1
|
Tuo X, Chen J, Hao C, Dai X, Zhu J, Tian S, Zhang Y, Wang F. Identification of GPNMB in endometrial cancer based on pan-cancer analysis and in vitro validation. Discov Oncol 2024; 15:489. [PMID: 39331202 PMCID: PMC11436576 DOI: 10.1007/s12672-024-01382-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND GPNMB is a type I transmembrane protein, and emerging evidence supports the relationship between GPNMB and cancers. OBJECTIVE Through a comprehensive pan-cancer analysis, we examined the expression levels, prognostic significance, and mutation profiles of GPNMB in different cancer types. Subsequently, utilizing in vitro experiments, we elucidated the impact of GPNMB in endometrial cancer (EC). METHODS TIMER2, GEPIA2, UALCAN and cBioPortal were used to analyze the expression pattern, prognostic values, and mutation status of GPNMB. HEC-1B and Ishikawa cells were used to conduct in vitro analyses of GPNMB overexpression. GeneMANIA and TIMER2 were used to evaluate the potential functions and correlations between GPNMB expression and tumor-infiltrating immune cells in EC. RESULTS GPNMB was found to be highly expressed in multiple cancers, where it was associated with poor prognosis. Additionally, GPNMB was downregulated at both mRNA and protein levels in EC. Overexpression of GPNMB inhibited the proliferation, migration, and invasion of HEC-1B and Ishikawa cells. Functional analysis showed that GPNMB was enriched in pathways associated with regulation of plasma lipoprotein particle levels. The expression of GPNMB was positively connected with B cell, CD8+ T cell, CD4+ T cell, Macrophage, Neutrophil, and Dendritic cell levels. CONCLUSION Through pan-cancer analysis, we identified the antitumor effect of GPNMB in EC and predicted the potential mechanisms between GPNMB expression and EC.
Collapse
Affiliation(s)
- Xiaoqian Tuo
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Jialan Chen
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Cuipei Hao
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Xiaole Dai
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Jiayi Zhu
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Siqi Tian
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Yan Zhang
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Fan Wang
- Department of Gynecology, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, People's Republic of China.
| |
Collapse
|
2
|
Barreto-Núñez R, Béland LC, Boutej H, Picher-Martel V, Dupré N, Barbeito L, Kriz J. Chronically activated microglia in ALS gradually lose their immune functions and develop unconventional proteome. Glia 2024; 72:1319-1339. [PMID: 38577970 DOI: 10.1002/glia.24531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/15/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
Neuroinflammation and chronic activation of microglial cells are the prominent features of amyotrophic lateral sclerosis (ALS) pathology. While alterations in the mRNA profile of diseased microglia have been well documented, the actual microglia proteome remains poorly characterized. Here we performed a functional characterization together with proteome analyses of microglial cells at different stages of disease in the SOD1-G93A model of ALS. Functional analyses of microglia derived from the lumbar spinal cord of symptomatic mice revealed: (i) remarkably high mitotic index (close to 100% cells are Ki67+) (ii) significant decrease in phagocytic capacity when compared to age-matched control microglia, and (iii) diminished response to innate immune challenges in vitro and in vivo. Proteome analysis revealed a development of two distinct molecular signatures at early and advanced stages of disease. While at early stages of disease, we identified several proteins implicated in microglia immune functions such as GPNMB, HMBOX1, at advanced stages of disease microglia signature at protein level was characterized with a robust upregulation of several unconventional proteins including rootletin, major vaults proteins and STK38. Upregulation of GPNMB and rootletin has been also found in the spinal cord samples of sporadic ALS. Remarkably, the top biological functions of microglia, in particular in the advanced disease, were not related to immunity/immune response, but were highly enriched in terms linked to RNA metabolism. Together, our results suggest that, over the course of disease, chronically activated microglia develop unconventional protein signatures and gradually lose their immune identity ultimately turning into functionally inefficient immune cells.
Collapse
Affiliation(s)
| | | | - Hejer Boutej
- CERVO Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Vincent Picher-Martel
- CERVO Research Centre, Laval University, Quebec City, Quebec, Canada
- Division of Neuroscence, Centre Hospitalier Universitaire de Québecṣ-Université Laval Research Center, Quebec City, Québec, Canada
| | - Nicolas Dupré
- Division of Neuroscence, Centre Hospitalier Universitaire de Québecṣ-Université Laval Research Center, Quebec City, Québec, Canada
- Department of Psychiatry and Neuroscience, Faculty Medicine, Laval University, Quebec City, Quebec, Canada
| | | | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec, Canada
- Department of Psychiatry and Neuroscience, Faculty Medicine, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
3
|
Klein S, Tolkach Y, Reinhardt HC, Buettner R, Quaas A, Helbig D. Proteomic analysis of pleomorphic dermal sarcoma reveals a fibroblastic cell of origin and distinct immune evasion mechanisms. Sci Rep 2024; 14:12516. [PMID: 38822058 PMCID: PMC11143252 DOI: 10.1038/s41598-024-62927-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Pleomorphic dermal sarcomas are infrequent neoplastic skin tumors, manifesting in regions of the skin exposed to ultraviolet radiation. Diagnosing the entity can be challenging and therapeutic options are limited. We analyzed 20 samples of normal healthy skin tissue (SNT), 27 malignant melanomas (MM), 20 cutaneous squamous cell carcinomas (cSCC), and 24 pleomorphic dermal sarcomas (PDS) using mass spectrometry. We explored a potential cell of origin in PDS and validated our findings using publicly available single-cell sequencing data. By correlating tumor purity (TP), inferred by both RNA- and DNA-sequencing, to protein abundance, we found that fibroblasts shared most of the proteins correlating to TP. This observation could also be made using publicly available SNT single cell sequencing data. Moreover, we studied relevant pathways of receptor/ligand (R/L) interactions. Analysis of R/L interactions revealed distinct pathways in cSCC, MM and PDS, with a prominent role of PDGFRB-PDGFD R/L interactions and upregulation of PI3K/AKT signaling pathway. By studying differentially expressed proteins between cSCC and PDS, markers such as MAP1B could differentiate between these two entities. To this end, we studied proteins associated with immunosuppression in PDS, uncovering that immunologically cold PDS cases shared a "negative regulation of interferon-gamma signaling" according to overrepresentation analysis.
Collapse
Affiliation(s)
- Sebastian Klein
- Department of Hematology and Stem Cell Transplantation, University Duisburg-Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
- West German Cancer Center Network, Partner Site Essen, Essen, Germany.
- Institute for Pathology and Neuropathology, University Hospital and Medical Faculty Cologne, Kerpenerstr 62, 50937, Cologne, Germany.
| | - Yuri Tolkach
- Institute for Pathology and Neuropathology, University Hospital and Medical Faculty Cologne, Kerpenerstr 62, 50937, Cologne, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Duisburg-Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
- West German Cancer Center Network, Partner Site Essen, Essen, Germany
| | - Reinhard Buettner
- Institute for Pathology and Neuropathology, University Hospital and Medical Faculty Cologne, Kerpenerstr 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute for Pathology and Neuropathology, University Hospital and Medical Faculty Cologne, Kerpenerstr 62, 50937, Cologne, Germany
| | - Doris Helbig
- Department of Dermatology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
4
|
Kimura T, Okita Y, Nagumo Y, Chin JM, Fikry MA, Shiga M, Kandori S, Kawahara T, Suzuki H, Nishiyama H, Kato M. Glycoprotein nonmetastatic melanoma protein B impacts the malignant potential of bladder cancer cells through its hem-immunoreceptor tyrosine-based activation motif. Pathol Int 2024; 74:262-273. [PMID: 38501371 DOI: 10.1111/pin.13419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
Bladder cancer is one of the most common cancers among men worldwide. Although multiple genomic mutations and epigenetic alterations have been identified, an efficacious molecularly targeted therapy has yet to be established. Therefore, a novel approach is anticipated. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane glycoprotein that is highly expressed in various cancers. In this study, we evaluated bladder cancer patient samples and found that GPNMB protein abundance is associated with high-grade tumors, and both univariate and multivariate analyses showed that GPNMB is a prognostic factor. Furthermore, the prognosis of patients with high GPNMB levels was significantly poorer in those with nonmuscle invasive bladder cancer (NMIBC) than in those with muscle invasive bladder cancer (MIBC). We then demonstrated that knockdown of GPNMB in MIBC cell lines with high GPNMB inhibits cellular migration and invasion, whereas overexpression of GPNMB further enhances cellular migration and invasion in MIBC cell lines with originally low GPNMB. Therefore, we propose that GPNMB is one of multiple driver molecules in the acquisition of cellular migratory and invasive potential in bladder cancers. Moreover, we revealed that the tyrosine residue in the hemi-immunoreceptor tyrosine-based activation motif (hemITAM) is required for GPNMB-induced cellular motility.
Collapse
Affiliation(s)
- Tomokazu Kimura
- Department of Urology, University of Tsukuba, Ibaraki, Japan
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yukari Okita
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | | | - Jas Min Chin
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Muhammad Ali Fikry
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masanobu Shiga
- Department of Urology, University of Tsukuba, Ibaraki, Japan
| | - Shuya Kandori
- Department of Urology, University of Tsukuba, Ibaraki, Japan
| | | | - Hiroyuki Suzuki
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Mitsuyasu Kato
- Department of Experimental Pathology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
5
|
Bhore N, Bogacki EC, O'Callaghan B, Plun-Favreau H, Lewis PA, Herbst S. Common genetic risk for Parkinson's disease and dysfunction of the endo-lysosomal system. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220517. [PMID: 38368938 PMCID: PMC10874702 DOI: 10.1098/rstb.2022.0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/18/2023] [Indexed: 02/20/2024] Open
Abstract
Parkinson's disease is a progressive neurological disorder, characterized by prominent movement dysfunction. The past two decades have seen a rapid expansion of our understanding of the genetic basis of Parkinson's, initially through the identification of monogenic forms and, more recently, through genome-wide association studies identifying common risk variants. Intriguingly, a number of cellular pathways have emerged from these analysis as playing central roles in the aetiopathogenesis of Parkinson's. In this review, the impact of data deriving from genome-wide analyses for Parkinson's upon our functional understanding of the disease will be examined, with a particular focus on examples of endo-lysosomal and mitochondrial dysfunction. The challenges of moving from a genetic to a functional understanding of common risk variants for Parkinson's will be discussed, with a final consideration of the current state of the genetic architecture of the disorder. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Noopur Bhore
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University of London, London WC1N 3BG, UK
| | - Erin C. Bogacki
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Benjamin O'Callaghan
- Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University of London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Helene Plun-Favreau
- Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University of London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Patrick A. Lewis
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University of London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Susanne Herbst
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University of London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
6
|
Capuozzo M, Celotto V, Santorsola M, Fabozzi A, Landi L, Ferrara F, Borzacchiello A, Granata V, Sabbatino F, Savarese G, Cascella M, Perri F, Ottaiano A. Emerging treatment approaches for triple-negative breast cancer. Med Oncol 2023; 41:5. [PMID: 38038783 DOI: 10.1007/s12032-023-02257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
Approximately, 15% of global breast cancer cases are diagnosed as triple-negative breast cancer (TNBC), identified as the most aggressive subtype due to the simultaneous absence of estrogen receptor, progesterone receptor, and HER2. This characteristic renders TNBC highly aggressive and challenging to treat, as it excludes the use of effective drugs such as hormone therapy and anti-HER2 agents. In this review, we explore standard therapies and recent emerging approaches for TNBC, including PARP inhibitors, immune checkpoint inhibitors, PI3K/AKT pathway inhibitors, and cytotoxin-conjugated antibodies. The mechanism of action of these drugs and their utilization in clinical practice is explained in a pragmatic and prospective manner, contextualized within the current landscape of standard therapies for this pathology. These advancements present a promising frontier for tailored interventions with the potential to significantly improve outcomes for TNBC patients. Interestingly, while TNBC poses a complex challenge, it also serves as a paradigm and an opportunity for translational research and innovative therapies in the field of oncology.
Collapse
Affiliation(s)
- Maurizio Capuozzo
- Pharmaceutical Department, ASL Napoli 3, Ercolano, 80056, Naples, Italy
| | - Venere Celotto
- Pharmaceutical Department, ASL Napoli 3, Ercolano, 80056, Naples, Italy
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Antonio Fabozzi
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Loris Landi
- Sanitary District, Ds. 58 ASL Napoli 3, Pompei, 80045, Naples, Italy
| | - Francesco Ferrara
- Pharmaceutical Department, ASL Napoli 3, Via Dell'amicizia 22, Nola, 80035, Naples, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Naples, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, 84081, Salerno, Italy
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale Srl, Via Padre Carmine Fico 24, Casalnuovo Di, 80013, Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy.
| |
Collapse
|
7
|
Gillett DA, Wallings RL, Uriarte Huarte O, Tansey MG. Progranulin and GPNMB: interactions in endo-lysosome function and inflammation in neurodegenerative disease. J Neuroinflammation 2023; 20:286. [PMID: 38037070 PMCID: PMC10688479 DOI: 10.1186/s12974-023-02965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Alterations in progranulin (PGRN) expression are associated with multiple neurodegenerative diseases (NDs), including frontotemporal dementia (FTD), Alzheimer's disease (AD), Parkinson's disease (PD), and lysosomal storage disorders (LSDs). Recently, the loss of PGRN was shown to result in endo-lysosomal system dysfunction and an age-dependent increase in the expression of another protein associated with NDs, glycoprotein non-metastatic B (GPNMB). MAIN BODY It is unclear what role GPNMB plays in the context of PGRN insufficiency and how they interact and contribute to the development or progression of NDs. This review focuses on the interplay between these two critical proteins within the context of endo-lysosomal health, immune function, and inflammation in their contribution to NDs. SHORT CONCLUSION PGRN and GPNMB are interrelated proteins that regulate disease-relevant processes and may have value as therapeutic targets to delay disease progression or extend therapeutic windows.
Collapse
Affiliation(s)
- Drew A Gillett
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Rebecca L Wallings
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Oihane Uriarte Huarte
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Li H, Argani P, Halper-Stromberg E, Lotan TL, Merino MJ, Reuter VE, Matoso A. Positive GPNMB Immunostaining Differentiates Renal Cell Carcinoma With Fibromyomatous Stroma Associated With TSC1/2/MTOR Alterations From Others. Am J Surg Pathol 2023; 47:1267-1273. [PMID: 37661807 PMCID: PMC10592185 DOI: 10.1097/pas.0000000000002117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Renal cell carcinoma with fibromyomatous stroma (RCCFMS) include ELOC/TCEB1 -mutated renal cell carcinoma (RCC) and those with TSC1/2 / MTOR alterations. Besides morphologic similarity, most of these tumors is known to be diffusely positive for carbonic anhydrase IX and cytokeratin 7 by immunohistochemistry. We previously showed strong and diffuse expression of GPNMB (glycoprotein nonmetastatic B) in translocation RCC and eosinophilic renal neoplasms with known TSC1/2/MTOR alterations. We retrospectively identified molecularly confirmed cases of TCEB1/ELOC -mutated RCC (7 tumors from 7 patients), and RCCFMS with alterations in TSC1/2/MTOR (6 tumors from 5 patients, 1 patient with tuberous sclerosis syndrome). In addition, we included 7 clear cell papillary renal cell tumors (CCPRCTs) and 8 clear cell RCC, as they can also present morphologic overlap with RCCFMS. Morphologically, RCCs with TSC1/2/MTOR alterations and those with TCEB1/ELOC mutations were indistinguishable and characterized by papillary, nested, or tubular architecture, with tumor cells with clear cytoplasm and low nuclear grade. By immunohistochemistry, cytokeratin 7 was positive in 5/7 (71%) of TCEB1/ELOC -mutated RCCs, 6/6 (100%) of RCCs with TSC1/2/mTOR alterations, and 7/7 (100%) of CCPRCTs ( P =not significant). Carbonic anhydrase IX was positive in 7/7 TCEB1/ELOC -mutated RCCs, 6/6 (100%) of RCCs with TSC1/2/MTOR alterations, and 7/7 (100%) of CCPRCTs ( P =NS). GPNMB was strongly and diffusely positive in all tumors with TSC1/2/MTOR alterations (6/6), while negative in all TCEB1/ELOC -mutated RCCs (0/6), or CCPRCTs (0/7) ( P =0.002). Two of 8 clear cell RCC showed focal weak staining, while 6/8 were negative. In conclusion, the results support the use of GPNMB to distinguish RCCFMS with TSC1/2/MTOR alterations from others with similar morphology.
Collapse
Affiliation(s)
- Huili Li
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | | | - Tamara L. Lotan
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | - Maria J. Merino
- Translational Surgical Pathology, Laboratory of Pathology, National Institutes of health, Bethesda, MD 20892
| | - Victor E. Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021
| | - Andres Matoso
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| |
Collapse
|
9
|
Venetis K, Sajjadi E, Ivanova M, Peccatori FA, Fusco N, Guerini-Rocco E. Characterization of the immune environment in pregnancy-associated breast cancer. Future Oncol 2023. [PMID: 37376974 DOI: 10.2217/fon-2022-1321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Abstract
Pregnancy-associated breast cancer (PrBC) is a rare and clinically challenging condition. Specific immune mechanisms and pathways are involved in maternal-fetal tolerance and tumor-host immunoediting. The comprehension of the molecular processes underpinning this immune synergy in PrBC is needed to improve patients' clinical management. Only a few studies focused on the immune biology of PrBC and attempted to identify bona fide biomarkers. Therefore, clinically actionable information remains extremely puzzling for these patients. In this review article, we discuss the current knowledge on the immune environment of PrBC, in comparison with pregnancy-unrelated breast cancer and in the context of maternal immune changes during pregnancy. A particular emphasis is given to the actual role of potential immune-related biomarkers for PrBC clinical management.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
- Department of Oncology & Hemato-Oncology, University of Milan, Milan, 20122, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
| | - Fedro Alessandro Peccatori
- Fertility & Procreation Unit, Division of Gynecologic Oncology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
- Department of Oncology & Hemato-Oncology, University of Milan, Milan, 20122, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, 20141, Italy
- Department of Oncology & Hemato-Oncology, University of Milan, Milan, 20122, Italy
| |
Collapse
|
10
|
Sánchez-León ML, Jiménez-Cortegana C, Silva Romeiro S, Garnacho C, de la Cruz-Merino L, García-Domínguez DJ, Hontecillas-Prieto L, Sánchez-Margalet V. Defining the Emergence of New Immunotherapy Approaches in Breast Cancer: Role of Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:5208. [PMID: 36982282 PMCID: PMC10048951 DOI: 10.3390/ijms24065208] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Breast cancer (BC) continues to be the most diagnosed tumor in women and a very heterogeneous disease both inter- and intratumoral, mainly given by the variety of molecular profiles with different biological and clinical characteristics. Despite the advancements in early detection and therapeutic strategies, the survival rate is low in patients who develop metastatic disease. Therefore, it is mandatory to explore new approaches to achieve better responses. In this regard, immunotherapy arose as a promising alternative to conventional treatments due to its ability to modulate the immune system, which may play a dual role in this disease since the relationship between the immune system and BC cells depends on several factors: the tumor histology and size, as well as the involvement of lymph nodes, immune cells, and molecules that are part of the tumor microenvironment. Particularly, myeloid-derived suppressor cell (MDSC) expansion is one of the major immunosuppressive mechanisms used by breast tumors since it has been associated with worse clinical stage, metastatic burden, and poor efficacy of immunotherapies. This review focuses on the new immunotherapies in BC in the last five years. Additionally, the role of MDSC as a therapeutic target in breast cancer will be described.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carlos Jiménez-Cortegana
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Silvia Silva Romeiro
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Víctor Sánchez-Margalet
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| |
Collapse
|
11
|
Mamais A, Wallings R, Rocha EM. Disease mechanisms as subtypes: Lysosomal dysfunction in the endolysosomal Parkinson's disease subtype. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:33-51. [PMID: 36803821 DOI: 10.1016/b978-0-323-85555-6.00009-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Parkinson's disease (PD) remains one of the most prevalent neurodegenerative disorders. It has become increasingly recognized that PD is not one disease but a constellation of many, with distinct cellular mechanisms driving pathology and neuronal loss in each given subtype. Endolysosomal trafficking and lysosomal degradation are crucial to maintain neuronal homeostasis and vesicular trafficking. It is clear that deficits in endolysosomal signaling data support the existence of an endolysosomal PD subtype. This chapter describes how cellular pathways involved in endolysosomal vesicular trafficking and lysosomal degradation in neurons and immune cells can contribute to PD. Last, as inflammatory processes including phagocytosis and cytokine release are central in glia-neuron interactions, a spotlight on the role of neuroinflammation plays in the pathogenesis of this PD subtype is also explored.
Collapse
Affiliation(s)
- Adamantios Mamais
- Department of Neurology, College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, United States; Center for Translational Research in Neurodegenerative disease, University of Florida, Gainesville, FL, United States
| | - Rebecca Wallings
- Department of Neurology, College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, United States; Center for Translational Research in Neurodegenerative disease, University of Florida, Gainesville, FL, United States
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
12
|
Son S, Kim H, Lim H, Lee JH, Lee KM, Shin I. CCN3/NOV promotes metastasis and tumor progression via GPNMB-induced EGFR activation in triple-negative breast cancer. Cell Death Dis 2023; 14:81. [PMID: 36737605 PMCID: PMC9898537 DOI: 10.1038/s41419-023-05608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. TNBC patients typically exhibit unfavorable outcomes due to its rapid growth and metastatic potential. Here, we found overexpression of CCN3 in TNBC patients. We identified that CCN3 knockdown diminished cancer stem cell formation, metastasis, and tumor growth in vitro and in vivo. Mechanistically, ablation of CCN3 reduced activity of the EGFR/MAPK pathway. Transcriptome profiling revealed that CCN3 induces glycoprotein nonmetastatic melanoma protein B (GPNMB) expression, which in turn activates the EGFR pathway. An interrogation of the TCGA dataset further supported the transcriptional regulation of GPNMB by CCN3. Finally, we showed that CCN3 activates Wnt signaling through a ligand-dependent or -independent mechanism, which increases microphthalmia-associated transcription factor (MITF) protein, a transcription factor inducing GPNMB expression. Together, our findings demonstrate the oncogenic role of CCN3 in TNBC, and we propose CCN3 as a putative therapeutic target for TNBC.
Collapse
Affiliation(s)
- Seogho Son
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Hogeun Lim
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Joo-Hyung Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
| | - Kyung-Min Lee
- Department of Life Science, Hanyang University, Seoul, 04763, Korea
- Natural Science Institute, Hanyang University, Seoul, 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul, 04763, Korea.
- Natural Science Institute, Hanyang University, Seoul, 04763, Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
13
|
Al Jarroudi O, El Bairi K, Curigliano G, Afqir S. Antibody-Drug Conjugates: A New Therapeutic Approach for Triple-Negative Breast Cancer. Cancer Treat Res 2023; 188:1-27. [PMID: 38175340 DOI: 10.1007/978-3-031-33602-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subset associated with a worse prognosis and poor response to conventional chemotherapy. Despite recent advances in drug discovery, its management is still a challenge for clinicians, illuminating the unmet need to develop novel treatment approaches. Antibody-drug conjugates (ADC) are innovative oncology drugs that combine the specificity of monoclonal antibodies and the high efficacy of anticancer payloads, to deliver cytotoxic drugs selectively to cancer cells. Various ADCs were investigated for TNBC and have provided a promise for this aggressive women's cancer including the FDA-approved sacituzumab govitecan. In this chapter, we reviewed different ADCs studied for TNBC including their mechanisms of action, efficacy, and tolerability. Moreover, we have also discussed their therapeutic potential based on combinatorial approaches with other targeted therapies in early and metastatic TNBC.
Collapse
Affiliation(s)
- Ouissam Al Jarroudi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco.
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco.
| | - Khalid El Bairi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Said Afqir
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
14
|
Chen N, Michaels E, Howard F, Nanda R. The evolving therapeutic landscape of antibody-drug conjugates in breast cancer. Expert Rev Anticancer Ther 2022; 22:1325-1331. [PMID: 36408586 PMCID: PMC9833603 DOI: 10.1080/14737140.2022.2147510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) are a relatively new class of anti-cancer therapies approved for a number of malignancies, including breast cancer. Their unique structure, consisting of a monoclonal antibody connected via a linker to a toxic payload, combines characteristics of both targeted therapy and chemotherapy. AREAS COVERED In this review, we discuss the unique molecular structure and pharmacologic principles of ADCs and present the clinical efficacy and relevant toxicities of ADCs both approved and in development. While HER2 is the most studied target with approved agents for both HER2-positive and HER2-low expressing tumors, novel targets in HER2-negative disease have expanded our therapeutic capabilities significantly. EXPERT OPINION ADCs are a promising, novel drug class with significant efficacy in all breast cancer subtypes. They are generally safe and well-tolerated. However, further research is necessary to improve their therapeutic potential. The development of predictive biomarkers to identify patients with greatest benefit, improved understanding of drug resistance to advance combination therapies, and novel targets are needed to further the field.
Collapse
Affiliation(s)
- Nan Chen
- Department of Internal Medicine, University of Chicago
| | | | | | - Rita Nanda
- Department of Internal Medicine, University of Chicago
| |
Collapse
|
15
|
Yazdi SI, Sadeghi M, Saeedzadeh E, Jalilifar M. Radiation dosimetry of 89Zr labeled antibody estimated using the MIRD method and MCNP code. NUCLEAR ENGINEERING AND TECHNOLOGY 2022. [DOI: 10.1016/j.net.2022.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Antibody-Drug Conjugates in Breast Cancer: What Is Beyond HER2? Cancer J 2022; 28:436-445. [DOI: 10.1097/ppo.0000000000000629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Ribeiro R, Carvalho MJ, Goncalves J, Moreira JN. Immunotherapy in triple-negative breast cancer: Insights into tumor immune landscape and therapeutic opportunities. Front Mol Biosci 2022; 9:903065. [PMID: 36060249 PMCID: PMC9437219 DOI: 10.3389/fmolb.2022.903065] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a clinically aggressive subtype of breast cancer that represents 15-20% of breast tumors and is more prevalent in young pre-menopausal women. It is the subtype of breast cancers with the highest metastatic potential and recurrence at the first 5 years after diagnosis. In addition, mortality increases when a complete pathological response is not achieved. As TNBC cells lack estrogen, progesterone, and HER2 receptors, patients do not respond well to hormone and anti-HER2 therapies, and conventional chemotherapy remains the standard treatment. Despite efforts to develop targeted therapies, this disease continues to have a high unmet medical need, and there is an urgent demand for customized diagnosis and therapeutics. As immunotherapy is changing the paradigm of anticancer treatment, it arises as an alternative treatment for TNBC patients. TNBC is classified as an immunogenic subtype of breast cancer due to its high levels of tumor mutational burden and presence of immune cell infiltrates. This review addresses the implications of these characteristics for the diagnosis, treatment, and prognosis of the disease. Herein, the role of immune gene signatures and tumor-infiltrating lymphocytes as biomarkers in TNBC is reviewed, identifying their application in patient diagnosis and stratification, as well as predictors of efficacy. The expression of PD-L1 expression is already considered to be predictive of response to checkpoint inhibitor therapy, but the challenges regarding its value as biomarker are described. Moreover, the rationales for different formats of immunotherapy against TNBC currently under clinical research are discussed, and major clinical trials are highlighted. Immune checkpoint inhibitors have demonstrated clinical benefit, particularly in early-stage tumors and when administered in combination with chemotherapy, with several regimens approved by the regulatory authorities. The success of antibody-drug conjugates and research on other emerging approaches, such as vaccines and cell therapies, will also be addressed. These advances give hope on the development of personalized, more effective, and safe treatments, which will improve the survival and quality of life of patients with TNBC.
Collapse
Affiliation(s)
- Rita Ribeiro
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Coimbra, Portugal
- iMed.ULisboa—Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
| | - Maria João Carvalho
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
- CHUC—Coimbra Hospital and University Centre, Department of Gynaecology, Coimbra, Portugal
- Univ Coimbra—University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- iCBR—Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - João Goncalves
- iMed.ULisboa—Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - João Nuno Moreira
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Coimbra, Portugal
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
18
|
Capici S, Ammoni LC, Meli N, Cogliati V, Pepe FF, Piazza F, Cazzaniga ME. Personalised Therapies for Metastatic Triple-Negative Breast Cancer: When Target Is Not Everything. Cancers (Basel) 2022; 14:cancers14153729. [PMID: 35954393 PMCID: PMC9367432 DOI: 10.3390/cancers14153729] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The purpose of the present review is to shed light on new molecular biomarkers in triple-negative breast cancer (TNBC), showing emerging therapeutic approaches related to specific molecular signatures and their mechanisms of action. A general overview of ongoing clinical trials, future perspectives and differences in approval by American and European regulatory authorities is provided. Abstract Triple-negative breast cancer—defined by the absence of oestrogen/progesterone receptors and human epidermal growth factor receptor 2 expression—is a complex and heterogeneous type of tumour characterised by poor prognosis, aggressive behaviour and lack of effective therapeutic strategies. The identification of new biomarkers and molecular signatures is leading to development of new therapeutic strategies including immunotherapy, targeted therapy and antibody-drug conjugates (ADCs). Against a background where chemotherapy has always been considered the standard of care, evolution towards a precision medicine approach could improve TNBC clinical practice in a complex scenario, with many therapeutic options and new drugs. The aim of this review was to focus on emerging therapeutic targets and their related specific therapy, discussing available and emerging drugs, underlining differences in approval by American and European regulatory authorities and showing the future perspective in the large number of ongoing clinical trials.
Collapse
Affiliation(s)
- Serena Capici
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Luca Carlofrancesco Ammoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
- Correspondence:
| | - Nicole Meli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| | - Viola Cogliati
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Francesca Fulvia Pepe
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
| | - Francesca Piazza
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| | - Marina Elena Cazzaniga
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.); (F.F.P.); (M.E.C.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (N.M.); (F.P.)
| |
Collapse
|
19
|
Tuo X, Zhou Y, Yang X, Ma S, Liu D, Zhang X, Hou H, Wang R, Li X, Zhao L. miR-532-3p suppresses proliferation and invasion of ovarian cancer cells via GPNMB/HIF-1α/HK2 axis. Pathol Res Pract 2022; 237:154032. [PMID: 35914373 DOI: 10.1016/j.prp.2022.154032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Identifying a new target of miR-532-3p and studying its functional mechanism to explore the detailed anti-tumor mechanism of miR-532-3p in ovarian cancer. METHODS Biological and molecular methods including real-time quantitative PCR (RT-qPCR), Western blotting, colony formation, in vitro migration and invasion assays, glucose consumption and lactate production assays, RNA interference and tumor xenograft mouse models were used to study the role of miR-532-3p and its target in ovarian cancer. mRNA sequencing, dual-luciferase reporter assay and immunohistochemistry (IHC) were used to identify miR-532-3p target. STRING dataset analysis, qPCR and Western blotting were used to investigate the downstream pathway of the target of miR-532-3p. RESULTS Forced expression of miR-532-3p inhibited the proliferation, migration and invasion of ovarian cancer cells in vitro and the tumor growth in nude mice. RNA sequencing found 299 mRNAs were downregulated in miR-532-3p-overexpressed ovarian cancer cells, and bioinformatic analysis indicated Glycoprotein Nonmetastatic Melanoma Protein B (GPNMB), a type I membrane glycoprotein, was the potential target of miR-532-3p. GPNMB was reduced at both RNA and protein levels in miR-532-3p-overexpressed ovarian cancer cells. Dual-luciferase reporter assay determined GPNMB as the target of miR-532-3p. Interference of GPNMB inhibited the proliferation, migration, invasion, glucose consumption and lactate production of ovarian cancer cells. Knocking down of GPNMB reduced the protein level of HIF-1α without affecting HIF-1α mRNA level. Overexpression of GPNMB reversed the antitumor effect of miR-532-3p. CONCLUSION miR-532-3p exerted the anti-cancer effect by targeting GPNMB/ HIF-1α/ HK2 pathway to inhibit aerobic glycolysis in ovarian cancer.
Collapse
Affiliation(s)
- Xiaoqian Tuo
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yuanyuan Zhou
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiling Yang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Sijia Ma
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Dan Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Xiaoling Zhang
- Department of Obstetrics and Gynecology, Xi'an Daxing Hospital, Xi'an, Shaanxi 710002, PR China
| | - Huilian Hou
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Ruiqi Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, Shaanxi 710061, PR China
| | - Xu Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, Shaanxi 710061, PR China.
| | - Le Zhao
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
20
|
Wang SH, Wu CH, Tsai CC, Chen TY, Tsai KJ, Hung CM, Hsu CY, Wu CW, Hsieh TH. Effects of Luteolin on Human Breast Cancer Using Gene Expression Array: Inferring Novel Genes. Curr Issues Mol Biol 2022; 44:2107-2121. [PMID: 35678671 PMCID: PMC9164068 DOI: 10.3390/cimb44050142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 11/24/2022] Open
Abstract
Taraxacum officinale (dandelion) is often used in traditional Chinese medicine for the treatment of cancer; however, the downstream regulatory genes and signaling pathways mediating its effects on breast cancer remain unclear. The present study aimed to explore the effects of luteolin, the main biologically active compound of T. officinale, on gene expression profiles in MDA-MB-231 and MCF-7 breast cancer cells. The results revealed that luteolin effectively inhibited the proliferation and motility of the MDA-MB-231 and MCF-7 cells. The mRNA expression profiles were determined using gene expression array analysis and analyzed using a bioinformatics approach. A total of 41 differentially expressed genes (DEGs) were found in the luteolin-treated MDA-MB-231 and MCF-7 cells. A Gene Ontology analysis revealed that the DEGs, including AP2B1, APP, GPNMB and DLST, mainly functioned as oncogenes. The human protein atlas database also found that AP2B1, APP, GPNMB and DLST were highly expressed in breast cancer and that AP2B1 (cut-off value, 75%) was significantly associated with survival rate (p = 0.044). In addition, a Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the DEGs were involved in T-cell leukemia virus 1 infection and differentiation. On the whole, the findings of the present study provide a scientific basis that may be used to evaluate the potential benefits of luteolin in human breast cancer. Further studies are required, however, to fully elucidate the role of the related molecular pathways.
Collapse
Affiliation(s)
- Shih-Ho Wang
- Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
- Division of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chin-Hu Wu
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-H.W.); (C.-Y.H.)
| | - Chin-Chuan Tsai
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; (C.-C.T.); (T.-Y.C.)
| | - Tai-Yu Chen
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan; (C.-C.T.); (T.-Y.C.)
| | - Kuen-Jang Tsai
- Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (K.-J.T.); (C.-M.H.)
| | - Chao-Ming Hung
- Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (K.-J.T.); (C.-M.H.)
| | - Chia-Yi Hsu
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-H.W.); (C.-Y.H.)
| | - Chia-Wei Wu
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
| | - Tsung-Hua Hsieh
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
- Correspondence: ; Tel.: +886-7-6151100 (ext. 5072)
| |
Collapse
|
21
|
Suda M, Shimizu I, Katsuumi G, Hsiao CL, Yoshida Y, Matsumoto N, Yoshida Y, Katayama A, Wada J, Seki M, Suzuki Y, Okuda S, Ozaki K, Nakanishi-Matsui M, Minamino T. Glycoprotein nonmetastatic melanoma protein B regulates lysosomal integrity and lifespan of senescent cells. Sci Rep 2022; 12:6522. [PMID: 35444208 PMCID: PMC9021310 DOI: 10.1038/s41598-022-10522-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/30/2022] [Indexed: 12/31/2022] Open
Abstract
Accumulation of senescent cells in various tissues has been reported to have a pathological role in age-associated diseases. Elimination of senescent cells (senolysis) was recently reported to reversibly improve pathological aging phenotypes without increasing rates of cancer. We previously identified glycoprotein nonmetastatic melanoma protein B (GPNMB) as a seno-antigen specifically expressed by senescent human vascular endothelial cells and demonstrated that vaccination against Gpnmb eliminated Gpnmb-positive senescent cells, leading to an improvement of age-associated pathologies in mice. The aim of this study was to elucidate whether GPNMB plays a role in senescent cells. We examined the potential role of GPNMB in senescent cells by testing the effects of GPNMB depletion and overexpression in vitro and in vivo. Depletion of GPNMB from human vascular endothelial cells shortened their replicative lifespan and increased the expression of negative cell cycle regulators. Conversely, GPNMB overexpression protected these cells against stress-induced premature senescence. Depletion of Gpnmb led to impairment of vascular function and enhanced atherogenesis in mice, whereas overexpression attenuated dietary vascular dysfunction and atherogenesis. GPNMB was upregulated by lysosomal stress associated with cellular senescence and was a crucial protective factor in maintaining lysosomal integrity. GPNMB is a seno-antigen that acts as a survival factor in senescent cells, suggesting that targeting seno-antigens such as GPNMB may be a novel strategy for senolytic treatments.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Chieh Lun Hsiao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Advanced Senotherapeautics, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Naomi Matsumoto
- Division of Biochemistry, School of Pharmacy, Iwate Medical University, Iwate, 028-3694, Japan
| | - Yutaka Yoshida
- Department of Structural Pathology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Akihiro Katayama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8561, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8561, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Kazuyuki Ozaki
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Mayumi Nakanishi-Matsui
- Division of Biochemistry, School of Pharmacy, Iwate Medical University, Iwate, 028-3694, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan.
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
22
|
Manevich L, Okita Y, Okano Y, Sugasawa T, Kawanishi K, Poullikkas T, Dang Cao LTL, Zheng L, Nakayama M, Matsumoto S, Tabuchi K, Kato M. Glycoprotein NMB promotes tumor formation and malignant progression of laryngeal squamous cell carcinoma. Cancer Sci 2022; 113:3244-3254. [PMID: 35365934 PMCID: PMC9459245 DOI: 10.1111/cas.15359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC), although one of the most common head and neck cancers, has a static or slightly decreased survival rate because of difficulties in early diagnosis, lack of effective molecular targeting therapy, and severe dysfunction after radical surgical treatments. Therefore, a novel therapeutic target is crucial to increase treatment efficacy and survival rates in these patients. Glycoprotein NMB (GPNMB), whose role in LSCC remains elusive, is a type 1 transmembrane protein involved in malignant progression of various cancers, and its high expression is thought to be a poor prognostic factor. In this study, we showed that GPNMB expression levels in LSCC samples are significantly higher than those in normal tissues, and GPNMB expression is observed mostly in growth‐arrested cancer cells. Furthermore, knockdown of GPNMB reduces monolayer cellular proliferation, cellular migration, and tumorigenic growth, while GPNMB protein displays an inverse relationship with Ki‐67 levels. Therefore, we conclude that GPNMB may be an attractive target for future LSCC therapy.
Collapse
Affiliation(s)
- Lev Manevich
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yukari Okita
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Yasuhito Okano
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan.,Otolaryngology, Head and Neck Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takehito Sugasawa
- Laboratory of Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kunio Kawanishi
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Thanasis Poullikkas
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Human Biology, School of integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Linda T L Dang Cao
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Ph.D. Program in Humanics, School of Integrative and Global Majors (SIGMA), University of Tsukuba, Ibaraki, Japan.,Life and Science Center of Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan
| | - Ling Zheng
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masahiro Nakayama
- Otolaryngology, Head and Neck Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shin Matsumoto
- Otolaryngology, Head and Neck Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Keiji Tabuchi
- Otolaryngology, Head and Neck Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Mitsuyasu Kato
- Experimental Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Division of Cell Dynamics, Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
23
|
Ziad A, Abdurahman A, Misako N. A comprehensive review on antibody-drug conjugates (ADCs) in the treatment landscape of non-small cell lung cancer (NSCLC). Cancer Treat Rev 2022; 106:102393. [DOI: 10.1016/j.ctrv.2022.102393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/02/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022]
|
24
|
Fertal SA, Poterala JE, Ponik SM, Wisinski KB. Stromal Characteristics and Impact on New Therapies for Metastatic Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1238. [PMID: 35267548 PMCID: PMC8909697 DOI: 10.3390/cancers14051238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
The heterogenous nature of triple-negative breast cancer (TNBC) is an underlying factor in therapy resistance, metastasis, and overall poor patient outcome. The lack of hormone and growth factor receptors lends to the use of chemotherapy as the first-line treatment for TNBC. However, the failure of chemotherapy demonstrates the need to develop novel immunotherapies, antibody-drug conjugates (ADCs), and other tumor- and stromal-targeted therapeutics for TNBC patients. The potential for stromal-targeted therapy is driven by studies indicating that the interactions between tumor cells and the stromal extracellular matrix (ECM) activate mechanisms of therapy resistance. Here, we will review recent outcomes from clinical trials targeting metastatic TNBC with immunotherapies aimed at programed death ligand-receptor interactions, and ADCs specifically linked to trophoblast cell surface antigen 2 (Trop-2). We will discuss how biophysical and biochemical cues from the ECM regulate the pathophysiology of tumor and stromal cells toward a pro-tumor immune environment, therapy resistance, and poor TNBC patient outcome. Moreover, we will highlight how ECM-mediated resistance is motivating the development of new stromal-targeted therapeutics with potential to improve therapy for this disease.
Collapse
Affiliation(s)
- Shelby A. Fertal
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Johanna E. Poterala
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| | - Suzanne M. Ponik
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kari B. Wisinski
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
25
|
Biondini M, Kiepas A, El-Houjeiri L, Annis MG, Hsu BE, Fortier AM, Morin G, Martina JA, Sirois I, Aguilar-Mahecha A, Gruosso T, McGuirk S, Rose AAN, Tokat UM, Johnson RM, Sahin O, Bareke E, St-Pierre J, Park M, Basik M, Majewski J, Puertollano R, Pause A, Huang S, Keler T, Siegel PM. HSP90 inhibitors induce GPNMB cell-surface expression by modulating lysosomal positioning and sensitize breast cancer cells to glembatumumab vedotin. Oncogene 2022; 41:1701-1717. [PMID: 35110681 DOI: 10.1038/s41388-022-02206-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/29/2021] [Accepted: 01/20/2022] [Indexed: 12/18/2022]
Abstract
Transmembrane glycoprotein NMB (GPNMB) is a prognostic marker of poor outcome in patients with triple-negative breast cancer (TNBC). Glembatumumab Vedotin, an antibody drug conjugate targeting GPNMB, exhibits variable efficacy against GPNMB-positive metastatic TNBC as a single agent. We show that GPNMB levels increase in response to standard-of-care and experimental therapies for multiple breast cancer subtypes. While these therapeutic stressors induce GPNMB expression through differential engagement of the MiTF family of transcription factors, not all are capable of increasing GPNMB cell-surface localization required for Glembatumumab Vedotin inhibition. Using a FACS-based genetic screen, we discovered that suppression of heat shock protein 90 (HSP90) concomitantly increases GPNMB expression and cell-surface localization. Mechanistically, HSP90 inhibition resulted in lysosomal dispersion towards the cell periphery and fusion with the plasma membrane, which delivers GPNMB to the cell surface. Finally, treatment with HSP90 inhibitors sensitizes breast cancers to Glembatumumab Vedotin in vivo, suggesting that combination of HSP90 inhibitors and Glembatumumab Vedotin may be a viable treatment strategy for patients with metastatic TNBC.
Collapse
Affiliation(s)
- Marco Biondini
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Alex Kiepas
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Leeanna El-Houjeiri
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Matthew G Annis
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Brian E Hsu
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Anne-Marie Fortier
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Geneviève Morin
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Isabelle Sirois
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada
| | - Adriana Aguilar-Mahecha
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada
| | - Tina Gruosso
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Shawn McGuirk
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - April A N Rose
- Department of Oncology and Surgery, McGill University, Montreal, QC, Canada
| | - Unal M Tokat
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | | | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, USA
| | - Eric Bareke
- Genome Québec Innovation Center, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Morag Park
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Mark Basik
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC, Canada.,Department of Oncology and Surgery, McGill University, Montreal, QC, Canada
| | - Jacek Majewski
- Genome Québec Innovation Center, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Arnim Pause
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Sidong Huang
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | | | - Peter M Siegel
- Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada. .,Department of Medicine, McGill University, Montreal, QC, Canada. .,Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
26
|
TROP-2, Nectin-4, GPNMB, and B7-H3 Are Potentially Therapeutic Targets for Anaplastic Thyroid Carcinoma. Cancers (Basel) 2022; 14:cancers14030579. [PMID: 35158847 PMCID: PMC8833363 DOI: 10.3390/cancers14030579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anaplastic thyroid carcinoma is a highly aggressive thyroid tumor with a poor prognosis. There are limited choices for the effective treatment of this type of carcinoma. Whether the targets of antibody–drug conjugates are expressed in anaplastic thyroid carcinoma remains unclear. Therefore, we examined expression rates of the following antibody–drug conjugate targets using the tissue microarrays of anaplastic thyroid carcinomas: human epidermal growth factor receptor 2, nectin-4, trophoblast cell surface antigen 2, glycoprotein non-metastatic B, and B7-H3. We found that glycoprotein non-metastatic B and B7-H3 were expressed in most anaplastic thyroid carcinoma tissues. Trophoblast cell surface antigen 2 and nectin-4 were expressed in 65% and 59% of anaplastic thyroid carcinoma tissues, respectively. Trophoblast cell surface antigen 2 was high expressed in anaplastic thyroid carcinoma undifferentiated from papillary thyroid carcinoma. In contrast, nectin-4 expression was high in patients with de novo anaplastic thyroid carcinoma. These cell membrane proteins are potential therapeutic targets for anaplastic thyroid carcinoma. Abstract Background: Anaplastic thyroid carcinoma (ATC) is a highly aggressive thyroid tumor with a poor prognosis. However, there are limited choices for ATC treatment. Recently, the effectiveness of antibody–drug conjugates has been demonstrated in various carcinomas. Whether the targets of antibody–drug conjugates are expressed in anaplastic thyroid carcinoma remains unclear. Methods: Fifty-four patients with ATC were enrolled in this study. Tissue microarrays were constructed using the archives of formalin-fixed paraffin-embedded tissue blocks. All sections were stained with the following antibody–drug conjugate targets: human epidermal growth factor receptor 2 (HER2), nectin-4, trophoblast cell surface antigen 2 (TROP-2), glycoprotein non-metastatic B (GPNMB), and B7-H3. Results: HER2 was negative in all tissues, whereas GPNMB and B7-H3 were expressed in most ATC tissues. TROP-2 and nectin-4 were expressed in 65% and 59% of ATC tissues, respectively. TROP-2 was expressed at significantly higher levels in ATC undifferentiated from papillary thyroid carcinoma than in ATC undifferentiated from follicular thyroid carcinoma and de novo ATC. In contrast, nectin-4 expression was markedly higher in patients with de novo ATC than in those with papillary and follicular thyroid carcinoma. Conclusions: TROP-2 and nectin-4 are potential therapeutic targets for ATC undifferentiated from papillary thyroid carcinoma and de novo ATC, respectively. GPNMB and B7-H3 potential for treating all types of ATC.
Collapse
|
27
|
Lee S, Cavaliere A, Gallezot JD, Keler T, Michelhaugh SK, Belitzky E, Liu M, Mulnix T, Maher SE, Bothwell ALM, Li F, Phadke M, Mittal S, Marquez-Nostra B. [89Zr]ZrDFO-CR011 positron emission tomography correlates with response to glycoprotein non-metastatic melanoma B-targeted therapy in triple negative breast cancer. Mol Cancer Ther 2022; 21:440-447. [PMID: 35027482 DOI: 10.1158/1535-7163.mct-21-0590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/14/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022]
Abstract
There is a need for prognostic markers to select patients most likely to benefit from antibody drug conjugate (ADC) therapy. We quantified the relationship between pre-treatment positron emission tomography (PET) imaging of glycoprotein non-metastatic melanoma B (gpNMB) with 89Zr-labeled anti-gpNMB antibody ([89Zr]ZrDFO-CR011) and response to ADC therapy (CDX-011) in triple negative breast cancer (TNBC). First, we compared different PET imaging metrics and found that standardized uptake values (SUV) and tumor-to-heart SUV ratios (SUVR) were sufficient to delineate differences in radiotracer uptake in the tumor of four different cell- and patient-derived tumor models and achieved high standardized effect sizes. These tumor models with varying levels of gpNMB expression were imaged with [89Zr]ZrDFO-CR011 followed by treatment with a single bolus injection of CDX-011. The percent change in tumor volume relative to baseline (% CTV) was then correlated with SUVmean of [89Zr]ZrDFO-CR011 uptake in the tumor. All gpNMB-positive tumor models responded to CDX-011 over 6 weeks of treatment, except one patient-derived tumor re-grew after 4 weeks of treatment. As expected, the gpNMB-negative tumor increased in volume by 130 {plus minus} 59 % at endpoint. The magnitude of pre-treatment SUV had the strongest inverse correlation with the % CTV at 2 - 4 weeks after treatment with CDX-011 (Spearman ρ = -0.8). However, pre-treatment PET imaging with [89Zr]ZrDFO-CR011 did not inform on which tumor types will re-grow over time. Other methods will be needed to predict resistance to treatment.
Collapse
Affiliation(s)
- Supum Lee
- Radiology and Biomedical Imaging, Yale University
| | | | | | | | | | | | - Michael Liu
- Radiology and Biomedical Imaging, Yale University
| | | | | | | | - Fangyong Li
- Yale Center for Analytic Sciences, Yale University School of Public Health
| | - Manali Phadke
- Yale Center for Analytical Sciences, Yale School of Medicine
| | - Sandeep Mittal
- Neurosurgery, Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine
| | | |
Collapse
|
28
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
29
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
30
|
Zhai JP, Liu ZH, Wang HD, Huang GL, Man LB. GPNMB overexpression is associated with extensive bone metastasis and poor prognosis in renal cell carcinoma. Oncol Lett 2021; 23:36. [PMID: 34966452 DOI: 10.3892/ol.2021.13154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Glycoprotein non-metastatic protein B (GPNMB) promotes bone metastasis (BM) in various types of cancer. However, GPNMB expression and its function in patients with renal cell carcinoma (RCC) and BM is still unknown. Therefore, the clinical significance of GPNMB and its biological function in RCC with BM was investigated in the present study. A total of 31 patients with RCC and BM were retrospectively collected. The association between GPNMB protein expression level on the primary tumor and the clinicopathological characteristics of the patients was analyzed. Kaplan-Meier analysis was used to investigate the association between GPNMB expression and the prognosis of the patients. The effects of GPNMB inhibition on cell proliferation, migration and invasion in RCC cells were investigated using short hairpin (sh)RNA. High GPNMB expression level was significantly associated with the number (P=0.001) and the extent of BM (P=0.001), Fuhrman grade (P=0.037), and ERK expression level (P=0.003) of the primary tumor. In addition, GPNMB overexpression was significantly associated with poor prognosis with respect to overall survival time (P=0.001). Furthermore, a specific shRNA sequence targeting the GPNMB gene was constructed and transduced into the ACHN cell line, using a lentivirus vector to obtain a stable cell line with low mRNA expression level of GPNMB. Low GPNMB expression level inhibited RCC cell proliferation, which was measured using a Cell Counting Kit-8 assay. Cell migration and invasion ability was significantly decreased in GPNMB knockdown RCC cells compared with that in cells transduced with the negative control shRNA. In addition, the protein expression levels of phosphorylated ERK were lower in the GPNMB shRNA-transduced ACHN cells compared with those in the control cells. Therefore, these results suggested that GPNMB plays an important role in tumor progression in RCC with BM. Furthermore, it might serve as a predictive marker for BM and as a poor prognostic factor in RCC with BM. GPNMB downregulation suppressed the proliferation, migration and invasion of the RCC cells, which may be mediated through the inhibition of the ERK signaling pathway.
Collapse
Affiliation(s)
- Jian-Po Zhai
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Zhen-Hua Liu
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Hai-Dong Wang
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Guang-Lin Huang
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| | - Li-Bo Man
- Department of Urology, Beijing Jishuitan Hospital, Beijing 102200, P.R. China
| |
Collapse
|
31
|
Guo Z, Xing R, Zhao M, Li Y, Lu H, Liu Z. Controllable Engineering and Functionalizing of Nanoparticles for Targeting Specific Proteins towards Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101713. [PMID: 34725943 PMCID: PMC8693047 DOI: 10.1002/advs.202101713] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/15/2021] [Indexed: 05/14/2023]
Abstract
Nanoparticles have been widely used in important biomedical applications such as imaging, drug delivery, and disease therapy, in which targeting toward specific proteins is often essential. However, current targeting strategies mainly rely on surface modification with bioligands, which not only often fail to provide desired properties but also remain challenging. Here an unprecedented approach is reported, called reverse microemulsion-confined epitope-oriented surface imprinting and cladding (ROSIC), for facile, versatile, and controllable engineering coreless and core/shell nanoparticles with tunable monodispersed size as well as specific targeting capability toward proteins and peptides. Via engineering coreless imprinted and cladded silica nanoparticles, the effectiveness and superiority over conventional imprinting of the proposed approach are first verified. The prepared nanoparticles exhibit both high specificity and high affinity. Using quantum dots, superparamagnetic nanoparticles, silver nanoparticles, and upconverting nanoparticles as a representative set of core substrates, a variety of imprinted and cladded single-core/shell nanoparticles are then successfully prepared. Finally, using imprinted and cladded fluorescent nanoparticles as probes, in vitro targeted imaging of triple-negative breast cancer (TNBC) cells and in vivo targeted imaging of TNBC-bearing mice are achieved. This approach opens a new avenue to engineering of nanoparticles for targeting specific proteins, holding great prospects in biomedical applications.
Collapse
Affiliation(s)
- Zhanchen Guo
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Rongrong Xing
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Menghuan Zhao
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Ying Li
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Haifeng Lu
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| |
Collapse
|
32
|
Khan SA, Sun Z, Dahlberg S, Malhotra J, Keresztes R, Ikpeazu C, Ma P, Ramalingam SS, Pillai R. Efficacy and Safety of Glembatumumab Vedotin in Patients With Advanced or Metastatic Squamous Cell Carcinoma of the Lung (PrECOG 0504). JTO Clin Res Rep 2021; 2:100166. [PMID: 34590018 PMCID: PMC8474292 DOI: 10.1016/j.jtocrr.2021.100166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction Glycoprotein NMB is a transmembrane protein linked with poor prognosis and is expressed in most squamous lung cancer. Glembatumumab vedotin is an antibody-drug conjugate targeting glycoprotein NMB, administered intravenously every 3 weeks in this phase 1 study to determine the safety, tolerability, and maximum tolerated dose in patients who had progressed on any number of previous therapies. Results A total of 13 patients were enrolled; adverse events (of any grade) including dyspnea, neutropenia, respiratory failure, anemia, increased aspartate transaminase/alanine transaminase, diarrhea, and hypophosphatemia were seen in 15% of patients. Grade 5 events included two cases of respiratory failure, either completely or partially attributed to cancer progression. The only other grade 5 event was “disease progression.” The most common adverse events (23%) were decreased appetite, fatigue, rash, and weight loss. The median overall and progression-free survivals were 5.7 months (90% confidence interval: 2.5–16.8) and 2.5 months (90% confidence interval: 1.6–5.8) respectively. Conclusions Glembatumumab vedotin exhibited no serious or unexpected toxicity in this heavily pretreated population, except those caused by disease progression. Modest anticancer activity was observed with a recommendation for a phase 2 dose of 1.9 mg/kg. This portion of the study was not undertaken owing to the company’s decision to discontinue drug development.
Collapse
Affiliation(s)
- Saad A. Khan
- Harold C. Simmons Comprehensive Cancer Center, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Current Affiliation: Cancer Institute, Stanford Medicine, Stanford University, Stanford, California
- Corresponding author. Address for correspondence: Saad A. Khan, MD, Division of Oncology, Department of Medicine, 875 Blake Wilbur Avenue, Stanford, CA 94304.
| | - Zhuoxin Sun
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Suzanne Dahlberg
- Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, Massachusetts
- Current Affiliation: Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jyoti Malhotra
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Roger Keresztes
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York
| | - Chukwuemeka Ikpeazu
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick Ma
- Department of Medicine, Penn State University, State College, Pennsylvania
| | - Suresh S. Ramalingam
- Department of Hematology/Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rathi Pillai
- Department of Hematology/Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
33
|
Allavena P, Digifico E, Belgiovine C. Macrophages and cancer stem cells: a malevolent alliance. Mol Med 2021; 27:121. [PMID: 34583655 PMCID: PMC8480058 DOI: 10.1186/s10020-021-00383-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Myeloid cells infiltrating tumors are gaining ever growing attention in the last years because their pro-tumor and immunosuppressive functions are relevant for disease progression and therapeutic responses. The functional ambiguity of tumor-associated macrophages (TAMs), mostly promoting tumor evolution, is a challenging hurdle. This is even more evident in the case of cancer stem cells (CSCs); as active participants in the specialized environment of the cancer stem cell niche, TAMs initiate a reciprocal conversation with CSCs. TAMs contribute to protect CSCs from the hostile environment (exogenous insults, toxic compounds, attacks from the immune cells), and produce several biologically active mediators that modulate crucial developmental pathways that sustain cancer cell stemness. In this review, we have focused our attention on the interaction between TAMs and CSCs; we describe how TAMs impact on CSC biology and, in turn, how CSCs exploit the tissue trophic activity of macrophages to survive and progress. Since CSCs are responsible for therapy resistance and tumor recurrence, they are important therapeutic targets. In view of the recent success in oncology obtained by stimulating the immune system, we discuss some macrophage-targeted therapeutic strategies that may also affect the CSCs and interrupt their malevolent alliance.
Collapse
Affiliation(s)
- Paola Allavena
- Humanitas Clinical and Research Center -IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy.
| | - Elisabeth Digifico
- Humanitas Clinical and Research Center -IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Cristina Belgiovine
- Humanitas Clinical and Research Center -IRCCS, via Manzoni 56, 20089, Rozzano, MI, Italy
| |
Collapse
|
34
|
Wang Z, Ran X, Qian S, Hou H, Dong M, Wu S, Ding M, Zhang Y, Zhang X, Zhang M, Chen Q. GPNMB promotes the progression of diffuse large B cell lymphoma via YAP1-mediated activation of the Wnt/β-catenin signaling pathway. Arch Biochem Biophys 2021; 710:108998. [PMID: 34280359 DOI: 10.1016/j.abb.2021.108998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 01/06/2023]
Abstract
Glycoprotein non-metastatic melanoma protein B (GPNMB) has been confirmed to be related to the pathogenesis of tumors. However, the potential impact of GPNMB on the progression of diffuse large B-cell lymphoma (DLBCL) is unclear. In this study, the expression levels of GPNMB and Yes-associated protein (YAP) were analyzed using qRT-PCT and Western blot assay. Cell counting kit-8, EdU, and flow cytometry assays were used to detect the proliferation and apoptosis of DLBCL cells. A nude mice xenograft model was established for in vivo research. Results showed that GPNMB and YAP1 were upregulated in DLBCL cell lines. Knockdown of GPNMB inhibited cell proliferation and promoted apoptosis in DLBCL cells. Additionally, the expression levels of YAP1 and the downstream effector of Hippo pathway (c-myc) were markedly decreased when GPNMB was knocked down. Moreover, knockdown of GPNMB inhibited the nuclear translocation of β-catenin protein, which could be abolished by YAP1 overexpression. Simultaneously, the anti-proliferative and pro-apoptotic effects of GPNMB knockdown could be reversed by YAP1 overexpression or LiCl (the activator of Wnt/β-catenin pathway). Furthermore, the mice xenograft model confirmed that inhibition of GPNMB restrained the tumorigenesis of DLBCL in vivo. In conclusion, GPNMB could partly activate the Wnt/β-catenin signaling pathway by targeting YAP1, so as to participate in tumorigenesis of DLBCL.
Collapse
Affiliation(s)
- Zeyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Xianting Ran
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, China
| | - Siyu Qian
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Huting Hou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Meng Dong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Shaoxuan Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Mengjie Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Yue Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China
| | - Qingjiang Chen
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, China.
| |
Collapse
|
35
|
O'Neill RS, Stoita A. Biomarkers in the diagnosis of pancreatic cancer: Are we closer to finding the golden ticket? World J Gastroenterol 2021; 27:4045-4087. [PMID: 34326612 PMCID: PMC8311531 DOI: 10.3748/wjg.v27.i26.4045] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/24/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer related mortality on a global scale. The disease itself is associated with a dismal prognosis, partly due to its silent nature resulting in patients presenting with advanced disease at the time of diagnosis. To combat this, there has been an explosion in the last decade of potential candidate biomarkers in the research setting in the hope that a diagnostic biomarker may provide a glimmer of hope in what is otherwise quite a substantial clinical dilemma. Currently, serum carbohydrate antigen 19-9 is utilized in the diagnostic work-up of patients diagnosed with PC however this biomarker lacks the sensitivity and specificity associated with a gold-standard marker. In the search for a biomarker that is both sensitive and specific for the diagnosis of PC, there has been a paradigm shift towards a focus on liquid biopsy and the use of diagnostic panels which has subsequently proved to have efficacy in the diagnosis of PC. Currently, promising developments in the field of early detection on PC using diagnostic biomarkers include the detection of microRNA (miRNA) in serum and circulating tumour cells. Both these modalities, although in their infancy and yet to be widely accepted into routine clinical practice, possess merit in the early detection of PC. We reviewed over 300 biomarkers with the aim to provide an in-depth summary of the current state-of-play regarding diagnostic biomarkers in PC (serum, urinary, salivary, faecal, pancreatic juice and biliary fluid).
Collapse
Affiliation(s)
- Robert S O'Neill
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| | - Alina Stoita
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| |
Collapse
|
36
|
Huang YH, Chu PY, Chen JL, Huang CT, Huang CC, Tsai YF, Wang YL, Lien PJ, Tseng LM, Liu CY. Expression pattern and prognostic impact of glycoprotein non-metastatic B (GPNMB) in triple-negative breast cancer. Sci Rep 2021; 11:12171. [PMID: 34108545 PMCID: PMC8190094 DOI: 10.1038/s41598-021-91588-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 05/28/2021] [Indexed: 12/31/2022] Open
Abstract
Glycoprotein non-metastatic B (GPNMB) is a transmembrane protein overexpressed in numerous cancers including triple-negative breast cancers (TNBC). It has been linked to promote cancer aggressiveness and implicated as a novel target for GPNMB-expressing cancers. In current study, we aimed to explore the clinical significance of GPNMB in TNBC. Among 759 specimens, immunohistochemistry (IHC) exhibited GPNMB expressions were variable in different subtypes and significantly higher in TNBC. Kaplan-Meier analysis revealed GPNMB overexpression in TNBC was associated with worse prognosis especially distant metastasis (P = 0.020, HR = 2.515, CI 1.154-5.480). Multivariate analysis showed GPNMB expression was an independent prognostic factor in terms of recurrence and distant metastasis (P = 0.008, HR = 3.22, CI 1.36-7.61; P = 0.017, HR = 3.08, CI 1.22-7.74). In silico analysis showed high mRNA expression of GPNMB was associated with distant metastasis and GPNMB was overexpressed in TNBC. Furthermore, GPNMB positively correlated with epithelial-mesenchymal transition (EMT) regulators, mesenchymal marker vimentin, MMP and integrins. The protein levels of Twist and MMP2 were upregulated by GPNMB overexpression in TNBC cells. GPNMB-enhanced cell invasion was attenuated by broad spectrum MMP inhibitor (GM 6001) and the selective inhibitor of MMP-2 (ARP100). In summary, GPNMB expression is prevalent in TNBC and may be implicated as a prognostic biomarker in patients with TNBC.
Collapse
Affiliation(s)
- Yu-Hsiang Huang
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua City, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ji-Lin Chen
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Teng Huang
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Division of Hematology & Oncology, Department of Medicine, Yang-Ming Branch of Taipei City Hospital, Taipei, Taiwan
| | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yi-Fang Tsai
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Ling Wang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Experimental Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pei-Ju Lien
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Nursing, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ling-Ming Tseng
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Experimental Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Yu Liu
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan. .,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan. .,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan.
| |
Collapse
|
37
|
Vahdat LT, Schmid P, Forero-Torres A, Blackwell K, Telli ML, Melisko M, Möbus V, Cortes J, Montero AJ, Ma C, Nanda R, Wright GS, He Y, Hawthorne T, Bagley RG, Halim AB, Turner CD, Yardley DA. Glembatumumab vedotin for patients with metastatic, gpNMB overexpressing, triple-negative breast cancer ("METRIC"): a randomized multicenter study. NPJ Breast Cancer 2021; 7:57. [PMID: 34016993 PMCID: PMC8137923 DOI: 10.1038/s41523-021-00244-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 02/16/2021] [Indexed: 12/09/2022] Open
Abstract
The METRIC study (NCT#0199733) explored a novel antibody–drug conjugate, glembatumumab vedotin (GV), targeting gpNMB that is overexpressed in ~40% of patients with triple-negative breast cancer (TNBC) and associated with poor prognosis. The study was a randomized, open-label, phase 2b study that evaluated progression-free survival (PFS) of GV compared with capecitabine in gpNMB-overexpressing TNBC. Patients who had previously received anthracycline and taxane-based therapy were randomized 2:1 to receive, GV (1.88 mg/kg IV q21 days) or capecitabine (2500 mg/m2 PO daily d1–14 q21 days). The primary endpoint was RECIST 1.1 PFS per independent, blinded central review. In all, 327 patients were randomized to GV (213 treated) or capecitabine (92 treated). Median PFS was 2.9 months for GV vs. 2.8 months for capecitabine. The most common grade ≥3 toxicities for GV were neutropenia, rash, and leukopenia, and for capecitabine were fatigue, diarrhea, and palmar-plantar erythrodysesthesia. The study did not meet the primary endpoint of improved PFS over capecitabine or demonstrate a relative risk/benefit improvement over capecitabine.
Collapse
Affiliation(s)
| | - Peter Schmid
- Center for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| | | | | | | | - Michelle Melisko
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - Javier Cortes
- IOB Institute of Oncology, Quironsalud Group, Madrid & Barcelona, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Cynthia Ma
- Washington University, St. Louis, MO, USA
| | | | - Gail S Wright
- Florida Cancer Specialists, New Port Richey, FL, USA
| | - Yi He
- Celldex Therapeutics, Inc., Hampton, NJ, USA.,AstraZeneca, Gaithersburg, MD, USA
| | | | - Rebecca G Bagley
- Celldex Therapeutics, Inc., Hampton, NJ, USA.,Syndax, Waltham, MA, USA
| | - Abdel-Baset Halim
- Celldex Therapeutics, Inc., Hampton, NJ, USA.,Taiho Oncology, Princeton, NJ, USA
| | - Christopher D Turner
- Celldex Therapeutics, Inc., Hampton, NJ, USA.,Blueprint Medicines, Inc., Cambridge, MA, USA
| | - Denise A Yardley
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN, USA
| |
Collapse
|
38
|
Abstract
Breast cancer, as a heterogeneous disease, includes a wide range of pathological and clinical behaviors. Current treatment protocols, including radiotherapy, chemotherapy, and hormone replacement therapy, are mainly associated with poor response and high rate of recurrence. Therefore, more efforts are needed to develop alternative therapies for this type of cancer. Immunotherapy, as a novel strategy in cancer treatment, has a potential in treating breast cancer patients. Although breast cancer has long been considered problematic to treat with immunotherapy, as it is immunologically "cold," numerous newer preclinical and clinical reports now recommend that immunotherapy has the capability to treat breast cancer patients. In this review, we highlight the different immunotherapy strategies in breast cancer treatment.
Collapse
|
39
|
Shiomi A, Kusuhara M, Sugino T, Sugiura T, Ohshima K, Nagashima T, Urakami K, Serizawa M, Saya H, Yamaguchi K. Comprehensive genomic analysis contrasting primary colorectal cancer and matched liver metastases. Oncol Lett 2021; 21:466. [PMID: 33907576 DOI: 10.3892/ol.2021.12727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
Recent studies have revealed that colorectal cancer (CRC) displays intratumor genetic heterogeneity, and that the cancer microenvironment plays an important role in the proliferation, invasion and metastasis of CRC. The present study performed genomic analysis on paired primary CRC and synchronous colorectal liver metastasis (CRLM) tissues collected from 22 patients using whole-exome sequencing, cancer gene panels and microarray gene expression profiling. In addition, immunohistochemical analysis was used to confirm the protein expression levels of genes identified as highly expressed in CRLM by DNA microarray analysis. The present study identified 10 genes that were highly expressed in CRLM compared with in CRC, from 36,022 probes obtained from primary CRC, CRLM and normal liver tissues by gene expression analysis with DNA microarrays. Of the 10 genes identified, five were classified as encoding 'matricellular proteins' [(osteopontin, periostin, thrombospondin-2, matrix Gla protein (MGP) and glycoprotein nonmetastatic melanoma protein B (GPNMB)] and were selected for immunohistochemical analysis. Osteopontin was strongly expressed in CRLM (6 of 22 cases: 27.3%), but not in CRC (0 of 22: 0%; P=0.02). Periostin also exhibited strong immunoreactivity in CRLM (17 of 22: 68.2%) compared with in CRC (7 of 22: 31.8%; P=0.006). Thrombospondin-2 exhibited strong immunoreactivity in both CRC and CRLM (54.5% in CRC, 45.5% in CRLM; P=0.55). GPNMB and MGP were rarely positive for both CRC and CRLM. A comparison of immunoreactive positive factors for these five genes revealed the complexities of gene expression in CRLM. Of the cases examined, 16 (72.7%) cases of CRC showed zero or only one positive immunoreactive factor. By contrast, CRLM showed more frequent and multiple immunoreactive factors; for example, 16 cases (72.7%) shared two or more factors, which was statistically more frequent than in CRC (P=0.007). The present study revealed the genomic heterogeneity between paired primary CRC and CRLM, in terms of cancer cell microenvironment. This finding may lead to novel diagnostic and therapeutic targets in the era of genome-guided personalized cancer treatment.
Collapse
Affiliation(s)
- Akio Shiomi
- Division of Colon and Rectal Surgery, Shizuoka Cancer Center, Shizuoka 411-8777, Japan.,Division of Gene Regulation Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-858, Japan
| | - Masatoshi Kusuhara
- Regional Resources Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center, Shizuoka 411-8777, Japan
| | - Teiichi Sugiura
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka 411-8777, Japan
| | - Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Takeshi Nagashima
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan.,SRL, Inc., Tokyo 163-0409, Japan
| | - Kenichi Urakami
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Masakuni Serizawa
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Hideyuki Saya
- Division of Gene Regulation Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-858, Japan
| | | |
Collapse
|
40
|
Sukumar J, Gast K, Quiroga D, Lustberg M, Williams N. Triple-negative breast cancer: promising prognostic biomarkers currently in development. Expert Rev Anticancer Ther 2021; 21:135-148. [PMID: 33198517 PMCID: PMC8174647 DOI: 10.1080/14737140.2021.1840984] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer associated with poor prognosis and limited treatment options. Validated prognostic and predictive biomarkers are needed to guide treatment decisions and prognostication.Areas covered: In this review, we discuss established and developing prognostic and predictive biomarkers in TNBC and associated emerging and approved therapies. Biomarkers reviewed include epidermal growth factor receptor (EGFR), vascular endothelial growth factors (VEGF), fibroblast growth factor receptor (FGFR), human epidermal growth factor receptor 2 (HER2), androgen receptor, NOTCH signaling, oxidative stress/redox signaling, microRNAs, TP53 mutation, breast cancer susceptibility gene 1 or 2 (BRCA1/2) mutation/homologous recombination deficiency (HRD), NTRK gene fusion, PI3K/AKT/mTOR, immune biomarkers (programmed death-ligand 1 (PDL1), tumor-infiltrating lymphocytes (TILs), tumor mutational burden (TMB), neoantigens, defects in DNA mismatch repair proteins (dMMR)/microsatellite instability-high (MSI-H)), circulating tumor cells/cell-free DNA, novel targets of antibody-drug conjugates, and residual disease.Expert opinion: Biomarker-driven care in the management of TNBC is increasing and has helped expand options for patients diagnosed with this subtype of breast cancer. Research efforts are ongoing to identify additional biomarkers and targeted treatment options with the ultimate goal of improving clinical outcomes and survivorship.
Collapse
Affiliation(s)
- Jasmine Sukumar
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Kelly Gast
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Maryam Lustberg
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Nicole Williams
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| |
Collapse
|
41
|
Abstract
ABSTRACT Triple-negative breast cancer is increasingly recognized as a heterogeneous entity that can be categorized according to histologic, molecular, and clinical subtypes. While chemotherapy remains the backbone of treatment for this disease, there are now several available targeted agents including immunotherapy, poly(adenosine diphosphate-ribose) polymerase inhibitors, and most recently a Food and Drug Administration-approved antibody-drug conjugate sacituzumab govitecan-hziy as a third-line treatment of metastatic triple-negative breast cancer. We review several actionable targets for triple-negative breast cancer and describe promising nonimmunotherapeutic agents including cyclin-dependent kinase inhibitors, androgen receptor inhibitors, mitogen-activated protein kinase inhibitors, phosphoinositide 3-kinase inhibitors, AKT (also known as protein kinase B) inhibitors, and antibody-drug conjugates.
Collapse
|
42
|
Dass SA, Tan KL, Selva Rajan R, Mokhtar NF, Mohd Adzmi ER, Wan Abdul Rahman WF, Tengku Din TADAA, Balakrishnan V. Triple Negative Breast Cancer: A Review of Present and Future Diagnostic Modalities. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:62. [PMID: 33445543 PMCID: PMC7826673 DOI: 10.3390/medicina57010062] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast type of cancer with no expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). It is a highly metastasized, heterogeneous disease that accounts for 10-15% of total breast cancer cases with a poor prognosis and high relapse rate within five years after treatment compared to non-TNBC cases. The diagnostic and subtyping of TNBC tumors are essential to determine the treatment alternatives and establish personalized, targeted medications for every TNBC individual. Currently, TNBC is diagnosed via a two-step procedure of imaging and immunohistochemistry (IHC), which are operator-dependent and potentially time-consuming. Therefore, there is a crucial need for the development of rapid and advanced technologies to enhance the diagnostic efficiency of TNBC. This review discusses the overview of breast cancer with emphasis on TNBC subtypes and the current diagnostic approaches of TNBC along with its challenges. Most importantly, we have presented several promising strategies that can be utilized as future TNBC diagnostic modalities and simultaneously enhance the efficacy of TNBC diagnostic.
Collapse
Affiliation(s)
- Sylvia Annabel Dass
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Kim Liu Tan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Rehasri Selva Rajan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia; (N.F.M.); (E.R.M.A.)
| | - Elis Rosliza Mohd Adzmi
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia; (N.F.M.); (E.R.M.A.)
| | - Wan Faiziah Wan Abdul Rahman
- Department of Pathology, School of Medical Sciences, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia;
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
| | - Tengku Ahmad Damitri Al-Astani Tengku Din
- Breast Cancer Awareness & Research Unit, Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
- Chemical Pathology Department, School of Medical Sciences, Health Campus, Kubang Kerian, Kelantan 16150, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, USM, Penang 11800, Malaysia; (S.A.D.); (K.L.T.); (R.S.R.)
| |
Collapse
|
43
|
Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer 2020; 6:54. [PMID: 33088912 PMCID: PMC7568552 DOI: 10.1038/s41523-020-00197-2] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is not a unique disease, encompassing multiple entities with marked histopathological, transcriptomic and genomic heterogeneity. Despite several efforts, transcriptomic and genomic classifications have remained merely theoretic and most of the patients are being treated with chemotherapy. Driver alterations in potentially targetable genes, including PIK3CA and AKT, have been identified across TNBC subtypes, prompting the implementation of biomarker-driven therapeutic approaches. However, biomarker-based treatments as well as immune checkpoint inhibitor-based immunotherapy have provided contrasting and limited results so far. Accordingly, a better characterization of the genomic and immune contexture underpinning TNBC, as well as the translation of the lessons learnt in the metastatic disease to the early setting would improve patients' outcomes. The application of multi-omics technologies, biocomputational algorithms, assays for minimal residual disease monitoring and novel clinical trial designs are strongly warranted to pave the way toward personalized anticancer treatment for patients with TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| |
Collapse
|
44
|
Liu J, Li G, He Y, He G, Zhang P, Shen X, Zhang W, Chen S, Cui S, Tan Y. The Association Analysis of GPNMB rs156429 With Clinical Manifestations in Chinese Population With Parkinson's Disease. Front Genet 2020; 11:952. [PMID: 32983228 PMCID: PMC7492656 DOI: 10.3389/fgene.2020.00952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/29/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jin Liu
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gen Li
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixi He
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiying He
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingchen Zhang
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Shen
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weishan Zhang
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengdi Chen,
| | - Shishuang Cui
- Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shishuang Cui,
| | - Yuyan Tan
- Department of Neurology & Collaborative Innovation Center for Brain Science, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Yuyan Tan,
| |
Collapse
|
45
|
The soluble glycoprotein NMB (GPNMB) produced by macrophages induces cancer stemness and metastasis via CD44 and IL-33. Cell Mol Immunol 2020; 18:711-722. [PMID: 32728200 DOI: 10.1038/s41423-020-0501-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.
Collapse
|
46
|
Cocco S, Piezzo M, Calabrese A, Cianniello D, Caputo R, Di Lauro V, Fusco G, di Gioia G, Licenziato M, de Laurentiis M. Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives. Int J Mol Sci 2020; 21:E4579. [PMID: 32605126 PMCID: PMC7369987 DOI: 10.3390/ijms21134579] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors characterized by aggressive behavior, high risk of distant recurrence, and poor survival. Chemotherapy is still the main therapeutic approach for this subgroup of patients, therefore, progress in the treatment of TNBC remains an important challenge. Data derived from molecular technologies have identified TNBCs with different gene expression and mutation profiles that may help developing targeted therapies. So far, however, only a few of these have shown to improve the prognosis and outcomes of TNBC patients. Robust predictive biomarkers to accelerate clinical progress are needed. Herein, we review prognostic and predictive biomarkers in TNBC, discuss the current evidence supporting their use, and look at the future of this research field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michelino de Laurentiis
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola, 53, 80131 Napoli NA, Italy; (S.C.); (M.P.); (A.C.); (D.C.); (R.C.); (V.D.L.); (G.F.); (G.d.G.); (M.L.)
| |
Collapse
|
47
|
Gadag S, Sinha S, Nayak Y, Garg S, Nayak UY. Combination Therapy and Nanoparticulate Systems: Smart Approaches for the Effective Treatment of Breast Cancer. Pharmaceutics 2020; 12:E524. [PMID: 32521684 PMCID: PMC7355786 DOI: 10.3390/pharmaceutics12060524] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer has become one of the biggest concerns for oncologists in the past few decades because of its unpredictable etiopathology and nonavailability of personalized translational medicine. The number of women getting affected by breast cancer has increased dramatically, owing to lifestyle and environmental changes. Besides, the development of multidrug resistance has become a challenge in the therapeutic management of breast cancer. Studies reveal that the use of monotherapy is not effective in the management of breast cancer due to high toxicity and the development of resistance. Combination therapies, such as radiation therapy with adjuvant therapy, endocrine therapy with chemotherapy, and targeted therapy with immunotherapy, are found to be effective. Thus, multimodal and combination treatments, along with nanomedicine, have emerged as a promising strategy with minimum side effects and drug resistance. In this review, we emphasize the multimodal approaches and recent advancements in breast cancer treatment modalities, giving importance to the current data on clinical trials. The novel treatment approach by targeted therapy, according to type, such as luminal, HER2 positive, and triple-negative breast cancer, are discussed. Further, passive and active targeting technologies, including nanoparticles, bioconjugate systems, stimuli-responsive, and nucleic acid delivery systems, including siRNA and aptamer, are explained. The recent research exploring the role of nanomedicine in combination therapy and the possible use of artificial intelligence in breast cancer therapy is also discussed herein. The complexity and dynamism of disease changes require the constant upgrading of knowledge, and innovation is essential for future drug development for treating breast cancer.
Collapse
Affiliation(s)
- Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Shristi Sinha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Sanjay Garg
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Usha Y. Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| |
Collapse
|
48
|
Tsou PS, Sawalha AH. Glycoprotein nonmetastatic melanoma protein B: A key mediator and an emerging therapeutic target in autoimmune diseases. FASEB J 2020; 34:8810-8823. [PMID: 32445534 DOI: 10.1096/fj.202000651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022]
Abstract
The glycoprotein nonmetastatic melanoma protein B (GPNMB, also known as osteoactivin) is highly expressed in many cell types and regulates the homeostasis in various tissues. In different physiological contexts, it functions as a melanosome-associated protein, membrane-bound surface receptor, soluble ligand, or adhesion molecule. Therefore, GPNMB is involved in cell differentiation, migration, inflammation, metabolism, and neuroprotection. Because of its various involvement in different physiological conditions, GPNMB has been implicated in many diseases, including cancer, neurological disorders, and more recently immune-mediated diseases. This review summarizes the regulation and function of GPNMB in normal physiology, and discusses the involvement of GPNMB in disease conditions with a particular focus on its potential role and therapeutic implications in autoimmunity.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.,Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
49
|
Nagayama A, Vidula N, Ellisen L, Bardia A. Novel antibody-drug conjugates for triple negative breast cancer. Ther Adv Med Oncol 2020; 12:1758835920915980. [PMID: 32426047 PMCID: PMC7222243 DOI: 10.1177/1758835920915980] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogenous subtype of breast cancer often associated with an aggressive phenotype and poor prognosis. Antibody–drug conjugate (ADC), comprising of a monoclonal antibody linked to a cytotoxic payload by a linker, is gaining increasing traction as an anti-cancer therapeutic. Emerging ADC drugs such as sacituzumab govitecan (IMMU-132) and trastuzumab deruxtecan (DS-8201a) are in late stages of clinical development for patients with metastatic breast cancer, including TNBC. In this article, we review and discuss the development and clinical application of ADCs in patients with advanced TNBC.
Collapse
Affiliation(s)
- Aiko Nagayama
- Department of Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Neelima Vidula
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Leif Ellisen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 10 North Grove Street, Boston, MA 02114-2621, USA
| |
Collapse
|
50
|
Liu LY, Ma XZ, Ouyang B, Ings DP, Marwah S, Liu J, Chen AY, Gupta R, Manuel J, Chen XC, Gage BK, Cirlan I, Khuu N, Chung S, Camat D, Cheng M, Sekhon M, Zagorovsky K, Abdou Mohamed MA, Thoeni C, Atif J, Echeverri J, Kollmann D, Fischer S, Bader GD, Chan WCW, Michalak TI, McGilvray ID, MacParland SA. Nanoparticle Uptake in a Spontaneous and Immunocompetent Woodchuck Liver Cancer Model. ACS NANO 2020; 14:4698-4715. [PMID: 32255624 DOI: 10.1021/acsnano.0c00468] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
There is a tremendous focus on the application of nanomaterials for the treatment of cancer. Nonprimate models are conventionally used to assess the biomedical utility of nanomaterials. However, these animals often lack an intact immunological background, and the tumors in these animals do not develop spontaneously. We introduce a preclinical woodchuck hepatitis virus-induced liver cancer model as a platform for nanoparticle (NP)-based in vivo experiments. Liver cancer development in these out-bred animals occurs as a result of persistent viral infection, mimicking human hepatitis B virus-induced HCC development. We highlight how this model addresses key gaps associated with other commonly used tumor models. We employed this model to (1) track organ biodistribution of gold NPs after intravenous administration, (2) examine their subcellular localization in the liver, (3) determine clearance kinetics, and (4) characterize the identity of hepatic macrophages that take up NPs using RNA-sequencing (RNA-seq). We found that the liver and spleen were the primary sites of NP accumulation. Subcellular analyses revealed accumulation of NPs in the lysosomes of CD14+ cells. Through RNA-seq, we uncovered that immunosuppressive macrophages within the woodchuck liver are the major cell type that take up injected NPs. The woodchuck-HCC model has the potential to be an invaluable tool to examine NP-based immune modifiers that promote host anti-tumor immunity.
Collapse
Affiliation(s)
- Lewis Y Liu
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Xue-Zhong Ma
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Ben Ouyang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
| | - Danielle P Ings
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Sagar Marwah
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Jeff Liu
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 230, Toronto, Ontario, Canada M5S 3E1
| | - Annie Y Chen
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Rahul Gupta
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Justin Manuel
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Xu-Chun Chen
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Blair K Gage
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- McEwen Stem Cell Institute, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Iulia Cirlan
- Princess Margaret Genomics Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Nicholas Khuu
- Princess Margaret Genomics Centre, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | - Sai Chung
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Damra Camat
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Michael Cheng
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
| | - Manmeet Sekhon
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Kyryl Zagorovsky
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
| | - Mohamed A Abdou Mohamed
- Luna Nanotech, Toronto, Ontario, Canada M5G 1Y8
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt 44519
| | - Cornelia Thoeni
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Jawairia Atif
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Juan Echeverri
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Dagmar Kollmann
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Sandra Fischer
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| | - Gary D Bader
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 230, Toronto, Ontario, Canada M5S 3E1
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, Ontario, Canada M5S 3G9
- Department of Materials Science and Engineering, University of Toronto, 160 College Street, Room 450, Toronto, Ontario, Canada M5S 3E1
| | - Tomasz I Michalak
- Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, 300 Prince Philip Drive, St. John's, Newfoundland, Canada A1B 3V6
| | - Ian D McGilvray
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
| | - Sonya A MacParland
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, Ontario, Canada M5G 2C4
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 6271, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|