1
|
Banerjee N, Roy L, Panda S, Roychowdhury T, Chatterjee S. In Silico-Designed G-Quadruplex Targeting Peptide Attenuates VEGF-A Expression, Preventing Angiogenesis in Cancer Cells. Chem Biol Drug Des 2024; 104:e70018. [PMID: 39704035 DOI: 10.1111/cbdd.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 12/21/2024]
Abstract
Vascular endothelial growth factor-A (VEGF-A) is a growth factor and pluripotent cytokine that promotes angiogenesis in cancer cells, transitioning to an angiogenic phenotype. The binding of VEGF-A protein to VEGF receptors (VEGFR-1 and VEGFR-2) initiates a cascade of events that stimulates angiogenesis by facilitating the migration and enhancing the permeability of endothelial cells. The proximal promoter of the VEGF gene encompasses a 36-base pair region (from -85 to -50) that can form a stable G-quadruplex (G4) structure in specific conditions. The activity of the VEGF promoter is reliant on this structure. During cancer progression, the VEGF-A G4 succumbs to cellular pressure and fails to maintain a stable structure. This shifts the balance to form a duplex structure, increasing the transcription rate. Earlier research has tried to develop small-molecule ligands to target and stabilise G4, demonstrating the possibility of suppressing VEGF expression. However, they either lack specificity or toxic. Peptides, on the other hand, are significantly less studied as G4 binders. Here, we designed a peptide that successfully binds and stabilises the VEGF-A G4 while reducing its gene expression. This further alters the expression fate of the VEGF-A signalling cascade and blocks angiogenesis in cancer cells. We employed high-resolution nuclear magnetic resonance (NMR) spectroscopy and molecular dynamics simulation to elucidate the chemical details of G4-peptide interaction. In addition, we used qPCR and western blot techniques to investigate the expression pattern of the molecules implicated in the VEGF-A signalling cascade. The study explores the intricate relationship between peptides and quadruplex structures, revealing valuable insights that can improve the design of pharmacophores targeting the dynamic quadruplex structure. The results of our study are encouraging, opening possibilities for advancements in, the characterisation and optimisation of peptides as G-quadruplex ligands in view of their potential therapeutic uses.
Collapse
Affiliation(s)
- Nilanjan Banerjee
- Non-Coding Genome Group, Department of Structural Biology, CEITEC-Central European Institute of Technology, Brno, Czech Republic
| | - Laboni Roy
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| | - Suman Panda
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Subhrangsu Chatterjee
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| |
Collapse
|
2
|
Zhuang X, Guo Y, Sun X, Chen J, Xie S, Yang F, Li J. Promising Proteolysis-Targeting Chimera for Mutant p53-R175H. ACS OMEGA 2024; 9:45138-45146. [PMID: 39554460 PMCID: PMC11561642 DOI: 10.1021/acsomega.4c06177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
The tumor suppressor protein p53 is among the most commonly mutated proteins across a variety of cancer types. Notably, the p53 R175H mutation ranks as one of the most prevalent hotspot mutations. Proteolysis-targeting chimeras (PROTACs) represent a class of bifunctional molecules capable of harnessing the cellular ubiquitin-proteasome pathway to facilitate targeted protein degradation. Despite the potential of PROTACs, limited research has been directed toward the degradation of the p53-R175H mutant protein. In this study, we developed a series of peptide-based PROTACs, leveraging known peptide ligands for both the p53-R175H mutation and the E3 ubiquitin ligase VHL. Our findings indicate that one of these peptide-based PROTACs is capable of directing the p53-R175H protein to the proteasome for degradation within a recombinant expression system. Moreover, by synthesizing a fusion peptide PROTAC molecule that incorporates a membrane-penetrating peptide, we have demonstrated its ability to traverse cellular membranes and subsequently reduce the levels of the p53-R175H mutant protein. Importantly, the degradation of p53-R175H was found to mitigate the cellular migration and invasion. In summary, our study introduces a novel class of protein degraders and establishes a foundational framework for the therapeutic management of cancers associated with p53 mutations.
Collapse
Affiliation(s)
- Xinzhe Zhuang
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| | - Yidan Guo
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| | - Xiaozi Sun
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| | - Jie Chen
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| | - Songbo Xie
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
- Department
of Ophthalmology, Tianjin Medical University General Hospital, Laboratory
of Molecular Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University, Tianjin 300052, China
| | - Fengtang Yang
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| | - Jingrui Li
- School
of Life Sciences and Medicine, Shandong
University of Technology, Zibo 255000, China
| |
Collapse
|
3
|
Carrera-Aubesart A, Gallo M, Defaus S, Todorovski T, Andreu D. Topoisomeric Membrane-Active Peptides: A Review of the Last Two Decades. Pharmaceutics 2023; 15:2451. [PMID: 37896211 PMCID: PMC10610229 DOI: 10.3390/pharmaceutics15102451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
In recent decades, bioactive peptides have been gaining recognition in various biomedical areas, such as intracellular drug delivery (cell-penetrating peptides, CPPs) or anti-infective action (antimicrobial peptides, AMPs), closely associated to their distinct mode of interaction with biological membranes. Exploiting the interaction of membrane-active peptides with diverse targets (healthy, tumoral, bacterial or parasitic cell membranes) is opening encouraging prospects for peptides in therapeutics. However, ordinary peptides formed by L-amino acids are easily decomposed by proteases in biological fluids. One way to sidestep this limitation is to use topoisomers, namely versions of the peptide made up of D-amino acids in either canonic (enantio) or inverted (retroenantio) sequence. Rearranging peptide sequences in this fashion provides a certain degree of native structure mimicry that, in appropriate contexts, may deliver desirable biological activity while avoiding protease degradation. In this review, we will focus on recent accounts of membrane-active topoisomeric peptides with therapeutic applications as CPP drug delivery vectors, or as antimicrobial and anticancer candidates. We will also discuss the most common modes of interaction of these peptides with their membrane targets.
Collapse
Affiliation(s)
- Adam Carrera-Aubesart
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Maria Gallo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Sira Defaus
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Toni Todorovski
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - David Andreu
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| |
Collapse
|
4
|
Di Stasi R, De Rosa L, D'Andrea LD. Structure-Based Design of Peptides Targeting VEGF/VEGFRs. Pharmaceuticals (Basel) 2023; 16:851. [PMID: 37375798 DOI: 10.3390/ph16060851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/03/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) and its receptors (VEGFRs) play a main role in the regulation of angiogenesis and lymphangiogenesis. Furthermore, they are implicated in the onset of several diseases such as rheumatoid arthritis, degenerative eye conditions, tumor growth, ulcers and ischemia. Therefore, molecules able to target the VEGF and its receptors are of great pharmaceutical interest. Several types of molecules have been reported so far. In this review, we focus on the structure-based design of peptides mimicking VEGF/VEGFR binding epitopes. The binding interface of the complex has been dissected and the different regions challenged for peptide design. All these trials furnished a better understanding of the molecular recognition process and provide us with a wealth of molecules that could be optimized to be exploited for pharmaceutical applications.
Collapse
Affiliation(s)
| | - Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, 80131 Napoli, Italy
| | | |
Collapse
|
5
|
Guo L, Overholser J, Good AJ, Ede NJ, Kaumaya PTP. Preclinical Studies of a Novel Human PD-1 B-Cell Peptide Cancer Vaccine PD1-Vaxx From BALB/c Mice to Beagle Dogs and to Non-Human Primates (Cynomolgus Monkeys). Front Oncol 2022; 12:826566. [PMID: 35646678 PMCID: PMC9137037 DOI: 10.3389/fonc.2022.826566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
Abstract
Immunotherapy with monoclonal antibodies to checkpoint inhibitors against the PD-1/PD-L1 signaling pathway is a landmark achievement in cancer therapy. Some anti-PD-1 inhibitors such as nivolumab and pembrolizumab have shown clinical success, in a percentage of patients with prolonged survival rates. However, adverse effects accompany these benefits. In this case, strategies with lower toxicity and increased specificity are urgently required. Cancer vaccines have the ability to stimulate the native immune system and in particular, an engineered B-cell epitope can elicit high-affinity polyclonal antibodies with similar efficacy to PD-1 monoclonal antibodies in murine animal models. We have previously designed and synthesized a unique B-cell vaccine, PD1-Vaxx [MVF-PD-1(92-110)], and we have tested the immunogenicity and antitumor properties in CT26 colon cancer BALB/c syngeneic mice model. This manuscript provides results from comprehensive preclinical pharmacology studies encompassing primary and secondary pharmacodynamics, biodistribution, and safety studies. The results from these preclinical studies support the use of PD1-Vaxx in a first-in-human clinical trial in patients with non-small cell lung cancer (NSCLC). A phase I trial in patients with NSCLC has commenced.
Collapse
Affiliation(s)
- Linlin Guo
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jay Overholser
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | | | | - Pravin T. P. Kaumaya
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- James Comprehensive Cancer Center, Columbus, OH, United States
- *Correspondence: Pravin T. P. Kaumaya, ; orcid.org/0000-0002-8647-3911
| |
Collapse
|
6
|
Recent Applications of Retro-Inverso Peptides. Int J Mol Sci 2021; 22:ijms22168677. [PMID: 34445382 PMCID: PMC8395423 DOI: 10.3390/ijms22168677] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022] Open
Abstract
Natural and de novo designed peptides are gaining an ever-growing interest as drugs against several diseases. Their use is however limited by the intrinsic low bioavailability and poor stability. To overcome these issues retro-inverso analogues have been investigated for decades as more stable surrogates of peptides composed of natural amino acids. Retro-inverso peptides possess reversed sequences and chirality compared to the parent molecules maintaining at the same time an identical array of side chains and in some cases similar structure. The inverted chirality renders them less prone to degradation by endogenous proteases conferring enhanced half-lives and an increased potential as new drugs. However, given their general incapability to adopt the 3D structure of the parent peptides their application should be careful evaluated and investigated case by case. Here, we review the application of retro-inverso peptides in anticancer therapies, in immunology, in neurodegenerative diseases, and as antimicrobials, analyzing pros and cons of this interesting subclass of molecules.
Collapse
|
7
|
Guo L, Kaumaya PTP. First prototype checkpoint inhibitor B-cell epitope vaccine (PD1-Vaxx) en route to human Phase 1 clinical trial in Australia and USA: exploiting future novel synergistic vaccine combinations. Br J Cancer 2021; 125:152-154. [PMID: 33772155 DOI: 10.1038/s41416-021-01342-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 11/09/2022] Open
Abstract
We developed a PD-1 B-cell epitope vaccine (PD1-Vaxx) to rival nivolumab therapy which has received ethics approvals for a Phase 1 clinical trial in Australia. The US FDA granted Investigational New Drug approval to Imugene Ltd for clinical testing in NSCLC. We demonstrated synergistic vaccine combinations with an HER-2 targeted vaccine (B-Vaxx).
Collapse
Affiliation(s)
- Linlin Guo
- Department of Obstetrics and Gynecology, Division of Vaccine Research, The Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Pravin T P Kaumaya
- Department of Obstetrics and Gynecology, Division of Vaccine Research, The Ohio State Wexner Medical Center, Columbus, OH, USA. .,Arthur G. James Cancer Hospital & Solove Research, Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
8
|
Malik AJ, Aronica PGA, Verma CS. DStabilize: A Web Resource to Generate Mirror Images of Biomolecules. Structure 2020; 28:1358-1360.e2. [PMID: 32783952 DOI: 10.1016/j.str.2020.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/13/2020] [Accepted: 07/23/2020] [Indexed: 10/23/2022]
Abstract
Peptides comprising D-amino acids have been shown to be resistant to proteolysis. This makes them potential candidates as probes of cellular interactions, notably protein-biomolecule interactions. However, the empirical conversion of the amino acids that constitute a peptide from L-forms to D-forms will result in abrogation of the normal interactions made by the L-amino acids due to side-chain orientation changes that are associated with the changes in chirality. These interactions can be preserved by reversing the sequence of the D-peptide. We present a web server (http://dstabilize.bii.a-star.edu.sg/) that allows users to convert between L-proteins and D-proteins and for sequence reversal of D-peptides, along with the capability of performing other empirical geometric transforms. This resource allows the user to generate structures of interest easily for subsequent in silico processing.
Collapse
Affiliation(s)
- Ashar J Malik
- Bioinformatics Institute (A(∗)STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore
| | - Pietro G A Aronica
- Bioinformatics Institute (A(∗)STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore
| | - Chandra S Verma
- Bioinformatics Institute (A(∗)STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
9
|
Kaumaya PTP. B-cell epitope peptide cancer vaccines: a new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol 2020; 16:1767-1791. [PMID: 32564612 PMCID: PMC7426751 DOI: 10.2217/fon-2020-0224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
In light of the numerous US FDA-approved humanized monoclonal antibodies (mAbs) for cancer immunotherapy, it is surprising that the advancement of B-cell epitope vaccines designed to elicit a natural humoral polyclonal antibody response has not gained traction in the immune-oncology landscape. Passive immunotherapy with humanized mAbs (Trastuzumab [Herceptin®]; Pertuzumab [Perjeta®]) has provided clinical benefit to breast cancer patients, albeit with significant shortcomings including toxicity problems and resistance, high costs, sophisticated therapeutic regimen and long half-life. The role of B-cell humoral immunity in cancer is under appreciated and underdeveloped. We have advanced the idea of active immunotherapy with chimeric B-cell epitope peptides incorporating a 'promiscuous' T-cell epitope that elicits a polyclonal antibody response, which provides safe, cost-effective therapeutic advantage over mAbs. We have created a portfolio of validated B-cell peptide epitopes against multiple receptor tyrosine kinases (HER-1, HER-3, IGF-1R and VEGF). We have successfully translated two HER-2 combination B-cell peptide vaccines in Phase I and II clinical trials. We have recently developed an effective novel PD-1 vaccine. In this article, I will review our approaches and strategies that focus on B-cell epitope cancer vaccines.
Collapse
Affiliation(s)
- Pravin TP Kaumaya
- Department of Obstetrics & Gynecology, College of Medicine, Wexner Medical Center, The James Cancer Hospital & Solove Research Institute, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Qu J, Ren X, Xue F, He Y, Zhang R, Zheng Y, Huang H, Wang W, Zhang J. Specific Knockdown of α-Synuclein by Peptide-Directed Proteasome Degradation Rescued Its Associated Neurotoxicity. Cell Chem Biol 2020; 27:751-762.e4. [DOI: 10.1016/j.chembiol.2020.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/21/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022]
|
11
|
Ghadam M, Sardari S, Shokrgozar MA, Mahdavi MS. Design of Anti-Angiogenic Peptidomimetics and Evaluation their Biological Activity by In Vitro Assays. Avicenna J Med Biotechnol 2020; 12:91-98. [PMID: 32431793 PMCID: PMC7229457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND One of the important therapeutic approaches in cancer field is development of compounds which can block the initial tumor growth and the progression of tumor metastasis with no side effects. Thus, the recent study was carried out to design anti-VEGFR2-peptidomimetics as the most significant factor of angiogenesis process- and evaluate their biological activity by in vitro assays. METHODS We designed anti-VEGFR2 peptidomimetics with anti-angiogenic activity, including compound P (lactam derivative) and compound T (indole derivative) by using in silico methods. Then, the inhibitory activity on angiogenesis was evaluated by using angiogenesis specific assays such as Human Umbilical Vein Endothelial Cell (HUVEC) proliferation, tube formation in Matrigel, MTT and Real-Time PCR. IC50 values of the compounds were also determined by cytotoxicity plot in MTT assay. RESULTS Compounds P and T inhibited HUVEC cell proliferation and viability in a dose-dependent manner. The IC50 for compound T and compound P in HUVEC cell line were 113 and 115 μg/ml, respectively. Tube formation assay revealed that both compounds can inhibit angiogenesis effectively. The results of Real-Time PCR also showed these compounds are able to inhibit the expression of CD31 gene in HUVEC cell line. CONCLUSION Our study suggested that compounds P and T may act as therapeutic molecules, or lead compounds for development of angiogenesis inhibitors in VEGF-related diseases.
Collapse
Affiliation(s)
- Mona Ghadam
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soroush Sardari
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran,Corresponding authors: Soroush Sardari, Ph.D., Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Ali Shokrgozar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran,Mohammad Ali Shokrgozar, Ph.D., National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran, Tel: +98 9122632484, E-mail: ;
| | - Mahdiyeh Sadat Mahdavi
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
12
|
Laengsri V, Nantasenamat C, Schaduangrat N, Nuchnoi P, Prachayasittikul V, Shoombuatong W. TargetAntiAngio: A Sequence-Based Tool for the Prediction and Analysis of Anti-Angiogenic Peptides. Int J Mol Sci 2019; 20:E2950. [PMID: 31212918 PMCID: PMC6628072 DOI: 10.3390/ijms20122950] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 11/21/2022] Open
Abstract
Cancer remains one of the major causes of death worldwide. Angiogenesis is crucial for the pathogenesis of various human diseases, especially solid tumors. The discovery of anti-angiogenic peptides is a promising therapeutic route for cancer treatment. Thus, reliably identifying anti-angiogenic peptides is extremely important for understanding their biophysical and biochemical properties that serve as the basis for the discovery of new anti-cancer drugs. This study aims to develop an efficient and interpretable computational model called TargetAntiAngio for predicting and characterizing anti-angiogenic peptides. TargetAntiAngio was developed using the random forest classifier in conjunction with various classes of peptide features. It was observed via an independent validation test that TargetAntiAngio can identify anti-angiogenic peptides with an average accuracy of 77.50% on an objective benchmark dataset. Comparisons demonstrated that TargetAntiAngio is superior to other existing methods. In addition, results revealed the following important characteristics of anti-angiogenic peptides: (i) disulfide bond forming Cys residues play an important role for inhibiting blood vessel proliferation; (ii) Cys located at the C-terminal domain can decrease endothelial formatting activity and suppress tumor growth; and (iii) Cyclic disulfide-rich peptides contribute to the inhibition of angiogenesis and cell migration, selectivity and stability. Finally, for the convenience of experimental scientists, the TargetAntiAngio web server was established and made freely available online.
Collapse
Affiliation(s)
- Vishuda Laengsri
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Pornlada Nuchnoi
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
13
|
Vascular Endothelial Growth Factor Sequestration Enhances In Vivo Cartilage Formation. Int J Mol Sci 2017; 18:ijms18112478. [PMID: 29160845 PMCID: PMC5713444 DOI: 10.3390/ijms18112478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/01/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Autologous chondrocyte transplantation for cartilage repair still has unsatisfactory clinical outcomes because of inter-donor variability and poor cartilage quality formation. Re-differentiation of monolayer-expanded human chondrocytes is not easy in the absence of potent morphogens. The Vascular Endothelial Growth Factor (VEGF) plays a master role in angiogenesis and in negatively regulating cartilage growth by stimulating vascular invasion and ossification. Therefore, we hypothesized that its sole microenvironmental blockade by either VEGF sequestration by soluble VEGF receptor-2 (Flk-1) or by antiangiogenic hyperbranched peptides could improve chondrogenesis of expanded human nasal chondrocytes (NC) freshly seeded on collagen scaffolds. Chondrogenesis of several NC donors was assessed either in vitro or ectopically in nude mice. VEGF blockade appeared not to affect NC in vitro differentiation, whereas it efficiently inhibited blood vessel ingrowth in vivo. After 8 weeks, in vivo glycosaminoglycan deposition was approximately two-fold higher when antiangiogenic approaches were used, as compared to the control group. Our data indicates that the inhibition of VEGF signaling, independently of the specific implementation mode, has profound effects on in vivo NC chondrogenesis, even in the absence of chondroinductive signals during prior culture or at the implantation site.
Collapse
|
14
|
|
15
|
Tino AB, Chitcholtan K, Sykes PH, Garrill A. Resveratrol and acetyl-resveratrol modulate activity of VEGF and IL-8 in ovarian cancer cell aggregates via attenuation of the NF-κB protein. J Ovarian Res 2016; 9:84. [PMID: 27906095 PMCID: PMC5134119 DOI: 10.1186/s13048-016-0293-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/26/2016] [Indexed: 12/14/2022] Open
Abstract
Background Key features of advanced ovarian cancer include metastasis via cell clusters in the abdominal cavity and increased chemoresistance. Resveratrol and derivatives of resveratrol have been shown to have antitumour properties. The purpose of this study was to investigate the effect of resveratrol and acetyl-resveratrol on 3D cell aggregates of ovarian cancer, and establish if NF-κB signalling may be a potential target. Methods Poly-HEMA coated wells were used to produce 3D aggregates of two ovarian cancer cell lines, SKOV-3 and OVCAR-5. The aggregates were exposed to 10, 20 or 30 μM resveratrol or acetyl-resveratrol for 2, 4 or 6 days. Cell growth and metabolism were measured then ELISA, western blot and immunofluorescence were utilised to evaluate VEGF, IL-8 and NF-κB levels. Results Resveratrol and acetyl-resveratrol reduced cell growth and metabolism of SKOV-3 aggregates in a dose- and time-dependent manner. After 6 days all three doses of both compounds inhibited cell growth. This growth inhibition correlated with the attenuated secretion of VEGF and a decrease of NF-κB protein levels. Conversely, the secretion of IL-8 increased with treatment. The effects of the compounds were limited in OVCAR-5 cell clusters. Conclusions The results suggest that resveratrol and its derivative acetyl-resveratrol may inhibit in vitro 3D cell growth of certain subtypes of ovarian cancer, and growth restriction may be associated with the secretion of VEGF under the control of the NF-κB protein.
Collapse
Affiliation(s)
- Alexandria B Tino
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Kenny Chitcholtan
- Obstetrics and Gynaecology Department Christchurch Women's Hospital, Private Bag 4711, Christchurch, 8140, New Zealand.
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8140, New Zealand
| |
Collapse
|
16
|
Kaumaya PTP. A paradigm shift: Cancer therapy with peptide-based B-cell epitopes and peptide immunotherapeutics targeting multiple solid tumor types: Emerging concepts and validation of combination immunotherapy. Hum Vaccin Immunother 2016; 11:1368-86. [PMID: 25874884 DOI: 10.1080/21645515.2015.1026495] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is a recognizable and urgent need to speed the development and application of novel, more efficacious anti-cancer vaccine therapies that inhibit tumor progression and prevent acquisition of tumor resistance. We have created and established a portfolio of validated peptide epitopes against multiple receptor tyrosine kinases and we have identified the most biologically effective combinations of EGFR (HER-1), HER-2, HER-3, VEGF and IGF-1R peptide vaccines/mimics to selectively inhibit multiple receptors and signaling pathways. The strategy is based on the use of chimeric conformational B-cell epitope peptides incorporating "promiscuous" T-cell epitopes that afford the possibility of generating an enduring immune response, eliciting protein-reactive high-affinity anti-peptide antibodies as potential vaccines and peptide mimics that act as antagonists to receptor signaling that drive cancer metastasis. In this review we will summarize our ongoing studies based on the development of combinatorial immunotherapeutic strategies that act synergistically to enhance immune-mediated tumor killing aimed at addressing mechanisms of tumor resistance for several tumor types.
Collapse
Affiliation(s)
- Pravin T P Kaumaya
- a Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center ; Columbus , OH , USA
| |
Collapse
|
17
|
González PL, Carvajal C, Cuenca J, Alcayaga-Miranda F, Figueroa FE, Bartolucci J, Salazar-Aravena L, Khoury M. Chorion Mesenchymal Stem Cells Show Superior Differentiation, Immunosuppressive, and Angiogenic Potentials in Comparison With Haploidentical Maternal Placental Cells. Stem Cells Transl Med 2015; 4:1109-21. [PMID: 26273064 PMCID: PMC4572900 DOI: 10.5966/sctm.2015-0022] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/22/2015] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Mesenchymal stem cells (MSCs) of placental origin have become increasingly translational owing to their abundance and accessibility. MSCs of different origin share several features but also present biological differences that might point to distinct clinical properties. Hence, mixing fetal and maternal cells from the same placenta can lead to contradicting results. We analyzed the biological characteristics of haploidentical MSCs isolated from fetal sources, including the umbilical cord (UC-MSCs) and chorion (Ch-MSCs), compared with maternal decidua MSCs (Dc-MSCs). All MSCs were analyzed for general stem cell properties. In addition, immunosuppressive capacity was assessed by the inhibition of T-cell proliferation, and angiogenic potential was evaluated in a Matrigel transplantation assay. The comparison between haploidentical MSCs displayed several distinct features, including (a) marked differences in the expression of CD56, (b) a higher proliferative capacity for Dc-MSCs and UC-MSCs than for Ch-MSCs, (c) a diversity of mesodermal differentiation potential in favor of fetal MSCs, (d) a higher capacity for Ch-MSCs to inhibit T-cell proliferation, and (e) superior angiogenic potential of Ch-MSCs evidenced by a higher capability to form tubular vessel-like structures and an enhanced release of hepatocyte growth factor and vascular endothelial growth factor under hypoxic conditions. Our results suggest that assessing the prevalence of fetomaternal contamination within placental MSCs is necessary to increase robustness and limit side effects in their clinical use. Finally, our work presents evidence positioning fetoplacental cells and notably Ch-MSCs in the forefront of the quest for cell types that are superior for applications in regenerative medicine. SIGNIFICANCE This study analyzed the biological characteristics of mesenchymal stem cells (MSCs) isolated from fetal and maternal placental origins. The findings can be summarized as follows: (a) important differences were found in the expression of CD56, (b) a different mesodermal differentiation potential was found in favor of fetal MSCs, (c) a higher immunosuppressive capacity for chorion MSCs was noted, and (d) superior angiogenic potential of Ch-MSCs was observed. These results suggest that assessing the prevalence of fetomaternal contamination within placental MSCs is necessary to increase robustness and limit side effects in their clinical use. The evidence should allow clinicians to view fetoplacental cells, notably Ch-MSCs, favorably as candidates for use in regenerative medicine.
Collapse
Affiliation(s)
- Paz L González
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Catalina Carvajal
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Fernando E Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Jorge Bartolucci
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Lorena Salazar-Aravena
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Cells for Cells, Santiago, Chile; Facultad de Medicina Universidad de los Andes y Clínica Universidad de Los Andes, Santiago, Chile; Consorcio Regenero, Santiago, Chile
| |
Collapse
|
18
|
Panagopoulos V, Zinonos I, Leach DA, Hay SJ, Liapis V, Zysk A, Ingman WV, DeNichilo MO, Evdokiou A. Uncovering a new role for peroxidase enzymes as drivers of angiogenesis. Int J Biochem Cell Biol 2015; 68:128-38. [PMID: 26386352 DOI: 10.1016/j.biocel.2015.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 09/01/2015] [Accepted: 09/16/2015] [Indexed: 10/23/2022]
Abstract
Peroxidases are heme-containing enzymes released by activated immune cells at sites of inflammation. To-date their functional role in human health has mainly been limited to providing a mechanism for oxidative defence against invading bacteria and other pathogenic microorganisms. Our laboratory has recently identified a new functional role for peroxidase enzymes in stimulating fibroblast migration and collagen biosynthesis, offering a new insight into the causative association between inflammation and the pro-fibrogenic events that mediate tissue repair and regeneration. Peroxidases are found at elevated levels within and near blood vessels however, their direct involvement in angiogenesis has never been reported. Here we report for the first time that myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are readily internalised by human umbilical vein endothelial cells (HUVEC) where they promote cellular proliferation, migration, invasion, and stimulate angiogenesis both in vitro and in vivo. These pro-angiogenic effects were attenuated using the specific peroxidase inhibitor 4-ABAH, indicating the enzyme's catalytic activity is essential in mediating this response. Mechanistically, we provide evidence that MPO and EPO regulate endothelial FAK, Akt, p38 MAPK, ERK1/2 phosphorylation and stabilisation of HIF-2α, culminating in transcriptional regulation of key angiogenesis pathways. These findings uncover for the first time an important and previously unsuspected role for peroxidases as drivers of angiogenesis, and suggest that peroxidase inhibitors may have therapeutic potential for the treatment of angiogenesis related diseases driven by inflammation.
Collapse
Affiliation(s)
- Vasilios Panagopoulos
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Irene Zinonos
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Damien A Leach
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Shelley J Hay
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Vasilios Liapis
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Aneta Zysk
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Wendy V Ingman
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Mark O DeNichilo
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia
| | - Andreas Evdokiou
- School of Medicine, Discipline of Surgery, Basil Hetzel Institute, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
19
|
Overholser J, Ambegaokar KH, Eze SM, Sanabria-Figueroa E, Nahta R, Bekaii-Saab T, Kaumaya PTP. Anti-Tumor Effects of Peptide Therapeutic and Peptide Vaccine Antibody Co-targeting HER-1 and HER-2 in Esophageal Cancer (EC) and HER-1 and IGF-1R in Triple-Negative Breast Cancer (TNBC). Vaccines (Basel) 2015; 3:519-43. [PMID: 26350593 PMCID: PMC4586465 DOI: 10.3390/vaccines3030519] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/29/2015] [Accepted: 06/29/2015] [Indexed: 12/19/2022] Open
Abstract
Despite the promise of targeted therapies, there remains an urgent need for effective treatment for esophageal cancer (EC) and triple-negative breast cancer (TNBC). Current FDA-approved drugs have significant problems of toxicity, safety, selectivity, efficacy and development of resistance. In this manuscript, we demonstrate that rationally designed peptide vaccines/mimics are a viable therapeutic strategy for blocking aberrant molecular signaling pathways with high affinity, specificity, potency and safety. Specifically, we postulate that novel combination treatments targeting members of the EGFR family and IGF-1R will yield significant anti-tumor effects in in vitro models of EC and TNBC possibly overcoming mechanisms of resistance. We show that the combination of HER-1 and HER-2 or HER-1 and IGF-1R peptide mimics/vaccine antibodies exhibited enhanced antitumor properties with significant inhibition of tumorigenesis in OE19 EC and MDA-MB-231 TNBC cell lines. Our work elucidates the mechanisms of HER-1/IGF-1R and HER-1/HER-2 signaling in these cancer cell lines, and the promising results support the rationale for dual targeting with HER-1 and HER-2 or IGF-1R as an improved treatment regimen for advanced therapy tailored to difference types of cancer.
Collapse
Affiliation(s)
- Jay Overholser
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Kristen Henkins Ambegaokar
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Siobhan M Eze
- Department of Pharmacology, Emory University and Winship Cancer Institute, Atlanta, GA 30322, USA.
| | - Eduardo Sanabria-Figueroa
- Molecular and Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Rita Nahta
- Department of Pharmacology, Emory University and Winship Cancer Institute, Atlanta, GA 30322, USA.
- Molecular and Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Tanios Bekaii-Saab
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Pravin T P Kaumaya
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Foy KC, Miller MJ, Overholser J, Donnelly SM, Nahta R, Kaumaya PT. IGF-1R peptide vaccines/mimics inhibit the growth of BxPC3 and JIMT-1 cancer cells and exhibit synergistic antitumor effects with HER-1 and HER-2 peptides. Oncoimmunology 2014; 3:e956005. [PMID: 25941587 DOI: 10.4161/21624011.2014.956005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022] Open
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) plays a crucial role in cellular growth, proliferation, transformation, and inhibition of apoptosis. A myriad of human cancer types have been shown to overexpress IGF-1R, including breast and pancreatic adenocarcinoma. IGF-1R signaling interferes with numerous receptor pathways, rendering tumor cells resistant to chemotherapy, anti-hormonal therapy, and epidermal growth factor receptor (EGFR, also known as HER-1) and v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2, (ERBB2, best known as HER-2) -targeted therapies. Targeting the IGF:IGF-1R axis with innovative peptide inhibitors and vaccine antibodies thus represents a promising therapeutic strategy to overcome drug resistance and to provide new avenues for individualized and combinatorial treatment strategies. In this study, we designed, synthesized, and characterized several B-cell epitopes from the IGF-1:IGF-1R axis. The chimeric peptide epitopes were highly immunogenic in outbred rabbits, eliciting high levels of peptide vaccine antibodies. The IGF-1R peptide antibodies and peptide mimics inhibited cell proliferation and receptor phosphorylation, induced apoptosis and antibody-dependent cellular cytotoxicity (ADCC), and significantly inhibited tumor growth in the transplantable BxPC-3 pancreatic and JIMT-1 breast cancer models. Our results showed that the peptides and antibodies targeting residues 56-81 and 233-251 are potential therapeutic and vaccine candidates for the treatment of IGF-1R-expressing cancers, including those that are resistant to the HER-2-targeted antibody, trastuzumab. Additionally, we found additive antitumor effects for the combination treatment of the IGF-1R 56-81 epitope with HER-1-418 and HER-2-597 epitopes. Treatment with the IGF-1R/HER-1 or IGF-1R/HER-2 combination inhibited proliferation, invasion, and receptor phosphorylation, and induced apoptosis and ADCC, to a greater degree than single agents.
Collapse
Affiliation(s)
- Kevin Chu Foy
- Department of Obstetrics and Gynecology; The Ohio State University ; Columbus, OH USA
| | - Megan J Miller
- Department of Obstetrics and Gynecology; The Ohio State University ; Columbus, OH USA ; Department of Microbiology; The Ohio State University ; Columbus, OH USA
| | - Jay Overholser
- Department of Obstetrics and Gynecology; The Ohio State University ; Columbus, OH USA
| | | | - Rita Nahta
- Department of Pharmacology; Emory University ; Atlanta, GA USA
| | - Pravin Tp Kaumaya
- Department of Obstetrics and Gynecology; The Ohio State University ; Columbus, OH USA ; Department of Microbiology; The Ohio State University ; Columbus, OH USA ; James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center; The Ohio State University ; Columbus, OH USA
| |
Collapse
|
21
|
Miller MJ, Foy KC, Overholser JP, Nahta R, Kaumaya PT. HER-3 peptide vaccines/mimics: Combined therapy with IGF-1R, HER-2, and HER-1 peptides induces synergistic antitumor effects against breast and pancreatic cancer cells. Oncoimmunology 2014; 3:e956012. [PMID: 25941588 DOI: 10.4161/21624011.2014.956012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022] Open
Abstract
The human epidermal growth factor receptor 3 (HER-3/ErbB3) is a unique member of the human epidermal growth factor family of receptors, because it lacks intrinsic kinase activity and ability to heterodimerize with other members. HER-3 is frequently upregulated in cancers with epidermal growth factor receptor (EGFR/HER-1/ErbB1) or human epidermal growth factor receptor 2 (HER-2/ErBB2) overexpression, and targeting HER-3 may provide a route for overcoming resistance to agents that target EGFR or HER-2. We have previously developed vaccines and peptide mimics for HER-1, HER-2 and vascular endothelial growth factor (VEGF). In this study, we extend our studies by identifying and evaluating novel HER-3 peptide epitopes encompassing residues 99-122, 140-162, 237-269 and 461-479 of the HER-3 extracellular domain as putative B-cell epitopes for active immunotherapy against HER-3 positive cancers. We show that the HER-3 vaccine antibodies and HER-3 peptide mimics induced antitumor responses: inhibition of cancer cell proliferation, inhibition of receptor phosphorylation, induction of apoptosis and antibody dependent cellular cytotoxicity (ADCC). Two of the HER-3 epitopes 237-269 (domain II) and 461-479 (domain III) significantly inhibited growth of xenografts originating from both pancreatic (BxPC3) and breast (JIMT-1) cancers. Combined therapy of HER-3 (461-471) epitope with HER-2 (266-296), HER-2 (597-626), HER-1 (418-435) and insulin-like growth factor receptor type I (IGF-1R) (56-81) vaccine antibodies and peptide mimics show enhanced antitumor effects in breast and pancreatic cancer cells. This study establishes the hypothesis that combination immunotherapy targeting different signal transduction pathways can provide effective antitumor immunity and long-term control of HER-1 and HER-2 overexpressing cancers.
Collapse
Key Words
- ADCC, antibody dependent, cellular cytotoxicity
- Antibodies
- ECD, extracellular domain
- ELISA, enzyme-linked immunosorbent assay
- FDA, Federal Drug Administration
- HER-1
- HER-1 (EGFR or ErbB1), human epidermal growth factor receptor
- HER-2
- HER-2 (ErbB2), human epidermal growth factor receptor 2
- HER-3 (ErbB3), human epidermal growth factor receptor 3
- HER-3 (erbb3)
- HER-4 (ErbB4), human epidermal growth factor receptor 4
- HPLC, high-pressure liquid chromatography
- IGF-1R
- Immunotherapy
- MALDI, matrix-assisted laser desorption/ionization
- MVF, Measles virus fusion protein
- RTK, receptor tyrosine kinase
- TKIs, Tyrosine kinase inhibitors.
- immunogenicity
- mAb, monocolonal antibody
- peptide vaccines
- peptidomimetics
- receptor tyrosine kinases
Collapse
Affiliation(s)
- Megan Jo Miller
- Department of Microbiology; The Ohio State University , Columbus, OH USA
| | - Kevin C Foy
- Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center ; Columbus, OH USA
| | - Jay P Overholser
- Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center ; Columbus, OH USA
| | - Rita Nahta
- Department of Pharmacology; Emory University , Atlanta, GA USA
| | - Pravin Tp Kaumaya
- Department of Microbiology; The Ohio State University , Columbus, OH USA ; Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center ; Columbus, OH USA ; The James Cancer Hospital and Solove Research Institute; and the Comprehensive Cancer Center; The Ohio State University , Columbus, OH USA
| |
Collapse
|
22
|
Su T, Long Y, Deng C, Feng L, Zhang X, Chen Z, Li C. Construction of a two-in-one liposomal system (TWOLips) for tumor-targeted combination therapy. Int J Pharm 2014; 476:241-52. [DOI: 10.1016/j.ijpharm.2014.09.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/13/2014] [Accepted: 09/28/2014] [Indexed: 10/24/2022]
|
23
|
Zhou S, Zhang P, Liang P, Huang X. The expression of miR-125b regulates angiogenesis during the recovery of heat-denatured HUVECs. Burns 2014; 41:803-11. [PMID: 25468475 DOI: 10.1016/j.burns.2014.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/23/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023]
Abstract
BACKGROUND In previous studies we found that miR-125b was down-regulated in denatured dermis of deep partial thickness burn patients. Moreover, miR-125b inhibited tumor-angiogenesis associated with the decrease of ERBB2 and VEGF expression in ovarian cancer cells and breast cancer cells, etc. In this study, we investigated the expression patterns and roles of miR-125b during the recovery of denatured dermis and heat-denatured human umbilical vein endothelial cells (HUVECs). METHODS Deep partial thickness burns in Sprague-Dawley rats and the heat-denatured cells (52°C, 35 s) were used for analysis. Western blot analysis and real-time PCR were applied to evaluate the expression of miR-125b and ERBB2 and VEGF. The ability of angiogenesis in heat-denatured HUVECs was analyzed by scratch wound healing and tube formation assay after pri-miR-125b or anti-miR-125b transfection. RESULTS miR-125b expression was time-dependent during the recovery of heat-denatured dermis and HUVECs. Moreover, miR-125b regulated ERBB2 mRNA and Protein Expression and regulated angiogenesis association with regulating the expression of VEGF in heat-denatured HUVECs. CONCLUSIONS Taken together our results show that the expression of miR-125b is time-dependent and miR-125b plays a regulatory role of angiogenesis during wound healing after burns.
Collapse
Affiliation(s)
- Situo Zhou
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Pengfei Liang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Xiaoyuan Huang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
24
|
Foy KC, Miller MJ, Moldovan N, Bozanovic T, Carson WE, Kaumaya PTP. Immunotherapy with HER-2 and VEGF peptide mimics plus metronomic paclitaxel causes superior antineoplastic effects in transplantable and transgenic mouse models of human breast cancer. Oncoimmunology 2014; 1:1004-1016. [PMID: 23170249 PMCID: PMC3494615 DOI: 10.4161/onci.21057] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
HER-2 and the vascular endothelial factor receptor (VEGF) represent validated targets for the therapy of multiple tumor types and inhibitors of these receptors have gained increasing importance in the clinic. In this context, novel bioactive agents associated with better therapeutic outcomes and improved safety profile are urgently required. Specifically engineered HER-2- and VEGF-derived peptides in combination with low-dose chemotherapy might provide a substantial impact on tumor metastasis and cancer progression. We tested the antitumor effects of HER-2 and VEGF peptide mimics in combination with metronomic paclitaxel in both PyMT and Balb/c murine model challenged with TUBO cells. The combination of low-dose paclitaxel and HER-2 or VEGF peptide mimics had greater inhibitory effects than either agent alone. Peptide treatment caused virtually no cardiotoxic effects, while paclitaxel and the anti-HER-2 antibody trastuzumab (Herceptin), exerted consistent cardiotoxicity. The combination regimen also promoted significant reductions in tumor burden and prolonged survival rates in both transgenic and transplantable tumor models. Tumor weights were significantly reduced in mice treated with HER-2 peptides alone, and even more in animals that received HER-2 peptide with low-dose paclitaxel, which alone had no significant effects on tumor growth in the transgenic model. Specifically engineered native peptide sequences from HER-2 and VEGF used in combination with metronomic paclitaxel demonstrate enhanced anticancer efficacy and an encouraging safety profile. This novel approach to targeted therapy may offer new avenues for the treatment of breast cancer and other solid tumors that overexpress HER-2 and VEGF.
Collapse
Affiliation(s)
- Kevin C Foy
- Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center; Columbus, OH USA ; Department of Microbiology; The Ohio State University Wexner Medical Center; Columbus, OH USA
| | - Megan J Miller
- Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center; Columbus, OH USA ; Department of Microbiology; The Ohio State University Wexner Medical Center; Columbus, OH USA
| | - Nicanor Moldovan
- Department of Internal Medicine; The Ohio State University Wexner Medical Center; Columbus, OH USA
| | - Tatjana Bozanovic
- Department of Gynecology and Obstetrics; School of Medicine; University of Belgrade; Belgrade, Serbia
| | - William E Carson
- James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center; The Ohio State University; Columbus, OH USA
| | - Pravin T P Kaumaya
- Department of Obstetrics and Gynecology; The Ohio State University Wexner Medical Center; Columbus, OH USA ; Department of Microbiology; The Ohio State University Wexner Medical Center; Columbus, OH USA ; James Cancer Hospital and Solove Research Institute and the Comprehensive Cancer Center; The Ohio State University; Columbus, OH USA
| |
Collapse
|
25
|
Kaumaya PTP. Bridging oncology and immunology: expanding horizons with innovative peptide vaccines and peptidomimetics. Immunotherapy 2014; 5:1159-63. [PMID: 24188668 DOI: 10.2217/imt.13.128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Pravin T P Kaumaya
- The Ohio State University Wexner Medical Center, Department of Obstetrics & Gynecology, 410 W 10th Avenue N729, Columbus, OH 43210, USA and The Comprehensive Cancer Center, The Ohio State University, 300 W 10th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
26
|
Schnatbaum K, Schmoldt HU, Daneschdar M, Plum LM, Jansong J, Zerweck J, Kühne Y, Masch A, Wenschuh H, Fiedler M, Türeci Ö, Sahin U, Reimer U. Peptide microarrays enable rapid mimotope optimization for pharmacokinetic analysis of the novel therapeutic antibody IMAB362. Biotechnol J 2014; 9:545-54. [DOI: 10.1002/biot.201300456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/06/2013] [Accepted: 02/04/2014] [Indexed: 01/27/2023]
|
27
|
Rapid and reversible knockdown of endogenous proteins by peptide-directed lysosomal degradation. Nat Neurosci 2014; 17:471-80. [PMID: 24464042 PMCID: PMC3937121 DOI: 10.1038/nn.3637] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/16/2013] [Indexed: 12/13/2022]
Abstract
Rapid and reversible methods for altering the level of endogenous proteins are critically important for studying biological systems and developing therapeutics. Here, we describe a membrane permeable targeting peptide-based method that rapidly and reversibly knocks down endogenous proteins through chaperone-mediated autophagy in vitro and in vivo. We demonstrated the specificity, efficacy and generalizability of the method by showing efficient knockdown of various proteins including death associated protein kinase 1 (160kDa), scaffolding protein PSD-95 (95kDa) and α-synuclein (18kDa) with their respective targeting peptides in a dose-, time- and lysosomal activity-dependent manner in neuronal cultures. More significantly, we showed that when given systemically, the peptide system efficiently knocked down the targeted protein in the brain of intact rats. Our study provides a robust and convenient research tool to manipulate endogenous protein levels, and may also lead to the development of protein knockdown-based novel therapeutics for treating various human diseases.
Collapse
|
28
|
Cao Z, Shang B, Zhang G, Miele L, Sarkar FH, Wang Z, Zhou Q. Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1836:273-286. [PMID: 23933263 DOI: 10.1016/j.bbcan.2013.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/22/2013] [Accepted: 08/01/2013] [Indexed: 12/12/2022]
Abstract
Robust neovascularization and lymphangiogenesis have been found in a variety of aggressive and metastatic tumors. Endothelial sprouting angiogenesis is generally considered to be the major mechanism by which new vasculature forms in tumors. However, increasing evidence shows that tumor vasculature is not solely composed of endothelial cells (ECs). Some tumor cells acquire processes similar to embryonic vasculogenesis and produce new vasculature through vasculogenic mimicry, trans-differentiation of tumor cells into tumor ECs, and tumor cell-EC vascular co-option. In addition, tumor cells secrete various vasculogenic factors that induce sprouting angiogenesis and lymphangiogenesis. Vasculogenic tumor cells actively participate in the formation of vascular cancer stem cell niche and a premetastatic niche. Therefore, tumor cell-mediated neovascularization and lymphangiogenesis are closely associated with tumor progression, cancer metastasis, and poor prognosis. Vasculogenic tumor cells have emerged as key players in tumor neovascularization and lymphangiogenesis and play pivotal roles in tumor progression and cancer metastasis. However, the mechanisms underlying tumor cell-mediated vascularity as they relate to tumor progression and cancer metastasis remain unclear. Increasing data have shown that various intrinsic and extrinsic factors activate oncogenes and vasculogenic genes, enhance vasculogenic signaling pathways, and trigger tumor neovascularization and lymphangiogenesis. Collectively, tumor cells are the instigators of neovascularization. Therefore, targeting vasculogenic tumor cells, genes, and signaling pathways will open new avenues for anti-tumor vasculogenic and metastatic drug discovery. Dual targeting of endothelial sprouting angiogenesis and tumor cell-mediated neovascularization and lymphangiogenesis may overcome current clinical problems with anti-angiogenic therapy, resulting in significantly improved anti-angiogenesis and anti-cancer therapies.
Collapse
Affiliation(s)
- Zhifei Cao
- Cyrus Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu 215006, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Crosstalk between HER2 signaling and angiogenesis in breast cancer: molecular basis, clinical applications and challenges. Curr Opin Oncol 2013; 25:313-24. [PMID: 23518595 DOI: 10.1097/cco.0b013e32835ff362] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Angiogenesis is an essential hallmark of cancer. Targeting angiogenesis has proven its efficacy in the modern therapeutic paradigm. HER2 positive breast cancer, in particular, is a challenging disease in which resistance to standard therapy has been attributed to parallel and downstream signaling cascades including angiogenesis. This review explores the molecular mechanisms underlying crosstalk between HER2 signaling and angiogenesis. It highlights the role of angiogenesis in the emerging resistance to anti-HER2 therapy. It surveys the current repertoire of clinical trials involving use of combination of anti-HER2 and antiangiogenic therapies. Finally, it entertains the hopes and challenges posed by this novel therapeutic approach. RECENT FINDINGS HER2 signaling upregulates angiogenesis at different levels and by different mechanisms. A large number of clinical trials were conducted in attempt to exploit the potential benefit of the combination. Results of early phase trials were promising. However, in the late phase clinical trials, the AVEREL trial did not demonstrate a consistent benefit for bevacizumab in the HER2 positive breast cancer patient population. The BETH trial is ongoing and recruiting patients. Safety issues regarding cardiovascular toxicity of the combination have been already raised. Negative experience of dual EGFR and VEGF targeting in colon cancer cannot be overlooked. SUMMARY Angiogenesis and HER2 signaling are closely related at the molecular level. Appraisal of efficacy of antiangiogenic therapies requires revisit of the current literature as well as following the results of ongoing trials.
Collapse
|
30
|
Prats AC, Van den Berghe L, Rayssac A, Ainaoui N, Morfoisse F, Pujol F, Legonidec S, Bikfalvi A, Prats H, Pyronnet S, Garmy-Susini B. CXCL4L1-fibstatin cooperation inhibits tumor angiogenesis, lymphangiogenesis and metastasis. Microvasc Res 2013; 89:25-33. [PMID: 23747987 DOI: 10.1016/j.mvr.2013.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/28/2013] [Accepted: 05/20/2013] [Indexed: 01/12/2023]
Abstract
Anti-angiogenic and anti-lymphangiogenic drugs slow tumor progression and dissemination. However, an important difficulty is that a tumor reacts and compensates to obtain the blood supply needed for tumor growth and lymphatic vessels to escape to distant loci. Therefore, there is a growing consensus on the requirement of multiple anti-(lymph)angiogenic molecules to stop cell invasion efficiently. Here we studied the cooperation between endogenous anti-angiogenic molecules, endostatin and fibstatin, and a chemokine, the Platelet Factor-4 variant 1, CXCL4L1. Anti-angiogenic factors were co-expressed by IRES-based bicistronic vectors and their cooperation was analyzed either by local delivery following transduction of pancreatic adenocarcinoma cells with lentivectors, or by distant delivery resulting from intramuscular administration in vivo of adeno-associated virus derived vectors followed by tumor subcutaneous injection. In this study, fibstatin and CXCL4L1 cooperate to inhibit endothelial cell proliferation, migration and tubulogenesis in vitro. No synergistic effect was found for fibstatin-endostatin combination. Importantly, we demonstrated for the first time that fibstatin and CXCL4L1 not only inhibit in vivo angiogenesis, but also lymphangiogenesis and tumor spread to the lymph nodes, whereas no beneficial effect was found on tumor growth inhibition using molecule combinations compared to molecules alone. These data reveal the synergy of CXCL4L1 and fibstatin in inhibition of tumor angiogenesis, lymphangiogenesis and metastasis and highlight the potential of IRES-based vectors to develop anti-metastasis combined gene therapies.
Collapse
Affiliation(s)
- A C Prats
- Université de Toulouse, UPS, TRADGENE, EA4554, F-31432 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Foy KC, Wygle RM, Miller MJ, Overholser JP, Bekaii-Saab T, Kaumaya PTP. Peptide vaccines and peptidomimetics of EGFR (HER-1) ligand binding domain inhibit cancer cell growth in vitro and in vivo. THE JOURNAL OF IMMUNOLOGY 2013; 191:217-27. [PMID: 23698748 DOI: 10.4049/jimmunol.1300231] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Epidermal growth factor receptor (EGFR) is a validated target for several cancers including lung, colorectal, and certain subtypes of breast cancer. Cetuximab targets ligand binding of EGFR, but major problems like high cost, short t1/2, toxicity, and emergence of resistance are associated with the drug. Immunization with EGFR B cell epitopes will train the immune system to produce specific Abs that can kill cancer cells. Also, therapy with stable, less-expensive, and nontoxic EGFR peptide mimics will block EGFR signaling and inhibit cancer growth. We designed three peptides based on the contact sites between EGF and EGFR. The B cell epitopes were synthesized alone and also linked with the measles virus T cell epitope to produce a chimeric peptide vaccine. The peptide vaccines were immunogenic in both mice and rabbits and Abs raised against the vaccine specifically bound EGFR-expressing cells and recombinant human EGFR protein. The peptide mimics and the anti-peptide Abs were able to inhibit EGFR signaling pathways. Immunization with the peptide vaccine or treatment with the B cell epitopes significantly reduced tumor growth in both transplantable breast and lung cancer models. Immunohistochemical analysis also showed significant reductions in microvascular density and actively dividing cells in the tumor sections after treatment in the FVB/n breast cancer model. The 418-435 B cell epitope was the best candidate both as a vaccine or peptide mimic because it caused significant inhibition in the two mouse models. Our results show that this novel EGFR B cell epitope has great potential to be used as a vaccine or treatment option for EGFR-expressing cancers.
Collapse
Affiliation(s)
- Kevin Chu Foy
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
32
|
Haen SP, Rammensee HG. The repertoire of human tumor-associated epitopes--identification and selection of antigens and their application in clinical trials. Curr Opin Immunol 2013; 25:277-83. [PMID: 23619309 DOI: 10.1016/j.coi.2013.03.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/14/2013] [Accepted: 03/21/2013] [Indexed: 11/26/2022]
Abstract
In cancer patients, active immunotherapy has gained significant importance in recent years by implementation of novel substances into standard clinical care. These new drugs represent strategies which either use defined cancer associated antigens as vaccines or induce tumor-directed immune responses through generation of a general inflammatory state which has extensive autoinflammatory side effects by induction of autoreactive immune cells. Hence, the definition of suitable target antigens for immunotherapy remains a major challenge. These antigens should ideally be specific markers for individual tumors or should be at least structures overexpressed on the tumor as compared to normal cells. Recent approaches have defined algorithms and refined analytical methods for antigen identification and immunological validation that have already been evaluated in clinical studies. This article summarizes recent developments in tissue analysis on genome, transcriptome and HLA-ligandome levels and of antigen application in recent clinical vaccination trials.
Collapse
Affiliation(s)
- Sebastian P Haen
- Medizinische Universitätsklinik Tübingen, Abteilung II für Onkologie, Hämatologie, Immunologie, Rheumatologie und Pulmologie, Otfried Müller Str. 10, D-72076 Tübingen, Germany
| | | |
Collapse
|
33
|
Kaumaya PTP, Foy KC. Peptide vaccines and targeting HER and VEGF proteins may offer a potentially new paradigm in cancer immunotherapy. Future Oncol 2012; 8:961-87. [PMID: 22894670 DOI: 10.2217/fon.12.95] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ErbB family (HER-1, HER-2, HER-3 and HER-4) of receptor tyrosine kinases has been the focus of cancer immunotherapeutic strategies while antiangiogenic therapies have focused on VEGF and its receptors VEGFR-1 and VEGFR-2. Agents targeting receptor tyrosine kinases in oncology include therapeutic antibodies to receptor tyrosine kinase ligands or the receptors themselves, and small-molecule inhibitors. Many of the US FDA-approved therapies targeting HER-2 and VEGF exhibit unacceptable toxicities, and show problems of efficacy, development of resistance and unacceptable safety profiles that continue to hamper their clinical progress. The combination of different peptide vaccines and peptidomimetics targeting specific molecular pathways that are dysregulated in tumors may potentiate anticancer immune responses, bypass immune tolerance and circumvent resistance mechanisms. The focus of this review is to discuss efforts in our laboratory spanning two decades of rationally developing peptide vaccines and therapeutics for breast cancer. This review highlights the prospective benefit of a new, untapped category of therapies biologically targeted to EGF receptor (HER-1), HER-2 and VEGF with potential peptide 'blockbusters' that could lay the foundation of a new paradigm in cancer immunotherapy by creating clinical breakthroughs for safe and efficacious cancer cures.
Collapse
Affiliation(s)
- Pravin T P Kaumaya
- Departments of Obstetrics & Gynecology, OSU Wexner Medical Center, James Cancer Hospital & Solove Research Institute & the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | | |
Collapse
|
34
|
Krupinski J, Abudawood M, Matou-Nasri S, Al-Baradie R, Petcu EB, Justicia C, Planas A, Liu D, Rovira N, Grau-Slevin M, Secades J, Slevin M. Citicoline induces angiogenesis improving survival of vascular/human brain microvessel endothelial cells through pathways involving ERK1/2 and insulin receptor substrate-1. Vasc Cell 2012; 4:20. [PMID: 23227823 PMCID: PMC3554547 DOI: 10.1186/2045-824x-4-20] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023] Open
Abstract
Background Citicoline is one of the neuroprotective agents that have been used as a therapy in stroke patients. There is limited published data describing the mechanisms through which it acts. Methods We used in vitro angiogenesis assays: migration, proliferation, differentiation into tube-like structures in Matrigel™ and spheroid development assays in human brain microvessel endothelial cells (hCMEC/D3). Western blotting was performed on protein extraction from hCMEC/D3 stimulated with citicoline. An analysis of citicoline signalling pathways was previously studied using a Kinexus phospho-protein screening array. A staurosporin/calcium ionophore-induced apoptosis assay was performed by seeding hCMEC/D3 on to glass coverslips in serum poor medium. In a pilot in vivo study, transient MCAO in rats was carried out with and without citicoline treatment (1000 mg/Kg) applied at the time of occlusion and subsequently every 3 days until euthanasia (21 days). Vascularity of the stroke-affected regions was examined by immunohistochemistry. Results Citicoline presented no mitogenic and chemotactic effects on hCMEC/D3; however, it significantly increased wound recovery, the formation of tube-like structures in Matrigel™ and enhanced spheroid development and sprouting. Citicoline induced the expression of phospho-extracellular-signal regulated kinase (ERK)-1/2. Kinexus assays showed an over-expression of insulin receptor substrate-1 (IRS-1). Knock-down of IRS-1 with targeted siRNA in our hCMEC/D3 inhibited the pro-angiogenic effects of citicoline. The percentage of surviving cells was higher in the presence of citicoline. Citicoline treatment significantly increased the numbers of new, active CD105-positive microvessels following MCAO. Conclusions The findings demonstrate both a pro-angiogenic and protective effect of citicoline on hCMEC/D3 in vitro and following middle cerebral artery occlusion (MCAO) in vivo.
Collapse
Affiliation(s)
- Jerzy Krupinski
- Cerebrovascular Diseases, Department of Neurology, Hospital Universitari Mútua Terrassa, Terrassa, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Koskimaki JE, Lee E, Chen W, Rivera CG, Rosca EV, Pandey NB, Popel AS. Synergy between a collagen IV mimetic peptide and a somatotropin-domain derived peptide as angiogenesis and lymphangiogenesis inhibitors. Angiogenesis 2012; 16:159-70. [PMID: 23053781 DOI: 10.1007/s10456-012-9308-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 09/15/2012] [Indexed: 01/13/2023]
Abstract
Angiogenesis is central to many physiological and pathological processes. Here we show two potent bioinformatically-identified peptides, one derived from collagen IV and translationally optimized, and one from a somatotropin domain-containing protein, synergize in angiogenesis and lymphangiogenesis assays including cell adhesion, migration and in vivo Matrigel plugs. Peptide-peptide combination therapies have recently been applied to diseases such as human immunodeficiency virus (HIV), but remain uncommon thus far in cancer, age-related macular degeneration and other angiogenesis-dependent diseases. Previous work from our group has shown that the collagen IV-derived peptide primarily binds β1 integrins, while the receptor for the somatotropin-derived peptide remains unknown. We investigate these peptides' mechanisms of action and find both peptides affect the vascular endothelial growth factor (VEGF) pathway as well as focal adhesion kinase (FAK) by changes in phosphorylation level and total protein content. Blocking of FAK both through binding of β1 integrins and through inhibition of VEGFR2 accounts for the synergy we observe. Since resistance through activation of multiple signaling pathways is a central problem of anti-angiogenic therapies in diseases such as cancer, we suggest that peptide combinations such as these are an approach that should be considered as a means to sustain anti-angiogenic and anti-lymphangiogenic therapy and improve efficacy of treatment.
Collapse
Affiliation(s)
- Jacob E Koskimaki
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Mizuguchi T, Ohara N, Iida M, Ninomiya R, Wada S, Kiso Y, Saito K, Akaji K. Evaluation of dimerization-inhibitory activities of cyclic peptides containing a β-hairpin loop sequence of the EGF receptor. Bioorg Med Chem 2012; 20:5730-7. [PMID: 22959765 DOI: 10.1016/j.bmc.2012.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 10/28/2022]
Abstract
Structure-activity relationships of cyclic peptides mimicking the β-hairpin structure of the 'dimerization arm' at residues 242-259 of the EGF receptor are examined. Cyclic peptides containing the arm head of the β-hairpin loop showed inhibitory activity toward the EGF receptor's dimerization. Cyclic peptides containing a Retro-Inverso sequence of the dimerization arm showed clear inhibitory effects on the dimerization in vitro and efficiently suppressed the proliferation of A431 cells, which abundantly express the EGF receptor on their surface. The effects at a specific hydrophobic site of the loop structure were expected to enhance the interactions with the receptor.
Collapse
Affiliation(s)
- Takaaki Mizuguchi
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, Kyoto Pharmaceutical University, Kyoto 607-8412, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Nahta R. Pharmacological strategies to overcome HER2 cross-talk and Trastuzumab resistance. Curr Med Chem 2012; 19:1065-75. [PMID: 22229414 DOI: 10.2174/092986712799320691] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/28/2011] [Accepted: 12/29/2011] [Indexed: 01/23/2023]
Abstract
Approximately 20-30% of breast cancers show increased expression of the HER2 receptor tyrosine kinase. Trastuzumab (Herceptin) is a clinically approved anti-HER2 monoclonal antibody. Many patients with HER2-overexpressing metastatic breast cancer respond to trastuzumab; however, a subset display primary drug resistance. In addition, many patients who initially respond to trastuzumab ultimately develop disease progression. Multiple molecular mechanisms contributing to trastuzumab resistance have been proposed in the literature. These mechanisms include cross-signaling from related HER/erbB receptors and compensatory signaling from receptors outside of the HER/erbB family, including receptors for insulin-like growth factor-I, vascular endothelial growth factor, and transforming growth factor beta. The major downstream signaling pathway activated by HER2 cross-talk is PI3K/mTOR, and a potential integrator of receptor cross-talk is Src-focal adhesion kinase (FAK) signaling. PI3K, Src, and FAK have independently been implicated in trastuzumab resistance. In this review, we will discuss pharmacological inhibition of HER2 cross-talk as a strategy to treat trastuzumab-refractory HER2-overexpresssing breast cancer.
Collapse
Affiliation(s)
- R Nahta
- Departments of Pharmacology, Emory University School of Medicine, USA.
| |
Collapse
|
38
|
Rau KM, Huang CC, Chiu TJ, Chen YY, Lu CC, Liu CT, Pei SN, Wei YC. Neovascularization evaluated by CD105 correlates well with prognostic factors in breast cancers. Exp Ther Med 2012; 4:231-236. [PMID: 23139713 DOI: 10.3892/etm.2012.594] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 05/21/2012] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis is critical for the growth, invasion and metastasis of cancers. Extensive neovascularization and tumor thrombus also correlate with a poor prognosis in breast cancer (BC). Although anti-angiogenic agents have been the therapies of choice for BC, in particular for triple-negative BCs, predictive markers for anti-angiogenic agents are lacking. Microvascular density (MVD) is commonly used to assess the neovascularization in tumors. Compared with pan-endothelial markers such as CD31, CD34 and von Willebrand factor (vWF), CD105 has a higher specificity for MVD in tumor tissues. In this study, we aimed to determine the prognostic value of CD105 in BCs. Paraffin-embedded tissue blocks from 201 BC patients were formed into tissue microarrays. Evaluation of MVD revealed that a median of 11 microvessels determined by CD105 staining correlated significantly with the pathological characteristics of BCs and also with the survival of patients. The expression of CD105 correlated inversely with hormone receptor (HR) expression but positively with Her-2 expression. Univariate analysis indicated that CD105 is a superior predictor of disease-free survival (DFS) in stage I and II diseases; multivariate analysis indicated that only hormone receptors (HRs) are suitable for predicting overall survival (OS) in stage III disease. These findings reveal for the first time that MVD measured by CD105 staining correlates positively with Her-2 expression but negatively with HR expression. The significance of MVD on OS is more apparent in early stage BCs. CD105 has the potential to be used as a predictive marker for anti-angiogenic agents; the targeting of CD105 may also be a potential anticancer strategy.
Collapse
Affiliation(s)
- Kun-Ming Rau
- Department of Internal Medicine, Division of Hematology-Oncology, and ; Chang Gung University, College of Medicine, Tao-Yuan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Human papillomavirus-16 E5 protein: oncogenic role and therapeutic value. Cell Oncol (Dordr) 2012; 35:67-76. [DOI: 10.1007/s13402-011-0069-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2011] [Indexed: 10/14/2022] Open
|
40
|
Kaumaya PT. Could precision-engineered peptide epitopes/vaccines be the key to a cancer cure? Future Oncol 2011; 7:807-10. [PMID: 21732751 DOI: 10.2217/fon.11.60] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
41
|
Gompels LL, Malik NM, Madden L, Jin P, Feldmann M, Shepard HM, Paleolog EM. Human epidermal growth factor receptor bispecific ligand trap RB200: abrogation of collagen-induced arthritis in combination with tumour necrosis factor blockade. Arthritis Res Ther 2011; 13:R161. [PMID: 21982514 PMCID: PMC3308094 DOI: 10.1186/ar3480] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 07/29/2011] [Accepted: 10/07/2011] [Indexed: 11/22/2022] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic disease associated with inflammation and destruction of bone and cartilage. Although inhibition of TNFα is widely used to treat RA, a significant number of patients do not respond to TNFα blockade, and therefore there is a compelling need to continue to identify alternative therapeutic strategies for treating chronic inflammatory diseases such as RA. The anti-epidermal growth factor (anti-EGF) receptor antibody trastuzumab has revolutionised the treatment of patients with EGF receptor-positive breast cancer. Expression of EGF ligands and receptors (known as HER) has also been documented in RA. The highly unique compound RB200 is a bispecific ligand trap that is composed of full-length extracellular domains of HER1 and HER3 EGF receptors. Because of its pan-HER specificity, RB200 inhibits responses mediated by HER1, HER2 and HER3 in vitro and in vivo. The objective of this study was to assess the effect of RB200 combined with TNF blockade in a murine collagen-induced arthritis (CIA) model of RA. Methods Arthritic mice were treated with RB200 alone or in combination with the TNF receptor fusion protein etanercept. We performed immunohistochemistry to assess CD31 and in vivo fluorescent imaging using anti-E-selectin antibody labelled with fluorescent dye to elucidate the effect of RB200 on the vasculature in CIA. Results RB200 significantly abrogated CIA by reducing paw swelling and clinical scores. Importantly, low-dose RB200 combined with a suboptimal dose of etanercept led to complete abrogation of arthritis. Moreover, the combination of RB200 with etanercept abrogated the intensity of the E-selectin-targeted signal to the level seen in control animals not immunised to CIA. Conclusions The human pan-EGF receptor bispecific ligand trap RB200, when combined with low-dose etanercept, abrogates CIA, suggesting that inhibition of events downstream of EGF receptor activation, in combination with TNFα inhibitors, may hold promise as a future therapy for patients with RA.
Collapse
Affiliation(s)
- Luke L Gompels
- Faculty of Medicine, Kennedy Institute of Rheumatology, 65 Aspenlea Road, London, W6 8LH, Imperial College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen Y, Luo W, Song H, Yin B, Tang J, Chen Y, Ng MH, Yeo AET, Zhang J, Xia N. Mimotope ELISA for detection of broad spectrum antibody against avian H5N1 influenza virus. PLoS One 2011; 6:e24144. [PMID: 21912666 PMCID: PMC3166295 DOI: 10.1371/journal.pone.0024144] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 08/01/2011] [Indexed: 11/18/2022] Open
Abstract
Background We have raised a panel of broad spectrum neutralizing monoclonal antibodies against the highly pathogenic H5N1 avian influenza virus, which neutralize the infectivity of, and afford protection against infection by, most of the major genetic groups of the virus evolved since 1997. Peptide mimics reactive with one of these broad spectrum H5N1 neutralizing antibodies, 8H5, were identified from random phage display libraries. Method The amino acid residues of the most reactive 12mer peptide, p125 (DTPLTTAALRLV), were randomly substituted to improve its mimicry of the natural 8H5 epitope. Result 133 reactive peptides with unique amino acid sequences were identified from 5 sub-libraries of p125. Four residues (2,4,5.9) of the parental peptide were preserved among all the derived peptides and probably essential for 8H5 binding. These are interspersed among four other residues (1,3,8,10), which exhibit restricted substitution and probably could contribute to binding, and another four (6,7,11,12) which could be randomly substituted and probably are not essential for binding. One peptide, V-1b, derived by substituting 5 of the latter residues is the most reactive and has a binding constant of 3.16×10−9 M, which is 38 fold higher than the affinity of the parental p125. Immunoassay produced with this peptide is specifically reactive with 8H5 but not also the other related broad spectrum H5N1 avian influenza virus neutralizing antibodies. Serum samples from 29 chickens infected with H5N1 avian influenza virus gave a positive result by this assay and those from 12 uninfected animals gave a negative test result. Conclusion The immunoassay produced with the 12 mer peptide,V1-b, is specific for the natural 8H5 epitope and can be used for detection of antibody against the broad spectrum neutralization site of H5N1 avian influenza virus.
Collapse
Affiliation(s)
- Yingwei Chen
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wenxin Luo
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
- * E-mail: (NX); (WL)
| | - Huijuan Song
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Boyuan Yin
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jixian Tang
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yixin Chen
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mun Hon Ng
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Anthony E. T. Yeo
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jun Zhang
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
| | - Ningshao Xia
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, The Key Laboratory of Education Minister for Cell Biology and Tumor Cell Engineering of Xiamen University, School of Life Sciences, Xiamen University, Xiamen, China
- * E-mail: (NX); (WL)
| |
Collapse
|