1
|
McDonald ES, Pan TC, Pant DK, Troester MA, Kossenkov AV, Mankoff DA, Mach RH, Chodosh LA. Ternary Complex Components Responsible for Rapid LDL Internalization as Biomarkers for Breast Cancer Associated with Proliferation and Early Recurrence. CANCER RESEARCH COMMUNICATIONS 2025; 5:226-239. [PMID: 39804138 PMCID: PMC11791746 DOI: 10.1158/2767-9764.crc-23-0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/16/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
SIGNIFICANCE This first large-scale analysis of the putative ternary complex responsible for rapid low-density lipoprotein internalization in breast cancer reveals a link between component expression and recurrence, with prognostic implications for identifying patients needing supplemental posttreatment surveillance and/or additional therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth S. McDonald
- Division of Breast Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tien-Chi Pan
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dhruv K. Pant
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melissa A. Troester
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Andrew V. Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - David A. Mankoff
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H. Mach
- Radiochemistry, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lewis A. Chodosh
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Toàn NM. Novel Molecular Classification of Breast Cancer with PET Imaging. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2099. [PMID: 39768978 PMCID: PMC11678748 DOI: 10.3390/medicina60122099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Breast cancer is a heterogeneous disease characterized by a wide range of biomarker expressions, resulting in varied progression, behavior, and prognosis. While traditional biopsy-based molecular classification is the gold standard, it is invasive and limited in capturing tumor heterogeneity, especially in deep or metastatic lesions. Molecular imaging, particularly positron emission tomography (PET) imaging, offering a non-invasive alternative, potentially plays a crucial role in the classification and management of breast cancer by providing detailed information about tumor location, heterogeneity, and progression. This narrative review, which focuses on both clinical patients and preclinical studies, explores the latest advancements in PET imaging for breast cancer, emphasizing the development of new tracers targeting hormone receptors such as the estrogen alpha receptor, progesterone receptor, androgen receptor, estrogen beta receptor, as well as the ErbB family of receptors, VEGF/VEGFR, PARP1, PD-L1, and markers for indirectly assessing Ki-67. These innovative radiopharmaceuticals have the potential to guide personalized treatment approaches based on the unique tumor profiles of individual patients. Additionally, they may improve the assessment of treatment efficacy, ultimately leading to better outcomes for those diagnosed with breast cancer.
Collapse
Affiliation(s)
- Ngô Minh Toàn
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
3
|
Li Y, Liang L. Transcription Impairment of TMEM208 by ZBTB14 Suppresses Breast cancer Radiotherapy Resistance. J Mammary Gland Biol Neoplasia 2024; 29:20. [PMID: 39692812 DOI: 10.1007/s10911-024-09573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
Zinc finger and BTB domain-containing protein (ZBTB) proteins have been implicated in different cellular processes, including DNA damage responses and cell cycle progression. However, the mechanism by which ZBTB14 modulates radiotherapy (RT) radioresistance (RT-R) remains to be elucidated. We aimed to elucidate the regulation mechanism of ZBTB14 in breast cancer (BC) RT-R. Using integrated bioinformatics prediction, ZBTB14 was identified as a hub transcription factor related to RT-R in BC. ZBTB14 was significantly under-expressed in non-responders and RT-R/BC cells, whereas its target transmembrane protein 208 (TMEM208) was significantly overexpressed in non-responders and RT-R/BC cells. Chromatin immunoprecipitation-qPCR and luciferase reporter assays revealed that ZBTB14 downregulated TMEM208 expression through transcriptional repression. Overexpression of ZBTB14 significantly inhibited the malignant biological behavior of BC cells and tumor growth in vivo, and further upregulation of TMEM208 reversed the biological activity and radiotherapy resistance of RT-R/BC cells inhibited by overexpression of ZBTB14.
Collapse
Affiliation(s)
- Yan Li
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Harbin, Heilongjiang, 150040, PR China
| | - Lili Liang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
4
|
Edmonds CE, O'Brien SR, McDonald ES, Mankoff DA, Pantel AR. PET Imaging of Breast Cancer: Current Applications and Future Directions. JOURNAL OF BREAST IMAGING 2024; 6:586-600. [PMID: 39401324 DOI: 10.1093/jbi/wbae053] [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: 04/23/2024] [Indexed: 11/07/2024]
Abstract
As molecular imaging use expands for patients with breast cancer, it is important for breast radiologists to have a basic understanding of molecular imaging, including PET. Although breast radiologists may not directly interpret such studies, basic knowledge of molecular imaging will enable the radiologist to better direct diagnostic workup of patients as well as discuss diagnostic imaging with the patient and other treating physicians. Several new tracers are now available to complement imaging glucose metabolism with FDG. Because it provides a noninvasive assessment of disease status across the whole body, PET offers specific advantages over tissue-based assays. Paired with targeted therapy, molecular imaging has the potential to guide personalized treatment of breast cancer, including guiding dosing during drug trials as well as predicting and assessing clinical response. This review discusses the current established applications of FDG, which remains the most widely used PET radiotracer for malignancy, including breast cancer, and highlights potential areas for expanded use based on recent research. It also summarizes research to date on the U.S. Food and Drug Administration (FDA)-approved PET tracer 16α-18F-fluoro-17β-estradiol (FES), which targets ER, including the current guidelines from the Society of Nuclear Medicine and Molecular Imaging on the appropriate use of FES-PET/CT for breast cancer as well as areas of active investigation for other potential applications. Finally, the review highlights several of the most promising novel PET tracers that are poised for clinical translation in the near future.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth S McDonald
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Takchi R, Prudner BC, Gong Q, Hagi T, Newcomer KF, Jin LX, Vangveravong S, Van Tine BA, Hawkins WG, Spitzer D. Cytotoxic sigma-2 ligands trigger cancer cell death via cholesterol-induced-ER-stress. Cell Death Dis 2024; 15:309. [PMID: 38697978 PMCID: PMC11066049 DOI: 10.1038/s41419-024-06693-8] [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: 07/25/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Sigma-2-ligands (S2L) are characterized by high binding affinities to their cognate sigma-2 receptor, overexpressed in rapidly proliferating tumor cells. As such, S2L were developed as imaging probes (ISO1) or as cancer therapeutics, alone (SV119 [C6], SW43 [C10]) and as delivery vehicles for cytotoxic drug cargoes (C6-Erastin, C10-SMAC). However, the exact mechanism of S2L-induced cytotoxicity remains to be fully elucidated. A series of high-affinity S2L were evaluated regarding their cytotoxicity profiles across cancer cell lines. While C6 and C10 displayed distinct cytotoxicities, C0 and ISO1 were essentially non-toxic. Confocal microscopy and lipidomics analysis in cellular and mouse models revealed that C10 induced increases in intralysosomal free cholesterol and in cholesterol esters, suggestive of unaltered intracellular cholesterol trafficking. Cytotoxicity was caused by cholesterol excess, a phenomenon that contrasts the effects of NPC1 inhibition. RNA-sequencing revealed gene clusters involved in cholesterol homeostasis and ER stress response exclusively by cytotoxic S2L. ER stress markers were confirmed by qPCR and their targeted modulation inhibited or enhanced cytotoxicity of C10 in a predicted manner. Moreover, C10 increased sterol regulatory element-binding protein 2 (SREBP2) and low-density lipoprotein receptor (LDLR), both found to be pro-survival factors activated by ER stress. Furthermore, inhibition of downstream processes of the adaptive response to S2L with simvastatin resulted in synergistic treatment outcomes in combination with C10. Of note, the S2L conjugates retained the ER stress response of the parental ligands, indicative of cholesterol homeostasis being involved in the overall cytotoxicity of the drug conjugates. Based on these findings, we conclude that S2L-mediated cell death is due to free cholesterol accumulation that leads to ER stress. Consequently, the cytotoxic profiles of S2L drug conjugates are proposed to be enhanced via concurrent ER stress inducers or simvastatin, strategies that could be instrumental on the path toward tumor eradication.
Collapse
Affiliation(s)
- Rony Takchi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Bethany C Prudner
- Department of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qingqing Gong
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Takaomi Hagi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Kenneth F Newcomer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Linda X Jin
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Suwanna Vangveravong
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Van Tine
- Department of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatric Hematology/Oncology, St. Louis Children's Hospital, St. Louis, MO, USA
- Alvin J Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - William G Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Alvin J Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA.
| | - Dirk Spitzer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Alvin J Siteman Cancer Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
6
|
Shields AF, Chen DL. Positron Emission Tomography Imaging of Tumor Proliferation and DNA Repair. Cancer J 2024; 30:170-175. [PMID: 38753751 DOI: 10.1097/ppo.0000000000000724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Positron emission tomography (PET) is an established tool for molecular imaging of cancers, and its role in diagnosis, staging, and phenotyping continues to evolve and expand rapidly. PET imaging of increased glucose utilization with 18F-fluorodeoxyglucose is now entrenched in clinical oncology practice for improving prognostication and treatment response assessment. Additional critical processes for cancer cell survival can also be imaged by PET, helping to inform individualized treatment selections for patients by improving our understanding of cell survival mechanisms and identifying relevant active mechanisms in each patient. The critical importance of quantifying cell proliferation and DNA repair pathways for prognosis and treatment selection is highlighted by the nearly ubiquitous use of the Ki-67 index, an established histological quantitative measure of cell proliferation, and BRCA mutation testing for treatment selection. This review focuses on PET advances in imaging and quantifying cell proliferation and poly(ADP-ribose)polymerase expression that can be used to complement cancer phenotyping approaches that will identify the most effective treatments for each individual patient.
Collapse
Affiliation(s)
- Anthony F Shields
- From the Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Delphine L Chen
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
7
|
Alluri SR, Zheng M, Holden D, Zhang Y, Zhang L, Felchner Z, Li S, Ropchan J, Carson R, Jia H, Huang Y. Evaluation of a First PET Tracer Suitable for Imaging the Sigma-2 Receptor in the Brain of Nonhuman Primates. Mol Pharm 2024; 21:194-200. [PMID: 38013422 DOI: 10.1021/acs.molpharmaceut.3c00750] [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: 11/29/2023]
Abstract
The sigma-2 receptor (σ2R), recently identified as transmembrane protein 97, is expressed in many cell types and mediates important functions in both the peripheral and central nervous systems. Over the years, σ2R has emerged as a potential therapeutic target for cancer and neurological disorders such as Alzheimer's disease (AD). The currently available σ2R radiotracers have been developed primarily for cancer imaging with limited brain uptake. Here, we report the evaluation of the first brain penetrant 18F-labeled radiotracer suitable for positron emission tomography (PET) imaging of σ2R in nonhuman primate brain.
Collapse
Affiliation(s)
| | - MingQiang Zheng
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Daniel Holden
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Ying Zhang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Li Zhang
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Zachary Felchner
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Jim Ropchan
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Richard Carson
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| | - Hongmei Jia
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yiyun Huang
- PET Center, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
8
|
Dashtestani P, Karami L. The molecular mechanism of the effects of the anti-neuropathic ligands on the modulation of the Sigma-2 receptor: An in-silico study. Int J Biol Macromol 2024; 254:127925. [PMID: 37944735 DOI: 10.1016/j.ijbiomac.2023.127925] [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: 05/24/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Neuropathic pain (NP) is a prevalent medical condition that lacks an effective treatment. Recently, the Sigma-2 receptor (S2R) has been proposed as a potential therapeutic target for NP. Some highly-selective S2R ligands (UKH1114, CM398, and YTD) have shown promising results in vivo, but the molecular interaction between the S2R and these ligands has been scarcely investigated. This work explores changes in the S2R upon interaction with the three mentioned ligands using in silico approaches. The results indicated that the ICL1, H1, ICL2, and ECL are the most dynamic regions of S2R in all systems. Binding interaction analysis identified amino acids with significant contribution to the binding free energy. Notably, the UKH1114-S2R simulation trajectory revealed that small alterations in the ICL1, H1, ICL2, and ECL form a new stable opening in the S2R, linking the occluded S2R binding pocket to the endoplasmic reticulum lumen, providing more evidence for the assumptions about the EBP and S2R mechanism of function. Further, the agreement between the membrane parameters in our study and experimental values confirms the validity of the MD simulations. Overall, this study provides new insights into the interaction between anti-NP ligands and the S2R.
Collapse
Affiliation(s)
- Parisa Dashtestani
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Leila Karami
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
9
|
Wang T, Wang J, Chen L, Zhang X, Mou T, An X, Zhang J, Zhang X, Deuther-Conrad W, Huang Y, Jia H. Development of a Highly Specific 18F-Labeled Radioligand for Imaging of the Sigma-2 Receptor in Brain Tumors. J Med Chem 2023; 66:12840-12857. [PMID: 37704582 DOI: 10.1021/acs.jmedchem.3c00735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Novel ligands with the 6,7-dimethoxy-1,2,3,4-tetrahydroisoquinoline or 5,6-dimethoxyisoindoline pharmacophore were designed and synthesized for evaluation of their structure-activity relationship to the sigma-2 (σ2) receptor and developed as suitable PET radioligands. Compound 1 was found to possess nanomolar affinity (Ki(σ1) = 2.57 nM) for the σ2 receptor, high subtype selectivity (>2000-fold), and high selectivity over 40 other receptors and transporters. Radioligand [18F]1 was prepared with radiochemical yield of 37-54%, > 99% radiochemical purity, and molar activity of 107-189 GBq/μmol. Biodistribution and blocking studies in mice and micro-PET/CT imaging of [18F]1 in rats indicated excellent binding specificity to the σ2 receptors in vivo. Micro-PET/CT imaging of [18F]1 in the U87MG glioma xenograft model demonstrated clear tumor visualization with high tumor uptake and tumor-to-background ratio. Co-injection with CM398 (5 μmol/kg) led to a remarkable reduction of tumor uptake (80%, 60-70 min), indicating high specific binding of [18F]1 in U87MG glioma xenografts.
Collapse
Affiliation(s)
- Tao Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
- Department of Nuclear Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jingqi Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Leyuan Chen
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaodan An
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Winnie Deuther-Conrad
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520-8048, United States
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
10
|
Sorbi C, Belluti S, Atene CG, Marocchi F, Linciano P, Roy N, Paradiso E, Casarini L, Ronsisvalle S, Zanocco-Marani T, Brasili L, Lanfrancone L, Imbriano C, Di Rocco G, Franchini S. BS148 Reduces the Aggressiveness of Metastatic Melanoma via Sigma-2 Receptor Targeting. Int J Mol Sci 2023; 24:ijms24119684. [PMID: 37298633 DOI: 10.3390/ijms24119684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The management of advanced-stage melanoma is clinically challenging, mainly because of its resistance to the currently available therapies. Therefore, it is important to develop alternative therapeutic strategies. The sigma-2 receptor (S2R) is overexpressed in proliferating tumor cells and represents a promising vulnerability to target. Indeed, we have recently identified a potent S2R modulator (BS148) that is effective in melanoma. To elucidate its mechanism of action, we designed and synthesized a BS148 fluorescent probe that enters SK-MEL-2 melanoma cells as assessed using confocal microscopy analysis. We show that S2R knockdown significantly reduces the anti-proliferative effect induced by BS148 administration, indicating the engagement of S2R in BS148-mediated cytotoxicity. Interestingly, BS148 treatment showed similar molecular effects to S2R RNA interference-mediated knockdown. We demonstrate that BS148 administration activates the endoplasmic reticulum stress response through the upregulation of protein kinase R-like ER kinase (PERK), activating transcription factor 4 (ATF4) genes, and C/EBP homologous protein (CHOP). Furthermore, we show that BS148 treatment downregulates genes related to the cholesterol pathway and activates the MAPK signaling pathway. Finally, we translate our results into patient-derived xenograft (PDX) cells, proving that BS148 treatment reduces melanoma cell viability and migration. These results demonstrate that BS148 is able to inhibit metastatic melanoma cell proliferation and migration through its interaction with the S2R and confirm its role as a promising target to treat cancer.
Collapse
Affiliation(s)
- Claudia Sorbi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Claudio Giacinto Atene
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Federica Marocchi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Neena Roy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Simone Ronsisvalle
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Tommaso Zanocco-Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Livio Brasili
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
11
|
Edmonds CE, O'Brien SR, Mankoff DA, Pantel AR. Novel applications of molecular imaging to guide breast cancer therapy. Cancer Imaging 2022; 22:31. [PMID: 35729608 PMCID: PMC9210593 DOI: 10.1186/s40644-022-00468-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
The goals of precision oncology are to provide targeted drug therapy based on each individual’s specific tumor biology, and to enable the prediction and early assessment of treatment response to allow treatment modification when necessary. Thus, precision oncology aims to maximize treatment success while minimizing the side effects of inadequate or suboptimal therapies. Molecular imaging, through noninvasive assessment of clinically relevant tumor biomarkers across the entire disease burden, has the potential to revolutionize clinical oncology, including breast oncology. In this article, we review breast cancer positron emission tomography (PET) imaging biomarkers for providing early response assessment and predicting treatment outcomes. For 2-18fluoro-2-deoxy-D-glucose (FDG), a marker of cellular glucose metabolism that is well established for staging multiple types of malignancies including breast cancer, we highlight novel applications for early response assessment. We then review current and future applications of novel PET biomarkers for imaging the steroid receptors, including the estrogen and progesterone receptors, the HER2 receptor, cellular proliferation, and amino acid metabolism.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
12
|
Discovery of novel, selective, functionalized 5-(2-(5-arylhexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)ethyl)-γ-butyrolactone sigma-2 ligands. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02909-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Discovery and development of brain-penetrant 18F-labeled radioligands for neuroimaging of the sigma-2 receptors. Acta Pharm Sin B 2022; 12:1406-1415. [PMID: 35530149 PMCID: PMC9069315 DOI: 10.1016/j.apsb.2021.08.029] [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: 05/28/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022] Open
Abstract
We have discovered and synthesized a series of indole-based derivatives as novel sigma-2 (σ 2) receptor ligands. Two ligands with high σ 2 receptor affinity and subtype selectivity were then radiolabeled with F-18 in good radiochemical yields and purities, and evaluated in rodents. In biodistribution studies in male ICR mice, radioligand [18F]9, or 1-(4-(5,6-dimethoxyisoindolin-2-yl)butyl)-4-(2-[18F]fluoroethoxy)-1H-indole, was found to display high brain uptake and high brain-to-blood ratio. Pretreatment of animals with the selective σ 2 receptor ligand CM398 led to significant reductions in both brain uptake (29%-54%) and brain-to-blood ratio (60%-88%) of the radioligand in a dose-dependent manner, indicating high and saturable specific binding of [18F]9 to σ 2 receptors in the brain. Further, ex vivo autoradiography in male ICR mice demonstrated regionally heterogeneous specific binding of [18F]9 in the brain that is consistent with the distribution pattern of σ 2 receptors. Dynamic positron emission tomography imaging confirmed regionally distinct distribution and high levels of specific binding for [18F]9 in the rat brain, along with appropriate tissue kinetics. Taken together, results from our current study indicated the novel radioligand [18F]9 as the first highly specific and promising imaging agent for σ 2 receptors in the brain.
Collapse
|
14
|
|
15
|
Balma M, Liberini V, Racca M, Laudicella R, Bauckneht M, Buschiazzo A, Nicolotti DG, Peano S, Bianchi A, Albano G, Quartuccio N, Abgral R, Morbelli SD, D'Alessandria C, Terreno E, Huellner MW, Papaleo A, Deandreis D. Non-conventional and Investigational PET Radiotracers for Breast Cancer: A Systematic Review. Front Med (Lausanne) 2022; 9:881551. [PMID: 35492341 PMCID: PMC9039137 DOI: 10.3389/fmed.2022.881551] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is one of the most common malignancies in women, with high morbidity and mortality rates. In breast cancer, the use of novel radiopharmaceuticals in nuclear medicine can improve the accuracy of diagnosis and staging, refine surveillance strategies and accuracy in choosing personalized treatment approaches, including radioligand therapy. Nuclear medicine thus shows great promise for improving the quality of life of breast cancer patients by allowing non-invasive assessment of the diverse and complex biological processes underlying the development of breast cancer and its evolution under therapy. This review aims to describe molecular probes currently in clinical use as well as those under investigation holding great promise for personalized medicine and precision oncology in breast cancer.
Collapse
Affiliation(s)
- Michele Balma
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
- *Correspondence: Michele Balma
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| | - Manuela Racca
- Nuclear Medicine Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | | | - Simona Peano
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Andrea Bianchi
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Giovanni Albano
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Civico di Cristina and Benfratelli Hospitals, Palermo, Italy
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| | - Silvia Daniela Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | | | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular & Preclinical Imaging Centers, University of Turin, Turin, Italy
| | - Martin William Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alberto Papaleo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Désirée Deandreis
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| |
Collapse
|
16
|
Bhandare RR, Sigalapalli DK, Shaik AB, Canney DJ, Blass BE. Selectivity profile comparison for certain γ-butyrolactone and oxazolidinone-based ligands on a sigma 2 receptor over sigma 1: a molecular docking approach. RSC Adv 2022; 12:20096-20109. [PMID: 35919619 PMCID: PMC9272471 DOI: 10.1039/d2ra03497b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/04/2022] [Indexed: 11/21/2022] Open
Abstract
Sigma receptors (σ1 R and σ2 R) are pharmacologically characterized membrane-bound receptors that bind a wide range of chemical compounds. Alzheimer's disease, traumatic brain injury, schizophrenia, and neuropathic pain have all been associated with abnormal σ2 activity. The σ2 receptor has recently been identified as a potential therapeutic target for inhibiting the formation of amyloid plaques. Numerous laboratories are now investigating the potential of σ2 ligands. Small molecule discovery is the focus of current research, with the goal of using target-based action to treat a variety of illnesses and ailments. Functionalized γ-butyrolactone and oxazolidinone-based ligands, in particular, are pharmacologically important scaffolds in drug discovery research and have been thoroughly examined for σ2 receptor efficacy. The purpose of this study was to evaluate the pharmacophoric features of different σ2 receptor ligands using in silico techniques. This study used a library of 58 compounds having a γ-butyrolactone and oxazolidinone core. To investigate the binding characteristics of the ligands with the σ2 receptor, a 3D homology model was developed. To understand the binding pattern of the γ-butyrolactone and oxazolidinone based ligands, molecular docking studies were performed on both σ1 and σ2 receptors. Furthermore, MM/GBSA binding energy calculations were used to confirm the binding of ligands on the σ2 over σ1 receptor. These in silico findings will aid in the discovery of selective σ2 ligands with good pharmacophoric properties and potency in the future. Selective action of γ-butyrolactones and oxazolidinones towards σ2 receptor.![]()
Collapse
Affiliation(s)
- Richie R. Bhandare
- College of Pharmacy & Health Sciences, Ajman University, PO Box 340, Ajman, United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Dilep Kumar Sigalapalli
- Department of Pharmaceutical Chemistry, Vignan Pharmacy College, Jawaharlal Nehru Technological University, Vadlamudi, 522213, Andhra Pradesh, India
| | - Afzal B. Shaik
- Department of Pharmaceutical Chemistry, Vignan Pharmacy College, Jawaharlal Nehru Technological University, Vadlamudi, 522213, Andhra Pradesh, India
| | - Daniel J. Canney
- Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 194140, USA
| | - Benjamin E. Blass
- Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 194140, USA
| |
Collapse
|
17
|
Yu M, Zhang Q, Xu S, Yin T, Li F. Successful treatment of refractory retroperitoneal Epstein-Barr virus-positive diffuse large B-cell lymphoma with secondary hemophagocytic syndrome by sequential combination regimen of PD-1 blockade and chimeric antigen receptor T cells: a case report. Anticancer Drugs 2022; 33:e769-e775. [PMID: 34387604 DOI: 10.1097/cad.0000000000001187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Epstein-Barr virus (EBV) is convincingly contributed to the development of several types of lymphomas such as NK/T cell lymphoma, Burkitt lymphoma, plasmablastic lymphoma, and diffuse large B cell lymphoma (DLBCL). Herein, we reported an atypical case of EBV-positive DLBCL in an immunocompetent young male patient who presented with epistaxis due to hypergammaglobulinemia. 2-Deoxy-2-[fluorine-8] fluoro-d-glucose PET/computed tomography showed multiple highly metabolic retroperitoneal tissue masses with the involvement of bilateral adrenal gland. Ultrasonography-guided biopsy revealed a significant number of lymphocytes and plasma-like cells that are immunopositive for plasma-cell markers and partly positive for pan-B cell markers. The Ki-67 proliferation index was 20%. The extensive distribution of EBV-encoded small RNAs was confirmed by in-situ hybridization. Due to atypical/overlapping pathological characteristics, it was initially misdiagnosed as extramedullary plasmacytoma and treated with two cycles of bortezomib, lenalidomide, and dexamethasone. Disease progression occurred and pathology consultation for the retroperitoneal biopsies modified the diagnosis to EBV-positive DLBCL with plasma cell differentiation. The treatment was adjusted to etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, rituximab, and lenalidomide (R2-EPOCH), but no response was observed after three cycles of treatment and he developed hemophagocytic syndrome during treatment. A monotherapy of anti-programmed cell death-1 (PD-1) treatment with tiririzumab was administered, successfully controlling hemophagocytic syndrome and EBV infection. The response assessment was partial for EBV-positive DLBCL, subsequent anti-CD19 chimeric antigen receptor-T (CAR-T) cell therapy resulted in complete remission including lumps, immunoglobulins, and negative EBV-DNA 1.5 months later. The present case study proved the possibility of PD-1 blockade in controlling EBV infection and associated hemophagocytic syndrome and offered an example of the combination of CAR-T therapy and PD-1 blockade for refractory EBV-positive DLBCL in clinic.
Collapse
Affiliation(s)
- Min Yu
- Department of Hematology, The First Affiliated Hospital of Nanchang University
- Department of Lymphomatous diseases, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province
| | - Qian Zhang
- Department of Hematology, The First Affiliated Hospital of Nanchang University
- Department of Lymphomatous diseases, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province
| | - Shan Xu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Ting Yin
- Department of Hematology, The First Affiliated Hospital of Nanchang University
- Department of Lymphomatous diseases, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University
- Department of Lymphomatous diseases, Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province
| |
Collapse
|
18
|
QSAR-Based Computational Approaches to Accelerate the Discovery of Sigma-2 Receptor (S2R) Ligands as Therapeutic Drugs. Molecules 2021; 26:molecules26175270. [PMID: 34500703 PMCID: PMC8434483 DOI: 10.3390/molecules26175270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
S2R overexpression is associated with various forms of cancer as well as both neuropsychiatric disorders (e.g., schizophrenia) and neurodegenerative diseases (Alzheimer’s disease: AD). In the present study, three ligand-based methods (QSAR modeling, pharmacophore mapping, and shape-based screening) were implemented to select putative S2R ligands from the DrugBank library comprising 2000+ entries. Four separate optimization algorithms (i.e., stepwise regression, Lasso, genetic algorithm (GA), and a customized extension of GA called GreedGene) were adapted to select descriptors for the QSAR models. The subsequent biological evaluation of selected compounds revealed that three FDA-approved drugs for unrelated therapeutic indications exhibited sub-1 uM binding affinity for S2R. In particular, the antidepressant drug nefazodone elicited a S2R binding affinity Ki = 140 nM. A total of 159 unique S2R ligands were retrieved from 16 publications for model building, validation, and testing. To our best knowledge, the present report represents the first case to develop comprehensive QSAR models sourced by pooling and curating a large assemblage of structurally diverse S2R ligands, which should prove useful for identifying new drug leads and predicting their S2R binding affinity prior to the resource-demanding tasks of chemical synthesis and biological evaluation.
Collapse
|
19
|
Fallica AN, Pittalà V, Modica MN, Salerno L, Romeo G, Marrazzo A, Helal MA, Intagliata S. Recent Advances in the Development of Sigma Receptor Ligands as Cytotoxic Agents: A Medicinal Chemistry Perspective. J Med Chem 2021; 64:7926-7962. [PMID: 34076441 PMCID: PMC8279423 DOI: 10.1021/acs.jmedchem.0c02265] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Since their discovery
as distinct receptor proteins, the specific
physiopathological role of sigma receptors (σRs) has been deeply
investigated. It has been reported that these proteins, classified
into two subtypes indicated as σ1 and σ2, might play a pivotal role in cancer growth, cell proliferation,
and tumor aggressiveness. As a result, the development of selective
σR ligands with potential antitumor properties attracted significant
attention as an emerging theme in cancer research. This perspective
deals with the recent advances of σR ligands as novel cytotoxic
agents, covering articles published between 2010 and 2020. An up-to-date
description of the medicinal chemistry of selective σ1R and σ2R ligands with antiproliferative and cytotoxic
activities has been provided, including major pharmacophore models
and comprehensive structure–activity relationships for each
main class of σR ligands.
Collapse
Affiliation(s)
- Antonino N Fallica
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Mohamed A Helal
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, sixth of October, Giza 12578, Egypt.,Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
20
|
Faheem, Karan Kumar B, Venkata Gowri Chandra Sekhar K, Chander S, Kunjiappan S, Murugesan S. 1,2,3,4-Tetrahydroisoquinoline (THIQ) as privileged scaffold for anticancer de novo drug design. Expert Opin Drug Discov 2021; 16:1119-1147. [PMID: 33908322 DOI: 10.1080/17460441.2021.1916464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Cancer is a dreadful disorder that is emerging as one of the leading causes of mortality across the globe. The complex tumor environment, supplemented with drawbacks of the existing drugs, has made it a global health concern. The Tetrahydroisoquinoline (THIQ) ring holds an important position in medicinal chemistry due to its wide range of pharmacological properties. Several THIQ based natural products have been previously explored for their antitumor properties, making it a vital scaffold for anticancer drug design.Areas covered: This review article addresses the potential of THIQ as anticancer agents. Various medicinal chemistry strategies employed for the design and development of THIQ analogs as inhibitors or modulators of relevant anticancer targets have been discussed in detail. Moreover, the common strategies employed for the synthesis of the core scaffold are also highlighted.Expert opinion: Evidently, THIQs have tremendous potential in anticancer drug design. Some of these analogs exhibited potent activity against various cancer molecular targets. However, there are some drawbacks, such as selectivity that need addressing. The synthetic ease for constructing the core scaffold complimented with its reactivity makes it ideal for further structure-activity relationship studies. For these reasons, THIQ is a privileged scaffold for the design and development of novel anticancer agents.
Collapse
Affiliation(s)
- Faheem
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | | | - Subhash Chander
- Amity Institute of Phytomedicine and Phytochemistry, Amity University Uttar Pradesh, Noida, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| |
Collapse
|
21
|
Romine PE, Peterson LM, Kurland BF, Byrd DW, Novakova-Jiresova A, Muzi M, Specht JM, Doot RK, Link JM, Krohn KA, Kinahan PE, Mankoff DA, Linden HM. 18F-fluorodeoxyglucose (FDG) PET or 18F-fluorothymidine (FLT) PET to assess early response to aromatase inhibitors (AI) in women with ER+ operable breast cancer in a window-of-opportunity study. Breast Cancer Res 2021; 23:88. [PMID: 34425871 PMCID: PMC8381552 DOI: 10.1186/s13058-021-01464-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE This study evaluated the ability of 18F-Fluorodeoxyglucose (FDG) and 18F-Fluorothymidine (FLT) imaging with positron emission tomography (PET) to measure early response to endocrine therapy from baseline to just prior to surgical resection in estrogen receptor positive (ER+) breast tumors. METHODS In two separate studies, women with early stage ER+ breast cancer underwent either paired FDG-PET (n = 22) or FLT-PET (n = 27) scans prior to endocrine therapy and again in the pre-operative setting. Tissue samples for Ki-67 were taken for all patients both prior to treatment and at the time of surgery. RESULTS FDG maximum standardized uptake value (SUVmax) declined in 19 of 22 lesions (mean 17% (range -45 to 28%)). FLT SUVmax declined in 24 of 27 lesions (mean 26% (range -77 to 7%)). The Ki-67 index declined in both studies, from pre-therapy (mean 23% (range 1 to 73%)) to surgery [mean 8% (range < 1 to 41%)]. Pre- and post-therapy PET measures showed strong rank-order agreement with Ki-67 percentages for both tracers; however, the percent change in FDG or FLT SUVmax did not demonstrate a strong correlation with Ki-67 index change or Ki-67 at time of surgery. CONCLUSIONS A window-of-opportunity approach using PET imaging to assess early response of breast cancer therapy is feasible. FDG and FLT-PET imaging following a short course of neoadjuvant endocrine therapy demonstrated measurable changes in SUVmax in early stage ER+ positive breast cancers. The percentage change in FDG and FLT-PET uptake did not correlate with changes in Ki-67; post-therapy SUVmax for both tracers was significantly associated with post-therapy Ki-67, an established predictor of endocrine therapy response.
Collapse
Affiliation(s)
- Perrin E. Romine
- grid.34477.330000000122986657Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, 1144 Eastlake (Mail Stop LG-200), Seattle, WA 98109-1023 USA
| | - Lanell M. Peterson
- grid.34477.330000000122986657Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, 1144 Eastlake (Mail Stop LG-200), Seattle, WA 98109-1023 USA
| | - Brenda F. Kurland
- grid.21925.3d0000 0004 1936 9000University of Pittsburgh, Pittsburgh, PA USA
| | - Darrin W. Byrd
- grid.34477.330000000122986657Department of Radiology, University of Washington, Seattle, WA USA
| | - Alena Novakova-Jiresova
- grid.4491.80000 0004 1937 116XDepartment of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Mark Muzi
- grid.34477.330000000122986657Department of Radiology, University of Washington, Seattle, WA USA
| | - Jennifer M. Specht
- grid.34477.330000000122986657Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, 1144 Eastlake (Mail Stop LG-200), Seattle, WA 98109-1023 USA
| | - Robert K. Doot
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA USA
| | - Jeanne M. Link
- grid.5288.70000 0000 9758 5690Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR USA
| | - Kenneth A. Krohn
- grid.5288.70000 0000 9758 5690Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR USA
| | - Paul E. Kinahan
- grid.34477.330000000122986657Department of Radiology, University of Washington, Seattle, WA USA
| | - David A. Mankoff
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA USA
| | - Hannah M. Linden
- grid.34477.330000000122986657Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, 1144 Eastlake (Mail Stop LG-200), Seattle, WA 98109-1023 USA
| |
Collapse
|
22
|
Sigma-2 Receptor-A Potential Target for Cancer/Alzheimer's Disease Treatment via Its Regulation of Cholesterol Homeostasis. Molecules 2020; 25:molecules25225439. [PMID: 33233619 PMCID: PMC7699687 DOI: 10.3390/molecules25225439] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
The sigma receptors were classified into sigma-1 and sigma-2 receptor based on their different pharmacological profiles. In the past two decades, our understanding of the biological and pharmacological properties of the sigma-1 receptor is increasing; however, little is known about the sigma-2 receptor. Recently, the molecular identity of the sigma-2 receptor has been identified as TMEM97. Although more and more evidence has showed that sigma-2 ligands have the ability to treat cancer and Alzheimer’s disease (AD), the mechanisms connecting these two diseases are unknown. Data obtained over the past few years from human and animal models indicate that cholesterol homeostasis is altered in AD and cancer, underscoring the importance of cholesterol homeostasis in AD and cancer. In this review, based on accumulated evidence, we proposed that the beneficial roles of sigma-2 ligands in cancer and AD might be mediated by their regulation of cholesterol homeostasis.
Collapse
|
23
|
Cordone P, Namballa HK, Harding WW. First synthesis of thiazepino[3,4‐a]isoquinolines, a facile new synthetic route to diazepino[3,4‐a]isoquinolines and assessment of their dopamine and σ receptor affinities. J Heterocycl Chem 2020; 57:3709-3713. [DOI: 10.1002/jhet.4086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Pierpaolo Cordone
- Department of Chemistry Hunter College, City University of New York New York New York USA
- Ph.D. Program in Biochemistry CUNY Graduate Center New York New York USA
| | - Hari Krishna Namballa
- Department of Chemistry Hunter College, City University of New York New York New York USA
| | - Wayne Wesley Harding
- Department of Chemistry Hunter College, City University of New York New York New York USA
- Ph.D. Program in Biochemistry CUNY Graduate Center New York New York USA
- Ph.D. Program in Chemistry CUNY Graduate Center New York New York USA
| |
Collapse
|
24
|
Zeng C, Riad A, Mach RH. The Biological Function of Sigma-2 Receptor/TMEM97 and Its Utility in PET Imaging Studies in Cancer. Cancers (Basel) 2020; 12:E1877. [PMID: 32668577 PMCID: PMC7409002 DOI: 10.3390/cancers12071877] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 11/16/2022] Open
Abstract
The sigma-2 receptor was originally defined pharmacologically and recently identified as TMEM97. TMEM97 has been validated as a biomarker of proliferative status and the radioligand of TMEM97, [18F]ISO-1, has been developed and validated as a PET imaging biomarker of proliferative status of tumors and as a predictor of the cancer therapy response. [18F]ISO-1 PET imaging should be useful to guide treatment for cancer patients. TMEM97 is a membrane-bound protein and localizes in multiple subcellular organelles including endoplasmic reticulum and lysosomes. TMEM97 plays distinct roles in cancer. It is reported that TMEM97 is upregulated in some tumors but downregulated in other tumors and it is required for cell proliferation in certain tumor cells. TMEM97 plays important roles in cholesterol homeostasis. TMEM97 expression is regulated by cholesterol-regulating signals such as sterol depletion and SREBP expression levels. TMEM97 regulates cholesterol trafficking processes such as low density lipoprotein (LDL) uptake by forming complexes with PGRMC1 and low density lipoprotein receptor (LDLR), as well as cholesterol transport out of lysosome by interacting with and regulating NPC1 protein. Understanding molecular functions of TMEM97 in proliferation and cholesterol metabolism will be important to develop strategies to diagnose and treat cancer and cholesterol disorders using a rich collection of TMEM97 radiotracers and ligands.
Collapse
Affiliation(s)
| | | | - Robert H. Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.Z.); (A.R.)
| |
Collapse
|
25
|
Amata E, Dichiara M, Gentile D, Marrazzo A, Turnaturi R, Arena E, La Mantia A, Tomasello BR, Acquaviva R, Di Giacomo C, Rescifina A, Prezzavento O. Sigma Receptor Ligands Carrying a Nitric Oxide Donor Nitrate Moiety: Synthesis, In Silico, and Biological Evaluation. ACS Med Chem Lett 2020; 11:889-894. [PMID: 32435401 DOI: 10.1021/acsmedchemlett.9b00661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
We report the development of molecular hybrids in which a nitrate group serving as nitric oxide (NO) donor is covalently joined to σ receptor ligands to give candidates for double-targeted cancer therapy. The compounds have been evaluated in radioligand binding assay at both σ receptors and selected compounds tested for NO release. Compounds 9, 15, 18, 19, and 21 were subjected to MTT test. Compound 15 produced a significant reduction of MCF-7 and Caco-2 cellular viability with comparable IC50 as doxorubicin, being also not toxic for fibroblast HFF-1 cells. Compound 15 has shown a σ1 receptor antagonist/σ2 receptor agonist profile. Two derivatives of compound 15 lacking the nitrate group did not induce a reduction of MCF-7 cellular viability, suggesting a potential synergistic effect between the σ receptors and the NO-mediated events. Overall, the combination of NO donor and σ receptors ligands provided compounds with beneficial effects for the treatment of cancer.
Collapse
Affiliation(s)
- Emanuele Amata
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Davide Gentile
- Department of Drug Sciences, Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Emanuela Arena
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Alfonsina La Mantia
- Department of Drug Sciences, Biochemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Barbara Rita Tomasello
- Department of Drug Sciences, Biochemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Rosaria Acquaviva
- Department of Drug Sciences, Biochemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Claudia Di Giacomo
- Department of Drug Sciences, Biochemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
| |
Collapse
|