1
|
Gálvez‐Montosa F, Peduzzi G, Sanchez‐Maldonado JM, ter Horst R, Cabrera‐Serrano AJ, Gentiluomo M, Macauda A, Luque N, Ünal P, García‐Verdejo FJ, Li Y, López López JA, Stein A, Bueno‐de‐Mesquita HB, Arcidiacono PG, Zanette DL, Kahlert C, Perri F, Soucek P, Talar‐Wojnarowska R, Theodoropoulos GE, Izbicki JR, Tamás H, Van Laarhoven H, Nappo G, Petrone MC, Lovecek M, Vermeulen RCH, Adamonis K, Reyes‐Zurita FJ, Holleczek B, Sumskiene J, Mohelníková‐Duchoňová B, Lawlor RT, Pezzilli R, Aoki MN, Pasquali C, Petrenkiene V, Basso D, Bunduc S, Comandatore A, Brenner H, Ermini S, Vanella G, Goetz MR, Archibugi L, Lucchesi M, Uzunoglu FG, Busch O, Milanetto AC, Puzzono M, Kupcinskas J, Morelli L, Sperti C, Carrara S, Capurso G, van Eijck CHJ, Oliverius M, Roth S, Tavano F, Kaaks R, Szentesi A, Vodickova L, Luchini C, Schöttker B, Landi S, Dohan O, Tacelli M, Greenhalf W, Gazouli M, Neoptolemos JP, Cavestro GM, Boggi U, Latiano A, Hegyi P, Ginocchi L, Netea MG, Sánchez‐Rovira P, Canzian F, Campa D, Sainz J. Polymorphisms within autophagy-related genes as susceptibility biomarkers for pancreatic cancer: A meta-analysis of three large European cohorts and functional characterization. Int J Cancer 2025; 156:339-352. [PMID: 39319538 PMCID: PMC11578083 DOI: 10.1002/ijc.35196] [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: 04/04/2024] [Revised: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers with patients having unresectable or metastatic disease at diagnosis, with poor prognosis and very short survival. Given that genetic variation within autophagy-related genes influences autophagic flux and susceptibility to solid cancers, we decided to investigate whether 55,583 single nucleotide polymorphisms (SNPs) within 234 autophagy-related genes could influence the risk of developing PDAC in three large independent cohorts of European ancestry including 12,754 PDAC cases and 324,926 controls. The meta-analysis of these populations identified, for the first time, the association of the BIDrs9604789 variant with an increased risk of developing the disease (ORMeta = 1.31, p = 9.67 × 10-6). We also confirmed the association of TP63rs1515496 and TP63rs35389543 variants with PDAC risk (OR = 0.89, p = 6.27 × 10-8 and OR = 1.16, p = 2.74 × 10-5). Although it is known that BID induces autophagy and TP63 promotes cell growth, cell motility and invasion, we also found that carriers of the TP63rs1515496G allele had increased numbers of FOXP3+ Helios+ T regulatory cells and CD45RA+ T regulatory cells (p = 7.67 × 10-4 and p = 1.56 × 10-3), but also decreased levels of CD4+ T regulatory cells (p = 7.86 × 10-4). These results were in agreement with research suggesting that the TP63rs1515496 variant alters binding sites for FOXA1 and CTCF, which are transcription factors involved in modulating specific subsets of regulatory T cells. In conclusion, this study identifies BID as new susceptibility locus for PDAC and confirms previous studies suggesting that the TP63 gene is involved in the development of PDAC. This study also suggests new pathogenic mechanisms of the TP63 locus in PDAC.
Collapse
Affiliation(s)
| | | | - José Manuel Sanchez‐Maldonado
- Department of Biochemistry and Molecular Biology IUniversity of GranadaGranadaSpain
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Rob ter Horst
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Antonio J. Cabrera‐Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
| | | | - Angelica Macauda
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Natalia Luque
- Department of Medical OncologyComplejo Hospitalario de JaénJaénSpain
| | - Pelin Ünal
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | | | - Angelika Stein
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Paolo Giorgio Arcidiacono
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in HealthCarlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz)CuritibaBrazil
| | - Christoph Kahlert
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Francesco Perri
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | | | - George E. Theodoropoulos
- Colorectal Unit, First Department of Propaedeutic SurgeryMedical School of National and Kapodistrian University of Athens, Hippocration General HospitalAthensGreece
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hussein Tamás
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Hanneke Van Laarhoven
- Department of Medical OncologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamImaging and BiomarkersAmsterdamThe Netherlands
| | - Gennaro Nappo
- Pancreatic UnitIRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | - Maria Chiara Petrone
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - Martin Lovecek
- Department of Surgery IUniversity Hospital OlomoucOlomoucCzech Republic
| | | | - Kestutis Adamonis
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | | | - Bernd Holleczek
- Saarland Cancer RegistrySaarbrückenGermany
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jolanta Sumskiene
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | | | - Rita T. Lawlor
- ARC‐Net Centre for Applied Research on Cancer University of VeronaVeronaItaly
- Department of Diagnostics and Public Health, Section of PathologyUniversity of VeronaVeronaItaly
| | | | - Mateus Nobrega Aoki
- Laboratory for Applied Science and Technology in HealthCarlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz)CuritibaBrazil
| | | | - Vitalija Petrenkiene
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Daniela Basso
- Department of DIMEDLaboratory Medicine, University of PadovaPadovaItaly
| | - Stefania Bunduc
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
- Carol Davila University of Medicine and PharmacyBucharestRomania
- Digestive Diseases and Liver Transplantation CenterFundeni Clinical InstituteBucharestRomania
| | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in MedicineUniversity of PisaPisaItaly
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Giuseppe Vanella
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | - Mara R. Goetz
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Livia Archibugi
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | | | - Faik Guntac Uzunoglu
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Olivier Busch
- Cancer Center AmsterdamImaging and BiomarkersAmsterdamThe Netherlands
- Department of Medical OncologyAmsterdam UMC Location University of AmsterdamAmsterdamThe Netherlands
| | | | - Marta Puzzono
- Gastroenterology and Gastrointestinal Endoscopy UnitVita‐Salute San Raffaele University, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Juozas Kupcinskas
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in MedicineUniversity of PisaPisaItaly
| | | | - Silvia Carrara
- Department of GastroenterologyIRCCS Humanitas Research Hospital – Endoscopic UnitMilanItaly
| | - Gabriele Capurso
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | | | - Martin Oliverius
- Department of Surgery, University Hospital Kralovske Vinohrady, Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Susanne Roth
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Francesca Tavano
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Rudolf Kaaks
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Andrea Szentesi
- Institute for Translational Medicine, Medical SchoolUniversity of PécsPécsHungary
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental MedicineCzech Academy of SciencesPragueCzech Republic
- Institute of Biology and Medical Genetics, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Faculty of Medicine and Biomedical Center in PilsenCharles UniversityPilsenCzech Republic
| | - Claudio Luchini
- ARC‐Net Centre for Applied Research on Cancer University of VeronaVeronaItaly
- Department of Engineering for Innovation in MedicineUniversity of VeronaVeronaItaly
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Orsolya Dohan
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Matteo Tacelli
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - William Greenhalf
- Institute for Health Research Liverpool Pancreas Biomedical Research UnitUniversity of LiverpoolLiverpoolUK
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - John P. Neoptolemos
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy UnitVita‐Salute San Raffaele University, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Ugo Boggi
- Division of General and Transplant SurgeryPisa University HospitalPisaItaly
| | - Anna Latiano
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Péter Hegyi
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
- Institute for Translational Medicine, Medical SchoolUniversity of PécsPécsHungary
- János Szentágothai Research CenterUniversity of PécsPécsHungary
| | - Laura Ginocchi
- Oncologia Massa CarraraAzienda USL Toscana Nord OvestCarraraItaly
| | - Mihai G. Netea
- Centre for Individualised Infection Medicine (CiiM) & TWINCOREjoint Ventures Between the Helmholtz‐Centre for Infection Research (HZI) and the Hannover Medical School (MHH)HannoverGermany
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES)University of BonnBonnGermany
| | | | - Federico Canzian
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Juan Sainz
- Department of Biochemistry and Molecular Biology IUniversity of GranadaGranadaSpain
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP)BarcelonaSpain
| |
Collapse
|
2
|
Gehrke NR, Feng D, Ayub Ali M, Maalouf MA, Holstein SA, Wiemer DF. α-Amino bisphosphonate triazoles serve as GGDPS inhibitors. Bioorg Med Chem Lett 2024; 102:129659. [PMID: 38373465 PMCID: PMC10981527 DOI: 10.1016/j.bmcl.2024.129659] [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: 11/13/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Depletion of cellular levels of geranylgeranyl diphosphate by inhibition of the enzyme geranylgeranyl diphosphate synthase (GGDPS) is a potential strategy for disruption of protein transport by limiting the geranylgeranylation of the Rab proteins that regulate intracellular trafficking. As such, there is interest in the development of GGDPS inhibitors for the treatment of malignancies characterized by abnormal protein production, including multiple myeloma. Our previous work has explored the structure-function relationship of a series of isoprenoid triazole bisphosphonate-based GGDPS inhibitors, with modifications having impact on enzymatic, cellular and in vivo activities. We have synthesized a new series of α-amino bisphosphonates to understand the impact of modifying the alpha position with a moiety that is potentially linkable to other agents. Bioassays evaluating the enzymatic and cellular activities of these compounds demonstrate that incorporation of the α-amino group affords compounds with GGDPS inhibitory activity which is modulated by isoprenoid tail chain length and olefin stereochemistry. These studies provide further insight into the complexity of the structure-function relationship and will enable future efforts focused on tumor-specific drug delivery.
Collapse
Affiliation(s)
- Nathaniel R Gehrke
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, US
| | - Dan Feng
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, US
| | - Md Ayub Ali
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, US; Department of Chemistry, Bangladesh University of Engineering and Technology (BUET), Dhaka-1000, Bangladesh
| | - Mona A Maalouf
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, US
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, US; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, US
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, US; Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, US.
| |
Collapse
|
3
|
Pham AC, Holstein SA, Borgstahl GE. Structural Insight into Geranylgeranyl Diphosphate Synthase (GGDPS) for Cancer Therapy. Mol Cancer Ther 2024; 23:14-23. [PMID: 37756579 PMCID: PMC10762340 DOI: 10.1158/1535-7163.mct-23-0358] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023]
Abstract
Geranylgeranyl diphosphate synthase (GGDPS), the source of the isoprenoid donor in protein geranylgeranylation reactions, has become an attractive target for anticancer therapy due to the reliance of cancers on geranylgeranylated proteins. Current GGDPS inhibitor development focuses on optimizing the drug-target enzyme interactions of nitrogen-containing bisphosphonate-based drugs. To advance GGDPS inhibitor development, understanding the enzyme structure, active site, and ligand/product interactions is essential. Here we provide a comprehensive structure-focused review of GGDPS. We reviewed available yeast and human GGDPS structures and then used AlphaFold modeling to complete unsolved structural aspects of these models. We delineate the elements of higher-order structure formation, product-substrate binding, the electrostatic surface, and small-molecule inhibitor binding. With the rise of structure-based drug design, the information provided here will serve as a valuable tool for rationally optimizing inhibitor selectivity and effectiveness.
Collapse
Affiliation(s)
- Andrew C. Pham
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sarah A. Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gloria E.O. Borgstahl
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- The Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
4
|
Kozalak G, Koşar A. Autophagy-related mechanisms for treatment of multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:838-857. [PMID: 38239705 PMCID: PMC10792488 DOI: 10.20517/cdr.2023.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Multiple myeloma (MM) is a type of hematological cancer that occurs when B cells become malignant. Various drugs such as proteasome inhibitors, immunomodulators, and compounds that cause DNA damage can be used in the treatment of MM. Autophagy, a type 2 cell death mechanism, plays a crucial role in determining the fate of B cells, either promoting their survival or inducing cell death. Therefore, autophagy can either facilitate the progression or hinder the treatment of MM disease. In this review, autophagy mechanisms that may be effective in MM cells were covered and evaluated within the contexts of unfolded protein response (UPR), bone marrow microenvironment (BMME), drug resistance, hypoxia, DNA repair and transcriptional regulation, and apoptosis. The genes that are effective in each mechanism and research efforts on this subject were discussed in detail. Signaling pathways targeted by new drugs to benefit from autophagy in MM disease were covered. The efficacy of drugs that regulate autophagy in MM was examined, and clinical trials on this subject were included. Consequently, among the autophagy mechanisms that are effective in MM, the most suitable ones to be used in the treatment were expressed. The importance of 3D models and microfluidic systems for the discovery of new drugs for autophagy and personalized treatment was emphasized. Ultimately, this review aims to provide a comprehensive overview of MM disease, encompassing autophagy mechanisms, drugs, clinical studies, and further studies.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
5
|
Clavero E, Sanchez-Maldonado JM, Macauda A, Ter Horst R, Sampaio-Marques B, Jurczyszyn A, Clay-Gilmour A, Stein A, Hildebrandt MAT, Weinhold N, Buda G, García-Sanz R, Tomczak W, Vogel U, Jerez A, Zawirska D, Wątek M, Hofmann JN, Landi S, Spinelli JJ, Butrym A, Kumar A, Martínez-López J, Galimberti S, Sarasquete ME, Subocz E, Iskierka-Jażdżewska E, Giles GG, Rybicka-Ramos M, Kruszewski M, Abildgaard N, Verdejo FG, Sánchez Rovira P, da Silva Filho MI, Kadar K, Razny M, Cozen W, Pelosini M, Jurado M, Bhatti P, Dudzinski M, Druzd-Sitek A, Orciuolo E, Li Y, Norman AD, Zaucha JM, Reis RM, Markiewicz M, Rodríguez Sevilla JJ, Andersen V, Jamroziak K, Hemminki K, Berndt SI, Rajkumar V, Mazur G, Kumar SK, Ludovico P, Nagler A, Chanock SJ, Dumontet C, Machiela MJ, Varkonyi J, Camp NJ, Ziv E, Vangsted AJ, Brown EE, Campa D, Vachon CM, Netea MG, Canzian F, Försti A, Sainz J. Polymorphisms within Autophagy-Related Genes as Susceptibility Biomarkers for Multiple Myeloma: A Meta-Analysis of Three Large Cohorts and Functional Characterization. Int J Mol Sci 2023; 24:ijms24108500. [PMID: 37239846 DOI: 10.3390/ijms24108500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Multiple myeloma (MM) arises following malignant proliferation of plasma cells in the bone marrow, that secrete high amounts of specific monoclonal immunoglobulins or light chains, resulting in the massive production of unfolded or misfolded proteins. Autophagy can have a dual role in tumorigenesis, by eliminating these abnormal proteins to avoid cancer development, but also ensuring MM cell survival and promoting resistance to treatments. To date no studies have determined the impact of genetic variation in autophagy-related genes on MM risk. We performed meta-analysis of germline genetic data on 234 autophagy-related genes from three independent study populations including 13,387 subjects of European ancestry (6863 MM patients and 6524 controls) and examined correlations of statistically significant single nucleotide polymorphisms (SNPs; p < 1 × 10-9) with immune responses in whole blood, peripheral blood mononuclear cells (PBMCs), and monocyte-derived macrophages (MDM) from a large population of healthy donors from the Human Functional Genomic Project (HFGP). We identified SNPs in six loci, CD46, IKBKE, PARK2, ULK4, ATG5, and CDKN2A associated with MM risk (p = 4.47 × 10-4-5.79 × 10-14). Mechanistically, we found that the ULK4rs6599175 SNP correlated with circulating concentrations of vitamin D3 (p = 4.0 × 10-4), whereas the IKBKErs17433804 SNP correlated with the number of transitional CD24+CD38+ B cells (p = 4.8 × 10-4) and circulating serum concentrations of Monocyte Chemoattractant Protein (MCP)-2 (p = 3.6 × 10-4). We also found that the CD46rs1142469 SNP correlated with numbers of CD19+ B cells, CD19+CD3- B cells, CD5+IgD- cells, IgM- cells, IgD-IgM- cells, and CD4-CD8- PBMCs (p = 4.9 × 10-4-8.6 × 10-4) and circulating concentrations of interleukin (IL)-20 (p = 0.00082). Finally, we observed that the CDKN2Ars2811710 SNP correlated with levels of CD4+EMCD45RO+CD27- cells (p = 9.3 × 10-4). These results suggest that genetic variants within these six loci influence MM risk through the modulation of specific subsets of immune cells, as well as vitamin D3-, MCP-2-, and IL20-dependent pathways.
Collapse
Affiliation(s)
- Esther Clavero
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain
| | - José Manuel Sanchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
| | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasias Center, Department of Hematology, Jagiellonian University Medical College, 31-066 Kraków, Poland
| | - Alyssa Clay-Gilmour
- Department of Biostatistics and Epidemiology, Arnold School of Public Health, University of South Carolina, Greenville, SC 29208, USA
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Angelika Stein
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Michelle A T Hildebrandt
- Department of Lymphoma-Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabriele Buda
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - Ramón García-Sanz
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Waldemar Tomczak
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland
| | - Ulla Vogel
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark
| | - Andrés Jerez
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, 08035 Barcelona, Spain
| | - Daria Zawirska
- Department of Hematology, University Hospital, 30-688 Kraków, Poland
| | - Marzena Wątek
- Holycross Medical Oncology Center, 25-735 Kielce, Poland
- Institute of Hematology and Transfusion Medicine, 00-791 Warsaw, Poland
| | - Jonathan N Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stefano Landi
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - John J Spinelli
- Division of Population Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Aleksandra Butrym
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Alfred Sokolowski Specialist Hospital in Walbrzych Oncology Support Centre for Clinical Trials, 58-309 Walbrzych, Poland
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | | | - Sara Galimberti
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - María Eugenia Sarasquete
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Edyta Subocz
- Department of Hematology, Military Institute of Medicine, 04-141 Warsaw, Poland
| | | | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Malwina Rybicka-Ramos
- Department of Hematology, Specialist Hospital No. 1 in Bytom, Academy of Silesia, Faculty of Medicine, 40-055 Katowice, Poland
| | - Marcin Kruszewski
- Department of Hematology, University Hospital No. 2, 85-168 Bydgoszcz, Poland
| | - Niels Abildgaard
- Department of Hematology, Odense University Hospital, DK-5000 Odense, Denmark
| | | | - Pedro Sánchez Rovira
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain
| | - Miguel Inacio da Silva Filho
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany
| | | | - Małgorzata Razny
- Department of Hematology, Rydygier Hospital, 31-826 Cracow, Poland
| | - Wendy Cozen
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, Department of Pathology, School of Medicine, Susan and Henry Samueli College of Health Sciences, Chao Family Comprehensive Cancer Center, University of California at Irvine, Irvine, CA 92697, USA
| | - Matteo Pelosini
- U.O. Dipartimento di Ematologia, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Manuel Jurado
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Medicine, University of Granada, 18012 Granada, Spain
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Program in Epidemiology, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Marek Dudzinski
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Agnieszka Druzd-Sitek
- Department of Lymphoproliferative Diseases, Maria Skłodowska Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Enrico Orciuolo
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Aaron D Norman
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
- Genetic Epidemiology and Risk Assessment Program, Mayo Clinic Comprehensive Cancer Center, Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal and ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
| | - Miroslaw Markiewicz
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | | | - Vibeke Andersen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University Hospital of Southern Denmark, DK-6200 Aabenraa, Denmark
| | - Krzysztof Jamroziak
- Department of Hematology, Transplantology and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Kari Hemminki
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vicent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Grzegorz Mazur
- Department of Internal Diseases, Occupational Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Shaji K Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charles Dumontet
- UMR INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, 69008 Lyon, France
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Nicola J Camp
- Division of Hematology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94143, USA
| | - Annette Juul Vangsted
- Department of Hematology, Rigshospitalet, Copenhagen University, DK-2100 Copenhagen, Denmark
| | - Elizabeth E Brown
- Department of Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Celine M Vachon
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Asta Försti
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| |
Collapse
|
6
|
Muehlebach ME, Holstein SA. Geranylgeranyl diphosphate synthase: Role in human health, disease and potential therapeutic target. Clin Transl Med 2023; 13:e1167. [PMID: 36650113 PMCID: PMC9845123 DOI: 10.1002/ctm2.1167] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023] Open
Abstract
Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthesis pathway, is responsible for the production of geranylgeranyl pyrophosphate (GGPP). GGPP serves as a substrate for the post-translational modification (geranylgeranylation) of proteins, including those belonging to the Ras superfamily of small GTPases. These proteins play key roles in signalling pathways, cytoskeletal regulation and intracellular transport, and in the absence of the prenylation modification, cannot properly localise and function. Aberrant expression of GGDPS has been implicated in various human pathologies, including liver disease, type 2 diabetes, pulmonary disease and malignancy. Thus, this enzyme is of particular interest from a therapeutic perspective. Here, we review the physiological function of GGDPS as well as its role in pathophysiological processes. We discuss the current GGDPS inhibitors under development and the therapeutic implications of targeting this enzyme.
Collapse
Affiliation(s)
- Molly E. Muehlebach
- Cancer Research Doctoral ProgramUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sarah A. Holstein
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
7
|
Haney SL, Varney ML, Williams JT, Smith LM, Talmon G, Holstein SA. Geranylgeranyl diphosphate synthase inhibitor and proteasome inhibitor combination therapy in multiple myeloma. Exp Hematol Oncol 2022; 11:5. [PMID: 35139925 PMCID: PMC8827146 DOI: 10.1186/s40164-022-00261-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/25/2022] [Indexed: 11/11/2022] Open
Abstract
Background Multiple myeloma (MM) remains an incurable malignancy, despite the advent of therapies such as proteosome inhibitors (PIs) that disrupt protein homeostasis and induce ER stress. We have pursued inhibition of geranylgeranyl diphosphate synthase (GGDPS) as a novel mechanism by which to target protein homeostasis in MM cells. GGDPS inhibitors (GGSI) disrupt Rab geranylgeranylation, which in turn results in perturbation of Rab-mediated protein trafficking, leading to accumulation of intracellular monoclonal protein, induction of ER stress and apoptosis. Our lead GGSI, RAM2061, has demonstrated favorable pharmacokinetic properties and in vivo efficacy. Here we sought to evaluate if combination therapy with GGSI and PI would result in enhanced disruption of the unfolded protein response (UPR) and increase anti-MM efficacy. Methods MTT assays were conducted to evaluate the cytotoxic effects of combining RAM2061 with bortezomib in human MM cells. The effects of RAM2061 and/or PI (bortezomib or carfilzomib) on markers of UPR and apoptosis were evaluated by a combination of immunoblot (ATF4, IRE1, p-eIF2a, cleaved caspases and PARP), RT-PCR (ATF4, ATF6, CHOP, PERK, IRE1) and flow cytometry (Annexin-V). Induction of immunogenic cell death (ICD) was assessed by immunoblot (HMGB1 release) and flow cytometry (calreticulin translocation). Cell assays were performed using both concurrent and sequential incubation with PIs. To evaluate the in vivo activity of GGSI/PI, a flank xenograft using MM.1S cells was performed. Results Isobologram analysis of cytotoxicity data revealed that sequential treatment of bortezomib with RAM2061 has a synergistic effect in MM cells, while concurrent treatment was primarily additive or mildly antagonistic. The effect of PIs on augmenting RAM2061-induced upregulation of UPR and apoptotic markers was dependent on timing of the PI exposure. Combination treatment with RAM2061 and bortezomib enhanced activation of ICD pathway markers. Lastly, combination treatment slowed MM tumor growth and lengthened survival in a MM xenograft model without evidence of off-target toxicity. Conclusion We demonstrate that GGSI/PI treatment can potentiate activation of the UPR and apoptotic pathway, as well as induce upregulation of markers associated with the ICD pathway. Collectively, these findings lay the groundwork for future clinical studies evaluating combination GGSI and PI therapy in patients with MM. Supplementary Information The online version contains supplementary material available at 10.1186/s40164-022-00261-6.
Collapse
Affiliation(s)
- Staci L Haney
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Michelle L Varney
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jacob T Williams
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Lynette M Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sarah A Holstein
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
8
|
Fairweather AER, Goetz DB, Schroeder CM, Bhuiyan NH, Varney ML, Wiemer DF, Holstein SA. Impact of α-modifications on the activity of triazole bisphosphonates as geranylgeranyl diphosphate synthase inhibitors. Bioorg Med Chem 2021; 44:116307. [PMID: 34298413 DOI: 10.1016/j.bmc.2021.116307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Agents that inhibit the enzyme geranylgeranyl diphosphate synthase (GGDPS) have anti-cancer activity and our prior studies have investigated the structure-function relationship for a family of isoprenoid triazole bisphosphonates as GGDPS inhibitors. To further explore this structure-function relationship, a series of novel α-modified triazole phosphonates was prepared and evaluated for activity as GGDPS inhibitors in enzyme and cell-based assays. These studies revealed flexibility at the α position of the bisphosphonate derivatives with respect to being able to accommodate a variety of substituents without significantly affecting potency compared to the parent unsubstituted inhibitor. However, the monophosphonate derivatives lacked activity. These studies further our understanding of the structure-function relationship of the triazole-based GGDPS inhibitors and lay the foundation for future studies evaluating the impact of α-modifications on in vivo activity.
Collapse
Affiliation(s)
| | - Daniel B Goetz
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Chloe M Schroeder
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Nazmul H Bhuiyan
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Michelle L Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA; Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
9
|
Sun S, Tao J, Sedghizadeh PP, Cherian P, Junka AF, Sodagar E, Xing L, Boeckman RK, Srinivasan V, Yao Z, Boyce BF, Lipe B, Neighbors JD, Russell RGG, McKenna CE, Ebetino FH. Bisphosphonates for delivering drugs to bone. Br J Pharmacol 2021; 178:2008-2025. [PMID: 32876338 DOI: 10.1111/bph.15251] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
Advances in the design of potential bone-selective drugs for the treatment of various bone-related diseases are creating exciting new directions for multiple unmet medical needs. For bone-related cancers, off-target/non-bone toxicities with current drugs represent a significant barrier to the quality of life of affected patients. For bone infections and osteomyelitis, bacterial biofilms on infected bones limit the efficacy of antibiotics because it is hard to access the bacteria with current approaches. Promising new experimental approaches to therapy, based on bone-targeting of drugs, have been used in animal models of these conditions and demonstrate improved efficacy and safety. The success of these drug-design strategies bodes well for the development of therapies with improved efficacy for the treatment of diseases affecting the skeleton. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.
Collapse
Affiliation(s)
| | - Jianguo Tao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Parish P Sedghizadeh
- Center for Biofilms, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | | | - Adam F Junka
- Department of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw; Wroclaw Research Centre EIT, Wroclaw, Poland
| | - Esmat Sodagar
- Center for Biofilms, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Robert K Boeckman
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | | | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea Lipe
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jeffrey D Neighbors
- BioVinc, Pasadena, CA, USA.,Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - R Graham G Russell
- The Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK.,Department of Oncology and Metabolism, The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, California, USA
| | - Frank H Ebetino
- BioVinc, Pasadena, CA, USA.,Department of Chemistry, University of Rochester, Rochester, NY, USA.,Department of Oncology and Metabolism, The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
10
|
Bhuiyan NH, Varney ML, Bhattacharya DS, Payne WM, Mohs AM, Holstein SA, Wiemer DF. ω-Hydroxy isoprenoid bisphosphonates as linkable GGDPS inhibitors. Bioorg Med Chem Lett 2019; 29:126633. [PMID: 31474482 DOI: 10.1016/j.bmcl.2019.126633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 08/20/2019] [Indexed: 02/08/2023]
Abstract
The enzyme geranylgeranyl diphosphate synthase (GGDPS) is a potential therapeutic target for multiple myeloma. Malignant plasma cells produce and secrete large amounts of monoclonal protein, and inhibition of GGDPS results in disruption of protein geranylgeranylation which in turn impairs intracellular protein trafficking. Our previous work has demonstrated that some isoprenoid triazole bisphosphonates are potent and selective inhibitors of GGDPS. To explore the possibility of selective delivery of such compounds to plasma cells, new analogues with an ω-hydroxy group have been synthesized and examined for their enzymatic and cellular activity. These studies demonstrate that incorporation of the ω-hydroxy group minimally impairs GGDPS inhibitory activity. Furthermore conjugation of one of the novel ω-hydroxy GGDPS inhibitors to hyaluronic acid resulted in enhanced cellular activity. These results will allow future studies to focus on the in vivo biodistribution of HA-conjugated GGDPS inhibitors.
Collapse
Affiliation(s)
- Nazmul H Bhuiyan
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, United States
| | - Michelle L Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Deep S Bhattacharya
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - William M Payne
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, United States; Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, United States.
| |
Collapse
|
11
|
Qi W, Yan L, Liu Y, Zhou X, Li R, Wang Y, Bai L, Chen J, Nie X. Simvastatin aggravates impaired autophagic flux in NSC34-hSOD1G93A cells through inhibition of geranylgeranyl pyrophosphate synthesis. Neuroscience 2019; 409:130-141. [DOI: 10.1016/j.neuroscience.2019.04.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/05/2019] [Accepted: 04/13/2019] [Indexed: 11/28/2022]
|
12
|
Zheng Z, Fan S, Zheng J, Huang W, Gasparetto C, Chao NJ, Hu J, Kang Y. Inhibition of thioredoxin activates mitophagy and overcomes adaptive bortezomib resistance in multiple myeloma. J Hematol Oncol 2018; 11:29. [PMID: 29482577 PMCID: PMC5828316 DOI: 10.1186/s13045-018-0575-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/12/2018] [Indexed: 11/25/2022] Open
Abstract
Background Although current chemotherapy using bortezomib (Velcade) against multiple myeloma in adults has achieved significant responses and even remission, a majority of patients will develop acquired resistance to bortezomib. Increased thioredoxin level has been reported to be associated with carcinogenesis; however, the role of thioredoxin in bortezomib drug resistance of myeloma remains unclear. Methods We generated several bortezomib-resistant myeloma cell lines by serially passaging with increased concentrations of bortezomib over a period of 1.5 years. Thioredoxin expression was measured by real-time PCR and western blot. Results The role of thioredoxin in the survival of bortezomib-resistant myeloma cells was determined by specific shRNA knockdown in vitro and in vivo. Thioredoxin inhibitor (PX12) was used to determine the effectiveness of thioredoxin inhibition in the treatment of bortezomib-resistant myeloma cells. The effect of thioredoxin inhibition on mitophagy induction was examined. The correlation of thioredoxin expression with patient overall survival was interrogated. Thioredoxin expression was significantly upregulated in bortezomib-resistant myeloma cells and the change correlated with the increase of bortezomib concentration. Thioredoxin gene knockdown using specific shRNA sensitized bortezomib-resistant myeloma cells to bortezomib efficiency in vitro and in vivo. Similarly, pharmacological inhibition with PX12 inhibited the growth of bortezomib-resistant myeloma cells and overcame bortezomib resistance in vitro and in vivo. Furthermore, inhibition of thioredoxin resulted in the activation of mitophagy and blockage of mitophagy prevented the effects of PX12 on bortezomib-resistant myeloma cells, indicating that mitophagy is the important molecular mechanism for the induction of cell death in bortezomib-resistant myeloma cells by PX12. Moreover, inhibition of thioredoxin resulted in downregulation of phosphorylated mTOR and ERK1/2. Finally, thioredoxin was overexpressed in primary myeloma cells isolated from bortezomib-resistant myeloma patients and overexpression of thioredoxin correlated with poor overall survival in patients with multiple myeloma. Conclusions Our findings demonstrated that increased thioredoxin plays a critical role in bortezomib resistance in multiple myeloma through mitophagy inactivation and increased mTOR and ERK1/2 phosphorylation. Thioredoxin provides a potential target for clinical therapeutics against multiple myeloma, particularly for bortezomib-resistant/refractory myeloma patients. Electronic supplementary material The online version of this article (10.1186/s13045-018-0575-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhihong Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.,Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Shengjun Fan
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jing Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei Huang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Cristina Gasparetto
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Nelson J Chao
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jianda Hu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA.
| |
Collapse
|
13
|
Multi-kinase inhibitors interact with sildenafil and ERBB1/2/4 inhibitors to kill tumor cells in vitro and in vivo. Oncotarget 2018; 7:40398-40417. [PMID: 27259258 PMCID: PMC5130016 DOI: 10.18632/oncotarget.9752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 05/20/2016] [Indexed: 01/12/2023] Open
Abstract
We have recently demonstrated that multi-kinase inhibitors such as sorafenib and pazopanib can suppress the detection of chaperones by in situ immuno-fluorescence, which is further enhanced by phosphodiesterase 5 inhibitors. Sorafenib and pazopanib inhibited the HSP90 ATPase activity with IC50 values of ~1.0 μM and ~75 nM, respectively. Pazopanib docked in silico with two possible poses into the HSP90 ATP binding pocket. Pazopanib and sildenafil combined to reduce the total protein levels of HSP1H/p105 and c-MYC and to reduce their co-localization. Sorafenib/pazopanib combined with sildenafil in a [GRP78+HSP27] –dependent fashion to: (i) profoundly activate an eIF2α/Beclin1 pathway; (ii) profoundly inactivate mTOR and increase ATG13 phosphorylation, collectively resulting in the formation of toxic autophagosomes. In a fresh PDX isolate of NSCLC combined knock down of [ERBB1+ERBB3] or use of the ERBB1/2/4 inhibitor afatinib altered cell morphology, enhanced ATG13 phosphorylation, inactivated NFκB, and further enhanced [sorafenib/pazopanib + sildenafil] lethality. Identical data to that with afatinib were obtained knocking down PI3K p110α/β or using buparlisib, copanlisib or the specific p110α inhibitor BYL719. Afatinib adapted NSCLC clones were resistant to buparlisib or copanlisib but were more sensitive than control clones to [sorafenib + sildenafil] or [pazopanib + sildenafil]. Lapatinib significantly enhanced the anti-tumor effect of [regorafenib + sildenafil] in vivo; afatinib and BYL719 enhanced the anti-tumor effects of [sorafenib + sildenafil] and [pazopanib] in vivo, respectively.
Collapse
|
14
|
Matthiesen RA, Varney ML, Xu PC, Rier AS, Wiemer DF, Holstein SA. α-Methylation enhances the potency of isoprenoid triazole bisphosphonates as geranylgeranyl diphosphate synthase inhibitors. Bioorg Med Chem 2018; 26:376-385. [PMID: 29248353 PMCID: PMC5752576 DOI: 10.1016/j.bmc.2017.10.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
Disruption of protein geranylgeranylation via inhibition of geranylgeranyl diphosphate synthase (GGDPS) represents a novel therapeutic strategy for a variety of malignancies, especially those characterized by excessive protein secretion such as multiple myeloma. Our work has demonstrated that some isoprenoid triazole bisphosphonates are potent and selective inhibitors of GGDPS. Here we present the synthesis and biological evaluation of a new series of isoprenoid triazoles modified by incorporation of a methyl group at the α-carbon. These studies reveal that incorporation of an α-methyl substituent enhances the potency of these compounds as GGDPS inhibitors, and, in the case of the homogeranyl/homoneryl series, abrogates the effects of olefin stereochemistry on inhibitory activity. The incorporation of the methyl group allowed preparation of a POM-prodrug, which displayed a 10-fold increase in cellular activity compared to the corresponding salt. These studies form the basis for future preclinical studies investigating the anti-myeloma activity of these novel α-methyl triazole bisphosphonates.
Collapse
Affiliation(s)
- Robert A Matthiesen
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, United States
| | - Michelle L Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Pauline C Xu
- College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Alex S Rier
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, United States
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, United States; Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, United States
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
15
|
Ranieri R, Ciaglia E, Amodio G, Picardi P, Proto MC, Gazzerro P, Laezza C, Remondelli P, Bifulco M, Pisanti S. N6-isopentenyladenosine dual targeting of AMPK and Rab7 prenylation inhibits melanoma growth through the impairment of autophagic flux. Cell Death Differ 2017; 25:353-367. [PMID: 29027991 DOI: 10.1038/cdd.2017.165] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/03/2017] [Accepted: 08/06/2017] [Indexed: 12/12/2022] Open
Abstract
Targeting the autophagic process is considered a promising therapeutic strategy in cancer since a great number of tumors, including melanoma, show high basal levels of protective autophagy that contributes to tumor progression and chemoresistance. Here, exploiting both in vitro and in vivo approaches, we identified N6-isopentenyladenosine (iPA), an end product of the mevalonate pathway, as a novel autophagy inhibitor with an interesting anti-melanoma activity. iPA, after being phosphorylated by adenosine kinase into 5'-iPA-monophosphate, induces autophagosome accumulation through AMPK activation, measured by increased fluorescent GFP-LC3 puncta and enhanced conversion into the lipidated autophagosome-associated LC3-II. However, at a later stage iPA blocks the autophagic flux monitored by p62 accumulation, Luciferase reporter-based assay for LC3 turnover in living cells and fluorescence of a tandem RFP-GFP-LC3 construct. Impaired autophagic flux is due to the block of autophagosome-lysosome fusion through the defective localization and function of Rab7, whose prenylation is inhibited by iPA, resulting in a net inhibition of autophagy completion that finally leads to melanoma apoptotic cell death. AMPK silencing prevents apoptosis upon iPA treatment, whereas basal autophagosome turnover is still inhibited due to unprenylated Rab7. These results strongly support the advantage of targeting autophagy for therapeutic gain in melanoma and provide the preclinical rational to further investigate the antitumor action of iPA, able to coordinately induce autophagosome accumulation and inhibit the autophagic flux, independently targeting AMPK and Rab7 prenylation. This property may be particularly useful for the selective killing of tumors, like melanoma, that frequently develop chemotherapy resistance due to protective autophagy activation.
Collapse
Affiliation(s)
- Roberta Ranieri
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Giuseppina Amodio
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Paola Picardi
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | | | | | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Paolo Remondelli
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Maurizio Bifulco
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Salerno, Italy
| |
Collapse
|
16
|
Haney SL, Allen C, Varney ML, Dykstra KM, Falcone ER, Colligan SH, Hu Q, Aldridge AM, Wright DL, Wiemer AJ, Holstein SA. Novel tropolones induce the unfolded protein response pathway and apoptosis in multiple myeloma cells. Oncotarget 2017; 8:76085-76098. [PMID: 29100294 PMCID: PMC5652688 DOI: 10.18632/oncotarget.18543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Tropolones are small organic compounds with metal-directing moieties. Tropolones inhibit the proliferation of cancer cell lines, possibly through their effects on metalloenzymes such as select histone deacetylases (HDACs). Pan-HDAC inhibitors are therapeutically beneficial in the treatment of multiple myeloma, however there is interest in the use of more selective HDAC inhibitor therapy to minimize adverse side effects. We hypothesized that tropolones might have anti-myeloma activities. To this end, a series of novel α-substituted tropolones were evaluated for effects on multiple myeloma cells. While all tested tropolones showed some level of cytotoxicity, MO-OH-Nap had consistently low IC50 values between 1-11 μM in all three cell lines tested and was used for subsequent experiments. MO-OH-Nap was found to induce apoptosis in a concentration-dependent manner. Time course experiments demonstrated that MO-OH-Nap promotes caspase cleavage in a time frame that was distinct from the pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). Furthermore, MO-OH-Nap- and SAHA-treated cells possess unique gene expression patterns, suggesting they promote apoptosis via different mechanisms. In particular, MO-OH-Nap increases the expression of markers associated with endoplasmic reticulum stress and the unfolded protein response. Synergistic cytotoxic effects were observed when cells were treated with the combination of MO-OH-Nap and the proteasome inhibitor bortezomib. However, treatment with MO-OH-Nap did not abrogate the bortezomib-induced increase in aggresomes, consistent with an HDAC6-independent mechanism for the observed synergy. Collectively, these finding support further investigation into the usefulness of α-substituted tropolones as anti-myeloma agents.
Collapse
Affiliation(s)
- Staci L. Haney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cheryl Allen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Michelle L. Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Eric R. Falcone
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Sean H. Colligan
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Qiang Hu
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Dennis L. Wright
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Andrew J. Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Sarah A. Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
17
|
Targeting autophagy in multiple myeloma. Leuk Res 2017; 59:97-104. [PMID: 28599191 DOI: 10.1016/j.leukres.2017.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
Abstract
Autophagy plays an important role in plasma cell ontogeny and in the pathophysiology of multiple myeloma. Autophagy is usually considered a pro-survival mechanism, and cooperates with the ubiquitin proteasome system in maintaining the homeostasis of myeloma cells by degrading excessive and misfolded proteins for energy recycling. Therefore, the inhibition of autophagy could effectively induce death in myeloma cells, and could synergize with proteasome inhibitors. However, the excessive activation of autophagy could also lead to the extreme degradation of the organelles that induce autophagic cell death. Hence, the activation of autophagic cell death might also represent a promising approach for treating myeloma. Recent studies have demonstrated that autophagy also mediates drug resistance in myeloma cells and the complications of myeloma, while the inhibition of autophagy may reverse the response to drugs. In this study, we have mainly reviewed recent research on autophagy in relationship to the therapeutic effect, the reversal of drug resistance, and the mediation of complications.
Collapse
|
18
|
Recent Advances in the Development of Mammalian Geranylgeranyl Diphosphate Synthase Inhibitors. Molecules 2017; 22:molecules22060886. [PMID: 28555000 PMCID: PMC5902023 DOI: 10.3390/molecules22060886] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 11/17/2022] Open
Abstract
The enzyme geranylgeranyl diphosphate synthase (GGDPS) catalyzes the synthesis of the 20-carbon isoprenoid geranylgeranyl diphosphate (GGPP). GGPP is the isoprenoid donor for protein geranylgeranylation reactions catalyzed by the enzymes geranylgeranyl transferase (GGTase) I and II. Inhibitors of GGDPS result in diminution of protein geranylgeranylation through depletion of cellular GGPP levels, and there has been interest in GGDPS inhibitors as potential anti-cancer agents. Here we discuss recent advances in the development of GGDPS inhibitors, including insights gained by structure-function relationships, and review the preclinical data that support the continued development of this novel class of drugs.
Collapse
|
19
|
Wills VS, Metzger JI, Allen C, Varney ML, Wiemer DF, Holstein SA. Bishomoisoprenoid triazole bisphosphonates as inhibitors of geranylgeranyl diphosphate synthase. Bioorg Med Chem 2017; 25:2437-2444. [PMID: 28302510 PMCID: PMC5450914 DOI: 10.1016/j.bmc.2017.02.066] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 11/23/2022]
Abstract
Protein geranylgeranylation reactions are dependent on the availability of geranylgeranyl diphosphate (GGDP), which serves as the isoprenoid donor. Inhibition of GGDP synthase (GGDPS) is of interest from a drug development perspective as GGDPS inhibition results in impaired protein geranylgeranylation, which in multiple myeloma, disrupts monoclonal protein trafficking and induces apoptosis. We have recently reported a series of isoprenoid triazole bisphosphonates and have demonstrated that a 3:1 mixture of homogeranyl and homoneryl isomers potently, and in a synergistic manner, inhibits GGDPS. We now present the synthesis and biological evaluation of a novel series of bishomoisoprenoid triazoles which furthers our understanding of the structure-function relationship of this class. These studies demonstrate the importance of chain length and olefin stereochemistry on inhibitory activity.
Collapse
Affiliation(s)
- Veronica S Wills
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Joseph I Metzger
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Cheryl Allen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Michelle L Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA; Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
20
|
Allen C, Kortagere S, Tong H, Matthiesen RA, Metzger JI, Wiemer DF, Holstein SA. Olefin Isomers of a Triazole Bisphosphonate Synergistically Inhibit Geranylgeranyl Diphosphate Synthase. Mol Pharmacol 2017; 91:229-236. [PMID: 28057800 DOI: 10.1124/mol.116.107326] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/28/2016] [Indexed: 11/22/2022] Open
Abstract
The isoprenoid donor for protein geranylgeranylation reactions, geranylgeranyl diphosphate (GGDP), is the product of the enzyme GGDP synthase (GGDPS) that condenses farnesyl diphosphate (FDP) and isopentenyl pyrophosphate. GGDPS inhibition is of interest from a therapeutic perspective for multiple myeloma because we have shown that targeting Rab GTPase geranylgeranylation impairs monoclonal protein trafficking, leading to endoplasmic reticulum stress and apoptosis. We reported a series of triazole bisphosphonate GGDPS inhibitors, of which the most potent was a 3:1 mixture of homogeranyl (HG) and homoneryl (HN) isomers. Here we determined the activity of the individual olefin isomers. Enzymatic and cellular assays revealed that although HN is approximately threefold more potent than HG, HN is not more potent than the original mixture. Studies in which cells were treated with varying concentrations of each isomer alone and in different combinations revealed that the two isomers potentiate the induced-inhibition of protein geranylgeranylation when used in a 3:1 HG:HN combination. A synergistic interaction was observed between the two isomers in the GGDPS enzyme assay. These results suggested that the two isomers bind simultaneously to the enzyme but within different domains. Computational modeling studies revealed that HN is preferred at the FDP site, that HG is preferred at the GGDP site, and that both isomers may bind to the enzyme simultaneously. These studies are the first to report a set of olefin isomers that synergistically inhibit GGDPS, thus establishing a new paradigm for the future development of GGDPS inhibitors.
Collapse
Affiliation(s)
- Cheryl Allen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - Sandhya Kortagere
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - Huaxiang Tong
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - Robert A Matthiesen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - Joseph I Metzger
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - David F Wiemer
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| | - Sarah A Holstein
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York (C.A.); Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.K.); Penn State Cancer Institute, Hershey, Pennsylvania (H.T.); Department of Chemistry, University of Iowa, Iowa City, Iowa (R.A.M., J.I.M., D.F.W.); and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska (S.A.H.)
| |
Collapse
|
21
|
Agabiti SS, Liang Y, Wiemer AJ. Molecular mechanisms linking geranylgeranyl diphosphate synthase to cell survival and proliferation. Mol Membr Biol 2016; 33:1-11. [PMID: 27537059 DOI: 10.1080/09687688.2016.1213432] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Geranylgeranyl diphosphate is a 20-carbon isoprenoid phospholipid whose lipid moiety can be post-translationally incorporated into proteins to promote membrane association. The process of geranylgeranylation has been implicated in anti-proliferative effects of clinical agents that inhibit enzymes of the mevalonate pathway (i.e. statins and nitrogenous bisphosphonates) as well as experimental agents that deplete geranylgeranyl diphosphate. Inhibitors of geranylgeranyl diphosphate synthase are an attractive way to block geranylgeranylation because they possess a calcium-chelating substructure to allow localization to bone and take advantage of a unique position of the enzyme within the biosynthetic pathway. Here, we describe recent advances in geranylgeranyl diphosphate synthase expression and inhibitor development with a particular focus on the molecular mechanisms that link geranylgeranyl diphosphate to cell proliferation via geranylgeranylated small GTPases.
Collapse
Affiliation(s)
- Sherry S Agabiti
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Yilan Liang
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA
| | - Andrew J Wiemer
- a Department of Pharmaceutical Sciences , University of Connecticut , Storrs , CT , USA.,b Institute for Systems Genomics, University of Connecticut , Storrs , CT , USA
| |
Collapse
|
22
|
Signore IA, Jerez C, Figueroa D, Suazo J, Marcelain K, Cerda O, Colombo Flores A. Inhibition of the 3-hydroxy-3-methyl-glutaryl-CoA reductase induces orofacial defects in zebrafish. ACTA ACUST UNITED AC 2016; 106:814-830. [PMID: 27488927 DOI: 10.1002/bdra.23546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/13/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Orofacial clefts (OFCs) are common birth defects, which include a range of disorders with a complex etiology affecting formation of craniofacial structures. Some forms of syndromic OFCs are produced by defects in the cholesterol pathway. The principal enzyme of the cholesterol pathway is the 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR). Our aim is to study whether defects of HMGCR function would produce orofacial malformation similar to those found in disorders of cholesterol synthesis. METHODS We used zebrafish hmgcrb mutants and HMGCR inhibition assay using atorvastatin during early and late stages of orofacial morphogenesis in zebrafish. To describe craniofacial phenotypes, we stained cartilage and bone and performed in situ hybridization using known craniofacial markers. Also, we visualized neural crest cell migration in a transgenic fish. RESULTS Our results showed that mutants displayed loss of cartilage and diminished orofacial outgrowth, and in some cases palatal cleft. Late treatments with statin show a similar phenotype. Affected-siblings displayed a moderate phenotype, whereas early-treated embryos had a minor cleft. We found reduced expression of the downstream component of Sonic Hedgehog-signaling gli1 in ventral brain, oral ectoderm, and pharyngeal endoderm in mutants and in late atorvastatin-treated embryos. CONCLUSION Our results suggest that HMGCR loss-of-function primarily affects postmigratory cranial neural crest cells through abnormal Sonic Hedgehog signaling, probably induced by reduction in metabolites of the cholesterol pathway. Malformation severity correlates with the grade of HMGCR inhibition, developmental stage of its disruption, and probably with availability of maternal lipids. Together, our results might help to understand the spectrum of orofacial phenotypes found in cholesterol synthesis disorders. Birth Defects Research (Part A) 106:814-830, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iskra A Signore
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Filosofía y Ciencias de la Complejidad (IFICC), Santiago, Chile
| | - Carolina Jerez
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Diego Figueroa
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - José Suazo
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Katherine Marcelain
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Oscar Cerda
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo Flores
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile. .,Servicio de Anatomía Patológica, Hospital Clínico de la Universidad de Chile, Santiago, Chile.
| |
Collapse
|