1
|
VandenHeuvel SN, Nash LL, Raghavan SA. Dormancy in Metastatic Colorectal Cancer: Tissue Engineering Opportunities for In Vitro Modeling. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195931 DOI: 10.1089/ten.teb.2025.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Colorectal cancer (CRC) recurs at a striking rate, specifically in patients with liver metastasis. Dormant CRC cells disseminated following initial primary tumor resection or treatment often resurface years later to form aggressive, therapy-resistant tumors that result in high patient mortality. Routine imaging-based screenings often fail to detect dormant cancer cell clusters, and there are no overt symptomatic presentations, making dormant CRC a major clinical challenge to diagnose and treat. Tissue engineering approaches are ideally suited to model dormant cancer cells and enable the discovery of therapeutic vulnerabilities or unique mechanistic dependencies of dormant CRC. Emerging evidence suggests that tissue-engineered approaches have been successfully used to model dormant breast and lung cancer. With CRC responsible for the second most cancer-related deaths worldwide and CRC patients commonly experiencing recurrence, it is essential to expand dormancy models to understand this phenomenon in the context of CRC. Most published in vitro models of CRC dormancy simplify the complex tumor microenvironment with two-dimensional culture systems to elucidate dormancy-driving mechanisms. Building on this foundation, future research should apply tissue engineering methods to this growing field to generate competent three-dimensional models and increase mechanistic knowledge. This review summarizes the current state of in vitro CRC dormancy models, highlighting the techniques utilized to give rise to dormant CRC cells: nutrient depletion, anticancer drugs, physical extracellular matrix interactions, and genetic manipulation. The metrics used to validate dormancy within each model are also consolidated to demonstrate the lack of established standards and the ambiguity around comparing studies that have been validated differently. The methods of these studies are organized in this review to increase comprehensibility and identify needs and opportunities for future bioengineered in vitro models to address dormancy-driven mortality in patients with CRC liver metastasis. Impact Statement Dormant cancer drives high patient mortality, especially in metastatic colorectal cancer, owing to the clinical inability to identify dormant cells prior to their overt recurrence. Lacking clinical insights, in vitro modeling for mechanistic and therapeutic discovery is hindered. Here, we review models and methods of inducing colorectal cancer dormancy with the goal of consolidating findings for reference. We also highlight the need for advanced, tissue-engineered models to better mimic the organ-specific 3D microenvironment of metastatic colorectal cancer. New models would enable breakthroughs in understanding mechanisms driving dormancy progression and reversal, thereby providing context for therapeutic advances to improve patient survival.
Collapse
Affiliation(s)
| | - Lucia L Nash
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Regional Excellence Center in Cancer, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
2
|
Shevtsov M, Yudintceva N, Bobkov D, Likhomanova R, Nechaeva A, Mikhailova E, Oganesyan E, Fedorov V, Kurkin A, Lukacheva A, Fofanov G, Kim A, Fedorov E, Sitovskaya D, Ulitin A, Mikhailova N, Anufriev I, Istomina M, Murashko E, Kessenikh E, Aksenov N, Vakhitova Y, Samochernykh K, Pitkin E, Shlyakhto E, Combs SE. RAS70 peptide targets multiforme glioblastoma by binding to the plasma membrane heat shock protein HSP70. Front Oncol 2025; 15:1543657. [PMID: 40196735 PMCID: PMC11973282 DOI: 10.3389/fonc.2025.1543657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
Multiforme glioblastoma-homing peptides, particularly targeting plasma membrane-bound heat shock protein mHsp70, demonstrate great application potential for tumor theranostics. In the current study, to further increase the bioavailability as well as penetration capacity through the blood-brain barrier (BBB) of the mHsp70-targeted peptide TKDNNLLGRFELSG, which is known to bind to the oligomerization sequence of mHsp70 chaperone, the latter was conjugated with tripeptide RGD (forming chimeric peptide termed RAS70). In the model BBB system RAS70 efficiently crossed the barrier accumulating in the glioblastoma cells. Subsequently, in the orthotopic glioma models, intravenous administration of the fluorescently labeled agent (RAS70-sCy7.5) resulted in the tumor retention of peptide (further confirmed by histological studies). Thus, as shown by the biodistribution studies employing epifluorescence imaging, accumulation of RAS70-sCy7.5 in C6 glioma was significantly enhanced as compared to scramble peptide. Local application of the RAS70-sCy7.5 peptide that was sprayed over the dissected brain tissues helped to efficiently delineate the tumors in glioma-bearing animals employing an intraoperative fluorescent imaging system. Tumor-specific internalization of the peptide was further confirmed on the ex vivo primary GBM samples obtained from adult neurooncological patients. In conclusion, RAS70 peptide demonstrated high glioma-homing properties which could be employed for the intraoperative tumor visualization as well as for developing a potential carrier for drug delivery.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Natalia Yudintceva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Danila Bobkov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| | - Ruslana Likhomanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Anastasiya Nechaeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elena Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elena Oganesyan
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Andrey Kurkin
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Anastasiya Lukacheva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Georgii Fofanov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Aleksander Kim
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Evegeniy Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Daria Sitovskaya
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Alexey Ulitin
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Natalia Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Ilya Anufriev
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Maria Istomina
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Ekaterina Murashko
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elizaveta Kessenikh
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Nikolay Aksenov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Yulia Vakhitova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Emil Pitkin
- Department of Statistics and Data Science, Wharton School, University of Pennsylvania, Philadelphia, PA, United States
| | - Evgeny Shlyakhto
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
3
|
Xiang D, Fu L, Yang Y, Liu C, He Y. Evaluating the diagnostic accuracy of heat shock proteins and their combination with Alpha-Fetoprotein in the detection of hepatocellular carcinoma: a meta-analysis. BMC Gastroenterol 2024; 24:178. [PMID: 38773451 PMCID: PMC11110180 DOI: 10.1186/s12876-024-03260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 05/10/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND A growing body of research suggests that heat shock proteins (HSPs) may serve as diagnostic biomarkers for hepatocellular carcinoma (HCC), but their results are still controversial. This meta-analysis endeavors to evaluate the diagnostic accuracy of HSPs both independently and in conjunction with alpha-fetoprotein (AFP) as novel biomarkers for HCC detection. METHODS Pooled statistical indices, including sensitivity, specificity, diagnostic odds ratio (DOR), positive likelihood ratio (PLR), and negative likelihood ratio (NLR) with 95% confidence intervals (CI), were computed to assess the diagnostic accuracy of HSPs, AFP, and their combinations. Additionally, the area under the summary receiver operating characteristic (SROC) curve (AUC) was determined. RESULTS A total of 2013 HCC patients and 1031 control subjects from nine studies were included in this meta-analysis. The summary estimates for HSPs and AFP are as follows: sensitivity of 0.78 (95% CI: 0.69-0.85) compared to 0.73 (95% CI: 0.65-0.80); specificity of 0.89 (95% CI: 0.81-0.95) compared to 0.86 (95% CI: 0.77-0.91); PLR of 7.4 (95% CI: 3.7-14.9) compared to 5.1 (95% CI: 3.3-8.1); NLR of 0.24 (95% CI: 0.16-0.37) compared to 0.31 (95% CI: 0.24-0.41); DOR of 30.19 (95% CI: 10.68-85.37) compared to 16.34 (95% CI: 9.69-27.56); and AUC of 0.90 (95% CI: 0.87-0.92) compared to 0.85 (95% CI: 0.82-0.88). The pooled sensitivity, specificity, PLR, NLR, DOR and AUC were 0.90 (95% CI: 0.82-0.95), 0.94 (95% CI: 0.82-0.98), 14.5 (95% CI: 4.6-45.4), 0.11 (95% CI: 0.06-0.20), 133.34 (95% CI: 29.65-599.61), and 0.96 (95% CI: 0.94-0.98) for the combination of HSPs and AFP. CONCLUSION Our analysis suggests that HSPs have potential as a biomarker for clinical use in the diagnosis of HCC, and the concurrent utilization of HSPs and AFP shows notable diagnostic effectiveness for HCC.
Collapse
Affiliation(s)
- Dan Xiang
- Department of Laboratory Medicine, Ya'an People's Hospital, Yaan, 625000, China
| | - Lifang Fu
- Department of Laboratory Medicine, Ya'an People's Hospital, Yaan, 625000, China
| | - Ying Yang
- Department of Clinical Laboratory, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
| | - ChengJiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Anqing, 246000, China.
| | - Yong He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Hu B, Liu G, Zhao K, Zhang G. Diversity of extracellular HSP70 in cancer: advancing from a molecular biomarker to a novel therapeutic target. Front Oncol 2024; 14:1388999. [PMID: 38646439 PMCID: PMC11026673 DOI: 10.3389/fonc.2024.1388999] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Heat shock protein 70 (HSP70) is a highly conserved protein functioning as a "molecular chaperone", which is integral to protein folding and maturation. In addition to its high expression within cells upon stressful challenges, HSP70 can be translocated to the cell membrane or released from cells in free form or within extracellular vesicles (EVs). Such trafficking of HSP70 is also present in cancer cells, as HSP70 is overexpressed in various types of patient samples across a range of common malignancies, signifying that extracellular HSP70 (eHSP70) can serve as a tumor biomarker. eHSP70 is involved in a broad range of cancer-related events, including cell proliferation and apoptosis, extracellular matrix (ECM) remodeling, epithelial-mesenchymal transition (EMT), angiogenesis, and immune response. eHSP70 can also induce cancer cell resistance to various treatments, such as chemotherapy, radiotherapy, and anti-programmed death-1 (PD-1) immunotherapy. Though the role of eHSP70 in tumors is contradictory, characterized by both pro-tumor and anti-tumor effects, eHSP70 serves as a promising target in cancer treatment. In this review, we comprehensively summarized the current knowledge about the role of eHSP70 in cancer progression and treatment resistance and discussed the feasibility of eHSP70 as a cancer biomarker and therapeutic target.
Collapse
Affiliation(s)
- Binbin Hu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guihong Liu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kejia Zhao
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Chengdu, Sichuan, China
| | - Gao Zhang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Yazdi M, Hasanzadeh Kafshgari M, Khademi Moghadam F, Zarezade V, Oellinger R, Khosravi M, Haas S, Hoch CC, Pockley AG, Wagner E, Wollenberg B, Multhoff G, Bashiri Dezfouli A. Crosstalk Between NK Cell Receptors and Tumor Membrane Hsp70-Derived Peptide: A Combined Computational and Experimental Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305998. [PMID: 38298098 PMCID: PMC11005703 DOI: 10.1002/advs.202305998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Natural killer (NK) cells are central components of the innate immunity system against cancers. Since tumor cells have evolved a series of mechanisms to escape from NK cells, developing methods for increasing the NK cell antitumor activity is of utmost importance. It is previously shown that an ex vivo stimulation of patient-derived NK cells with interleukin (IL)-2 and Hsp70-derived peptide TKD (TKDNNLLGRFELSG, aa450-461) results in a significant upregulation of activating receptors including CD94 and CD69 which triggers exhausted NK cells to target and kill malignant solid tumors expressing membrane Hsp70 (mHsp70). Considering that TKD binding to an activating receptor is the initial step in the cytolytic signaling cascade of NK cells, herein this interaction is studied by molecular docking and molecular dynamics simulation computational modeling. The in silico results showed a crucial role of the heterodimeric receptor CD94/NKG2A and CD94/NKG2C in the TKD interaction with NK cells. Antibody blocking and CRISPR/Cas9-mediated knockout studies verified the key function of CD94 in the TKD stimulation and activation of NK cells which is characterized by an increased cytotoxic capacity against mHsp70 positive tumor cells via enhanced production and release of lytic granules and pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Mina Yazdi
- Pharmaceutical BiotechnologyDepartment of PharmacyLudwig‐Maximilians‐Universität (LMU)81377MunichGermany
| | - Morteza Hasanzadeh Kafshgari
- Heinz‐Nixdorf‐Chair of Biomedical ElectronicsCampus Klinikum München rechts der IsarTranslaTUMTechnische Universität München81675MunichGermany
| | | | - Vahid Zarezade
- Behbahan Faculty of Medical SciencesBehbahan6361796819Iran
| | - Rupert Oellinger
- Institute of Molecular Oncology and Functional GenomicsSchool of MedicineTechnische Universität München81675MunichGermany
- Central Institute for Translational Cancer Research (TranslaTUM)School of MedicineTechnische Universität München81675MunichGermany
| | - Mohammad Khosravi
- Department of PathobiologyFaculty of Veterinary MedicineShahid Chamran University of AhvazAhvaz6135783151Iran
| | - Stefan Haas
- Department of Radiation OncologySchool of MedicineTechnische Universität München81675MunichGermany
- Department of OtorhinolaryngologySchool of MedicineTechnische Universität München81675MunichGermany
| | - Cosima C. Hoch
- Department of OtorhinolaryngologySchool of MedicineTechnische Universität München81675MunichGermany
| | - Alan Graham Pockley
- John van Geest Cancer Research CentreSchool of Science and TechnologyNottingham Trent UniversityNottinghamNG11 8NSUK
| | - Ernst Wagner
- Pharmaceutical BiotechnologyDepartment of PharmacyLudwig‐Maximilians‐Universität (LMU)81377MunichGermany
| | - Barbara Wollenberg
- Department of OtorhinolaryngologySchool of MedicineTechnische Universität München81675MunichGermany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research (TranslaTUM)School of MedicineTechnische Universität München81675MunichGermany
- Department of Radiation OncologySchool of MedicineTechnische Universität München81675MunichGermany
| | - Ali Bashiri Dezfouli
- Central Institute for Translational Cancer Research (TranslaTUM)School of MedicineTechnische Universität München81675MunichGermany
- Department of Radiation OncologySchool of MedicineTechnische Universität München81675MunichGermany
- Department of OtorhinolaryngologySchool of MedicineTechnische Universität München81675MunichGermany
| |
Collapse
|
6
|
Thomsen AR, Sahlmann J, Bronsert P, Schilling O, Poensgen F, May AM, Timme-Bronsert S, Grosu AL, Vaupel P, Gebbers JO, Multhoff G, Lüchtenborg AM. Protocol of the HISTOTHERM study: assessing the response to hyperthermia and hypofractionated radiotherapy in recurrent breast cancer. Front Oncol 2023; 13:1275222. [PMID: 38169879 PMCID: PMC10759986 DOI: 10.3389/fonc.2023.1275222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Breast cancer is globally the leading cancer in women, and despite the high 5-year survival rate the most frequent cause of cancer related deaths. Surgery, systemic therapy and radiotherapy are the three pillars of curative breast cancer treatment. However, locoregional recurrences frequently occur after initial treatment and are often challenging to treat, amongst others due to high doses of previous radiotherapy treatments. Radiotherapy can be combined with local hyperthermia to sensitize tumor cells to radiation and thereby significantly reduce the required radiation dose. Therefore, the combination treatment of mild local hyperthermia, i.e. locally heating of the tissue to 39-43°C, and re-irradiation with a reduced total dose is a relevant treatment option for previously irradiated patients. The mechanisms of this effect in the course of the therapy are to date not well understood and will be investigated in the HISTOTHERM study. Methods and analyses Patients with local or (loco)regional recurrent breast cancer with macroscopic tumors are included in the study. Local tumor control is evaluated clinically and histologically during the course of a combination treatment of 60 minutes mild superficial hyperthermia (39 - 43°C) using water-filtered infrared A (wIRA) irradiation, immediately followed by hypofractionated re-irradiation with a total dose of 20-24 Gy, administered in weekly doses of 4 Gy. Tumor and tumor stroma biopsies as well as blood samples will be collected prior to treatment, during therapy (at a dose of 12 Gy) and in the follow-up to monitor therapy response. The treatment represents the standard operating procedure for hyperthermia plus re-irradiation. Various tissue and blood-based markers are analyzed. We aim at pinpointing key mechanisms and markers for therapy response which may help guiding treatment decisions in future. In addition, quality of life in the course of treatment will be assessed and survival data will be evaluated. Registration The study is registered at the German Clinical Trials Register, Deutsches Register Klinischer Studien (DRKS00029221).
Collapse
Affiliation(s)
- Andreas R. Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Jörg Sahlmann
- Institute for Medical Biometry and Statistics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felicia Poensgen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Pediatric Department, Black Forest Baar Clinic, Villingen-Schwenningen, Germany
| | - Annette M. May
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Medizinisches Versorgungszentrum Laaff, Freiburg, Germany
| | - Sylvia Timme-Bronsert
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| | - Jan-Olaf Gebbers
- Department of Pathology, Working Group Digital Pathology, University of Berne, Bern, Switzerland
| | - Gabriele Multhoff
- Center for Translational Cancer Research, Klinikum rechts der Isar, Department of Radiation Oncology, Technical University Munich (TUM), Munich, Germany
| | - Anne-Marie Lüchtenborg
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Maher J, Davies DM. CAR-Based Immunotherapy of Solid Tumours-A Survey of the Emerging Targets. Cancers (Basel) 2023; 15:1171. [PMID: 36831514 PMCID: PMC9953954 DOI: 10.3390/cancers15041171] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Immunotherapy with CAR T-cells has revolutionised the treatment of B-cell and plasma cell-derived cancers. However, solid tumours present a much greater challenge for treatment using CAR-engineered immune cells. In a partner review, we have surveyed data generated in clinical trials in which patients with solid tumours that expressed any of 30 discrete targets were treated with CAR-based immunotherapy. That exercise confirms that efficacy of this approach falls well behind that seen in haematological malignancies, while significant toxic events have also been reported. Here, we consider approximately 60 additional candidates for which such clinical data are not available yet, but where pre-clinical data have provided support for their advancement to clinical evaluation as CAR target antigens.
Collapse
Affiliation(s)
- John Maher
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne BN21 2UD, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| | - David M. Davies
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
8
|
Abd El-Fadeal NM, Ellawindy A, Jeraiby MA, Qusti SY, Alshammari EM, Alzahrani AK, Ismail EA, Ehab Z, Toraih EA, Fawzy MS, Mohamed MH. HSP70 Expression Signature in Renal Cell Carcinoma: A Clinical and Bioinformatic Analysis Approach. Genes (Basel) 2023; 14:genes14020355. [PMID: 36833281 PMCID: PMC9957082 DOI: 10.3390/genes14020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Heat shock proteins (HSPs) are cytoprotective against stressful conditions, as in the case of cancer cell metabolism. Scientists proposed that HSP70 might be implicated in increased cancer cell survival. This study aimed to investigate the HSP70 (HSPA4) gene expression signature in patients with renal cell carcinoma (RCC) in correlation to cancer subtype, stage, grade, and recurrence, combining both clinicopathological and in silico analysis approaches. One hundred and thirty archived formalin-fixed paraffin-embedded samples, including 65 RCC tissue specimens and their paired non-cancerous tissues, were included in the study. Total RNA was extracted from each sample and analyzed using TaqMan quantitative Real-Time Polymerase Chain Reaction. Correlation and validation to the available clinicopathological data and results were executed. Upregulated HSP70 (HSPA4) gene expression was evident in RCC compared to non-cancer tissues in the studied cohort and was validated by in silico analysis. Furthermore, HSP70 expression levels showed significant positive correlations with cancer size, grade, and capsule infiltration, as well as recurrence in RCC patients. The expression levels negatively correlated with the overall survival (r = -0.87, p < 0.001). Kaplan-Meier curves showed lower survival rates in high HSP70 expressor group compared to the low expressors. In conclusion, the HSP70 expression levels are associated with poor RCC prognosis in terms of advanced grade, capsule infiltration, recurrence, and short survival.
Collapse
Affiliation(s)
- Noha M. Abd El-Fadeal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Center of Excellence in Molecular and Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Alia Ellawindy
- Medical Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt
| | - Mohammed A. Jeraiby
- Department of Biochemistry, Faculty of Medicine, Jazan University, Jazan 82621, Saudi Arabia
| | - Safaa Y. Qusti
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Eida M. Alshammari
- Department of Chemistry, College of Sciences, University of Ha’il, Ha’il 2440, Saudi Arabia
| | - Ahmad Khuzaim Alzahrani
- Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Ezzat A. Ismail
- Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ziad Ehab
- Faculty of Medicine, Mansoura University, Mansoura 21955, Egypt
| | - Eman A. Toraih
- Medical Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia 41522, Egypt
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Correspondence: (E.A.T.); (M.S.F.)
| | - Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
- Correspondence: (E.A.T.); (M.S.F.)
| | - Marwa Hussein Mohamed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
9
|
Jung S, Jiang L, Zhao J, Shultz LD, Greiner DL, Bae M, Li X, Ordikhani F, Kuai R, Joseph J, Kasinath V, Elmaleh DR, Abdi R. Clathrin light chain-conjugated drug delivery for cancer. Bioeng Transl Med 2023; 8:e10273. [PMID: 36684105 PMCID: PMC9842032 DOI: 10.1002/btm2.10273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/25/2023] Open
Abstract
Targeted drug delivery systems hold the remarkable potential to improve the therapeutic index of anticancer medications markedly. Here, we report a targeted delivery platform for cancer treatment using clathrin light chain (CLC)-conjugated drugs. We conjugated CLC to paclitaxel (PTX) through a glutaric anhydride at high efficiency. Labeled CLCs localized to 4T1 tumors implanted in mice, and conjugation of PTX to CLC enhanced its delivery to these tumors. Treatment of three different mouse models of cancer-melanoma, breast cancer, and lung cancer-with CLC-PTX resulted in significant growth inhibition of both the primary tumor and metastatic lesions, as compared to treatment with free PTX. CLC-PTX treatment caused a marked increase in apoptosis of tumor cells and reduction of tumor angiogenesis. Our data suggested HSP70 as a binding partner for CLC. Our study demonstrates that CLC-based drug-conjugates constitute a novel drug delivery platform that can augment the effects of chemotherapeutics in treating a variety of cancers. Moreover, conjugation of therapeutics with CLC may be used as means by which drugs are delivered specifically to primary tumors and metastatic lesions, thereby prolonging the survival of cancer patients.
Collapse
Affiliation(s)
- Sungwook Jung
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Liwei Jiang
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Institute of Health and Medical TechnologyHefei Institutes of Physical Science, Chinese Academy of SciencesBostonHefeiChina
| | - Jing Zhao
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Dale L. Greiner
- Department of Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Munhyung Bae
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Xiaofei Li
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Farideh Ordikhani
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Rui Kuai
- Center for Nanomedicine and Division of Engineering in Medicine, Department of MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - John Joseph
- Center for Nanomedicine and Division of Engineering in Medicine, Department of MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Vivek Kasinath
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - David R. Elmaleh
- Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Reza Abdi
- Transplantation Research Center, Renal DivisionBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
10
|
Dezfouli AB, Stangl S, Foulds GA, Lennartz P, Pilkington GJ, Pockley AG, Multhoff G. Immunohistochemical, Flow Cytometric, and ELISA-Based Analyses of Intracellular, Membrane-Expressed, and Extracellular Hsp70 as Cancer Biomarkers. Methods Mol Biol 2023; 2693:307-324. [PMID: 37540444 DOI: 10.1007/978-1-0716-3342-7_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
The major stress-inducible 70 kDa heat shock (stress) protein 70 (Hsp70) is frequently overexpressed in highly aggressive tumor cells and thus might serve as a tumor-specific biomarker of aggressive disease and/or therapeutic resistance. We have previously shown that, in contrast to normal cells, tumor cells present Hsp70 on their plasma membrane. In order to elucidate the role of intracellular, membrane-bound and extracellular Hsp70 as a potential tumor biomarker in cancer, herein we describe protocols for the staining of cytosolic Hsp70 in tumor formalin-fixed paraffin-embedded (FFPE) sections from patients with glioblastoma multiforme using immunohistochemistry, for detecting the expression of plasma membrane-bound Hsp70 by a range of cancer-derived cells using multi-parametric flow cytometry using the cmHsp70.1 monoclonal antibody (mAb) and for the measurement of free and vesicular-associated Hsp70 in the circulation of patients with cancer using a unique enzyme-linked immunosorbent assay (ELISA).
Collapse
Affiliation(s)
- Ali Bashiri Dezfouli
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Stefan Stangl
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Philipp Lennartz
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre, School of Pharmacy and Biomedical Sciences, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - A Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Gabriele Multhoff
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.
| |
Collapse
|
11
|
Ghazi B, El Ghanmi A, Kandoussi S, Ghouzlani A, Badou A. CAR T-cells for colorectal cancer immunotherapy: Ready to go? Front Immunol 2022; 13:978195. [PMID: 36458008 PMCID: PMC9705989 DOI: 10.3389/fimmu.2022.978195] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/14/2022] [Indexed: 08/12/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cells represent a new genetically engineered cell-based immunotherapy tool against cancer. The use of CAR T-cells has revolutionized the therapeutic approach for hematological malignancies. Unfortunately, there is a long way to go before this treatment can be developed for solid tumors, including colorectal cancer. CAR T-cell therapy for colorectal cancer is still in its early stages, and clinical data are scarce. Major limitations of this therapy include high toxicity, relapses, and an impermeable tumor microenvironment for CAR T-cell therapy in colorectal cancer. In this review, we summarize current knowledge, highlight challenges, and discuss perspectives regarding CAR T-cell therapy in colorectal cancer.
Collapse
Affiliation(s)
- Bouchra Ghazi
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Adil El Ghanmi
- Mohammed VI International University Hospital, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Sarah Kandoussi
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| |
Collapse
|
12
|
Albakova Z, Mangasarova Y, Sapozhnikov A. Impaired Heat Shock Protein Expression in Activated T Cells in B-Cell Lymphoma. Biomedicines 2022; 10:2747. [PMID: 36359267 PMCID: PMC9687880 DOI: 10.3390/biomedicines10112747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 09/24/2023] Open
Abstract
Heat shock proteins (HSPs) are molecular chaperones that act in a variety of cellular processes, ensuring protein homeostasis and integrity. HSPs play critical roles in the modulation of various immune cells. However, the role of HSPs in T cell activation is largely unknown. We show that HSPs are upregulated following CD3/CD28 stimulation, suggesting that HSP expression might be regulated via TCR. We found that B-cell lymphoma (BCL) patients have dysregulated expression of intracellular and extracellular HSPs, immune checkpoints PD-1, CTLA-4, and STAT3 in CD3/CD28-activated T cells. Consistent with previous findings, we show that HSP90 inhibition downregulated CD4 and CD8 surface markers in healthy controls and BCL patients. HSP90 inhibition alone or in combination with PD-1 or CTLA-4 inhibitors differentially affected CD4+ and CD8+ T cell degranulation responses when stimulated with allogeneic DCs or CD3/CD28 in BCL patients. Additionally, we showed that HSP90 inhibition does not significantly affect intracellular PD-1 and CTLA-4 expression in CD3/CD28-activated T cells. These findings may provide the basis for the discovery of novel immunological targets for the treatment of cancer patients and improve our understanding of HSP functions in immune cells.
Collapse
Affiliation(s)
- Zarema Albakova
- Department of Biology, Lomonosov Moscow State University, Moscow 119192, Russia
- Chokan Limited Liability Partnership (LLP), Almaty 050039, Kazakhstan
| | - Yana Mangasarova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Alexander Sapozhnikov
- Department of Biology, Lomonosov Moscow State University, Moscow 119192, Russia
- Department of Immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
13
|
Ammerata G, Filippo R, Laface C, Memeo R, Solaini L, Cavaliere D, Navarra G, Ranieri G, Currò G, Ammendola M. Hyperthermic intraperitoneal chemotherapy and colorectal cancer: From physiology to surgery. World J Clin Cases 2022; 10:10852-10861. [PMID: 36338235 PMCID: PMC9631165 DOI: 10.12998/wjcc.v10.i30.10852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/23/2022] [Accepted: 08/13/2022] [Indexed: 02/05/2023] Open
Abstract
The pursuit of this paper is to collect principal reviews and systematic reviews about hyperthermic intraperitoneal chemotherapy (HIPEC) and cytoreductive surgery (CRS) used in colorectal cancer (CRC). We focus on principal biological aspects of CRC, hyperthermia effects, and surgical procedures. We searched PubMed/MEDLINE for the principal reviews and systematic reviews published from 2010 to 2021 regarding the bimodal treatment (CRS + HIPEC) against local and advanced CRC. In the literature, from several studies, it seems that the efficacy of bimodal treatment with an accurate CRS can extend overall survival. Despite these studies, there are not still any straight guidelines more detailed and scheduled about the use of combined treatment in patients with CRC. Even if the concept is still not very clear and shared, after a careful evaluation of the published data, and after some technical and pathophysiological descriptions, we concluded that it is possible to improve the overall survival and quality of life and to reduce the tumor relapse in patients affected by locally advanced (pT4) CRC with peritoneal metastases.
Collapse
Affiliation(s)
- Giorgio Ammerata
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Rosalinda Filippo
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Carmelo Laface
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre “Giovanni Paolo II”, Bari 70124, Italy
| | - Riccardo Memeo
- Hepato-Biliary and Pancreatic Surgical Unit, “F. Miulli” Hospital, Acquaviva delle Fonti, Bari 70124, Italy
| | - Leonardo Solaini
- Department of Medical and Surgical Sciences, University of Bologna, Forlì 40126, Italy
| | - Davide Cavaliere
- Department of General and Oncologic Surgery, Morgagni-Pierantoni Hospital, Forlì 47121, Italy
| | - Giuseppe Navarra
- Department of Human Pathology of Adult and Evolutive Age, Surgical Oncology Division, “G. Martino” Hospital, University of Messina, Messina 98122, Italy
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre “Giovanni Paolo II”, Bari 70124, Italy
| | - Giuseppe Currò
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Michele Ammendola
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| |
Collapse
|
14
|
Javid H, Hashemian P, Yazdani S, Sharbaf Mashhad A, Karimi-Shahri M. The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem 2022; 123:1704-1735. [PMID: 36063530 DOI: 10.1002/jcb.30326] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023]
Abstract
Heat shock proteins (HSPs) are a large molecular chaperone family classified by their molecular weights, including HSP27, HSP40, HSP60, HSP70, HSP90, and HSP110. HSPs are likely to have antiapoptotic properties and participate actively in various processes such as tumor cell proliferation, invasion, metastases, and death. In this review, we discuss comprehensively the functions of HSPs associated with the progression of colorectal cancer (CRC) and metastasis and resistance to cancer therapy. Taken together, HSPs have numerous clinical applications as biomarkers for cancer diagnosis and prognosis and potential therapeutic targets for CRC and its related metastases.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Pedram Hashemian
- Jahad Daneshgahi Research Committee, Jahad Daneshgahi Institute, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Laboratory Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
15
|
Hu C, Yang J, Qi Z, Wu H, Wang B, Zou F, Mei H, Liu J, Wang W, Liu Q. Heat shock proteins: Biological functions, pathological roles, and therapeutic opportunities. MedComm (Beijing) 2022; 3:e161. [PMID: 35928554 PMCID: PMC9345296 DOI: 10.1002/mco2.161] [Citation(s) in RCA: 235] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
The heat shock proteins (HSPs) are ubiquitous and conserved protein families in both prokaryotic and eukaryotic organisms, and they maintain cellular proteostasis and protect cells from stresses. HSP protein families are classified based on their molecular weights, mainly including large HSPs, HSP90, HSP70, HSP60, HSP40, and small HSPs. They function as molecular chaperons in cells and work as an integrated network, participating in the folding of newly synthesized polypeptides, refolding metastable proteins, protein complex assembly, dissociating protein aggregate dissociation, and the degradation of misfolded proteins. In addition to their chaperone functions, they also play important roles in cell signaling transduction, cell cycle, and apoptosis regulation. Therefore, malfunction of HSPs is related with many diseases, including cancers, neurodegeneration, and other diseases. In this review, we describe the current understandings about the molecular mechanisms of the major HSP families including HSP90/HSP70/HSP60/HSP110 and small HSPs, how the HSPs keep the protein proteostasis and response to stresses, and we also discuss their roles in diseases and the recent exploration of HSP related therapy and diagnosis to modulate diseases. These research advances offer new prospects of HSPs as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Chen Hu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Jing Yang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Ziping Qi
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Hong Wu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Fengming Zou
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
- Precision Medicine Research Laboratory of Anhui ProvinceHefeiAnhuiP. R. China
| |
Collapse
|
16
|
Bashiri Dezfouli A, Yazdi M, Benmebarek MR, Schwab M, Michaelides S, Miccichè A, Geerts D, Stangl S, Klapproth S, Wagner E, Kobold S, Multhoff G. CAR T Cells Targeting Membrane-Bound Hsp70 on Tumor Cells Mimic Hsp70-Primed NK Cells. Front Immunol 2022; 13:883694. [PMID: 35720311 PMCID: PMC9198541 DOI: 10.3389/fimmu.2022.883694] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022] Open
Abstract
Strategies to boost anti-tumor immunity are urgently needed to treat therapy-resistant late-stage cancers, including colorectal cancers (CRCs). Cytokine stimulation and genetic modifications with chimeric antigen receptors (CAR) represent promising strategies to more specifically redirect anti-tumor activities of effector cells like natural killer (NK) and T cells. However, these approaches are critically dependent on tumor-specific antigens while circumventing the suppressive power of the solid tumor microenvironment and avoiding off-tumor toxicities. Previously, we have shown that the stress-inducible heat shock protein 70 (Hsp70) is frequently and specifically expressed on the cell surface of many different, highly aggressive tumors but not normal tissues. We could take advantage of tumors expressing Hsp70 on their membrane (‘mHsp70’) to attract and engage NK cells after in vitro stimulation with the 14-mer Hsp70 peptide TKDNNLLGRFELSG (TKD) plus low dose interleukin (IL)-2. However, a potential limitation of activated primary NK cells after adoptive transfer is their comparably short life span. T cells are typically long-lived but do not recognize mHsp70 on tumor cells, even after stimulation with TKD/IL-2. To combine the advantages of mHsp70-specificity with longevity, we constructed a CAR having specificity for mHsp70 and retrovirally transduced it into primary T cells. Co-culture of anti-Hsp70 CAR-transduced T cells with mHsp70-positive tumor cells stimulates their functional responsiveness. Herein, we demonstrated that human CRCs with a high mHsp70 expression similarly attract TKD/IL-2 stimulated NK cells and anti-Hsp70 CAR T cells, triggering the release of their lytic effector protein granzyme B (GrB) and the pro-inflammatory cytokine interferon (IFN)-γ, after 4 and 24 hours, respectively. In sum, stimulated NK cells and anti-Hsp70 CAR T cells demonstrated comparable anti-tumor effects, albeit with somewhat differing kinetics. These findings, together with the fact that mHsp70 is expressed on a large variety of different cancer entities, highlight the potential of TKD/IL-2 pre-stimulated NK, as well as anti-Hsp70 CAR T cells to provide a promising direction in the field of targeted, cell-based immunotherapies which can address significant unmet clinical needs in a wide range of cancer settings.
Collapse
Affiliation(s)
- Ali Bashiri Dezfouli
- Central Institute for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Munich, Germany
| | - Melissa Schwab
- Central Institute for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| | - Stefanos Michaelides
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Munich, Germany
| | | | | | - Stefan Stangl
- Central Institute for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany.,Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sarah Klapproth
- Institute of Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research Deutsches Zentrum für Lungenforschung (DZL), Munich, Germany.,German Center for Translational Cancer Research Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research Technische Universität München (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
17
|
Forcing the Antitumor Effects of HSPs Using a Modulated Electric Field. Cells 2022; 11:cells11111838. [PMID: 35681533 PMCID: PMC9180583 DOI: 10.3390/cells11111838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/10/2022] Open
Abstract
The role of Heat Shock Proteins (HSPs) is a “double-edged sword” with regards to tumors. The location and interactions of HSPs determine their pro- or antitumor activity. The present review includes an overview of the relevant functions of HSPs, which could improve their antitumor activity. Promoting the antitumor processes could assist in the local and systemic management of cancer. We explore the possibility of achieving this by manipulating the electromagnetic interactions within the tumor microenvironment. An appropriate electric field may select and affect the cancer cells using the electric heterogeneity of the tumor tissue. This review describes the method proposed to effect such changes: amplitude-modulated radiofrequency (amRF) applied with a 13.56 MHz carrier frequency. We summarize the preclinical investigations of the amRF on the HSPs in malignant cells. The preclinical studies show the promotion of the expression of HSP70 on the plasma membrane, participating in the immunogenic cell death (ICD) pathway. The sequence of guided molecular changes triggers innate and adaptive immune reactions. The amRF promotes the secretion of HSP70 also in the extracellular matrix. The extracellular HSP70 accompanied by free HMGB1 and membrane-expressed calreticulin (CRT) form damage-associated molecular patterns encouraging the dendritic cells’ maturing for antigen presentation. The process promotes killer T-cells. Clinical results demonstrate the potential of this immune process to trigger a systemic effect. We conclude that the properly applied amRF promotes antitumor HSP activity, and in situ, it could support the tumor-specific immune effects produced locally but acting systemically for disseminated cells and metastatic lesions.
Collapse
|
18
|
Albakova Z, Mangasarova Y. The HSP Immune Network in Cancer. Front Immunol 2021; 12:796493. [PMID: 34917098 PMCID: PMC8669653 DOI: 10.3389/fimmu.2021.796493] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Heat shock proteins are molecular chaperones which support tumor development by regulating various cellular processes including unfolded protein response, mitochondrial bioenergetics, apoptosis, autophagy, necroptosis, lipid metabolism, angiogenesis, cancer cell stemness, epithelial-mesenchymal transition and tumor immunity. Apart from their intracellular activities, HSPs have also distinct extracellular functions. However, the role that HSP chaperones play in the regulation of immune responses inside and outside the cell is not yet clear. Herein, we explore the intracellular and extracellular immunologic functions of HSPs in cancer. A broader understanding of how HSPs modulate immune responses may provide critical insights for the development of effective immunotherapies.
Collapse
Affiliation(s)
- Zarema Albakova
- Department of Immunology, Lomonosov Moscow State University, Moscow, Russia
| | | |
Collapse
|
19
|
Wang X, Xie L, Zhu L. Clinicopathological significance of HSP70 expression in gastric cancer: a systematic review and meta-analysis. BMC Gastroenterol 2021; 21:437. [PMID: 34809574 PMCID: PMC8609754 DOI: 10.1186/s12876-021-01990-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background Heat shock protein 70 (HSP70) has been associated with the clinicopathological characteristics and prognosis of many cancers types, implying that it is a potential cancer biomarker. However, no consensus has been reached regarding its clinicopathological and prognostic significance in patients with gastric cancer. To address this gap, we performed a systematic review and meta-analysis. Methods We searched PubMed, Embase, and the Cochrane Library for full-text literature according to the eligibility criteria. We used the odds ratio and hazard ratio as the suitable parameters to evaluate the clinicopathological and prognostic significance of HSP70. The statistical analysis was performed using STATA 15.0. Results After inclusion and exclusion of studies based on the eligibility criteria, data of 1,307 patients with gastric cancer from 9 studies were finally included. The pooled outcomes implied that HSP70 expression was significantly correlated with higher differentiation degrees, intestinal gastric cancer, and lymphovascular invasion but not with age, gender, depth of invasion, Helicobacter pylori infection, lymph node invasion, TNM stages, and metastasis. The pooled HR showed no significant correlation between HSP70 expression and overall survival of gastric cancer patients. Conclusions Our meta-analysis showed that HSP70 plays a complicated role in the development of gastric cancer. It may be directly engaged in tumour differentiation and distant invasion but cannot be considered a biomarker for predicting the prognosis of gastric cancer.
Collapse
Affiliation(s)
- Xiaolu Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Li Xie
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lijing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
20
|
The plant diterpene epoxysiderol targets Hsp70 in cancer cells, affecting its ATPase activity and reducing its translocation to plasma membrane. Int J Biol Macromol 2021; 189:262-270. [PMID: 34437915 DOI: 10.1016/j.ijbiomac.2021.08.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 11/23/2022]
Abstract
The ATP-dependent molecular chaperone Hsp70 is over-expressed in cancer cells where it plays pivotal roles in stabilization of onco-proteins, promoting cell proliferation and protecting cells from apoptosis and necrosis. Moreover, a relationship between the ability of cancer cells to migrate and the abundance of membrane-associated Hsp70 was shown. However, although Hsp70 is a promising target for cancer therapy, there is a still unsatisfied requirement of inhibitors possibly blocking its cancer-associated activities. Moving from the evidence that the plant diterpene oridonin efficiently targets Hsp70 1A in cancer cells, we set up a small kaurane diterpenoids collection and subjected it to a Surface Plasmon Resonance-screening, to identify new putative inhibitors of this chaperone. The results obtained suggested epoxysiderol as an effective Hsp70 1A interactor; therefore, using a combination of bioanalytical, biochemical and bioinformatics approaches, this compound was shown to bind the nucleotide-binding-domain of the chaperone, thus affecting its ATPase activity. The interaction between epoxysiderol and Hsp70 1A was also demonstrated to actually occur inside cancer cells, significantly reduced the translocation of the chaperone to the cell membrane, thus suggesting a possible role of epoxysiderol as an anti-metastasis agent.
Collapse
|
21
|
Heat Shock Proteins 27, 70, and 110: Expression and Prognostic Significance in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13174407. [PMID: 34503216 PMCID: PMC8431468 DOI: 10.3390/cancers13174407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/26/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Heat shock proteins (HSPs) are evolutionarily conserved chaperones occurring in virtually all living organisms playing a key role in the maintenance of cellular homeostasis. They are constitutively expressed to prevent and repair protein damage following various physiological and environmental stressors. HSPs are overexpressed in various types of cancers to provide cytoprotective function, and they have been described to influence prognosis and response to therapy. Moreover, they have been used as a tumor marker in blood serum biochemistry and they represent a potentially promising therapeutic target. To clarify prognostic significance of two canonical HSPs (27 and 70) and less known HSP110 (previously known as HSP105) in colorectal carcinoma (CRC), we retrospectively performed HSP immunohistochemistry on tissue microarrays from formalin-fixed paraffin-embedded tumor tissue from 297 patients with known follow-up. Survival analysis (univariate Kaplan-Meier analysis with the log-rank test and multivariate Cox regression) revealed significantly shorter overall survival (OS, mean 5.54 vs. 7.07, p = 0.033) and borderline insignificantly shorter cancer specific survival (CSS, mean 6.3 vs. 7.87 years, p = 0.066) in patients with HSP70+ tumors. In the case of HSP27+ tumors, there was an insignificantly shorter OS (mean 6.36 vs. 7.13 years, p = 0.2) and CSS (mean 7.17 vs. 7.95 years, p = 0.2). HSP110 showed no significant impact on survival. Using Pearson's chi-squared test, there was a significant association of HSP27 and HSP70 expression with advanced cancer stage. HSP27+ tumors were more frequently mismatch-repair proficient and vice versa (p = 0.014), and they occurred more often in female patients and vice versa (p = 0.015). There was an enrichment of left sided tumors with HSP110+ compared to the right sided (p = 0.022). In multivariate Cox regression adjusted on the UICC stage, grade and right/left side; both HSPs 27 and 70 were not independent survival predictors (p = 0.616 & p = 0.586). In multivariate analysis, only advanced UICC stage (p = 0) and right sided localization (p = 0.04) were independent predictors of worse CSS. In conclusion, from all three HSPs examined in our study, only HSP70 expression worsened CRC prognosis, although stage-dependent. The contribution of this article may be seen as a large survival analysis of HSPs 27 and 70 and the largest analysis of HSP110 described in CRC.
Collapse
|
22
|
Hsp70 in Liquid Biopsies-A Tumor-Specific Biomarker for Detection and Response Monitoring in Cancer. Cancers (Basel) 2021; 13:cancers13153706. [PMID: 34359606 PMCID: PMC8345117 DOI: 10.3390/cancers13153706] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
In contrast to normal cells, tumor cells of multiple entities overexpress the Heat shock protein 70 (Hsp70) not only in the cytosol, but also present it on their plasma membrane in a tumor-specific manner. Furthermore, membrane Hsp70-positive tumor cells actively release Hsp70 in small extracellular vesicles with biophysical characteristics of exosomes. Due to conformational changes of Hsp70 in a lipid environment, most commercially available antibodies fail to detect membrane-bound and vesicular Hsp70. To fill this gap and to assess the role of vesicular Hsp70 in circulation as a potential tumor biomarker, we established the novel complete (comp)Hsp70 sandwich ELISA, using two monoclonal antibodies (mAbs), that is able to recognize both free and lipid-associated Hsp70 on the cell surface of viable tumor cells and on small extracellular vesicles. The epitopes of the mAbs cmHsp70.1 (aa 451-461) and cmHsp70.2 (aa 614-623) that are conserved among different species reside in the substrate-binding domain of Hsp70 with measured affinities of 0.42 nM and 0.44 nM, respectively. Validation of the compHsp70 ELISA revealed a high intra- and inter-assay precision, linearity in a concentration range of 1.56 to 25 ng/mL, high recovery rates of spiked liposomal Hsp70 (>84%), comparable values between human serum and plasma samples and no interference by food intake or age of the donors. Hsp70 concentrations in the circulation of patients with glioblastoma, squamous cell or adeno non-small cell lung carcinoma (NSCLC) at diagnosis were significantly higher than those of healthy donors. Hsp70 concentrations dropped concomitantly with a decrease in viable tumor mass upon irradiation of patients with approximately 20 Gy (range 18-22.5 Gy) and after completion of radiotherapy (60-70 Gy). In summary, the compHsp70 ELISA presented herein provides a sensitive and reliable tool for measuring free and vesicular Hsp70 in liquid biopsies of tumor patients, levels of which can be used as a tumor-specific biomarker, for risk assessment (i.e., differentiation of grade III vs. IV adeno NSCLC) and monitoring of therapeutic outcomes.
Collapse
|
23
|
Albakova Z, Siam MKS, Sacitharan PK, Ziganshin RH, Ryazantsev DY, Sapozhnikov AM. Extracellular heat shock proteins and cancer: New perspectives. Transl Oncol 2020; 14:100995. [PMID: 33338880 PMCID: PMC7749402 DOI: 10.1016/j.tranon.2020.100995] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
High expression of extracellular heat shock proteins (HSPs) indicates highly aggressive tumors. HSP profiling of extracellular vesicles (EVs) derived from various biological fluids and released by immune cells may open new perspectives for an identification of diagnostic, prognostic and predictive biomarkers of cancer. Identification of specific microRNAs targeting HSPs in EVs may be a promising strategy for the discovery of novel biomarkers of cancer.
Heat shock proteins (HSPs) are a large family of molecular chaperones aberrantly expressed in cancer. The expression of HSPs in tumor cells has been shown to be implicated in the regulation of apoptosis, immune responses, angiogenesis and metastasis. Given that extracellular vesicles (EVs) can serve as potential source for the discovery of clinically useful biomarkers and therapeutic targets, it is of particular interest to study proteomic profiling of HSPs in EVs derived from various biological fluids of cancer patients. Furthermore, a divergent expression of circulating microRNAs (miRNAs) in patient samples has opened new opportunities in exploiting miRNAs as diagnostic tools. Herein, we address the current literature on the expression of extracellular HSPs with particular interest in HSPs in EVs derived from various biological fluids of cancer patients and different types of immune cells as promising targets for identification of clinical biomarkers of cancer. We also discuss the emerging role of miRNAs in HSP regulation for the discovery of blood-based biomarkers of cancer. We outline the importance of understanding relationships between various HSP networks and co-chaperones and propose the model for identification of HSP signatures in cancer. Elucidating the role of HSPs in EVs from the proteomic and miRNAs perspectives may provide new opportunities for the discovery of novel biomarkers of cancer.
Collapse
Affiliation(s)
- Zarema Albakova
- Department of Biology, Lomonosov Moscow State University, 199192 Moscow, Russia.
| | | | - Pradeep Kumar Sacitharan
- The Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom; Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Rustam H Ziganshin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Dmitriy Y Ryazantsev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexander M Sapozhnikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
24
|
Vostakolaei MA, Hatami-Baroogh L, Babaei G, Molavi O, Kordi S, Abdolalizadeh J. Hsp70 in cancer: A double agent in the battle between survival and death. J Cell Physiol 2020; 236:3420-3444. [PMID: 33169384 DOI: 10.1002/jcp.30132] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/23/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
The heat shock protein (Hsps) superfamily, also known as molecular chaperones, are highly conserved and present in all living organisms and play vital roles in protein fate. The HspA1A (Hsp70-1), called Hsp70 in this review, is expressed at low or undetectable levels in most unstressed normal cells, but numerous studies have shown that diverse types of tumor cells express Hsp70 at the plasma membrane that leads to resistance to programmed cell death and tumor progression. Hsp70 is released into the extracellular milieu in three forms including free soluble, complexed with cancer antigenic peptides, and exosome forms. Therefore, it seems to be a promising therapeutic target in human malignancies. However, a great number of studies have indicated that both intracellular and extracellular Hsp70 have a dual function. A line of evidence presented that intracellular Hsp70 has a cytoprotective function via suppression of apoptosis and lysosomal cell death (LCD) as well as that extracellular Hsp70 can promote tumorigenesis and angiogenesis. Other evidence showed intracellular Hsp70 can promote apoptosis and membrane-associated/extracellular Hsp70 can elicit antitumor innate and adaptive immune responses. Given the contradictory functions, as a "double agent," could Hsp70 be a promising tool in the future of targeted cancer therapies? To answer this question, in this review, we will discuss the functions of Hsp70 in cancers besides inhibition and stimulation strategies for targeting Hsp70 along with their challenges.
Collapse
Affiliation(s)
- Mehdi A Vostakolaei
- Digestive Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Hatami-Baroogh
- Department of Reproduction and Development, Royan Institute for Animal Biotechnology, ACER, Isfahan, Iran
| | - Ghader Babaei
- Department of Biochemistry, Urmia University Medical Sciences, Urmia, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirafkan Kordi
- Antimicrobial Resistance Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Membrane-Associated Heat Shock Proteins in Oncology: From Basic Research to New Theranostic Targets. Cells 2020; 9:cells9051263. [PMID: 32443761 PMCID: PMC7290778 DOI: 10.3390/cells9051263] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Heat shock proteins (HSPs) constitute a large family of conserved proteins acting as molecular chaperones that play a key role in intracellular protein homeostasis, regulation of apoptosis, and protection from various stress factors (including hypoxia, thermal stress, oxidative stress). Apart from their intracellular localization, members of different HSP families such as small HSPs, HSP40, HSP60, HSP70 and HSP90 have been found to be localized on the plasma membrane of malignantly transformed cells. In the current article, the role of membrane-associated molecular chaperones in normal and tumor cells is comprehensively reviewed with implications of these proteins as plausible targets for cancer therapy and diagnostics.
Collapse
|
26
|
Labavić D, Ladjimi MT, Courtade E, Pfeuty B, Thommen Q. Mammalian cell sensitivity to hyperthermia in various cell lines: a new universal and predictive description. Int J Hyperthermia 2020; 37:506-516. [DOI: 10.1080/02656736.2020.1762005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- D. Labavić
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - M. T. Ladjimi
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - E. Courtade
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - B. Pfeuty
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - Q. Thommen
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| |
Collapse
|
27
|
Albakova Z, Armeev GA, Kanevskiy LM, Kovalenko EI, Sapozhnikov AM. HSP70 Multi-Functionality in Cancer. Cells 2020; 9:cells9030587. [PMID: 32121660 PMCID: PMC7140411 DOI: 10.3390/cells9030587] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/20/2022] Open
Abstract
The 70-kDa heat shock proteins (HSP70s) are abundantly present in cancer, providing malignant cells selective advantage by suppressing multiple apoptotic pathways, regulating necrosis, bypassing cellular senescence program, interfering with tumor immunity, promoting angiogenesis and supporting metastasis. This direct involvement of HSP70 in most of the cancer hallmarks explains the phenomenon of cancer "addiction" to HSP70, tightly linking tumor survival and growth to the HSP70 expression. HSP70 operates in different states through its catalytic cycle, suggesting that it can multi-function in malignant cells in any of these states. Clinically, tumor cells intensively release HSP70 in extracellular microenvironment, resulting in diverse outcomes for patient survival. Given its clinical significance, small molecule inhibitors were developed to target different sites of the HSP70 machinery. Furthermore, several HSP70-based immunotherapy approaches were assessed in clinical trials. This review will explore different roles of HSP70 on cancer progression and emphasize the importance of understanding the flexibility of HSP70 nature for future development of anti-cancer therapies.
Collapse
Affiliation(s)
- Zarema Albakova
- Department of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia; (G.A.A.); (A.M.S.)
- Department of Immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.M.K.); (E.I.K.)
- Correspondence:
| | - Grigoriy A. Armeev
- Department of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia; (G.A.A.); (A.M.S.)
| | - Leonid M. Kanevskiy
- Department of Immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.M.K.); (E.I.K.)
| | - Elena I. Kovalenko
- Department of Immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.M.K.); (E.I.K.)
| | - Alexander M. Sapozhnikov
- Department of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia; (G.A.A.); (A.M.S.)
- Department of Immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.M.K.); (E.I.K.)
| |
Collapse
|
28
|
Multhoff G, Vaupel P. Hypoxia Compromises Anti-Cancer Immune Responses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1232:131-143. [PMID: 31893404 DOI: 10.1007/978-3-030-34461-0_18] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hypoxia, one of the hallmarks of cancer, is caused by an insufficient oxygen supply, mostly due to a chaotic, deficient tumor microcirculation. Apart from a hypoxia-mediated resistance to standard therapies, modulated gene and protein expression, genetic instability and malignant progression, hypoxia also plays a pivotal role in anti-cancer immune responses by (a) reducing survival, cytolytic and migratory activity of effector cells such as CD4+ cells, CD8+ cytotoxic T cells, natural killer-like T cells and natural killer cells, (b) reducing the production and release of effector cytokines, (c) supporting immunosuppressive cells such as regulatory T cells, myeloid-derived suppressor cells and M2 macrophages, (d) increasing the production and release of immunosuppressive cytokines, and (e) inducing the expression of immune checkpoint inhibitors. In this minireview, immunosuppressive effects of hypoxia- and HIF-1a-driven traits in cancers are described.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Center for Translational Cancer Research (TranslaTUM), Radiation Immuno-Oncology Group, Klinikum rechts der Isar, TU Munich (TUM), München, Germany.
| | - Peter Vaupel
- Department of Radiation Oncology, Klinikum rechts der Isar, TU München (TUM), München, Germany
| |
Collapse
|
29
|
De Andrade WP, Da Conceição Braga L, Gonçales NG, Silva LM, Da Silva Filho AL. HSPA1A, HSPA1L and TRAP1 heat shock genes may be associated with prognosis in ovarian epithelial cancer. Oncol Lett 2019; 19:359-367. [PMID: 31897148 PMCID: PMC6923843 DOI: 10.3892/ol.2019.11095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/12/2019] [Indexed: 12/24/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, with the presence of chemoresistance contributing to the poor prognosis. Heat Shock Proteins (HSPs) genes are activated in response to pathophysiological stress and serve a role in a variety of stages in carcinogenesis, acting primarily as anti-apoptotic agents and in chemotherapy resistance in a variety of tumor types. The current study evaluated the HSP gene expression profile in women with ovarian cancer (OC) and their correlation with clinical and pathological aspects of patients with OC. A total of 51 patients included in the current study were divided into four groups: Primary Epithelial Ovarian Cancer (EOC; n=14), metastatic EOC (n=11), ovarian serous cystadenoma (n=7) and no evidence of ovarian malignancy or control groups (n=19). RNA extraction and reverse transcription-quantitative (RT-q) PCR was then performed on the samples obtained. RT-qPCR was performed to compare TNF receptor associated protein 1 (TRAP1), heat shock protein family (HSP) HSPB1, HSPD1, HSPA1A and HSPA1L expression in primary and metastatic EOCs. TRAP1, HSPB1, HSPD1, HSPA1A and HSPA1L gene expression did not differ among groups. HSPA1A, HSPA1L and TRAP1 were revealed to be underexpressed in the primary and metastatic EOC groups, with HSPA1L exhibiting the lowest expression. TRAP1 expression was higher in tumors at stages I/II compared with those at stages III/IV. No correlation was exhibited between HSP expression and age, menarche, menopause, parity, period after menopause initiation, cytoreduction, CA-125 or overall and disease-free survival. HSPA1A was negatively correlated with the risk of mortality from OC. The results indicated that the downregulation of HSPA1A, HSPA1L and TRAP1 could be associated with the clinical prognostic features of women with EOC.
Collapse
Affiliation(s)
- Warne Pedro De Andrade
- Oncology Service, Vera Cruz Hospital, Belo Horizonte, Minas Gerais 30180-090, Brazil.,Department of Obstetrics and Gynecology, School of Medicine, São Paulo State University, Botucatu, Sao Paulo 18618-687, Brazil
| | - Letícia Da Conceição Braga
- Department of Obstetrics and Gynecology, School of Medicine, São Paulo State University, Botucatu, Sao Paulo 18618-687, Brazil.,Cellular Biology Service, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Nikole Gontijo Gonçales
- Cellular Biology Service, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Luciana Maria Silva
- Cellular Biology Service, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Agnaldo Lopes Da Silva Filho
- Department of Obstetrics and Gynecology, School of Medicine, São Paulo State University, Botucatu, Sao Paulo 18618-687, Brazil.,Department of Obstetrics and Gynecology, School of Medicine, Minas Gerais Federal University, Belo Horizonte, Minas Gerais 30130-100, Brazil
| |
Collapse
|
30
|
Vostakolaei MA, Molavi O, Hejazi MS, Kordi S, Rahmati S, Barzegari A, Abdolalizadeh J. Isolation and characterization of a novel scFv antibody fragments specific for Hsp70 as a tumor biomarker. J Cell Biochem 2019; 120:14711-14724. [DOI: 10.1002/jcb.28732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/12/2019] [Accepted: 03/22/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Mehdi Asghari Vostakolaei
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
- Biotechnology Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Saeid Hejazi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
| | - Shirafkan Kordi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Saman Rahmati
- Medical Biotechnology Department Biotechnology Research Center, Pasteur Institute of Iran Tehran Iran
| | - Abolfazl Barzegari
- Research Centre for Pharmaceutical Nanotechnology Tabriz University of Medical Sciences Tabriz Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
- Paramedical Faculty Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
31
|
Gráf L, Barabás L, Madaras B, Garam N, Maláti É, Horváth L, Prohászka Z, Horváth Z, Kocsis J. High serum Hsp70 level predicts poor survival in colorectal cancer: Results obtained in an independent validation cohort. Cancer Biomark 2019; 23:539-547. [PMID: 30452400 DOI: 10.3233/cbm-181683] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Hsp70 plays important role in the development and progression of cancer. Previously we described the association between serum Hsp70 levels and mortality of colorectal cancer. OBJECTIVE In this new prospective study we aimed to confirm and extend our previous findings in a larger cohort of patients, based on a longer follow-up period. METHODS Two hundred and thirty-two patients diagnosed with colorectal cancer were enrolled in the study. Baseline serum Hsp70 level and classical biomarker levels were measured. Patients were treated according to stage of the tumor and follow-up lasted for a median 46.4 months. RESULTS We found that serum Hsp70 concentrations increase significantly with stage of the disease (1.79; 2.23 and 3.21 ng/ml in stage I+II, III and IV respectively, p= 0.012 and 0.002, Mann-Whitney test) and with other known biomarkers of the disease. We managed to confirm our previous findings that high baseline serum Hsp70 level (> 1.64 ng/ml) predicted poor 5-year survival (risk of death HR: 1.94 CI: 1.294-2.909; univariate; HR: 2.418 CI: 1.373-4.258; multivariate Cox regression analysis) in the whole patient population and also in subgroups of stage IV and stage III disease. The strongest association was observed in women under age of 70 (HR: 8.12, CI: 2.02-35.84; p= 0.004; multivariate Cox regression). The power of this colorectal cancer prognostic model could be amplified by combining Hsp70 levels and inflammatory markers. Patients with high Hsp70, CRP and high baseline WBC or platelet count had 5-times higher risk of death (HR: 5.07 CI: 2.74-9.39, p< 0.0001; and HR: 4.98 CI: 3.08-8.06, p< 0.0001 respectively). CONCLUSIONS These results confirm and validate our previous findings that serum Hsp70 is a useful biomarker of colorectal cancer.
Collapse
Affiliation(s)
- László Gráf
- 3rd Department of Internal Medicine, Semmelweis University, Budapest 1125, Hungary
| | - Lóránd Barabás
- 2nd Department of Surgery, Semmelweis University, Budapest 1125, Hungary
| | | | - Nóra Garam
- 3rd Department of Internal Medicine, Semmelweis University, Budapest 1125, Hungary
| | - Éva Maláti
- János Balassa Hospital, County Hospital Tolna, Szekszárd 7100, Hungary
| | - Laura Horváth
- 3rd Department of Internal Medicine, Semmelweis University, Budapest 1125, Hungary
| | - Zoltán Prohászka
- 3rd Department of Internal Medicine, Semmelweis University, Budapest 1125, Hungary
| | - Zsolt Horváth
- Department of Oncoradiology, Bács-Kiskun County Hospital, Kecskemét H6000, Hungary
| | - Judit Kocsis
- 3rd Department of Internal Medicine, Semmelweis University, Budapest 1125, Hungary.,Department of Oncoradiology, Bács-Kiskun County Hospital, Kecskemét H6000, Hungary
| |
Collapse
|
32
|
Shevtsov M, Stangl S, Nikolaev B, Yakovleva L, Marchenko Y, Tagaeva R, Sievert W, Pitkin E, Mazur A, Tolstoy P, Galibin O, Ryzhov V, Steiger K, Smirnov O, Khachatryan W, Chester K, Multhoff G. Granzyme B Functionalized Nanoparticles Targeting Membrane Hsp70-Positive Tumors for Multimodal Cancer Theranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900205. [PMID: 30828968 DOI: 10.1002/smll.201900205] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/11/2019] [Indexed: 05/20/2023]
Abstract
Functionalized superparamagnetic iron oxide nanoparticles (SPIONs) have emerged as potential clinical tools for cancer theranostics. Membrane-bound 70 kDa heat shock protein (mHsp70) is ubiquitously expressed on the cell membrane of various tumor types but not normal cells and therefore provides a tumor-specific target. The serine protease granzyme B (GrB) that is produced as an effector molecule by activated T and NK cells has been shown to specifically target mHsp70 on tumor cells. Following binding to Hsp70, GrB is rapidly internalized into tumor cells. Herein, it is demonstrated that GrB functionalized SPIONs act as a contrast enhancement agent for magnetic resonance imaging and induce specific tumor cell apoptosis. Combinatorial regimens employing stereotactic radiotherapy and/or magnetic targeting are found to further enhance the therapeutic efficacy of GrB-SPIONs in different tumor mouse models.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Radiation Immuno-Oncology group, Klinikum rechts der Isar, Einsteinstr. 25, 81675, Munich, Germany
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky ave., 4, St. Petersburg, 194064, Russia
- First Pavlov State Medical University of St. Petersburg, L'va Tolstogo str. 6/8, St. Petersburg, 197022, Russia
- Almazov National Medical Research Centre, Russian Polenov Neurosurgical Institute, Mayakovskogo str. 12, St. Petersburg, 191104, Russia
| | - Stefan Stangl
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Radiation Immuno-Oncology group, Klinikum rechts der Isar, Einsteinstr. 25, 81675, Munich, Germany
| | - Boris Nikolaev
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, St. Petersburg, 191014, Russia
| | - Ludmila Yakovleva
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, St. Petersburg, 191014, Russia
| | - Yaroslav Marchenko
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, St. Petersburg, 191014, Russia
| | - Ruslana Tagaeva
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, St. Petersburg, 191014, Russia
| | - Wolfgang Sievert
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Radiation Immuno-Oncology group, Klinikum rechts der Isar, Einsteinstr. 25, 81675, Munich, Germany
| | - Emil Pitkin
- Wharton School, University of Pennsylvania, Walnut Street 3730, Philadelphia, PA, 19104, USA
| | - Anton Mazur
- Saint Petersburg State University, Universitetskaya nab. 7-9, St. Petersburg, 199034, Russia
| | - Peter Tolstoy
- Saint Petersburg State University, Universitetskaya nab. 7-9, St. Petersburg, 199034, Russia
| | - Oleg Galibin
- First Pavlov State Medical University of St. Petersburg, L'va Tolstogo str. 6/8, St. Petersburg, 197022, Russia
| | - Vyacheslav Ryzhov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - Katja Steiger
- Institute of Pathology, Technische Universität München, Trogerstr. 18, 81675, Munich, Germany
| | - Oleg Smirnov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - William Khachatryan
- Almazov National Medical Research Centre, Russian Polenov Neurosurgical Institute, Mayakovskogo str. 12, St. Petersburg, 191104, Russia
| | - Kerry Chester
- UCL Cancer Institute, University College London, 72 Huntley Street, WC1E 6DD, London, UK
| | - Gabriele Multhoff
- Center for Translational Cancer Research Technische Universität München (TranslaTUM), Radiation Immuno-Oncology group, Klinikum rechts der Isar, Einsteinstr. 25, 81675, Munich, Germany
| |
Collapse
|
33
|
Breuninger S, Stangl S, Werner C, Sievert W, Lobinger D, Foulds GA, Wagner S, Pickhard A, Piontek G, Kokowski K, Pockley AG, Multhoff G. Membrane Hsp70-A Novel Target for the Isolation of Circulating Tumor Cells After Epithelial-to-Mesenchymal Transition. Front Oncol 2018; 8:497. [PMID: 30443493 PMCID: PMC6223102 DOI: 10.3389/fonc.2018.00497] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022] Open
Abstract
The presence of circulating tumor cells (CTCs) in the peripheral blood is a pre-requisite for progression, invasion, and metastatic spread of cancer. Consequently, the enumeration and molecular characterization of CTCs from the peripheral blood of patients with solid tumors before, during and after treatment serves as a valuable tool for categorizing disease, evaluating prognosis and for predicting and monitoring therapeutic responsiveness. Many of the techniques for isolating CTCs are based on the expression of epithelial cell surface adhesion molecule (EpCAM, CD326) on tumor cells. However, the transition of adherent epithelial cells to migratory mesenchymal cells (epithelial-to-mesenchymal transition, EMT)-an essential element of the metastatic process-is frequently associated with a loss of expression of epithelial cell markers, including EpCAM. A highly relevant proportion of mesenchymal CTCs cannot therefore be isolated using techniques that are based on the "capture" of cells expressing EpCAM. Herein, we provide evidence that a monoclonal antibody (mAb) directed against a membrane-bound form of Hsp70 (mHsp70)-cmHsp70.1-can be used for the isolation of viable CTCs from peripheral blood of tumor patients of different entities in a more quantitative manner. In contrast to EpCAM, the expression of mHsp70 remains stably upregulated on migratory, mesenchymal CTCs, metastases and cells that have been triggered to undergo EMT. Therefore, we propose that approaches for isolating CTCs based on the capture of cells that express mHsp70 using the cmHsp70.1 mAb are superior to those based on EpCAM expression.
Collapse
Affiliation(s)
- Stephanie Breuninger
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| | - Stefan Stangl
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| | - Caroline Werner
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| | - Wolfgang Sievert
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| | - Dominik Lobinger
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, College of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Sarah Wagner
- John van Geest Cancer Research Centre, College of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Anja Pickhard
- Department of Otolaryngology Head and Neck Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Guido Piontek
- Department of Otolaryngology Head and Neck Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Konrad Kokowski
- Department of Pneumology and Pneumologic Oncology, Klinikum Bogenhausen, Munich, Germany
| | - Alan G Pockley
- John van Geest Cancer Research Centre, College of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Gabriele Multhoff
- Center for Translational Cancer Research TU München (TranslaTUM), Klinikum rechts der Isar, TUM, Munich, Germany
| |
Collapse
|
34
|
Stangl S, Tei L, De Rose F, Reder S, Martinelli J, Sievert W, Shevtsov M, Öllinger R, Rad R, Schwaiger M, D'Alessandria C, Multhoff G. Preclinical Evaluation of the Hsp70 Peptide Tracer TPP-PEG 24-DFO[ 89Zr] for Tumor-Specific PET/CT Imaging. Cancer Res 2018; 78:6268-6281. [PMID: 30228173 DOI: 10.1158/0008-5472.can-18-0707] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/04/2018] [Accepted: 09/14/2018] [Indexed: 11/16/2022]
Abstract
High precision in vivo PET/CT imaging of solid tumors improves diagnostic credibility and clinical outcome of patients. An epitope of the oligomerization domain of Hsp70 is exclusively exposed on the membrane of a large variety of tumor types, but not on normal cells, and thus provides a universal tumor-specific target. Here we developed a novel PET tracer TPP-PEG24-DFO[89Zr] based on the tumor cell-penetrating peptide probe TPP, which specifically recognizes membrane Hsp70 (mHsp70) on tumor cells. The implemented PEG24 moiety supported tracer stability and improved biodistribution characteristics in vivo The K d of the tracer ranged in the low nanomolar range (18.9 ± 11.3 nmol/L). Fluorescein isothiocyanate (FITC)-labeled derivatives TPP-[FITC] and TPP-PEG24-[FITC] revealed comparable and specific binding to mHsp70-positive 4T1, 4T1+, a derivative of the 4T1 cell line sorted for high Hsp70 expression, and CT26 tumor cells, but not to mHsp70-negative normal fibroblasts. The rapid internalization kinetics of mHsp70 into the cytosol and the favorable biodistribution of the peptide-based tracer TPP-PEG24-DFO[89Zr] in vivo enabled a tumor-specific accumulation with a high tumor-to-background contrast and renal body clearance. The tumor-specific enrichment of the tracer in 4T1+ (6.2 ± 1.1%ID/g), 4T1 (4.3 ± 0.7%ID/g), and CT26 (2.6 ± 0.6%ID/g) mouse tumors with very high, high, and intermediate mHsp70 densities, respectively, reflected mHsp70 expression profiles of the different tumor types, whereas benign mHsp70-negative fibroblastic hyperplasia showed no tracer accumulation (0.2 ± 0.03%ID/g). The ability of our chemically optimized peptide-based tracer TPP-PEG24-DFO[89Zr] to detect mHsp70 in vivo suggests its broad applicability in targeting and imaging with high specificity for any tumor type that exhibits surface expression of Hsp70.Significance: A novel peptide-based PET tracer against the oligomerization domain of Hsp70 has potential for universal tumor-specific imaging in vivo across many tumor type. Cancer Res; 78(21); 6268-81. ©2018 AACR.
Collapse
Affiliation(s)
- Stefan Stangl
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Lorenzo Tei
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Francesco De Rose
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Sybille Reder
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Jonathan Martinelli
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Wolfgang Sievert
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Maxim Shevtsov
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia.,Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Rupert Öllinger
- Medical Department II, Translational Gastroenterological Oncology, Centre for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Roland Rad
- Medical Department II, Translational Gastroenterological Oncology, Centre for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Calogero D'Alessandria
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Gabriele Multhoff
- Radiation Immuno Oncology Group, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.
| |
Collapse
|
35
|
Circulating Heat Shock Protein 70 Is a Novel Biomarker for Early Diagnosis of Lung Cancer. DISEASE MARKERS 2018; 2018:6184162. [PMID: 30245753 PMCID: PMC6136573 DOI: 10.1155/2018/6184162] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/05/2018] [Accepted: 06/11/2018] [Indexed: 02/01/2023]
Abstract
Heat shock protein 70 (HSP70) was a highly conserved protein which was significantly induced in response to cellular stresses. HSP70 played an important role in the pathogenesis of cancer which stabilized the production of large amount of oncogenic proteins and finally supported growth and survival of tumor. However, there was no report about the diagnosis of circulating HSP70 in lung cancer patients. In this study, a total of 297 participants (lung cancer: 197, healthy control: 100) were enrolled in the detection of circulating HSP70 level in plasma by ELISA assay. The results indicated that circulating HSP70 significantly decreased in lung cancer patients compared to healthy controls (P < 0.0001). Receiver operating characteristic (ROC) analysis showed that HSP70 (AUC: 82.2%, SN: 74.1%, SP: 80.0%) had higher diagnosis value than clinical existing biomarkers CEA (AUC: 80.1%, SN: 76.8%, SP: 67.3%) and CA 19-9 (AUC: 63.7%, SN: 64.2%, SP: 54.0%). In the analysis of early lung cancer patients, ROC results also revealed that HSP70 (AUC: 83.8%, SN: 71.2%, SP: 84.0%) have higher sensitivity, specificity, and AUC than CEA (AUC: 73.7%, SN: 73.2%, SP: 69.1%) and CA 19-9 (AUC: 61.5%, SN: 69.4%, SP: 53.4%). In analysis of specific histological classifications, HSP70 showed more valuable in the diagnosis of SCC (AUC: 85.9%, SN: 86.1.9%, SP: 81.0%) than ADC (AUC: 81.0%, SN: 69.1%, SP: 81.0%). Combined analysis of HSP70 and existing biomarker: CEA and CA 19-9 exhibited that HSP70 combined CEA and CA 19-9 showed the highest AUC (0.945, 95% CI, 0.855–1.000). The importance of our results was that we found decreased circulating HSP70, in combination with elevated CEA and CA 19-9, could be utilized in the diagnosis of early (stage I and II) lung cancer.
Collapse
|
36
|
Stangl S, Foulds GA, Fellinger H, Pilkington GJ, Pockley AG, Multhoff G. Immunohistochemical and Flow Cytometric Analysis of Intracellular and Membrane-Bound Hsp70, as a Putative Biomarker of Glioblastoma Multiforme, Using the cmHsp70.1 Monoclonal Antibody. Methods Mol Biol 2018; 1709:307-320. [PMID: 29177668 DOI: 10.1007/978-1-4939-7477-1_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
The major stress-inducible 70 kDa heat shock (stress) protein 70 (Hsp70) is frequently overexpressed in highly aggressive tumor cells and thus might serve as a tumor-specific biomarker of aggressive disease. We have previously shown that, in contrast to normal cells, tumor cells present Hsp70 on their plasma membrane. In order to elucidate the role of intracellular and membrane-bound Hsp70 as a potential tumor biomarker in glioblastoma multiforme, herein, we describe protocols for the staining of cytosolic Hsp70 in tumor formalin fixed paraffin-embedded (FFPE) sections using immunohistochemistry, and for plasma membrane-bound Hsp70 by multi-parametric flow cytometry using the cmHsp70.1 monoclonal antibody (mAb).
Collapse
Affiliation(s)
- Stefan Stangl
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, Munich, 81675, Germany
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK
| | - Helena Fellinger
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, Munich, 81675, Germany
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - A Graham Pockley
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, Munich, 81675, Germany.,John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technische Universität München, Ismaninger Str. 22, Munich, 81675, Germany. .,CCG-Innate Immunity in Tumor Biology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg, 85764, Germany.
| |
Collapse
|
37
|
Lamprecht C, Gehrmann M, Madl J, Römer W, Multhoff G, Ebner A. Molecular AFM imaging of Hsp70-1A association with dipalmitoyl phosphatidylserine reveals membrane blebbing in the presence of cholesterol. Cell Stress Chaperones 2018; 23:673-683. [PMID: 29404895 PMCID: PMC6045550 DOI: 10.1007/s12192-018-0879-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/10/2018] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
Hsp70-1A-the major stress-inducible member of the HSP70 chaperone family-is being implicated in cancer diseases with the development of resistances to standard therapies. In normal cells, the protein is purely cytosolic, but in a growing number of tumor cells, a significant fraction can be identified on to the cell surface. The anchoring mechanism is still under debate, as Hsp70-1A lacks conventional signaling sequences for translocation from the cytosol to exoplasmic leaflet of the plasma membrane and common membrane binding domains. Recent reports propose a lipid-mediated anchoring mechanism based on a specific interaction with charged, saturated lipids such as dipalmitoyl phosphatidylserine (DPPS). Here, we prepared planar supported lipid bilayers (SLBs) to visualize the association of Hsp70-1A directly and on the single molecule level by atomic force microscopy (AFM). The single molecule sensitivity of our approach allowed us to explore the low concentration range of 0.05 to 1.0 μg/ml of Hsp70-1A which was not studied before. We compared the binding of the protein to bilayers with 20% DPPS lipid content both in the absence and presence of cholesterol. Hsp70-1A inserted exclusively into DPPS domains and assembled in clusters with increasing protein density. A critical density was reached for incubation with 0.5 μg/ml (7 nM); at higher concentrations, membrane defects were observed that originated from cluster centers. In the presence of cholesterol, this critical concentration leads to the formation of membrane blebs, which burst at higher concentrations supporting a previously proposed non-classical pathway for the export of Hsp70-1A by tumor cells. In the discussion of our data, we attempt to link the lipid-mediated plasma membrane localization of Hsp70-1A to its potential involvement in the development of resistances to radiation and chemotherapy based on our own findings and the current literature.
Collapse
Affiliation(s)
- Constanze Lamprecht
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria.
- Institute of Physics, Experimental Polymer Physics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Str. 3, 79104, Freiburg, Germany.
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany.
| | - Mathias Gehrmann
- Department of Radiotherapy and Radiooncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Josef Madl
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Winfried Römer
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Gabriele Multhoff
- Department of Radiotherapy and Radiooncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Andreas Ebner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| |
Collapse
|
38
|
Pockley AG, Henderson B. Extracellular cell stress (heat shock) proteins-immune responses and disease: an overview. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0522. [PMID: 29203707 DOI: 10.1098/rstb.2016.0522] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2017] [Indexed: 12/11/2022] Open
Abstract
Extracellular cell stress proteins are highly conserved phylogenetically and have been shown to act as powerful signalling agonists and receptors for selected ligands in several different settings. They also act as immunostimulatory 'danger signals' for the innate and adaptive immune systems. Other studies have shown that cell stress proteins and the induction of immune reactivity to self-cell stress proteins can attenuate disease processes. Some proteins (e.g. Hsp60, Hsp70, gp96) exhibit both inflammatory and anti-inflammatory properties, depending on the context in which they encounter responding immune cells. The burgeoning literature reporting the presence of stress proteins in a range of biological fluids in healthy individuals/non-diseased settings, the association of extracellular stress protein levels with a plethora of clinical and pathological conditions and the selective expression of a membrane form of Hsp70 on cancer cells now supports the concept that extracellular cell stress proteins are involved in maintaining/regulating organismal homeostasis and in disease processes and phenotype. Cell stress proteins, therefore, form a biologically complex extracellular cell stress protein network having diverse biological, homeostatic and immunomodulatory properties, the understanding of which offers exciting opportunities for delivering novel approaches to predict, identify, diagnose, manage and treat disease.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- A Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Brian Henderson
- Division of Microbial Diseases, UCL Eastman Dental Institute, London WC1X 8LD, UK
| |
Collapse
|
39
|
Zhang L, Zhao H, Cui Z, Lv Y, Zhang W, Ma X, Zhang J, Sun B, Zhou D, Yuan L. A peptide derived from apoptin inhibits glioma growth. Oncotarget 2018; 8:31119-31132. [PMID: 28415709 PMCID: PMC5458194 DOI: 10.18632/oncotarget.16094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 03/02/2017] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma (GBM) is associated with poor prognosis due to its resistance to surgery, irradiation, and conventional chemotherapy. Thus, efficient therapeutic approaches for the treatment of GBM are urgently needed. HSP70 is an antiapoptotic protein that participates in the inhibition of both mitochondrial and membrane receptor apoptosis pathways and is highly expressed in glioma tissues. Here, we investigated a derivative of apoptin; specifically, a chicken anemia viral protein with selective toxicity toward cancer cells that can inhibit hyperactive molecules, including HSP70. Our earlier studies demonstrated that apoptin directly binds to the promoter of HSP70 and inhibits HSP70 transcription, which contributes to HSP70 downregulation. This study provides the first demonstration of the therapeutic potential of an apoptin-derived peptide for the treatment of GBM by identifying the minimal region of the apoptin domain required for interaction with the heat-shock element (HSE). This apoptin-derived peptide (ADP) inhibits glioma cell proliferation and tumor growth as well as exhibits an increased ability to promote apoptosis in GBM cells compared with rapamycin and temozolomide. ADP treatment inhibited xenograft tumor growth and increased the overall health and survival of nude mice implanted with GBM cells. These effects were measured in tumors obtained from cell lines and were observed in both intracranial and subcutaneous xenografts. In conclusion, we provide the first demonstration that ADP has therapeutic potential for the treatment of human GBM. Specifically, this study suggests that ADP is a potent candidate for drug development based on its favorable toxicity and pharmacokinetic profiles as well as its time- and cost-saving benefits.
Collapse
Affiliation(s)
- Liqiu Zhang
- Teaching Experiment Center of Biotechnology, Harbin Medical University, Harbin, P.R. China
| | - Hengyu Zhao
- Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Zhongqi Cui
- Department of Biochemistry and Molecular Biology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| | - Yueshan Lv
- Department of Immunology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| | - Wenjia Zhang
- Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Xiaoyu Ma
- Beijing Sun Palace Community Health Center, P.R. China
| | - Jianan Zhang
- Department of Biochemistry and Molecular Biology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| | - Banghao Sun
- Department of Biochemistry and Molecular Biology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| | - Danyang Zhou
- Department of Biochemistry and Molecular Biology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| | - Lijie Yuan
- Department of Biochemistry and Molecular Biology, Daqing Campus, Harbin Medical University, Daqing, Heilongjiang, Daqing, P.R. China
| |
Collapse
|
40
|
Stangl S, Tontcheva N, Sievert W, Shevtsov M, Niu M, Schmid TE, Pigorsch S, Combs SE, Haller B, Balermpas P, Rödel F, Rödel C, Fokas E, Krause M, Linge A, Lohaus F, Baumann M, Tinhofer I, Budach V, Stuschke M, Grosu AL, Abdollahi A, Debus J, Belka C, Maihöfer C, Mönnich D, Zips D, Multhoff G. Heat shock protein 70 and tumor-infiltrating NK cells as prognostic indicators for patients with squamous cell carcinoma of the head and neck after radiochemotherapy: A multicentre retrospective study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer 2017; 142:1911-1925. [PMID: 29235112 PMCID: PMC5873418 DOI: 10.1002/ijc.31213] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/28/2022]
Abstract
Tumor cells frequently overexpress heat shock protein 70 (Hsp70) and present it on their cell surface, where it can be recognized by pre-activated NK cells. In our retrospective study the expression of Hsp70 was determined in relation to tumor-infiltrating CD56+ NK cells in formalin-fixed paraffin embedded (FFPE) tumor specimens of patients with SCCHN (N = 145) as potential indicators for survival and disease recurrence. All patients received radical surgery and postoperative cisplatin-based radiochemotherapy (RCT). In general, Hsp70 expression was stronger, but with variable intensities, in tumor compared to normal tissues. Patients with high Hsp70 expressing tumors (scores 3-4) showed significantly decreased overall survival (OS; p = 0.008), local progression-free survival (LPFS; p = 0.034) and distant metastases-free survival (DMFS; p = 0.044), compared to those with low Hsp70 expression (scores 0-2), which remained significant after adjustment for relevant prognostic variables. The adverse prognostic value of a high Hsp70 expression for OS was also observed in patient cohorts with p16- (p = 0.001), p53- (p = 0.0003) and HPV16 DNA-negative (p = 0.001) tumors. The absence or low numbers of tumor-infiltrating CD56+ NK cells also correlated with significantly decreased OS (p = 0.0001), LPFS (p = 0.0009) and DMFS (p = 0.0001). A high Hsp70 expression and low numbers of tumor-infiltrating NK cells have the highest negative predictive value (p = 0.00004). In summary, a strong Hsp70 expression and low numbers of tumor-infiltrating NK cells correlate with unfavorable outcome following surgery and RCT in patients with SCCHN, and thus serve as negative prognostic markers.
Collapse
Affiliation(s)
- Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | | | - Wolfgang Sievert
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Minli Niu
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Steffi Pigorsch
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Technische Universität München (TUM), Munich, Germany
| | - Panagiotis Balermpas
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Emmanouil Fokas
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Mechthild Krause
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology, Berlin, Germany
| | - Annett Linge
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany
| | - Fabian Lohaus
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany
| | - Michael Baumann
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany
| | - Inge Tinhofer
- Department of Radiooncology and Radiotherapy, Charite University Hospital Berlin, Berlin, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Berlin, Germany
| | - Volker Budach
- Department of Radiooncology and Radiotherapy, Charite University Hospital Berlin, Berlin, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Berlin, Germany
| | - Martin Stuschke
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Essen, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Faculty, Medical Centre, University of Freiburg, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Freiburg, Germany
| | - Amir Abdollahi
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg Ion Therapy Centre (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School, Heidelberg, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany
| | - Claus Belka
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital LMU Munich, Munich, Germany.,Clinical Cooperation Group (CCG) Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
| | - Cornelius Maihöfer
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital LMU Munich, Munich, Germany.,Clinical Cooperation Group (CCG) Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
| | - David Mönnich
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany.,DKTK Consortium Tübingen, Tübingen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany.,DKTK Consortium Tübingen, Tübingen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Tübingen, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| |
Collapse
|
41
|
Zingoni A, Fionda C, Borrelli C, Cippitelli M, Santoni A, Soriani A. Natural Killer Cell Response to Chemotherapy-Stressed Cancer Cells: Role in Tumor Immunosurveillance. Front Immunol 2017; 8:1194. [PMID: 28993779 PMCID: PMC5622151 DOI: 10.3389/fimmu.2017.01194] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are innate cytotoxic lymphoid cells that actively prevent neoplastic development, growth, and metastatic dissemination in a process called cancer immunosurveillance. An equilibrium between immune control and tumor growth is maintained as long as cancer cells evade immunosurveillance. Therapies designed to kill cancer cells and to simultaneously sustain host antitumor immunity are an appealing strategy to control tumor growth. Several chemotherapeutic agents, depending on which drugs and doses are used, give rise to DNA damage and cancer cell death by means of apoptosis, immunogenic cell death, or other forms of non-apoptotic death (i.e., mitotic catastrophe, senescence, and autophagy). However, it is becoming increasingly clear that they can trigger additional stress responses. Indeed, relevant immunostimulating effects of different therapeutic programs include also the activation of pathways able to promote their recognition by immune effector cells. Among stress-inducible immunostimulating proteins, changes in the expression levels of NK cell-activating and inhibitory ligands, as well as of death receptors on tumor cells, play a critical role in their detection and elimination by innate immune effectors, including NK cells. Here, we will review recent advances in chemotherapy-mediated cellular stress pathways able to stimulate NK cell effector functions. In particular, we will address how these cytotoxic lymphocytes sense and respond to different types of drug-induced stresses contributing to anticancer activity.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristiana Borrelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,Neuromed I.R.C.C.S. - Istituto Neurologico Mediterraneo, Pozzilli, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
42
|
Brodt P. Role of the Microenvironment in Liver Metastasis: From Pre- to Prometastatic Niches. Clin Cancer Res 2016; 22:5971-5982. [PMID: 27797969 DOI: 10.1158/1078-0432.ccr-16-0460] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 01/14/2023]
Abstract
Liver metastases remain a major barrier to successful management of malignant disease, particularly for cancers of the gastrointestinal tract but also for other malignancies, such as breast carcinoma and melanoma. The ability of metastatic cells to survive and proliferate in the liver is determined by the outcome of complex, reciprocal interactions between tumor cells and different local resident subpopulations, including the sinusoidal endothelium, stellate, Kupffer, and inflammatory cells that are mediated through cell-cell and cell-extracellular matrix adhesion and the release of soluble factors. Cross-communication between different hepatic resident cells in response to local tissue damage and inflammation and the recruitment of bone marrow cells further enhance this intercellular communication network. Both resident and recruited cells can play opposing roles in the progression of metastasis, and the balance of these divergent effects determines whether the tumor cells will die, proliferate, and colonize the new site or enter a state of dormancy. Moreover, this delicate balance can be tilted in favor of metastasis, if factors produced by the primary tumor precondition the microenvironment to form niches of activated resident cells that promote tumor expansion. This review aims to summarize current knowledge on these diverse interactions and the impact they can have on the clinical management of hepatic metastases. Clin Cancer Res; 22(24); 5971-82. ©2016 AACR.
Collapse
Affiliation(s)
- Pnina Brodt
- Departments of Surgery, Medicine, and Oncology, McGill University and the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
43
|
Radons J. The human HSP70 family of chaperones: where do we stand? Cell Stress Chaperones 2016; 21:379-404. [PMID: 26865365 PMCID: PMC4837186 DOI: 10.1007/s12192-016-0676-6] [Citation(s) in RCA: 394] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 01/23/2023] Open
Abstract
The 70-kDa heat shock protein (HSP70) family of molecular chaperones represents one of the most ubiquitous classes of chaperones and is highly conserved in all organisms. Members of the HSP70 family control all aspects of cellular proteostasis such as nascent protein chain folding, protein import into organelles, recovering of proteins from aggregation, and assembly of multi-protein complexes. These chaperones augment organismal survival and longevity in the face of proteotoxic stress by enhancing cell viability and facilitating protein damage repair. Extracellular HSP70s have a number of cytoprotective and immunomodulatory functions, the latter either in the context of facilitating the cross-presentation of immunogenic peptides via major histocompatibility complex (MHC) antigens or in the context of acting as "chaperokines" or stimulators of innate immune responses. Studies have linked the expression of HSP70s to several types of carcinoma, with Hsp70 expression being associated with therapeutic resistance, metastasis, and poor clinical outcome. In malignantly transformed cells, HSP70s protect cells from the proteotoxic stress associated with abnormally rapid proliferation, suppress cellular senescence, and confer resistance to stress-induced apoptosis including protection against cytostatic drugs and radiation therapy. All of the cellular activities of HSP70s depend on their adenosine-5'-triphosphate (ATP)-regulated ability to interact with exposed hydrophobic surfaces of proteins. ATP hydrolysis and adenosine diphosphate (ADP)/ATP exchange are key events for substrate binding and Hsp70 release during folding of nascent polypeptides. Several proteins that bind to distinct subdomains of Hsp70 and consequently modulate the activity of the chaperone have been identified as HSP70 co-chaperones. This review focuses on the regulation, function, and relevance of the molecular Hsp70 chaperone machinery to disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jürgen Radons
- Scientific Consulting International, Mühldorfer Str. 64, 84503, Altötting, Germany.
| |
Collapse
|
44
|
Meng Y, Wang S, Li C, Qian M, Zheng Y, Yan X, Huang R. TKD peptide as a ligand targeting drug delivery systems to memHsp70-positive breast cancer. Int J Pharm 2016; 498:40-8. [DOI: 10.1016/j.ijpharm.2015.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/14/2015] [Accepted: 12/06/2015] [Indexed: 11/28/2022]
|
45
|
Muth C, Rubner Y, Semrau S, Rühle PF, Frey B, Strnad A, Buslei R, Fietkau R, Gaipl US. Primary glioblastoma multiforme tumors and recurrence : Comparative analysis of the danger signals HMGB1, HSP70, and calreticulin. Strahlenther Onkol 2015; 192:146-55. [PMID: 26646311 DOI: 10.1007/s00066-015-0926-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/11/2015] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor. Despite improved multimodal therapies, the tumor recurs in most cases. Diverging patient survival suggests great tumor heterogeneity and different therapy responses. Danger signals such as high-mobility group box protein 1 (HMGB1), heat shock protein 70 (HSP70), and calreticulin (CRT) are biomarker candidates, due to their association with tumor progression versus induction of antitumor immune responses. Overexpression of these danger signals has been reported for various types of tumors; however, their role in GBM is still elusive. A direct comparison of their expression in the primary tumor versus the corresponding relapse is still lacking for most tumor entities. PATIENTS AND METHODS We therefore performed an expression analysis by immunohistochemistry of the danger signals HMGB1, HSP70, and CRT in primary tumors and the corresponding relapses of 9 patients with de novo GBM. RESULTS HMGB1 was highly expressed in primary tumors with a significant reduction in the respective relapse. The extracellular HSP70 expression was significantly increased in the relapse compared to the primary tumor. CRT was generally highly expressed in the primary tumor, with a slight increase in the relapse. CONCLUSION The combination of a decreased expression of HMGB1, an increased expression of extracellular HSP70, and an increased expression of CRT in the relapse seems to be beneficial for patient survival. HMGB1, extracellular HSP70, and CRT could be taken into concerted consideration as potential biomarkers for the prognosis of patients with GBM.
Collapse
Affiliation(s)
- Carolin Muth
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yvonne Rubner
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Paul-Friedrich Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Annedore Strnad
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rolf Buslei
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
46
|
Gunther S, Ostheimer C, Stangl S, Specht HM, Mozes P, Jesinghaus M, Vordermark D, Combs SE, Peltz F, Jung MP, Multhoff G. Correlation of Hsp70 Serum Levels with Gross Tumor Volume and Composition of Lymphocyte Subpopulations in Patients with Squamous Cell and Adeno Non-Small Cell Lung Cancer. Front Immunol 2015; 6:556. [PMID: 26579130 PMCID: PMC4629690 DOI: 10.3389/fimmu.2015.00556] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/17/2015] [Indexed: 12/16/2022] Open
Abstract
Heat-shock protein 70 (Hsp70) is frequently found on the plasma membrane of a large number of malignant tumors including non-small cell lung cancer (NSCLC) and gets released into the blood circulation in lipid vesicles. On the one hand, a membrane (m)Hsp70-positive phenotype correlates with a high aggressiveness of the tumor; on the other hand, mHsp70 serves as a target for natural killer (NK) cells that had been pre-stimulated with Hsp70-peptide TKD plus low-dose interleukin-2 (TKD/IL-2). Following activation, NK cells show an up-regulated expression of activatory C-type lectin receptors, such as CD94/NKG2C, NKG2D, and natural cytotoxicity receptors (NCRs; NKp44, NKp46, and NKp30) and thereby gain the capacity to kill mHsp70-positive tumor cells. With respect to these results, the efficacy of ex vivo TKD/IL-2 stimulated, autologous NK cells is currently tested in a proof-of-concept phase II clinical trial in patients with squamous cell NSCLC after radiochemotherapy (RCT) at the TUM. Inclusion criteria are histological proven, non-resectable NSCLC in stage IIIA/IIIB, clinical responses to RCT and a mHsp70-positive tumor phenotype. The mHsp70 status is determined in the serum of patients using the lipHsp70 ELISA test, which enables the quantification of liposomal and free Hsp70. Squamous cell and adeno NSCLC patients had significantly higher serum Hsp70 levels than healthy controls. A significant correlation of serum Hsp70 levels with the gross tumor volume was shown for adeno and squamous cell NSCLC. However, significantly elevated ratios of activated CD69(+)/CD94(+) NK cells that are associated with low serum Hsp70 levels were observed only in patients with squamous cell lung cancer. These data might provide a first hint that squamous cell NSCLC is more immunogenic than adeno NSCLC.
Collapse
Affiliation(s)
- Sophie Gunther
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Christian Ostheimer
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg , Halle (Saale) , Germany
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Hanno M Specht
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Petra Mozes
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Moritz Jesinghaus
- Department of Pathology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Dirk Vordermark
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg , Halle (Saale) , Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany ; Department of Innovative Radiotherapy (iRT), Helmholtz Zentrum München , Oberschleißheim , Germany ; Department of Radiation Sciences (DRS), Helmholtz Zentrum München , Oberschleißheim , Germany
| | - Friedhelm Peltz
- Pulmonary Division, 1 Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Max P Jung
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM) , München , Germany ; Department of Innovative Radiotherapy (iRT), Helmholtz Zentrum München , Oberschleißheim , Germany ; Department of Radiation Sciences (DRS), Helmholtz Zentrum München , Oberschleißheim , Germany
| |
Collapse
|
47
|
Wang Y, Wang X, Ferrone CR, Schwab JH, Ferrone S. Intracellular antigens as targets for antibody based immunotherapy of malignant diseases. Mol Oncol 2015; 9:1982-93. [PMID: 26597109 DOI: 10.1016/j.molonc.2015.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022] Open
Abstract
This review discusses the potential use of intracellular tumor antigens as targets of antibody-based immunotherapy for the treatment of solid tumors. In addition, it describes the characteristics of the intracellular tumor antigens targeted with antibodies which have been described in the literature and have been identified in the authors' laboratory. Finally, the mechanism underlying the trafficking of the intracellular tumor antigens to the plasma membrane of tumor cells are reviewed.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Joseph H Schwab
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States; Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
48
|
Derer A, Deloch L, Rubner Y, Fietkau R, Frey B, Gaipl US. Radio-Immunotherapy-Induced Immunogenic Cancer Cells as Basis for Induction of Systemic Anti-Tumor Immune Responses - Pre-Clinical Evidence and Ongoing Clinical Applications. Front Immunol 2015; 6:505. [PMID: 26500646 PMCID: PMC4597129 DOI: 10.3389/fimmu.2015.00505] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/16/2015] [Indexed: 01/18/2023] Open
Abstract
Radiotherapy (RT) primarily aims to locally destroy the tumor via the induction of DNA damage in the tumor cells. However, the so-called abscopal, namely systemic and immune–mediated, effects of RT move over more and more in the focus of scientists and clinicians since combinations of local irradiation with immune therapy have been demonstrated to induce anti-tumor immunity. We here summarize changes of the phenotype and microenvironment of tumor cells after exposure to irradiation, chemotherapeutic agents, and immune modulating agents rendering the tumor more immunogenic. The impact of therapy-modified tumor cells and damage-associated molecular patterns on local and systemic control of the primary tumor, recurrent tumors, and metastases will be outlined. Finally, clinical studies affirming the bench-side findings of interactions and synergies of radiation therapy and immunotherapy will be discussed. Focus is set on combination of radio(chemo)therapy (RCT) with immune checkpoint inhibitors, growth factor inhibitors, and chimeric antigen receptor T-cell therapy. Well-deliberated combination of RCT with selected immune therapies and growth factor inhibitors bear the great potential to further improve anti-cancer therapies.
Collapse
Affiliation(s)
- Anja Derer
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Lisa Deloch
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Yvonne Rubner
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
49
|
Murakami N, Kühnel A, Schmid TE, Ilicic K, Stangl S, Braun IS, Gehrmann M, Molls M, Itami J, Multhoff G. Role of membrane Hsp70 in radiation sensitivity of tumor cells. Radiat Oncol 2015. [PMID: 26197988 PMCID: PMC4511458 DOI: 10.1186/s13014-015-0461-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The major stress-inducible heat shock protein 70 (Hsp70) is frequently overexpressed in the cytosol and integrated in the plasma membrane of tumor cells via lipid anchorage. Following stress such as non-lethal irradiation Hsp70 synthesis is up-regulated. Intracellular located Hsp70 is known to exert cytoprotective properties, however, less is known about membrane (m)Hsp70. Herein, we investigate the role of mHsp70 in the sensitivity towards irradiation in tumor sublines that differ in their cytosolic and/or mHsp70 levels. METHODS The isogenic human colon carcinoma sublines CX(+) with stable high and CX(-) with stable low expression of mHsp70 were generated by fluorescence activated cell sorting, the mouse mammary carcinoma sublines 4 T1 (4 T1 ctrl) and Hsp70 knock-down (4 T1 Hsp70 KD) were produced using the CRISPR/Cas9 system, and the Hsp70 down-regulation in human lung carcinoma sublines H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/EPLC-272H HSF-1 KD was achieved by small interfering (si)RNA against Heat shock factor 1 (HSF-1). Cytosolic and mHsp70 was quantified by Western blot analysis/ELISA and flow cytometry; double strand breaks (DSBs) and apoptosis were measured by flow cytometry using antibodies against γH2AX and real-time PCR (RT-PCR) using primers and antibodies directed against apoptosis related genes; and radiation sensitivity was determined using clonogenic cell surviving assays. RESULTS CX(+)/CX(-) tumor cells exhibited similar cytosolic but differed significantly in their mHsp70 levels, 4 T1 ctrl/4 T1 Hsp70 KD cells showed significant differences in their cytosolic and mHsp70 levels and H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/EPLC-272H HSF-1 KD lung carcinoma cell sublines had similar mHsp70 but significantly different cytosolic Hsp70 levels. γH2AX was significantly up-regulated in irradiated CX(-) and 4 T1 Hsp70 KD with low basal mHsp70 levels, but not in their mHsp70 high expressing counterparts, irrespectively of their cytosolic Hsp70 content. After irradiation γH2AX, Caspase 3/7 and Annexin V were up-regulated in the lung carcinoma sublines, but no significant differences were observed in H1339 ctrl/H1339 HSF-1 KD, and EPLC-272H ctrl/EPLC-272H HSF-1 KD that exhibit identical mHsp70 but different cytosolic Hsp70 levels. Clonogenic cell survival was significantly lower in CX(-) and 4 T1 Hsp70 KD cells with low mHsp70 expression, than in CX+ and 4 T1 ctrl cells, whereas no difference in clonogenic cell survival was observed in H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/ EPLC-272H HSF-1 KD sublines with identical mHsp70 but different cytosolic Hsp70 levels. CONCLUSION In summary, our results indicate that mHsp70 has an impact on radiation resistance.
Collapse
Affiliation(s)
- Naoya Murakami
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Annett Kühnel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katarina Ilicic
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Isabella S Braun
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Mathias Gehrmann
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Molls
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jun Itami
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. .,Clinical Cooperation Group - Innate Immunity in Tumor Biology, Institute of Biomedical Imaging (IBMI), Helmholtz Zentrum München, Munich, Germany.
| |
Collapse
|
50
|
Abstract
Although heat-shock (cell stress) proteins are commonly considered as being intracellular molecular chaperones that undertake a number of cytoprotective and cellular housekeeping functions, there is now a wealth of evidence to indicate that these proteins can be released by cells via active processes. Many molecular chaperones are secreted, or exist as cell surface proteins which can act as powerful signalling agonists and also as receptors for selected ligands. Levels of heat-shock (cell stress) proteins in biological fluids are now being associated with a plethora of clinical conditions, and these proteins therefore have potential utility as biomarkers of disease and/or response to therapeutic intervention. The present article summarizes current knowledge relating to extracellular cell stress proteins as biomarkers of human disease.
Collapse
|