1
|
Leylek O, Honeywell ME, Lee MJ, Hemann MT, Ozcan G. Functional genomics reveals an off-target dependency of drug synergy in gastric cancer therapy. Gastric Cancer 2024; 27:1201-1219. [PMID: 39033209 PMCID: PMC11513712 DOI: 10.1007/s10120-024-01537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Integrating molecular-targeted agents into combination chemotherapy is transformative for enhancing treatment outcomes in cancer. However, realizing the full potential of this approach requires a clear comprehension of the genetic dependencies underlying drug synergy. While the interactions between conventional chemotherapeutics are well-explored, the interplay of molecular-targeted agents with conventional chemotherapeutics remains a frontier in cancer treatment. Hence, we leveraged a powerful functional genomics approach to decode genomic dependencies that drive synergy in molecular-targeted agent/chemotherapeutic combinations in gastric adenocarcinoma, addressing a critical need in gastric cancer therapy. METHODS We screened pharmacological interactions between fifteen molecular-targeted agent/conventional chemotherapeutic pairs in gastric adenocarcinoma cells, and examined the genome-scale genetic dependencies of synergy integrating genome-wide CRISPR screening with the shRNA-based signature assay. We validated the synergy in cell death using fluorescence-based and lysis-dependent inference of cell death kinetics assay, and validated the genetic dependencies by single-gene knockout experiments. RESULTS Our combination screen identified SN-38/erlotinib as the drug pair with the strongest synergism. Functional genomics assays unveiled a genetic dependency signature of SN-38/erlotinib identical to SN-38. Remarkably, the enhanced cell death with improved kinetics induced by SN-38/erlotinib was attributed to erlotinib's off-target effect, inhibiting ABCG2, rather than its on-target effect on EGFR. CONCLUSION In the era of precision medicine, where emphasis on primary drug targets prevails, our research challenges this paradigm by showcasing a robust synergy underpinned by an off-target dependency. Further dissection of the intricate genetic dependencies that underlie synergy can pave the way to developing more effective combination strategies in gastric cancer therapy.
Collapse
Affiliation(s)
- Ozen Leylek
- Koç University Research Center for Translational Medicine, 34450, Istanbul, Turkey
| | - Megan E Honeywell
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Michael J Lee
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
| | - Michael T Hemann
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- MIT Koch Institute for Integrative Cancer Research, Cambridge, MA, 02139, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA.
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine, 34450, Istanbul, Turkey.
- Department of Medical Pharmacology, Koç University School of Medicine, 34450, Istanbul, Turkey.
| |
Collapse
|
2
|
Abd-Elhakim YM, Mohamed AAR, Khamis T, Metwally MMM, El-Shetry ES, Albaqami A, Mawkili W, Alosaimi ME, Alotaibi BS, ElAshmouny N, Dahran N, Alsharif G, Samak MA. Alleviative effects of green-fabricated zinc oxide nanoparticles on acrylamide-induced oxidative and inflammatory reactions in the rat stomach via modulating gastric neuroactive substances and the MiR-27a-5p/ROS/NF-κB axis. Tissue Cell 2024; 91:102574. [PMID: 39353228 DOI: 10.1016/j.tice.2024.102574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Little is known about the effects of acrylamide (AMD) on the stomach. So, this study evaluated the effect of oral AMD exposure (20 mg/kg b.wt) on oxidative status, apoptotic, and inflammatory reactions in rat's stomach for 60 days. To explore novel targets of AMD toxicity, a more detailed molecular and immune-expression study was performed. Besides, the possible protective effect of green synthesized zinc oxide nanoparticles (G-ZNP) (10 mg/kg b.wt) was explored. The results revealed that AMD significantly provoked oxidative and lipid peroxidative damage of the stomach in terms of increased ROS and MDA but reduced SOD, CAT, GSH, and GSH/GSSG. Additionally, the stomachs of AMD-exposed rats showed a significant increment of PGE2 but reduced NO. Histopathologically, AMD induced a significant increase in PAS stain and the immunoexpression of iNOS and NF-κB in the glandular stomach. A significant upregulation of CART, VACHT, EGFR, caspase-3, NOS-1, and miR-27a-5p was evident in the stomach of the AMD group. Yet, G-ZNP oral dosing significantly rescued the AMD-induced oxidative damage, apoptotic reaction, inflammatory effect, and altered miR-27a-5p and gene expressions in the stomach. Conclusively, these findings demonstrated the efficacy of G-ZNP in protecting against the harmful impacts of acrylamide on stomach tissues.
Collapse
Affiliation(s)
- Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed M M Metwally
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, King Salman International University, Ras Sidr, Egypt; Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Eman S El-Shetry
- Department of Anatomy, College of Medicine, University of Hail, Hail, Saudi Arabia; Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amirah Albaqami
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, Taif 21944, Saudi Arabia
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Manal E Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O Box 84428, Riyadh 11671, Saudi Arabia.
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Naira ElAshmouny
- Department of Histology and cell biology, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Naief Dahran
- Department of Basic Medical Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Ghadi Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, P.O.Box 9515, Jeddah 21423, Saudi Arabia; Department of Biomedical Research, King Abdullah International Medical Research Center, P.O.Box 9515, Jeddah 21423, Saudi Arabia
| | - Mai A Samak
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; College of medicine, University of Ha'il, Ha'il 2240, Saudi Arabia
| |
Collapse
|
3
|
Shaker F, Razi S, Rezaei N. Circulating miRNA and circulating tumor DNA application as liquid biopsy markers in gastric cancer. Clin Biochem 2024; 129:110767. [PMID: 38705444 DOI: 10.1016/j.clinbiochem.2024.110767] [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: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Liquid biopsy has been investigated as a novel method to overcome the numerous challenges in gastric cancer (GC) management. This non-invasive, feasible, and easy-to-repeat method has been shown to be cost-effective and capable of increasing diagnostic sensitivity and prognostic assessment. Additionally, it is potentially accurate to aid decision-making and personalized treatment planning. MicroRNA (miRNA) and circulating tumor DNA (ctDNA) markers can enhance GC management in various aspects, including diagnosis (mainly earlier diagnosis and the ability to perform population-based screening), prognosis (more precise stratification of prognosis), and treatment (including more accurate prediction of treatment response and earlier detection of resistance to the treatment). Concerning the treatment-related application, miRNAs' mimics and antagonists (by using two main strategies of restoring tumor suppressor miRNAs and inhibiting oncogene miRNAs) have been shown to be effective therapeutic agents. However, these need to be further validated in clinical trials. Furthermore, novel delivery systems, such as lipid-based vectors, polymeric-based vectors, and exosome-based delivery, have been developed to enhance the performance of these agents. Moreover, this paper explores the current detection and measuring methods for these markers. These approaches are categorized into direct methods (e.g., Chem-NAT, HTG EdgeSeq, and Multiplex Circulating Fireplex) and indirect methods (e.g., Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), qPCR, microarray, and NGS) for miRNA detection. For ctDNA measurement, main core technologies like NGS, digital PCR, real-time PCR, and mass spectrometry are suggested.
Collapse
Affiliation(s)
- Farhad Shaker
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
4
|
Wang Y, Li J, Liu X, Zhang Y, Wang C, Guo Q, Wang Y, Jiang B, Jin X, Liu Y. Elucidation of the anti-gastric cancer mechanism of Guiqi Baizhu Formula by integrative approach of chemical bioinformatics. Int Immunopharmacol 2024; 134:112245. [PMID: 38749334 DOI: 10.1016/j.intimp.2024.112245] [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: 02/26/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 06/03/2024]
Abstract
Gastric cancer (GC) has posed a great threat to the lives of people around the world. To date, safer and more cost-effective therapy for GC is lacking. Traditional Chinese medicine (TCM) may provide some new options for this. Guiqi Baizhu Formula (GQBZF), a classic TCM formula, has been extensively used to treat GC, while its bioactive components and therapeutic mechanisms remain unclear. In this study, we evaluated the underlying mechanisms of GQBZF in treating GC by integrative approach of chemical bioinformatics. GQBZF lyophilized powder (0.0625 mg/mL, 0.125 mg/mL) significantly attenuated the expression of p-IGF1R, PI3K, p-PDK1, p-VEGFR2 to inhibit the proliferation, migration and induce apoptosis of gastric cancer cells, which was consistent with the network pharmacology. Additionally, atractylenolide Ⅰ, quercetin, glycyrol, physcione and aloe-emodin, emodin, kaempferol, licoflavone A were found to be the key compounds of GQBZF regulating IGF1R and VEGFR2, respectively. And among which, glycyrol and emodin were determined as key active compounds against GC by farther vitro experiments and LC/MS. Meanwhile, we also found that glycyrol inhibited MKN-45 cells proliferation and enhanced apoptosis, which might be related to the inhibition of IGF1R/PI3K/PDK1, and emodin could significantly attenuate the MKN-45 cells migration, which might be related to the inhibition of VEGFR2-related signaling pathway. These results were verified again by molecular dynamics simulation and binding interaction pattern. In summary, this study suggested that GQBZF and its key active components (glycyrol and emodin) can suppress IGF1R/PI3K/PDK1 and VEGFR2-related signaling pathway, thereby inhibiting tumor cell proliferation and migration and inducing apoptosis. These findings provided an important strategy for developing new agents and facilitated clinical use of GQBZF against GC.
Collapse
Affiliation(s)
- Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jiawei Li
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiuzhu Liu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yixi Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Chao Wang
- College of Medical, Shanxi Datong University, Datong 037000, China
| | - Qingyang Guo
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yan Wang
- Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Bing Jiang
- Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiaojie Jin
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Dunhuang Medical and Transformation, Ministry of Education of The People's Republic of China, Lanzhou 730000, China.
| | - Yongqi Liu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Dunhuang Medical and Transformation, Ministry of Education of The People's Republic of China, Lanzhou 730000, China.
| |
Collapse
|
5
|
Yan-Rui W, Xue-Er Y, Mao-Yu D, Ya-Ting L, Bo-Heng L, Miao-Jie Z, Li Z. Research on the signaling pathway and the related mechanism of traditional Chinese medicine intervention in chronic gastritis of the "inflammation-cancer transformation". Front Pharmacol 2024; 15:1338471. [PMID: 38698812 PMCID: PMC11063381 DOI: 10.3389/fphar.2024.1338471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Objective: The aim of this study is to uncover the traditional Chinese medicine (TCM) treatments for chronic gastritis and their potential targets and pathways involved in the "inflammation-cancer" conversion in four stages. These findings can provide further support for future research into TCM and its active components. Materials and methods: The literature search encompassed PubMed, Web of Science, Google Scholar, CNKI, WanFang, and VIP, employing keywords such as "chronic gastritis", "gastric cancer", "traditional Chinese medicine", "medicinal herb", "Chinese herb", and "natural plant". Results: Herbal remedies may regulate the signaling pathways linked to the advancement of chronic gastritis. Under the multi-target and multi-pathway independent or combined reaction, the inflammatory microenvironment may be enhanced, leading to repair of damaged gastric mucosal cells, buffering the progress of mucosal atrophic degeneration via the decrease of inflammatory factor expression, inhibition of oxidative stress-induced damage, facilitation of microvascular neovascularization in the gastric mucosa and regulation of the processes of gastric mucosal cell differentiation and proliferation. Simultaneously, the decreased expression of inflammatory factors may impact the expression of associated oncogenes and regulate the malignant proliferation of cells, thereby achieving the treatment and prevention objectives of gastric cancer through the reduction of cell metastasis and apoptosis. Conclusion: Chinese medicine formulations and individual drugs can be utilised at various stages of the "inflammation-cancer" progression of chronic gastritis to prevent and treat gastric cancer in a multi-level, multi-targeted, and multi-directional fashion. This can provide guidance for the accurate application of medicines during different stages of "inflammation-cancer" transformation. New insights into the mechanism of inflammation-cancer transformation and the development of novel drugs for chronic gastritis can be gained through an extensive investigation of TCM treatment in this condition.
Collapse
Affiliation(s)
- Wang Yan-Rui
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Yan Xue-Er
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Ding Mao-Yu
- Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Lu Ya-Ting
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Lu Bo-Heng
- Beijing University of Chinese Medicine, Beijing, China
| | - Zhai Miao-Jie
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Zhu Li
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Leylek O, Honeywell ME, Lee MJ, Hemann MT, Ozcan G. Functional genomics reveals an off-target dependency of drug synergy in gastric cancer therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.07.561351. [PMID: 37873383 PMCID: PMC10592690 DOI: 10.1101/2023.10.07.561351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The rational combination of anticancer agents is critical to improving patient outcomes in cancer. Nonetheless, most combination regimens in the clinic result from empirical methodologies disregarding insight into the mechanism of action and missing the opportunity to improve therapy outcomes incrementally. Deciphering the genetic dependencies and vulnerabilities responsible for synergistic interactions is crucial for rationally developing effective anticancer drug combinations. Hence, we screened pairwise pharmacological interactions between molecular-targeted agents and conventional chemotherapeutics and examined the genome-scale genetic dependencies in gastric adenocarcinoma cell models. Since this type of cancer is mainly chemoresistant and incurable, clinical situations demand effective combination strategies. Our pairwise combination screen revealed SN38/erlotinib as the drug pair with the most robust synergism. Genome-wide CRISPR screening and a shRNA-based signature assay indicated that the genetic dependency/vulnerability signature of SN38/erlotinib is the same as SN38 alone. Additional investigation revealed that the enhanced cell death with improved death kinetics caused by the SN38/erlotinib combination is surprisingly due to erlotinib's off-target effect that inhibits ABCG2 but not its on-target effect on EGFR. Our results confirm that a genetic dependency signature different from the single-drug application may not be necessary for the synergistic interaction of molecular-targeted agents with conventional chemotherapeutics in gastric adenocarcinoma. The findings also demonstrated the efficacy of functional genomics approaches in unveiling biologically validated mechanisms of pharmacological interactions.
Collapse
Affiliation(s)
- Ozen Leylek
- Koç University Research Center for Translational Medicine, Istanbul, 34450 Turkiye
| | - Megan E Honeywell
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605 USA
| | - Michael J Lee
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605 USA
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605 USA
| | - Michael T Hemann
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
- MIT Koch Institute for Integrative Cancer Research, Cambridge, MA, 02139 USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139 USA
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine, Istanbul, 34450 Turkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, 34450 Turkiye
| |
Collapse
|
7
|
Lee JE, Kim KT, Shin SJ, Cheong JH, Choi YY. Genomic and evolutionary characteristics of metastatic gastric cancer by routes. Br J Cancer 2023; 129:672-682. [PMID: 37422528 PMCID: PMC10421927 DOI: 10.1038/s41416-023-02338-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND In gastric cancer (GC) patients, metastatic progression through the lymphatic, hematogenous, peritoneal, and ovarian routes, is the ultimate cause of death. However, the genomic and evolutionary characteristics of metastatic GC have not been widely evaluated. METHODS Whole-exome sequencing data were analyzed for 99 primary and paired metastatic gastric cancers from 15 patients who underwent gastrectomy and metastasectomy. RESULTS Hematogenous metastatic tumors were associated with increased chromosomal instability and de novo gain/amplification in cancer driver genes, whereas peritoneal/ovarian metastasis was linked to sustained chromosomal stability and de novo somatic mutations in driver genes. The genomic distance of the hematogenous and peritoneal metastatic tumors was found to be closer to the primary tumors than lymph node (LN) metastasis, while ovarian metastasis was closer to LN and peritoneal metastasis than the primary tumor. Two migration patterns for metastatic GCs were identified; branched and diaspora. Both molecular subtypes of the metastatic tumors, rather than the primary tumor, and their migration patterns were related to patient survival. CONCLUSIONS Genomic characteristics of metastatic gastric cancer is distinctive by routes and associated with patients' prognosis along with genomic evolution pattenrs, indicating that both primary and metastatic gastric cancers require genomic evaluation.
Collapse
Affiliation(s)
- Jae Eun Lee
- Portrai Inc., Seoul, Korea
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, Seoul, South Korea
| | - Ki Tae Kim
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry, Seoul National University, Seoul, South Korea
- Dental Research Institute and Dental Multi-omics Center, Seoul National University, Seoul, South Korea
| | - Su-Jin Shin
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, Seoul, South Korea.
| | - Yoon Young Choi
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea.
| |
Collapse
|
8
|
Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals (Basel) 2023; 16:ph16040614. [PMID: 37111371 PMCID: PMC10144220 DOI: 10.3390/ph16040614] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: The oncology field has drastically changed with the advent of precision medicine, led by the discovery of druggable genes or immune targets assessed through next-generation sequencing. Biomarker-based treatments are increasingly emerging, and currently, six tissue-agnostic therapies are FDA-approved. (2) Methods: We performed a review of the literature and reported the trials that led to the approval of tissue-agnostic treatments and ongoing clinical trials currently investigating novel biomarker-based approaches. (3) Results: We discussed the approval of agnostic treatments: pembrolizumab and dostarlimab for MMRd/MSI-H, pembrolizumab for TMB-H, larotrectinib and entrectinib for NTRK-fusions, dabrafenib plus trametinib for BRAF V600E mutation, and selpercatinib for RET fusions. In addition, we reported novel clinical trials of biomarker-based approaches, including ALK, HER2, FGFR, and NRG1. (4) Conclusions: Precision medicine is constantly evolving, and with the improvement of diagnostic tools that allow a wider genomic definition of the tumor, tissue-agnostic targeted therapies are a promising treatment strategy tailored to the specific tumor genomic profile, leading to improved survival outcomes.
Collapse
Affiliation(s)
- Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40127 Bologna, Italy
| | - Razelle Kurzrock
- MCW Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, San Diego, CA 92093, USA
- Department of Oncology, University of Nebraska, Omaha, NE 68198, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
9
|
Adashek JJ, Subbiah V, Westphalen CB, Naing A, Kato S, Kurzrock R. Cancer: slaying the nine-headed Hydra. Ann Oncol 2023; 34:61-69. [PMID: 35931318 PMCID: PMC10923524 DOI: 10.1016/j.annonc.2022.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 02/03/2023] Open
Abstract
Modern medicine continues to evolve, and the treatment armamentarium for various diseases grows more individualized across a breadth of medical disciplines. Cure rates for infectious diseases that were previously pan-fatal approach 100% because of the identification of the specific pathogen(s) involved and the use of appropriate combinations of drugs, where needed, to completely extinguish infection and hence prevent emergence of resistant strains. Similarly, with the assistance of technologies such as next-generation sequencing and immunomic analysis as part of the contemporary oncology armory, therapies can be tailored to each tumor. Importantly, molecular interrogation has revealed that metastatic cancers are distinct from each other and complex. Therefore, it is conceivable that rational personalized drug combinations will be needed to eradicate cancers, and eradication will be necessary to mitigate clonal evolution and resistance.
Collapse
Affiliation(s)
- J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore.
| | - V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C B Westphalen
- Comprehensive Cancer Center Munich and Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - A Naing
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California, San Diego
| | - R Kurzrock
- WIN Consortium, San Diego; MCW Cancer Center, Milwaukee; University of Nebraska, Omaha, USA.
| |
Collapse
|
10
|
Li X, Xu J, Xie J, Yang W. Research progress in targeted therapy and immunotherapy for gastric cancer. Chin Med J (Engl) 2022; 135:1299-1313. [PMID: 35830242 PMCID: PMC9433086 DOI: 10.1097/cm9.0000000000002185] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Gastric cancer (GC) is one of the most common malignant tumors worldwide. Its incidence ranks the 5th among all malignant tumors globally, and it is the 3rd leading cause of death among patients with cancer. Surgical treatment is the first choice in clinical practice. However, targeted therapy, immunotherapy, and other treatment methods have also become research hotspots at home and abroad with the development of individualized precision therapy in recent years, besides traditional radiotherapy and chemotherapy. At present, targeted therapy and immunotherapy are methods used for treating GC, and they have important clinical application value and prospects. This study aimed to review the research progress of targeted therapy and immunotherapy for GC, focusing on its mechanism of action and related important clinical trials, hoping to provide references for the clinical treatment of GC.
Collapse
Affiliation(s)
- Xuewei Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xu
- Department of Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Wenhui Yang
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
| |
Collapse
|
11
|
Li Z, Qin T, Li Z, Zhao X, Zhang X, Zhao T, Yang N, Miao J, Ma J, Zhang Z. Discovery of quinazoline derivatives as a novel class of potent and in vivo efficacious LSD1 inhibitors by drug repurposing. Eur J Med Chem 2021; 225:113778. [PMID: 34416665 DOI: 10.1016/j.ejmech.2021.113778] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 01/09/2023]
Abstract
Histone lysine-specific demethylase 1 (LSD1) is an important epigenetic modulator, and is implicated in malignant transformation and tumor pathogenesis in different ways. Therefore, the inhibition of LSD1 provides an attractive therapeutic target for cancer therapy. Based on drug repurposing strategy, we screened our in-house chemical library toward LSD1, and found that the EGFR inhibitor erlotinib, an FDA-approved drug for lung cancer, possessed low potency against LSD1 (IC50 = 35.80 μM). Herein, we report our further medicinal chemistry effort to obtain a highly water-soluble erlotinib analog 5k (>100 mg/mL) with significantly enhanced inhibitory activity against LSD1 (IC50 = 0.69 μM) as well as higher specificity. In MGC-803 cells, 5k suppressed the demethylation of LSD1, indicating its cellular activity against the enzyme. In addition, 5k had a remarkable capacity to inhibit colony formation, suppress migration and induce apoptosis of MGC803 cells. Furthermore, in MGC-803 xenograft mouse model, 5k treatment resulted in significant reduction in tumor size by 81.6% and 96.1% at dosages of 40 and 80 mg/kg/d, respectively. Our findings indicate that erlotinib-based analogs provide a novel structural set of LSD1 inhibitors with potential for further investigation, and may serve as novel candidates for the treatment of LSD1-overexpressing cancers.
Collapse
Affiliation(s)
- Zhonghua Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Tingting Qin
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Zhongrui Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xuan Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xinhui Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Taoqian Zhao
- Department of Chemistry, National University of Singapore, 117543, Singapore
| | - Nian Yang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jinlian Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
12
|
Liu X, Wang S, Li J, Zhang J, Liu D. Regulatory effect of traditional Chinese medicines on signaling pathways of process from chronic atrophic gastritis to gastric cancer. CHINESE HERBAL MEDICINES 2021; 14:5-19. [PMID: 36120132 PMCID: PMC9476726 DOI: 10.1016/j.chmed.2021.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/23/2021] [Accepted: 10/12/2021] [Indexed: 01/30/2023] Open
Affiliation(s)
- Xinnan Liu
- Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin 300380, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuping Wang
- Department of Pharmacy, Tianjin Provincial Corps Hospital, Chinese People’s Armed Police Forces, Tianjin 300162, China
| | - Jingyang Li
- Logistics College of Chinese People’s Armed Police Forces, Tianjin 300309, China
| | - Jingze Zhang
- Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin 300380, China
- Corresponding authors.
| | - Dailin Liu
- Tianjin Modern Innovation Chinese Medicine Technology Co., Ltd., Tianjin 300380, China
- Corresponding authors.
| |
Collapse
|
13
|
Cadamuro M, Lasagni A, Lamarca A, Fouassier L, Guido M, Sarcognato S, Gringeri E, Cillo U, Strazzabosco M, Marin JJ, Banales JM, Fabris L. Targeted therapies for extrahepatic cholangiocarcinoma: preclinical and clinical development and prospects for the clinic. Expert Opin Investig Drugs 2021; 30:377-388. [PMID: 33622120 DOI: 10.1080/13543784.2021.1880564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Until recently, cholangiocarcinoma (CCA) was a largely overlooked disease, and among CCAs, extrahepatic CCA (eCCA) was even more neglected. Despite the growing impact of molecularly targeted therapies and immunotherapy, prognosis of eCCA is dismal. Therefore, unraveling the complex molecular landscape of eCCA has become an urgent need. Deep phenotyping studies have revealed that eCCA is a heterogeneous tumor, harboring specific alterations categorizable into four classes, 'Mesenchymal', 'Proliferation', 'Immune', 'Metabolic'. Molecular alterations convey the activation of several pro-oncogenic pathways, where either actionable drivers or outcome predictors can be identified.Areas covered: We offer insights on perturbed pathways, molecular profiling, and actionable targets in eCCA and present a perspective on the potential stepping-stones to future progress. A systematic literature search in PubMed/ClinicalTrials.gov websites was performed by authors from different disciplines according to their specific topic knowledge to identify the newest and most relevant advances in precision medicine of eCCA.Expert opinion: eCCA is a distinct entity with unique features in terms of molecular classes, oncogenic drivers, and tumor microenvironment. Since more prevalent mutations are currently undruggable, and immunotherapy can be offered only to a minority of patients, international collaborations are instrumental to improve the understanding of the molecular underpins of this disease.
Collapse
Affiliation(s)
- Massimiliano Cadamuro
- Department of Molecular Medicine (DMM), University of Padua, Padua. Italy.,International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy
| | - Alberto Lasagni
- Division of General Medicine, Padua University-Hospital, Padua, Italy
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation, Manchester, United Kingdom.,Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Enrico Gringeri
- Hepatobiliary Surgery and Liver Transplantation, Padua University-Hospital, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplantation, Padua University-Hospital, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Mario Strazzabosco
- International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy.,Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US
| | - Jose Jg Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), IBSAL, CIBERehd, University of Salamanca, Salamanca, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital -, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Luca Fabris
- Department of Molecular Medicine (DMM), University of Padua, Padua. Italy.,International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy.,Division of General Medicine, Padua University-Hospital, Padua, Italy.,Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US
| |
Collapse
|
14
|
Zhang Y, Tan J, Zhou L, Shan X, Liu J, Ma Y. Synthesis and Application of AS1411-Functionalized Gold Nanoparticles for Targeted Therapy of Gastric Cancer. ACS OMEGA 2020; 5:31227-31233. [PMID: 33324832 PMCID: PMC7726946 DOI: 10.1021/acsomega.0c04605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/10/2020] [Indexed: 05/09/2023]
Abstract
Gastric cancer therapy is still a big challenge, and nanomedicines bring much more hope. It is essential to develop multifunctional nanoparticles, especially those with high targeted capacity and antitumor effects, to improve gastric cancer therapy. In this study, we constructed AS1411 aptamer-based gold nanoparticles with appropriate size facilitating endocytosis and actively targeted drug delivery for gastric cancer cells via the specific AS1411-nucleolin interaction. The AS1411-based nanoparticles showed obviously increased targeted capacity towards AGS cells compared to random ssDNA-based nanoparticles. Meanwhile, compared to L929 cells, the AS1411-based nanoparticles showed selective drug uptake and delivery for AGS cells. Importantly, the AS1411-based nanoparticles exhibited significantly stronger antitumor effects on AGS cells under laser irradiation compared to chemotherapy alone. Our nanoparticles combined targeted drug delivery and efficient antitumor effects within one single nanoplatform, which are promising to be applied as targeted nanomedicines against gastric cancer.
Collapse
Affiliation(s)
- Yajie Zhang
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
- Department
of Gastroenterology, Shengjing Hospital
of China Medical University, Shenyang 110004, China
| | - Jingwei Tan
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Lu Zhou
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Xiaoqing Shan
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Jianling Liu
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Yong Ma
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
15
|
Costantini M, Amoreo CA, Torregrossa L, Alì G, Munari E, Jeronimo C, Henrique R, Petronilho S, Capitanio U, Lucianò R, Suardi N, Landi MT, Anceschi U, Brassetti A, Fazio VM, Gallucci M, Simone G, Sentinelli S, Poeta ML. Assessment of HER2 Protein Overexpression and Gene Amplification in Renal Collecting Duct Carcinoma: Therapeutic Implication. Cancers (Basel) 2020; 12:E3345. [PMID: 33198197 PMCID: PMC7697829 DOI: 10.3390/cancers12113345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 01/31/2023] Open
Abstract
Collecting duct carcinoma (CDC) is rare and aggressive histology of kidney cancers. Although different therapeutic approaches have been tested, the 2-year survival remains very poor. Since CDC exhibits overlapping features with urothelial carcinoma, the analysis of shared molecular alterations could provide new insights into the understanding of this rare disease and also therapeutic options. We collected 26 CDC cases, and we assessed HER2 protein expression by immunohistochemistry (IHC) and gene amplification by fluorescence in-situ hybridization (FISH) according to 2018 ASCO/CAP HER2-testing recommendations. Six out of twenty-six (23%) tumors showed HER2 positive staining. In particular, 3+ score was present in 2/6 cases (33%), 2+ in 3/6 cases (50%) and 1+ in 1/6 cases (17%). The 6 HER2+ tumors were also analyzed by FISH to assess gene copy number. One out of six CDC with IHC 3+ was also HER2 amplified, showing an average HER2 copy number ≥4.0 (10.85) and a HER2/CEP17 ratio ≥ (5.63), while the 5/6 cases were HER2 negative. Based on the 2018 ASCO/CAP guidelines overall, 2/26 CDC cases (8%) were HER2+. The present study provides evidence for testing, in future studies, HER2 to assess its clinical value as a novel target for the treatment of this highly malignant cancer.
Collapse
Affiliation(s)
- Manuela Costantini
- Department of Urology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy; (M.C.); (U.A.); (A.B.); (G.S.)
| | - Carla Azzurra Amoreo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy;
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Anatomic Pathology Section, 56126 Pisa, Italy; (L.T.); (G.A.)
| | - Greta Alì
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Anatomic Pathology Section, 56126 Pisa, Italy; (L.T.); (G.A.)
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria, 37024 Negrar, Italy;
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Carmen Jeronimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal; (C.J.); (R.H.); (S.P.)
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal; (C.J.); (R.H.); (S.P.)
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Sara Petronilho
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal; (C.J.); (R.H.); (S.P.)
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Umberto Capitanio
- Unit of Urology, Division of Experimental Oncology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (U.C.); (N.S.)
| | - Roberta Lucianò
- Unit of Pathology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Nazareno Suardi
- Unit of Urology, Division of Experimental Oncology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (U.C.); (N.S.)
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD 20892, USA;
| | - Umberto Anceschi
- Department of Urology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy; (M.C.); (U.A.); (A.B.); (G.S.)
| | - Aldo Brassetti
- Department of Urology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy; (M.C.); (U.A.); (A.B.); (G.S.)
| | - Vito Michele Fazio
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
- CNR-Institute of Translational Pharmacology, 00133 Roma, Italy
| | - Michele Gallucci
- Department of Urology, University of Rome, La Sapienza, Rome, Viale del Policlinico 155, 00161 Rome, Italy;
| | - Giuseppe Simone
- Department of Urology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy; (M.C.); (U.A.); (A.B.); (G.S.)
| | - Steno Sentinelli
- Department of Pathology, IRCCS Regina Elena National Cancer Institute—Rome, via Elio Chianesi 53, 00144 Rome, Italy;
| | - Maria Luana Poeta
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, via Orabona 4, 70126 Bari, Italy
| |
Collapse
|