1
|
Han Q, Shi J, Liu J, Fu Y, Li Z, Guo H, Guan X, Xue X, Liu H, Zhao L, Zhang C. Decoding the research landscape of drug resistance and therapeutic approaches in head and neck cancer: a bibliometric analysis from 2000 to 2023. Front Pharmacol 2024; 15:1375110. [PMID: 38645557 PMCID: PMC11026562 DOI: 10.3389/fphar.2024.1375110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction Head and neck cancer is one of the most common tumors worldwide. However, drug resistance in its treatment has become a major factor limiting the efficacy. This study aims to comprehensively understand the current status of research in this field. Methods The study analyzes papers related to therapeutic resistance in head and neck cancer published between 2000 and 2023 in the Web of Science Core Collection To achieve the research objectives, we searched the WoSCC for research and review papers on therapeutic resistance in head and neck cancer from 2000 to 2023, screened the English literature, and analyzed the research hotspots, academic collaborations, and trends in detail using tools such as Citespace, SCImago Graphica, and VOS viewer. Results This study summarizes 787 head and neck cancer treatment resistance publications from WoSCC. The analysis showed that China and the United States are the major contributors in this field, and Grandis Jennifer R and Yang Jai-Sing are the key scholars. Keyword analysis showed that "cisplatin resistance" is a continuing focus of attention, while "Metastasis" and "Ferroptosis" may be emerging research hotspots. Literature clustering analysis pointed out that "Ferroptosis", "Immunotherapy" and "ERK signaling" were the recent hotspots that received extensive attention and citations. Finally, we discuss the current status and challenges in drug-resistant therapies for head and neck cancer. Conclusion This study is the first comprehensive bibliometric analysis of drug resistance in head and neck cancer. Reveals current trends and helps researchers grasp cutting-edge hotspots in the field.
Collapse
Affiliation(s)
- Qi Han
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqi Shi
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jiaojiao Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yang Fu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, China
| | - Zhongxun Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Huina Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoya Guan
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
- Department of Cell Biology and Genetics, the Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liting Zhao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Jagadeeshan S, Novoplansky OZ, Cohen O, Kurth I, Hess J, Rosenberg AJ, Grandis JR, Elkabets M. New insights into RAS in head and neck cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188963. [PMID: 37619805 DOI: 10.1016/j.bbcan.2023.188963] [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: 06/29/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
RAS genes are known to be dysregulated in cancer for several decades, and substantial effort has been dedicated to develop agents that reduce RAS expression or block RAS activation. The recent introduction of RAS inhibitors for cancer patients highlights the importance of comprehending RAS alterations in head and neck cancer (HNC). In this regard, we examine the published findings on RAS alterations and pathway activations in HNC, and summarize their role in HNC initiation, progression, and metastasis. Specifically, we focus on the intrinsic role of mutated-RAS on tumor cell signaling and its extrinsic role in determining tumor-microenvironment (TME) heterogeneity, including promoting angiogenesis and enhancing immune escape. Lastly, we summarize the intrinsic and extrinsic role of RAS alterations on therapy resistance to outline the potential of targeting RAS using a single agent or in combination with other therapeutic agents for HNC patients with RAS-activated tumors.
Collapse
Affiliation(s)
- Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| | - Ofra Z Novoplansky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| | - Oded Cohen
- Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Department of Otolaryngology- Head and Neck Surgery and Oncology, Soroka Medical Center, Beersheva, Israel.
| | - Ina Kurth
- Division of Radiooncology-Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| | - Jennifer R Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| |
Collapse
|
3
|
Chang CW, Chen C, Chang CW, Chiu PY, Yang JS, Chen FA. Effects of Tetrandrine on the Apoptosis of Cisplatin-resistant Oral Cancer Cells. Pharmacogn Mag 2023. [DOI: 10.1177/09731296231158699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Background Cisplatin, the first-line drug for chemotherapy, often has limited treatment efficacy because of resistance and cancer recurrence mechanisms. Tetrandrine is a unique secondary metabolite of Stephania tetrandra. As a traditional Chinese medicine agent, tetrandrine has been reported to have antioxidant, anti-inflammatory, antitumor, and antiangiogenesis activities and has been shown to inhibit the proliferation and angiogenesis of colorectal, lung, and breast cancer cells; potential mechanisms underlying its activities include the promotion of tumor cell apoptosis, promotion of cell cycle arrest, and intensification of reactive oxygen species (ROS) production. Objectives The main treatments for oral cancer are chemotherapy, surgery, and radiotherapy; these treatments are often used in combination. Cancer cells easily develop cisplatin resistance; therefore, we investigated tetrandrine’s potential as a therapy for overcoming resistance to oral cancer drugs. Materials and Methods We used the cisplatin-resistant oral cancer CAR cell line (CAL27) as a research objected and applied inhibitor treatment to clarify the role of tetrandrine in cell death and mitochondrial dysfunction. Results Tetrandrine could effectively inhibit CAR cell proliferation and induce apoptosis, with a corresponding increase in ROS production in mitochondria. Moreover, tetrandrine increased caspase-9 and caspase-3 activity in CAR cells and induced apoptotic mRNA, caspase-3/-9, AIF, and Endo G overexpression. Our results indicate that tetrandrine induces apoptosis in CAR cells through a mitochondrial-dependent signaling pathway.
Collapse
Affiliation(s)
- Chin-Wen Chang
- Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Chun Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Wei Chang
- Department of Pharmacy and Master Program, Tajen University, Pingtung, Taiwan
| | - Po-Yen Chiu
- Department of Pharmacy and Master Program, Tajen University, Pingtung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Fu-An Chen
- Department of Pharmacy and Master Program, Tajen University, Pingtung, Taiwan
| |
Collapse
|
4
|
Kang JJ, Ko A, Kil SH, Mallen-St Clair J, Shin DS, Wang MB, Srivatsan ES. EGFR pathway targeting drugs in head and neck cancer in the era of immunotherapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188827. [PMID: 36309124 DOI: 10.1016/j.bbcan.2022.188827] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/30/2022] [Accepted: 10/16/2022] [Indexed: 11/12/2022]
Abstract
Receptor tyrosine kinases (RTKs) are cell surface receptors that bind growth factor ligands and initiate cellular signaling. Of the 20 classes of RTKs, 7 classes, I-V, VIII, and X, are linked to head and neck cancers (HNCs). We focus on the first class of RTK, epidermal growth factor receptor (EGFR), as it is the most thoroughly studied class. EGFR overexpression is observed in 20% of tumors, and expression of EGFR variant III is seen in 15% of aggressive chemoradiotherapy resistant HNCs. Currently, the EGFR monoclonal antibody (mAb) cetuximab is the only FDA approved RTK-targeting drug for the treatment of HNCs. Clinical trials have also included EGFR mAbs, with tyrosine kinase inhibitors, and small molecule inhibitors targeting the EGFR, MAPK, and mTOR pathways. Additionally, Immunotherapy has been found to be effective in 15 to 20% of patients with recurrent or metastatic HNC as a monotherapy. Thus, attempts are underway for the combinatorial treatment of immunotherapy and EGFR mAbs to determine if the recruitment of immune cells in the tumor microenvironment can overcome EGFR resistance.
Collapse
Affiliation(s)
- James J Kang
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Albert Ko
- Department of Surgery, VA Greater Los Angeles Healthcare System/UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sang Hoon Kil
- Department of Surgery, VA Greater Los Angeles Healthcare System/UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jon Mallen-St Clair
- Department of Otolaryngology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel Sanghoon Shin
- Department of Medicine, VA Greater Los Angeles Healthcare System/UCLA David Geffen School of Medicine, Los Angeles, CA, USA; Molecular Biology Institute, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Marilene B Wang
- Department of Surgery, VA Greater Los Angeles Healthcare System/UCLA David Geffen School of Medicine, Los Angeles, CA, USA; Molecular Biology Institute, UCLA, Los Angeles, CA, USA; Department of Head and Neck Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Eri S Srivatsan
- Department of Surgery, VA Greater Los Angeles Healthcare System/UCLA David Geffen School of Medicine, Los Angeles, CA, USA; Molecular Biology Institute, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Integrin-Activating Peptides Derived from Tenascin-C in Cancer Aggression and New Anticancer Strategy Using the Fibronectin-Derived Integrin-Inactivating Peptide. Molecules 2020; 25:E3239. [PMID: 32708610 PMCID: PMC7396993 DOI: 10.3390/molecules25143239] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Matricellular proteins, which exist in association with the extracellular matrix (ECM) and ECM protein molecules, harbor functional sites within their molecular structures. These functional sites are released through proteolytic cleavage by inflammatory proteinases, such as matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), and the peptides containing these functional sites have unique biological activities that are often not detected in the parent molecules. We previously showed that tenascin-C (TNC) and plasma fibronectin (pFN), examples of matricellular proteins, have cryptic bioactive sites that have opposite effects on cell adhesion to the ECM. A peptide containing the bioactive site of TNC, termed TNIIIA2, which is highly released at sites of inflammation and in the tumor microenvironment (TME), has the ability to potently and persistently activate β1-integrins. In the opposite manner, the peptide FNIII14 containing the bioactive site of pFN has the ability to inactivate β1-integrins. This review highlights that peptide TNIIIA2 can act as a procancer factor and peptide FNIII14 can act as an anticancer agent, based on the regulation on β1-integrin activation. Notably, the detrimental effects of TNIIIA2 can be inhibited by FNIII14. These findings open the possibility for new therapeutic strategies based on the inactivation of β1-integrin by FNIII14.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (M.F.); (M.S.)
| | - Manabu Sasada
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (M.F.); (M.S.)
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda, Yamaguchi 756-0884, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (M.F.); (M.S.)
| |
Collapse
|
6
|
A comparative assessment of the effects of integrin inhibitor cilengitide on primary culture of head and neck squamous cell carcinoma (HNSCC) and HNSCC cell lines. Clin Transl Oncol 2019; 21:1052-1060. [DOI: 10.1007/s12094-018-02025-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/20/2018] [Indexed: 01/17/2023]
|
7
|
Zamora A, Gandioso A, Massaguer A, Buenestado S, Calvis C, Hernández JL, Mitjans F, Rodríguez V, Ruiz J, Marchán V. Toward Angiogenesis Inhibitors Based on the Conjugation of Organometallic Platinum(II) Complexes to RGD Peptides. ChemMedChem 2018; 13:1755-1762. [PMID: 29932312 DOI: 10.1002/cmdc.201800282] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/19/2018] [Indexed: 12/16/2022]
Abstract
A novel conjugate between a cyclometalated platinum(II) complex with dual antiangiogenic and antitumor activity and a cyclic peptide containing the RGD sequence (-Arg-Gly-Asp-) has been synthesized by combining solid- and solution-phase methodologies. Although peptide conjugation rendered a non-cytotoxic compound in all tested tumor cell lines (± αV β3 and αV β5 integrin receptors), the antiangiogenic activity of the Pt-c(RGDfK) conjugate in human umbilical vein endothelial cells at sub-cytotoxic concentrations opens the way to the design of a novel class of angiogenesis inhibitors through conjugation of metallodrugs with high antiangiogenic activity to cyclic RGD-containing peptides or peptidomimetic analogues.
Collapse
Affiliation(s)
- Ana Zamora
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB, University of Barcelona, Martí i Franquès 1-11, 08028, Barcelona, Spain.,Departamento de Química Inorgánica, Universidad de Murcia and Institute for Bio-Health, Research of Murcia (IMIB-Arrixaca), 30071, Murcia, Spain
| | - Albert Gandioso
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB, University of Barcelona, Martí i Franquès 1-11, 08028, Barcelona, Spain
| | - Anna Massaguer
- Departament de Biologia, Universitat de Girona, 17071, Girona, Spain
| | - Silvia Buenestado
- Biomed Division, LEITAT Technological Center, 08028, Barcelona, Spain
| | - Carme Calvis
- Biomed Division, LEITAT Technological Center, 08028, Barcelona, Spain
| | | | - Francesc Mitjans
- Biomed Division, LEITAT Technological Center, 08028, Barcelona, Spain
| | - Venancio Rodríguez
- Departamento de Química Inorgánica, Universidad de Murcia and Institute for Bio-Health, Research of Murcia (IMIB-Arrixaca), 30071, Murcia, Spain
| | - José Ruiz
- Departamento de Química Inorgánica, Universidad de Murcia and Institute for Bio-Health, Research of Murcia (IMIB-Arrixaca), 30071, Murcia, Spain
| | - Vicente Marchán
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB, University of Barcelona, Martí i Franquès 1-11, 08028, Barcelona, Spain
| |
Collapse
|
8
|
αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol Adv 2017; 36:208-227. [PMID: 29155160 DOI: 10.1016/j.biotechadv.2017.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
Integrins are cell adhesion receptors predominantly important during normal and tumor angiogenesis. A sequence present on several extracellular matrix proteins composed of Arg-Gly-Asp (RGD) has attracted attention due to its role in cell adhesion mediated by integrins. The development of ligands that can bind to integrins involved in tumor angiogenesis and brake disease progression has resulted in new investigational drug entities reaching the clinical trial phase in humans. The use of integrin-specific ligands can be useful for the vascularization of regenerative medicine constructs, which remains a major limitation for translation into clinical practice. In order to enhance vascularization, immobilization of integrin-specific RGD peptidomimetics within constructs is a recommended approach, due to their high specificity and selectivity towards certain desired integrins. This review endeavours to address the potential of peptidomimetic-coated biomaterials as vascular network promoters for regenerative medicine purposes. Clinical studies involving molecules tracking active integrins in cancer angiogenesis and reasons for their failure are also addressed.
Collapse
|
9
|
Cedra S, Wiegand S, Kolb M, Dietz A, Wichmann G. Reduced Cytokine Release in Ex Vivo Response to Cilengitide and Cetuximab Is a Marker for Improved Survival of Head and Neck Cancer Patients. Cancers (Basel) 2017; 9:cancers9090117. [PMID: 28872582 PMCID: PMC5615332 DOI: 10.3390/cancers9090117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/18/2017] [Accepted: 09/02/2017] [Indexed: 02/07/2023] Open
Abstract
Targeting of αVβ3 and αVβ5 integrins by cilengitide may reduce growth of solid tumors including head and neck squamous cell carcinoma (HNSCC). Preclinical investigations suggest increased activity of cilengitide in combination with other treatment modalities. The only published trial in HNSCC (ADVANTAGE) investigated cisplatin, 5-fluorouracil, and cetuximab (PFE) without or with once (PFE+CIL1W) or twice weekly cilengitide (PFE+CIL2W) in recurrent/metastatic HNSCC. ADVANTAGE showed good tolerability of the cilengitide arms and even lower adverse events (AEs) compared to PFE but not the benefit in overall survival expected based on preclinical data. As we found in the FLAVINO assay, a short-time ex vivo assay for prediction of chemosensitivity, only a subgroup of HNSCC had an increased suppressive effect of cilengitide containing combination therapies on colony formation of epithelial cells (CFec) and release of pro-angiogenetic and pro-inflammatory cytokines, whereas other HNSCC failed to respond. Response to αVβ3 and αVβ5 integrin targeting by cilengitide classifies HNSCC regarding outcome. We present FLAVINO data arguing for further development of cilengitide plus cetuximab in treatment of a subgroup of HNSCC potentially identified by the FLAVINO assay using a set of biomarkers for response evaluation.
Collapse
Affiliation(s)
- Susan Cedra
- Department of Otolaryngology, Head and Neck Surgery, University of Leipzig, 04103 Leipzig, Germany.
| | - Susanne Wiegand
- Department of Otolaryngology, Head and Neck Surgery, University of Leipzig, 04103 Leipzig, Germany.
| | - Marlen Kolb
- Department of Otolaryngology, Head and Neck Surgery, University of Leipzig, 04103 Leipzig, Germany.
| | - Andreas Dietz
- Department of Otolaryngology, Head and Neck Surgery, University of Leipzig, 04103 Leipzig, Germany.
| | - Gunnar Wichmann
- Department of Otolaryngology, Head and Neck Surgery, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
10
|
Ahmedah HT, Patterson LH, Shnyder SD, Sheldrake HM. RGD-Binding Integrins in Head and Neck Cancers. Cancers (Basel) 2017; 9:cancers9060056. [PMID: 28587135 PMCID: PMC5483875 DOI: 10.3390/cancers9060056] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alterations in integrin expression and function promote tumour growth, invasion, metastasis and neoangiogenesis. Head and neck cancers are highly vascular tumours with a tendency to metastasise. They express a wide range of integrin receptors. Expression of the αv and β1 subunits has been explored relatively extensively and linked to tumour progression and metastasis. Individual receptors αvβ3 and αvβ5 have proved popular targets for diagnostic and therapeutic agents but lesser studied receptors, such as αvβ6, αvβ8, and β1 subfamily members, also show promise. This review presents the current knowledge of integrin expression and function in squamous cell carcinoma of the head and neck (HNSCC), with a particular focus on the arginine-glycine-aspartate (RGD)-binding integrins, in order to highlight the potential of integrins as targets for personalised tumour-specific identification and therapy.
Collapse
Affiliation(s)
- Hanadi Talal Ahmedah
- Radiological Sciences Department, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia.
| | | | - Steven D Shnyder
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| | - Helen M Sheldrake
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| |
Collapse
|
11
|
Guimarães DDO, Lopes DS, Azevedo FVPV, Gimenes SNC, Silva MA, Achê DC, Gomes MSR, Vecchi L, Goulart LR, Yoneyama KAG, Rodrigues RS, Rodrigues VDM. In vitro antitumor and antiangiogenic effects of Bothropoidin, a metalloproteinase from Bothrops pauloensis snake venom. Int J Biol Macromol 2017; 97:770-777. [DOI: 10.1016/j.ijbiomac.2017.01.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
|
12
|
Avβ3 integrin: Pathogenetic role in osteotropic tumors. Crit Rev Oncol Hematol 2015; 96:183-93. [PMID: 26126493 DOI: 10.1016/j.critrevonc.2015.05.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 04/30/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022] Open
Abstract
The interplay of cancer cells and accessory cells within the microenvironment drives signals regulating the proliferation, migration and skeleton colonization. Osteotropism of tumor cells depends on chemokine activation, production of soluble factors and defective gene expression that cooperate within the metastatic niche to the bone resorbing functions of osteoclasts. Adhesion of cancer cells to the extracellular matrix is regulated by integrins as αvβ3 that enhances their invasiveness, pro-tumor angiogenesis and skeleton invasion. Therefore, αvβ3 signaling is implicated in enhancing osteotropism of breast and prostate cancers as well as of multiple myeloma. Targeting of αvβ3 has been adopted to restrain the tumor progression in several cancer models leading to improvement of overall survival as effect of the reduction of both tumor burden and osteotropism by malignant cells. Here, we review both the role of αvβ3 in malignant osteoclastogenesis and its potential targeting to restrain the bone colonization by skeleton invading cancers.
Collapse
|
13
|
Nakamura T, Shinriki S, Jono H, Ueda M, Nagata M, Guo J, Hayashi M, Yoshida R, Ota T, Ota K, Kawahara K, Nakagawa Y, Yamashita S, Nakayama H, Hiraki A, Shinohara M, Ando Y. Osteopontin-integrin α(v)β(3) axis is crucial for 5-fluorouracil resistance in oral squamous cell carcinoma. FEBS Lett 2014; 589:231-9. [PMID: 25497015 DOI: 10.1016/j.febslet.2014.12.004] [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] [Received: 09/12/2014] [Revised: 11/19/2014] [Accepted: 12/02/2014] [Indexed: 01/13/2023]
Abstract
Clinical applications of a chemotherapeutic agent, 5-fluorouracil (5-FU) in oral squamous cell carcinoma (OSCC) have been limited because of drug resistance. This study aimed to identify novel mechanisms of 5-FU resistance. Here we found increased osteopontin (OPN) gene expression in OSCC tissues with resistance to 5-FU-based chemoradiotherapy. OPN overexpression in OSCC cells led to 5-FU resistance and abrogated the prosurvival effect of the drug in a mouse xenograft model. OPN-induced 5-FU resistance required integrin αvβ3. Targeting integrin αvβ3 reversed the resistance in a 5-FU-resistant clone highly expressing OPN. Our data suggest that the OPN-integrin αvβ3 axis is crucial for 5-FU resistance in OSCC.
Collapse
Affiliation(s)
- Takuya Nakamura
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Satoru Shinriki
- Department of Laboratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan.
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Department of Pharmacy, Kumamoto University Hospital, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Masashi Nagata
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Jianying Guo
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Mitsuhiro Hayashi
- Department of Breast and Endocrine Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Ryoji Yoshida
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Tomoko Ota
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Kazutoshi Ota
- Department of Oral and Maxillofacial Surgery, Kumamoto City Hospital, Japan
| | - Kenta Kawahara
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Yoshihiro Nakagawa
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Satoshi Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Hideki Nakayama
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Akimitsu Hiraki
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Masanori Shinohara
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Japan
| |
Collapse
|
14
|
Iagaru A, Mosci C, Shen B, Chin FT, Mittra E, Telli ML, Gambhir SS. 18F-FPPRGD2 PET/CT: Pilot Phase Evaluation of Breast Cancer Patients. Radiology 2014; 273:549-59. [DOI: 10.1148/radiol.14140028] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
15
|
Unger GM, Kren BT, Korman VL, Kimbrough TG, Vogel RI, Ondrey FG, Trembley JH, Ahmed K. Mechanism and efficacy of sub-50-nm tenfibgen nanocapsules for cancer cell-directed delivery of anti-CK2 RNAi to primary and metastatic squamous cell carcinoma. Mol Cancer Ther 2014; 13:2018-29. [PMID: 24867250 DOI: 10.1158/1535-7163.mct-14-0166] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Improved survival for patients with head and neck cancers (HNC) with recurrent and metastatic disease warrants that cancer therapy is specific, with protected delivery of the therapeutic agent to primary and metastatic cancer cells. A further objective should be that downregulation of the intracellular therapy target leads to cell death without compensation by an alternate pathway. To address these goals, we report the utilization of a sub-50-nm tenfibgen (s50-TBG) nanocapsule that delivers RNAi oligonucleotides directed against the essential survival signal protein kinase CK2 (RNAi-CK2) in a cancer cell-specific manner. We have evaluated mechanism and efficacy of using s50-TBG-RNAi-CK2 nanocapsules for therapy of primary and metastatic head and neck squamous cell carcinoma (HNSCC). s50-TBG nanocapsules enter cancer cells via the lipid raft/caveolar pathway and deliver their cargo (RNAi-CK2) preferentially to malignant but not normal tissues in mice. Our data suggest that RNAi-CK2, a unique single-stranded oligonucleotide, co-opts the argonaute 2/RNA-induced silencing complex pathway to target the CK2αα' mRNAs. s50-TBG-RNAi-CK2 inhibited cell growth corresponding with reduced CK2 expression in targeted tumor cells. Treatment of three xenograft HNSCC models showed that primary tumors and metastases responded to s50-TBG-RNAi-CK2 therapy, with tumor shrinkage and 6-month host survival that was achieved at relatively low doses of the therapeutic agent without any adverse toxic effect in normal tissues in the mice. We suggest that our nanocapsule technology and anti-CK2 targeting combine into a therapeutic modality with a potential of significant translational promise.
Collapse
Affiliation(s)
| | - Betsy T Kren
- Medicine, Masonic Cancer Center, University of Minnesota; and
| | | | | | | | | | - Janeen H Trembley
- Laboratory Medicine and Pathology; Masonic Cancer Center, University of Minnesota; and Cellular and Molecular Biochemistry Research Laboratory (151), Minneapolis VA Health Care System, Minneapolis, Minnesota
| | - Khalil Ahmed
- Otolaryngology, and Laboratory Medicine and Pathology; Masonic Cancer Center, University of Minnesota; and Cellular and Molecular Biochemistry Research Laboratory (151), Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
16
|
D'Onofrio N, Caraglia M, Grimaldi A, Marfella R, Servillo L, Paolisso G, Balestrieri ML. Vascular-homing peptides for targeted drug delivery and molecular imaging: meeting the clinical challenges. Biochim Biophys Acta Rev Cancer 2014; 1846:1-12. [PMID: 24704283 DOI: 10.1016/j.bbcan.2014.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/20/2014] [Accepted: 03/22/2014] [Indexed: 12/12/2022]
Abstract
The vasculature of each organ expresses distinct molecular signatures critically influenced by the pathological status. The heterogeneous profile of the vascular beds has been successfully unveiled by the in vivo phage display, a high-throughput tool for mapping normal, diseased, and tumor vasculature. Specific challenges of this growing field are targeted therapies against cancer and cardiovascular diseases, as well as novel bioimaging diagnostic tools. Tumor vasculature-homing peptides have been extensively evaluated in several preclinical and clinical studies both as targeted-therapy and diagnosis. To date, results from several Phase I and II trials have been reported and many other trials are currently ongoing or recruiting patients. In this review, advances in the identification of novel peptide ligands and their corresponding receptors on tumor endothelium through the in vivo phage display technology are discussed. Emphasis is given to recent findings in the clinical setting of vascular-homing peptides selected by in vivo phage display for the treatment of advanced malignancies and their altered vascular beds.
Collapse
Affiliation(s)
- Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Raffaele Marfella
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy
| | - Giuseppe Paolisso
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. de Crecchio 7, 80138 Naples, Italy.
| |
Collapse
|
17
|
Sheldrake HM, Patterson LH. Strategies to inhibit tumor associated integrin receptors: rationale for dual and multi-antagonists. J Med Chem 2014; 57:6301-15. [PMID: 24568695 DOI: 10.1021/jm5000547] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The integrins are a family of 24 heterodimeric transmembrane cell surface receptors. Involvement in cell attachment to the extracellular matrix, motility, and proliferation identifies integrins as therapeutic targets in cancer and associated conditions: thrombosis, angiogenesis, and osteoporosis. The most reported strategy for drug development is synthesis of an agent that is highly selective for a single integrin receptor. However, the ability of cancer cells to change their integrin repertoire in response to drug treatment renders this approach vulnerable to the development of resistance and paradoxical promotion of tumor growth. Here, we review progress toward development of antagonists targeting two or more members of the Arg-Gly-Asp (RGD) binding integrins, notably αvβ3, αvβ5, αvβ6, αvβ8, α5β1, and αIIbβ3, as anticancer therapeutics.
Collapse
Affiliation(s)
- Helen M Sheldrake
- Institute of Cancer Therapeutics, University of Bradford , Bradford, BD7 1DP, U.K
| | | |
Collapse
|
18
|
Cheng NC, van Zandwijk N, Reid G. Cilengitide inhibits attachment and invasion of malignant pleural mesothelioma cells through antagonism of integrins αvβ3 and αvβ5. PLoS One 2014; 9:e90374. [PMID: 24595274 PMCID: PMC3940880 DOI: 10.1371/journal.pone.0090374] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 01/31/2014] [Indexed: 11/18/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an almost invariably fatal, asbestos-related malignancy arising from the mesothelial membrane lining the thoracic cavities. Despite some improvements in treatment, therapy is not considered curative and median survival following diagnosis is less than 1 year. Although still classed as a rare cancer, the incidence of MPM is increasing, and the limited progress in treating the disease makes the identification of new therapies a priority. As there is evidence for expression of the integrins αvβ3 and αvβ5 in MPM, there is a rationale for investigating the effects on MPM of cilengitide, a synthetic peptide inhibitor of integrin αv heterodimer with high specificity for αvβ3 and αvβ5. In mesothelial cells (MC) and 7 MPM cell lines, growth inhibition by cilengitide was associated with the expression level of its target integrins. Furthermore, cilengitide caused cell detachment and subsequent death of anoikis-sensitive cells. It also suppressed invasion of MPM cells in monolayer and three-dimensional cultures. Gene knockdown experiments indicated that these effects of cilengitide were, at least partly, due to antagonism of αvβ3 and αvβ5.
Collapse
Affiliation(s)
- Ngan Ching Cheng
- Asbestos Diseases Research Institute, University of Sydney, Concord, New South Wales, Australia
- * E-mail:
| | - Nico van Zandwijk
- Asbestos Diseases Research Institute, University of Sydney, Concord, New South Wales, Australia
| | - Glen Reid
- Asbestos Diseases Research Institute, University of Sydney, Concord, New South Wales, Australia
| |
Collapse
|
19
|
Vermorken JB, Peyrade F, Krauss J, Mesía R, Remenar E, Gauler TC, Keilholz U, Delord JP, Schafhausen P, Erfán J, Brümmendorf TH, Iglesias L, Bethe U, Hicking C, Clement PM. Cisplatin, 5-fluorouracil, and cetuximab (PFE) with or without cilengitide in recurrent/metastatic squamous cell carcinoma of the head and neck: results of the randomized phase I/II ADVANTAGE trial (phase II part). Ann Oncol 2014; 25:682-688. [PMID: 24567516 PMCID: PMC3933250 DOI: 10.1093/annonc/mdu003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 12/23/2013] [Accepted: 12/31/2013] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recurrent and/or metastatic squamous cell carcinoma of the head and neck (R/M-SCCHN) overexpresses αvβ5 integrin. Cilengitide selectively inhibits αvβ3 and αvβ5 integrins and is investigated as a treatment strategy. PATIENTS AND METHODS The phase I/II study ADVANTAGE evaluated cilengitide combined with cisplatin, 5-fluorouracil, and cetuximab (PFE) in R/M-SCCHN. The phase II part reported here was an open-label, randomized, controlled trial investigating progression-free survival (PFS). Patients received up to six cycles of PFE alone or combined with cilengitide 2000 mg once (CIL1W) or twice (CIL2W) weekly. Thereafter, patients received maintenance therapy (cilengitide arms: cilengitide plus cetuximab; PFE-alone arm: cetuximab only) until disease progression or unacceptable toxicity. RESULTS One hundred and eighty-two patients were treated. Median PFS per investigator read was similar for CIL1W + PFE, CIL2W + PFE, and PFE alone (6.4, 5.6, and 5.7 months, respectively). Accordingly, median overall survival and objective response rates were not improved with cilengitide (12.4 months/47%, 10.6 months/27%, and 11.6 months/36%, respectively). No clinically meaningful safety differences were observed between groups. None of the tested biomarkers (expression of integrins, CD31, Ki-67, vascular endothelial growth factor receptor 2, vascular endothelial-cadherin, type IV collagen, epidermal growth factor receptor, or p16 for human papillomavirus) were predictive of outcome. CONCLUSION Neither of the cilengitide-containing regimens demonstrated a PFS benefit over PFE alone in R/M-SCCHN patients.
Collapse
Affiliation(s)
- J B Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium.
| | - F Peyrade
- Medical Oncology Service, Center Antoine Lacassagne, Nice, France
| | - J Krauss
- Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - R Mesía
- Medical Oncology Service, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - E Remenar
- Head and Neck Surgery, National Oncology Institute, Budapest, Hungary
| | - T C Gauler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen
| | - U Keilholz
- Department of Hematology and Medical Oncology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - J P Delord
- Clinical Research Unit, Institute Claudius Regaud, Toulouse, France
| | - P Schafhausen
- II Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - J Erfán
- Onco-radiology, Jósa András Teaching Hospital, Nyíregyháza, Hungary
| | - T H Brümmendorf
- Department of Hematology and Oncology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - L Iglesias
- Lung and Head and Neck Cancer Unit, Hospital 12 de Octubre, Madrid, Spain
| | - U Bethe
- Merck KGaA, Darmstadt, Germany
| | | | - P M Clement
- Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Heiduschka G, Lill C, Schneider S, Seemann R, Kornek G, Schmid R, Kotowski U, Thurnher D. The effect of cilengitide in combination with irradiation and chemotherapy in head and neck squamous cell carcinoma cell lines. Strahlenther Onkol 2014; 190:472-9. [PMID: 24557056 DOI: 10.1007/s00066-014-0600-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 11/08/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Integrins are highly attractive targets in oncology due to their involvement in angiogenesis in a wide spectrum of cancer entities. Among several integrin inhibitors under clinical evaluation, cilengitide is the most promising compound. However, little is known about the cellular processes induced during cilengitide therapy in combination with irradiation and cisplatin in head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS The cytostatic effect of cilengitide was assessed by proliferation assay in the three HNSCC cell lines SCC25, FaDu and CAL27. Combination experiments with cisplatin and irradiation were performed. Possible synergistic effects were calculated in combination index (CI) analyses. Colony forming inhibition was investigated in clonogenic assays. Real-time PCR arrays were used to evaluate target protein gene expression patterns. Flow cytometry was used to detect apoptosis. RESULTS Used alone, cilengitide has only minor cytotoxic effects in HNSCC cell lines. However, combination with cisplatin resulted in synergistic growth inhibition in all three cell lines. Irradiation showed synergism in short-term experiments and in colony forming assays, an additive effect was detected. Real-time PCR assay detected downregulation of the antiapoptotic protein Bcl-2 after exposure of cells to cilengitide. CONCLUSION Cilengitide in combination with cisplatin and irradiation may be a feasible option for the treatment of patients with head and neck cancer. However, further investigations are required to understand the exact mechanism that leads to synergistic cytotoxicity.
Collapse
Affiliation(s)
- G Heiduschka
- Department of Otorhinolaryngology, Head and Neck Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
21
|
El-Kenawi AE, El-Remessy AB. Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol 2013; 170:712-29. [PMID: 23962094 PMCID: PMC3799588 DOI: 10.1111/bph.12344] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/25/2013] [Accepted: 07/30/2013] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis, a process of new blood vessel formation, is a prerequisite for tumour growth to supply the proliferating tumour with oxygen and nutrients. The angiogenic process may contribute to tumour progression, invasion and metastasis, and is generally accepted as an indicator of tumour prognosis. Therefore, targeting tumour angiogenesis has become of high clinical relevance. The current review aimed to highlight mechanistic details of anti-angiogenic therapies and how they relate to classification and treatment rationales. Angiogenesis inhibitors are classified into either direct inhibitors that target endothelial cells in the growing vasculature or indirect inhibitors that prevent the expression or block the activity of angiogenesis inducers. The latter class extends to include targeted therapy against oncogenes, conventional chemotherapeutic agents and drugs targeting other cells of the tumour micro-environment. Angiogenesis inhibitors may be used as either monotherapy or in combination with other anticancer drugs. In this context, many preclinical and clinical studies revealed higher therapeutic effectiveness of the combined treatments compared with individual treatments. The proper understanding of synergistic treatment modalities of angiogenesis inhibitors as well as their wide range of cellular targets could provide effective tools for future therapies of many types of cancer.
Collapse
Affiliation(s)
- Asmaa E El-Kenawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura UniversityMansoura, Egypt
| | - Azza B El-Remessy
- Center for Pharmacy and Experimental Therapeutics, University of GeorgiaAugusta, GA, USA
- Department of Pharmacology and Toxicology, Georgia Regents UniversityAugusta, GA, USA
- Charlie Norwood VA Medical CenterAugusta, GA, USA
| |
Collapse
|
22
|
Duperret EK, Ridky TW. Focal adhesion complex proteins in epidermis and squamous cell carcinoma. Cell Cycle 2013; 12:3272-85. [PMID: 24036537 DOI: 10.4161/cc.26385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions (FAs) are large, integrin-containing, multi-protein assemblies spanning the plasma membrane that link the cellular cytoskeleton to surrounding extracellular matrix. They play critical roles in adhesion and cell signaling and are major regulators of epithelial homeostasis, tissue response to injury, and tumorigenesis. Most integrin subunits and their associated FA proteins are expressed in skin, and murine genetic models have provided insight into the functional roles of FAs in normal and neoplastic epidermis. Here, we discuss the roles of these proteins in normal epidermal proliferation, adhesion, wound healing, and cancer. While many downstream signaling mechanisms remain unclear, the critically important roles of FAs are highlighted by the development of therapeutics targeting FAs for human cancer.
Collapse
|
23
|
Scaringi C, Enrici RM, Minniti G. Combining molecular targeted agents with radiation therapy for malignant gliomas. Onco Targets Ther 2013; 6:1079-95. [PMID: 23966794 PMCID: PMC3745290 DOI: 10.2147/ott.s48224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The expansion in understanding the molecular biology that characterizes cancer cells has led to the rapid development of new agents to target important molecular pathways associated with aberrant activation or suppression of cellular signal transduction pathways involved in gliomagenesis, including epidermal growth factor receptor, vascular endothelial growth factor receptor, mammalian target of rapamycin, and integrins signaling pathways. The use of antiangiogenic agent bevacizumab, epidermal growth factor receptor tyrosine kinase inhibitors gefitinib and erlotinib, mammalian target of rapamycin inhibitors temsirolimus and everolimus, and integrin inhibitor cilengitide, in combination with radiation therapy, has been supported by encouraging preclinical data, resulting in a rapid translation into clinical trials. Currently, the majority of published clinical studies on the use of these agents in combination with radiation and cytotoxic therapies have shown only modest survival benefits at best. Tumor heterogeneity and genetic instability may, at least in part, explain the poor results observed with a single-target approach. Much remains to be learned regarding the optimal combination of targeted agents with conventional chemoradiation, including the use of multipathways-targeted therapies, the selection of patients who may benefit from combined treatments based on molecular biomarkers, and the verification of effective blockade of signaling pathways.
Collapse
Affiliation(s)
- Claudia Scaringi
- Department of Radiation Oncology, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | | | | |
Collapse
|
24
|
Feasibility of 6-month maintenance cetuximab after adjuvant concurrent chemoradiation plus cetuximab in squamous cell carcinoma of the head and neck. Strahlenther Onkol 2013; 189:625-31. [DOI: 10.1007/s00066-013-0378-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/06/2013] [Indexed: 10/26/2022]
|
25
|
Bábíčková J, Tóthová Ľ, Boor P, Celec P. In vivo phage display--a discovery tool in molecular biomedicine. Biotechnol Adv 2013; 31:1247-59. [PMID: 23623852 DOI: 10.1016/j.biotechadv.2013.04.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/15/2013] [Accepted: 04/15/2013] [Indexed: 12/13/2022]
Abstract
In vivo phage display is a high-throughput method for identifying target ligands specific for different vascular beds. Targeting is possible due to the heterogeneous expression of receptors and other antigens in a particular vascular bed. Such expression is additionally influenced by the physiological or pathological status of the vasculature. In vivo phage display represents a technique that is usable in both, vascular mapping and targeted drug development. In this review, several important methodological aspects of in vivo phage display experiments are discussed. These include choosing an appropriate phage library, an appropriate animal model and the route of phage library administration. In addition, peptides or antibodies identified by in vivo phage display homing to specific types of vascular beds, including the altered vasculature present in several types of diseases are summarized. Still, confirmation in independent experiments and reproduction of identified sequences are needed for enhancing the clinical applicability of in vivo phage display research.
Collapse
Affiliation(s)
- Janka Bábíčková
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia; Division of Nephrology, RWTH University, Aachen, Germany
| | | | | | | |
Collapse
|
26
|
Abstract
In locally advanced cervical cancer, 18F-fluorodeoxyglucose (FDG) positron emission tomography – computed tomography (PET/CT) has become important in the initial evaluation of disease extent. It is superior to other imaging modalities for lymph node status and distant metastasis. PET-defined cervical tumor volume predicts progression-free and overall survival. Higher FDG uptake in both primary and regional lymph nodes is strongly predictive of worse outcome. FDG-PET is useful for assessing treatment response 3 months after completing concurrent chemo-radiotherapy (CRT) and predicting long-term survival, and in suspected disease recurrence. In the era of image-guided adaptive radiotherapy, accurately defining disease areas is critical to avoid irradiating normal tissue. Based on additional information provided by FDG-PET, radiation treatment volumes can be modified and higher doses to FDG-positive lymph nodes safely delivered. FDG-PET/CT has been used for image-guided brachytherapy of FDG-avid tumor volume, while respecting low doses to bladder and rectum. Despite survival improvements due to CRT in cervical cancer, disease recurrences continue to be a major problem. Biological rationale exists for combining novel non-cytotoxic agents with CRT, and drugs targeting specific molecular pathways are under clinical development. The integration of these targeted therapies in clinical trials, and the need for accurate predictors of radio-curability is essential. New molecular imaging tracers may help identifying more aggressive tumors. 64Cu-labeled diacetyl-di(N(4)-methylthiosemicarbazone) is taken up by hypoxic tissues, which may be valuable for prognostication and radiation treatment planning. PET/CT imaging with novel radiopharmaceuticals could further impact cervical cancer treatment as surrogate markers of drug activity at the tumor microenvironment level. The present article reviews the current and emerging role of PET/CT in the management of cervical cancer.
Collapse
Affiliation(s)
- Fernanda G Herrera
- Department of Radiation Oncology, Lausanne University Hospital Lausanne, Switzerland
| | | |
Collapse
|
27
|
Phase Ib trial of the Toll-like receptor 9 agonist IMO-2055 in combination with 5-fluorouracil, cisplatin, and cetuximab as first-line palliative treatment in patients with recurrent/metastatic squamous cell carcinoma of the head and neck. Invest New Drugs 2013; 31:1207-16. [PMID: 23397499 DOI: 10.1007/s10637-013-9933-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/24/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND This Phase Ib trial assessed the maximum tolerated dose (MTD) and safety of the Toll-like receptor 9 agonist IMO-2055 combined with 5-fluorouracil, cisplatin, and cetuximab (PFE) as first-line palliative treatment in patients with relapsed and/or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN). METHODS A standard 3+3 study design was used. Patients were sequentially enrolled to be treated with IMO-2055 (0.16, 0.32, or 0.48 mg/kg/day; days 1, 8, 15), 5-fluorouracil (1,000 mg/m(2)/day; days 1-4), cisplatin (100 mg/m(2)/day; day 1) and cetuximab (400 mg/m(2)/day first dose; then 250 mg/m(2)/day; days 1, 8, 15) every 3 weeks. RESULTS Thirteen patients received IMO-2055. Dose-limiting toxicities (DLTs; ie, any Grade [G]3/4 treatment-related adverse events [TEAEs] in cycle 1) occurred in 2/4 patients treated with IMO-2055 0.32 mg/kg (G4 hypokalemia and hypomagnesemia [n=1]; G4 septicemia, hyperthermia, febrile neutropenia, and G3 hypotension [n=1]). In the IMO-2055 0.16-mg/kg expansion cohort, 1 patient experienced DLTs of G3 sepsis, bacteremia, and hyperthermia. The most common G ≥ 3 TEAEs were neutropenia (n=9; not including febrile neutropenia [n=1]), hypokalemia (n=5), and hypomagnesemia (n=4). Serious adverse events (SAEs) occurred in 8 patients, including 4 with SAEs considered IMO-2055 related; 1 of these patients died. Best response achieved overall was partial response in 3 patients and stable disease in 9 patients. The overall safety profile led to early trial termination; the safety monitoring committee did not confirm the MTD (formally IMO-2055 0.16 mg/kg). CONCLUSIONS Regimens combining IMO-2055 and PFE cannot be recommended for further development in R/M SCCHN patients.
Collapse
|
28
|
Ruiz-Esparza GU, Flores-Arredondo JH, Segura-Ibarra V, Torre-Amione G, Ferrari M, Blanco E, Serda RE. The physiology of cardiovascular disease and innovative liposomal platforms for therapy. Int J Nanomedicine 2013; 8:629-40. [PMID: 23413209 PMCID: PMC3572823 DOI: 10.2147/ijn.s30599] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Heart disease remains the major cause of death in males and females, emphasizing the need for novel strategies to improve patient treatment and survival. A therapeutic approach, still in its infancy, is the development of site-specific drug-delivery systems. Nanoparticle-based delivery systems, such as liposomes, have evolved into robust platforms for site-specific delivery of therapeutics. In this review, the clinical impact of cardiovascular disease and the pathophysiology of different subsets of the disease are described. Potential pathological targets for therapy are introduced, and promising advances in nanotherapeutic cardiovascular applications involving liposomal platforms are presented.
Collapse
|
29
|
Hoffmann TK. Systemic therapy strategies for head-neck carcinomas: Current status. GMS CURRENT TOPICS IN OTORHINOLARYNGOLOGY, HEAD AND NECK SURGERY 2012; 11:Doc03. [PMID: 23320055 PMCID: PMC3544206 DOI: 10.3205/cto000085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Head and neck cancers, most of which are squamous cell tumours, have an unsatisfactory prognosis despite intensive local treatment. This can be attributed, among other factors, to tumour recurrences inside or outside the treated area, and metastases at more distal locations. These tumours therefore require not only the standard surgical and radiation treatments, but also effective systemic modalities. The main option here is antineoplastic chemotherapy, which is firmly established in the palliative treatment of recurrent or metastatic stages of disease, and is used with curative intent in the form of combined simultaneous or adjuvant chemoradiotherapy in patients with inoperable or advanced tumour stages. Neoadjuvant treatment strategies for tumour reduction before surgery have yet to gain acceptance. Induction chemotherapy protocols before radiotherapy have to date been used in patients at high risk of distant metastases or as an aid for decision-making ("chemoselection") in those with extensive laryngeal cancers, prior to definitive chemoradiotherapy or laryngectomy. Triple-combination induction therapy (taxanes, cisplatin, 5-fluorouracil) shows high remission rates with significant toxicity and, in combination with (chemo-)radiotherapy, is currently being compared with simultaneous chemoradiotherapy; the current gold standard with regards to efficacy and long-term toxicity.A further systemic treatment strategy, called "targeted therapy", has been developed to help increase specificity and reduce toxicity. An example of targeted therapy, EGFR-specific antibodies, can be used in palliative settings and, in combination with radiotherapy, to treat advanced head and neck cancers. A series of other novel biologicals such as signal cascade inhibitors, genetic agents, or immunotherapies, are currently being evaluated in large-scale clinical studies, and could prove useful in patients with advanced, recurring or metastatic head and neck cancers. When developing a lasting, individualised systemic tumour therapy, the critical evaluation criteria are not only efficacy and acute toxicity but also (long-term) quality-of-life and the identification of dedicated predictive biomarkers.
Collapse
|
30
|
Schuler PJ, Hoffmann TK, Gauler TC, Bergmann C, Brandau S, Lang S. [Immunotherapy of head and neck cancer. Current developments]. HNO 2012; 61:559-72. [PMID: 23247754 DOI: 10.1007/s00106-012-2635-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In order to improve the prognosis for patients with head and neck squamous cell cancer (HNSCC) the introduction of new therapeutic strategies is necessary. The concept of immunotherapy has been applied and improved for several years and recent studies have used tumor-specific antigens which facilitates targeted oncologic therapy. However, immunotherapy is hampered by the fact that immunosuppressive mechanisms are pronounced and relevant effector cells are suppressed, especially in patients with HNSCC. Successful immunotherapy could induce an antitumor immune response by restitution of these cell populations. Current anti-tumor immunotherapy includes unspecific immune stimulation, genetic modification of tumor and immune cells, the use of monoclonal antibodies, e.g. cetuximab, adoptive cell transfer and tumor vaccination. In the future, these biologic therapies alone or in combination with conventional therapeutic regimens could present a valuable therapeutic option for HNSCC patients.
Collapse
Affiliation(s)
- P J Schuler
- Klinik und Poliklinik für Hals-, Nasen- und Ohrenkrankheiten, Universität Duisburg-Essen.
| | | | | | | | | | | |
Collapse
|
31
|
Ten Hagen TLM, Seynhaeve ALB, de Wiel-Ambagtsheer GA, de Bruijn EA, van Tiel ST, Ruegg C, Meyring M, Grell M, Goodman SL, Eggermont AMM. The αVβ3/αVβ5 integrin inhibitor cilengitide augments tumor response to melphalan isolated limb perfusion in a sarcoma model. Int J Cancer 2012; 132:2694-704. [PMID: 23152080 DOI: 10.1002/ijc.27940] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 10/15/2012] [Indexed: 01/13/2023]
Abstract
Isolated limb perfusion (ILP) with melphalan and tumor necrosis factor (TNF)-α is used to treat bulky, locally advanced melanoma and sarcoma. However, TNF toxicity suggests a need for better-tolerated drugs. Cilengitide (EMD 121974), a novel cyclic inhibitor of alpha-V integrins, has both anti-angiogenic and direct anti-tumor effects and is a possible alternative to TNF in ILP. In this study, rats bearing a hind limb soft tissue sarcoma underwent ILP using different combinations of melphalan, TNF and cilengitide in the perfusate. Further groups had intra-peritoneal (i.p.) injections of cilengitide or saline 2 hr before and 3 hr after ILP. A 77% response rate (RR) was seen in animals treated i.p. with cilengitide and perfused with melphalan plus cilengitide. The RR was 85% in animals treated i.p. with cilengitide and ILP using melphalan plus both TNF and cilengitide. Both RRs were significantly greater than those seen with melphalan or cilengitide alone. Histopathology showed that high RRs were accompanied by disruption of tumor vascular endothelium and tumor necrosis. Compared with ILP using melphalan alone, the addition of cilengitide resulted in a three to sevenfold increase in melphalan concentration in tumor but not in muscle in the perfused limb. Supportive in vitro studies indicate that cilengitide both inhibits tumor cell attachment and increases endothelial permeability. Since cilengitide has low toxicity, these data suggest the agent is a good alternative to TNF in the ILP setting.
Collapse
Affiliation(s)
- Timo L M Ten Hagen
- Department of Surgery, Section Surgical Oncology, Laboratory Experimental Surgical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Schöpper S, Laban S, Güldenzoph B, Münscher A, Knecht R, Kurzweg T. [New aspects in the therapy of recurrence and metastasis of head and neck cancer: highlights from the 2012 ASCO meeting]. HNO 2012; 60:957-61. [PMID: 23114541 DOI: 10.1007/s00106-012-2596-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The majority of patients with a squamous cell carcinoma of the head and neck region (HNSCC) initially present in a locally advanced stage of the disease. Despite aggressive protocols in first-line treatment, some of these patients develop locoregional recurrences or metastases and are, in particular, extraordinarily challenging for the multidisciplinary treatment team. Therefore, among resectability and prior therapy, age, performance status and individual expectations of the patient have to be taken into account. Apart from surgical options like salvage surgery, chemotherapy and target therapy as well as reirradiation are possible treatment concepts. Unfortunately, most treatment options offer only little to no survival benefit.
Collapse
Affiliation(s)
- S Schöpper
- Klinik und Poliklinik für Hals-, Nasen- und Ohrenheilkunde, Kopf-Hals-Tumorzentrum des Universitären Cancer Center Hamburg, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Deutschland.
| | | | | | | | | | | |
Collapse
|
33
|
Antiangiogenic therapy for glioma. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:483040. [PMID: 22830012 PMCID: PMC3399341 DOI: 10.1155/2012/483040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/27/2012] [Accepted: 05/02/2012] [Indexed: 01/18/2023]
Abstract
Currently, antiangiogenic agents are routinely used for the treatment of patients with glioma. However, despite advances in pharmacological and surgical therapy, glioma remains an incurable disease. Indeed, the formation of an abnormal tumor vasculature and the invasion of glioma cells along neuronal tracts are proposed to comprise the major factors that are attributed to the therapeutic resistance of these tumors. The development of curative therapeutic modalities for the treatment of glioma requires further investigation of the molecular mechanisms regulating angiogenesis and invasion. In this review, we discuss the molecular characteristics of angiogenesis and invasion in human malignant glioma, we present several available drugs that are used or can potentially be utilized for the inhibition of angiogenesis in glioma, and we focus our attention on the key mediators of the molecular mechanisms underlying the resistance of glioma to antiangiogenic therapy.
Collapse
|
34
|
Saunders NA, Simpson F, Thompson EW, Hill MM, Endo-Munoz L, Leggatt G, Minchin RF, Guminski A. Role of intratumoural heterogeneity in cancer drug resistance: molecular and clinical perspectives. EMBO Mol Med 2012; 4:675-84. [PMID: 22733553 PMCID: PMC3494067 DOI: 10.1002/emmm.201101131] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 04/22/2012] [Accepted: 04/30/2012] [Indexed: 12/15/2022] Open
Abstract
Drug resistance continues to be a major barrier to the delivery of curative therapies in cancer. Historically, drug resistance has been associated with over-expression of drug transporters, changes in drug kinetics or amplification of drug targets. However, the emergence of resistance in patients treated with new-targeted therapies has provided new insight into the complexities underlying cancer drug resistance. Recent data now implicate intratumoural heterogeneity as a major driver of drug resistance. Single cell sequencing studies that identified multiple genetically distinct variants within human tumours clearly demonstrate the heterogeneous nature of human tumours. The major contributors to intratumoural heterogeneity are (i) genetic variation, (ii) stochastic processes, (iii) the microenvironment and (iv) cell and tissue plasticity. Each of these factors impacts on drug sensitivity. To deliver curative therapies to patients, modification of current therapeutic strategies to include methods that estimate intratumoural heterogeneity and plasticity will be essential.
Collapse
Affiliation(s)
- Nicholas A Saunders
- Epithelial Cancer Program, University of Queensland, Diamantine Institute, Princess Alexandra Hospital, Queensland, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Specenier P, Vermorken JB. Biologic therapy in head and neck cancer: a road with hurdles. ISRN ONCOLOGY 2012; 2012:163752. [PMID: 22745915 PMCID: PMC3382358 DOI: 10.5402/2012/163752] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 04/11/2012] [Indexed: 12/28/2022]
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in the vast majority of cases of squamous cell carcinoma of the head and neck (SCCHN). A high EGFR expression is associated with an unfavorable prognosis. Cetuximab is a chimeric human/murine IgG1 antibody which binds with high affinity to the EGFR. It is the only targeted agent which got approval for the treatment of SCCHN from the regulatory agencies of Europe and the United States, both in locoregionally advanced disease, in association with radiation, and in recurrent/metastatic disease. The outcome of trials involving other EGFR-directed monoclonal antibodies, that is, zalutumumab and panitumumab, was consistent with the results with cetuximab. However these trials failed to meet their primary endpoint. The results with EGFR-directed tyrosine kinase inhibitors have been disappointing. Other potential targets for treatment in SCCHN include the entire ErbB family, the vascular endothelial growth factor (VEGF) and its receptor (VEGFR), the insulin-like growth factor 1 receptor (IGF-1R), the insulin receptor (IR), histone deacetylases (HDAC), the mammalian target of rapamycin (mTOR), the platelet-derived growth factor receptor (PDGFR), heat-shock protein 90 (HSP90), nuclear factor-kappa B (NF-κB), aurora A or B, and phosphatidylinositol 3-kinase (PIK3CA).
Collapse
Affiliation(s)
- Pol Specenier
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Jan B. Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| |
Collapse
|
36
|
Kurzweg T, Möckelmann N, Laban S, Knecht R. Current treatment options for recurrent/metastatic head and neck cancer: a post-ASCO 2011 update and review of last year's literature. Eur Arch Otorhinolaryngol 2012; 269:2157-67. [PMID: 22437252 DOI: 10.1007/s00405-012-1998-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 03/07/2012] [Indexed: 10/28/2022]
Abstract
The majority of patients with a squamous cell carcinoma of the head and neck present with locally advanced tumors. The first-line treatment of locally advanced tumor stages consists of a combined modality management. Despite these aggressive protocols, many patients develop locoregional recurrences or metastasis and place particularly high demands on the interdisciplinary treatment team. Treatment with a curative intent must be differentiated from a palliative one. In addition to prior treatment, resectability, age and performance status, patient wishes must be taken into consideration in treatment planning, especially considering that most therapies offer little to no overall survival benefit. Salvage surgery, chemo- and target therapies, and reirradiation are head and neck surgeon's and radiooncologist's weapons in the fight against these strong opponents. This review focuses on publications and meeting news from last year and reviews the current status of the clinical application of each treatment modality in recurrent or metastatic head and neck cancer.
Collapse
Affiliation(s)
- T Kurzweg
- Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | | | | | | |
Collapse
|
37
|
Prager GW, Poettler M, Unseld M, Zielinski CC. Angiogenesis in cancer: Anti-VEGF escape mechanisms. Transl Lung Cancer Res 2012; 1:14-25. [PMID: 25806151 PMCID: PMC4367591 DOI: 10.3978/j.issn.2218-6751.2011.11.02] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 11/29/2011] [Indexed: 12/21/2022]
Abstract
It is now widely accepted that tumor-angiogenesis plays a crucial role in tumor growth, tumor propagation and metastasis formation. Among several angiogenic activators, the vascular endothelial growth factor (VEGF) and its receptors represent one of the major inducers of tumor angiogenesis. Thus, this system has become the focus of therapeutic interventions, which led to the approval of the anti-VEGF blocking antibody bevacizumab and the VEGFR-2 pathway inhibitors pazopanib, sorafenib and sunitinib. However, not every cancer patient benefits from such treatment or finally becomes resistant to anti-VEGF approaches; others are suffering from adverse effects. Thus, there is an urgent need for a better understanding of VEGF-independent mechanisms leading to angiogenesis in cancer. This review focuses on anti-VEGF escape mechanisms of tumor cells and its microenvironment.
Collapse
Affiliation(s)
- Gerald W Prager
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Marina Poettler
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Matthias Unseld
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Christoph C Zielinski
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| |
Collapse
|
38
|
Aziz K, Nowsheen S, Pantelias G, Iliakis G, Gorgoulis VG, Georgakilas AG. Targeting DNA damage and repair: embracing the pharmacological era for successful cancer therapy. Pharmacol Ther 2011; 133:334-50. [PMID: 22197993 DOI: 10.1016/j.pharmthera.2011.11.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 11/30/2011] [Indexed: 12/19/2022]
Abstract
DNA is under constant assault from genotoxic agents which creates different kinds of DNA damage. The precise replication of the genome and the continuous surveillance of its integrity are critical for survival and the avoidance of carcinogenesis. Cells have evolved an arsenal of repair pathways and cell cycle checkpoints to detect and repair DNA damage. When repair fails, typically cell cycle progression is halted and apoptosis is initiated. Here, we review the different sources and types of DNA damage including DNA replication stress and oxidative stress, the repair pathways that cells utilize to repair damaged DNA, and discuss their biological significance, especially with reference to cancer induction and cancer therapy. We also describe the main methodologies currently used for the detection of DNA damage with their strengths and limitations. We conclude with an outline as to how this information can be used to identify novel pharmacological targets for DNA repair pathways or enhancers of DNA damage to develop improved treatment strategies that will benefit cancer patients.
Collapse
Affiliation(s)
- K Aziz
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
39
|
Prager GW, Poettler M. Angiogenesis in cancer. Basic mechanisms and therapeutic advances. Hamostaseologie 2011; 32:105-14. [PMID: 21837355 DOI: 10.5482/ha-1163] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022] Open
Abstract
Etiological concepts on cancer development, malignant growth and tumour propagation have undergone a revolutionary development during recent years: Among other aspects, the discovery of angiogenesis - the growth of new blood vessels from pre-existing vasculature - as a key element in the pathogenesis of malignancy has opened an abundance of biologic insights and subsequent therapeutic options, which have led to improved prognosis in many cancers including those originating from colon, lung, breast and kidney. Thereby, targeting the major pro-angiogenic stimulus vascular endothelial growth factor (VEGF) became the focus for therapeutic interventions. However, the use of VEGF-targeting drugs has been shown to be of limited efficacy, which might lie in the fact that tumor angiogenesis is mediated by a variety of different subcellular systems. This review focuses on the basic mechanisms involved in angiogenesis, which potentially represent novel targets for pharmacological agents in the treatment of malignancies.
Collapse
Affiliation(s)
- G W Prager
- Comprehensive Cancer Center Vienna, Department of Medicine I, Medical University of Vienna, Austria.
| | | |
Collapse
|