1
|
Ge M, Zhu Y, Wei M, Piao H, He M. Improving the efficacy of anti-EGFR drugs in GBM: Where we are going? Biochim Biophys Acta Rev Cancer 2023; 1878:188996. [PMID: 37805108 DOI: 10.1016/j.bbcan.2023.188996] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/11/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
The therapies targeting mutations of driver genes in cancer have advanced into clinical trials for a variety of tumors. In glioblastoma (GBM), epidermal growth factor receptor (EGFR) is the most commonly mutated oncogene, and targeting EGFR has been widely investigated as a promising direction. However, the results of EGFR pathway inhibitors have not been satisfactory. Limited blood-brain barrier (BBB) permeability, drug resistance, and pathway compensation mechanisms contribute to the failure of anti-EGFR therapies. This review summarizes recent research advances in EGFR-targeted therapy for GBM and provides insight into the reasons for the unsatisfactory results of EGFR-targeted therapy. By combining the results of preclinical studies with those of clinical trials, we discuss that improved drug penetration across the BBB, the use of multi-target combinations, and the development of peptidomimetic drugs under the premise of precision medicine may be promising strategies to overcome drug resistance in GBM.
Collapse
Affiliation(s)
- Manxi Ge
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Yan Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, China.
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China.
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China; Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.
| |
Collapse
|
2
|
Abstract
Metastatic breast cancer (BC) is an aggressive form of cancer and is an absolute challenge to treat. This review discusses the standard treatments available for metastatic BC. It further highlights the rationale for targeting oncodrivers, tumor-associated antigens, and neoantigens in BC. Explaining the significance of immune response in successful immunotherapeutic studies, it draws attention towards how adoptive cell therapy can be a useful immunotherapeutic tool. We focus on adoptive cell therapy in BC covering tumor-infiltrating lymphocyte therapy, engineered T cell receptor therapy, chimeric antigen receptor therapy, dendritic cell therapy and natural killer cell therapy. In this work, we aim to provide an overview of clinical data regarding the use of cellular immunotherapies in BC. Eventually, we conclude by proposing future adoptive cell therapy approaches, which can be used to cure BC.
Collapse
|
3
|
Geskovski N, Matevska-Geshkovska N, Dimchevska Sazdovska S, Glavas Dodov M, Mladenovska K, Goracinova K. The impact of molecular tumor profiling on the design strategies for targeting myeloid leukemia and EGFR/CD44-positive solid tumors. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:375-401. [PMID: 33981532 PMCID: PMC8093552 DOI: 10.3762/bjnano.12.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/12/2021] [Indexed: 05/21/2023]
Abstract
Nanomedicine has emerged as a novel cancer treatment and diagnostic modality, whose design constantly evolves towards increasing the safety and efficacy of the chemotherapeutic and diagnostic protocols. Molecular diagnostics, which create a great amount of data related to the unique molecular signatures of each tumor subtype, have emerged as an important tool for detailed profiling of tumors. They provide an opportunity to develop targeting agents for early detection and diagnosis, and to select the most effective combinatorial treatment options. Alongside, the design of the nanoscale carriers needs to cope with novel trends of molecular screening. Also, multiple targeting ligands needed for robust and specific interactions with the targeted cell populations have to be introduced, which should result in substantial improvements in safety and efficacy of the cancer treatment. This article will focus on novel design strategies for nanoscale drug delivery systems, based on the unique molecular signatures of myeloid leukemia and EGFR/CD44-positive solid tumors, and the impact of novel discoveries in molecular tumor profiles on future chemotherapeutic protocols.
Collapse
Affiliation(s)
- Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Nadica Matevska-Geshkovska
- Center for Pharmaceutical Biomolecular Analyses, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Simona Dimchevska Sazdovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- Department of Nanobiotechnology, Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Kristina Mladenovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- College of Pharmacy, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
4
|
Guryanova SV, Khaitov RM. Strategies for Using Muramyl Peptides - Modulators of Innate Immunity of Bacterial Origin - in Medicine. Front Immunol 2021; 12:607178. [PMID: 33959120 PMCID: PMC8093441 DOI: 10.3389/fimmu.2021.607178] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/06/2021] [Indexed: 12/22/2022] Open
Abstract
The spread of infectious diseases is rampant. The emergence of new infections, the irrational use of antibiotics in medicine and their widespread use in agriculture contribute to the emergence of microorganisms that are resistant to antimicrobial drugs. By 2050, mortality from antibiotic-resistant strains of bacteria is projected to increase up to 10 million people per year, which will exceed mortality from cancer. Mutations in bacteria and viruses are occurring faster than new drugs and vaccines are being introduced to the market. In search of effective protection against infections, new strategies and approaches are being developed, one of which is the use of innate immunity activators in combination with etiotropic chemotherapy drugs. Muramyl peptides, which are part of peptidoglycan of cell walls of all known bacteria, regularly formed in the body during the breakdown of microflora and considered to be natural regulators of immunity. Their interaction with intracellular receptors launches a sequence of processes that ultimately leads to the increased expression of genes of MHC molecules, pro-inflammatory mediators, cytokines and their soluble and membrane-associated receptors. As a result, all subpopulations of immunocompetent cells are activated: macrophages and dendritic cells, neutrophils, T-, B- lymphocytes and natural killer cells for an adequate response to foreign or transformed antigens, manifested both in the regulation of the inflammatory response and in providing immunological tolerance. Muramyl peptides take part in the process of hematopoiesis, stimulating production of colony-stimulating factors, which is the basis for their use in the treatment of oncological diseases. In this review we highlight clinical trials of drugs based on muramyl peptides, as well as clinical efficacy of drugs mifamurtide, lycopid, liasten and polimuramil. Such a multifactorial effect of muramyl peptides and a well-known mechanism of activity make them promising drugs in the treatment and preventing of infectious, allergic and oncological diseases, and in the composition of vaccines.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (RAS), Moscow, Russia
- Department of Biology and General Genetics, Medical Institute, RUDN University, Moscow, Russia
| | - Rahim M. Khaitov
- National Research Center – Institute of Immunology of Federal Medico-Biological Agency, Moscow, Russia
- Department of Immunology, Moscow State University of Medicine and Dentistry, Moscow, Russia
| |
Collapse
|
5
|
Molecular Targeting of Epidermal Growth Factor Receptor (EGFR) and Vascular Endothelial Growth Factor Receptor (VEGFR). Molecules 2021; 26:molecules26041076. [PMID: 33670650 PMCID: PMC7922143 DOI: 10.3390/molecules26041076] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) are two extensively studied membrane-bound receptor tyrosine kinase proteins that are frequently overexpressed in many cancers. As a result, these receptor families constitute attractive targets for imaging and therapeutic applications in the detection and treatment of cancer. This review explores the dynamic structure and structure-function relationships of these two growth factor receptors and their significance as it relates to theranostics of cancer, followed by some of the common inhibition modalities frequently employed to target EGFR and VEGFR, such as tyrosine kinase inhibitors (TKIs), antibodies, nanobodies, and peptides. A summary of the recent advances in molecular imaging techniques, including positron emission tomography (PET), single-photon emission computerized tomography (SPECT), computed tomography (CT), magnetic resonance imaging (MRI), and optical imaging (OI), and in particular, near-IR fluorescence imaging using tetrapyrrolic-based fluorophores, concludes this review.
Collapse
|
6
|
Fidelle M, Yonekura S, Picard M, Cogdill A, Hollebecque A, Roberti MP, Zitvogel L. Resolving the Paradox of Colon Cancer Through the Integration of Genetics, Immunology, and the Microbiota. Front Immunol 2020; 11:600886. [PMID: 33381121 PMCID: PMC7768083 DOI: 10.3389/fimmu.2020.600886] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
While colorectal cancers (CRC) are paradigmatic tumors invaded by effector memory lymphocytes, the mechanisms accounting for the relative resistance of MSI negative CRC to immunogenic cell death mediated by oxaliplatin and immune checkpoint inhibitors has remained an open conundrum. Here, we propose the viewpoint where its microenvironmental contexture could be explained -at least in part- by macroenvironmental cues constituted by the complex interplay between the epithelial barrier, its microbial ecosystem, and the local immune system. Taken together this dynamic ménage-à-trois offers novel coordinated actors of the humoral and cellular immune responses actionable to restore sensitivity to immune checkpoint inhibition. Solving this paradox involves breaking tolerance to crypt stem cells by inducing the immunogenic apoptosis of ileal cells in the context of an ileal microbiome shifted towards immunogenic bacteria using cytotoxicants. This manoeuver results in the elicitation of a productive Tfh and B cell dialogue in mesenteric lymph nodes culminating in tumor-specific memory CD8+ T cell responses sparing the normal epithelium.
Collapse
Affiliation(s)
- Marine Fidelle
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Satoru Yonekura
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Marion Picard
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Unit Biology and Genetics of the Bacterial Cell Wall, Institut Pasteur, Paris, France
| | - Alexandria Cogdill
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Antoine Hollebecque
- Gustave Roussy, Villejuif, France
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| |
Collapse
|
7
|
Kaumaya PTP, Guo L, Overholser J, Penichet ML, Bekaii-Saab T. Immunogenicity and antitumor efficacy of a novel human PD-1 B-cell vaccine (PD1-Vaxx) and combination immunotherapy with dual trastuzumab/pertuzumab-like HER-2 B-cell epitope vaccines (B-Vaxx) in a syngeneic mouse model. Oncoimmunology 2020; 9:1818437. [PMID: 33117602 PMCID: PMC7553530 DOI: 10.1080/2162402x.2020.1818437] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Therapeutic blockade of PD-1/PD-L1 signaling with monoclonal antibodies (mAbs) has shown clinical success and activity across a broad set of cancer subtypes. However, monotherapy with PD-1/PD-L1 inhibitors are only effective in a subset of patients and ongoing studies show efficacy of treatment depends on a combinatorial approach. Contrary to mAbs chimeric B-cell cancer vaccines incorporating a “promiscuous” T-cell epitope have the advantage of producing a polyclonal B-cell antibody that can potentially induce memory B- and T-cell responses, while reducing immune evasion and suppression. Here, we describe a novel PD-1 B-cell peptide epitope vaccine (amino acid 92–110; PD1-Vaxx) linked to a measles virus fusion peptide (MVF) amino acid 288–302 via a four amino acid residue (GPSL) emulsified in Montanide ISA 720VG that aims to induce the production of polyclonal antibodies that block PD-1 signaling and thus trigger anticancer effects similar to nivolumab. In preclinical studies, the PD1-Vaxx outperformed the standard anti-mouse PD-1 antibody (mAb 29F.1A12) in a mouse model of human HER-2 expressing colon carcinoma. Furthermore, the combination of PD1-Vaxx with combo HER-2 peptide vaccine (B-Vaxx) showed enhanced inhibition of tumor growth in colon carcinoma BALB/c model challenged with CT26/HER-2 cells. The PD-1 or combined vaccines were safe with no evidence of toxicity or autoimmunity.
Collapse
Affiliation(s)
- Pravin T P Kaumaya
- Department of Obstetrics & Gynecology.,The Wexner Medical Center and the Arthur G. James Cancer Hospital, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | | | - Manuel L Penichet
- Division of Surgical Oncology Department of Surgery, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
8
|
Establishment and validation of an immune-associated signature in lung adenocarcinoma. Int Immunopharmacol 2020; 88:106867. [PMID: 32799112 DOI: 10.1016/j.intimp.2020.106867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/18/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
Recent studies demonstrated that immune associated genes (IAGs) played an important role in the treatment of lung adenocarcinoma (LUAD). In the research, we established an IAGs signature and validated its prognostic value in LUAD by using bioinformatic methods and public databases. Based on the RNA-Seq samples from The Cancer Genome Atlas (TCGA), 576 differentially expressed IAGs were firstly identified. The R package coxph was used to select significant prognostic IAGs using both univariate and multivariate analyses. As a result, four IAGs (SCG2, CCL20, CAT, S100P) were finally screened in an IAGs signature. Based on these four IAGs, LASSO (least absolute shrinkage and selection operator) Cox regression analysis was used to construct a Risk score prognostic model and survival analysis revealed that high risk score was significantly associated with poor survival outcomes, which was validated in the external datasets GSE68465 and GSE31210. In addition, Risk score was found to be significantly associated with stage, lymphatic involvement, tumor metastasis and immune cells (B cells and dendritic cells) infiltration. Moreover, it was found that TP53 and EGFR had a higher mutation frequency in high risk group. Then a nomogram with clinical characteristics was established to superiorly predict prognosis of LUAD patients, and calibration plots and ROC analysis proved its accuracy. We believe that our findings can be conveniently used for individualized prediction of the clinical prognosis for LUAD patients, but further clinical trials and experimental exploration are needed to validate our observations.
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: 8] [Impact Index Per Article: 2.0] [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
|
Yang CY, Liao WY, Ho CC, Chen KY, Tsai TH, Hsu CL, Su KY, Chang YL, Wu CT, Hsu CC, Liao BC, Hsu WH, Lee JH, Lin CC, Shih JY, Yang JCH, Yu CJ. Association between programmed death-ligand 1 expression, immune microenvironments, and clinical outcomes in epidermal growth factor receptor mutant lung adenocarcinoma patients treated with tyrosine kinase inhibitors. Eur J Cancer 2020; 124:110-122. [DOI: 10.1016/j.ejca.2019.10.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/06/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022]
|
11
|
Sachdeva S, Joo H, Tsai J, Jasti B, Li X. A Rational Approach for Creating Peptides Mimicking Antibody Binding. Sci Rep 2019; 9:997. [PMID: 30700733 PMCID: PMC6353898 DOI: 10.1038/s41598-018-37201-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
This study reports a novel method to design peptides that mimic antibody binding. Using the Knob-Socket model for protein-protein interaction, the interaction surface between Cetuximab and EGFR was mapped. EGFR binding peptides were designed based on geometry and the probability of the mapped knob-sockets pairs. Designed peptides were synthesized and then characterized for binding specificity, affinity, cytotoxicity of drug-peptide conjugate and inhibition of phosphorylation. In cell culture studies, designed peptides specifically bind and internalize to EGFR overexpressing cells with three to four-fold higher uptake compared to control cells that do not overexpress EGFR. The designed peptide, Pep11, bound to EGFR with KD of 252 nM. Cytotoxicity of Monomethyl Auristatin E (MMAE)-EGFR-Pep11 peptide-drug conjugate was more than 2,000 fold higher against EGFR overexpressing cell lines A431, MDA MB 468 than control HEK 293 cells which lack EGFR overexpression. MMAE-EGFR-Pep11 conjugate also showed more than 90-fold lower cytotoxicity towards non-EGFR overexpressing HEK 293 cells when compared with cytotoxicity of MMAE itself. In conclusion, a method that can rationally design peptides using knob-socket model is presented. This method was successfully applied to create peptides based on the antigen-antibody interaction to mimic the specificity, affinity and functionality of antibody.
Collapse
Affiliation(s)
- Sameer Sachdeva
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA, 95211, USA.,Amneal Pharmaceuticals, Piscataway, NJ, 08854, USA
| | - Hyun Joo
- Department of Chemistry, University of the Pacific, Stockton, CA, 95211, USA
| | - Jerry Tsai
- Department of Chemistry, University of the Pacific, Stockton, CA, 95211, USA
| | - Bhaskara Jasti
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA, 95211, USA
| | - Xiaoling Li
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA, 95211, USA.
| |
Collapse
|
12
|
Shen M, Jiang YZ, Wei Y, Ell B, Sheng X, Esposito M, Kang J, Hang X, Zheng H, Rowicki M, Zhang L, Shih WJ, Celià-Terrassa T, Liu Y, Cristea II, Shao ZM, Kang Y. Tinagl1 Suppresses Triple-Negative Breast Cancer Progression and Metastasis by Simultaneously Inhibiting Integrin/FAK and EGFR Signaling. Cancer Cell 2019; 35:64-80.e7. [PMID: 30612941 DOI: 10.1016/j.ccell.2018.11.016] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/13/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancer (TNBC) patients have the worst prognosis and distant metastasis-free survival among all major subtypes of breast cancer. The poor clinical outlook is further exacerbated by a lack of effective targeted therapies for TNBC. Here we show that ectopic expression and therapeutic delivery of the secreted protein Tubulointerstitial nephritis antigen-like 1 (Tinagl1) suppresses TNBC progression and metastasis through direct binding to integrin α5β1, αvβ1, and epidermal growth factor receptor (EGFR), and subsequent simultaneous inhibition of focal adhesion kinase (FAK) and EGFR signaling pathways. Moreover, Tinagl1 protein level is associated with good prognosis and reversely correlates with FAK and EGFR activation status in TNBC. Our results suggest Tinagl1 as a candidate therapeutic agent for TNBC by dual inhibition of integrin/FAK and EGFR signaling pathways.
Collapse
Affiliation(s)
- Minhong Shen
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Brian Ell
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Xinlei Sheng
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Mark Esposito
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Jooeun Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Xiang Hang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Hanqiu Zheng
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Michelle Rowicki
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Weichung J Shih
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, 683 Hoes Lane West, Piscataway, NJ 08854, USA; Division of Biometrics, Rutgers Cancer Institute of New Jersey Rutgers, New Brunswick, NJ 08901, USA
| | - Toni Celià-Terrassa
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Yirong Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - IIeana Cristea
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA; Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
13
|
Wagner S, Mullins CS, Linnebacher M. Colorectal cancer vaccines: Tumor-associated antigens vs neoantigens. World J Gastroenterol 2018; 24:5418-5432. [PMID: 30622371 PMCID: PMC6319136 DOI: 10.3748/wjg.v24.i48.5418] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
Therapeutic options for the treatment of colorectal cancer (CRC) are diverse but still not always satisfying. Recent success of immune checkpoint inhibition treatment for the subgroup of CRC patients suffering from hyper-mutated tumors suggests a permanent role of immune therapy in the clinical management of CRC. Substantial improvement in treatment outcome could be achieved by development of efficient patient-individual CRC vaccination strategies. This mini-review summarizes the current knowledge on the two general classes of targets: tumor-associated antigens (TAAs) and tumor-specific antigens. TAAs like carcinoembryonic antigen and melanoma associated antigen are present in and shared by a subgroup of patients and a variety of clinical studies examined the efficacy of different TAA-derived peptide vaccines. Combinations of several TAAs as the next step and the development of personalized TAA-based peptide vaccines are discussed. Improvements of peptide-based vaccines achievable by adjuvants and immune-stimulatory chemotherapeutics are highlighted. Finally, we sum up clinical studies using tumor-specific antigens - in CRC almost exclusively neoantigens - which revealed promising results; particularly no severe adverse events were reported so far. Critical progress for clinical outcomes can be expected by individualizing neoantigen-based peptide vaccines and combining them with immune-stimulatory chemotherapeutics and immune checkpoint inhibitors. In light of these data and latest developments, truly personalized neoantigen-based peptide vaccines can be expected to fulfill modern precision medicine’s requirements and will manifest as treatment pillar for routine clinical management of CRC.
Collapse
Affiliation(s)
- Sandra Wagner
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| | - Christina S Mullins
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| | - Michael Linnebacher
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| |
Collapse
|
14
|
Wang SS, Liu W, Ly D, Xu H, Qu L, Zhang L. Tumor-infiltrating B cells: their role and application in anti-tumor immunity in lung cancer. Cell Mol Immunol 2018; 16:6-18. [PMID: 29628498 DOI: 10.1038/s41423-018-0027-x] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023] Open
Abstract
Evidence indicates that lung cancer development is a complex process that involves interactions between tumor cells, stromal fibroblasts, and immune cells. Tumor-infiltrating immune cells play a significant role in the promotion or inhibition of tumor growth. As an integral component of the tumor microenvironment, tumor-infiltrating B lymphocytes (TIBs) exist in all stages of cancer and play important roles in shaping tumor development. Here, we review recent clinical and preclinical studies that outline the role of TIBs in lung cancer development, assess their prognostic significance, and explore the potential benefit of B cell-based immunotherapy for lung cancer treatment.
Collapse
Affiliation(s)
- Si-Si Wang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130061, China
| | - Wei Liu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130061, China. .,Department of Thoracic surgery, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Dalam Ly
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 1L7, Canada.,Departments of Laboratory Medicine and Pathobiology, Immunology, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Hao Xu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130061, China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 1L7, Canada. .,Departments of Laboratory Medicine and Pathobiology, Immunology, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
15
|
Wang A, Cui M, Qu H, Di J, Wang Z, Xing J, Wu F, Wu W, Wang X, Shen L, Jiang B, Su X. Induction of anti-EGFR immune response with mimotopes identified from a phage display peptide library by panitumumab. Oncotarget 2018; 7:75293-75306. [PMID: 27659529 PMCID: PMC5342741 DOI: 10.18632/oncotarget.12167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in several epithelial tumors. Anti-EGFR humanized monoclonal antibodies, cetuximab and panitumumab, in combination with chemotherapy have improved the prognosis for patients with wild-type RAS tumors. To identify mimotopes of EGFR and develop mimotope-based EGFR vaccines, we screened a phage display peptide library with panitumumab. Two EGFR mimotopes P19 and P26, which could be recognized by panitumumab specifically, were isolated. To enhance the immune responses, we generated recombinant proteins of P19 or P26 fused to a heat-shock cognate protein 70 (Hsc70), and evaluated the efficacy of Hsc70-P19 and Hsc70-P26 as vaccines in vivo. Immunization with Hsc70-P19 or Hsc70-P26 fusion protein stimulated the immune system to produce specific antibodies against peptides as well as EGFR. Moreover, antibodies elicited against mimotopes could induce antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and inhibit the proliferation of EGFR-overexpressing A431 cells. Treatment with Hsc70-P19 and Hsc70-P26 significantly reduced tumor growth in BALB/c transplantable lung cancer models. Although there was no sequence homology between the phage-derived peptides and EGFR by alignments, both peptides mimic the conformational structure of EGFR binding to panitumumab. In conclusion, the mimotopes we identified from phage display peptide library could be promising candidate vaccines for active anti-EGFR immunotherapy against cancers.
Collapse
Affiliation(s)
- Aidong Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ming Cui
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Jiabo Di
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zaozao Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiadi Xing
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fan Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wei Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xicheng Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Beihai Jiang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiangqian Su
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
16
|
Hayes DA, Kunde DA, Taylor RL, Pyecroft SB, Sohal SS, Snow ET. ERBB3: A potential serum biomarker for early detection and therapeutic target for devil facial tumour 1 (DFT1). PLoS One 2017; 12:e0177919. [PMID: 28591206 PMCID: PMC5462353 DOI: 10.1371/journal.pone.0177919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Devil Facial Tumour 1 (DFT1) is one of two transmissible neoplasms of Tasmanian devils (Sarcophilus harrisii) predominantly affecting their facial regions. DFT1's cellular origin is that of Schwann cell lineage where lesions are evident macroscopically late in the disease. Conversely, the pre-clinical timeframe from cellular transmission to appearance of DFT1 remains uncertain demonstrating the importance of an effective pre-clinical biomarker. We show that ERBB3, a marker expressed normally by the developing neural crest and Schwann cells, is immunohistohemically expressed by DFT1, therefore the potential of ERBB3 as a biomarker was explored. Under the hypothesis that serum ERBB3 levels may increase as DFT1 invades local and distant tissues our pilot study determined serum ERBB3 levels in normal Tasmanian devils and Tasmanian devils with DFT1. Compared to the baseline serum ERBB3 levels in unaffected Tasmanian devils, Tasmanian devils with DFT1 showed significant elevation of serum ERBB3 levels. Interestingly Tasmanian devils with cutaneous lymphoma (CL) also showed elevation of serum ERBB3 levels when compared to the baseline serum levels of Tasmanian devils without DFT1. Thus, elevated serum ERBB3 levels in otherwise healthy looking devils could predict possible DFT1 or CL in captive or wild devil populations and would have implications on the management, welfare and survival of Tasmanian devils. ERBB3 is also a therapeutic target and therefore the potential exists to consider modes of administration that may eradicate DFT1 from the wild.
Collapse
Affiliation(s)
- Dane A. Hayes
- Department of Primary Industries, Parks Water and Environment, Animal Health Laboratory, Launceston, Tasmania, Australia
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Dale A. Kunde
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Robyn L. Taylor
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- Department of Primary Industries, Parks Water and Environment, Resource Management and Conservation, Hobart, Tasmania, Australia
| | - Stephen B. Pyecroft
- School of Animal & Veterinary Sciences, Faculty of Science, University of Adelaide, Roseworthy Campus, Roseworthy, South Australia
| | - Sukhwinder Singh Sohal
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Elizabeth T. Snow
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
17
|
Jin D, Yu X, Chen B, Li Z, Ding J, Zhao X, Qi G. Combined immunotherapy of breast cancer with EGF and VEGF vaccines from DNA shuffling in a mouse model. Immunotherapy 2017; 9:537-553. [PMID: 28509606 DOI: 10.2217/imt-2017-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Development of EGF and VEGF vaccines with high antigenicity for combined immunotherapy of EGF-EGFR signaling-dependent epithelial tumors such as breast cancer. METHOD EGF genes from mouse, human and chicken were randomly assembled to chimeric genes by DNA shuffling, then a chimeric EGF was selected out by PCR, SDS-PAGE and immunization for combined immunotherapy of breast cancer with a previously constructed chimeric VEGF vaccine from shuffling. RESULTS Combined vaccination with chimeric EGF and VEGF from shuffling could induce high titer of antibodies against EGF and VEGF to inhibit tumor growth and angiogenesis, and improve the survival rate of mice with breast cancer. CONCLUSION Combined vaccination with EGF and VEGF from shuffling showed better immunotherapy on EGF-EGFR signaling-dependent epithelial tumors such as breast cancer than the single-agent EGF vaccination.
Collapse
Affiliation(s)
- Dong Jin
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xin Yu
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Bing Chen
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhitao Li
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jia Ding
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuyun Zhao
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Gaofu Qi
- College of Life Science & Technology, Huazhong Agricultural University, Wuhan 430070, China.,Biomedical Center, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
18
|
Alanazi IO, Khan Z. Understanding EGFR Signaling in Breast Cancer and Breast Cancer Stem Cells: Overexpression and Therapeutic Implications. Asian Pac J Cancer Prev 2017; 17:445-53. [PMID: 26925626 DOI: 10.7314/apjcp.2016.17.2.445] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Epidermal growth factor receptors (EGFRs/HERs) and downstream signaling pathways have been implicated in the pathogenesis of several malignancies including breast cancer and its resistance to treatment with chemotherapeutic drugs. Consequently, several monoclonal antibodies as well as small molecule inhibitors targeting these pathways have emerged as therapeutic tools in the recent past. However, studies have shown that utilizing these molecules in combination with chemotherapy has yielded only limited success. This review describes the current understanding of EGFRs/HERs and associated signaling pathways in relation to development of breast cancer and responses to various cancer treatments in the hope of pointing to improved prevention, diagnosis and treatment. Also, we review the role of breast cancer stem cells (BCSCs) in disease and the potential to target these cells.
Collapse
Affiliation(s)
- Ibrahim O Alanazi
- King Abdulaziz City for Science and Technology, Genome Center, King Saud University, Riyadh, Kingdom of Saudi Arabia E-mail :
| | | |
Collapse
|
19
|
Barth RF, Wu G, Meisen WH, Nakkula RJ, Yang W, Huo T, Kellough DA, Kaumaya P, Turro C, Agius LM, Kaur B. Design, synthesis, and evaluation of cisplatin-containing EGFR targeting bioconjugates as potential therapeutic agents for brain tumors. Onco Targets Ther 2016; 9:2769-81. [PMID: 27274273 PMCID: PMC4869632 DOI: 10.2147/ott.s99242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to evaluate four different platinated bioconjugates containing a cisplatin (cis-diamminedichloroplatinum [cis-DDP]) fragment and epidermal growth factor receptor (EGFR)-targeting moieties as potential therapeutic agents for the treatment of brain tumors using a human EGFR-expressing transfectant of the F98 rat glioma (F98EGFR) to assess their efficacy. The first two bioconjugates employed the monoclonal antibody cetuximab (C225 or Erbitux®) as the targeting moiety, and the second two used genetically engineered EGF peptides. C225-G5-Pt was produced by reacting cis-DDP with a fifth-generation polyamidoamine dendrimer (G5) and then linking it to C225 by means of two heterobifunctional reagents. The second bioconjugate (C225-PG-Pt) employed the same methodology except that polyglutamic acid was used as the carrier. The third and fourth bioconjugates used two different EGF peptides, PEP382 and PEP455, with direct coordination to the Pt center of the cis-DDP fragment. In vivo studies with C225-G5-Pt failed to demonstrate therapeutic activity following intracerebral (ic) convection-enhanced delivery (CED) to F98EGFR glioma-bearing rats. The second bioconjugate, C225-PG-Pt, failed to show in vitro cytotoxicity. Furthermore, because of its high molecular weight, we decided that lower molecular weight peptides might provide better targeting and microdistribution within the tumor. Both PEP382-Pt and PEP455-Pt bioconjugates were cytotoxic in vitro and, based on this, a pilot study was initiated using PEP455-Pt. The end point for this study was tumor size at 6 weeks following tumor cell implantation and 4 weeks following ic CED of PEP455-Pt to F98 glioma-bearing rats. Neuropathologic examination revealed that five of seven rats were either tumor-free or only had microscopic tumors at 42 days following tumor implantation compared to a mean survival time of 20.5 and 26.3 days for untreated controls. In conclusion, we have succeeded in reformatting the toxicity profile of cis-DDP and demonstrated the therapeutic efficacy of the PEP455-Pt bioconjugate in F98 glioma-bearing rats.
Collapse
Affiliation(s)
- Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Gong Wu
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - W Hans Meisen
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Robin J Nakkula
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Weilian Yang
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Tianyao Huo
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - David A Kellough
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Pravin Kaumaya
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH, USA; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Claudia Turro
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Lawrence M Agius
- Department of Pathology, Mater Dei Hospital, University of Malta Medical School, Msida, Malta
| | - Balveen Kaur
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
20
|
111In-cetuximab as a diagnostic agent by accessible epidermal growth factor (EGF) receptor targeting in human metastatic colorectal carcinoma. Oncotarget 2016; 6:16601-10. [PMID: 26062654 PMCID: PMC4599292 DOI: 10.18632/oncotarget.3968] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal adenocarcinoma is a common cause death cancer in the whole world. The aim of this study is to define the 111In-cetuximab as a diagnosis tracer of human colorectal adenocarcinoma. In this research, cell uptake, nano-SPECT/CT scintigraphy, autoradiography, biodistribution and immunohitochemical staining of EGF receptor were included. HCT-116 and HT-29 cell expressed a relatively high and moderate level of EGF receptor, respectively. The nano-SPECT/CT image of 111In-cetuximab showed tumor radiation uptake of subcutaneous HCT-116 xenograft tumor was higher than SW-620. Autoradiography image also showed that tumor of HCT-116 had high 111In-cetuximab uptake. Mice that bearing CT-26 in situ xenograft colorectal tumors showed similar high uptake in vivo and ex vivo through nano-SPECT/CT imaging at 72 hours. Metastatic HCT-116/Luc tumors demonstrated the highest uptake at 72 hours after the injection of 111In-cetuximab. Relatively, results of 111In-DTPA showed that metabolism through urinary system, especially in the kidney. The quantitative analysis of biodistribution showed count value of metastatic HCT-116/Luc tumors that treated with 111In-cetuximab had a significant difference (P < 0.05) compared with that treated with 111In-DTPA after injection 72 hours. Result of immunohistologic staining of EGF receptor also showed high EGF receptor expression and uptake in metastatic colorectal tumors. In summary, we suggested that 111In-cetuximab will be a potential tool for detecting EGF receptor expression in human metastatic colorectal carcinoma.
Collapse
|
21
|
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: 1.8] [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
|
22
|
|
23
|
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: 14] [Impact Index Per Article: 1.6] [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
|
24
|
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: 1.1] [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
|
25
|
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: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022] 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
|
26
|
Amin M, Lockhart AC. The potential role of immunotherapy to treat colorectal cancer. Expert Opin Investig Drugs 2014; 24:329-44. [PMID: 25519074 DOI: 10.1517/13543784.2015.985376] [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] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the fourth most common cancer and the second leading cause of cancer-related death worldwide. Surgery, chemotherapy, radiation therapy and anti-angiogenic therapies form the backbone of treatment for CRC in various stages. Immunotherapy is frequently used either alone or in combination with chemotherapy for the treatment of various cancers such as melanoma, prostate cancer and renal cell cancer. Current CRC research is moving forward to discover ways to incorporate immunotherapies into the treatment of CRC. AREAS COVERED The aim of this review is to summarize the potential role of immunotherapy in CRC. Herein, the authors provide a brief overview of immune modulatory cells, immune surveillance and escape in CRC. They also review vaccine trials in addition to cytokines and monoclonal antibodies. This coverage includes ongoing trials and checkpoint inhibitors such as cytotoxic T lymphocyte antigen-1, programmed cell death-1, and PDL1. EXPERT OPINION Checkpoint inhibitors in combination with either chemotherapy or chemo-antiangiogenic-therapy may represent a future therapeutic approach for CRC incorporating immune system targeting. Given the success of immune-based therapy in other tumor types, the authors anticipate that a similar breakthrough in CRC will be forthcoming.
Collapse
Affiliation(s)
- Manik Amin
- Washington University, Siteman Cancer Center , 660 S. Euclid Ave, Box 8056, St. Louis, MO 63110 , USA
| | | |
Collapse
|
27
|
Kanthala S, Banappagari S, Gokhale A, Liu YY, Xin G, Zhao Y, Jois S. Novel Peptidomimetics for Inhibition of HER2:HER3 Heterodimerization in HER2-Positive Breast Cancer. Chem Biol Drug Des 2014; 85:702-714. [PMID: 25346057 DOI: 10.1111/cbdd.12453] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/23/2014] [Accepted: 10/15/2014] [Indexed: 01/06/2023]
Abstract
The current approach to treating HER2-overexpressed breast cancer is the use of monoclonal antibodies or a combination of antibodies with traditional chemotherapeutic agents or kinase inhibitors. Our approach is to target clinically validated HER2 domain IV with peptidomimetics and inhibit the protein-protein interactions (PPI) of HERs. Unlike antibodies, peptidomimetics have advantages in terms of stability, modification, and molecular size. We have designed peptidomimetics (compounds 5 and 9) that bind to HER2 domain IV, inhibit protein-protein interactions, and decrease cell viability in breast cancer cells with HER2 overexpression. We have shown, using enzyme fragment complementation and proximity ligation assays, that peptidomimetics inhibit the PPI of HER2:HER3. Compounds 5 and 9 suppressed the tumor growth in a xenograft mouse model. Furthermore, we have shown that these compounds inhibit PPI of HER2:HER3 and phosphorylation of HER2 as compared to control in tissue samples derived from in vivo studies. The stability of the compounds was also investigated in mouse serum, and the compounds exhibited stability with a half-life of up to 3 h. These results suggest that the novel peptidomimetics we have developed target the extracellular domain of HER2 protein and inhibit HER2:HER3 interaction, providing a novel method to treat HER2-positive cancer.
Collapse
Affiliation(s)
- Shanthi Kanthala
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201
| | - Sashikanth Banappagari
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201
| | - Ameya Gokhale
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201
| | - Yong-Yu Liu
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201
| | - Gu Xin
- Department of Pharmacology, LSU Health Sciences Center, Shreveport, LA 71103
| | - Yunfeng Zhao
- Department of Pharmacology, LSU Health Sciences Center, Shreveport, LA 71103
| | - Seetharama Jois
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201
| |
Collapse
|
28
|
Pandey JP, Kistner-Griffin E, Radwan FF, Kaur N, Namboodiri AM, Black L, Butler MA, Carreon T, Ruder AM. Endogenous antibody responsiveness to epidermal growth factor receptor is associated with immunoglobulin allotypes and overall survival of patients with glioblastoma. Neuro Oncol 2014; 17:678-84. [PMID: 25326496 DOI: 10.1093/neuonc/nou298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/23/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Immunoglobulin γ marker (GM) and κ marker (KM) allotypes, hereditary antigenic determinants of γ and κ chains, respectively, have been shown to be associated with immunity to a variety of self and nonself antigens, but their possible contribution to immunity to the tumor-associated antigens epidermal growth factor receptor (EGFR) and EGFR variant (v)III has not been evaluated. The aim of the present investigation was to determine whether the interindividual variation in endogenous antibody responsiveness to EGFR and EGFRvIII is associated with particular GM, KM, and Fcγ receptor (FcγR) genotypes and whether antibody levels were associated with the overall survival of patients with glioblastoma. METHODS A total of 126 Caucasian participants with glioblastoma were genotyped for several GM, KM, and FcγR alleles and characterized for IgG antibodies to EGFR and EGFRvIII antigens. RESULTS The anti-EGFR antibody levels associated with GM 3/3 homozygotes and GM 3/17 heterozygotes were similar (15.9 vs 16.4 arbitrary units [AU]/µL) and significantly lower than those associated with GM 17/17 homozygotes (19.6 AU/µL; nominal P = .007). Participants homozygous for the GM 21 allele also had significantly higher levels of anti-EGFR antibodies than GM 5/5 homozygotes and GM 5/21 heterozygotes (20.1 vs 16.0 and 16.3 AU/µL; nominal P = .005). Similar associations were found with immune responsiveness to EGFRvIII. Higher anti-EGFR and anti-EGFRvIII antibody levels were associated with enhanced overall survival (16 vs 11 mo, nominal P = .038 and 20 vs 11 mo, nominal P = .004, respectively). CONCLUSIONS GM allotypes contribute to humoral immunity to EGFR in glioblastoma.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Emily Kistner-Griffin
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Faisal F Radwan
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Navtej Kaur
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Aryan M Namboodiri
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Laurel Black
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Mary Ann Butler
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Tania Carreon
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| | - Avima M Ruder
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina (J.P.P., F.F.R., N.K., A.M.N., L.B.); Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina (E.K-G.); Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Cincinnati, Ohio (M.A.B., T.C., A.M.R.)
| |
Collapse
|
29
|
Navari M, Zare M, Javanmardi M, Asadi-Ghalehni M, Modjtahedi H, Rasaee MJ. Epitope mapping of epidermal growth factor receptor (EGFR) monoclonal antibody and induction of growth-inhibitory polyclonal antibodies by vaccination with EGFR mimotope. Immunopharmacol Immunotoxicol 2014; 36:309-15. [PMID: 25070131 DOI: 10.3109/08923973.2014.945127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One of the proposed approaches in cancer therapy is to induce and direct the patient's own immune system against cancer cells. In this study, we determined the epitope mapping of the rat anti-human epidermal growth factor receptor (EGFR) monoclonal antibody ICR-62 using a phage display of random peptide library and identified a 12 amino acids peptide, which was recognized as a mimotope. The peptide was synthesized and conjugated to bovine serum albumin (BSA) as carrier protein (P-BSA). We have shown that ICR-62 can react specifically with P-BSA as well as native EGFR. Two rabbits were immunized either by BSA or P-BSA and the rabbits IgGs were purified and examined for binding to the antigens, mimotope and the EGFR protein purified from the EGFR overexpressing A431 cell line. We showed that the rabbit IgG generated against the mimotope is capable of inhibiting the growth of A431 cells by 15%, but does not have any effect on the growth of EGFR-negative MDA-MB-453 cell line in vitro. Our results support the need for further investigations on the potential of vaccination with either mimotope of the EGFR or epitope displayed on the surface of phage particles for use in active immunotherapy of cancer.
Collapse
Affiliation(s)
- Mohsen Navari
- Department of Medical Biotechnology, School of Medical Sciences, Tarbiat Modares University , Tehran , Islamic Republic of Iran and
| | | | | | | | | | | |
Collapse
|
30
|
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: 0.7] [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
|
31
|
Shin SY, Yoon H, Hwang D, Ahn S, Kim DW, Koh D, Lee YH, Lim Y. Benzochalcones bearing pyrazoline moieties show anti-colorectal cancer activities and selective inhibitory effects on aurora kinases. Bioorg Med Chem 2013; 21:7018-24. [PMID: 24095020 DOI: 10.1016/j.bmc.2013.09.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/06/2013] [Accepted: 09/06/2013] [Indexed: 12/22/2022]
|