.W., et al. Mutations of PIK3CA in gastric adenocarcinoma.BMC Cancer2005•••5[14]Kita D., et al. PIK3CA alterations in primary (de novo) and secondary glioblastomas.Acta Neuropathol.20071133295302[15]Gallia G.L., et al. PIK3CA gene mutations in pediatric and adult glioblastoma multiforme.Mol. Cancer Res.2006410709714[16]Dam V., et al. Mutations in PIK3CA are infrequent in neuroblastoma.BMC Cancer2006•••6[17]Pang J.C.S., et al. Rare mutation of PIK3CA in meningiomas.Acta Neuropathol.20061113284285[18]Hartmann C., et al. PIK3CA mutations in oligodendroglial tumours.Neuropathol. Appl. Neurobiol.2006322209212[19]Hartmann C., et al. PIK3CA mutations in glioblastoma multiforme.Acta Neuropathol.20051096639642[20]Mueller W., et al. Mutations of the PIK3CA gene are rare in human glioblastoma.Acta Neuropathol.20051096654655[21]Broderick D.K., et al. Mutations of PIK3CA in anaplastic oligodendrogliomas, high-grade astrocytomas, and medulloblastomas.Cancer Res.2004641550485050[22]Campbell I.G., et al. Mutation of the PIK3CA gene in ovarian and breast cancer.Cancer Res.2004642176787681[23]Gustin J.P., Cosgrove D.P., Park B.H.. The PIK3CA gene as a mutated target for cancer therapy.Curr. Cancer Drug Targets200888733740[24]Isakoff S.J., et al. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells.Cancer Res.200565231099211000[25]Guo X.N., et al. Mutant PIK3CA-bearing colon cancer cells display increased metastasis in an orthotopic model.Cancer Res.2007671258515858[26]Ikenoue T., et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer.Cancer Res.2005651145624567[27]Samuels Y., et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells.Cancer Cell200576561573[28]Isakoff S.J., et al. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells.Cancer Res.200565231099211000[29]Zhao J.J., et al. The oncogenic properties of mutant p110 alpha and p110 beta phosphatidylinositol 3-kinases in human mammary epithelial cells.Proc. Natl. Acad. Sci. USA2005102511844318448[30]Kang S.Y., Bader A.G., Vogt P.K.. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic.Proc. Natl. Acad. Sci. USA20051023802807[31]Bader A.G., Kang S.Y., Vogt P.K.. Cancer-specific mutations in PIK3CA are oncogenic in vivo.Proc. Natl. Acad. Sci. USA2006103514751479[32]Miled N., et al. Mechanism of two classes of cancer mutations in the phosphoinositide 3-kinase catalytic subunit.Science20073175835239242[33]Kang S., et al. Mutated PI 3-kinases - Cancer targets on a silver platter.Cell Cycle200544578581[34]Berns K., et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer.Cancer Cell2007124395402[35]Maruyama N., et al. Clinicopathologic analysis of breast cancers with PIK3CA mutations in Japanese women.Clin. Cancer Res.2007132408414[36]Pérez-Tenorio G., et al. PIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancer.Clin. Cancer Res.2007131235773584[37]Li S.Y., et al. PIK3CA mutations in breast cancer are associated with poor outcome.Breast Cancer Res. Treat.20069619195[38]Kato S., et al. PIK3CA mutation is predictive of poor survival in patients with colorectal cancer.Int. J. Cancer2007121817711778[39]Barbareschi M., et al. Different Prognostic Roles of Mutations in the Helical and Kinase Domains of the PIK3CA Gene in Breast Carcinomas.Clin. Cancer Res.2007132060646069[40]Davies M., Hennessy B., Mills G.B.. Point mutations of protein kinases ion and individualised cancer therapy.Expert Opin. Pharmacother.200671622432261[41]Pao W., et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib.Proc. Natl. Acad. Sci. USA2004101361330613311[42]Weinstein I.B.. Cancer: Addiction to oncogenes - The Achilles heal of cancer.Science200229755786364[43]Huang C.H., et al. The structure of a human p110alpha/p85alpha complex elucidates the effects of oncogenic PI3Kalpha mutations.Science2007318585717441748[44]Schultz R.M., et al. In vitro and in vivo Antitumor-Activity of the Phosphatidylinositol-3-Kinase Inhibitor, Wortmannin.Anticancer Res.199515411351139[45]Semba S., et al. The in vitro and in vivo effects of 2-(4-morpholinyl)-8-phenyl-chromone (LY294002), a specific inhibitor of phosphatidylinositol 3 '-kinase, in human colon cancer cells.Clin. Cancer Res.20028619571963[46]Hu L.M., et al. In vivo and in vitro ovarian carcinoma growth inhibition by a phosphatidylinositol 3-kinase inhibitor (LY294002).Clin. Cancer Res.200063880886[47]Workman P.. Inhibiting the phosphoinositide 3-kinase pathway for cancer treatment.Biochem. Soc. Trans.200432393396[48]Gharbi S.I., et al. Exploring the specificity of the PI3K family inhibitor LY294002.Biochem. J.20074041521[49]Sarkaria J.N., et al. Inhibition of phosphoinositide 3-kinase related kinases by the radiosensitizing agent wortmannin.Cancer Res.1998581943754382[50]Banin S., et al. Enhanced phosphorylation of p53 by ATN in response to DNA damage.Science1998281538316741677[51]Brunn G.J., et al. Direct inhibition of the signaling functions of the mammalian target of rapamycin by the phosphoinositide 3-kinase inhibitors, wortmannin and LY294002.EMBO J.1996151952565267[52]Hartley K.O., et al. DNA-Dependent Protein-Kinase Catalytic Subunit - a Relative of Phosphatidylinositol 3-Kinase and the Ataxia-Telangiectasia Gene-Product.Cell1995825849856[53]Arcaro A., Wymann M.P.. Wortmannin is a potent phosphatidylinositol 3-kinase inhibitor: the role of phosphatidylinositol 3,4,5-trisphosphate in neutrophil responses.Biochem. J.1993296Pt 2297301[54]Vlahos C.J., et al. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002).J. Biol. Chem.1994269752415248[55]Garlich J.R., et al. A vascular targeted pan phosphoinositide 3-kinase inhibitor prodrug, SF1126, with antitumor and antiangiogenic activity.Cancer Res.2008681206215[56]Ihle N.T., et al. Molecular pharmacology and antitumor activity of PX-866, a novel inhibitor of phosphoinositide-3-kinase signaling.Mol. Cancer Ther.200437763772[57]Hayakawa M., et al. Synthesis and biological evaluation of sulfonylhydrazone-substituted imidazo 1,2-a pyridines as novel PI3 kinase p110 alpha inhibitors.Bioorg. Med. Chem.2007151758375844[58]Hayakawa M., et al. Synthesis and biological evaluation of pyrido 3 ',2′: 4,5 furo 3,2-d pyrimidine derivatives as novel PI3 kinase p110 alpha inhibitors.Bioorg. Med. Chem. Lett.200717924382442[59]Hayakawa M., et al. Synthesis and biological evaluation of imidazo 1,2-a pyridine derivatives as novel PI3 kinase p110 alpha inhibitors.Bioorg. Med. Chem.2007151403412[60]Hayakawa M., et al. Synthesis and biological evaluation of 4-morpholino-2-phenylquinazolines and related derivatives as novel PI3 kinase p110 alpha inhibitors.Bioorg. Med. Chem.2006142068476858[61]Marion F., et al. Liphagal, a selective inhibitor of PI3 kinase alpha isolated from the sponge Aka coralliphaga: Structure elucidation and biomimetic synthesis.Org. Lett.200682321324[62]Ihle N.T., et al. The phosphatidylinositol-3-kinase inhibitor PX-866 overcomes resistance to the epidermal growth factor receptor inhibitor gefitinib in A-549 human non-small cell lung cancer xenografts.Mol. Cancer Ther.20054913491357[63]Knight Z.A., et al. A pharmacological map of the PI3-K family defines a role for p110 alpha in insulin signaling.Cell20061254733747[64]Ihle N.T., et al. Molecular pharmacology and antitumor activity of PX-866, a novel inhibitor of phosphoinositide-3-kinase signaling.Mol. Cancer Ther.200437763772[65]Garcia-Echeverria C., Sellers W.R.. Drug discovery approaches targeting the PI3K/Akt pathway in cancer.Oncogene2008274155115526[66]O’Brien C., et al. Predictive biomarkers of sensitivity to the phosphatidylinositol 3′ kinase inhibitor GDC-0941 in breast cancer preclinical models.Clin. Cancer Res.2010161436703683[67]Chakrabarty A., et al. Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors.Proc. Natl. Acad. Sci. USA2012109827182723[68]Maira S.M., et al. Identification and characterization of NVP-BKM120, an orally available pan class I PI3-Kinase inhibitor.Mol. Cancer Ther.2012[69]Castillo J.J., Furman M., Winer E.S.. CAL-101: a phosphatidylinositol-3-kinase p110-delta inhibitor for the treatment of lymphoid malignancies.Expert Opin. Investig. Drugs20122111522[70]Howes A.L., et al. The phosphatidylinositol 3-kinase inhibitor, PX-866, is a potent inhibitor of cancer cell motility and growth in three-dimensional cultures.Mol. Cancer Ther.20076925052514[71]Miller T.W., et al. Mutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer.Breast Cancer Res.2011136224[72]Arteaga C.L.. Abstracts of the IX Madrid Breast Cancer ConferenceMadrid, SpainNovember 16-17, 2011O115112[73]Lin H., et al. Synthesis and structure-activity relationships of imidazo[1,2-a]pyrimidin-5(1H)-ones as a novel series of beta isoform selective phosphatidylinositol 3-kinase inhibitors.Bioorg. Med. Chem. Lett.2012[74]Guerreiro A.S., et al. A sensitized RNA interference screen identifies a novel role for the PI3K p110gamma isoform in medulloblastoma cell proliferation and chemoresistance.Mol. Cancer Res.201197925935[75]Tanaka H., et al. The selective class I PI3K inhibitor CH5132799 targets human cancers harboring oncogenic PIK3CA mutations.Clin. Cancer Res.2011171032723281[76]Carvalho S., et al. PIKing the right isoform: the emergent role of the p110beta subunit in breast cancer.Virchows Arch.20104563235243[77]Engelman J.A., Luo J., Cantley L.C.. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism.Nat. Rev. Genet.200678606619[78]McMullen J.R., Jay P.Y.. PI3K(p110 alpha) inhibitors as anti-cancer agents - Minding the heart.Cell Cycle200768910913[79]Altomare D.A., Testa J.R.. Perturbations of the AKT signaling pathway in human cancer.Oncogene2005245074557464[80]Staal S.P.. Molecular-Cloning of the Akt Oncogene and Its Human Homologs Akt1 and Akt2 - Amplification of Akt1 in a Primary Human Gastric Adenocarcinoma.Proc. Natl. Acad. Sci. USA1987841450345037[81]Lin X.D., et al. Discovery of 2-pyrimidyl-5-amidothiophenes as potent inhibitors for AKT: Synthesis and SAR studies.Bioorg. Med. Chem. Lett.2006161641634168[82]Zhu G.D., et al. Isoquinoline-pyridine-based protein kinase B/Akt antagonists: SAR and in vivo antitumor activity.Bioorg. Med. Chem. Lett.2006161231503155[83]Hideshima T., et al. Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells.Blood20061071040534062[84]Mandal M., et al. The Akt inhibitor KP372-1 inhibits proliferation and induces apoptosis and anoikis in squamous cell carcinoma of the head and neck.Oral Oncol.2006424430439[85]Zeng Z.H., et al. Simultaneous inhibition of PDK1/AKT and Fms-like tyrosine kinase 3 signaling by a small-molecule KP372-1 induces mitochondrial dysfunction and apoptosis in acute myelogenous leukemia.Cancer Res.200666737373746[86]Koul D., et al. Inhibition of Akt survival pathway by a small-molecule inhibitor in human glioblastoma.Mol. Cancer Ther.200653637644[87]Gills J.J., et al. Spectrum of activity and molecular correlates of response to phosphatidylinositol ether lipid analogues, novel lipid-based inhibitors of Akt.Mol. Cancer Ther.200653713722[88]Li Q., et al. Synthesis and structure-activity relationship of 3,4 '-bispyridinylethylenes: Discovery of a potent 3-isoquinolinylpyridine inhibitor of protein kinase B (PKB/Akt) for the treatment of cancer.Bioorg. Med. Chem. Lett.200616720002007[89]Li Q., et al. Discovery of trans-3,4 '-bispyridinylethylenes as potent and novel inhibitors of protein kinase B (PKB/Akt) for the treatment of cancer: Synthesis and biological evaluation.Bioorg. Med. Chem. Lett.200616616791685[90]Tang H.J., et al. A small molecule compound inhibits AKT pathway in ovarian cancer cell lines.Gynecol. Oncol.20061002308317[91]Georgakis G.V., et al. Inhibition of the phosphatidylinositol-3 kinase/Akt promotes G1 cell cycle arrest and apoptosis in Hodgkin lymphoma.Br. J. Haematol.20061324503511[92]Shi Y., et al. Optimal classes of chemotherapeutic agents sensitized by specific small-molecule inhibitors of Akt in vitro and in vivo.Neoplasia20057119921000[93]Luo Y., et al. Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo.Mol. Cancer Ther.200546977986[94]Mandal M., et al. The Akt inhibitor KP372-1 suppresses Akt activity and cell proliferation and induces apoptosis in thyroid cancer cells.Br. J. Cancer2005921018991905[95]Shin I., et al. Proapoptotic activity of cell-permeable anti-Akt single-chain antibodies.Cancer Res.200565728152824[96]Lindsley C.W., et al. Allosteric Akt (PKB) inhibitors: discovery and SAR of isozyme selective inhibitors.Bioorg. Med. Chem. Lett.2005153761764[97]Barnett S.F., et al. Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors.Biochem. J.2005385399408[98]Hiromura M., et al. Inhibition of Akt kinase activity by a peptide spanning the beta A strand of the proto-oncogene TCL1.J. Biol. Chem.2004279515340753418[99]Meuillet E.J., et al. In vivo molecular pharmacology and antitumor activity of the targeted Akt inhibitor PX-316.Oncol. Res.20041410513527[100]Jin X., et al. Inhibition of AKT survival pathway by a small molecule inhibitor in human endometrial cancer cells.Br. J. Cancer2004911018081812[101]Shaw Y.J., et al. Pharmacological exploitation of the alpha 1-adrenoreceptor antagonist doxazosin to develop a novel class of antitumor agents that block intracellular protein kinase B/Akt activation.J. Med. Chem.2004471844534462[102]Yang L., et al. Akt/protein kinase B signaling inhibitor-2, a selective small molecule inhibitor of Akt signaling with antitumor activity in cancer cells overexpressing Akt.Cancer Res.2004641343944399[103]Castillo S.S., et al. Preferential inhibition of Akt and killing of Akt-dependent cancer cells by rationally designed phosphatidylinositol ether lipid analogues.Cancer Res.200464827822792[104]Luo Y., et al. Pseudosubstrate peptides inhibit Akt and induce cell growth inhibition.Biochemistry200443512541263[105]Kondapaka S.B., et al. Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation.Mol. Cancer Ther.200321110931103[106]Meuillet E.J., et al. Specific inhibition of the Akt1 pleckstrin homology domain by D-3-deoxy-phosphatidyl-myo-inositol analogues.Mol. Cancer Ther.200324389399[107]Ruiter G.A., et al. Anti-cancer alkyl-lysophospholipids inhibit the phosphatidylinositol 3-kinase-Akt/PKB survival pathway.Anticancer Drugs2003142167173[108]Reuveni H., et al. Toward a PKB inhibitor: Modification of a selective PKA inhibitor by rational design.Biochemistry200241321030410314[109]Vink S.R., et al. Radiosensitization of squamous cell carcinoma by the alkylphospholipid perifosine in cell culture and xenografts.Clin. Cancer Res.200612516151622[110]Li X., et al. Enhancement of antitumor activity of the anti-EGF receptor monoclonal antibody cetuximab/C225 by perifosine in PTEN-deficient cancer cells.Oncogene2006254525535[111]Rahmani M., et al. Coadministration of histone deacetylase inhibitors and perifosine synergistically induces apoptosis in human leukemia cells through Akt and ERK1/2 inactivation and the generation of ceramide and reactive oxygen species.Cancer Res.200565624222432[112]Smorenburg C.H., et al. Phase II study of miltefosine 6% solution as topical treatment of skin metastases in breast cancer patients.Anticancer Drugs20001110825828[113]Clive S., Gardiner J., Leonard R.C.F.. Miltefosine as a topical treatment for cutaneous metastases in breast carcinoma.Cancer Chemother. Pharmacol.199944S29S30[114]Terwogt J.M.M., et al. Phase II trial of topically applied miltefosine solution in patients with skin-metastasized breast cancer.Br. J. Cancer1999797-811581161[115]Dummer R., et al. Topical Administration of Hexadecylphosphocholine in Patients with Cutaneous Lymphomas - Results of a Phase-I/Ii Study.J. Am. Acad. Dermatol.1993296963970[116]Feun L.G., et al. A Phase-Ii Trial of Tricyclic Nucleoside Phosphate in Patients with Advanced Squamous-Cell Carcinoma of the Cervix - a Gynecologic-Oncology-Group Study.Cancer Clin. Trials1993166506508[117]Feun L.G., et al. Phase-1 Study of Tricyclic Nucleoside Phosphate Using a 5-Day Continuous Infusion Schedule.Cancer Res.198444836083612[118]Carol H., et al. Initial testing (stage 1) of the Akt inhibitor GSK690693 by the pediatric preclinical testing program.Pediatr. Blood Cancer201055713291337[119]Chan T.O., et al. Resistance of Akt kinases to dephosphorylation through ATP-dependent conformational plasticity.Proc. Natl. Acad. Sci. USA201110846E1120E1127[120]Yap T.A., et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors.J. Clin. Oncol.2011293546884695[121]Yoeli-Lerner M., et al. Akt blocks breast cancer cell motility and invasion through the transcription factor NFAT.Mol. Cell2005204539550[122]Chen W.S., et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the akt1 gene.Genes Dev.2001151722032208[123]Cho H., et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB beta).Science2001292552217281731[124]Tschopp O., et al. Essential role of protein kinase B gamma (PKB gamma/Akt3) in postnatal brain development but not in glucose homeostasis.Development20051321329432954[125]Wullschleger S., Loewith R., Hall M.N.. TOR signaling in growth and metabolism.Cell20061243471484[126]Bernardi R., et al. PML inhibits HIF-1 alpha translation and neoangiogenesis through repression of mTOR.Nature20064427104779785[127]Sehgal S.N., Baker H., Vezina C.. Rapamycin (Ay-22,989), a New Antifungal Antibiotic.2. Fermentation, Isolation and Characterization.J. Antibiot. (Tokyo)19752810727732[128]Neuhaus P., Klupp J., Langrehr J.M.. mTOR inhibitors: An overview.Liver Transpl.200176473484[129]Hudes G., et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma.N. Engl. J. Med.20073562222712281[130]Baselga J., et al. Everolimus in Postmenopausal Hormone-Receptor-Positive Advanced Breast Cancer.N. Engl. J. Med.2011[131]Wiederrecht G.J., et al. Mechanism of action of rapamycin: new insights into the regulation of G1-phase progression in eukaryotic cells.Prog. Cell Cycle Res.199515371[132]Sarbassov D.D., Ali S.M., Sabatini D.M.. Growing roles for the mTOR pathway.Curr. Opin. Cell Biol.2005176596603[133]Manning B.D.. Balancing Akt with S6K: implications for both metabolic diseases and tumorigenesis.J. Cell Biol.20041673399403[134]Sarbassov D.D., et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex.Science2005307571210981101[135]Sarbassov D.D., et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB.Mol. Cell2006222159168[136]Benjamin D., et al. Rapamycin passes the torch: a new generation of mTOR inhibitors.Nat. Rev. Drug Discov.20111011868880[137]O’Regan R., Hawk N.N.. mTOR inhibition in breast cancer: unraveling the complex mechanisms of mTOR signal transduction and its clinical implications in therapy.Expert Opin. Ther. Targets2011157859872[138]Fan Q.W., et al. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma.Cancer Cell200695341349[139]Raynaud F.I., et al. Pharmacologic characterization of a potent inhibitor of class I phosphatidylinositide 3-kinases.Cancer Res.2007671258405850[140]Whyte D.B., Holbeck S.L.. Correlation of PIK3Ca mutations with gene expression and drug sensitivity in NCI-60 cell lines.Biochem. Biophys. Res. Commun.20063402469475[141]Saal L.H., et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma.Cancer Res.200565725542559[142]Chan C.T., Metz M.Z., Kane S.E.. Differential sensitivities of trastuzumab (Herceptin (R))-resistant human breast cancer cells to phosphoinositide-3 kinase (PI-3K) and epidermal growth factor receptor (EGFR) kinase inhibitors.Breast Cancer Res. Treat.2005912187201[143]Jordan N.J., et al. Increased constitutive activity of PKB/Akt in tamoxifen resistant breast cancer MCF-7 cells.Breast Cancer Res. Treat.2004872167180[144]Yakes F.M., et al. Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt is required for antibody-mediated effects on p27, cyclin D1, and antitumor action.Cancer Res.2002621441324141[145]Perez-Tenorio G., Stal O.. Activation of AKT/PKB in breast cancer predicts a worse outcome among endocrine treated patients.Br. J. Cancer2002864540545[146]Sanchez C.G., et al. Preclinical modeling of combined phosphatidylinositol-3-kinase inhibition with endocrine therapy for estrogen receptor-positive breast cancer.Breast Cancer Res.2011132R21[147]Gupta S., et al. Binding of ras to phosphoinositide 3-kinase p110alpha is required for ras-driven tumorigenesis in mice.Cell20071295957968[148]Higgins M.J., et al. PIK3CA mutations and EGFR overexpression predict for lithium sensitivity in human breast epithelial cells.Cancer Biol. Ther.2011113358367

AKT Signaling in Regulating Angiogenesis

Bing-Hua Jiang*,Ling-Zhi Liu*
Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA19107, USA

Abstract